1
|
Xu G, Ge R, Zhang C, Zhao Z, Han L, Zhang W, Yue W, Zhang J, Zhao Y, Hou S, Li L, Wang P. Promotion of nerve regeneration and motor function recovery in SCI rats using LOCAS-iPSCs-NSCs. Stem Cell Res Ther 2024; 15:376. [PMID: 39444002 PMCID: PMC11515548 DOI: 10.1186/s13287-024-03999-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 10/14/2024] [Indexed: 10/25/2024] Open
Abstract
BACKGROUND Spinal cord injury (SCI) is a severe traumatic spinal condition with a poor prognosis. In this study, a scaffold called linearly ordered collagen aggregates (LOCAS) was created and loaded with induced pluripotent stem cells (iPSCs)-derived neural stem cells (NSCs) from human umbilical cord blood derived mesenchymal stem cells (hUCB-MSCs) to treat SCI in a rat model. METHODS The rats underwent a complete transection SCI resulting in a 3-mm break at either the T9 or T10 level of the spinal cord. RESULTS Scanning electron microscope analysis revealed a uniform pore structure on the coronal plane of the scaffold. The LOCAS had a porosity of 88.52% and a water absorption of 1161.67%. Its compressive modulus and stress were measured at 4.1 MPa and 205 kPa, respectively, with a degradation time of 10 weeks. After 12 weeks, rats in the LOCAS-iPSCs-NSCs group exhibited significantly higher BBB scores (8.6) compared to the LOCAS-iPSCs-NSCs group (5.6) and the Model group (4.2). The CatWalk analysis showed improved motion trajectory, regularity index (RI), and swing speed in the LOCAS-iPSCs-NSCs group compared to the other groups. Motor evoked potentials latency was lower and amplitude was higher in the LOCAS-iPSCs-NSCs group, indicating better neural function recovery. Histological analysis demonstrated enhanced neuronal differentiation of NSCs and nerve fiber regeneration promoted by LOCAS-iPSCs-NSCs, leading to improved motor function recovery in rats. The LOCAS scaffold facilitated ordered neurofilament extension and guided nerve regeneration. CONCLUSIONS The combination of LOCAS and iPSCs-NSCs demonstrated a positive therapeutic impact on motor function recovery and tissue repair in rats with SCI. This development offers a more resilient bionic microenvironment and presents novel possibilities for clinical SCI repair.
Collapse
Affiliation(s)
- Gang Xu
- Department of Orthopaedics, First Affiliated Hospital of Dalian Medical University, Dalian, 116011, Liaoning Province, China.
- Key Laboratory of Molecular Mechanism for Repair and Remodeling of Orthopaedic Diseases, Liaoning Province, Dalian, 116011, Liaoning Province, China.
| | - Rui Ge
- Department of Orthopaedics, First Affiliated Hospital of Dalian Medical University, Dalian, 116011, Liaoning Province, China
- Key Laboratory of Molecular Mechanism for Repair and Remodeling of Orthopaedic Diseases, Liaoning Province, Dalian, 116011, Liaoning Province, China
| | - Chunli Zhang
- Senior Department of Orthopedics, The Fourth Medical Center of PLA General Hospital, Beijing, 100048, China
- Beijing Engineering Research Center of Orthopedics Implants, Beijing, 100048, China
| | - Ziteng Zhao
- Senior Department of Orthopedics, The Fourth Medical Center of PLA General Hospital, Beijing, 100048, China
- Beijing Engineering Research Center of Orthopedics Implants, Beijing, 100048, China
| | - Liwei Han
- Senior Department of Orthopedics, The Fourth Medical Center of PLA General Hospital, Beijing, 100048, China
- Beijing Engineering Research Center of Orthopedics Implants, Beijing, 100048, China
| | - Wanhao Zhang
- Department of Orthopaedics, First Affiliated Hospital of Dalian Medical University, Dalian, 116011, Liaoning Province, China
| | - WenJie Yue
- Department of Orthopaedics, First Affiliated Hospital of Dalian Medical University, Dalian, 116011, Liaoning Province, China
| | - Jing Zhang
- Department of Orthopaedics, First Affiliated Hospital of Dalian Medical University, Dalian, 116011, Liaoning Province, China
| | - Yantao Zhao
- Senior Department of Orthopedics, The Fourth Medical Center of PLA General Hospital, Beijing, 100048, China
- Beijing Engineering Research Center of Orthopedics Implants, Beijing, 100048, China
| | - Shuxun Hou
- Senior Department of Orthopedics, The Fourth Medical Center of PLA General Hospital, Beijing, 100048, China
- Beijing Engineering Research Center of Orthopedics Implants, Beijing, 100048, China
| | - Li Li
- Senior Department of Orthopedics, The Fourth Medical Center of PLA General Hospital, Beijing, 100048, China.
- Beijing Engineering Research Center of Orthopedics Implants, Beijing, 100048, China.
| | - Peng Wang
- Department of Neurosurgery, The First Medical Center of PLA General Hospital, Beijing, 100853, China.
| |
Collapse
|
2
|
Cao Y, Wang Y, Xia D, Fan Z. KDM2B and its peptides promote the stem cells from apical papilla mediated nerve injury repair in rats by intervening EZH2 function. Cell Prolif 2024:e13756. [PMID: 39358887 DOI: 10.1111/cpr.13756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 09/09/2024] [Accepted: 09/14/2024] [Indexed: 10/04/2024] Open
Abstract
How to improve the neurogenic potential of mesenchymal stem cells (MSCs) and develop biological agent based on the underlying epigenetic mechanism remains a challenge. Here, we investigated the effect of histone demethylase Lysine (K)-specific demethylase 2B (KDM2B) on neurogenic differentiation and nerve injury repair by using MSCs from dental apical papilla (SCAP). We found that KDM2B promoted the neurogenic indicators expression and neural spheres formation in SCAP, and modified the Histone H3K4 trimethylation (H3K4me3) methylation on neurogenesis-related genes. KDM2B improved the SCAP mediated recovery of motor ability at the early healing stage of spinal cord injury rats. Meanwhile, KDM2B acted as a negative regulator to its partner EZH2 during neurogenic differentiation, enhancer of zeste homologue 2 (EZH2) suppressed the neurogenic ability of SCAP. Further, the protein interaction between KDM2B and EZH2 was identified which decreased during neurogenic differentiation. On this basis, we revealed seven key protein binding sequences of KDM2B to EZH2, and synthesized KDM2B-peptides based on these sequences. By the usage of KDM2B-peptides, EZH2 function was effectively intervened and the neurogenic ability of SCAP was promoted. More, KDM2B-peptides significantly improved the SCAP mediated functional recovery at SCI early phase. Our study revealed that KDM2B acted as a promotor to neurogenic differentiation ability of dental MSCs through binding and negatively regulating EZH2, and provided the KDM2B-peptides as candidate agents for improving the neurogenic ability of MSCs and nerve injury repair.
Collapse
Affiliation(s)
- Yangyang Cao
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, China
| | - Yantong Wang
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, China
| | - Dengsheng Xia
- Department of General Dentistry and Integrated Emergency Dental Care, Capital Medical University School of Stomatology, Beijing, China
| | - Zhipeng Fan
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, China
- Beijing Laboratory of Oral Health, Capital Medical University School, Beijing, China
- Research Unit of Tooth Development and Regeneration, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
3
|
Xie Y, Ma C, Zhu Q, Fu T, Bai L, Lan X, Liu L, Xiao J. Facial nerve regeneration via body-brain crosstalk: The role of stem cells and biomaterials. Neurobiol Dis 2024; 200:106650. [PMID: 39197536 DOI: 10.1016/j.nbd.2024.106650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 08/24/2024] [Accepted: 08/25/2024] [Indexed: 09/01/2024] Open
Abstract
The human body is a complex, integral whole, and disruptions in one organ can lead to dysfunctions in other parts of the organ network. The facial nerve, as the seventh cranial nerve, arises from the brainstem, controls facial expression muscles and plays a crucial role in brain-body communication. This vulnerable nerve can be damaged by trauma, inflammation, tumors, and congenital diseases, often impairing facial expression. Stem cells have gained significant attention for repairing peripheral nerve injuries due to their multidirectional differentiation potential. Additionally, various biomaterials have been used in tissue engineering for regeneration and repair. However, the therapeutic potential of stem cells and biomaterials in treating facial nerve injuries requires further exploration. In this review, we summarize the roles of stem cells and biomaterials in the regeneration and repair of damaged facial nerves, providing a theoretical basis for the recovery and reconstruction of body-brain crosstalk between the brain and facial expression muscles.
Collapse
Affiliation(s)
- Yuping Xie
- Department of Oral Implantology, The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou 646000, China; Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou 646000, China
| | - Chuan Ma
- Department of Oral Implantology, The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou 646000, China
| | - Qiang Zhu
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou 646000, China; Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou 646000, China
| | - Ting Fu
- Department of Oral Implantology, The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou 646000, China
| | - Long Bai
- Department of Oral Implantology, The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou 646000, China; Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou 646000, China
| | - Xiaorong Lan
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou 646000, China
| | - Lin Liu
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou 646000, China; Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou 646000, China.
| | - Jingang Xiao
- Department of Oral Implantology, The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou 646000, China; Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou 646000, China; Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou 646000, China.
| |
Collapse
|
4
|
Ma D, Fu C, Li F, Ruan R, Lin Y, Li X, Li M, Zhang J. Functional biomaterials for modulating the dysfunctional pathological microenvironment of spinal cord injury. Bioact Mater 2024; 39:521-543. [PMID: 38883317 PMCID: PMC11179178 DOI: 10.1016/j.bioactmat.2024.04.015] [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: 01/13/2024] [Revised: 04/13/2024] [Accepted: 04/14/2024] [Indexed: 06/18/2024] Open
Abstract
Spinal cord injury (SCI) often results in irreversible loss of sensory and motor functions, and most SCIs are incurable with current medical practice. One of the hardest challenges in treating SCI is the development of a dysfunctional pathological microenvironment, which mainly comprises excessive inflammation, deposition of inhibitory molecules, neurotrophic factor deprivation, glial scar formation, and imbalance of vascular function. To overcome this challenge, implantation of functional biomaterials at the injury site has been regarded as a potential treatment for modulating the dysfunctional microenvironment to support axon regeneration, remyelination at injury site, and functional recovery after SCI. This review summarizes characteristics of dysfunctional pathological microenvironment and recent advances in biomaterials as well as the technologies used to modulate inflammatory microenvironment, regulate inhibitory microenvironment, and reshape revascularization microenvironment. Moreover, technological limitations, challenges, and future prospects of functional biomaterials to promote efficient repair of SCI are also discussed. This review will aid further understanding and development of functional biomaterials to regulate pathological SCI microenvironment.
Collapse
Affiliation(s)
- Dezun Ma
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, 1 Qiuyang Road, Fuzhou, Fujian, 350122, PR China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, PR China
| | - Changlong Fu
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, 1 Qiuyang Road, Fuzhou, Fujian, 350122, PR China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, PR China
| | - Fenglu Li
- College of Chemical Engineering, Fuzhou University, 2 Xueyuan Road, Fuzhou, 350108, PR China
- Qingyuan Innovation Laboratory, 1 Xueyuan Road, Quanzhou, 362801, PR China
| | - Renjie Ruan
- College of Chemical Engineering, Fuzhou University, 2 Xueyuan Road, Fuzhou, 350108, PR China
- Qingyuan Innovation Laboratory, 1 Xueyuan Road, Quanzhou, 362801, PR China
| | - Yanming Lin
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, 1 Qiuyang Road, Fuzhou, Fujian, 350122, PR China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, PR China
| | - Xihai Li
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, 1 Qiuyang Road, Fuzhou, Fujian, 350122, PR China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, PR China
| | - Min Li
- Fujian Children's Hospital, Fujian Branch of Shanghai Children's Medical Center, 966 Hengyu Road, Fuzhou, 350014, PR China
- Fujian Maternity and Child Health Hospital, 111 Daoshan Road, Fuzhou, 350005, PR China
- College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, 111 Daoshan Road, Fuzhou, 350005, PR China
| | - Jin Zhang
- College of Chemical Engineering, Fuzhou University, 2 Xueyuan Road, Fuzhou, 350108, PR China
- Qingyuan Innovation Laboratory, 1 Xueyuan Road, Quanzhou, 362801, PR China
| |
Collapse
|
5
|
Xiao L, Duan T, Xia Y, Chen Y, Sun Y, Xu Y, Xu L, Yan X, Hu J. [Linarin inhibits microglia activation-mediated neuroinflammation and neuronal apoptosis in mouse spinal cord injury by inhibiting the TLR4/NF-κB pathway]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2024; 44:1589-1598. [PMID: 39276055 PMCID: PMC11378057 DOI: 10.12122/j.issn.1673-4254.2024.08.18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 09/16/2024]
Abstract
OBJECTIVE To investigate the mechanism underlying the neuroprotective effect of linarin (LIN) against microglia activation-mediated inflammation and neuronal apoptosis following spinal cord injury (SCI). METHODS Fifty C57BL/6J mice (8- 10 weeks old) were randomized to receive sham operation, SCI and linarin treatment at 12.5, 25, and 50 mg/kg following SCI (n=10). Locomotor function recovery of the SCI mice was assessed using the Basso Mouse Scale, inclined plane test, and footprint analysis, and spinal cord tissue damage and myelination were evaluated using HE and LFB staining. Nissl staining, immunofluorescence assay and Western blotting were used to observe surviving anterior horn motor neurons in injured spinal cord tissue. In cultured BV2 cells, the effects of linarin against lipopolysaccharide (LPS)‑induced microglia activation, inflammatory factor release and signaling pathway changes were assessed with immunofluorescence staining, Western blotting, RT-qPCR, and ELISA. In a BV2 and HT22 cell co-culture system, Western blotting was performed to examine the effect of linarin against HT22 cell apoptosis mediated by LPS-induced microglia activation. RESULTS Linarin treatment significantly improved locomotor function (P < 0.05), reduced spinal cord damage area, increased spinal cord myelination, and increased the number of motor neurons in the anterior horn of the SCI mice (P < 0.05). In both SCI mice and cultured BV2 cells, linarin effectively inhibited glial cell activation and suppressed the release of iNOS, COX-2, TNF-α, IL-6, and IL-1β, resulting also in reduced neuronal apoptosis in SCI mice (P < 0.05). Western blotting suggested that linarin-induced microglial activation inhibition was mediated by inhibition of the TLR4/NF- κB signaling pathway. In the cell co-culture experiments, linarin treatment significantly decreased inflammation-mediated apoptosis of HT22 cells (P < 0.05). CONCLUSION The neuroprotective effect of linarin is medicated by inhibition of microglia activation via suppressing the TLR4/NF‑κB signaling pathway, which mitigates neural inflammation and reduce neuronal apoptosis to enhance motor function of the SCI mice.
