1
|
Shortiss C, Howard L, Mcmahon SS. Viability and Suitability Of Rat Pup Organotypic Spinal Cord Slice Cultures In a Transection Injury Model. J Neurosci Methods 2025:110499. [PMID: 40419025 DOI: 10.1016/j.jneumeth.2025.110499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2025] [Revised: 05/09/2025] [Accepted: 05/23/2025] [Indexed: 05/28/2025]
Abstract
BACKGROUND Ex vivo organotypic spinal cord slice culture (OSC) models are advantageous for spinal cord injury (SCI) research. They retain in vivo cellular interactions but involve fewer ethical concerns and are easier to manipulate than in vivo models. Due to improved viability OSCs derived from early postnatal animals are preferred. A transection injury OSC model derived from postnatal day (P) 4 rats has been validated, however as rodent spinal cord development changes significantly in early postnatal weeks, older postnatal day animals may be more appropriate for OSC models. NEW METHOD This study compared the viability of OSCs derived from P11 rats with the established P4 transection model. RESULTS No significant differences in cell viability were found between P4 and P11 control slices, or between control and injured slices within each age group. P11 OSCs also demonstrated strong viable cell ingrowth into a transection injury gap post-injury, comparable to P4 OSCs, and increased their volume fraction of immunostaining for the glial scar markers GFAP and CSPGs along transection gap edges. Sex was not found to affect any of these parameters. COMPARISON WITH EXISTING METHODS Our findings demonstrate that P11 OSCs maintain viability and cellular ingrowth post transection injury comparable to that of the well-established P4 derived OSCs. P11 OSCs were also found to exhibit a glial scarring response post transection injury. CONCLUSIONS OSCs from P11 animals maintain viability comparable to P4 OSCs while also exhibiting a scarring response. This study concludes that P11 OSCs are a valid alternate to P4 OSC transection SCI model.
Collapse
Affiliation(s)
- Ciara Shortiss
- Anatomy, School of Medicine, University of Galway, Galway, Ireland; Regenerative Medicine Institute, University of Galway, Galway, Ireland
| | - Linda Howard
- Regenerative Medicine Institute, University of Galway, Galway, Ireland
| | - Siobhan S Mcmahon
- Anatomy, School of Medicine, University of Galway, Galway, Ireland; Galway Neuroscience Centre, University of Galway, Galway, Ireland
| |
Collapse
|
2
|
Singh N, Pathak Z, Kumar H. Rab27a-mediated extracellular vesicle release drives astrocytic CSPG secretion and glial scarring in spinal cord injury. BIOMATERIALS ADVANCES 2025; 176:214357. [PMID: 40414081 DOI: 10.1016/j.bioadv.2025.214357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 05/15/2025] [Accepted: 05/16/2025] [Indexed: 05/27/2025]
Abstract
Traumatic spinal cord injury (SCI) prevents axonal regeneration by impairing neuronal function and causing glial scarring. Chondroitin sulfate proteoglycans (CSPGs) from astrocytes drive this process, yet the release mechanism, potentially involving extracellular vesicles (EVs), remains unclear. Rab27a releases EVs from multivesicular bodies (MVBs) by enabling their docking and fusion with the plasma membrane. We confirmed Rab27a expression, and its localization, subsequently, EV release was validated with CD9, Alix, and TSG101 markers. Rab27a-mediated EV release was confirmed in both Rab27a-induced and Rab27a-siRNA-treated cells. Rab27a-derived EVs inhibited neuronal cell growth, while Rab27a-siRNA EVs promoted neuronal growth. Our study also observed upregulated Rab27a expression in the rat contusion model of SCI. Further analysis showed increased CSPG expression in Rab27a-induced conditions via the Rho/ROCK pathway, with altered pAkt, and β-tubulin III, levels. Immunohistochemistry confirmed CSPG/Rab27a/GFAP and CSPG/CD9 co-localization in tissue sections, verifying that Rab27a mediates EV release containing CSPG from astrocytes. These findings suggest that Rab27a plays a crucial role in CSPG release via EVs and scar formation. Functional recovery was significantly improved with Rab27a-siRNA treatment, suggesting Rab27a as a potential target for astrocytic scar modulation in SCI. The study reveals the detailed mechanistic insight of Rab27a-dependent CSPG release via EVs for sub-acute scar formation in contusion SCI.
Collapse
Affiliation(s)
- Nidhi Singh
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER) Ahmedabad, India
| | - Zarna Pathak
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER) Ahmedabad, India
| | - Hemant Kumar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER) Ahmedabad, India.
| |
Collapse
|
3
|
Gu Q, Fan H, Zhang S, Xia S, Tan X, Zhou X. Analysis of the Autophagy-related Gene NLRC4 in spinal cord injury. Neurol Res 2025:1-16. [PMID: 40357725 DOI: 10.1080/01616412.2025.2503462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2025] [Accepted: 04/30/2025] [Indexed: 05/15/2025]
Abstract
BACKGROUND/OBJECTIVES To investigate the role of the autophagy-related gene NLR Family CARD Domain Containing 4 (NLRC4) in spinal cord injury via bioinformatics methods, which may provide new targets for the diagnosis and treatment of spinal cord injury. METHODS This analysis is based on the GEO database dataset GSE151371. To identify potential autophagy-related genes involved in SCI, protein‒protein interaction (PPI) networks were analyzed. Immune microenvironment analysis (LM22) was performed via the CIBERSORTx database to determine the makeup of 22 immune cell types. Furthermore, a rat spinal cord injury model was generated, and the expression of selected autophagy-related genes was validated via immunofluorescence labeling and Western blotting. RESULTS Disease enrichment analysis via the Metascape database revealed enrichment for diseases related to the spinal cord, inflammation, infection, and immunity, which aligns with the functional analysis results of previously identified genes. Through the PPI and autophagy-related genes, we identified NLRC4 within the key subnetwork of the PPI network, highlighting its significance as a key signature gene associated with SCI. NLRC4 expression was significantly increased in the three groups, which was correlated with the severity of SCI. In the rat SCI model, NLRC4 protein expression was significantly greater in the SCI group than in the sham group (p < 0.001), confirming the validity of the model. CONCLUSIONS Since NLRC4 is an important gene involved in the autophagy that leads to spinal cord damage, it can be utilized to illuminate the optimal approach to immunotherapy for individuals with SCI and uncover new targets for therapy.
Collapse
Affiliation(s)
- Qie Gu
- Department of Spine Surgery, Center for Orthopedic Surgery, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Hongbo Fan
- Department of Neurocritical Care Medicine, The Second Hospital of Dalian Medical University, Dalian, China
| | - Siqi Zhang
- Department of Spine Surgery, Center for Orthopedic Surgery, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Shuaishuai Xia
- Department of Spine Surgery, Center for Orthopedic Surgery, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Xuemei Tan
- Department of Spine Surgery, Center for Orthopedic Surgery, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Xiang Zhou
- Department of Spine Surgery, Center for Orthopedic Surgery, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
4
|
Lin M, Alimerzaloo F, Wang X, Alhalabi O, Krieg SM, Skutella T, Younsi A. Harnessing stem cell-derived exosomes: a promising cell-free approach for spinal cord injury. Stem Cell Res Ther 2025; 16:182. [PMID: 40247394 PMCID: PMC12004558 DOI: 10.1186/s13287-025-04296-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Accepted: 03/25/2025] [Indexed: 04/19/2025] Open
Abstract
Spinal cord injury (SCI) is a severe injury to the central nervous system that often results in permanent neurological dysfunction. Current treatments have limited efficacy and face challenges in restoring neurological function after injury. Recently, stem cell-derived exosomes have gained attention as an experimental treatment for SCI due to their unique properties, including superior biocompatibility, minimal immunogenicity and non-tumorigenicity. With their potential as a cell-free therapy, exosomes promote SCI repair by enhancing nerve regeneration, reducing inflammation and stabilizing the blood-spinal cord barrier. This review summarizes advances in stem cell-derived exosome research for SCI over the past years, focusing on their mechanisms and future prospects. Despite their promising therapeutic potential, clinical translation remains challenging due to standardization of exosome isolation protocols, compositional consistency and long-term safety profiles that require further investigation.
Collapse
Affiliation(s)
- Miaoman Lin
- Department of Neurosurgery, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120, Heidelberg, Germany
- Medical Faculty, Heidelberg University, Heidelberg, Germany
| | - Farzaneh Alimerzaloo
- Medical Faculty, Heidelberg University, Heidelberg, Germany
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Xingjin Wang
- Department of Neurosurgery, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120, Heidelberg, Germany
- Medical Faculty, Heidelberg University, Heidelberg, Germany
| | - Obada Alhalabi
- Department of Neurosurgery, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120, Heidelberg, Germany
- Medical Faculty, Heidelberg University, Heidelberg, Germany
| | - Sandro M Krieg
- Department of Neurosurgery, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120, Heidelberg, Germany
- Medical Faculty, Heidelberg University, Heidelberg, Germany
| | - Thomas Skutella
- Medical Faculty, Heidelberg University, Heidelberg, Germany
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Alexander Younsi
- Department of Neurosurgery, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120, Heidelberg, Germany.
- Medical Faculty, Heidelberg University, Heidelberg, Germany.
| |
Collapse
|
5
|
Shen Z, Feng B, Lim WL, Woo T, Liu Y, Vicenzi S, Wang J, Kwon BK, Zou Y. Astrocytic Ryk signaling coordinates scarring and wound healing after spinal cord injury. Proc Natl Acad Sci U S A 2025; 122:e2417400122. [PMID: 40208942 PMCID: PMC12012454 DOI: 10.1073/pnas.2417400122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 02/25/2025] [Indexed: 04/12/2025] Open
Abstract
Wound healing after spinal cord injury involves highly coordinated interactions among multiple cell types, which are poorly understood. Astrocytes play a central role in creating a border against the non-neural lesion core. To do so, astrocytes undergo dramatic morphological changes by first thickening and elongating their processes and then overlapping them to form a physical barrier. We show here that the expression of a cell-surface receptor, Ryk, is induced in astrocytes after injury in both rodent and human spinal cords. Astrocyte-specific knockout of Ryk dramatically elongated the reactive astrocytes, accelerated the formation of the border, and reduced the size of the scar. Astrocyte-specific knockout of Ryk also accelerated the injury responses of multiple cell types. Single-cell transcriptomics analyses revealed a broad range of changes in cell signaling among astrocytes, microglia, fibroblasts, and endothelial cells after astrocyte-specific Ryk knockout, suggesting that Ryk not only regulates injury responses of astrocytes but may also regulate signals emanating from astrocytes and coordinate the responses of these cell types. The elongation of astrocyte processes is mediated by NrCAM, a cell adhesion molecule induced by astrocyte-specific conditional knockout of Ryk after spinal cord injury. Our findings suggest that Ryk is a promising therapeutic target to accelerate wound healing, promote neuronal survival, and enhance functional recovery.
Collapse
Affiliation(s)
- Zhe Shen
- Department of Neurobiology, School of Biological Sciences, University of California, San Diego, La Jolla, CA92093
| | - Bo Feng
- Department of Neurobiology, School of Biological Sciences, University of California, San Diego, La Jolla, CA92093
| | - Wei Ling Lim
- Department of Neurobiology, School of Biological Sciences, University of California, San Diego, La Jolla, CA92093
| | - Timothy Woo
- Department of Neurobiology, School of Biological Sciences, University of California, San Diego, La Jolla, CA92093
| | - Yanlin Liu
- Department of Neurobiology, School of Biological Sciences, University of California, San Diego, La Jolla, CA92093
| | - Silvia Vicenzi
- Department of Neurobiology, School of Biological Sciences, University of California, San Diego, La Jolla, CA92093
| | - Jingyi Wang
- Department of Neurobiology, School of Biological Sciences, University of California, San Diego, La Jolla, CA92093
| | - Brian K. Kwon
- Department of Orthopaedics, International Collaboration on Repair Discoveries, University of British Columbia, Vancouver, BCV5Z 1M9, Canada
| | - Yimin Zou
- Department of Neurobiology, School of Biological Sciences, University of California, San Diego, La Jolla, CA92093
| |
Collapse
|
6
|
He C, Wang Y, Gong W, Zhang S. Targeted Delivery of Acid-Responsive Rutin Nanoparticles Based on Aldehyde Adsorption for the Treatment of Spinal Cord Injury in Rats. ACS Biomater Sci Eng 2025; 11:2192-2202. [PMID: 40167167 DOI: 10.1021/acsbiomaterials.5c00038] [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: 04/02/2025]
Abstract
Spinal cord injury (SCI) can cause irreversible nerve damage, imposing a significant burden on both patients and society. Methylprednisolone (MP), the recommended clinical drug, possesses antioxidant, anti-inflammatory, and antiapoptotic effects. It improves nerve damage by inhibiting secondary pathological processes. However, high-dose MP administration may result in side effects, including diabetes, femoral head necrosis, and infections. Therefore, there is a need to identify safer alternatives to mitigate the issues associated with MP administration. Rutin, a natural small molecule, exhibits multifaceted therapeutic capabilities and high biosafety, making it a promising alternative to MP treatment. However, its poor solubility and rapid metabolism limit its in vivo bioavailability. In this study, a drug-free polypeptide (PAH) containing hydrazide groups on the side chains is designed, which can be used for mitigating secondary SCI through scavenging toxic aldehydes. Then, we utilize PAH to encapsulate rutin and develop aldehyde-responsive nanomedicine for intravenous administration in SCI rats, providing a novel approach for the clinical replacement of MP.
