1
|
Song X, Li R, Chu X, Li Q, Li R, Li Q, Tong KY, Gu X, Ming D. Multilevel analysis of the central-peripheral-target organ pathway: contributing to recovery after peripheral nerve injury. Neural Regen Res 2025; 20:2807-2822. [PMID: 39435615 PMCID: PMC11826472 DOI: 10.4103/nrr.nrr-d-24-00641] [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: 06/11/2024] [Revised: 08/06/2024] [Accepted: 09/23/2024] [Indexed: 10/23/2024] Open
Abstract
Peripheral nerve injury is a common neurological condition that often leads to severe functional limitations and disabilities. Research on the pathogenesis of peripheral nerve injury has focused on pathological changes at individual injury sites, neglecting multilevel pathological analysis of the overall nervous system and target organs. This has led to restrictions on current therapeutic approaches. In this paper, we first summarize the potential mechanisms of peripheral nerve injury from a holistic perspective, covering the central nervous system, peripheral nervous system, and target organs. After peripheral nerve injury, the cortical plasticity of the brain is altered due to damage to and regeneration of peripheral nerves; changes such as neuronal apoptosis and axonal demyelination occur in the spinal cord. The nerve will undergo axonal regeneration, activation of Schwann cells, inflammatory response, and vascular system regeneration at the injury site. Corresponding damage to target organs can occur, including skeletal muscle atrophy and sensory receptor disruption. We then provide a brief review of the research advances in therapeutic approaches to peripheral nerve injury. The main current treatments are conducted passively and include physical factor rehabilitation, pharmacological treatments, cell-based therapies, and physical exercise. However, most treatments only partially address the problem and cannot complete the systematic recovery of the entire central nervous system-peripheral nervous system-target organ pathway. Therefore, we should further explore multilevel treatment options that produce effective, long-lasting results, perhaps requiring a combination of passive (traditional) and active (novel) treatment methods to stimulate rehabilitation at the central-peripheral-target organ levels to achieve better functional recovery.
Collapse
Affiliation(s)
- Xizi Song
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
- Haihe Laboratory of Brain-Machine Interface and Human-Machine Fusion, Tianjin, China
| | - Ruixin Li
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
- Haihe Laboratory of Brain-Machine Interface and Human-Machine Fusion, Tianjin, China
| | - Xiaolei Chu
- Department of Rehabilitation, Tianjin University Tianjin Hospital, Tianjin, China
| | - Qi Li
- Department of Rehabilitation, Tianjin University Tianjin Hospital, Tianjin, China
| | - Ruihua Li
- Department of Hand Microsurgery, Tianjin University Tianjin Hospital, Tianjin, China
| | - Qingwen Li
- School of Exercise and Health, Tianjin University of Sport, Tianjin, China
| | - Kai-Yu Tong
- Department of Biomedical Engineering, the Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Xiaosong Gu
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
- Haihe Laboratory of Brain-Machine Interface and Human-Machine Fusion, Tianjin, China
| | - Dong Ming
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
- Haihe Laboratory of Brain-Machine Interface and Human-Machine Fusion, Tianjin, China
| |
Collapse
|
2
|
Liu N, Ma Y, Gong W, Shao X, Shi T, Li L, Wu W, Chen X, Shi Y, Zhang P, Lin J, Wang C, Fang D, Yang L, Wang P, Gao W, He Y, An X, Du R, Chen Y, Liu B, Qin J, Chen D, Cai P, Jiang Q, Guo B. Osteocyte-derived extracellular vesicles mediate the bone-to-cartilage crosstalk and promote osteoarthritis progression. Nat Commun 2025; 16:4746. [PMID: 40399261 PMCID: PMC12095588 DOI: 10.1038/s41467-025-59861-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 05/02/2025] [Indexed: 05/23/2025] Open
Abstract
Osteoarthritis is a common degenerative joint disease, in which mechanical overloading disrupts subchondral bone remodeling before cartilage degeneration and the osteocytes in the subchondral bone are mainly responsible for mechanosensing. However, their functional role in the early osteoarthritis is still unclear. Here we show that mechanical stress induces osteocytes in subchondral bone to secrete extracellular vesicles that accelerate cartilage metabolic dysregulation in patients with both sexes and male mice. The miR-23b-3p in extracellular vesicles promotes cartilage catabolism and inhibits anabolism by targeting OTUD4, disrupting mitophagy in chondrocytes. Inhibiting miR-23b-3p in osteocytes or chondrocytes reduces cartilage degeneration and osteoarthritis progression in male mice. Together, our findings highlight that osteocyte-derived extracellular vesicles mediate communication with chondrocytes and suggest miR-23b-3p as a potential therapeutic target for osteoarthritis.