Collapse
Affiliation(s)
- L Xiao
- Department of Rehabilitation Medicine, First Affiliated Hospital of Bengbu Medical University, Bengbu 233004, China
| | - T Duan
- Department of Emergency Medicine, First Affiliated Hospital of Bengbu Medical University, Bengbu 233004, China
| | - Y Xia
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical University, Bengbu 233004, China
| | - Y Chen
- Department of Rehabilitation Medicine, First Affiliated Hospital of Bengbu Medical University, Bengbu 233004, China
| | - Y Sun
- Department of Rehabilitation Medicine, First Affiliated Hospital of Bengbu Medical University, Bengbu 233004, China
| | - Y Xu
- Department of Histology and Embryology, College of Basic Medical Sciences, Bengbu Medical University, Bengbu 233030, China
| | - L Xu
- Department of Rehabilitation Medicine, First Affiliated Hospital of Bengbu Medical University, Bengbu 233004, China
| | - X Yan
- Department of Rehabilitation Medicine, First Affiliated Hospital of Bengbu Medical University, Bengbu 233004, China
| | - J Hu
- Anhui Provincial Key Laboratory of Basic and Translational Research of Inflammation-related Diseases
- Clinical Laboratory, First Affiliated Hospital of Bengbu Medical University, Bengbu 233004, China
| |
Collapse
|
6
|
Ye Z, Zheng Y, Li N, Zhang H, Li Q, Wang X. Repair of spinal cord injury by bone marrow mesenchymal stem cell-derived exosomes: a systematic review and meta-analysis based on rat models. Front Mol Neurosci 2024; 17:1448777. [PMID: 39169950 PMCID: PMC11335736 DOI: 10.3389/fnmol.2024.1448777] [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: 06/15/2024] [Accepted: 07/25/2024] [Indexed: 08/23/2024] Open
Abstract
Objective This study aims to systematically evaluate the efficacy of bone marrow mesenchymal stem cell-derived exosomes (BMSCs-Exo) in improving spinal cord injury (SCI) to mitigate the risk of translational discrepancies from animal experiments to clinical applications. Methods We conducted a comprehensive literature search up to March 2024 using PubMed, Embase, Web of Science, and Scopus databases. Two researchers independently screened the literature, extracted data, and assessed the quality of the studies. Data analysis was performed using STATA16 software. Results A total of 30 studies were included. The results indicated that BMSCs-Exo significantly improved the BBB score in SCI rats (WMD = 3.47, 95% CI [3.31, 3.63]), inhibited the expression of the pro-inflammatory cytokine TNF-α (SMD = -3.12, 95% CI [-3.57, -2.67]), and promoted the expression of anti-inflammatory cytokines IL-10 (SMD = 2.76, 95% CI [1.88, 3.63]) and TGF-β (SMD = 3.89, 95% CI [3.02, 4.76]). Additionally, BMSCs-Exo significantly reduced apoptosis levels (SMD = -4.52, 95% CI [-5.14, -3.89]), promoted the expression of axonal regeneration markers NeuN cells/field (SMD = 3.54, 95% CI [2.65, 4.42]), NF200 (SMD = 4.88, 95% CI [3.70, 6.05]), and the number of Nissl bodies (SMD = 1.89, 95% CI [1.13, 2.65]), and decreased the expression of astrogliosis marker GFAP (SMD = -5.15, 95% CI [-6.47, -3.82]). The heterogeneity among studies was primarily due to variations in BMSCs-Exo transplantation doses, with efficacy increasing with higher doses. Conclusion BMSCs-Exo significantly improved motor function in SCI rats by modulating inflammatory responses, reducing apoptosis, inhibiting astrogliosis, and promoting axonal regeneration. However, the presence of selection, performance, and detection biases in current animal experiments may undermine the quality of evidence in this study.
Collapse
Affiliation(s)
- Zhongduo Ye
- The First Hospital of Lanzhou University, Lanzhou, China
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Yukun Zheng
- The First Hospital of Lanzhou University, Lanzhou, China
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Ningning Li
- Lanzhou Maternal and Child Health Hospital, Lanzhou, China
| | - Huaibin Zhang
- The First Hospital of Lanzhou University, Lanzhou, China
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Qiangqiang Li
- The First Hospital of Lanzhou University, Lanzhou, China
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Xiong Wang
- The First Hospital of Lanzhou University, Lanzhou, China
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
| |
Collapse
|
7
|
Ju D, Dong C. The combined application of stem cells and three-dimensional bioprinting scaffolds for the repair of spinal cord injury. Neural Regen Res 2024; 19:1751-1758. [PMID: 38103241 PMCID: PMC10960285 DOI: 10.4103/1673-5374.385842] [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: 02/13/2023] [Revised: 06/07/2023] [Accepted: 08/04/2023] [Indexed: 12/18/2023] Open
Abstract
Spinal cord injury is considered one of the most difficult injuries to repair and has one of the worst prognoses for injuries to the nervous system. Following surgery, the poor regenerative capacity of nerve cells and the generation of new scars can make it very difficult for the impaired nervous system to restore its neural functionality. Traditional treatments can only alleviate secondary injuries but cannot fundamentally repair the spinal cord. Consequently, there is a critical need to develop new treatments to promote functional repair after spinal cord injury. Over recent years, there have been several developments in the use of stem cell therapy for the treatment of spinal cord injury. Alongside significant developments in the field of tissue engineering, three-dimensional bioprinting technology has become a hot research topic due to its ability to accurately print complex structures. This led to the loading of three-dimensional bioprinting scaffolds which provided precise cell localization. These three-dimensional bioprinting scaffolds could repair damaged neural circuits and had the potential to repair the damaged spinal cord. In this review, we discuss the mechanisms underlying simple stem cell therapy, the application of different types of stem cells for the treatment of spinal cord injury, and the different manufacturing methods for three-dimensional bioprinting scaffolds. In particular, we focus on the development of three-dimensional bioprinting scaffolds for the treatment of spinal cord injury.
Collapse
Affiliation(s)
- Dingyue Ju
- Department of Anatomy, Medical College of Nantong University, Nantong, Jiangsu Province, China
| | - Chuanming Dong
- Department of Anatomy, Medical College of Nantong University, Nantong, Jiangsu Province, China
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| |
Collapse
|
8
|
Lin K, Zhang Y, Shen Y, Xu Y, Huang M, Liu X. Hydrogen Sulfide can Scavenge Free Radicals to Improve Spinal Cord Injury by Inhibiting the p38MAPK/mTOR/NF-κB Signaling Pathway. Neuromolecular Med 2024; 26:26. [PMID: 38907170 DOI: 10.1007/s12017-024-08794-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Accepted: 05/31/2024] [Indexed: 06/23/2024]
Abstract
Spinal cord injury (SCI) causes irreversible cell loss and neurological dysfunctions. Presently, there is no an effective clinical treatment for SCI. It can be the only intervention measure by relieving the symptoms of patients such as pain and fever. Free radical-induced damage is one of the validated mechanisms in the complex secondary injury following primary SCI. Hydrogen sulfide (H2S) as an antioxidant can effectively scavenge free radicals, protect neurons, and improve SCI by inhibiting the p38MAPK/mTOR/NF-κB signaling pathway. In this report, we analyze the pathological mechanism of SCI, the role of free radical-mediated the p38MAPK/mTOR/NF-κB signaling pathway in SCI, and the role of H2S in scavenging free radicals and improving SCI.
Collapse
Affiliation(s)
- Kexin Lin
- Department of Histology and Embryology, School of Medicine, Shaoxing University, Shaoxing, 312000, Zhejiang Province, China
| | - Yong Zhang
- Department of Histology and Embryology, School of Medicine, Shaoxing University, Shaoxing, 312000, Zhejiang Province, China
| | - Yanyang Shen
- Department of Histology and Embryology, School of Medicine, Shaoxing University, Shaoxing, 312000, Zhejiang Province, China
| | - Yiqin Xu
- Department of Histology and Embryology, School of Medicine, Shaoxing University, Shaoxing, 312000, Zhejiang Province, China
| | - Min Huang
- Department of Histology and Embryology, School of Medicine, Shaoxing University, Shaoxing, 312000, Zhejiang Province, China
| | - Xuehong Liu
- Department of Histology and Embryology, School of Medicine, Shaoxing University, Shaoxing, 312000, Zhejiang Province, China.
| |
Collapse
|
9
|
Huang Y, Hu R, Wu L, He K, Ma R. Immunoregulation of Glia after spinal cord injury: a bibliometric analysis. Front Immunol 2024; 15:1402349. [PMID: 38938572 PMCID: PMC11208308 DOI: 10.3389/fimmu.2024.1402349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Accepted: 05/27/2024] [Indexed: 06/29/2024] Open
Abstract
Objective Immunoregulation is a complex and critical process in the pathological process of spinal cord injury (SCI), which is regulated by various factors and plays an important role in the functional repair of SCI. This study aimed to explore the research hotspots and trends of glial cell immunoregulation after SCI from a bibliometric perspective. Methods Data on publications related to glial cell immunoregulation after SCI, published from 2004 to 2023, were obtained from the Web of Science Core Collection. Countries, institutions, authors, journals, and keywords in the topic were quantitatively analyzed using the R package "bibliometrix", VOSviewer, Citespace, and the Bibliometrics Online Analysis Platform. Results A total of 613 papers were included, with an average annual growth rate of 9.39%. The papers came from 36 countries, with the United States having the highest output, initiating collaborations with 27 countries. Nantong University was the most influential institution. We identified 3,177 authors, of whom Schwartz, m, of the Weizmann Institute of Science, was ranked first regarding both field-specific H-index (18) and average number of citations per document (151.44). Glia ranked first among journals with 2,574 total citations. The keywords "microglia," "activation," "macrophages," "astrocytes," and "neuroinflammation" represented recent hot topics and are expected to remain a focus of future research. Conclusion These findings strongly suggest that the immunomodulatory effects of microglia, astrocytes, and glial cell interactions may be critical in promoting nerve regeneration and repair after SCI. Research on the immunoregulation of glial cells after SCI is emerging, and there should be greater cooperation and communication between countries and institutions to promote the development of this field and benefit more SCI patients.
Collapse
Affiliation(s)
- Yi Huang
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, The Third School of Clinical Medicine (School of Rehabilitation Medicine), Zhejiang Chinese Medical University, Hangzhou, China
| | - Rong Hu
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, The Third School of Clinical Medicine (School of Rehabilitation Medicine), Zhejiang Chinese Medical University, Hangzhou, China
| | - Lei Wu
- Department of Acupuncture, The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Kelin He
- Department of Acupuncture, The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Ruijie Ma
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, The Third School of Clinical Medicine (School of Rehabilitation Medicine), Zhejiang Chinese Medical University, Hangzhou, China
- Department of Acupuncture, The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
10
|
Tahmasebi F, Asl ER, Vahidinia Z, Faghihi F, Barati S. The comparative effects of bone marrow mesenchymal stem cells and supernatant transplantation on demyelination and inflammation in cuprizone model. Mol Biol Rep 2024; 51:674. [PMID: 38787497 DOI: 10.1007/s11033-024-09628-w] [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: 11/21/2023] [Accepted: 05/08/2024] [Indexed: 05/25/2024]
Abstract
BACKGROUND Multiple sclerosis (MS) is a chronic demyelinating disease of the central nervous system (CNS) with inflammation and immune dysfunction. OBJECTIVES We compared the remyelination and immunomodulation properties of mesenchymal stem cells (MSCs) with their conditioned medium (CM) in the cuprizone model. METHODS Twenty-four C57BL/ 6 mice were divided into four groups. After cuprizone demyelination, MSCs and their CM were injected into the right lateral ventricle of mice. The expression level of IL-1β, TNF-α, and BDNF genes was evaluated using the qRT-PCR. APC antibody was used to assess the oligodendrocyte population using the immunofluorescent method. The remyelination and axonal repair were studied by specific staining of the LFB and electron microscopy techniques. RESULTS Transplantation of MSCs and CM increased the expression of the BDNF gene and decreased the expression of IL-1β and TNF-α genes compared to the cuprizone group, and these effects in the cell group were more than CM. Furthermore, cell transplantation resulted in a significant improvement in myelination and axonal repair, which was measured by luxol fast blue and transmission electron microscope images. The cell group had a higher number of oligodendrocytes than other groups. CONCLUSIONS According to the findings, injecting MSCs intraventricularly versus cell-conditioned medium can be a more effective approach to improving chronic demyelination in degenerative diseases like MS.
Collapse
Affiliation(s)
- Fatemeh Tahmasebi
- Department of Anatomy, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Elmira Roshani Asl
- Department of Biochemistry, Saveh University of Medical Sciences, Saveh, Iran
| | - Zeinab Vahidinia
- Anatomical Sciences Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Faezeh Faghihi
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
- Pad Nahad Tabiat Company, Ltd, Tehran, Iran
| | - Shirin Barati
- Department of Anatomy, Saveh University of Medical Sciences, Saveh, Iran.
| |
Collapse
|
11
|
Wang Z, Li J, Xu T, Guo B, Xie Z, Li M. The Efficacy of Different Material Scaffold-Guided Cell Transplantation in the Treatment of Spinal Cord Injury in Rats: A Systematic Review and Network Meta-analysis. Cell Mol Neurobiol 2024; 44:43. [PMID: 38703332 PMCID: PMC11069479 DOI: 10.1007/s10571-024-01465-6] [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: 12/17/2023] [Accepted: 02/23/2024] [Indexed: 05/06/2024]
Abstract
Cell transplantation is a promising treatment option for spinal cord injury (SCI). However, there is no consensus on the choice of carrier scaffolds to host the cells. This study aims to evaluate the efficacy of different material scaffold-mediated cell transplantation in treating SCI in rats. According to PRISMA's principle, Embase, PubMed, Web of Science, and Cochrane databases were searched, and relevant literature was referenced. Only original research on cell transplantation plus natural or synthetic scaffolds in SCI rats was included. Direct and indirect evidence for improving hind limb motor function was pooled through meta-analysis. A subgroup analysis of some factors that may affect the therapeutic effect was conducted to understand the results fully. In total, 25 studies met the inclusion criteria, in which 293 rats received sham surgery, 78 rats received synthetic material scaffolds, and 219 rats received natural materials scaffolds. The network meta-analysis demonstrated that although synthetic scaffolds were slightly inferior to natural scaffolds in terms of restoring motor function in cell transplantation of SCI rats, no statistical differences were observed between the two (MD: -0.35; 95% CI -2.6 to 1.9). Moreover, the subgroup analysis revealed that the type and number of cells may be important factors in therapeutic efficacy (P < 0.01). Natural scaffolds and synthetic scaffolds are equally effective in cell transplantation of SCI rats without significant differences. In the future, the findings need to be validated in multicenter, large-scale, randomized controlled trials in clinical practice. Trial registration: Registration ID CRD42024459674 (PROSPERO).
Collapse
Affiliation(s)
- Zhihua Wang
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, No.17, Yongwai Street, Nanchang, 330006, Jiangxi Province, China
- Postdoctoral Innovation Practice Base, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| | - Jun Li
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, No.17, Yongwai Street, Nanchang, 330006, Jiangxi Province, China
- Department of the Second Clinical Medical College of Nanchang University, No.460, BaYi Street, Nanchang, 330006, Jiangxi Province, China
| | - Tianqi Xu
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, No.17, Yongwai Street, Nanchang, 330006, Jiangxi Province, China
- Department of the Second Clinical Medical College of Nanchang University, No.460, BaYi Street, Nanchang, 330006, Jiangxi Province, China
| | - Boyu Guo
- Department of the First Clinical Medical College of Nanchang University, No.460, BaYi Street, Nanchang, 330006, Jiangxi Province, China
| | - Zhiping Xie
- Department of Neurosurgery, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, No.152 Aiguo Road, Nanchang, 330006, Jiangxi Province, China.