Collapse
Affiliation(s)
- Chuanyu He
- Department of Spinal Surgery, Center of Orthopedics, The First Hospital of Jilin University, Changchun, Jilin 130021, People's Republic of China
- Jilin Engineering Research Center for Spine and Spinal Cord Injury, Changchun 130021, China
- Department of Orthopedics, the Sixth Affiliated Hospital of Xinjiang Medical University, Urumqi 830002, China
| | - Yongjie Wang
- Department of Spinal Surgery, Center of Orthopedics, The First Hospital of Jilin University, Changchun, Jilin 130021, People's Republic of China
- Jilin Engineering Research Center for Spine and Spinal Cord Injury, Changchun 130021, China
| | - Weiquan Gong
- Department of Spinal Surgery, Center of Orthopedics, The First Hospital of Jilin University, Changchun, Jilin 130021, People's Republic of China
- Jilin Engineering Research Center for Spine and Spinal Cord Injury, Changchun 130021, China
| | - Shaokun Zhang
- Department of Spinal Surgery, Center of Orthopedics, The First Hospital of Jilin University, Changchun, Jilin 130021, People's Republic of China
- Jilin Engineering Research Center for Spine and Spinal Cord Injury, Changchun 130021, China
| |
Collapse
|
7
|
Cieri MB, Ramos AJ. Astrocytes, reactive astrogliosis, and glial scar formation in traumatic brain injury. Neural Regen Res 2025; 20:973-989. [PMID: 38989932 PMCID: PMC11438322 DOI: 10.4103/nrr.nrr-d-23-02091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 04/14/2024] [Indexed: 07/12/2024] Open
Abstract
Traumatic brain injury is a global health crisis, causing significant death and disability worldwide. Neuroinflammation that follows traumatic brain injury has serious consequences for neuronal survival and cognitive impairments, with astrocytes involved in this response. Following traumatic brain injury, astrocytes rapidly become reactive, and astrogliosis propagates from the injury core to distant brain regions. Homeostatic astroglial proteins are downregulated near the traumatic brain injury core, while pro-inflammatory astroglial genes are overexpressed. This altered gene expression is considered a pathological remodeling of astrocytes that produces serious consequences for neuronal survival and cognitive recovery. In addition, glial scar formed by reactive astrocytes is initially necessary to limit immune cell infiltration, but in the long term impedes axonal reconnection and functional recovery. Current therapeutic strategies for traumatic brain injury are focused on preventing acute complications. Statins, cannabinoids, progesterone, beta-blockers, and cerebrolysin demonstrate neuroprotective benefits but most of them have not been studied in the context of astrocytes. In this review, we discuss the cell signaling pathways activated in reactive astrocytes following traumatic brain injury and we discuss some of the potential new strategies aimed to modulate astroglial responses in traumatic brain injury, especially using cell-targeted strategies with miRNAs or lncRNA, viral vectors, and repurposed drugs.
Collapse
Affiliation(s)
- María Belén Cieri
- Laboratorio de Neuropatología Molecular, IBCN UBA-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | | |
Collapse
|
8
|
Yari-Ilkhchi A, Hamidi N, Mahkam M, Ebrahimi-Kalan A. Graphene-based materials: an innovative approach for neural regeneration and spinal cord injury repair. RSC Adv 2025; 15:9829-9853. [PMID: 40165920 PMCID: PMC11956154 DOI: 10.1039/d4ra07976k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Accepted: 02/17/2025] [Indexed: 04/02/2025] Open
Abstract
Spinal cord injury (SCI), the most serious disease affecting the central nervous system (CNS), is one of contemporary medicine's most difficult challenges, causing patients to suffer physically, emotionally, and socially. However, due to recent advances in medical science and biomaterials, graphene-based materials (GBMs) have tremendous potential in SCI therapy due to their wonderful and valuable properties, such as physicochemical properties, extraordinary electrical conductivity, distinct morphology, and high mechanical strength. This review discusses SCI pathology and GBM characteristics, as well as recent in vitro and in vivo findings on graphenic scaffolds, electrodes, and injectable achievements for SCI improvement using neuroprotective and neuroregenerative techniques to improve neural structural and functional repair. Additionally, it suggests possible ideas and desirable products for graphene-based technological advances, intending to reach therapeutic importance for SCI.
Collapse
Affiliation(s)
- Ayda Yari-Ilkhchi
- Chemistry Department, Faculty of Science, Azarbaijan Shahid Madani University 5375171379 Tabriz Iran
- Faculty of Chemical and Metallurgical Engineering, Department of Chemical Engineering, Istanbul Technical University Maslak 34469 Istanbul Turkey
- Faculty of Engineering and Natural Sciences, Sabanci University 34956 Istanbul Turkey
| | - Nazila Hamidi
- Department of Chemistry and Biochemistry, The University of Tulsa Tulsa OK 74104 USA
| | - Mehrdad Mahkam
- Chemistry Department, Faculty of Science, Azarbaijan Shahid Madani University 5375171379 Tabriz Iran
| | - Abbas Ebrahimi-Kalan
- Faculty of Advanced Medical Science, Tabriz University of Medical Sciences 5166614733 Tabriz Iran
| |
Collapse
|
9
|
Tang W, Wang A, Liu S, Wen G, Qi H, Gu Y, Xu C, Ren S, Zhang S, He Y. Calycosin regulates astrocyte reactivity and astrogliosis after spinal cord injury by targeting STAT3 phosphorylation. J Neuroimmunol 2025; 400:578535. [PMID: 39954615 DOI: 10.1016/j.jneuroim.2025.578535] [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: 09/12/2024] [Revised: 01/23/2025] [Accepted: 01/27/2025] [Indexed: 02/17/2025]
Abstract
BACKGROUND Astrocytes are the most populous glial cells in the central nervous system (CNS), which can exert detrimental effects through a process of reactive astrogliosis. Our previous study has indicated the potential effect of Calycosin in preventing spinal cord injury (SCI). This study aims to investigate the mechanism by which calycosin regulates the polarization of A1 astrocytes, a neurotoxic subtype of reactive astrocytes, in SCI models. MATERIALS AND METHODS The SCI model was induced by applying mechanical compression to the spinal cord using vascular clamps. A1 astrocyte differentiation was induced by treating astrocytes with microglia supernatant obtained after Lipopolysaccharide (LPS) stimulation. Key protein expression levels were analyzed by Western blotting, and astrocyte markers such as CS56, GFAP, C3, S100A10 were assessed through immunofluorescence staining. RESULTS Calycosin treatment significantly reduced glial scar formation and C3 expression in SCI rats. However, S100A10 expression remained unchanged. Further analysis showed that Calycosin inhibited A1 astrocyte activation, migration, and invasion, which was associated with STAT3 phosphorylation. Calycosin downregulated p-STAT3 levels in both A1 astrocytes and SCI rats. These effects were reversed by Colivelin (a STAT3 activator) in A1 astrocytes. CONCLUSION Calycosin treatment can modulate p-STAT3 expression, thereby altering the functionality of astrocytes during the recovery phase and positively impacting the treatment and rehabilitation of SCI.
Collapse
Affiliation(s)
- Wenhai Tang
- Department of Spine Surgery, The Second Affiliated Hospital of Hainan Medical University, Haikou 570216, China
| | - Aitao Wang
- Department of Anesthesiology, Hohhot First Hospital, Hohhot 010030, China
| | - Shengxing Liu
- Department of Spine Surgery, The Second Affiliated Hospital of Hainan Medical University, Haikou 570216, China
| | - Guangyu Wen
- Department of Spine Surgery, The Second Affiliated Hospital of Hainan Medical University, Haikou 570216, China
| | - Hao Qi
- Department of Spine Surgery, The Second Affiliated Hospital of Hainan Medical University, Haikou 570216, China
| | - Yuntao Gu
- Department of Spine Surgery, The Second Affiliated Hospital of Hainan Medical University, Haikou 570216, China
| | - Chunzhao Xu
- Department of Spine Surgery, The Second Affiliated Hospital of Hainan Medical University, Haikou 570216, China
| | - Shanwu Ren
- Department of Spine Surgery, The Second Affiliated Hospital of Hainan Medical University, Haikou 570216, China
| | - Shunli Zhang
- Department of Spine Surgery, The Second Affiliated Hospital of Hainan Medical University, Haikou 570216, China.
| | - Yongxiong He
- Department of Orthopedics, Beijing Chest Hospital, Capital Medical University, Beijing 101149, China.
| |
Collapse
|
10
|
Khavandegar A, Ahmadi NS, Mousavi MA, Ramezani Z, Khodadoust E, Hasan Zadeh Tabatabaei MS, Hasanpour Segherlou Z, Zeinaddini-Meymand A, Nasehi F, Moafi M, RayatSanati K, Masoomi R, Hamidi S, Pourkhodadad S, Rahimi-Movaghar V. The potential role of RhoA/ROCK-inhibition on locomotor recovery after spinal cord injury: a systematic review of in-vivo studies. Spinal Cord 2025; 63:95-126. [PMID: 39956860 DOI: 10.1038/s41393-025-01064-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Revised: 01/27/2025] [Accepted: 02/06/2025] [Indexed: 02/18/2025]
Abstract
STUDY DESIGN Systematic Review. OBJECTIVES To thoroughly assess the existing literature regarding the impact of anti-RhoA/ROCK agents or procedures on functional recovery in animal models of SCI. SETTING Sina Trauma and Surgery Research Center, Tehran University of Medical Sciences. METHODS A comprehensive search was conducted in Ovid MEDLINE, Embase, Scopus, and Web of Science Core Collection utilizing a combination of keywords. All in-vivo animal studies of acute or chronic SCI that evaluated the pharmacological effects of Rho/ROCK inhibitors in English literature were included in this study. RESULTS Totally, 2320 articles were identified, of which, 60 papers were included for further analysis. A total of 47 (78%) studies were conducted merely on rats, 9 (15%) on mice, 3 (5%) used both, and the remaining used other animals. Y-27632, Fasudil, C3 Transferase and its derivatives (C3-05/PEP-C3/CT04/C3bot154-182/C3bot26mer(156-181)), Ibuprofen, Electroacupuncture (EA), SiRhoA, miR-133b, miR-135-5p, miR-381, miR-30b, Statins, 17β-estradiol, β-elemene, Lentivirus-mediated PGC-1a, Repulsive guidance molecule (RGMa), Local profound hypothermia, Jisuikang (JSK), Hyperbaric oxygen (HBO), Lv-shRhoA (Notch-1 inhibitor), Anti-Ryk antibody, LINGO-antagonist, BA-210, p21Cip1/WAF1, ORL-1 antagonist, Epigallocatechin-3-gallate (EGCG), Tamsulosin, AAV.ULK1.DN, and Indomethacin were the 28 reported agents/procedures with anti-RhoA/ROCK effects. The pooled SMD for BBB scores was 0.41 (p = 0.048) in the first week, 0.85 (p < 0.001) in the second week, 1.22 (p = 0.010) in the third week, and 1.53 (p = 0.001) in the fourth week. CONCLUSION Of the 28 identified anti-RhoA/ROCK agents, all but two (C3bot and its derivatives and EGCG) demonstrated promising results. The results of the meta-analysis cautiously indicate a significant increase in BBB scores over time after SCI.
Collapse
Affiliation(s)
- Armin Khavandegar
- Sina Trauma and Surgery Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Negar Sadat Ahmadi
- Sina Trauma and Surgery Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Alsadat Mousavi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zahra Ramezani
- Sina Trauma and Surgery Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Elaheh Khodadoust
- Sina Trauma and Surgery Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | | | | | | | - Fatemeh Nasehi
- Sina Trauma and Surgery Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Maral Moafi
- Cell Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kimia RayatSanati
- Sina Trauma and Surgery Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Rasool Masoomi
- Sina Trauma and Surgery Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Sorour Hamidi
- Department of Neurosurgery, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Soheila Pourkhodadad
- Department of Pharmacy and Chemical Biology, Emory University, School of Medicine, Atlanta, GA, USA
| | - Vafa Rahimi-Movaghar
- Sina Trauma and Surgery Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
11
|
Gupta SJ, Churchward MA, Todd KG, Winship IR. A dual role for pleiotrophin in modulating inflammation and myelination in the presence of chondroitin sulfate proteoglycans after nervous system injury. Front Cell Neurosci 2025; 19:1549433. [PMID: 40083634 PMCID: PMC11903471 DOI: 10.3389/fncel.2025.1549433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Accepted: 02/10/2025] [Indexed: 03/16/2025] Open
Abstract
Chondroitin sulfate proteoglycans (CSPGs), key components of the extracellular matrix and the glial scar that forms around central nervous system (CNS) injuries, are recognized for hindering neuronal regeneration. We previously demonstrated the potential of pleiotrophin (PTN) to induce neurite outgrowth even in the presence of inhibitory CSPGs. The effects of PTN on microglia and oligodendrocytes are not well described. Here, we examined how PTN administration alters the differentiation of oligodendrocyte precursor cells (OPCs) into mature oligodendrocytes in the presence of CSPGs using in vitro cell culture model. Moreover, we explored the effects of PTN on the inflammatory activity of microglia with and without inflammatory stimulation (IFN-γ) in a CSPG-rich environment. The data showed that the CSPG matrix inhibited the differentiation of OPCs into mature oligodendrocytes. PTN induced dose-dependent differentiation of OPCs into mature oligodendrocytes, with an optimal effect at 10 nM PTN. Moreover, PTN modified the immunological response of microglia in the presence of CSPGs, with reduced proinflammatory activity that was further reduced by PTN administration, in contrast to the increased release of matrix metalloproteinases (MMP 9). However, when IFN-γ-activated microglia were treated with PTN, proinflammatory signaling was stimulated at higher PTN concentrations (10 nM and 100 nM). Overall, our results indicate that PTN can overcome the inhibitory effect of CSPGs on the differentiation of OPCs into oligodendrocytes and can modulate inflammation mediated by CSPGs from microglia. Collectively, these findings demonstrate that PTN can effectively counteract the inhibitory effects of CSPGs on the differentiation of OPCs into mature oligodendrocytes while also modulating microglial responses to reduce proinflammatory activity and increase MMP-9 release. Thus, PTN has great potential to improve remyelination and neuroprotective strategies in the treatment of demyelinating diseases or any injury.
Collapse
Affiliation(s)
- Somnath J. Gupta
- Neurochemical Research Unit, Department of Psychiatry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| | - Matthew A. Churchward
- Neurochemical Research Unit, Department of Psychiatry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
- Department of Biological Sciences, Concordia University of Edmonton, Edmonton, AB, Canada
| | - Kathryn G. Todd
- Neurochemical Research Unit, Department of Psychiatry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| | - Ian R. Winship
- Neurochemical Research Unit, Department of Psychiatry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
12
|
Nutt K, Dombros-Ryan Z, Birea R, Franks EV, Eastham S, Godwin M, Adams CF, Chari DM, Jenkins SI. Electrospun Polycaprolactone (PCL) Nanofibers Induce Elongation and Alignment of Co-Cultured Primary Cortical Astrocytes and Neurons. MICROMACHINES 2025; 16:256. [PMID: 40141867 PMCID: PMC11946388 DOI: 10.3390/mi16030256] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 01/27/2025] [Accepted: 02/18/2025] [Indexed: 03/28/2025]
Abstract
Neuromimetic in vitro models, simulating in vivo architecture/organization, are urgently needed to reduce experimental reliance on live animals. Our group recently reported a novel brain tissue derivation protocol, simultaneously deriving all major cortical cell types (including immune cells) in a facile protocol, generating a network of neurons in a single growth medium, which was interfaced with nanomaterials. This represents a significant advance, as tissue engineers overwhelmingly use diverse methods to derive and combine individual brain cells for materials-interfacing. However, this multicellular model lacked cellular directionality/structural organization (unlike the highly organized cortical circuits in vivo). Synthetic nanofiber constructs are of high value in tissue engineering, providing directional cues for cells. Most neuro-nanofiber studies employ simple monocultures of astrocytes/neurons and commonly use peripheral neurons rather than central nervous system populations. Here, we have interfaced our complex brain model (neurons/astrocytes derived simultaneously) with randomly oriented or aligned polycaprolactone (PCL) fiber meshes. Both cell types showed targeted extension along aligned fibers versus coverslips or random fibers. A new analysis method developed in-house demonstrated that peak orientations for astrocytes and neurons correlated with aligned nanofibers. Our data support the concept that nanofiber scaffolds can achieve organized growth of mixed cortical neural cell populations, mimicking neural architecture.