Collapse
Affiliation(s)
- Na Liu
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Nanjing, PR China
| | - Yuze Ma
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Nanjing, PR China
| | - Wang Gong
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Nanjing, PR China
| | - Xiaoyan Shao
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Nanjing, PR China
| | - Tianshu Shi
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Nanjing, PR China
| | - Lan Li
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Nanjing, PR China
| | - Wenshu Wu
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Nanjing, PR China
| | - Xiang Chen
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Nanjing, PR China
| | - Yong Shi
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Nanjing, PR China
| | - Pan Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Nanjing, PR China
| | - Jiaquan Lin
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Nanjing, PR China
| | - Chengzhi Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Nanjing, PR China
| | - Depeng Fang
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Nanjing, PR China
| | - Lin Yang
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Nanjing, PR China
| | - Pu Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Nanjing, PR China
| | - Wentian Gao
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Nanjing, PR China
| | - Yi He
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Nanjing, PR China
| | - Xueying An
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Nanjing, PR China
| | - Rui Du
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Nanjing, PR China
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Xuzhou Medical University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
| | - Ying Chen
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Nanjing, PR China
| | - Bin Liu
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Nanjing, PR China
| | - Jianghui Qin
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Nanjing, PR China
| | - Dongyang Chen
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Nanjing, PR China
| | - Pingqiang Cai
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China.
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Nanjing, PR China.
| | - Qing Jiang
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China.
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Nanjing, PR China.
| | - Baosheng Guo
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China.
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Nanjing, PR China.
| |
Collapse
|
3
|
Xiang B, Zhang S, Zhao IS, Gan X, Zhang Y. Microenvironmental Modulation for Therapeutic Efficacy of Extracellular Vesicles. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2503027. [PMID: 40145773 PMCID: PMC12079496 DOI: 10.1002/advs.202503027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 03/04/2025] [Indexed: 03/28/2025]
Abstract
Extracellular vesicles (EVs) hold significant promise for the prevention and treatment of various diseases. However, the translation of EV-based therapies into clinical practice faces considerable challenges, particularly in terms of production yield and therapeutic efficacy. Recent studies have emphasized the heterogeneity of EVs and the influence of parental cell microenvironmental signals on their biogenesis, cargo composition, and therapeutic outcomes. This review offers a comprehensive overview of strategies to optimize the therapeutic efficacy of EVs through physical, biochemical, and mechanical modulation. Additionally, it explores how microenvironmental signals affect EV cargoes and the mechanisms by which these signals can improve therapeutic efficacy. The review also addresses current challenges and potential solutions to accelerate the clinical translation of EV therapies. Ultimately, it highlights the potential of microenvironmental modulation in unlocking the full therapeutic capacity of EVs, providing key insights into their production and clinical use for treating various diseases.