- Department of Neurosurgery, Xiangya Hospital Jiangxi Hospital, Central South University, Nanchang, Jiangxi Province, China.
| | - Meihua Li
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, No.17, Yongwai Street, Nanchang, 330006, Jiangxi Province, China.
| |
Collapse
|
12
|
Jiu J, Liu H, Li D, Li J, Liu L, Yang W, Yan L, Li S, Zhang J, Li X, Li JJ, Wang B. 3D bioprinting approaches for spinal cord injury repair. Biofabrication 2024; 16:032003. [PMID: 38569491 DOI: 10.1088/1758-5090/ad3a13] [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: 10/10/2023] [Accepted: 04/03/2024] [Indexed: 04/05/2024]
Abstract
Regenerative healing of spinal cord injury (SCI) poses an ongoing medical challenge by causing persistent neurological impairment and a significant socioeconomic burden. The complexity of spinal cord tissue presents hurdles to successful regeneration following injury, due to the difficulty of forming a biomimetic structure that faithfully replicates native tissue using conventional tissue engineering scaffolds. 3D bioprinting is a rapidly evolving technology with unmatched potential to create 3D biological tissues with complicated and hierarchical structure and composition. With the addition of biological additives such as cells and biomolecules, 3D bioprinting can fabricate preclinical implants, tissue or organ-like constructs, andin vitromodels through precise control over the deposition of biomaterials and other building blocks. This review highlights the characteristics and advantages of 3D bioprinting for scaffold fabrication to enable SCI repair, including bottom-up manufacturing, mechanical customization, and spatial heterogeneity. This review also critically discusses the impact of various fabrication parameters on the efficacy of spinal cord repair using 3D bioprinted scaffolds, including the choice of printing method, scaffold shape, biomaterials, and biological supplements such as cells and growth factors. High-quality preclinical studies are required to accelerate the translation of 3D bioprinting into clinical practice for spinal cord repair. Meanwhile, other technological advances will continue to improve the regenerative capability of bioprinted scaffolds, such as the incorporation of nanoscale biological particles and the development of 4D printing.
Collapse
Affiliation(s)
- Jingwei Jiu
- Department of Orthopaedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- Department of Orthopaedic Surgery, Shanxi Medical University Second Affiliated Hospital, Taiyuan, People's Republic of China
| | - Haifeng Liu
- Department of Orthopaedic Surgery, Shanxi Medical University Second Affiliated Hospital, Taiyuan, People's Republic of China
| | - Dijun Li
- Department of Orthopaedic Surgery, Shanxi Medical University Second Affiliated Hospital, Taiyuan, People's Republic of China
| | - Jiarong Li
- School of Biomedical Engineering, Faculty of Engineering and IT, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Lu Liu
- School of Biomedical Engineering, Faculty of Engineering and IT, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Wenjie Yang
- School of Biomedical Engineering, Faculty of Engineering and IT, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Lei Yan
- Department of Orthopaedic Surgery, Shanxi Medical University Second Affiliated Hospital, Taiyuan, People's Republic of China
| | - Songyan Li
- Department of Orthopaedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Jing Zhang
- Department of Emergency Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550001, People's Republic of China
| | - Xiaoke Li
- Department of Orthopaedic Surgery, Shanxi Medical University Second Affiliated Hospital, Taiyuan, People's Republic of China
| | - Jiao Jiao Li
- School of Biomedical Engineering, Faculty of Engineering and IT, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Bin Wang
- Department of Orthopaedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| |
Collapse
|
13
|
Kun W, Xiaomei C, Lei Y, Huizhi Z. Modulating Th1/Th2 drift in asthma-related immune inflammation by enhancing bone mesenchymal stem cell homing through targeted inhibition of the Notch1/Jagged1 signaling pathway. Int Immunopharmacol 2024; 130:111713. [PMID: 38387192 DOI: 10.1016/j.intimp.2024.111713] [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: 12/10/2023] [Revised: 01/29/2024] [Accepted: 02/14/2024] [Indexed: 02/24/2024]
Abstract
Asthma, a disease intricately linked to immune inflammation, is significantly influenced by the immune regulatory effect of bone mesenchymal stem cells (BMSCs). This study aims to investigate changes in the homing of BMSCs in bronchial asthma, focusing on the Notch homolog (Notch)1/Jagged1 signaling pathway's role in regulating T helper 1(Th1)/T helper 2(Th2) drift. Additionally, we further explore the effects and mechanisms of homologous BMSCs implantation in asthma-related immune inflammation. Following intervention with BMSCs, a significant improvement in the pathology of rats with asthma was observed. Simultaneously, a reduction in the expression of inflammatory cells and inflammatory cytokines, including tumor necrosis factor-α (TNF-α), interleukin(IL)-4, and IL-13 was observed in bronchoalveolar lavage fluid (BALF). Furthermore, there was an increase in the expression of Th1 cytokine Interferon-γ(IFN-γ)and the transcription factor T-box expressed in T cell (T-bet), while the expression of Th2 cytokine IL-13 and transcription factor GATA binding protein (GATA)-3 decreased in lung tissue. This indicates that the Th1/Th2 drift leans towards Th1, which a crucial in ameliorating asthma inflammation. Importantly, inhibition of the Notch1 signaling pathway led to an increased expression of the Stromal cell-derived factor-1(SDF-1)/C-X-C motif chemokine receptor (CXCR)4 chemokine axis. Consequently, the homing ability of bone marrow mesenchymal stem cells to asthma-affected lung tissue was significantly enhanced. BMSCs demonstrated heightened efficacy in regulating the cytokine/chemokine network and Th1/Th2 balance, thereby restoring a stable state during the immune response process in asthma. In conclusion, inhibiting the Notch signaling pathway enhances the expression of the SDF-1 and CXCR4 chemokine axis, facilitating the migration of allogeneic BMSCs to injured lung tissues. This, in turn, promotes immune regulation and improves the Th1/Th2 imbalance, thereby enhancing the therapeutic effect on asthmatic airway inflammation.
Collapse
Affiliation(s)
- Wang Kun
- Huixue Research Center, Anhui University of Chinese Medicine, Hefei 230038, China; College of Traditional Chinese Medicine, Anhui University of Chinese Medicine, Hefei 230012, China; Key Laboratory of Xin'an Medical Science, Ministry of Education, Anhui University of Chinese Medicine, Hefei 230038, China
| | - Cao Xiaomei
- College of Traditional Chinese Medicine, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Yang Lei
- Intensive Care Department, The Second Affiliated Hospital of Anhui University of Chinese Medicine, Hefei 230061, China
| | - Zhu Huizhi
- The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei 230031, China.
| |
Collapse
|
14
|
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
|
15
|
Luan Z, Zhang J, Wang Y. Identification of marker genes for spinal cord injury. Front Med (Lausanne) 2024; 11:1364380. [PMID: 38463490 PMCID: PMC10921937 DOI: 10.3389/fmed.2024.1364380] [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: 01/06/2024] [Accepted: 02/07/2024] [Indexed: 03/12/2024] Open
Abstract
Introduction Spinal cord injury (SCI) is a profoundly disabling and devastating neurological condition, significantly impacting patients' quality of life. It imposes unbearable psychological and economic pressure on both patients and their families, as well as placing a heavy burden on society. Methods In this study, we integrated datasets GSE5296 and GSE47681 as training groups, analyzed gene variances between sham group and SCI group mice, and conducted Gene Ontology (GO) enrichment analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis based on the differentially expressed genes. Subsequently, we performed Weighted Gene Correlation Network Analysis (WGCNA) and Lasso regression analyses. Results We identified four characteristic disease genes: Icam1, Ch25h, Plaur and Tm4sf1. We examined the relationship between SCI and immune cells, and validated the expression of the identified disease-related genes in SCI rats using PCR and immunohistochemistry experiments. Discussion In conclusion, we have identified and verified four genes related to SCI: Icam1, Ch25h, Plaur and Tm4sf1, which could offer insights for SCI treatment.
Collapse
Affiliation(s)
- Zhiwei Luan
- Department of Orthopedic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China
| | - Jiayu Zhang
- Department of Hygienic Toxicology, College of Public Health, Harbin Medical University, Harbin, China
| | - Yansong Wang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- NHC Key Laboratory of Cell Transplantation, Harbin Medical University, Harbin, China
- Heilongjiang Provincial Key Laboratory of Hard Tissue Development and Regeneration, Harbin Medical University, Harbin, China
| |
Collapse
|
16
|
Zhang J, Zhang X, Jiang Q, Qu D, Hu Y, Qi C, Fu H. [Experimental study of M2 microglia transplantation promoting spinal cord injury repair in mice]. ZHONGGUO XIU FU CHONG JIAN WAI KE ZA ZHI = ZHONGGUO XIUFU CHONGJIAN WAIKE ZAZHI = CHINESE JOURNAL OF REPARATIVE AND RECONSTRUCTIVE SURGERY 2024; 38:198-205. [PMID: 38385233 PMCID: PMC10882233 DOI: 10.7507/1002-1892.202311093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 02/23/2024]
Abstract
Objective To investigate the effect of M2 microglia (M2-MG) transplantation on spinal cord injury (SCI) repair in mice. Methods Primary MG were obtained from the cerebral cortex of 15 C57BL/6 mice born 2-3 days old by pancreatic enzyme digestion and identified by immunofluorescence staining of Iba1. Then the primary MG were co-cultured with interleukin 4 for 48 hours (experimental group) to induce into M2 phenotype and identified by immunofluorescence staining of Arginase 1 (Arg-1) and Iba1. The normal MG were harvested as control (control group). The dorsal root ganglion (DRG) of 5 C57BL/6 mice born 1 week old were co-cultured with M2-MG for 5 days to observe the axon length, the DRG alone was used as control. Forty-two 6-week-old female C57BL/6 mice were randomly divided into sham group ( n=6), SCI group ( n=18), and SCI+M2-MG group ( n=18). In sham group, only the laminae of T 10 level were removed; SCI group and SCI+M2-MG group underwent SCI modeling, and SCI+M2-MG group was simultaneously injected with M2-MG. The survival of mice in each group was observed after operation. At immediate (0), 3, 7, 14, 21, and 28 days after operation, the motor function of mice was evaluated by Basso Mouse Scale (BMS) score, and the gait was evaluated by footprint experiment at 28 days. The spinal cord tissue was taken after operation for immunofluorescence staining, in which glial fibrillary acidic protein (GFAP) staining at 7, 14, and 28 days was used to observe the injured area of the spinal cord, neuronal nuclei antigen staining at 28 days was used to observe the survival of neurons, and GFAP/C3 double staining at 7 and 14 days was used to observe the changes in the number of A1 astrocytes. Results The purity of MG in vitro reached 90%, and the most of the cells were polarized into M2 phenotype identified by Arg-1 immunofluorescence staining. M2-MG promoted the axon growth when co-cultured with DRGs in vitro ( P<0.05). All groups of mice survived until the experiment was completed. The hind limb motor function of SCI group and SCI+M2-MG group gradually recovered over time. Among them, the SCI+M2-MG group had significantly higher BMS scores than the SCI group at 21 and 28 days ( P<0.05), and the dragging gait significantly improved at 28 days, but it did not reach the level of the sham group. Immunofluorescence staining showed that compared with the SCI group, the SCI+M2-MG group had a smaller injury area at 7, 14, and 28 days, an increase in neuronal survival at 28 days, and a decrease in the number of A1 astrocytes at 7 and 14 days, with significant differences ( P<0.05). Conclusion M2-MG transplantation improves the motor function of the hind limbs of SCI mice by promoting neuron survival and axon regeneration. This neuroprotective effect is related to the inhibition of A1 astrocytes polarization.
Collapse
Affiliation(s)
- Jing Zhang
- Qingdao Medical College of Qingdao University, Qingdao Shandong, 266073, P. R. China
| | - Xiaoyue Zhang
- Qingdao Medical College of Qingdao University, Qingdao Shandong, 266073, P. R. China
| | - Qi Jiang
- Qingdao Medical College of Qingdao University, Qingdao Shandong, 266073, P. R. China
| | - Di Qu
- Qingdao Medical College of Qingdao University, Qingdao Shandong, 266073, P. R. China
| | - Yusheng Hu
- Qingdao Medical College of Qingdao University, Qingdao Shandong, 266073, P. R. China
| | - Chao Qi
- Department of Sports Medicine, the Affiliated Hospital of Qingdao University, Qingdao Shandong, 266103, P. R. China
| | - Haitao Fu
- Department of Sports Medicine, the Affiliated Hospital of Qingdao University, Qingdao Shandong, 266103, P. R. China
| |
Collapse
|
17
|
Yari D, Saberi A, Salmasi Z, Ghoreishi SA, Etemad L, Movaffagh J, Ganjeifar B. Recent Advances in the Treatment of Spinal Cord Injury. THE ARCHIVES OF BONE AND JOINT SURGERY 2024; 12:380-399. [PMID: 38919744 PMCID: PMC11195032 DOI: 10.22038/abjs.2023.73944.3424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 10/07/2023] [Indexed: 06/27/2024]
Abstract
Spinal cord injury (SCI) is a complex, multifaceted, progressive, and yet incurable complication that can cause irreversible damage to the individual, family, and society. In recent years strategies for the management and rehabilitation of SCI besides axonal regeneration, remyelination, and neuronal plasticity of the injured spinal cord have significantly improved. Although most of the current research and therapeutic advances have been made in animal models, so far, no specific and complete treatment has been reported for SCI in humans. The failure to treat this complication has been due to the inherent neurological complexity and the structural, cellular, molecular, and biochemical characteristics of spinal cord injury. In this review, in addition to elucidating the causes of spinal cord injury from a molecular and pathophysiological perspective, the complexity and drawbacks of neural regeneration that lead to the failure in SCI treatment are described. Also, recent advances and cutting-edge strategies in most areas of SCI treatment are presented.
Collapse
Affiliation(s)
- Davood Yari
- Department of Clinical Biochemistry, Babol University of Medical Sciences, Babol, Iran
- Orthopedic Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Arezoo Saberi
- Department of Pharmaceutics, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zahra Salmasi
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Alireza Ghoreishi
- Orthopedic Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Orthopedic Surgery, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Leila Etemad
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Jebrail Movaffagh
- Department of Pharmaceutics, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
- Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Babak Ganjeifar
- Department of Neurosurgery, Ghaem Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
18
|
Golmakani H, Azimian A, Golmakani E. Newly discovered functions of miRNAs in neuropathic pain: Transitioning from recent discoveries to innovative underlying mechanisms. Mol Pain 2024; 20:17448069231225845. [PMID: 38148597 PMCID: PMC10851769 DOI: 10.1177/17448069231225845] [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: 08/21/2023] [Revised: 09/12/2023] [Accepted: 10/11/2023] [Indexed: 12/28/2023] Open
Abstract
Neuropathic pain is a widespread clinical issue caused by somatosensory nervous system damage, affecting numerous individuals. It poses considerable economic and public health challenges, and managing it can be challenging due to unclear underlying mechanisms. Nevertheless, emerging evidence suggests that neurogenic inflammation and neuroinflammation play a role in developing pain patterns. Emerging evidence suggests that neurogenic inflammation and neuroinflammation play significant roles in developing neuropathic pain within the nervous system. Increased/decreased miRNA expression patterns could affect the progression of neuropathic and inflammatory pain by controlling nerve regeneration, neuroinflammation, and the expression of abnormal ion channels. However, our limited knowledge of miRNA targets hinders a complete grasp of miRNA's functions. Meanwhile, exploring exosomal miRNA, a recently uncovered role, has significantly advanced our comprehension of neuropathic pain's pathophysiology in recent times. In this review, we present a comprehensive overview of the latest miRNA studies and explore the possible ways miRNAs might play a role in the development of neuropathic pain.