Collapse
Affiliation(s)
- Kayleigh Nutt
- Neural Tissue Engineering: Keele (NTEK), Keele University, Keele ST5 5BG, UK (Z.D.-R.); (E.V.F.); (C.F.A.)
- School of Life Sciences, Keele University, Keele ST5 5BG, UK
| | - Zoe Dombros-Ryan
- Neural Tissue Engineering: Keele (NTEK), Keele University, Keele ST5 5BG, UK (Z.D.-R.); (E.V.F.); (C.F.A.)
- School of Life Sciences, Keele University, Keele ST5 5BG, UK
| | - Ruxandra Birea
- Neural Tissue Engineering: Keele (NTEK), Keele University, Keele ST5 5BG, UK (Z.D.-R.); (E.V.F.); (C.F.A.)
- School of Life Sciences, Keele University, Keele ST5 5BG, UK
| | - Emily Victoria Franks
- Neural Tissue Engineering: Keele (NTEK), Keele University, Keele ST5 5BG, UK (Z.D.-R.); (E.V.F.); (C.F.A.)
- School of Medicine, Keele University, Keele ST5 5BG, UK
| | - Sarah Eastham
- Neural Tissue Engineering: Keele (NTEK), Keele University, Keele ST5 5BG, UK (Z.D.-R.); (E.V.F.); (C.F.A.)
- Department of Biomedical Engineering, University of Strathclyde, Glasgow G4 0NW, UK
| | - Morgan Godwin
- Neural Tissue Engineering: Keele (NTEK), Keele University, Keele ST5 5BG, UK (Z.D.-R.); (E.V.F.); (C.F.A.)
- School of Life Sciences, Keele University, Keele ST5 5BG, UK
| | - Chris F. Adams
- Neural Tissue Engineering: Keele (NTEK), Keele University, Keele ST5 5BG, UK (Z.D.-R.); (E.V.F.); (C.F.A.)
- School of Life Sciences, Keele University, Keele ST5 5BG, UK
| | - Divya Maitreyi Chari
- Neural Tissue Engineering: Keele (NTEK), Keele University, Keele ST5 5BG, UK (Z.D.-R.); (E.V.F.); (C.F.A.)
- School of Medicine, Keele University, Keele ST5 5BG, UK
| | - Stuart Iain Jenkins
- Neural Tissue Engineering: Keele (NTEK), Keele University, Keele ST5 5BG, UK (Z.D.-R.); (E.V.F.); (C.F.A.)
- School of Medicine, Keele University, Keele ST5 5BG, UK
| |
Collapse
|
13
|
Paracha M, Brezinski AN, Singh R, Sinson E, Satkunendrarajah K. Targeting Spinal Interneurons for Respiratory Recovery After Spinal Cord Injury. Cells 2025; 14:288. [PMID: 39996760 PMCID: PMC11854602 DOI: 10.3390/cells14040288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 02/07/2025] [Accepted: 02/11/2025] [Indexed: 02/26/2025] Open
Abstract
Spinal interneurons (SpINs) are pivotal to the function of neural circuits, orchestrating motor, sensory, and autonomic functions in the healthy, intact central nervous system. These interneurons (INs) are heterogeneous, with diverse types contributing to various neural systems, including those that control respiratory function. Research in the last few decades has highlighted the complex involvement of SpINs in modulating motor control. SpINs also partake in motor plasticity by aiding in adapting and rewiring neural circuits in response to injury or disease. This plasticity is crucial in the context of spinal cord injury (SCI), where damage often leads to severe and long-term breathing deficits. Such deficits are a leading cause of morbidity and mortality in individuals with SCI, emphasizing the need for effective interventions. This review will focus on SpIN circuits involved in the modulation of breathing and explore current and emerging approaches that leverage SpINs as therapeutic targets to promote respiratory recovery following SCI.
Collapse
Affiliation(s)
- Maha Paracha
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, WI 53226, USA; (M.P.); (A.N.B.); (R.S.); (E.S.)
- Clement J. Zablocki Veterans Affairs Medical Center, Milwaukee, WI 53295, USA
| | - Allison N. Brezinski
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, WI 53226, USA; (M.P.); (A.N.B.); (R.S.); (E.S.)
- Clement J. Zablocki Veterans Affairs Medical Center, Milwaukee, WI 53295, USA
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Rhea Singh
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, WI 53226, USA; (M.P.); (A.N.B.); (R.S.); (E.S.)
| | - Elizabeth Sinson
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, WI 53226, USA; (M.P.); (A.N.B.); (R.S.); (E.S.)
| | - Kajana Satkunendrarajah
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, WI 53226, USA; (M.P.); (A.N.B.); (R.S.); (E.S.)
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| |
Collapse
|
14
|
Gao Y, Wang H, Shi L, Lu P, Dai G, Zhang M, Han B, Cao M, Li Y, Rui Y. Erroneous Differentiation of Tendon Stem/Progenitor Cells in the Pathogenesis of Tendinopathy: Current Evidence and Future Perspectives. Stem Cell Rev Rep 2025; 21:423-453. [PMID: 39579294 DOI: 10.1007/s12015-024-10826-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] [Accepted: 11/20/2024] [Indexed: 11/25/2024]
Abstract
Tendinopathy is a condition characterized by persistent tendon pain, structural damage, and compromised functionality. Presently, the treatment for tendinopathy remains a formidable challenge, partly because of its unclear pathogenesis. Tendon stem/progenitor cells (TSPCs) are essential for tendon homeostasis, regeneration, remodeling, and repair. An innovative theory has been previously proposed, with insufficient evidence, that the erroneous differentiation of TSPCs may constitute one of the fundamental mechanisms underpinning tendinopathy. Over the past few years, there has been accumulating evidence for plausibility of this theory. In this review, we delve into alterations in the differentiation potential of TSPCs and the underlying mechanisms in the context of injury-induced tendinopathy, diabetic tendinopathy, and age-related tendinopathy to provide updated evidence on the erroneous differentiation theory. Despite certain limitations inherent in the existing body of evidence, the erroneous differentiation theory emerges as a promising and highly pertinent avenue for understanding tendinopathy. In the future, advanced methodologies will be harnessed to further deepen comprehension of this theory, paving the way for prospective developments in clinical therapies targeting TSPCs for the management of tendinopathy.
Collapse
Affiliation(s)
- Yucheng Gao
- Department of Orthopaedics, School of Medicine, Zhongda Hospital, Southeast University, No. 87 Ding Jia Qiao, Nanjing, 210009, Jiangsu, China
- School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China
| | - Hao Wang
- Department of Orthopaedics, School of Medicine, Zhongda Hospital, Southeast University, No. 87 Ding Jia Qiao, Nanjing, 210009, Jiangsu, China
- School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China
| | - Liu Shi
- Department of Orthopaedics, School of Medicine, Zhongda Hospital, Southeast University, No. 87 Ding Jia Qiao, Nanjing, 210009, Jiangsu, China
- School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China
| | - Panpan Lu
- Department of Orthopaedics, School of Medicine, Zhongda Hospital, Southeast University, No. 87 Ding Jia Qiao, Nanjing, 210009, Jiangsu, China
- School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China
| | - Guangchun Dai
- Department of Orthopaedics, School of Medicine, Zhongda Hospital, Southeast University, No. 87 Ding Jia Qiao, Nanjing, 210009, Jiangsu, China
- School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China
| | - Ming Zhang
- Department of Orthopaedics, School of Medicine, Zhongda Hospital, Southeast University, No. 87 Ding Jia Qiao, Nanjing, 210009, Jiangsu, China
- School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China
| | - Bowen Han
- School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China
| | - Mumin Cao
- Department of Orthopaedics, School of Medicine, Zhongda Hospital, Southeast University, No. 87 Ding Jia Qiao, Nanjing, 210009, Jiangsu, China
- School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China
| | - Yingjuan Li
- Department of Geriatrics, School of Medicine, Zhongda Hospital, Southeast University, Nanjing, 210009, Jiangsu, China
| | - Yunfeng Rui
- Department of Orthopaedics, School of Medicine, Zhongda Hospital, Southeast University, No. 87 Ding Jia Qiao, Nanjing, 210009, Jiangsu, China.
- School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China.
| |
Collapse
|
15
|
Hajinejad M, Far BF, Gorji A, Sahab-Negah S. The effects of self-assembling peptide on glial cell activation. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:1391-1402. [PMID: 39305327 DOI: 10.1007/s00210-024-03415-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Accepted: 08/26/2024] [Indexed: 02/14/2025]
Abstract
Glial cells play a critical role in the healthy and diseased phases of the central nervous system (CNS). CNS diseases involve a wide range of pathological conditions characterized by poor recovery of neuronal function. Glial cell-related target therapies are progressively gaining interest in inhibiting secondary injury-related death. Modulation of the extracellular matrix by artificial scaffolds plays a critical role in the behavior of glial cells after injury. Among numerous types of scaffolds, self-assembling peptides (SAPs) notably give attention to the design of a proper biophysical and biomechanical microenvironment for cellular homeostasis and tissue regeneration. Implementing SAPs in an injured brain can induce neural differentiation in transplanted stem cells, reducing inflammation and inhibiting glial scar formation. In this review, we investigate the recent findings to elucidate the pivotal role of SAPs in orchestrating the most pivotal secondary response following CNS injury. Notably, we explore their impact on the activation of glial cells and their modulatory effects on microglial and astrocytic polarization.
Collapse
Affiliation(s)
- Mehrdad Hajinejad
- Qaen Faculty of Medical Sciences, Birjand University of Medical Sciences, Birjand, Iran
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Bahareh Farasati Far
- Department of Chemistry, Iran University of Science and Technology, Tehran, Iran
| | - Ali Gorji
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran
- Epilepsy Research Center, Neurosurgery Department, Münster University, Münster, Germany
- Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Pardis Campus, Azadi Square, Kalantari Blvd., Mashhad, Iran
| | - Sajad Sahab-Negah
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
- Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Pardis Campus, Azadi Square, Kalantari Blvd., Mashhad, Iran.
| |
Collapse
|
16
|
Hajimirzaei P, Tabatabaei FSA, Nasibi-Sis H, Razavian RS, Nasirinezhad F. Schwann cell transplantation for remyelination, regeneration, tissue sparing, and functional recovery in spinal cord injury: A systematic review and meta-analysis of animal studies. Exp Neurol 2025; 384:115062. [PMID: 39579959 DOI: 10.1016/j.expneurol.2024.115062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 11/02/2024] [Accepted: 11/12/2024] [Indexed: 11/25/2024]
Abstract
INTRODUCTION Spinal cord injury (SCI) is a significant global health challenge that results in profound physical and neurological impairments. Despite progress in medical care, the treatment options for SCI are still restricted and often focus on symptom management rather than promoting neural repair and functional recovery. This study focused on clarifying the impact of Schwann cell (SC) transplantation on the molecular, cellular, and functional basis of recovery in animal models of SCI. MATERIAL AND METHODS Relevant studies were identified by conducting searches across multiple databases, which included PubMed, Web of Science, Scopus, and ProQuest. The data were analyzed via comprehensive meta-analysis software. We assessed the risk of bias via the SYRCLE method. RESULTS The analysis included 59 studies, 48 of which provided quantitative data. The results revealed significant improvements in various outcome variables, including protein zero structures (SMD = 1.66, 95 %CI: 0.96-2.36; p < 0.001; I2 = 49.8 %), peripherally myelinated axons (SMD = 1.81, 95 %CI: 0.99-2.63; p < 0.001; I2 = 39.3 %), biotinylated dextran amine-labeled CST only rostral (SMD = 1.31, 95 % CI: 0.50-2.12, p < 0.01, I2 = 49.7 %), fast blue-labeled reticular formation (SMD = 0.96, 95 %CI: 0.43-1.49, p < 0.001, I2 = 0.0 %), 5-hydroxytryptamine caudally (SMD = 0.83, 95 %CI: 0.36-1.29, p < 0.001, I2 = 17.2 %) and epicenter (SMD = 0.85, 95 %CI: 0.17-1.53, p < 0.05, I2 = 62.7 %), tyrosine hydroxylase caudally (SMD = 1.86, 95 %CI: 1.14-2.59, p < 0.001, I2 = 0.0 %) and epicenter (SMD = 1.82, 95 %CI: 1.18-2.47, p < 0.001, I2 = 0.0 %), cavity volume (SMD = -2.07, 95 %CI: -2.90 - -1.24, p < 0.001, I2 = 67.2 %), and Basso, Beattie, and Bresnahan (SMD = 1.26, 95 %CI: 0.93-1.58; p < 0.001; I2 = 79.4 %). CONCLUSIONS This study demonstrates the promising potential of SC transplantation as a therapeutic approach for SCI, clarifying its impact on various biological processes critical for recovery.
Collapse
Affiliation(s)
- Pooya Hajimirzaei
- Department of Radiation Sciences, Allied Medicine Faculty, Iran University of Medical Sciences, Tehran, Iran; Radiation Biology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | | | - Hamed Nasibi-Sis
- Department of Medical Library and Information Sciences, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Farinaz Nasirinezhad
- Department of Physiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran; Physiology Research Center, Iran University of Medical sciences, Tehran, Iran; Center of Experimental and Comparative Study, Iran University of Medical sciences, Tehran, Iran.
| |
Collapse
|
17
|
Poongodi R, Hsu YW, Yang TH, Huang YH, Yang KD, Lin HC, Cheng JK. Stem Cell-Derived Extracellular Vesicle-Mediated Therapeutic Signaling in Spinal Cord Injury. Int J Mol Sci 2025; 26:723. [PMID: 39859437 PMCID: PMC11765593 DOI: 10.3390/ijms26020723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 01/14/2025] [Accepted: 01/14/2025] [Indexed: 01/27/2025] Open
Abstract
Mesenchymal stem cell-derived extracellular vesicles (MSC-EVs) have emerged as a promising therapeutic strategy for spinal cord injury (SCI). These nanosized vesicles possess unique properties such as low immunogenicity and the ability to cross biological barriers, making them ideal carriers for delivering bioactive molecules to injured tissues. MSC-EVs have been demonstrated to exert multiple beneficial effects in SCI, including reducing inflammation, promoting neuroprotection, and enhancing axonal regeneration. Recent studies have delved into the molecular mechanisms underlying MSC-EV-mediated therapeutic effects. Exosomal microRNAs (miRNAs) have been identified as key regulators of various cellular processes involved in SCI pathogenesis and repair. These miRNAs can influence inflammation, oxidative stress, and apoptosis by modulating gene expression. This review summarized the current state of MSC-EV-based therapies for SCI, highlighting the underlying mechanisms and potential clinical applications. We discussed the challenges and limitations of translating these therapies into clinical practice, such as inconsistent EV production, complex cargo composition, and the need for targeted delivery strategies. Future research should focus on optimizing EV production and characterization, identifying key therapeutic miRNAs, and developing innovative delivery systems to maximize the therapeutic potential of MSC-EVs in SCI.