Collapse
Affiliation(s)
- Bilu Xiang
- School of DentistryShenzhen University Medical SchoolShenzhen518055China
- Institute of Oral ScienceShenzhen UniversityShenzhen518055China
| | - Shiying Zhang
- School of DentistryShenzhen University Medical SchoolShenzhen518055China
| | - Irene Shuping Zhao
- School of DentistryShenzhen University Medical SchoolShenzhen518055China
- Institute of Oral ScienceShenzhen UniversityShenzhen518055China
| | - Xueqi Gan
- State Key Laboratory of Oral DiseaseNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengdu610041China
| | - Yang Zhang
- School of DentistryShenzhen University Medical SchoolShenzhen518055China
- Institute of Oral ScienceShenzhen UniversityShenzhen518055China
- School of Biomedical EngineeringShenzhen University Medical SchoolShenzhen518055China
| |
Collapse
|
4
|
Khan A, Oliveira J, Lee YS, Guest JD, Silvera R, Pressman Y, Pearse DD, Nettina AE, Goldschmidt-Clermont PJ, Al-Ali H, Williams I, Levi AD, Dietrich WD. Human Schwann Cell-Derived Extracellular Vesicle Isolation, Bioactivity Assessment, and Omics Characterization. Int J Nanomedicine 2025; 20:4123-4144. [PMID: 40201152 PMCID: PMC11977562 DOI: 10.2147/ijn.s500159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 02/05/2025] [Indexed: 04/10/2025] Open
Abstract
Purpose Schwann cell-derived extracellular vesicles (SCEVs) have demonstrated favorable effects in spinal cord, peripheral nerve, and brain injuries. Herein, a scalable, standardized, and efficient isolation methodology of SCEVs obtaining a high yield with a consistent composition as measured by proteomic, lipidomic, and miRNA analysis of their content is described for future clinical use. Methods Human Schwann cells were obtained ethically from nine donors and cultured in a defined growth medium optimized for proliferation. At confluency, the culture was replenished with an isolation medium for 48 hours, then collected and centrifuged sequentially at low and ultra-high speeds to collect purified EVs. The EVs were characterized with mass spectrometry to identify and quantify proteins, lipidomic analysis to assess lipid composition, and next-generation sequencing to confirm miRNA profiles. Each batch of EVs was assessed to ensure their therapeutic potential in promoting neurite outgrowth and cell survival. Results High yields of SCEVs were consistently obtained with similar comprehensive molecular profiles across samples, indicating the reproducibility and reliability of the isolation method. Bioactivity to increase neurite process growth was confirmed in vitro. The predominance of triacylglycerol and phosphatidylcholine suggested its role in cellular membrane dynamics essential for axon regeneration and inflammation mitigation. Of the 2517 identified proteins, 136 were closely related to nervous system repair and regeneration. A total of 732 miRNAs were cataloged, with the top 30 miRNAs potentially contributing to axon growth, neuroprotection, myelination, angiogenesis, the attenuation of neuroinflammation, and key signaling pathways such as VEGFA-VEGFR2 and PI3K-Akt signaling, which are crucial for nervous system repair. Conclusion The study establishes a robust framework for SCEV isolation and their comprehensive characterization, which is consistent with their therapeutic potential in neurological applications. This work provides a valuable proteomic, lipidomic, and miRNA dataset to inform future advancements in applying SCEV to the experimental treatment of neurological injuries and diseases.