Collapse
Affiliation(s)
- Hasan Golmakani
- Department of Pediatrics, Faculty of Medicine, Mashhad Azad University, Mashhad, Iran
| | - Amir Azimian
- Department of Pathobiology and Laboratory Sciences, Faculty of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Ebrahim Golmakani
- Department of Anesthesiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
19
|
Wang Y, Ding Y, Guo C. Mesenchymal Stem Cells for the Treatment of Spinal Cord Injury in Rat Models: A Systematic Review and Network Meta-Analysis. Cell Transplant 2024; 33:9636897241262992. [PMID: 38910431 PMCID: PMC11265244 DOI: 10.1177/09636897241262992] [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: 04/09/2024] [Revised: 05/23/2024] [Accepted: 06/01/2024] [Indexed: 06/25/2024] Open
Abstract
Transplantation of mesenchymal stem cells (MSCs) is one of the hopeful treatments for spinal cord injury (SCI). Most current studies are in animals, and less in humans, and the optimal transplantation strategy for MSCs is still controversial. In this article, we explore the optimal transplantation strategy of MSCs through a network meta-analysis of the effects of MSCs on SCI in animal models. PubMed, Web of Science, Cochrane Library, Embase, China National Knowledge Infrastructure (CNKI), Wanfang Database, China Science and Technology Journal Database (VIP), and Chinese Biomedical Literature Service System (SinoMed) databases were searched by computer for randomized controlled studies on MSCs for SCI. Two investigators independently completed the literature screening and data extraction based on the inclusion and exclusion criteria. RevMan 5.4 software was used to assess the quality of the included literature. Stata 16.0 software was used for standard meta-analysis and network meta-analysis. Standardized mean difference (SMD) was used for continuous variables to combine the statistics and calculate 95% confidence interval (95% CI). P < 0.05 was considered a statistically significant difference. Cochrane's Q test and the I2 value were used to indicate the magnitude of heterogeneity. A random-effects model was used if I2 > 50% and P < 0.10 indicated significant heterogeneity between studies, and conversely, a fixed-effects model was used. Evidence network diagrams were drawn based on direct comparisons between various interventions. The surface under the cumulative ranking curve area (SUCRA) was used to predict the ranking of the treatment effects of each intervention. A total of 32 animal studies were included in this article for analysis. The results of the standard meta-analysis showed that MSCs improved motor ability after SCI. The network meta-analysis showed that the best treatment effect was achieved for adipose tissue-derived mesenchymal stromal cells (ADMSCs) in terms of cell source and intrathecal (IT) in terms of transplantation modality. For transplantation timing, the best treatment effect was achieved when transplantation was performed in the subacute phase. The available literature suggests that IT transplantation using ADMSCs in the subacute phase may be the best transplantation strategy to improve functional impairment after SCI. Future high-quality studies are still needed to further validate the results of this study to ensure the reliability of the results.
Collapse
Affiliation(s)
- Yueying Wang
- College of Rehabilitation Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yi Ding
- Department of Rehabilitation Medicine, The Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Chenchen Guo
- Department of Rehabilitation Medicine, Neck, Shoulder, Lumbago and Leg Pain Hospital Affiliated to Shandong First Medical University, Jinan, China
| |
Collapse
|
20
|
Wang Y, Xiong Z, Zhang Q, Liu M, Zhang J, Qi X, Jiang X, Yu W. Acetyl-11-Keto-β-Boswellic Acid Accelerates the Repair of Spinal Cord Injury in Rats by Resisting Neuronal Pyroptosis with Nrf2. Int J Mol Sci 2023; 25:358. [PMID: 38203528 PMCID: PMC10779011 DOI: 10.3390/ijms25010358] [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: 10/29/2023] [Revised: 12/14/2023] [Accepted: 12/21/2023] [Indexed: 01/12/2024] Open
Abstract
The primary aim of this study is to delve into the potential of Acetyl-11-keto-β-boswellic acid (AKBA) in ameliorating neuronal damage induced by acute spinal cord injury, as well as to unravel the intricate underlying mechanisms. A cohort of 40 Sprague-Dawley rats was meticulously categorized into four groups. Following a seven-day oral administration of AKBA, damaged spinal cord samples were meticulously procured for Nissl staining and electron microscopy to assess neuronal demise. Employing ELISA, immunofluorescence, Western blot (WB), and quantitative polymerase chain reaction (qPCR), the modulatory effects of AKBA within the context of spinal cord injury were comprehensively evaluated. Furthermore, employing an ex vivo extraction of spinal cord neurons, an ATP + LPS-induced pyroptotic injury model was established. The model was subsequently subjected to Nrf2 inhibition, followed by a battery of assessments involving ELISA, DCFH-DA staining, flow cytometry, immunofluorescence, and WB to decipher the effects of AKBA on the spinal cord neuron pyroptosis model. By engaging the Nrf2-ROS-NLRP3 pathway, AKBA exerted a repressive influence on the expression of the pyroptotic initiator protein Caspase-1, thereby mitigating the release of GSDMD and alleviating pyroptosis. Additionally, AKBA demonstrated the ability to attenuate the release of IL-18 and IL-1β, curbing neuronal loss and expediting the restorative processes within the context of spinal cord injury. Our study elucidates that AKBA can reduce spinal cord neuronal apoptosis, providing a basis for the development of AKBA as a clinical treatment for spinal cord injury.
Collapse
Affiliation(s)
- Yao Wang
- Department of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (Y.W.); (Z.X.); (Q.Z.); (M.L.); (J.Z.); (X.Q.)
| | - Zongliang Xiong
- Department of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (Y.W.); (Z.X.); (Q.Z.); (M.L.); (J.Z.); (X.Q.)
| | - Qiyuan Zhang
- Department of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (Y.W.); (Z.X.); (Q.Z.); (M.L.); (J.Z.); (X.Q.)
| | - Mengmeng Liu
- Department of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (Y.W.); (Z.X.); (Q.Z.); (M.L.); (J.Z.); (X.Q.)
| | - Jingjing Zhang
- Department of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (Y.W.); (Z.X.); (Q.Z.); (M.L.); (J.Z.); (X.Q.)
| | - Xinyue Qi
- Department of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (Y.W.); (Z.X.); (Q.Z.); (M.L.); (J.Z.); (X.Q.)
| | - Xiaowen Jiang
- Department of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (Y.W.); (Z.X.); (Q.Z.); (M.L.); (J.Z.); (X.Q.)
| | - Wenhui Yu
- Department of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (Y.W.); (Z.X.); (Q.Z.); (M.L.); (J.Z.); (X.Q.)
- Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Northeast Agricultural University, Harbin 150030, China
- Institute of Chinese Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| |
Collapse
|
21
|
Mi X, Ni C, Zhao J, Amin N, Jiao D, Fang M, Ye X. P2Y12 receptor mediates apoptosis and demyelination to affect functional recovery in mice with spinal cord injury. Neurochem Int 2023; 171:105641. [PMID: 37952830 DOI: 10.1016/j.neuint.2023.105641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 09/28/2023] [Accepted: 11/06/2023] [Indexed: 11/14/2023]
Abstract
Among diseases of the central nervous system (CNS), spinal cord injury (SCI) has a high fatality rate. It has been proven that P2Y G protein-coupled purinergic receptors have a neuroprotective role in apoptosis and regeneration inside the damaged spinal cord. The P2Y12 receptor (P2Y12R) has recently been linked to peripheral neuropathy and stroke. However, the role of P2Y12R after SCI remains unclear. Our study randomly divided C57BL/6J female mice into 3 groups: Sham+DMSO, SCI+DMSO, and SCI+MRS2395. MRS2395 as a P2Y12R inhibitor was intraperitoneally injected at a dose of 1.5 mg/kg once daily for 7 days. We showed that the P2Y12R was markedly activated after injury, and it was double labeled with the microglial and neuron. Behavioral tests were employed to assess motor function recovery. By using immunofluorescence staining, the NeuN expression level was detected. The morphology of neurons was observed by hematoxylin-eosin and Nissl staining. P2Y12R, Bax, GFAP, PCNA and calbindin expression levels were detected using Western blot. Meanwhile, mitochondria and myelin sheath were observed by transmission electron microscopy (TEM). Our findings demonstrated that MRS2395 significantly enhanced motor function induced by SCI and that was used to alleviate apoptosis and astrocyte scarring. NeuN positive cells in the SCI group were lower than in the therapy group, although Bax, GFAP, PCNA and calbindin expression levels were considerably higher. Moreover, following MRS2395 therapy, the histological damage was reversed. A notable improvement in myelin sheath and mitochondrial morphology was seen in the therapy group. Together, our findings indicate that activation of P2Y12R in damaged spinal cord may be a critical event and suggest that inhibition of P2Y12R might be a feasible therapeutic strategy for treating SCI.
Collapse
Affiliation(s)
- Xiaodan Mi
- Center for Rehabilitation Medicine, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Department of Rehabilitation Medicine, Zhejiang Provincial People's Hospital Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Chengtao Ni
- Graduate School, Bengbu Medical College, Bengbu, Anhui, China
| | - Jingting Zhao
- Center for Rehabilitation Medicine, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Department of Rehabilitation Medicine, Zhejiang Provincial People's Hospital Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Nashwa Amin
- Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China; Institute of System Medicine, Zhejiang University School of Medicine, Hangzhou, China; Department of Zoology, Faculty of Science, Aswan University, Egypt
| | - Dian Jiao
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, China
| | - Marong Fang
- Children's Hospital of Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China.
| | - Xiangming Ye
- Center for Rehabilitation Medicine, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Department of Rehabilitation Medicine, Zhejiang Provincial People's Hospital Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China.
| |
Collapse
|
22
|
He S, Wang Q, Chen L, He YJ, Wang X, Qu S. miR-100a-5p-enriched exosomes derived from mesenchymal stem cells enhance the anti-oxidant effect in a Parkinson's disease model via regulation of Nox4/ROS/Nrf2 signaling. J Transl Med 2023; 21:747. [PMID: 37875930 PMCID: PMC10594913 DOI: 10.1186/s12967-023-04638-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 10/17/2023] [Indexed: 10/26/2023] Open
Abstract
BACKGROUND The pathogenesis of Parkinson's disease (PD) has not been fully elucidated, and there are no effective disease-modifying drugs for the treatment of PD. Mesenchymal stem cells have been used to treat several diseases, but are not readily available. METHODS Here, we used phenotypically uniform trophoblast stage-derived mesenchymal stem cells (T-MSCs) from embryonic stem cells, which are capable of stable production, and their exosomes (T-MSCs-Exo) to explore the molecular mechanisms involved in dopaminergic (DA) neuron protection in PD models using experimental assays (e.g., western blotting, immunofluorescence and immunohistochemistry staining). RESULTS We assessed the levels of DA neuron injury and oxidative stress in MPTP-induced PD mice and MPP+-induced MN9D cells after treating them with T-MSCs or T-MSCs-Exo. Furthermore, T-MSCs-Exo miRNA sequencing analysis revealed that miR-100-5p-enriched T-MSCs-Exo directly targeted the 3' UTR of NOX4, which could protect against the loss of DA neurons, maintain nigro-striatal system function, ameliorate motor deficits, and reduce oxidative stress via the Nox4-ROS-Nrf2 axis in PD models. CONCLUSIONS The study suggests that miR-100-5p-enriched T-MSCs-Exo may be a promising biological agent for the treatment of PD. Schematic summary of the mechanism underlying the neuroprotective actions of T-MSCs-Exo in PD. T-MSCs Exo may inhibit the expression level of the target gene NOX4 by delivering miR-100-5p, thereby reducing ROS production and alleviating oxidative stress via the Nox4-ROS-Nrf2 axis, thus improving DA neuron damage in PD.
Collapse
Affiliation(s)
- Songzhe He
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
- Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangzhou, 510515, Guangdong, China
- Key Laboratory of Mental Health of the Ministry of Education, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Qiongqiong Wang
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
- Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangzhou, 510515, Guangdong, China
- Key Laboratory of Mental Health of the Ministry of Education, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Liankuai Chen
- ImStem Biotechnology, Inc., 400 Farmington Avenue R1808, Farmington, CT, 06030, USA
- Zhuhai Hengqin ImStem Biotechnology Co., Ltd, Hengqin New District Huandao Donglu 1889 Building 3, Zhuhai, 519000, Guangdong, China
| | - Yusheng Jason He
- ImStem Biotechnology, Inc., 400 Farmington Avenue R1808, Farmington, CT, 06030, USA
- Zhuhai Hengqin ImStem Biotechnology Co., Ltd, Hengqin New District Huandao Donglu 1889 Building 3, Zhuhai, 519000, Guangdong, China
| | - Xiaofang Wang
- ImStem Biotechnology, Inc., 400 Farmington Avenue R1808, Farmington, CT, 06030, USA
- Zhuhai Hengqin ImStem Biotechnology Co., Ltd, Hengqin New District Huandao Donglu 1889 Building 3, Zhuhai, 519000, Guangdong, China
| | - Shaogang Qu
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China.
- Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangzhou, 510515, Guangdong, China.
- Key Laboratory of Mental Health of the Ministry of Education, Southern Medical University, Guangzhou, 510515, Guangdong, China.
- Department of Neurology, Ganzhou Hospital-Nanfang Hospital, Southern Medical University, Ganzhou, 341000, Jiangxi, China.
| |
Collapse
|
23
|
Chen SY, Yang RL, Wu XC, Zhao DZ, Fu SP, Lin FQ, Li LY, Yu LM, Zhang Q, Zhang T. Mesenchymal Stem Cell Transplantation: Neuroprotection and Nerve Regeneration After Spinal Cord Injury. J Inflamm Res 2023; 16:4763-4776. [PMID: 37881652 PMCID: PMC10595983 DOI: 10.2147/jir.s428425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Accepted: 10/03/2023] [Indexed: 10/27/2023] Open
Abstract
Spinal Cord Injury (SCI), with its morbidity characteristics of high disability rate and high mortality rate, is a disease that is highly destructive to both the physiology and psychology of the patient, and for which there is still a lack of effective treatment. Following spinal cord injury, a cascade of secondary injury reactions known as ischemia, peripheral inflammatory cell infiltration, oxidative stress, etc. create a microenvironment that is unfavorable to neural recovery and ultimately results in apoptosis and necrosis of neurons and glial cells. Mesenchymal stem cell (MSC) transplantation has emerged as a more promising therapeutic options in recent years. MSC can promote spinal cord injury repair through a variety of mechanisms, including immunomodulation, neuroprotection, and nerve regeneration, giving patients with spinal cord injury hope. In this paper, it is discussed the neuroprotection and nerve regeneration components of MSCs' therapeutic method for treating spinal cord injuries.