Collapse
Affiliation(s)
- Raju Poongodi
- Department of Medical Research, MacKay Memorial Hospital, Taipei 10449, Taiwan; (R.P.); (T.-H.Y.)
| | - Yung-Wei Hsu
- Department of Anesthesiology, MacKay Memorial Hospital, Taipei 10449, Taiwan; (Y.-W.H.); (Y.-H.H.)
- Department of Medicine, MacKay Medical College, New Taipei City 25245, Taiwan
| | - Tao-Hsiang Yang
- Department of Medical Research, MacKay Memorial Hospital, Taipei 10449, Taiwan; (R.P.); (T.-H.Y.)
| | - Ya-Hsien Huang
- Department of Anesthesiology, MacKay Memorial Hospital, Taipei 10449, Taiwan; (Y.-W.H.); (Y.-H.H.)
- Department of Medicine, MacKay Medical College, New Taipei City 25245, Taiwan
| | - Kuender D. Yang
- Institute of Long-Term Care, MacKay Medical College, New Taipei City 25245, Taiwan;
- MacKay Children’s Hospital, Taipei 10449, Taiwan
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
| | - Hsin-Chieh Lin
- Department of Materials Science and Engineering, National Yang Ming Chiao Tung University, Hsinchu 300093, Taiwan;
- Center for Intelligent Drug Systems and Smart Bio-Devices (IDS2B), National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan
| | - Jen-Kun Cheng
- Department of Medical Research, MacKay Memorial Hospital, Taipei 10449, Taiwan; (R.P.); (T.-H.Y.)
- Department of Anesthesiology, MacKay Memorial Hospital, Taipei 10449, Taiwan; (Y.-W.H.); (Y.-H.H.)
- Department of Medicine, MacKay Medical College, New Taipei City 25245, Taiwan
| |
Collapse
|
18
|
Siebert JR, Kennedy K, Osterhout DJ. Neurons Are Not All the Same: Diversity in Neuronal Populations and Their Intrinsic Responses to Spinal Cord Injury. ASN Neuro 2025; 17:2440299. [PMID: 39819292 PMCID: PMC11877619 DOI: 10.1080/17590914.2024.2440299] [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: 01/19/2025] Open
Abstract
Functional recovery following spinal cord injury will require the regeneration and repair of damaged neuronal pathways. It is well known that the tissue response to injury involves inflammation and the formation of a glial scar at the lesion site, which significantly impairs the capacity for neuronal regeneration and functional recovery. There are initial attempts by both supraspinal and intraspinal neurons to regenerate damaged axons, often influenced by the neighboring tissue pathology. Many experimental therapeutic strategies are targeted to further stimulate the initial axonal regrowth, with little consideration for the diversity of the affected neuronal populations. Notably, recent studies reveal that the neuronal response to injury is variable, based on multiple factors, including the location of the injury with respect to the neuronal cell bodies and the affected neuronal populations. New insights into regenerative mechanisms have shown that neurons are not homogenous but instead exhibit a wide array of diversity in their gene expression, physiology, and intrinsic responses to injury. Understanding this diverse intrinsic response is crucial, as complete functional recovery requires the successful coordinated regeneration and reorganization of various neuron pathways.
Collapse
Affiliation(s)
- Justin R. Siebert
- Physician Assistant Studies Program, Department of Health Care and Administration, Slippery Rock University of Pennsylvania, Slippery Rock, PA, USA
| | - Kiersten Kennedy
- Norton College of Medicine, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Donna J. Osterhout
- Department of Cell & Developmental Biology, SUNY Upstate Medical University, Syracuse, NY, USA
| |
Collapse
|
19
|
Mao D, Chen Q, Tong S, Xu Z, Yu G, Chang C, Lv Y. Integrated bioinformatics analysis identified cuproptosis-related hub gene Mpeg1 as potential biomarker in spinal cord injury. Sci Rep 2025; 15:1993. [PMID: 39814871 PMCID: PMC11736097 DOI: 10.1038/s41598-025-86170-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 01/08/2025] [Indexed: 01/18/2025] Open
Abstract
Spinal cord injury (SCI) is a profound ailment lacking a well-defined molecular mechanism and effective treatments. Cuproptosis, identified as a recently discovered cell death pathway, exhibits diverse roles in various cancers. Nevertheless, its involvement in SCI is yet to be elucidated. Firstly, the RNA sequencing data of 1, 3, 7 dpi SCI samples were collected from GEO database. We performed differential expression analysis on these samples with varying cuproptosis-related scores calculating by ssGSEA. Subsequently, we conducted enrichment analyses with KEGG, GO, and GSEA. Simultaneously, we executed WGCNA analysis using cuproptosis-related scores, selecting the most relevant module for enrichment analysis. Hub genes were identified at the intersection of PPI analysis results from two modules and cuproptosis-related DEGs. Additionally, relying on the immune infiltration landscape associated with cuproptosis, we carried out immune cell correlation analysis on hub genes. Finally, to corroborate our earlier findings, we utilized single-cell RNA-seq analysis and in vitro experimental validation. Based on ssGSEA, differential expression analysis and WGCNA analysis, we identified two modules that were highly relevant to cell division and immune processes, respectively. From these modules, we identified two hub genes, Cd48 and Mpeg1, which exhibited a strong positive correlation (R = 0.92) and shared similar pathways. Furthermore, we observed a positive correlation between M2 macrophages and Cd48/Mpeg1. To validate our findings, we performed external cohort validation using a single-cell RNA sequencing dataset. The results confirmed that Mpeg1 was highly expressed in microglia (macrophages in center nervous system) following spinal cord injury. Additionally, we conducted in vitro experiments to further validate the molecular functions of Mpeg1 in SCI. In summary, targeting Mpeg1, as well as cuproptosis and immune cell infiltration, holds promise as a potential strategy for reducing spinal cord tissue damage and promoting recovery after SCI. These findings provide valuable insights for future therapeutic interventions.
Collapse
Affiliation(s)
- Dandan Mao
- Department of Neurosurgery, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, China
| | - Qi Chen
- Department of Nursing, Huashan Hospital, Fudan University, Shanghai, China
| | - Shuolan Tong
- The First Clinical Medical College, Wenzhou Medical University, Wenzhou, China
| | - Zixia Xu
- The First Clinical Medical College, Wenzhou Medical University, Wenzhou, China
| | - Guofeng Yu
- Department of Neurosurgery, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, China
| | - Chuan Chang
- Department of Neurosurgery, Huashan hospital, Fudan University, Shanghai, China.
| | - Yao Lv
- Department of Neurosurgery, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, China.
| |
Collapse
|
20
|
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
|
21
|
Sekiya T, Holley MC. The Glial Scar: To Penetrate or Not for Motor Pathway Restoration? Cell Transplant 2025; 34:9636897251315271. [PMID: 40152462 PMCID: PMC11951902 DOI: 10.1177/09636897251315271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 12/25/2024] [Accepted: 01/08/2025] [Indexed: 03/29/2025] Open
Abstract
Although notable progress has been made, restoring motor function from the brain to the muscles continues to be a substantial clinical challenge in motor neuron diseases/disorders such as spinal cord injury (SCI). While cell transplantation has been widely explored as a potential therapeutic method for reconstructing functional motor pathways, there remains considerable opportunity for enhancing its therapeutic effectiveness. We reviewed studies on motor pathway regeneration to identify molecular and ultrastructural cues that could enhance the efficacy of cell transplantation. While the glial scar is often cited as an intractable barrier to axon regeneration, this mainly applies to axons trying to penetrate its "core" to reach the opposite side. However, the glial scar exhibits a "duality," with an anti-regenerative core and a pro-regenerative "surface." This surface permissiveness is attributed to pro-regenerative molecules, such as laminin in the basement membrane (BM). Transplanting donor cells onto the BM, which forms plastically after injury, may significantly enhance the efficacy of cell transplantation. Specifically, forming detour pathways between transplanted cells and endogenous propriospinal neurons on the pro-regenerative BM may efficiently bypass the intractable scar core and promote motor pathway regeneration. We believe harnessing the tissue's innate repair capacity is crucial, and targeting post-injury plasticity in astrocytes and Schwann cells, especially those associated with the BM that has predominantly been overlooked in the field of SCI research, can advance motor system restoration to a new stage. A shift in cell delivery routes-from the traditional intra-parenchymal (InP) route to the transplantation of donor cells onto the pro-regenerative BM via the extra-parenchymal (ExP) route-may signify a transformative step forward in neuro-regeneration research. Practically, however, the complementary use of both InP and ExP methods may offer the most substantial benefit for restoring motor pathways. We aim for this review to deepen the understanding of cell transplantation and provide a framework for evaluating the efficacy of this therapeutic modality in comparison to others.
Collapse
Affiliation(s)
- Tetsuji Sekiya
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Department of Neurological Surgery, Hikone Chuo Hospital, Hikone, Japan
| | - Matthew C. Holley
- Department of Biomedical Science, University of Sheffield, Sheffield, England
| |
Collapse
|
22
|
Díaz-Pérez S, DeLong JH, Rivier CA, Lee CY, Askenase MH, Zhu B, Zhang L, Brennand KJ, Martins AJ, Sansing LH. Single-nucleus RNA sequencing of human periventricular white matter in vascular dementia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.06.627202. [PMID: 39713290 PMCID: PMC11661092 DOI: 10.1101/2024.12.06.627202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Vascular dementia (VaD) refers to a variety of dementias driven by cerebrovascular disease and is the second leading cause of dementia globally. VaD may be caused by ischemic strokes, intracerebral hemorrhage, and/or cerebral small vessel disease, commonly identified as white matter hyperintensities on MRI. The mechanisms underlying these white matter lesions in the periventricular brain are poorly understood. In this study we perform an extensive transcriptomic analysis on human postmortem periventricular white matter lesions in patients with VaD with the goal of identifying molecular pathways in the disease. We find increased cellular stress responses in astrocytes, oligodendrocytes, and oligodendrocyte precursor cells as well as transcriptional and translational repression in microglia in our dataset. We show that several genes identified by GWAS as being associated with white matter disease are differentially expressed in cells in VaD. Finally, we compare our dataset to an independent snRNAseq dataset of PVWM in VaD and a scRNAseq dataset on human iPSC-derived microglia exposed to oxygen glucose deprivation (OGD). We identify the increase of the heat shock protein response as a conserved feature of VaD across celltypes and show that this increase is not linked to OGD exposure. Overall, our study is the first to show that increased heat shock protein responses are a common feature of lesioned PVWM in VaD and may represent a potential therapeutic target.
Collapse
Affiliation(s)
| | - Jonathan H. DeLong
- Department of Neurology, Yale University School of Medicine, New Haven, CT
| | - Cyprien A. Rivier
- Department of Neurology, Yale University School of Medicine, New Haven, CT
| | - Chia-Yi Lee
- Department of Genetics, Yale University School of Medicine, New Haven, CT
| | - Michael H. Askenase
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT
| | - Biqing Zhu
- Program of Computational Biology and Bioinformatics, Yale University School of Medicine, New Haven, CT
| | - Le Zhang
- Department of Neurology, Yale University School of Medicine, New Haven, CT
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT
| | - Kristen J. Brennand
- Department of Genetics, Yale University School of Medicine, New Haven, CT
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT
| | - Andrew J. Martins
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT
| | - Lauren H. Sansing
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT
- Department of Neurology, Yale University School of Medicine, New Haven, CT
| |
Collapse
|
23
|
Sámano C, Mazzone GL. The role of astrocytes response triggered by hyperglycaemia during spinal cord injury. Arch Physiol Biochem 2024; 130:724-741. [PMID: 37798949 DOI: 10.1080/13813455.2023.2264538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 09/22/2023] [Indexed: 10/07/2023]
Abstract
OBJECTIVE This manuscript aimed to provide a comprehensive overview of the physiological, molecular, and cellular mechanisms triggered by reactive astrocytes (RA) in the context of spinal cord injury (SCI), with a particular focus on cases involving hyperglycaemia. METHODS The compilation of articles related to astrocyte responses in neuropathological conditions, with a specific emphasis on those related to SCI and hyperglycaemia, was conducted by searching through databases including Science Direct, Web of Science, and PubMed. RESULTS AND CONCLUSIONS This article explores the dual role of astrocytes in both neurophysiological and neurodegenerative conditions within the central nervous system (CNS). In the aftermath of SCI and hyperglycaemia, astrocytes undergo a transformation into RA, adopting a distinct phenotype. While there are currently no approved therapies for SCI, various therapeutic strategies have been proposed to alleviate the detrimental effects of RAs following SCI and hyperglycemia. These strategies show promising potential in the treatment of SCI and its likely comorbidities.
Collapse
Affiliation(s)
- C Sámano
- Departamento de Ciencias Naturales, Universidad Autónoma Metropolitana, Unidad Cuajimalpa (UAM-C), Ciudad de México, México
| | - G L Mazzone
- Instituto de Investigaciones en Medicina Traslacional (IIMT), CONICET-Universidad Austral, Pilar, Buenos Aires, Argentina
- Facultad de Ciencias Biomédicas, Universidad Austral, Pilar, Buenos Aires, Argentina
| |
Collapse
|
24
|
Zucha D, Abaffy P, Kirdajova D, Jirak D, Kubista M, Anderova M, Valihrach L. Spatiotemporal transcriptomic map of glial cell response in a mouse model of acute brain ischemia. Proc Natl Acad Sci U S A 2024; 121:e2404203121. [PMID: 39499634 PMCID: PMC11573666 DOI: 10.1073/pnas.2404203121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 09/30/2024] [Indexed: 11/07/2024] Open
Abstract
The role of nonneuronal cells in the resolution of cerebral ischemia remains to be fully understood. To decode key molecular and cellular processes that occur after ischemia, we performed spatial and single-cell transcriptomic profiling of the male mouse brain during the first week of injury. Cortical gene expression was severely disrupted, defined by inflammation and cell death in the lesion core, and glial scar formation orchestrated by multiple cell types on the periphery. The glial scar was identified as a zone with intense cell-cell communication, with prominent ApoE-Trem2 signaling pathway modulating microglial activation. For each of the three major glial populations, an inflammatory-responsive state, resembling the reactive states observed in neurodegenerative contexts, was observed. The recovered spectrum of ischemia-induced oligodendrocyte states supports the emerging hypothesis that oligodendrocytes actively respond to and modulate the neuroinflammatory stimulus. The findings are further supported by analysis of other spatial transcriptomic datasets from different mouse models of ischemic brain injury. Collectively, we present a landmark transcriptomic dataset accompanied by interactive visualization that provides a comprehensive view of spatiotemporal organization of processes in the postischemic mouse brain.