Collapse
Affiliation(s)
- Aisha Khan
- Interdisciplinary Stem Cell Institute, Miller School of Medicine, University of Miami, Miami, FL, USA
- The Miami Project to Cure Paralysis, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Julia Oliveira
- The Miami Project to Cure Paralysis, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Yee-Shuan Lee
- Interdisciplinary Stem Cell Institute, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - James D Guest
- The Miami Project to Cure Paralysis, Miller School of Medicine, University of Miami, Miami, FL, USA
- Neurological Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Risset Silvera
- The Miami Project to Cure Paralysis, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Yelena Pressman
- The Miami Project to Cure Paralysis, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Damien D Pearse
- The Miami Project to Cure Paralysis, Miller School of Medicine, University of Miami, Miami, FL, USA
- Neurological Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Adriana E Nettina
- Interdisciplinary Stem Cell Institute, Miller School of Medicine, University of Miami, Miami, FL, USA
- The Miami Project to Cure Paralysis, Miller School of Medicine, University of Miami, Miami, FL, USA
| | | | - Hassan Al-Ali
- The Miami Project to Cure Paralysis, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Indigo Williams
- The Miami Project to Cure Paralysis, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Allan D Levi
- The Miami Project to Cure Paralysis, Miller School of Medicine, University of Miami, Miami, FL, USA
- Neurological Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - W Dalton Dietrich
- The Miami Project to Cure Paralysis, Miller School of Medicine, University of Miami, Miami, FL, USA
- Neurological Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
| |
Collapse
|
5
|
Liang X, Wang Z, Wang S, Ruan F, Zhang Y, Shao D, Liu X, Chen F, Shi X. Magnetic mesoporous silica nanoparticles loaded with peptides for the targeted repair of cavernous nerve injury underlying erectile dysfunction. Biomaterials 2025; 314:122811. [PMID: 39265373 DOI: 10.1016/j.biomaterials.2024.122811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 08/22/2024] [Accepted: 09/02/2024] [Indexed: 09/14/2024]
Abstract
Erectile dysfunction (ED) is a common male sexual disorder characterized by repeated or persistent difficulty in achieving or maintaining an erection. It can arise from various factors, with cavernous nerve injury (CNI) from radical prostatectomy being a predominant cause of iatrogenic ED, posing significant clinical concerns. The complexity of cavernous tissue damage in CNI-induced ED (CNIED) often results in poor efficacy and resistance to conventional vascular ED treatments. To address CNI-induced ED, this study developed a system of magnetic mesoporous silica nanoparticles (MSNs) loaded with peptides for targeted treatment. Core-shell Fe3O4-coated MSNs were used as drug carriers and loaded with RADA16-I/RAD-RGI peptides (PD) to create a neurotrophic microenvironment to treat peripheral nerve defects. Furthermore, the neuro-targeting peptide HLNILSTLWKYR (PT) was grafted onto MSNs. The in vivo therapeutic effect was evaluated using a rat bilateral cavernous nerve injury (BCNI) model. The results showed that the neuro-targeted Fe3O4@SiO2-PT-PD nanoparticles significantly promoted regeneration of the cavernous nerve and restored erectile function. This promising strategy offers significant clinical potential for treating CNI-induced ED. Nanomedicine technology has the potential to not only improve treatment outcomes but also reduce side effects in healthy cells, paving the way for more accurate targeted repair of cavernous nerve damage.
Collapse
Affiliation(s)
- Xiaojie Liang
- National Engineering Research Centre for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, China; School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510640, China
| | - Zhu Wang
- Neonatology Department, Guangdong Women and Children Hospital, Guangzhou, 510010, China
| | - Shuting Wang
- National Engineering Research Centre for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, China; School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510640, China
| | - Feixia Ruan
- National Engineering Research Centre for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, China; School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou, Guangdong, 511442, China
| | - Yidan Zhang
- National Engineering Research Centre for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, China; School of Medicine, South China University of Technology, Guangzhou, Guangdong, 510006, China
| | - Dan Shao
- National Engineering Research Centre for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, China; School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou, Guangdong, 511442, China; School of Medicine, South China University of Technology, Guangzhou, Guangdong, 510006, China
| | - Xuemin Liu
- Department of Gynecology and Obstetrics, The Third Affiliated Hospital of Guangzhou Medical-University, Guangzhou, Guangdong, 510510, China.
| | - Fangman Chen
- National Engineering Research Centre for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, China; School of Medicine, South China University of Technology, Guangzhou, Guangdong, 510006, China.
| | - Xuetao Shi
- National Engineering Research Centre for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, China; School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510640, China; Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou, 510006, China.