Collapse
Affiliation(s)
- Si-Yu Chen
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, People’s Republic of China
| | - Rui-Lin Yang
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, People’s Republic of China
| | - Xiang-Chong Wu
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, People’s Republic of China
| | - De-Zhi Zhao
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, People’s Republic of China
| | - Sheng-Ping Fu
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, People’s Republic of China
| | - Feng-Qin Lin
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, People’s Republic of China
| | - Lin-Yan Li
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, People’s Republic of China
| | - Li-Mei Yu
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, People’s Republic of China
| | - Qian Zhang
- Department of Human Anatomy, Zunyi Medical University, Zunyi, Guizhou, People’s Republic of China
| | - Tao Zhang
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, People’s Republic of China
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, People’s Republic of China
| |
Collapse
|
24
|
Zeng CW. Advancing Spinal Cord Injury Treatment through Stem Cell Therapy: A Comprehensive Review of Cell Types, Challenges, and Emerging Technologies in Regenerative Medicine. Int J Mol Sci 2023; 24:14349. [PMID: 37762654 PMCID: PMC10532158 DOI: 10.3390/ijms241814349] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 09/17/2023] [Accepted: 09/19/2023] [Indexed: 09/29/2023] Open
Abstract
Spinal cord injuries (SCIs) can lead to significant neurological deficits and lifelong disability, with far-reaching physical, psychological, and economic consequences for affected individuals and their families. Current treatments for SCIs are limited in their ability to restore function, and there is a pressing need for innovative therapeutic approaches. Stem cell therapy has emerged as a promising strategy to promote the regeneration and repair of damaged neural tissue following SCIs. This review article comprehensively discusses the potential of different stem cell types, such as embryonic stem cells (ESCs), induced pluripotent stem cells (iPSCs), mesenchymal stem cells (MSCs), and neural stem/progenitor cells (NSPCs), in SCI treatment. We provide an in-depth analysis of the unique advantages and challenges associated with each stem cell type, as well as the latest advancements in the field. Furthermore, we address the critical challenges faced in stem cell therapy for SCIs, including safety concerns, ethical considerations, standardization of protocols, optimization of transplantation parameters, and the development of effective outcome measures. We also discuss the integration of novel technologies such as gene editing, biomaterials, and tissue engineering to enhance the therapeutic potential of stem cells. The article concludes by emphasizing the importance of collaborative efforts among various stakeholders in the scientific community, including researchers, clinicians, bioengineers, industry partners, and patients, to overcome these challenges and realize the full potential of stem cell therapy for SCI patients. By fostering such collaborations and advancing our understanding of stem cell biology and regenerative medicine, we can pave the way for the development of groundbreaking therapies that improve the lives of those affected by SCIs.
Collapse
Affiliation(s)
- Chih-Wei Zeng
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA;
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
25
|
Zeng CW, Tsai HJ. The Promising Role of a Zebrafish Model Employed in Neural Regeneration Following a Spinal Cord Injury. Int J Mol Sci 2023; 24:13938. [PMID: 37762240 PMCID: PMC10530783 DOI: 10.3390/ijms241813938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 09/07/2023] [Accepted: 09/09/2023] [Indexed: 09/29/2023] Open
Abstract
Spinal cord injury (SCI) is a devastating event that results in a wide range of physical impairments and disabilities. Despite the advances in our understanding of the biological response to injured tissue, no effective treatments are available for SCIs at present. Some studies have addressed this issue by exploring the potential of cell transplantation therapy. However, because of the abnormal microenvironment in injured tissue, the survival rate of transplanted cells is often low, thus limiting the efficacy of such treatments. Many studies have attempted to overcome these obstacles using a variety of cell types and animal models. Recent studies have shown the utility of zebrafish as a model of neural regeneration following SCIs, including the proliferation and migration of various cell types and the involvement of various progenitor cells. In this review, we discuss some of the current challenges in SCI research, including the accurate identification of cell types involved in neural regeneration, the adverse microenvironment created by SCIs, attenuated immune responses that inhibit nerve regeneration, and glial scar formation that prevents axonal regeneration. More in-depth studies are needed to fully understand the neural regeneration mechanisms, proteins, and signaling pathways involved in the complex interactions between the SCI microenvironment and transplanted cells in non-mammals, particularly in the zebrafish model, which could, in turn, lead to new therapeutic approaches to treat SCIs in humans and other mammals.
Collapse
Affiliation(s)
- Chih-Wei Zeng
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA;
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Huai-Jen Tsai
- Department of Life Science, Fu Jen Catholic University, New Taipei City 242062, Taiwan
| |
Collapse
|
26
|
Chi H, Song M, Zhang J, Zhou J, Liu D. Relationship between acute glucose variability and cognitive decline in type 2 diabetes: A systematic review and meta-analysis. PLoS One 2023; 18:e0289782. [PMID: 37656693 PMCID: PMC10473499 DOI: 10.1371/journal.pone.0289782] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 07/25/2023] [Indexed: 09/03/2023] Open
Abstract
BACKGROUND Cognitive decline is one of the most widespread chronic complications of diabetes, which occurs in more than half of the patients with type 2 diabetes (T2DM). Emerging evidences have suggested that glucose variability (GV) is associated with the pathogenesis of diabetic complications. However, the influence of acute GV on cognitive dysfunction in T2DM is still controversial. The aim of the study was to evaluate the association between acute GV and cognitive defect in T2DM, and provide a most recent and comprehensive summary of the evidences in this research field. METHODS PubMed, Cochrane library, EMBASE, Web of science, Sinomed, China National Knowledge Infrastructure (CNKI), and Wanfang were searched for articles that reported on the association between acute GV and cognitive impairment in T2DM. RESULTS 9 eligible studies were included, with a total of 1263 patients with T2DM involved. Results showed that summary Fisher's z value was -0.23 [95%CI (-0.39, -0.06)], suggesting statistical significance (P = 0.006). Summary r value was -0.22 [95%CI (-0.37, -0.06)]. A lower cognitive performance was found in the subjects with greater glucose variation, which has statistical significance. Mean amplitude of glycemic excursions (MAGE) was associated with a higher risk of poor functional outcomes. Fisher's z value was -0.35 [95%CI (-0.43, -0.25)], indicating statistical significance (P = 0.011). Sensitivity analyses by omitting individual studies showed stability of the results. CONCLUSIONS Overall, higher acute GV is associated with an increased risk of cognitive impairment in patients with T2DM. Further studies should be required to determine whether targeted intervention of reducing acute GV could prevent cognitive decline.
Collapse
Affiliation(s)
- Haiyan Chi
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
- Department of Endocrinology, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai, Shandong, China
| | - Min Song
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Jinbiao Zhang
- Department of Neurology, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai, Shandong, China
| | - Junyu Zhou
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Deshan Liu
- Department of Traditional Chinese Medicine, Qilu Hospital of Shandong University, Jinan, Shandong, China
| |
Collapse
|
27
|
Shimizu Y, Ntege EH, Azuma C, Uehara F, Toma T, Higa K, Yabiku H, Matsuura N, Inoue Y, Sunami H. Management of Rheumatoid Arthritis: Possibilities and Challenges of Mesenchymal Stromal/Stem Cell-Based Therapies. Cells 2023; 12:1905. [PMID: 37508569 PMCID: PMC10378234 DOI: 10.3390/cells12141905] [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: 05/06/2023] [Revised: 06/28/2023] [Accepted: 07/14/2023] [Indexed: 07/30/2023] Open
Abstract
Rheumatoid arthritis (RA) is a highly prevalent, chronic, and progressive autoimmune disorder primarily affecting joints and muscles. The associated inflammation, pain, and motor restriction negatively impact patient quality of life (QOL) and can even contribute to premature mortality. Further, conventional treatments such as antiinflammatory drugs are only symptomatic. Substantial progress has been made on elucidating the etiopathology of overt RA, in particular the contributions of innate and adaptive immune system dysfunction to chronic inflammation. Although the precise mechanisms underlying onset and progression remain elusive, the discovery of new drug targets, early diagnosis, and new targeted treatments have greatly improved the prognosis and QOL of patients with RA. However, a sizable proportion of patients develop severe adverse effects, exhibit poor responses, or cannot tolerate long-term use of these drugs, necessitating more effective and safer therapeutic alternatives. Mounting preclinical and clinical evidence suggests that the transplantation of multipotent adult stem cells such as mesenchymal stromal/stem cells is a safe and effective treatment strategy for controlling chronic inflammation and promoting tissue regeneration in patients with intractable diseases, including RA. This review describes the current status of MSC-based therapies for RA as well as the opportunities and challenges to broader clinical application.
Collapse
Affiliation(s)
- Yusuke Shimizu
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, University of the Ryukyus, Nishihara 903-0215, Japan
| | - Edward Hosea Ntege
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, University of the Ryukyus, Nishihara 903-0215, Japan
| | - Chinatsu Azuma
- Department of Orthopedic Surgery, Graduate School of Medicine, University of the Ryukyus, Nishihara 903-0215, Japan
| | - Fuminari Uehara
- Department of Orthopedic Surgery, Graduate School of Medicine, University of the Ryukyus, Nishihara 903-0215, Japan
| | - Takashi Toma
- Department of Orthopedic Surgery, Graduate School of Medicine, University of the Ryukyus, Nishihara 903-0215, Japan
| | - Kotaro Higa
- Department of Orthopedic Surgery, Graduate School of Medicine, University of the Ryukyus, Nishihara 903-0215, Japan
| | - Hiroki Yabiku
- Department of Orthopedic Surgery, Graduate School of Medicine, University of the Ryukyus, Nishihara 903-0215, Japan
| | - Naoki Matsuura
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, University of the Ryukyus, Nishihara 903-0215, Japan
| | - Yoshikazu Inoue
- Department of Plastic and Reconstructive Surgery, School of Medicine, Fujita Health University, Toyoake 470-1192, Japan
| | - Hiroshi Sunami
- Center for Advanced Medical Research, School of Medicine, University of the Ryukyus, Nishihara 903-0215, Japan
| |
Collapse
|
28
|
Cai M, Chen L, Wang T, Liang Y, Zhao J, Zhang X, Li Z, Wu H. Hydrogel scaffolds in the treatment of spinal cord injury: a review. Front Neurosci 2023; 17:1211066. [PMID: 37325033 PMCID: PMC10266534 DOI: 10.3389/fnins.2023.1211066] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 05/12/2023] [Indexed: 06/17/2023] Open
Abstract
Spinal cord injury (SCI) is a disease of the central nervous system often caused by accidents, and its prognosis is unsatisfactory, with long-term adverse effects on patients' lives. The key to its treatment lies in the improvement of the microenvironment at the injury and the reconstruction of axons, and tissue repair is a promising therapeutic strategy. Hydrogel is a three-dimensional mesh structure with high water content, which has the advantages of biocompatibility, degradability, and adjustability, and can be used to fill pathological defects by injectable flowing hydrophilic material in situ to accurately adapt to the size and shape of the injury. Hydrogels mimic the natural extracellular matrix for cell colonization, guide axon extension, and act as a biological scaffold, which can be used as an excellent carrier to participate in the treatment of SCI. The addition of different materials to make composite hydrogel scaffolds can further enhance their performance in all aspects. In this paper, we introduce several typical composite hydrogels and review the research progress of hydrogel for SCI to provide a reference for the clinical application of hydrogel therapy for SCI.
Collapse
Affiliation(s)
- Manqi Cai
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
- Department of Surgery, The Third Hospital of Guangdong Medical University (Longjiang Hospital of Shunde District), Foshan, China
| | - Liji Chen
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
| | - Tao Wang
- Department of Surgery, The Third Hospital of Guangdong Medical University (Longjiang Hospital of Shunde District), Foshan, China
| | - Yinru Liang
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
| | - Jie Zhao
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
| | - Xiaomin Zhang
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
| | - Ziyi Li
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
- The Second Clinical Medical College, Guangdong Medical University, Dongguan, China
| | - Hongfu Wu
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
| |
Collapse
|
29
|
Huang LY, Sun X, Pan HX, Wang L, He CQ, Wei Q. Cell transplantation therapies for spinal cord injury focusing on bone marrow mesenchymal stem cells: Advances and challenges. World J Stem Cells 2023; 15:385-399. [PMID: 37342219 PMCID: PMC10277963 DOI: 10.4252/wjsc.v15.i5.385] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 02/17/2023] [Accepted: 03/21/2023] [Indexed: 05/26/2023] Open
Abstract
Spinal cord injury (SCI) is a devastating condition with complex pathological mechanisms that lead to sensory, motor, and autonomic dysfunction below the site of injury. To date, no effective therapy is available for the treatment of SCI. Recently, bone marrow-derived mesenchymal stem cells (BMMSCs) have been considered to be the most promising source for cellular therapies following SCI. The objective of the present review is to summarize the most recent insights into the cellular and molecular mechanism using BMMSC therapy to treat SCI. In this work, we review the specific mechanism of BMMSCs in SCI repair mainly from the following aspects: Neuroprotection, axon sprouting and/or regeneration, myelin regeneration, inhibitory microenvironments, glial scar formation, immunomodulation, and angiogenesis. Additionally, we summarize the latest evidence on the application of BMMSCs in clinical trials and further discuss the challenges and future directions for stem cell therapy in SCI models.
Collapse
Affiliation(s)
- Li-Yi Huang
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital/West China School of Medicine, Sichuan University, Chengdu 610044, Sichuan Province, China
| | - Xin Sun
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital/West China School of Medicine, Sichuan University, Chengdu 610044, Sichuan Province, China
| | - Hong-Xia Pan
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital/West China School of Medicine, Sichuan University, Chengdu 610044, Sichuan Province, China
| | - Lu Wang
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital/West China School of Medicine, Sichuan University, Chengdu 610044, Sichuan Province, China
| | - Cheng-Qi He
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital/West China School of Medicine, Sichuan University, Chengdu 610044, Sichuan Province, China
| | - Quan Wei
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital/West China School of Medicine, Sichuan University, Chengdu 610044, Sichuan Province, China
| |
Collapse
|
30
|
Chen S, Saeed AFUH, Liu Q, Jiang Q, Xu H, Xiao GG, Rao L, Duo Y. Macrophages in immunoregulation and therapeutics. Signal Transduct Target Ther 2023; 8:207. [PMID: 37211559 DOI: 10.1038/s41392-023-01452-1] [Citation(s) in RCA: 312] [Impact Index Per Article: 312.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 03/06/2023] [Accepted: 04/26/2023] [Indexed: 05/23/2023] Open
Abstract
Macrophages exist in various tissues, several body cavities, and around mucosal surfaces and are a vital part of the innate immune system for host defense against many pathogens and cancers. Macrophages possess binary M1/M2 macrophage polarization settings, which perform a central role in an array of immune tasks via intrinsic signal cascades and, therefore, must be precisely regulated. Many crucial questions about macrophage signaling and immune modulation are yet to be uncovered. In addition, the clinical importance of tumor-associated macrophages is becoming more widely recognized as significant progress has been made in understanding their biology. Moreover, they are an integral part of the tumor microenvironment, playing a part in the regulation of a wide variety of processes including angiogenesis, extracellular matrix transformation, cancer cell proliferation, metastasis, immunosuppression, and resistance to chemotherapeutic and checkpoint blockade immunotherapies. Herein, we discuss immune regulation in macrophage polarization and signaling, mechanical stresses and modulation, metabolic signaling pathways, mitochondrial and transcriptional, and epigenetic regulation. Furthermore, we have broadly extended the understanding of macrophages in extracellular traps and the essential roles of autophagy and aging in regulating macrophage functions. Moreover, we discussed recent advances in macrophages-mediated immune regulation of autoimmune diseases and tumorigenesis. Lastly, we discussed targeted macrophage therapy to portray prospective targets for therapeutic strategies in health and diseases.