Collapse
Affiliation(s)
- Daniel Zucha
- Laboratory of Gene Expression, Institute of Biotechnology of the Czech Academy of Sciences, Vestec 25250, Czech Republic
- Department of Informatics and Chemistry, Faculty of Chemical Technology, University of Chemistry and Technology, Prague 16000, Czech Republic
| | - Pavel Abaffy
- Laboratory of Gene Expression, Institute of Biotechnology of the Czech Academy of Sciences, Vestec 25250, Czech Republic
| | - Denisa Kirdajova
- Laboratory of Gene Expression, Institute of Biotechnology of the Czech Academy of Sciences, Vestec 25250, Czech Republic
- Department of Cellular Neurophysiology, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague 14220, Czech Republic
| | - Daniel Jirak
- Department of Radiodiagnostic and Interventional Radiology, Institute of Clinical and Experimental Medicine, Prague 14021, Czech Republic
- Faculty of Health Studies, Technical University of Liberec, Liberec 46001, Czech Republic
| | - Mikael Kubista
- Laboratory of Gene Expression, Institute of Biotechnology of the Czech Academy of Sciences, Vestec 25250, Czech Republic
| | - Miroslava Anderova
- Department of Cellular Neurophysiology, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague 14220, Czech Republic
| | - Lukas Valihrach
- Laboratory of Gene Expression, Institute of Biotechnology of the Czech Academy of Sciences, Vestec 25250, Czech Republic
| |
Collapse
|
25
|
Mansor NI, Balqis TN, Lani MN, Lye KL, Nor Muhammad NA, Ismail WIW, Abidin SZ. Nature's Secret Neuro-Regeneration Pathway in Axolotls, Polychaetes and Planarians for Human Therapeutic Target Pathways. Int J Mol Sci 2024; 25:11904. [PMID: 39595973 PMCID: PMC11593954 DOI: 10.3390/ijms252211904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 11/03/2024] [Accepted: 11/04/2024] [Indexed: 11/28/2024] Open
Abstract
Despite significant improvements in the comprehension of neuro-regeneration, restoring nerve injury in humans continues to pose a substantial therapeutic difficulty. In the peripheral nervous system (PNS), the nerve regeneration process after injury relies on Schwann cells. These cells play a crucial role in regulating and releasing different extracellular matrix proteins, including laminin and fibronectin, which are essential for facilitating nerve regeneration. However, during regeneration, the nerve is required to regenerate for a long distance and, subsequently, loses its capacity to facilitate regeneration during this progression. Meanwhile, it has been noted that nerve regeneration has limited capabilities in the central nervous system (CNS) compared to in the PNS. The CNS contains factors that impede the regeneration of axons following injury to the axons. The presence of glial scar formation results from this unfavourable condition, where glial cells accumulate at the injury site, generating a physical and chemical barrier that hinders the regeneration of neurons. In contrast to humans, several species, such as axolotls, polychaetes, and planarians, possess the ability to regenerate their neural systems following amputation. This ability is based on the vast amount of pluripotent stem cells that have the remarkable capacity to differentiate and develop into any cell within their body. Although humans also possess these cells, their numbers are extremely limited. Examining the molecular pathways exhibited by these organisms has the potential to offer a foundational understanding of the human regeneration process. This review provides a concise overview of the molecular pathways involved in axolotl, polychaete, and planarian neuro-regeneration. It has the potential to offer a new perspective on therapeutic approaches for neuro-regeneration in humans.
Collapse
Affiliation(s)
- Nur Izzati Mansor
- Department of Nursing, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras 56000, Kuala Lumpur, Malaysia;
| | - Tengku Nabilatul Balqis
- Faculty of Science and Marine Environment, Universiti Malaysia Terengganu, Kuala Nerus 21030, Terengganu, Malaysia; (T.N.B.); (W.I.W.I.)
| | - Mohd Nizam Lani
- Faculty of Fisheries and Food Science, Universiti Malaysia Terengganu, Kuala Nerus 21030, Terengganu, Malaysia;
| | - Kwan Liang Lye
- ME Scientifique Sdn Bhd, Taman Universiti Indah, Seri Kembangan 43300, Selangor, Malaysia;
| | - Nor Azlan Nor Muhammad
- Institute of Systems Biology (INBIOSIS), Universiti Kebangsaan Malaysia, Bangi 43600, Selangor, Malaysia;
| | - Wan Iryani Wan Ismail
- Faculty of Science and Marine Environment, Universiti Malaysia Terengganu, Kuala Nerus 21030, Terengganu, Malaysia; (T.N.B.); (W.I.W.I.)
- Research Interest Group Biological Security and Sustainability (BIOSES), Faculty of Science and Marine Environment, Universiti Malaysia Terengganu, Kuala Nerus 21030, Terengganu, Malaysia
| | - Shahidee Zainal Abidin
- Faculty of Science and Marine Environment, Universiti Malaysia Terengganu, Kuala Nerus 21030, Terengganu, Malaysia; (T.N.B.); (W.I.W.I.)
- Research Interest Group Biological Security and Sustainability (BIOSES), Faculty of Science and Marine Environment, Universiti Malaysia Terengganu, Kuala Nerus 21030, Terengganu, Malaysia
| |
Collapse
|
26
|
Saadinam F, Azami M, Pedram MS, Sadeghinezhad J, Jabbari Fakhr M, Salimi A, Aminianfar H, Molazem M, Mokhber Dezfouli MR, Dehghan MM. Injectable alginate chitosan hydrogel as a promising bioengineered therapy for acute spinal cord injury. Sci Rep 2024; 14:26747. [PMID: 39500959 PMCID: PMC11538431 DOI: 10.1038/s41598-024-77995-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 10/28/2024] [Indexed: 11/08/2024] Open
Abstract
Dealing with spinal cord injuries presents problematic due to multiple secondary mechanisms. Beyond primary concerns like paralysis and disability, complications including urinary, gastrointestinal, cardiac, and respiratory disorders, along with substantial economic burdens may occur. Limited research focuses on modeling and treating contusion and compression injuries. Tissue engineering emerges as an innovative treatment, targeting lesion pathophysiology. This study was evaluated implanting injectable biomaterials into injury-induced cavity before glial scar formation, avoiding tissue incisions and minimizing further damage. The efficacy of injectable alginate/thiolated chitosan hydrogel was investigated for acute spinal cord injury induced by Vanický method in Wistar rats. Three days post-injury, hydrogel was administrated through microinjection after laminectomy. After 60 days, the hydrogel group demonstrated notable motor function enhancement compared to the control by the BBB locomotor test (P < 0.05). However, no statistically significant differences were observed in MRI assessment concerning lesion severity. Stereological and histopathological evaluations revealed a reduction in vacuole volume and the presence of axon profiles within the scaffold (P < 0.05), alongside reduced infiltration of inflammatory and Gitter cells in the hydrogel group, although the latter was not statistically significant compared to the control. Thiolated chitosan/ alginate hydrogel implantation may be regarded as a promising treatment to enhance motor function by restraining destructive processes post-acute spinal cord injury.
Collapse
Affiliation(s)
- Fatemeh Saadinam
- Department of Surgery and Diagnostic Imaging, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
- Institute of Biomedical Research, University of Tehran, Tehran, Iran
| | - Mahmoud Azami
- Department of Tissue Engineering and Applied Cell sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mir Sepehr Pedram
- Department of Surgery and Diagnostic Imaging, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
- Institute of Biomedical Research, University of Tehran, Tehran, Iran
| | - Javad Sadeghinezhad
- Department of Basic Sciences, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Massoumeh Jabbari Fakhr
- Institute of Biomedical Research, University of Tehran, Tehran, Iran
- Department of Tissue Engineering and Applied Cell sciences, School of Medicine, Qom University of Medical Science and Health Services, Qom, Iran
| | - Atena Salimi
- Department of Surgery and Diagnostic Imaging, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Hossein Aminianfar
- Institute of Biomedical Research, University of Tehran, Tehran, Iran
- Department of Pathology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Mohammad Molazem
- Department of Surgery and Diagnostic Imaging, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | | | - Mohammad Mehdi Dehghan
- Department of Surgery and Diagnostic Imaging, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran.
- Institute of Biomedical Research, University of Tehran, Tehran, Iran.
| |
Collapse
|
27
|
Jiang H, Zhang C, Lin M, Yin Y, Deng S, Liu W, Zhuo B, Tian G, Du Y, Meng Z. Deciphering the mechanistic impact of acupuncture on the neurovascular unit in acute ischemic stroke: Insights from basic research in a narrative review. Ageing Res Rev 2024; 101:102536. [PMID: 39384155 DOI: 10.1016/j.arr.2024.102536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 09/27/2024] [Accepted: 10/01/2024] [Indexed: 10/11/2024]
Abstract
Ischemic stroke(IS), a severe acute cerebrovascular disease, not only imposes a heavy economic burden on society but also presents numerous challenges in treatment. During the acute phase, while thrombolysis and thrombectomy serve as primary treatments, these approaches are restricted by a narrow therapeutic window. During rehabilitation, commonly used neuroprotective agents struggle with their low drug delivery efficiency and inadequate preclinical testing, and the long-term pharmacological and toxicity effects of nanomedicines remain undefined. Meanwhile, acupuncture as a therapeutic approach is widely acknowledged for its effectiveness in treating IS and has been recommended by the World Health Organization (WHO) as an alternative and complementary therapy, even though its exact mechanisms remain unclear. This review aims to summarize the known mechanisms of acupuncture and electroacupuncture (EA) in the treatment of IS. Research shows that acupuncture treatment mainly protects the neurovascular unit through five mechanisms: 1) reducing neuronal apoptosis and promoting neuronal repair and proliferation; 2) maintaining the integrity of the blood-brain barrier (BBB); 3) inhibiting the overactivation and polarization imbalance of microglia; 4) regulating the movement of vascular smooth muscle (VSM) cells; 5) promoting the proliferation of oligodendrocyte precursors. Through an in-depth analysis, this review reveals the multi-level, multi-dimensional impact of acupuncture treatment on the neurovascular unit (NVU) following IS, providing stronger evidence and a theoretical basis for its clinical application.
Collapse
Affiliation(s)
- Hailun Jiang
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Chao Zhang
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Mengxuan Lin
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Yu Yin
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Shizhe Deng
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Wei Liu
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Bifang Zhuo
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Guang Tian
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Yuzheng Du
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China.
| | - Zhihong Meng
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China.
| |
Collapse
|
28
|
Shen Z, Feng B, Lim WL, Woo T, Liu Y, Vicenzi S, Wang J, Kwon BK, Zou Y. Astrocytic Ryk signaling coordinates scarring and wound healing after spinal cord injury. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.16.618727. [PMID: 39463959 PMCID: PMC11507886 DOI: 10.1101/2024.10.16.618727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Wound healing after spinal cord injury involves highly coordinated interactions among multiple cell types, which is poorly understood. Astrocytes play a central role in creating a border against the non-neural lesion core. To do so, astrocytes undergo dramatic morphological changes by first thickening the processes and then elongating and overlap them. We show here show that the expression of a cell-surface receptor, Ryk, is induced in astrocytes after injury in both rodent and human spinal cord. Astrocyte-specific knockout of Ryk dramatically elongated the reactive astrocytes and accelerated the formation of the border and reduced the size of the scar. Astrocyte-specific knockout of Ryk also accelerated the injury responses of multiple cell types, including the resolution of neuroinflammation. Single cell transcriptomics analyses revealed a broad range of changes cell signaling among astrocytes, microglia, fibroblasts, endothelial cell, etc, after astrocyte-specific Ryk knockout, suggesting that Ryk not only regulates the injury response of astrocytes but may also regulate signals which coordinate the responses of multiple cell types. The elongation is mediated by NrCAM, a cell adhesion molecule induced by astrocyte-specific conditional knockout of Ryk after spinal cord injury. Our findings suggest a promising therapeutic target to accelerate wound healing and promote neuronal survival and enhance functional recovery.
Collapse
|
29
|
Hosseini SM, Nemati S, Karimi-Abdolrezaee S. Astrocytes originated from neural stem cells drive the regenerative remodeling of pathologic CSPGs in spinal cord injury. Stem Cell Reports 2024; 19:1451-1473. [PMID: 39303705 PMCID: PMC11561464 DOI: 10.1016/j.stemcr.2024.08.007] [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: 05/09/2024] [Revised: 08/20/2024] [Accepted: 08/21/2024] [Indexed: 09/22/2024] Open
Abstract
Neural degeneration is a hallmark of spinal cord injury (SCI). Multipotent neural precursor cells (NPCs) have the potential to reconstruct the damaged neuron-glia network due to their tri-lineage capacity to generate neurons, astrocytes, and oligodendrocytes. However, astrogenesis is the predominant fate of resident or transplanted NPCs in the SCI milieu adding to the abundant number of resident astrocytes in the lesion. How NPC-derived astrocytes respond to the inflammatory milieu of SCI and the mechanisms by which they contribute to the post-injury recovery processes remain largely unknown. Here, we uncover that activated NPC-derived astrocytes exhibit distinct molecular signature that is immune modulatory and foster neurogenesis, neuronal maturity, and synaptogenesis. Mechanistically, NPC-derived astrocytes perform regenerative matrix remodeling by clearing inhibitory chondroitin sulfate proteoglycans (CSPGs) from the injury milieu through LAR and PTP-σ receptor-mediated endocytosis and the production of ADAMTS1 and ADAMTS9, while most resident astrocytes are pro-inflammatory and contribute to the pathologic deposition of CSPGs. These novel findings unravel critical mechanisms of NPC-mediated astrogenesis in SCI repair.