| |
Collapse
|
6
|
Zhao X, Yao M, Wang Y, Feng C, Yang Y, Tian L, Bao C, Li X, Zhu X, Zhang X. Neuroregulation during Bone Formation and Regeneration: Mechanisms and Strategies. ACS APPLIED MATERIALS & INTERFACES 2025; 17:7223-7250. [PMID: 39869030 DOI: 10.1021/acsami.4c16786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
The skeleton is highly innervated by numerous nerve fibers. These nerve fibers, in addition to transmitting information within the bone and mediating bone sensations, play a crucial role in regulating bone tissue formation and regeneration. Traditional bone tissue engineering (BTE) often fails to achieve satisfactory outcomes when dealing with large-scale bone defects, which is frequently related to the lack of effective reconstruction of the neurovascular network. In recent years, increasing research has revealed the critical role of nerves in bone metabolism. Nerve fibers regulate bone cells through neurotransmitters, neuropeptides, and peripheral glial cells. Furthermore, nerves also coordinate with the vascular and immune systems to jointly construct a microenvironment favorable for bone regeneration. As a signaling driver of bone formation, neuroregulation spans the entire process of bone physiological activities from the embryonic formation to postmaturity remodeling and repair. However, there is currently a lack of comprehensive summaries of these regulatory mechanisms. Therefore, this review sketches out the function of nerves during bone formation and regeneration. Then, we elaborate on the mechanisms of neurovascular coupling and neuromodulation of bone immunity. Finally, we discuss several novel strategies for neuro-bone tissue engineering (NBTE) based on neuroregulation of bone, focusing on the coordinated regeneration of nerve and bone tissue.
Collapse
Affiliation(s)
- Xiangrong Zhao
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Med-X Center for Materials, Sichuan University, Chengdu 610041, Sichuan, China
| | - Meilin Yao
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Med-X Center for Materials, Sichuan University, Chengdu 610041, Sichuan, China
| | - Yuyi Wang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Cong Feng
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Yuhan Yang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Med-X Center for Materials, Sichuan University, Chengdu 610041, Sichuan, China
| | - Luoqiang Tian
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Chongyun Bao
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Med-X Center for Materials, Sichuan University, Chengdu 610041, Sichuan, China
| | - Xiangfeng Li
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Xiangdong Zhu
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| |
Collapse
|
7
|
Zhang L, Xie J, Dai W, Lu B, Yi S. Schwann cells in regeneration and cancer. Front Pharmacol 2025; 16:1506552. [PMID: 39981185 PMCID: PMC11840318 DOI: 10.3389/fphar.2025.1506552] [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: 10/05/2024] [Accepted: 01/09/2025] [Indexed: 02/22/2025] Open
Abstract
Schwann cells are specific peripheral glial cells with remarkable plasticity following peripheral nerve injury. Injury responses stimulate c-Jun activation in Schwann cells, drive epithelial-mesenchymal transition and cellular phenotypic changes, and induce the generation of reprogrammed repair Schwann cells to orchestrate peripheral nerve regeneration process. Schwann cells and/or Schwann cell-derived molecules are commonly used as supporting cells and/or neurotrophic factors to construct Schwann cell-based tissue-engineered nerve grafts for repairing severe peripheral nerve injury with long defects. Transplantation of Schwann cells and/or Schwann cell-derived molecules also serves as a helpful approach for the treatment of other injured tissues, such as the spinal cord, skin, digit tip, and bone. Schwann cells are not only associated with tissue regeneration but also involved in tumorigenesis and tumor progression. Schwann cells are the major cellular component of neurofibromatosis type 1 and the sole cell type in neurofibromatosis type 2 and schwannomatosis. In addition, Schwann cells also function as an important player in the tumor microenvironment and aid in the growth and invasiveness of many other solid cancers. In the present review, we outline the physiological and pathological activities of Schwann cells and discuss the functional roles of Schwann cells in homeostasis, regeneration, and cancer.