Collapse
Affiliation(s)
- Shanze Chen
- Department of Respiratory Diseases and Critic Care Unit, Shenzhen Institute of Respiratory Disease, Shenzhen Key Laboratory of Respiratory Disease, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, China
| | - Abdullah F U H Saeed
- Department of Cancer Biology, Beckman Research Institute of City of Hope National Medical Center, Los Angeles, CA, 91010, USA
| | - Quan Liu
- Department of Laboratory Medicine, Huazhong University of Science and Technology Union Shenzhen Hospital (Nanshan Hospital), Shenzhen University, Shenzhen, 518052, China
| | - Qiong Jiang
- Department of Respiratory Diseases and Critic Care Unit, Shenzhen Institute of Respiratory Disease, Shenzhen Key Laboratory of Respiratory Disease, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, China
| | - Haizhao Xu
- Department of Respiratory Diseases and Critic Care Unit, Shenzhen Institute of Respiratory Disease, Shenzhen Key Laboratory of Respiratory Disease, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, China
- Department of Respiratory, The First Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Gary Guishan Xiao
- State Key Laboratory of Fine Chemicals, Department of Pharmaceutical Sciences, School of Chemical Engineering, Dalian University of Technology, Dalian, China.
| | - Lang Rao
- Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen, 518132, China.
| | - Yanhong Duo
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
31
|
Serafini MA, Sirena DH, da Silveira ABT, Franco-da-Silva M, Aubin MR, Garcez TNA, Araújo A, dos Santos Pereira F, Hoogduijn MJ, da Costa Gonçalves F, Paz AH. Mesenchymal stromal cell-derived membrane particles: A novel cell-free therapy for inflammatory bowel diseases. Int Immunopharmacol 2023; 118:110076. [PMID: 37030123 DOI: 10.1016/j.intimp.2023.110076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 03/02/2023] [Accepted: 03/21/2023] [Indexed: 04/08/2023]
Abstract
Inflammatory bowel diseases (IBD), including ulcerative colitis, are chronic and idiopathic inflammations of the gastrointestinal tract. A disruption of the epithelial barrier and an imbalance between Th1 and Th2 subsets are associated with the onset and progression of these diseases. Mesenchymal stromal cells (MSC) are a promising therapy for IBD. However, cell-tracking studies have shown that intravenously infused MSC localize to the lungs and present short-term survival. To reduce practical complexities arising from living cells, we generated membrane particles (MP) from MSC membranes, which possess some of the immunomodulatory properties of MSC. This study investigated the effect of MSC-derived MP and conditioned media (CM) as cell-free therapies in the dextran sulfate sodium (DSS)-induced colitis model. Acute colitis was induced in C57BL/6 mice by oral administration of 2% DSS in drinking water ad libitum from days 0 to 7. Mice were treated with MP, CM, or living MSC on days 2 and 5. Our findings revealed that MP, CM, and living MSC ameliorated DSS-induced colitis by reducing colonic inflammation, the loss of colonic goblet cells, and intestinal mucosa permeability, preventing apoptosis of damaged colonic cells and balancing Th1 and Th2 activity. Therefore, MSC-derived MP have high therapeutic potential for treating IBD, overcoming the deficiencies of living MSC therapy, and opening novel frontiers in inflammatory diseases medicine.
Collapse
|
32
|
Lee JH, Kim EJ. A Comprehensive Review of the Effects of Extracorporeal Shock Wave Therapy on Stroke Patients: Balance, Pain, Spasticity. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:medicina59050857. [PMID: 37241089 DOI: 10.3390/medicina59050857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 04/24/2023] [Accepted: 04/26/2023] [Indexed: 05/28/2023]
Abstract
Stroke remains a leading cause of disability worldwide, with survivors often experiencing impairments in balance, pain, spasticity, and control that limit their ability to perform daily living activities. Extracorporeal shock wave therapy (ESWT) has emerged as a potential treatment modality to improve these outcomes in stroke patients. This review aims to provide a comprehensive examination of the effects of ESWT on stroke patients, focusing on the theoretical background, balance, pain reduction, muscle spasticity and control, and upper and lower extremities. This study reviewed the use of ESWT in treating balance, pain, and spasticity in stroke patients, focusing on articles published in PubMed between January 2003 and January 2023. Systematic reviews related to stroke were used to provide an overview of stroke, and a total of 33 articles related to balance, pain, and spasticity were selected. ESWT has several shock wave generation methods and application methods, and it has been shown to have positive therapeutic effects on various aspects of rehabilitation for stroke patients, such as improving balance, reducing pain, decreasing muscle spasticity and increasing control, and enhancing functional activities of the upper and lower extremities. The efficacy of ESWT may vary depending on the patient's condition, application method, and treatment area. Therefore, it is important to apply ESWT according to the individual characteristics of each patient in clinical practice to maximize its potential benefits.
Collapse
Affiliation(s)
- Jung-Ho Lee
- Department of Physical Therapy, Kyungdong University, 815, Gyeonhwon-ro, Munmak-eup, Wonju-si 26495, Gang-won-do, Republic of Korea
| | - Eun-Ja Kim
- Department of Physical Therapy, Kyungdong University, 815, Gyeonhwon-ro, Munmak-eup, Wonju-si 26495, Gang-won-do, Republic of Korea
| |
Collapse
|
33
|
Zeng CW. Multipotent Mesenchymal Stem Cell-Based Therapies for Spinal Cord Injury: Current Progress and Future Prospects. BIOLOGY 2023; 12:biology12050653. [PMID: 37237467 DOI: 10.3390/biology12050653] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 04/25/2023] [Accepted: 04/25/2023] [Indexed: 05/28/2023]
Abstract
Spinal cord injury (SCI) represents a significant medical challenge, often resulting in permanent disability and severely impacting the quality of life for affected individuals. Traditional treatment options remain limited, underscoring the need for novel therapeutic approaches. In recent years, multipotent mesenchymal stem cells (MSCs) have emerged as a promising candidate for SCI treatment due to their multifaceted regenerative capabilities. This comprehensive review synthesizes the current understanding of the molecular mechanisms underlying MSC-mediated tissue repair in SCI. Key mechanisms discussed include neuroprotection through the secretion of growth factors and cytokines, promotion of neuronal regeneration via MSC differentiation into neural cell types, angiogenesis through the release of pro-angiogenic factors, immunomodulation by modulating immune cell activity, axonal regeneration driven by neurotrophic factors, and glial scar reduction via modulation of extracellular matrix components. Additionally, the review examines the various clinical applications of MSCs in SCI treatment, such as direct cell transplantation into the injured spinal cord, tissue engineering using biomaterial scaffolds that support MSC survival and integration, and innovative cell-based therapies like MSC-derived exosomes, which possess regenerative and neuroprotective properties. As the field progresses, it is crucial to address the challenges associated with MSC-based therapies, including determining optimal sources, intervention timing, and delivery methods, as well as developing standardized protocols for MSC isolation, expansion, and characterization. Overcoming these challenges will facilitate the translation of preclinical findings into clinical practice, providing new hope and improved treatment options for individuals living with the devastating consequences of SCI.
Collapse
Affiliation(s)
- Chih-Wei Zeng
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
34
|
Li H, Wei J, Zhang Z, Li J, Ma Y, Zhang P, Lin J. Menstrual blood-derived endometrial stem cells alleviate neuroinflammation by modulating M1/M2 polarization in cell and rat Parkinson's disease models. Stem Cell Res Ther 2023; 14:85. [PMID: 37055866 PMCID: PMC10099022 DOI: 10.1186/s13287-023-03330-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 04/05/2023] [Indexed: 04/15/2023] Open
Abstract
BACKGROUND Neuroinflammation is closely related to the development of Parkinson's disease (PD). Because of the extensive sources, non-invasive and periodical collection method, human menstrual blood-derived endometrial stem cells (MenSCs) have been explored as a promising tool for treatment of PD. This study aimed to investigate if MenSCs could inhibit neuroinflammation in PD rats by regulating M1/M2 polarization and to excavate the underlying mechanisms. METHODS MenSCs were co-cultured with 6-OHDA-exposed microglia cell lines. Then the morphology of microglia cells and the level of inflammatory factors were assessed by immunofluorescence and qRT-PCR. After MenSCs were transplanted into the brain of PD rats, animal motor function, the expression of tyrosine hydroxylase, and the level of inflammatory factors in the cerebrospinal fluid (CSF) and serum were detected to evaluate the therapeutic potential of MenSCs. Meanwhile, the expression of M1/M2 phenotype related genes was detected by qRT-PCR. One protein array kit containing 1000 kinds of factors was used to detect the protein components in the conditioned medium of MenSCs. Finally, bioinformatic analysis was performed to analyze the function of factors secreted by MenSCs and the signal pathways involved in. RESULTS MenSCs could suppress 6-OHDA-induced microglia cell activation and significantly decrease inflammation in vitro. After transplantation into the brain of PD rats, MenSCs improved animal motor function, which was indicated by the increased movement distance, ambulatory episodes, exercise time on the rotarod, and less contralateral rotation. Additionally, MenSCs reduced the loss of dopaminergic neurons and down-regulated the level of pro-inflammatory factors in the CSF and serum. Moreover, q-PCR and WB results showed the transplantation of MenSCs significantly down-regulated the expression of M1 phenotype cell markers and meanwhile up-regulated the expression of M2 phenotype cell markers in the brain of PD rats. 176 biological processes including inflammatory response, negative regulation of apoptotic process, and microglial cell activation were enriched by GO-BP analysis. 58 signal pathways including PI3K/Akt and MAPK were enriched by KEGG analysis. CONCLUSIONS In conclusion, our results provide preliminary evidence for the anti-inflammation capacity of MenSCs by regulating M1/M2 polarization. We firstly demonstrated the biological process of factors secreted by MenSCs and the signal pathways involved in using protein array and bioinformatic analysis.
Collapse
Affiliation(s)
- Han Li
- Stem Cells and Biotherapy Engineering Research Center of Henan, National Joint Engineering Laboratory of Stem Cells and Biotherapy, School of Life Science and Technology, Xinxiang Medical University, Xinxiang, 453003, China
| | - Jinghui Wei
- Stem Cells and Biotherapy Engineering Research Center of Henan, National Joint Engineering Laboratory of Stem Cells and Biotherapy, School of Life Science and Technology, Xinxiang Medical University, Xinxiang, 453003, China
| | - Zhigang Zhang
- Department of Neurology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, 45003, China
| | - Junyao Li
- Stem Cells and Biotherapy Engineering Research Center of Henan, National Joint Engineering Laboratory of Stem Cells and Biotherapy, School of Life Science and Technology, Xinxiang Medical University, Xinxiang, 453003, China
| | - Yaokai Ma
- Stem Cells and Biotherapy Engineering Research Center of Henan, National Joint Engineering Laboratory of Stem Cells and Biotherapy, School of Life Science and Technology, Xinxiang Medical University, Xinxiang, 453003, China
| | - Ping Zhang
- Department of Neurology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, 45003, China
| | - Juntang Lin
- Henan Joint International Research Laboratory of Stem Cell Medicine, School of Medical Engineering, Xinxiang Medical University, Xinxiang, 453003, China.
| |
Collapse
|
35
|
Bai T, Duan H, Zhang B, Hao P, Zhao W, Gao Y, Yang Z, Li X. Neuronal differentiation and functional maturation of neurons from neural stem cells induced by bFGF-chitosan controlled release system. Drug Deliv Transl Res 2023:10.1007/s13346-023-01322-x. [PMID: 36943630 DOI: 10.1007/s13346-023-01322-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/28/2023] [Indexed: 03/23/2023]
Abstract
Available methods for differentiating stem cells into neurons require a large number of cytokines and neurotrophic factors, with complex steps and slow processes, and are inefficient to produce functional neurons and form synaptic contacts, which is expensive and impractical in clinical application. Here, we demonstrated a bioactive material, basic fibroblast growth factor (bFGF)-chitosan controlled release system, for facilitating neuronal differentiation from NSCs and the functional maturation of the induced neurons with high efficiency. We illustrated by immunostaining that the neurons derived from NSCs expressed mature immunomarkers of interneurons and excitatory neurons. And we found by patch-clamp that the induced neurons exhibited diverse electrophysiological properties as well as formed functional synapses. In vivo, we implanted bFGF-chitosan into lesion area in traumatic brain injury (TBI) mice and similarly observed abundance of neuroblasts in SVZ and the presence of newborn functional neurons in injury area, which integrated into synaptic networks. Taken together, our efficient and rapid tissue engineering approach may be a potential method for the generation of functional neuronal lineage cells from stem cells and a therapy of brain injury and disease.
Collapse
Affiliation(s)
- Tianyu Bai
- School of Biological Science and Medical Engineering, Beihang University, No. 37 Xueyuan Road, Haidian District, Beijing, 100083, People's Republic of China
| | - Hongmei Duan
- Department of Neurobiology, Fengtai District, Capital Medical University, No. 10 Xitoutiao Strip, Beijing, 100069, People's Republic of China
| | - Boya Zhang
- Department of Neurobiology, Fengtai District, Capital Medical University, No. 10 Xitoutiao Strip, Beijing, 100069, People's Republic of China
| | - Peng Hao
- Department of Neurobiology, Fengtai District, Capital Medical University, No. 10 Xitoutiao Strip, Beijing, 100069, People's Republic of China
| | - Wen Zhao
- Department of Neurobiology, Fengtai District, Capital Medical University, No. 10 Xitoutiao Strip, Beijing, 100069, People's Republic of China
| | - Yudan Gao
- Department of Neurobiology, Fengtai District, Capital Medical University, No. 10 Xitoutiao Strip, Beijing, 100069, People's Republic of China
| | - Zhaoyang Yang
- Department of Neurobiology, Fengtai District, Capital Medical University, No. 10 Xitoutiao Strip, Beijing, 100069, People's Republic of China.
| | - Xiaoguang Li
- School of Biological Science and Medical Engineering, Beihang University, No. 37 Xueyuan Road, Haidian District, Beijing, 100083, People's Republic of China.
- Department of Neurobiology, Fengtai District, Capital Medical University, No. 10 Xitoutiao Strip, Beijing, 100069, People's Republic of China.
| |
Collapse
|
36
|
Raue KD, David BT, Fessler RG. Spinal Cord-Gut-Immune Axis and its Implications Regarding Therapeutic Development for Spinal Cord Injury. J Neurotrauma 2023; 40:793-806. [PMID: 36509451 DOI: 10.1089/neu.2022.0264] [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] [Indexed: 12/14/2022] Open
Abstract
Spinal cord injury (SCI) affects ∼1,300,000 people living in the United States. Most research efforts have been focused on reversing paralysis, as this is arguably the most defining feature of SCI. The damage caused by SCI, however, extends past paralysis and includes other debilitating outcomes including immune dysfunction and gut dysbiosis. Recent efforts are now investigating the pathophysiology of and developing therapies for these more distal manifestations of SCI. One exciting avenue is the spinal cord-gut-immune axis, which proposes that gut dysbiosis amplifies lesion inflammation and impairs SCI recovery. This review will highlight the most recent findings regarding gut and immune dysfunction following SCI, and discuss how the central nervous system (CNS), gut, and immune system all coalesce to form a bidirectional axis that can impact SCI recovery. Finally, important considerations regarding how the spinal cord-gut-immune axis fits within the larger framework of therapeutic development (i.e., probiotics, fecal transplants, dietary modifications) will be discussed, emphasizing the lack of interdepartmental investigation and the missed opportunity to maximize therapeutic benefit in SCI.