Collapse
Affiliation(s)
- Seyed Mojtaba Hosseini
- Department of Physiology and Pathophysiology, Spinal Cord Research Centre, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada; Manitoba Multiple Sclerosis Research Center, Winnipeg, MB, Canada
| | - Shiva Nemati
- Department of Physiology and Pathophysiology, Spinal Cord Research Centre, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada; Manitoba Multiple Sclerosis Research Center, Winnipeg, MB, Canada
| | - Soheila Karimi-Abdolrezaee
- Department of Physiology and Pathophysiology, Spinal Cord Research Centre, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada; Manitoba Multiple Sclerosis Research Center, Winnipeg, MB, Canada; Children Hospital Research Institute of Manitoba, Winnipeg, MB, Canada.
| |
Collapse
|
30
|
Yue WWS, Touhara KK, Toma K, Duan X, Julius D. Endogenous opioid signalling regulates spinal ependymal cell proliferation. Nature 2024; 634:407-414. [PMID: 39294372 DOI: 10.1038/s41586-024-07889-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 07/31/2024] [Indexed: 09/20/2024]
Abstract
After injury, mammalian spinal cords develop scars to confine the lesion and prevent further damage. However, excessive scarring can hinder neural regeneration and functional recovery1,2. These competing actions underscore the importance of developing therapeutic strategies to dynamically modulate scar progression. Previous research on scarring has primarily focused on astrocytes, but recent evidence has suggested that ependymal cells also participate. Ependymal cells normally form the epithelial layer encasing the central canal, but they undergo massive proliferation and differentiation into astroglia following certain injuries, becoming a core scar component3-7. However, the mechanisms regulating ependymal proliferation in vivo remain unclear. Here we uncover an endogenous κ-opioid signalling pathway that controls ependymal proliferation. Specifically, we detect expression of the κ-opioid receptor, OPRK1, in a functionally under-characterized cell type known as cerebrospinal fluid-contacting neuron (CSF-cN). We also discover a neighbouring cell population that expresses the cognate ligand prodynorphin (PDYN). Whereas κ-opioids are typically considered inhibitory, they excite CSF-cNs to inhibit ependymal proliferation. Systemic administration of a κ-antagonist enhances ependymal proliferation in uninjured spinal cords in a CSF-cN-dependent manner. Moreover, a κ-agonist impairs ependymal proliferation, scar formation and motor function following injury. Together, our data suggest a paracrine signalling pathway in which PDYN+ cells tonically release κ-opioids to stimulate CSF-cNs and suppress ependymal proliferation, revealing an endogenous mechanism and potential pharmacological strategy for modulating scarring after spinal cord injury.
Collapse
Affiliation(s)
- Wendy W S Yue
- Department of Physiology, University of California, San Francisco, CA, USA.
| | - Kouki K Touhara
- Department of Physiology, University of California, San Francisco, CA, USA
| | - Kenichi Toma
- Department of Ophthalmology, University of California, San Francisco, CA, USA
| | - Xin Duan
- Department of Ophthalmology, University of California, San Francisco, CA, USA
| | - David Julius
- Department of Physiology, University of California, San Francisco, CA, USA.
| |
Collapse
|
31
|
Wen X, Ye Y, Yu Z, Shen H, Cui G, Chen G. The role of nitric oxide and hydrogen sulfide in spinal cord injury: an updated review. Med Gas Res 2024; 14:96-101. [PMID: 39073336 PMCID: PMC466995 DOI: 10.4103/2045-9912.385946] [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: 09/05/2022] [Revised: 12/16/2022] [Accepted: 04/04/2023] [Indexed: 07/30/2024] Open
Abstract
Medical gases play an important role in the pathophysiology of human diseases and have received extensive attention for their role in neuroprotection. Common pathological mechanisms of spinal cord injury include excitotoxicity, inflammation, cell death, glial scarring, blood-spinal cord barrier disruption, and ischemia/reperfusion injury. Nitric oxide and hydrogen sulfide are important gaseous signaling molecules in living organisms; their pathological role in spinal cord injury models has received more attention in recent years. This study reviews the possible mechanisms of spinal cord injury and the role of nitric oxide and hydrogen sulfide in spinal cord injury.
Collapse
Affiliation(s)
- Xiaoliang Wen
- Brain and Nerve Research Laboratory, Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Yang Ye
- Brain and Nerve Research Laboratory, Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Zhengquan Yu
- Brain and Nerve Research Laboratory, Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Haitao Shen
- Brain and Nerve Research Laboratory, Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Gang Cui
- Brain and Nerve Research Laboratory, Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Gang Chen
- Brain and Nerve Research Laboratory, Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| |
Collapse
|
32
|
Liu X, Xu Y, Wang Y, Peng X, Jian J, Wang X, Wang T. RETRACTED ARTICLE: Administration of methylprednisolone do not affect the spinal scar component of spinal cord injury. J Spinal Cord Med 2024:1-10. [PMID: 39167368 DOI: 10.1080/10790268.2024.2352929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/23/2024] Open
Abstract
We, the Editors and Publisher of the journal The Journal of Spinal Cord Medicine, have retracted the following article:Liu, X., Xu, Y., Wang, Y., Peng, X., Jian, J., Wang, X., & Wang, T. (2024). Administration of methylprednisolone do not affect the spinal scar component of spinal cord injury. The Journal of Spinal Cord Medicine, 1-10. https://doi.org/10.1080/10790268.2024.2352929Following publication, in November 2024, the authors notified the journal that they had identified areas in the article which required improvement, and requested retraction.Upon investigation by the journal, significant concerns were identified regarding the animal model and the integrity of Figure(s) 2 and 4.When approached for an explanation following their request to retract the article, the authors did not respond.As verifying the validity of published work is core to the integrity of the scholarly record, we are therefore retracting the article. The corresponding author listed in this publication has been informed. The author(s) agree with the retraction.We have been informed in our decision-making by our editorial policies and the COPE guidelines.The retracted article will remain online to maintain the scholarly record, but it will be digitally watermarked on each page as 'Retracted'.
Collapse
Affiliation(s)
- Xin Liu
- Institute of Base Medicine, Jinzhou Medical University, Jinzhou, People's Republic of China
| | - Yang Xu
- Institute of Neurological Disease, West China Hospital, Sichuan University & The Research Units of West China, Chinese Academy of Medical Sciences, Chengdu, People's Republic of China
| | - Yangyang Wang
- Institute of Neurological Disease, West China Hospital, Sichuan University & The Research Units of West China, Chinese Academy of Medical Sciences, Chengdu, People's Republic of China
| | - Xia Peng
- Institute of Neuroscience, Laboratory Zoology Department, Kunming Medical University, Kunming, People's Republic of China
| | - Jiao Jian
- Institute of Neuroscience, Laboratory Zoology Department, Kunming Medical University, Kunming, People's Republic of China
| | - Xuefang Wang
- Institute of Neurological Disease, West China Hospital, Sichuan University & The Research Units of West China, Chinese Academy of Medical Sciences, Chengdu, People's Republic of China
| | - Tinghua Wang
- Institute of Base Medicine, Jinzhou Medical University, Jinzhou, People's Republic of China
- Institute of Neurological Disease, West China Hospital, Sichuan University & The Research Units of West China, Chinese Academy of Medical Sciences, Chengdu, People's Republic of China
- Institute of Neuroscience, Laboratory Zoology Department, Kunming Medical University, Kunming, People's Republic of China
| |
Collapse
|
33
|
Saraswathy VM, Zhou L, Mokalled MH. Single-cell analysis of innate spinal cord regeneration identifies intersecting modes of neuronal repair. Nat Commun 2024; 15:6808. [PMID: 39147780 PMCID: PMC11327264 DOI: 10.1038/s41467-024-50628-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 07/11/2024] [Indexed: 08/17/2024] Open
Abstract
Adult zebrafish have an innate ability to recover from severe spinal cord injury. Here, we report a comprehensive single nuclear RNA sequencing atlas that spans 6 weeks of regeneration. We identify cooperative roles for adult neurogenesis and neuronal plasticity during spinal cord repair. Neurogenesis of glutamatergic and GABAergic neurons restores the excitatory/inhibitory balance after injury. In addition, a transient population of injury-responsive neurons (iNeurons) show elevated plasticity 1 week post-injury. We found iNeurons are injury-surviving neurons that acquire a neuroblast-like gene expression signature after injury. CRISPR/Cas9 mutagenesis showed iNeurons are required for functional recovery and employ vesicular trafficking as an essential mechanism that underlies neuronal plasticity. This study provides a comprehensive resource of the cells and mechanisms that direct spinal cord regeneration and establishes zebrafish as a model of plasticity-driven neural repair.
Collapse
Affiliation(s)
- Vishnu Muraleedharan Saraswathy
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA
| | - Lili Zhou
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA
| | - Mayssa H Mokalled
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA.
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO, USA.
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
34
|
Xu X, Liu R, Li Y, Zhang C, Guo C, Zhu J, Dong J, Ouyang L, Momeni MR. Spinal Cord Injury: From MicroRNAs to Exosomal MicroRNAs. Mol Neurobiol 2024; 61:5974-5991. [PMID: 38261255 DOI: 10.1007/s12035-024-03954-7] [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: 11/05/2023] [Accepted: 01/12/2024] [Indexed: 01/24/2024]
Abstract
Spinal cord injury (SCI) is an unfortunate experience that may generate extensive sensory and motor disabilities due to the destruction and passing of nerve cells. MicroRNAs are small RNA molecules that do not code for proteins but instead serve to regulate protein synthesis by targeting messenger RNA's expression. After SCI, secondary damage like apoptosis, oxidative stress, inflammation, and autophagy occurs, and differentially expressed microRNAs show a function in these procedures. Almost all animal and plant cells release exosomes, which are sophisticated formations of lipid membranes. These exosomes have the capacity to deliver significant materials, such as proteins, RNAs and lipids, to cells in need, regulating their functions and serving as a way of communication. This new method offers a fresh approach to treating spinal cord injury. Obviously, the exosome has the benefit of conveying the transported material across performing regulatory activities and the blood-brain barrier. Among the exosome cargoes, microRNAs, which modulate their mRNA targets, show considerable promise in the pathogenic diagnosis, process, and therapy of SCI. Herein, we describe the roles of microRNAs in SCI. Furthermore, we emphasize the importance of exosomal microRNAs in this disease.
Collapse
Affiliation(s)
- Xiangyang Xu
- Spinal Surgery, Henan Province Hospital of Traditional Chinese Medicine, The Second Affiliated Hospital of Henan University of Traditional Chinese Medicine), Zhengzhou, Henan, 450003, China
| | - Ruyin Liu
- Spinal Surgery, Henan Province Hospital of Traditional Chinese Medicine, The Second Affiliated Hospital of Henan University of Traditional Chinese Medicine), Zhengzhou, Henan, 450003, China
| | - Yunpeng Li
- Spinal Surgery, Henan Province Hospital of Traditional Chinese Medicine, The Second Affiliated Hospital of Henan University of Traditional Chinese Medicine), Zhengzhou, Henan, 450003, China
| | - Cheng Zhang
- College of Traditional Chinese Medicine Orthopedics and Traumatology, Henan University of Traditional Chinese Medicine, Zhengzhou, Henan, 450003, China
| | - Chuanghao Guo
- College of Traditional Chinese Medicine Orthopedics and Traumatology, Henan University of Traditional Chinese Medicine, Zhengzhou, Henan, 450003, China
| | - Jiong Zhu
- College of Traditional Chinese Medicine Orthopedics and Traumatology, Henan University of Traditional Chinese Medicine, Zhengzhou, Henan, 450003, China
| | - Jiaan Dong
- College of Traditional Chinese Medicine Orthopedics and Traumatology, Henan University of Traditional Chinese Medicine, Zhengzhou, Henan, 450003, China
| | - Liyun Ouyang
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Penang, 11700, Malaysia.
| | | |
Collapse
|
35
|
Jiang Y, Cai Y, Yang N, Gao S, Li Q, Pang Y, Su P. Molecular mechanisms of spinal cord injury repair across vertebrates: A comparative review. Eur J Neurosci 2024; 60:4552-4568. [PMID: 38978308 DOI: 10.1111/ejn.16462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 05/09/2024] [Accepted: 06/20/2024] [Indexed: 07/10/2024]
Abstract
In humans and other adult mammals, axon regeneration is difficult in axotomized neurons. Therefore, spinal cord injury (SCI) is a devastating event that can lead to permanent loss of locomotor and sensory functions. Moreover, the molecular mechanisms of axon regeneration in vertebrates are not very well understood, and currently, no effective treatment is available for SCI. In striking contrast to adult mammals, many nonmammalian vertebrates such as reptiles, amphibians, bony fishes and lampreys can spontaneously resume locomotion even after complete SCI. In recent years, rapid progress in the development of next-generation sequencing technologies has offered valuable information on SCI. In this review, we aimed to provide a comparison of axon regeneration process across classical model organisms, focusing on crucial genes and signalling pathways that play significant roles in the regeneration of individually identifiable descending neurons after SCI. Considering the special evolutionary location and powerful regenerative ability of lamprey and zebrafish, they will be the key model organisms for ongoing studies on spinal cord regeneration. Detailed study of SCI in these model organisms will help in the elucidation of molecular mechanisms of neuron regeneration across species.
Collapse
Affiliation(s)
- Ying Jiang
- College of Life Science, Liaoning Normal University, Dalian, China
- Lamprey Research Center, Liaoning Normal University, Dalian, China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, China
| | - Yang Cai
- College of Life Science, Liaoning Normal University, Dalian, China
- Lamprey Research Center, Liaoning Normal University, Dalian, China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, China
| | - Ning Yang
- College of Life Science, Liaoning Normal University, Dalian, China
- Lamprey Research Center, Liaoning Normal University, Dalian, China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, China
| | - Si Gao
- College of Life Science, Liaoning Normal University, Dalian, China
- Lamprey Research Center, Liaoning Normal University, Dalian, China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, China
| | - Qingwei Li
- College of Life Science, Liaoning Normal University, Dalian, China
- Lamprey Research Center, Liaoning Normal University, Dalian, China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, China
| | - Yue Pang
- College of Life Science, Liaoning Normal University, Dalian, China
- Lamprey Research Center, Liaoning Normal University, Dalian, China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, China
| | - Peng Su
- College of Life Science, Liaoning Normal University, Dalian, China
- Lamprey Research Center, Liaoning Normal University, Dalian, China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, China
| |
Collapse
|
36
|
Azargoonjahromi A. The duality of amyloid-β: its role in normal and Alzheimer's disease states. Mol Brain 2024; 17:44. [PMID: 39020435 PMCID: PMC11256416 DOI: 10.1186/s13041-024-01118-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 07/14/2024] [Indexed: 07/19/2024] Open
Abstract
Alzheimer's disease (AD) is a degenerative neurological condition that gradually impairs cognitive abilities, disrupts memory retention, and impedes daily functioning by impacting the cells of the brain. A key characteristic of AD is the accumulation of amyloid-beta (Aβ) plaques, which play pivotal roles in disease progression. These plaques initiate a cascade of events including neuroinflammation, synaptic dysfunction, tau pathology, oxidative stress, impaired protein clearance, mitochondrial dysfunction, and disrupted calcium homeostasis. Aβ accumulation is also closely associated with other hallmark features of AD, underscoring its significance. Aβ is generated through cleavage of the amyloid precursor protein (APP) and plays a dual role depending on its processing pathway. The non-amyloidogenic pathway reduces Aβ production and has neuroprotective and anti-inflammatory effects, whereas the amyloidogenic pathway leads to the production of Aβ peptides, including Aβ40 and Aβ42, which contribute to neurodegeneration and toxic effects in AD. Understanding the multifaceted role of Aβ, particularly in AD, is crucial for developing effective therapeutic strategies that target Aβ metabolism, aggregation, and clearance with the aim of mitigating the detrimental consequences of the disease. This review aims to explore the mechanisms and functions of Aβ under normal and abnormal conditions, particularly in AD, by examining both its beneficial and detrimental effects.