Collapse
Affiliation(s)
- Kristen D Raue
- Department of Neurosurgery, Rush University Medical Center, Chicago, Illinois, USA
| | - Brian T David
- Department of Neurosurgery, Rush University Medical Center, Chicago, Illinois, USA
| | - Richard G Fessler
- Department of Neurosurgery, Rush University Medical Center, Chicago, Illinois, USA
| |
Collapse
|
37
|
Xiang W, Cao H, Tao H, Jin L, Luo Y, Tao F, Jiang T. Applications of chitosan-based biomaterials: From preparation to spinal cord injury neuroprosthetic treatment. Int J Biol Macromol 2023; 230:123447. [PMID: 36708903 DOI: 10.1016/j.ijbiomac.2023.123447] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 12/04/2022] [Accepted: 01/24/2023] [Indexed: 01/27/2023]
Abstract
Spinal cord injury (SCI)-related disabilities are a serious problem in the modern society. Further, the treatment of SCI is highly challenging and is urgently required in clinical practice. Research on nerve tissue engineering is an emerging approach for improving the treatment outcomes of SCI. Chitosan (CS) is a cationic polysaccharide derived from natural biomaterials. Chitosan has been found to exhibit excellent biological properties, such as nontoxicity, biocompatibility, biodegradation, and antibacterial activity. Recently, chitosan-based biomaterials have attracted significant attention for SCI repair in nerve tissue engineering applications. These studies revealed that chitosan-based biomaterials have various functions and mechanisms to promote SCI repair, such as promoting neural cell growth, guiding nerve tissue regeneration, delivering nerve growth factors, and as a vector for gene therapy. Chitosan-based biomaterials have proven to have excellent potential for the treatment of SCI. This review aims to introduce the recent advances in chitosan-based biomaterials for SCI treatment and to highlight the prospects for further application.
Collapse
Affiliation(s)
- Wei Xiang
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan University, Wuhan 430060, China
| | - Hui Cao
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan University, Wuhan 430060, China
| | - Hai Tao
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan University, Wuhan 430060, China
| | - Lin Jin
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan University, Wuhan 430060, China
| | - Yue Luo
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan University, Wuhan 430060, China
| | - Fenghua Tao
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan University, Wuhan 430060, China.
| | - Ting Jiang
- Department of Neurological Rehabilitation, Zhongnan Hospital of Wuhan University, Wuhan 430071, China.
| |
Collapse
|
38
|
Mou C, Wang X, Li W, Li Z, Liu N, Xu Y. Efficacy of mesenchymal stromal cells intraspinal transplantation for patients with different degrees of spinal cord injury: A systematic review and meta-analysis. Cytotherapy 2023; 25:530-536. [PMID: 36805381 DOI: 10.1016/j.jcyt.2023.01.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 01/06/2023] [Accepted: 01/24/2023] [Indexed: 02/22/2023]
Abstract
BACKGROUND AIMS Several studies have reported that mesenchymal stromal cells (MSCs) may improve neurological functions in patients with spinal cord injury (SCI). In this study, we conducted a systematic review and meta-analysis to summarize the effects of MSC treatment on different degrees of severity of SCI. METHODS Systematic searching of studies reporting outcomes of MSCs on specific injury severities of patients with SCI was performed in The National Library of Medicine (MEDLINE), Embase and Cochrane for published articles up to the 6 July 2022. Two investigators independently reviewed the included studies and extracted the relevant data. The standardized mean differences of American Spinal Injury Association (ASIA) motor score, ASIA light touch scores, ASIA pinprick scores and the Barthel index between baseline and follow-ups were pooled. RESULTS A total of eight studies were included. A large majority focused on patients with ASIA grade A classification. The pooled mean differences of ASIA motor scores, ASIA light touch scores, ASIA pinprick scores and the Barthel index were -2.78 (95% confidence interval [CI] -5.12 to -0.43, P = 0.02), -18.26 (95% CI -26.09 to -10.43, P < 0.01), -17.08 (95% CI -24.10 to -10.07, P < 0.01) and -4.37 (95% CI -10.96 to 2.22, P = 0.19), respectively. CONCLUSIONS MSC transplantation was a significantly effective therapy for patients with SCI with ASIA grade A. In the future, further studies are warranted to confirm the potential beneficial effects of MSC therapy.
Collapse
Affiliation(s)
- Chunlin Mou
- Technology Department, Everunion Biotechnology Co. Ltd., Tianjin, China
| | - Xiujuan Wang
- Technology Department, Everunion Biotechnology Co. Ltd., Tianjin, China
| | - Wei Li
- Technology Department, Everunion Biotechnology Co. Ltd., Tianjin, China
| | - Zhengnan Li
- Technology Department, Everunion Biotechnology Co. Ltd., Tianjin, China
| | - Nian Liu
- Technology Department, Everunion Biotechnology Co. Ltd., Tianjin, China
| | - Yongsheng Xu
- Technology Department, Everunion Biotechnology Co. Ltd., Tianjin, China.
| |
Collapse
|
39
|
Zamanifard M, Khorasani MT, Daliri M. Hybrid electrospun polyhydroxybutyrate/gelatin/laminin/polyaniline scaffold for nerve tissue engineering application: Preparation, characterization, and in vitro assay. Int J Biol Macromol 2023; 235:123738. [PMID: 36805505 DOI: 10.1016/j.ijbiomac.2023.123738] [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: 11/07/2022] [Revised: 12/26/2022] [Accepted: 02/14/2023] [Indexed: 02/20/2023]
Abstract
Despite the widespread central nervous system injuries, treatment of these disorders is still an issue of concern due to the complexities. Natural recovery in these patients is rarely observed, which calls for developing new methods that address these problems. In this study, natural polymers of polyhydroxybutyrate (PHB) and gelatin were electrospun into scaffolds and cross-linked. In order to modify the PHB-based scaffold for nerve tissue engineering, the scaffold surface was modified by exposure to the ammonium gas plasma under controlled conditions, and the laminin as a promoter for neural cells was coated on the sample surface. Then, polyaniline nanoparticles were inkjet-printed on a sample surface as parallel lines to induce the differentiation of stem cells into neural cells. Infrared spectroscopy, absorption of PBS, AFM, degradation rate, contact angle, electron microscopy and optical microscopy, thermal and mechanical behavior, and analysis of the viability of L929 cells were investigated for the scaffolds. The results showed gelatin decreased the contact angle from 106.2° to 38° and increased the residual weight after PBS incubation from 82 % to 38 %. The moduli of the scaffold increased from 8.78 MPa for pure PHB to 28.74 for the modified scaffold. In addition, performed methods increased cell viability from 69 % for PHB to 89 % for modified scaffold and also had a favorable effect on cell adhesion. Investigation of culturing P19 stem cells demonstrated that they successfully differentiated into neural cells. Results show that the scaffolds prepared in this study were promising for nerve tissue engineering.
Collapse
Affiliation(s)
- Mohammad Zamanifard
- Department of Biomaterials, Faculty of Biomedical Engineering, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Mohammad Taghi Khorasani
- Biomaterials Department of Iran Polymer and Petrochemical Institute, P.O. BOX 14965/159, Tehran, Iran.
| | - Morteza Daliri
- Department of Animal and Marine Biotechnology, National Institute of Genetic Engineering and Biotechnology, Shahrak-e Pajoohesh Km 15, Tehran-Karaj Highway, Tehran, Iran
| |
Collapse
|
40
|
Medvediev VV, Oleksenko NP, Pichkur LD, Verbovska SA, Savosko SI, Draguntsova NG, Lontkovskyi YA, Vaslovych VV, Tsymbalyuk VI. Implantation Effect of a Fibrin Matrix Associated with Mesenchymal Wharton’s Jelly Stromal Cells on the Course of an Experimental Spinal Cord Injury. CYTOL GENET+ 2023. [DOI: 10.3103/s0095452723010073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/11/2023]
|
41
|
Suzuki H, Imajo Y, Funaba M, Ikeda H, Nishida N, Sakai T. Current Concepts of Biomaterial Scaffolds and Regenerative Therapy for Spinal Cord Injury. Int J Mol Sci 2023; 24:ijms24032528. [PMID: 36768846 PMCID: PMC9917245 DOI: 10.3390/ijms24032528] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/05/2023] [Accepted: 01/11/2023] [Indexed: 02/03/2023] Open
Abstract
Spinal cord injury (SCI) is a catastrophic condition associated with significant neurological deficit and social and financial burdens. It is currently being managed symptomatically, with no real therapeutic strategies available. In recent years, a number of innovative regenerative strategies have emerged and have been continuously investigated in preclinical research and clinical trials. In the near future, several more are expected to come down the translational pipeline. Among ongoing and completed trials are those reporting the use of biomaterial scaffolds. The advancements in biomaterial technology, combined with stem cell therapy or other regenerative therapy, can now accelerate the progress of promising novel therapeutic strategies from bench to bedside. Various types of approaches to regeneration therapy for SCI have been combined with the use of supportive biomaterial scaffolds as a drug and cell delivery system to facilitate favorable cell-material interactions and the supportive effect of neuroprotection. In this review, we summarize some of the most recent insights of preclinical and clinical studies using biomaterial scaffolds in regenerative therapy for SCI and summarized the biomaterial strategies for treatment with simplified results data. One hundred and sixty-eight articles were selected in the present review, in which we focused on biomaterial scaffolds. We conducted our search of articles using PubMed and Medline, a medical database. We used a combination of "Spinal cord injury" and ["Biomaterial", or "Scaffold"] as search terms and searched articles published up until 30 April 2022. Successful future therapies will require these biomaterial scaffolds and other synergistic approaches to address the persistent barriers to regeneration, including glial scarring, the loss of a structural framework, and biocompatibility. This database could serve as a benchmark to progress in future clinical trials for SCI using biomaterial scaffolds.
Collapse
|
42
|
Petinati N, Shipounova I, Sats N, Dorofeeva A, Sadovskaya A, Kapranov N, Tkachuk Y, Bondarenko A, Muravskaya M, Kotsky M, Kaplanskaya I, Vasilieva T, Drize N. Multipotent Mesenchymal Stromal Cells from Porcine Bone Marrow, Implanted under the Kidney Capsule, form an Ectopic Focus Containing Bone, Hematopoietic Stromal Microenvironment, and Muscles. Cells 2023; 12:268. [PMID: 36672203 PMCID: PMC9857022 DOI: 10.3390/cells12020268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/02/2023] [Accepted: 01/05/2023] [Indexed: 01/11/2023] Open
Abstract
Multipotent mesenchymal stromal cells (MSCs) are an object of intense investigation due to their therapeutic potential. MSCs have been well studied in vitro, while their fate after implantation in vivo has been poorly analyzed. We studied the properties of MSCs from the bone marrow (BM-MSC) before and after implantation under the renal capsule using a mini pig model. Autologous BM-MSCs were implanted under the kidney capsule. After 2.5 months, ectopic foci containing bones, foci of ectopic hematopoiesis, bone marrow stromal cells and muscle cells formed. Small pieces of the implant were cultivated as a whole. The cells that migrated out from these implants were cultured, cloned, analyzed and were proven to meet the most of criteria for MSCs, therefore, they are designated as MSCs from the implant-IM-MSCs. The IM-MSC population demonstrated high proliferative potential, similar to BM-MSCs. IM-MSC clones did not respond to adipogenic differentiation inductors: 33% of clones did not differentiate, and 67% differentiated toward an osteogenic lineage. The BM-MSCs revealed functional heterogeneity after implantation under the renal capsule. The BM-MSC population consists of mesenchymal precursor cells of various degrees of differentiation, including stem cells. These newly discovered properties of mini pig BM-MSCs reveal new possibilities in terms of their manipulation.
Collapse
Affiliation(s)
- Nataliya Petinati
- Laboratory for Physiology of Hematopoiesis, National Medical Research Center for Hematology, Ministry of Health of the Russian Federation, 125167 Moscow, Russia
| | - Irina Shipounova
- Laboratory for Physiology of Hematopoiesis, National Medical Research Center for Hematology, Ministry of Health of the Russian Federation, 125167 Moscow, Russia
| | - Natalia Sats
- Laboratory for Physiology of Hematopoiesis, National Medical Research Center for Hematology, Ministry of Health of the Russian Federation, 125167 Moscow, Russia
| | - Alena Dorofeeva
- Laboratory for Physiology of Hematopoiesis, National Medical Research Center for Hematology, Ministry of Health of the Russian Federation, 125167 Moscow, Russia
| | - Alexandra Sadovskaya
- Laboratory for Physiology of Hematopoiesis, National Medical Research Center for Hematology, Ministry of Health of the Russian Federation, 125167 Moscow, Russia
- Department of Immunology, Faculty of Biology, Federal State Budget Educational Institution of Higher Education M.V. Lomonosov Moscow State University, 119234 Moscow, Russia
| | - Nikolay Kapranov
- Laboratory for Physiology of Hematopoiesis, National Medical Research Center for Hematology, Ministry of Health of the Russian Federation, 125167 Moscow, Russia
| | - Yulia Tkachuk
- Bioclinic for Working with Animals, Federal State Budgetary Scientific Institution Izmerov Research Institute of Occupational Health, 105275 Moscow, Russia
| | - Anatoliy Bondarenko
- Bioclinic for Working with Animals, Federal State Budgetary Scientific Institution Izmerov Research Institute of Occupational Health, 105275 Moscow, Russia
| | - Margarita Muravskaya
- Bioclinic for Working with Animals, Federal State Budgetary Scientific Institution Izmerov Research Institute of Occupational Health, 105275 Moscow, Russia
| | - Michail Kotsky
- Bioclinic for Working with Animals, Federal State Budgetary Scientific Institution Izmerov Research Institute of Occupational Health, 105275 Moscow, Russia
| | - Irina Kaplanskaya
- MNIOI Them. P.A. Herzen—Branch of the Federal State Budgetary Institution “NMITs Radiology” of the Ministry of Health of Russia, Department of Pathomorphology, 125284 Moscow, Russia
| | - Tamara Vasilieva
- Department of Cell Biology, Faculty of Biology, Federal State Budget Educational Institution of Higher Education M.V. Lomonosov Moscow State University, 119234 Moscow, Russia
| | - Nina Drize
- Laboratory for Physiology of Hematopoiesis, National Medical Research Center for Hematology, Ministry of Health of the Russian Federation, 125167 Moscow, Russia
| |
Collapse
|
43
|
Rybachuk O, Savytska N, Pinet É, Yaminsky Y, Medvediev V. Heterogeneous pHPMA hydrogel promotes neuronal differentiation of bone marrow derived stromal cells in vitroand in vivo. Biomed Mater 2023; 18. [PMID: 36542861 DOI: 10.1088/1748-605x/acadc3] [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: 06/27/2022] [Accepted: 12/21/2022] [Indexed: 12/24/2022]
Abstract
Synthetic hydrogels composed of polymer pore frames are commonly used in medicine, from pharmacologically targeted drug delivery to the creation of bioengineering constructions used in implantation surgery. Among various possible materials, the most common are poly-[N(2-hydroxypropyl)methacrylamide] (pHPMA) derivatives. One of the pHPMA derivatives is biocompatible hydrogel, NeuroGel. Upon contact with nervous tissue, the NeuroGel's structure can support the chemical and physiological conditions of the tissue necessary for the growth of native cells. Owing to the different pore diameters in the hydrogel, not only macromolecules, but also cells can migrate. This study evaluated the differentiation of bone marrow stromal cells (BMSCs) into neurons, as well as the effectiveness of using this biofabricated system in spinal cord injuryin vivo. The hydrogel was populated with BMSCs by injection or rehydration. After cultivation, these fragments (hydrogel + BMSCs) were implanted into the injured rat spinal cord. Fragments were immunostained before implantation and seven months after implantation. During cultivation with the hydrogel, both variants (injection/rehydration) of the BMSCs culture retained their viability and demonstrated a significant number of Ki-67-positive cells, indicating the preservation of their proliferative activity. In hydrogel fragments, BMSCs also maintained their viability during the period of cocultivation and were Ki-67-positive, but in significantly fewer numbers than in the cell culture. In addition, in fragments of hydrogel with grafted BMSCs, both by the injection or rehydration versions, we observed a significant number up to 57%-63.5% of NeuN-positive cells. These results suggest that the heterogeneous pHPMA hydrogel promotes neuronal differentiation of bone marrow-derived stromal cells. Furthermore, these data demonstrate the possible use of NeuroGel implants with grafted BMSCs for implantation into damaged areas of the spinal cord, with subsequent nerve fiber germination, nerve cell regeneration, and damaged segment restoration.