Collapse
|
37
|
Rybachuk O, Nesterenko Y, Zhovannyk V. Modern advances in spinal cord regeneration: hydrogel combined with neural stem cells. Front Pharmacol 2024; 15:1419797. [PMID: 38994202 PMCID: PMC11236698 DOI: 10.3389/fphar.2024.1419797] [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: 04/18/2024] [Accepted: 06/11/2024] [Indexed: 07/13/2024] Open
Abstract
Severe spinal cord injuries (SCI) lead to loss of functional activity of the body below the injury site, affect a person's ability to self-care and have a direct impact on performance. Due to the structural features and functional role of the spinal cord in the body, the consequences of SCI cannot be completely overcome at the expense of endogenous regenerative potential and, developing over time, lead to severe complications years after injury. Thus, the primary task of this type of injury treatment is to create artificial conditions for the regenerative growth of damaged nerve fibers through the area of the SCI. Solving this problem is possible using tissue neuroengineering involving the technology of replacing the natural tissue environment with synthetic matrices (for example, hydrogels) in combination with stem cells, in particular, neural/progenitor stem cells (NSPCs). This approach can provide maximum stimulation and support for the regenerative growth of axons of damaged neurons and their myelination. In this review, we consider the currently available options for improving the condition after SCI (use of NSC transplantation or/and replacement of the damaged area of the SCI with a matrix, specifically a hydrogel). We emphasise the expediency and effectiveness of the hydrogel matrix + NSCs complex system used for the reconstruction of spinal cord tissue after injury. Since such a complex approach (a combination of tissue engineering and cell therapy), in our opinion, allows not only to creation of conditions for supporting endogenous regeneration or mechanical reconstruction of the spinal cord, but also to strengthen endogenous regeneration, prevent the spread of the inflammatory process, and promote the restoration of lost reflex, motor and sensory functions of the injured area of spinal cord.
Collapse
Affiliation(s)
- Oksana Rybachuk
- Bogomoletz Institute of Physiology NAS of Ukraine, Kyiv, Ukraine
- Institute of Genetic and Regenerative Medicine, M. D. Strazhesko National Scientific Center of Cardiology, Clinical and Regenerative Medicine, National Academy of Medical Sciences of Ukraine, Kyiv, Ukraine
| | | | | |
Collapse
|
38
|
Abbaszadeh F, Javadpour P, Mousavi Nasab MM, Jorjani M. The Role of Vitamins in Spinal Cord Injury: Mechanisms and Benefits. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2024; 2024:4293391. [PMID: 38938696 PMCID: PMC11211004 DOI: 10.1155/2024/4293391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 03/18/2024] [Accepted: 06/06/2024] [Indexed: 06/29/2024]
Abstract
Spinal cord injury (SCI) is a common neurological disease worldwide, often resulting in a substantial decrease in quality of life, disability, and in severe cases, even death. Unfortunately, there is currently no effective treatment for this disease. Nevertheless, current basic and clinical evidence suggests that vitamins, with their antioxidant properties and biological functions, may play a valuable role in improving the quality of life for individuals with SCI. They can promote overall health and facilitate the healing process. In this review, we discuss the mechanisms and therapeutic potential of vitamins in the treatment of SCI.
Collapse
Affiliation(s)
- Fatemeh Abbaszadeh
- Neurobiology Research CenterShahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Pegah Javadpour
- Neuroscience Research CenterShahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Masoumeh Jorjani
- Neurobiology Research CenterShahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of PharmacologySchool of MedicineShahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
39
|
Hosen S, Ikeda-Yorifuji I, Yamashita T. Asporin and CD109, expressed in the injured neonatal spinal cord, attenuate axonal re-growth in vitro. Neurosci Lett 2024; 833:137832. [PMID: 38796094 DOI: 10.1016/j.neulet.2024.137832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 05/17/2024] [Accepted: 05/22/2024] [Indexed: 05/28/2024]
Abstract
Axonal regeneration is restricted in adults and causes irreversible motor dysfunction following spinal cord injury (SCI). In contrast, neonates have prominent regenerative potential and can restore their neural function. Although the distinct cellular responses in neonates have been studied, how they contribute to neural recovery remains unclear. To assess whether the secreted molecules in neonatal SCI can enhance neural regeneration, we re-analyzed the previously performed single-nucleus RNA-seq (snRNA-seq) and focused on Asporin and Cd109, the highly expressed genes in the injured neonatal spinal cord. In the present study, we showed that both these molecules were expressed in the injured spinal cords of adults and neonates. We treated the cortical neurons with recombinant Asporin or CD109 to observe their direct effects on neurons in vitro. We demonstrated that these molecules enhance neurite outgrowth in neurons. However, these molecules did not enhance re-growth of severed axons. Our results suggest that Asporin and CD109 influence neurites at the lesion site, rather than promoting axon regeneration, to restore neural function in neonates after SCI.
Collapse
Affiliation(s)
- Sakura Hosen
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Iyo Ikeda-Yorifuji
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, Osaka, Japan.
| | - Toshihide Yamashita
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, Osaka, Japan; WPI Immunology Frontier Research Center, Osaka University, Suita, Japan; Department of Molecular Neuroscience, Graduate School of Frontier Biosciences, Osaka University, Suita, Japan; Department of Neuro-Medical Science, Graduate School of Medicine, Osaka University, Suita, Japan.
| |
Collapse
|
40
|
Zavvarian MM, Modi AD, Sadat S, Hong J, Fehlings MG. Translational Relevance of Secondary Intracellular Signaling Cascades Following Traumatic Spinal Cord Injury. Int J Mol Sci 2024; 25:5708. [PMID: 38891894 PMCID: PMC11172219 DOI: 10.3390/ijms25115708] [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: 04/09/2024] [Revised: 05/15/2024] [Accepted: 05/21/2024] [Indexed: 06/21/2024] Open
Abstract
Traumatic spinal cord injury (SCI) is a life-threatening and life-altering condition that results in debilitating sensorimotor and autonomic impairments. Despite significant advances in the clinical management of traumatic SCI, many patients continue to suffer due to a lack of effective therapies. The initial mechanical injury to the spinal cord results in a series of secondary molecular processes and intracellular signaling cascades in immune, vascular, glial, and neuronal cell populations, which further damage the injured spinal cord. These intracellular cascades present promising translationally relevant targets for therapeutic intervention due to their high ubiquity and conservation across eukaryotic evolution. To date, many therapeutics have shown either direct or indirect involvement of these pathways in improving recovery after SCI. However, the complex, multifaceted, and heterogeneous nature of traumatic SCI requires better elucidation of the underlying secondary intracellular signaling cascades to minimize off-target effects and maximize effectiveness. Recent advances in transcriptional and molecular neuroscience provide a closer characterization of these pathways in the injured spinal cord. This narrative review article aims to survey the MAPK, PI3K-AKT-mTOR, Rho-ROCK, NF-κB, and JAK-STAT signaling cascades, in addition to providing a comprehensive overview of the involvement and therapeutic potential of these secondary intracellular pathways following traumatic SCI.
Collapse
Affiliation(s)
- Mohammad-Masoud Zavvarian
- Division of Genetics and Development, Toronto Western Hospital, University Health Network, Toronto, ON M5T 2S8, Canada; (M.-M.Z.); (A.D.M.); (S.S.); (J.H.)
- Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Akshat D. Modi
- Division of Genetics and Development, Toronto Western Hospital, University Health Network, Toronto, ON M5T 2S8, Canada; (M.-M.Z.); (A.D.M.); (S.S.); (J.H.)
- Department of Biological Sciences, University of Toronto, Scarborough, ON M1C 1A4, Canada
- Department of Human Biology, University of Toronto, Toronto, ON M5S 3J6, Canada
| | - Sarah Sadat
- Division of Genetics and Development, Toronto Western Hospital, University Health Network, Toronto, ON M5T 2S8, Canada; (M.-M.Z.); (A.D.M.); (S.S.); (J.H.)
- Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - James Hong
- Division of Genetics and Development, Toronto Western Hospital, University Health Network, Toronto, ON M5T 2S8, Canada; (M.-M.Z.); (A.D.M.); (S.S.); (J.H.)
| | - Michael G. Fehlings
- Division of Genetics and Development, Toronto Western Hospital, University Health Network, Toronto, ON M5T 2S8, Canada; (M.-M.Z.); (A.D.M.); (S.S.); (J.H.)
- Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
- Department of Surgery, Faculty of Medicine, University of Toronto, Toronto, ON M5T 1P5, Canada
| |
Collapse
|
41
|
Sun Z, Chen Z, Yin M, Wu X, Guo B, Cheng X, Quan R, Sun Y, Zhang Q, Fan Y, Jin C, Yin Y, Hou X, Liu W, Shu M, Xue X, Shi Y, Chen B, Xiao Z, Dai J, Zhao Y. Harnessing developmental dynamics of spinal cord extracellular matrix improves regenerative potential of spinal cord organoids. Cell Stem Cell 2024; 31:772-787.e11. [PMID: 38565140 DOI: 10.1016/j.stem.2024.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 02/07/2024] [Accepted: 03/11/2024] [Indexed: 04/04/2024]
Abstract
Neonatal spinal cord tissues exhibit remarkable regenerative capabilities as compared to adult spinal cord tissues after injury, but the role of extracellular matrix (ECM) in this process has remained elusive. Here, we found that early developmental spinal cord had higher levels of ECM proteins associated with neural development and axon growth, but fewer inhibitory proteoglycans, compared to those of adult spinal cord. Decellularized spinal cord ECM from neonatal (DNSCM) and adult (DASCM) rabbits preserved these differences. DNSCM promoted proliferation, migration, and neuronal differentiation of neural progenitor cells (NPCs) and facilitated axonal outgrowth and regeneration of spinal cord organoids more effectively than DASCM. Pleiotrophin (PTN) and Tenascin (TNC) in DNSCM were identified as contributors to these abilities. Furthermore, DNSCM demonstrated superior performance as a delivery vehicle for NPCs and organoids in spinal cord injury (SCI) models. This suggests that ECM cues from early development stages might significantly contribute to the prominent regeneration ability in spinal cord.
Collapse
Affiliation(s)
- Zheng Sun
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhenni Chen
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Man Yin
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xianming Wu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Bo Guo
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Xiaokang Cheng
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Rui Quan
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yuting Sun
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qi Zhang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Yongheng Fan
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chen Jin
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yanyun Yin
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Xianglin Hou
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Weiyuan Liu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Muya Shu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Xiaoyu Xue
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Ya Shi
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Bing Chen
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Zhifeng Xiao
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Jianwu Dai
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China.
| | - Yannan Zhao
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China.
| |
Collapse
|
42
|
Liu T, Ma Z, Liu L, Pei Y, Wu Q, Xu S, Liu Y, Ding N, Guan Y, Zhang Y, Chen X. Conditioned medium from human dental pulp stem cells treats spinal cord injury by inhibiting microglial pyroptosis. Neural Regen Res 2024; 19:1105-1111. [PMID: 37862215 PMCID: PMC10749599 DOI: 10.4103/1673-5374.385309] [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: 11/19/2022] [Revised: 05/05/2023] [Accepted: 07/12/2023] [Indexed: 10/22/2023] Open
Abstract
Human dental pulp stem cell transplantation has been shown to be an effective therapeutic strategy for spinal cord injury. However, whether the human dental pulp stem cell secretome can contribute to functional recovery after spinal cord injury remains unclear. In the present study, we established a rat model of spinal cord injury based on impact injury from a dropped weight and then intraperitoneally injected the rats with conditioned medium from human dental pulp stem cells. We found that the conditioned medium effectively promoted the recovery of sensory and motor functions in rats with spinal cord injury, decreased expression of the microglial pyroptosis markers NLRP3, GSDMD, caspase-1, and interleukin-1β, promoted axonal and myelin regeneration, and inhibited the formation of glial scars. In addition, in a lipopolysaccharide-induced BV2 microglia model, conditioned medium from human dental pulp stem cells protected cells from pyroptosis by inhibiting the NLRP3/caspase-1/interleukin-1β pathway. These results indicate that conditioned medium from human dental pulp stem cells can reduce microglial pyroptosis by inhibiting the NLRP3/caspase-1/interleukin-1β pathway, thereby promoting the recovery of neurological function after spinal cord injury. Therefore, conditioned medium from human dental pulp stem cells may become an alternative therapy for spinal cord injury.
Collapse
Affiliation(s)
- Tao Liu
- Department of Orthopedic Surgery, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Ziqian Ma
- Department of Orthopedic Surgery, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Liang Liu
- Department of Orthopedic Surgery, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Yilun Pei
- Department of Orthopedic Surgery, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Qichao Wu
- Department of Orthopedic Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Songjie Xu
- Department of Orthopedic Surgery, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Yadong Liu
- Department of Orthopedic Surgery, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Nan Ding
- Department of Stomatology, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Yun Guan
- Department of Anesthesiology and Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
- Department of Neurological Surgery, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Yan Zhang
- Department of Orthopedic Surgery, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Xueming Chen
- Department of Orthopedic Surgery, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
43
|
Du J, Liu X, Marasini S, Wang Z, Dammen-Brower K, Yarema KJ, Jia X. Metabolically Glycoengineered Neural Stem Cells Boost Neural Repair After Cardiac Arrest. ADVANCED FUNCTIONAL MATERIALS 2024; 34:2309866. [PMID: 39071865 PMCID: PMC11281434 DOI: 10.1002/adfm.202309866] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Indexed: 07/30/2024]
Abstract
Cardiac arrest (CA)-induced cerebral ischemia remains challenging with high mortality and disability. Neural stem cell (NSC) engrafting is an emerging therapeutic strategy with considerable promise that, unfortunately, is severely compromised by limited cell functionality after in vivo transplantation. This groundbreaking report demonstrates that metabolic glycoengineering (MGE) using the "Ac5ManNTProp (TProp)" monosaccharide analog stimulates the Wnt/β-catenin pathway, improves cell adhesion, and enhances neuronal differentiation in human NSCs in vitro thereby substantially increasing the therapeutic potential of these cells. For the first time, MGE significantly enhances NSC efficacy for treating ischemic brain injury after asphyxia CA in rats. In particular, neurological deficit scores and neurobehavioral tests experience greater improvements when the therapeutic cells are pretreated with TProp than with "stand-alone" NSC therapy. Notably, the TProp-NSC group exhibits significantly stronger neuroprotective functions including enhanced differentiation, synaptic plasticity, and reduced microglia recruitment; furthermore, Wnt pathway agonists and inhibitors demonstrate a pivotal role for Wnt signaling in the process. These findings help establish MGE as a promising avenue for addressing current limitations associated with NSC transplantation via beneficially influencing neural regeneration and synaptic plasticity, thereby offering enhanced therapeutic options to boost brain recovery following global ischemia.