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, NAMS of Ukraine, Kyiv, Ukraine
| | - Natalia Savytska
- Bogomoletz Institute of Physiology NAS of Ukraine, Kyiv, Ukraine.,German Center for Neurodegenerative Diseases, Tübingen, Germany
| | | | - Yurii Yaminsky
- State Institution 'Romodanov Neurosurgery Institute, NAMS of Ukraine', Kyiv, Ukraine
| | - Volodymyr Medvediev
- Bogomoletz Institute of Physiology NAS of Ukraine, Kyiv, Ukraine.,Bogomolets National Medical University, Kyiv, Ukraine
| |
Collapse
|
44
|
Huang T, Wu J, Mu J, Gao J. Advanced Therapies for Traumatic Central Nervous System Injury: Delivery Strategy Reinforced Efficient Microglial Manipulation. Mol Pharm 2023; 20:41-56. [PMID: 36469398 DOI: 10.1021/acs.molpharmaceut.2c00605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Traumatic central nervous system (CNS) injuries, including spinal cord injury and traumatic brain injury, are challenging enemies of human health. Microglia, the main component of the innate immune system in CNS, can be activated postinjury and are key participants in the pathological procedure and development of CNS trauma. Activated microglia can be typically classified into pro-inflammatory (M1) and anti-inflammatory (M2) phenotypes. Reducing M1 polarization while promoting M2 polarization is thought to be promising for CNS injury treatment. However, obstacles such as the low permeability of the blood-brain barrier and short retention time in circulation limit the therapeutic outcomes of administrated drugs, and rational delivery strategies are necessary for efficient microglial regulation. To this end, proper administration methods and delivery systems like nano/microcarriers and scaffolds are investigated to augment the therapeutic effects of drugs, while some of these delivery systems have self-efficacies in microglial manipulation. Besides, systems based on cell and cell-derived exosomes also show impressive effects, and some underlying targeting mechanisms of these delivery systems have been discovered. In this review, we introduce the roles of microglia play in traumatic CNS injuries, discuss the potential targets for the polarization regulation of microglial phenotype, and summarize recent studies and clinical trials about delivery strategies on enhancing the effect of microglial regulation and therapeutic outcome, as well as targeting mechanisms post CNS trauma.
Collapse
Affiliation(s)
- Tianchen Huang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.,Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jiahe Wu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.,Department of Clinical Pharmacology, Key Laboratory of Clinical Cancer, Pharmacology and Toxicology Research of Zhejiang Province, Affiliated, Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Jiafu Mu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jianqing Gao
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.,Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.,Jinhua Institute of Zhejiang University, Jinhua 321002, China
| |
Collapse
|
45
|
Zhao YY, Wu ZJ, Zhu LJ, Niu TX, Liu B, Li J. Emerging roles of miRNAs in neuropathic pain: From new findings to novel mechanisms. Front Mol Neurosci 2023; 16:1110975. [PMID: 36873108 PMCID: PMC9981676 DOI: 10.3389/fnmol.2023.1110975] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 01/30/2023] [Indexed: 02/19/2023] Open
Abstract
Neuropathic pain, which results from damage to the somatosensory nervous system, is a global clinical condition that affects many people. Neuropathic pain imposes significant economic and public health burdens and is often difficult to manage because the underlying mechanisms remain unclear. However, mounting evidence indicates a role for neurogenic inflammation and neuroinflammation in pain pattern development. There is increasing evidence that the activation of neurogenic inflammation and neuroinflammation in the nervous system contribute to neuropathic pain. Altered miRNA expression profiles might be involved in the pathogenesis of both inflammatory and neuropathic pain by regulating neuroinflammation, nerve regeneration, and abnormal ion channel expression. However, the lack of knowledge about miRNA target genes prevents a full understanding of the biological functions of miRNAs. At the same time, an extensive study on exosomal miRNA, a newly discovered role, has advanced our understanding of the pathophysiology of neuropathic pain in recent years. This section provides a comprehensive overview of the current understanding of miRNA research and discusses the potential mechanisms of miRNAs in neuropathic pain.
Collapse
Affiliation(s)
- Yu-Ying Zhao
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China.,Tianjin Research Institute of Anesthesiology, Tianjin, China
| | - Zi-Jun Wu
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China.,Tianjin Research Institute of Anesthesiology, Tianjin, China
| | - Li-Juan Zhu
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China.,Tianjin Research Institute of Anesthesiology, Tianjin, China
| | - Tong-Xiang Niu
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China.,Tianjin Research Institute of Anesthesiology, Tianjin, China
| | - Bin Liu
- Department of Critical Care Medicine, General Hospital of Tianjin Medical University, Tianjin, China.,Center for Critical Care Medicine, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, China
| | - Jing Li
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China.,Tianjin Research Institute of Anesthesiology, Tianjin, China
| |
Collapse
|
46
|
Xia Y, Yang R, Wang H, Hou Y, Li Y, Zhu J, Xu F, Fu C. Biomaterials delivery strategies to repair spinal cord injury by modulating macrophage phenotypes. J Tissue Eng 2022; 13:20417314221143059. [PMID: 36600997 PMCID: PMC9806413 DOI: 10.1177/20417314221143059] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 11/17/2022] [Indexed: 12/28/2022] Open
Abstract
Spinal cord injury (SCI) causes tremendous harm to a patient's physical, mental, and financial health. Moreover, recovery of SCI is affected by many factors, inflammation is one of the most important as it engulfs necrotic tissue and cells during the early stages of injury. However, excessive inflammation is not conducive to damage repair. Macrophages are classified into either blood-derived macrophages or resident microglia based on their origin, their effects on SCI being two-sided. Microglia first activate and recruit blood-derived macrophages at the site of injury-blood-borne macrophages being divided into pro-inflammatory M1 phenotypes and anti-inflammatory M2 phenotypes. Among them, M1 macrophages secrete inflammatory factors such as interleukin-β (IL-β), tumor necrosis factor-α (TNF-α), IL-6, and interferon-γ (IFN-γ) at the injury site, which aggravates SCIs. M2 macrophages secrete IL-4, IL-10, IL-13, and neurotrophic factors to inhibit the inflammatory response and inhibit neuronal apoptosis. Consequently, modulating phenotypic differentiation of macrophages appears to be a meaningful therapeutic target for the treatment of SCI. Biomaterials are widely used in regenerative medicine and tissue engineering due to their targeting and bio-histocompatibility. In this review, we describe the effects of biomaterials applied to modulate macrophage phenotypes on SCI recovery and provide an outlook.
Collapse
Affiliation(s)
- Yuanliang Xia
- Department of Spine Surgery, The First
Hospital of Jilin University, Changchun, PR China
| | - Ruohan Yang
- Cancer Center, The First Hospital of
Jilin University, Changchun, PR China
| | - Hengyi Wang
- Department of Spine Surgery, The First
Hospital of Jilin University, Changchun, PR China
| | - Yulin Hou
- Depattment of Cardiology, Guangyuan
Central Hospital, Guangyuan, PR China
| | - Yuehong Li
- Department of Spine Surgery, The First
Hospital of Jilin University, Changchun, PR China
| | - Jianshu Zhu
- Department of Spine Surgery, The First
Hospital of Jilin University, Changchun, PR China
| | - Feng Xu
- Department of Spine Surgery, The First
Hospital of Jilin University, Changchun, PR China
| | - Changfeng Fu
- Department of Spine Surgery, The First
Hospital of Jilin University, Changchun, PR China,Changfeng Fu, Department of Spine Surgery,
The First Hospital of Jilin University, 1 Xinmin Street, Changchun 130021, PR
China.
| |
Collapse
|
47
|
Xu J, Xi K, Tang J, Wang J, Tang Y, Wu L, Xu Y, Xu Z, Chen L, Cui W, Gu Y. Engineered Living Oriented Electrospun Fibers Regulate Stem Cell Para-Secretion and Differentiation to Promote Spinal Cord Repair. Adv Healthc Mater 2022; 12:e2202785. [PMID: 36541060 DOI: 10.1002/adhm.202202785] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 12/16/2022] [Indexed: 12/24/2022]
Abstract
Living biomaterials directly couple with live cells to synthesize functional molecules and respond to dynamic environments, allowing the design, construction and application of next generation composite materials. Improving the coordination and communication between artificial materials and living cells is essential. In this study, collagen self-assembly and micro-sol electrospinning techniques are used to prepare oriented living fiber bundles that can increase the transplantation rate of stem cells in the early stages of inflammation, indirectly enhancing the dynamic regulation of stem cells during inflammation. Additionally, brain-derived neurotrophic factor (BDNF) contained in the fiber can improve the differentiation of bone marrow mesenchymal stem cells (BMSCs) into neurons once the inflammatory storm subsides. The living oriented fiber bundles fully simulate the 3D structure of the central nervous system, activate integrin β1, promote the growth and adhesion of stem cells in the acute stage of inflammation, upregulate anti-inflammatory genes by more than twofold via BMSCs in response to inflammation, and stably release BDNF for up to 4 weeks post-inflammation storm subsidence. Finally, the BDNF induces the differentiation of BMSCs to neurons by enhancing the expression of neural-related genes, which enables the recovery of neurological functions in the later stages of spinal cord injury.
Collapse
Affiliation(s)
- Jingzhi Xu
- Department of Orthopedic Surgery, Orthopedic Institute, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou, Jiangsu, 215006, P. R. China
| | - Kun Xi
- Department of Orthopedic Surgery, Orthopedic Institute, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou, Jiangsu, 215006, P. R. China
| | - Jincheng Tang
- Department of Orthopedic Surgery, Orthopedic Institute, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou, Jiangsu, 215006, P. R. China
| | - Juan Wang
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, P. R. China
| | - Yunkai Tang
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, P. R. China
| | - Liang Wu
- Department of Orthopedic Surgery, Orthopedic Institute, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou, Jiangsu, 215006, P. R. China
| | - Yichang Xu
- Department of Orthopedic Surgery, Orthopedic Institute, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou, Jiangsu, 215006, P. R. China
| | - Zonghan Xu
- Department of Orthopedic Surgery, Orthopedic Institute, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou, Jiangsu, 215006, P. R. China
| | - Liang Chen
- Department of Orthopedic Surgery, Orthopedic Institute, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou, Jiangsu, 215006, P. R. China
| | - Wenguo Cui
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, P. R. China
| | - Yong Gu
- Department of Orthopedic Surgery, Orthopedic Institute, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou, Jiangsu, 215006, P. R. China
| |
Collapse
|
48
|
Li M, Chen H, Zhu M. Mesenchymal stem cells for regenerative medicine in central nervous system. Front Neurosci 2022; 16:1068114. [PMID: 36583105 PMCID: PMC9793714 DOI: 10.3389/fnins.2022.1068114] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 11/28/2022] [Indexed: 12/15/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are multipotent stem cells, whose paracrine and immunomodulatory potential has made them a promising candidate for central nervous system (CNS) regeneration. Numerous studies have demonstrated that MSCs can promote immunomodulation, anti-apoptosis, and axon re-extension, which restore functional neural circuits. The therapeutic effects of MSCs have consequently been evaluated for application in various CNS diseases including spinal cord injury, cerebral ischemia, and neurodegenerative disease. In this review, we will focus on the research works published in the field of mechanisms and therapeutic effects of MSCs in CNS regeneration.
Collapse
Affiliation(s)
- Man Li
- Department of Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hong Chen
- Department of Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mingxin Zhu
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China,*Correspondence: Mingxin Zhu,
| |
Collapse
|
49
|
Wu Y, Tang Z, Zhang J, Wang Y, Liu S. Restoration of spinal cord injury: From endogenous repairing process to cellular therapy. Front Cell Neurosci 2022; 16:1077441. [PMID: 36523818 PMCID: PMC9744968 DOI: 10.3389/fncel.2022.1077441] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Accepted: 11/08/2022] [Indexed: 09/26/2023] Open
Abstract
Spinal cord injury (SCI) disrupts neurological pathways and impacts sensory, motor, and autonomic nerve function. There is no effective treatment for SCI currently. Numerous endogenous cells, including astrocytes, macrophages/microglia, and oligodendrocyte, are involved in the histological healing process following SCI. By interfering with cells during the SCI repair process, some advancements in the therapy of SCI have been realized. Nevertheless, the endogenous cell types engaged in SCI repair and the current difficulties these cells confront in the therapy of SCI are poorly defined, and the mechanisms underlying them are little understood. In order to better understand SCI and create new therapeutic strategies and enhance the clinical translation of SCI repair, we have comprehensively listed the endogenous cells involved in SCI repair and summarized the six most common mechanisms involved in SCI repair, including limiting the inflammatory response, protecting the spared spinal cord, enhancing myelination, facilitating neovascularization, producing neurotrophic factors, and differentiating into neural/colloidal cell lines.
Collapse
Affiliation(s)
| | | | | | | | - Shengwen Liu
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
50
|
Fan R, Wang L, Botchway BOA, Zhang Y, Liu X. Protective role of ethyl pyruvate in spinal cord injury by inhibiting the high mobility group box-1/toll-like receptor4/nuclear factor-kappa B signaling pathway. Front Mol Neurosci 2022; 15:1013033. [PMID: 36187352 PMCID: PMC9524569 DOI: 10.3389/fnmol.2022.1013033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Accepted: 08/29/2022] [Indexed: 11/13/2022] Open
Abstract
Spinal cord injury (SCI) is a high incident rate of central nervous system disease that usually causes paralysis below the injured level. The occurrence of chronic inflammation with the axonal regeneration difficulties are the underlying barriers for the recovery of SCI patients. Current studies have paid attention to controlling the instigative and developmental process of neuro-inflammation. Ethyl pyruvate, as a derivative of pyruvate, has strong anti-inflammatory and neuroprotective functions. Herein, we reviewed the recent studies of ethyl pyruvate and high mobility group box-1 (HMGB1). We think HMGB1 that is one of the main nuclear protein mediators to cause an inflammatory response. This protein induces astrocytic activation, and promotes glial scar formation. Interestingly, ethyl pyruvate has potent inhibitory effects on HMGB1 protein, as it inhibits chronic inflammatory response by modulating the HMGB1/TLR4/NF-κB signaling pathway. This paper discusses the potential mechanism of ethyl pyruvate in inhibiting chronic inflammation after SCI. Ethyl pyruvate can be a prospective therapeutic agent for SCI.
Collapse
Affiliation(s)
- Ruihua Fan
- Department of Histology and Embryology, Medical College, Shaoxing University, Shaoxing, China
- School of Life Sciences, Shaoxing University, Shaoxing, China
| | - Lvxia Wang
- Department of Histology and Embryology, Medical College, Shaoxing University, Shaoxing, China
- School of Life Sciences, Shaoxing University, Shaoxing, China
| | | | - Yong Zhang
- School of Life Sciences, Shaoxing University, Shaoxing, China
| | - Xuehong Liu
- Department of Histology and Embryology, Medical College, Shaoxing University, Shaoxing, China
- School of Life Sciences, Shaoxing University, Shaoxing, China
- *Correspondence: Xuehong Liu, ; orcid.org/0000-0003-4325-6762
| |
Collapse
|