Collapse
Affiliation(s)
- Jian Du
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Xiao Liu
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Subash Marasini
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Zhuoran Wang
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Kris Dammen-Brower
- Department of Biomedical Engineering, The Johns Hopkins School of Medicine, Baltimore, MD, 21205
- Translational Cell and Tissue Engineering Center, The Johns Hopkins School of Medicine, Baltimore, MD, 21231
| | - Kevin J. Yarema
- Department of Biomedical Engineering, The Johns Hopkins School of Medicine, Baltimore, MD, 21205
- Translational Cell and Tissue Engineering Center, The Johns Hopkins School of Medicine, Baltimore, MD, 21231
| | - Xiaofeng Jia
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201
- Department of Biomedical Engineering, The Johns Hopkins School of Medicine, Baltimore, MD, 21205
- Department of Orthopedics, University of Maryland School of Medicine, Baltimore, MD 21201
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD 21201
| |
Collapse
|
44
|
Rao Y, Li J, Qiao R, Luo J, Liu Y. Synergistic effects of tetramethylpyrazine and astragaloside IV on spinal cord injury via alteration of astrocyte A1/A2 polarization through the Sirt1-NF-κB pathway. Int Immunopharmacol 2024; 131:111686. [PMID: 38461631 DOI: 10.1016/j.intimp.2024.111686] [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: 11/07/2023] [Revised: 01/12/2024] [Accepted: 02/09/2024] [Indexed: 03/12/2024]
Abstract
OBJECTIVE Reactive astrocytes are hallmarks of traumatic spinal cord injury (T-SCI) and are associated with neuropathic pain (NP). Mediating the functional phenotype of reactive astrocytes helps neural repair and ameliorates NP in T-SCI. Here, we aimed to explore the role of tetramethylpyrazine (TMPZ) and astragaloside IV (AGS-IV) in astrocyte polarization and the underlying molecular mechanism in T-SCI. METHODS Primary cultured astrocytes from mice were treated with LPS or conditioned medium from "M1" polarized microglia (M1-CM), followed by TMPZ and/or AGS-IV administration. The expression levels of "A1" astrocyte-specific markers (including C3, GBP2, Serping1, iNOS), "A2" astrocyte-specific markers (including S100a10 and PTX3), Sirt1 and NF-κB were detected via western blotting. TNF-α and IL-1β levels were detected via ELISA. RT-PCR was used to evaluate OIP5-AS1 and miR-34a expression. si-OIP5-AS1 or the Sirt1 inhibitor EX-527 was administered to astrocytes. A spinal cord injury (SCI) model was constructed in Sprague-Dawley (SD) rats. Alterations in astrocytic "A1/A2" polarization in the spinal cord tissues were evaluated. RESULTS LPS and M1-CM induced "A1" polarization of primary astrocytes. TMPZ and ASG IV could substantially reduce the expression of "A1"-related biomarkers but enhance "A2"-related biomarkers. OIP5-AS1 and Sirt1 levels were reduced in "A1"-polarized astrocytes, while miR-34a and p-NF-κB p65 were elevated. TMPZ and ASG IV enhanced OIP5-AS1 and Sirt1 levels and, in contrast, attenuated the changes in miR-34a and p-NF-κB p65 levels. Notably, the TMPZ and ASG IV combination had stronger effects on astrocyte polarization than the single treatment with TMPZ or ASG IV. OIP5-AS1 knockdown and Sirt1 inhibition both reversed the regulatory effects of TMPZ and ASG IV in astrocytic polarization. According to the in vivo experiments, the expression of "A1"-associated markers was enhanced in the spinal cords of SCI rats. The TMPZ and ASG IV combination reduced astrocytic "A1" polarization and enhanced astrocytic "A2" polarization. The expression of lncRNA OIP5-AS1 and Sirt1 was enhanced by TMPZ and ASG IV, while that of miR-34a and p-NF-κB p65 was inhibited. CONCLUSION The combination of TMPZ and ASG IV can ameliorate dysregulated astrocytic polarization induced by spinal cord injury by affecting the lncRNA OIP5-AS1-Sirt1-NF-κB pathway.
Collapse
Affiliation(s)
- Yaojian Rao
- Department of Spine Surgery, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang 471003, China.
| | - Junjie Li
- Department of Spine Surgery, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang 471003, China
| | - Ruofei Qiao
- Department of Spine Surgery, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang 471003, China
| | - Jinxin Luo
- Department of Spine Surgery, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang 471003, China
| | - Yan Liu
- Department of Spine Surgery, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang 471003, China
| |
Collapse
|
45
|
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
|
46
|
Pushchina EV, Bykova ME, Varaksin AA. Post-Traumatic Expressions of Aromatase B, Glutamine Synthetase, and Cystathionine-Beta-Synthase in the Cerebellum of Juvenile Chum Salmon, Oncorhynchus keta. Int J Mol Sci 2024; 25:3299. [PMID: 38542274 PMCID: PMC10970380 DOI: 10.3390/ijms25063299] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 02/11/2024] [Accepted: 03/11/2024] [Indexed: 04/29/2025] Open
Abstract
In adult fish, neurogenesis occurs in many areas of the brain, including the cerebellum, with the ratio of newly formed cells relative to the total number of brain cells being several orders of magnitude greater than in mammals. Our study aimed to compare the expressions of aromatase B (AroB), glutamine synthetase (GS), and cystathionine-beta-synthase (CBS) in the cerebellum of intact juvenile chum salmon, Oncorhynchus keta. To identify the dynamics that determine the involvement of AroB, GS, and CBS in the cellular mechanisms of regeneration, we performed a comprehensive assessment of the expressions of these molecular markers during a long-term primary traumatic brain injury (TBI) and after a repeated acute TBI to the cerebellum of O. keta juveniles. As a result, in intact juveniles, weak or moderate expressions of AroB, GS, and CBS were detected in four cell types, including cells of the neuroepithelial type, migrating, and differentiated cells (graphic abstract, A). At 90 days post injury, local hypercellular areas were found in the molecular layer containing moderately labeled AroB+, GS+, and CBS+ cells of the neuroepithelial type and larger AroB+, GS+, and CBS+ cells (possibly analogous to the reactive glia of mammals); patterns of cells migration and neovascularization were also observed. A repeated TBI caused the number of AroB+, GS+, and CBS+ cells to further increase; an increased intensity of immunolabeling was recorded from all cell types (graphic abstract, C). Thus, the results of this study provide a better understanding of adult neurogenesis in teleost fishes, which is expected to clarify the issue of the reactivation of adult neurogenesis in mammalian species.
Collapse
Affiliation(s)
- Evgeniya V. Pushchina
- Zhirmunsky National Scientific Center of Marine Biology, Far Eastern Branch, Russian Academy of Sciences, 690041 Vladivostok, Russia; (M.E.B.); (A.A.V.)
| | | | | |
Collapse
|
47
|
Wu T, Li Y, Wu Z, Wang Z, Li Y, Jian K, He C, Zhang C, Shi L, Dai J. Enzyme-immobilized nanoclay hydrogel simultaneously reduces inflammation and scar deposition to treat spinal cord injury. CHEMICAL ENGINEERING JOURNAL 2024; 484:149642. [DOI: 10.1016/j.cej.2024.149642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
48
|
Liu J, Qi L, Bao S, Yan F, Chen J, Yu S, Dong C. The acute spinal cord injury microenvironment and its impact on the homing of mesenchymal stem cells. Exp Neurol 2024; 373:114682. [PMID: 38199509 DOI: 10.1016/j.expneurol.2024.114682] [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: 11/07/2023] [Revised: 12/08/2023] [Accepted: 01/02/2024] [Indexed: 01/12/2024]
Abstract
Spinal cord injury (SCI) is a highly debilitating condition that inflicts devastating harm on the lives of affected individuals, underscoring the urgent need for effective treatments. By activating inflammatory cells and releasing inflammatory factors, the secondary injury response creates an inflammatory microenvironment that ultimately determines whether neurons will undergo necrosis or regeneration. In recent years, mesenchymal stem cells (MSCs) have garnered increasing attention for their therapeutic potential in SCI. MSCs not only possess multipotent differentiation capabilities but also have homing abilities, making them valuable as carriers and mediators of therapeutic agents. The inflammatory microenvironment induced by SCI recruits MSCs to the site of injury through the release of various cytokines, chemokines, adhesion molecules, and enzymes. However, this mechanism has not been previously reported. Thus, a comprehensive exploration of the molecular mechanisms and cellular behaviors underlying the interplay between the inflammatory microenvironment and MSC homing is crucial. Such insights have the potential to provide a better understanding of how to harness the therapeutic potential of MSCs in treating inflammatory diseases and facilitating injury repair. This review aims to delve into the formation of the inflammatory microenvironment and how it influences the homing of MSCs.
Collapse
Affiliation(s)
- Jinyi Liu
- Department of Anatomy, Medical College of Nantong University, Nantong, China
| | - Longju Qi
- Affiliated Nantong Hospital 3 of Nantong University, Nantong, China
| | - Shengzhe Bao
- Department of Anatomy, Medical College of Nantong University, Nantong, China
| | - Fangsu Yan
- Department of Anatomy, Medical College of Nantong University, Nantong, China
| | - Jiaxi Chen
- Department of Anatomy, Medical College of Nantong University, Nantong, China
| | - Shumin Yu
- Department of Anatomy, Medical College of Nantong University, Nantong, China
| | - Chuanming Dong
- Department of Anatomy, Medical College of Nantong University, Nantong, China; Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China.
| |
Collapse
|
49
|
Xu ZX, Xu D, Fang F, Fan YJ, Wu B, Chen YF, Huang HE, Huang XH, Zhuang YH, Xu WH. Enhanced axon outgrowth of spinal motor neurons in co-culturing with dorsal root ganglions antagonizes the growth inhibitory environment. Regen Ther 2024; 25:68-76. [PMID: 38148872 PMCID: PMC10750115 DOI: 10.1016/j.reth.2023.11.013] [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: 09/23/2023] [Revised: 11/09/2023] [Accepted: 11/22/2023] [Indexed: 12/28/2023] Open
Abstract
Introduction Forming a bridge made of functional axons to span the lesion is essential to reconstruct the motor circuitry following spinal cord injury (SCI). Dorsal root ganglion (DRG) axons are robust in axon growth and have been proved to facilitate the growth of cortical neurons in a process of axon-facilitated axon regeneration. However, whether DRG transplantation affects the axon outgrowth of spinal motor neurons (SMNs) that play crucial roles in motor circuitry remains unclear. Methods We investigated the axonal growth patterns of co-cultured DRGs and SMN aggregates (SMNAs) taking advantage of a well-designed 3D-printed in vitro system. Chondroitin sulphate proteoglycans (CSPG) induced inhibitory matrix was introduced to imitate the inhibitory environment following SCI. Axonal lengths of DRG, SMNA or DRG & SMNA cultured on the permissive or CSPG induced inhibitory matrix were measured and compared. Results Our results indicated that under the guidance of full axonal connection generated from two opposing populations of DRGs, SMNA axons were growth-enhanced and elongated along the DRG axon bridge to distances that they could not otherwise reach. Quantitatively, the co-culture increased the SMNA axonal length by 32.1 %. Moreover, the CSPG matrix reduced the axonal length of DRGs and SMNAs by 46.2 % and 17.7 %, respectively. This inhibitory effect was antagonized by the co-culture of DRGs and SMNAs. Especially for SMNAs, they extended the axons across the CSPG-coating matrix, reached the lengths close to those of SMNAs cultured on the permissive matrix alone. Conclusions This study deepens our understanding of axon-facilitated reconstruction of the motor circuitry. Moreover, the results support SCI treatment utilizing the enhanced outgrowth of axons to restore functional connectivity in SCI patients.
Collapse
Affiliation(s)
- Zi-Xing Xu
- Department of Spinal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian Province, China
- Department of Orthopedics, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian Province, China
- Fujian Provincial Institute of Orthopedics, Fuzhou, Fujian Province, China
| | - Dan Xu
- Fujian Key Laboratory of Brain Aging and Neurodegenerative Diseases, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Fang Fang
- Department of Pharmacology, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Ying-Juan Fan
- Department of Pharmacology, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Bing Wu
- The Central Laboratory, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian Province, China
| | - Yu-Fan Chen
- Department of Spinal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian Province, China
| | - Hao-En Huang
- Department of Spinal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian Province, China
| | - Xin-Hao Huang
- Department of Spinal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian Province, China
| | - Yue-Hong Zhuang
- Fujian Key Laboratory of Brain Aging and Neurodegenerative Diseases, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Wei-Hong Xu
- Department of Spinal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian Province, China
- Department of Orthopedics, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian Province, China
- Fujian Provincial Institute of Orthopedics, Fuzhou, Fujian Province, China
| |
Collapse
|
50
|
Bolini L, Campos RMP, Spiess DA, Lima-Rosa FL, Dantas DP, Conde L, Mendez-Otero R, Vale AM, Pimentel-Coelho PM. Long-term recruitment of peripheral immune cells to brain scars after a neonatal insult. Glia 2024; 72:546-567. [PMID: 37987116 DOI: 10.1002/glia.24490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 10/23/2023] [Accepted: 10/31/2023] [Indexed: 11/22/2023]
Abstract
Although brain scars in adults have been extensively studied, there is less data available regarding scar formation during the neonatal period, and the involvement of peripheral immune cells in this process remains unexplored in neonates. Using a murine model of neonatal hypoxic-ischemic encephalopathy (HIE) and confocal microscopy, we characterized the scarring process and examined the recruitment of peripheral immune cells to cortical and hippocampal scars for up to 1 year post-insult. Regional differences in scar formation were observed, including the presence of reticular fibrotic networks in the cortex and perivascular fibrosis in the hippocampus. We identified chemokines with chronically elevated levels in both regions and demonstrated, through a parabiosis-based strategy, the recruitment of lymphocytes, neutrophils, and monocyte-derived macrophages to the scars several weeks after the neonatal insult. After 1 year, however, neutrophils and lymphocytes were absent from the scars. Our data indicate that peripheral immune cells are transient components of HIE-induced brain scars, opening up new possibilities for late therapeutic interventions.
Collapse
Affiliation(s)
- Lukas Bolini
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Daiane Aparecida Spiess
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Frederico Luis Lima-Rosa
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Danillo Pereira Dantas
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Luciana Conde
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Rosalia Mendez-Otero
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Andre M Vale
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | |
Collapse
|