Collapse
Affiliation(s)
- Lan Zhang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu, China
| | - Jiale Xie
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu, China
| | - Wenyu Dai
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu, China
| | - Bing Lu
- Department of Clinical Biobank and Institute of Oncology, Affiliated Hospital of Nantong University, Nantong University, Nantong, Jiangsu, China
| | - Sheng Yi
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu, China
| |
Collapse
|
8
|
Su Y, Liu T, Zhao M, Wu D, Wang Y, Wu X. Isoviolanthin promotes Schwann cells activity in peripheral nerve regeneration via Fhl3-mediated epithelial-mesenchymal transition-like process: An in vitro study. Heliyon 2025; 11:e41087. [PMID: 39811297 PMCID: PMC11731196 DOI: 10.1016/j.heliyon.2024.e41087] [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: 05/12/2024] [Revised: 12/07/2024] [Accepted: 12/08/2024] [Indexed: 01/16/2025] Open
Abstract
Schwann cells, as crucial regenerative cells, possess the ability to facilitate axon growth following peripheral nerve injury. However, the regeneration efficiency dominated by Schwann cells is impaired by factors such as the severity of peripheral nervous injury, aging, and metabolic disease. Cause the limitations of clinical treatments, it is necessary to urgently search for new substances that could reinforce the functionality of Schwann cells and promote nerve regeneration. We represented the first evidence that isoviolanthin possesses the capability to enhance Schwann cell proliferation and migration. Then, transcriptome sequencing was employed to examine the Differential Expressed Genes (DEGs), resulting in the identification of 193 DEGs. Following this, the expression levels of the top 5 up-regulated genes were confirmed through RT-qPCR, with Fhl3 demonstrating the most significant up-regulation. Schwann cells were transduced with virus particles made in HEK-293T/17 cells by transfection with lentivirus packaging plasmids containing Fhl3. A notable enhancement in Schwann cell proliferation and migration was observed following transduction. Furthermore, the Fhl3-up group exhibited a significant upregulation of Vimentin expression compared to the control group. These results suggested that isoviolanthin plays a positive role in enhancing Schwann cells' activity via increasing Fhl3 expression, and the mechanism may be related to the EMT(epithelial-mesenchymal transition)-like process.
Collapse
Affiliation(s)
- Yajuan Su
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Tiantian Liu
- Department of Orthopedic Rehabilitation, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, 200137, China
| | - Minjun Zhao
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Dandan Wu
- Department of Orthopedic Rehabilitation, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, 200137, China
| | - Yuehua Wang
- Department of Neurosurgery, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, 200137, China
| | - Xubo Wu
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Department of Orthopedic Rehabilitation, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, 200137, China
| |
Collapse
|
9
|
Koyama S, Etkins J, Jun J, Miller M, So GC, Gisch DL, Eadon MT. Utilization of Cannabidiol in Post-Organ-Transplant Care. Int J Mol Sci 2025; 26:699. [PMID: 39859413 PMCID: PMC11765766 DOI: 10.3390/ijms26020699] [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: 12/16/2024] [Revised: 01/06/2025] [Accepted: 01/07/2025] [Indexed: 01/27/2025] Open
Abstract
Cannabidiol (CBD) is one of the major phytochemical constituents of cannabis, Cannabis sativa, widely recognized for its therapeutic potential. While cannabis has been utilized for medicinal purposes since ancient times, its psychoactive and addictive properties led to its prohibition in 1937, with only the medical use being reauthorized in 1998. Unlike tetrahydrocannabinol (THC), CBD lacks psychoactive and addictive properties, yet the name that suggests its association with cannabis has significantly contributed to its public visibility. CBD exhibits diverse pharmacological properties, most notably anti-inflammatory effects. Additionally, it interacts with key drug-metabolizing enzyme families, including cytochrome P450 (CYP) and uridine 5'-diphospho-glucuronosyltransferase (UGT), which mediate phase I and phase II metabolism, respectively. By binding to these enzymes, CBD can inhibit the metabolism of co-administered drugs, which can potentially enhance their toxicity or therapeutic effects. Mild to moderate adverse events associated with CBD use have been reported. Advances in chemical formulation techniques have recently enabled strategies to minimize these effects. This review provides an overview of CBD, covering its historical background, recent clinical trials, adverse event profiles, and interactions with molecular targets such as receptors, channels, and enzymes. We particularly emphasize the mechanisms underlying its anti-inflammatory effects and interaction with drugs relevant to organ transplantation. Finally, we explore recent progress in the chemical formulation of CBD in order to enhance its bioavailability, which will enable decreasing the dose to use and increase its safety and efficacy.
Collapse
Affiliation(s)
- Sachiko Koyama
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (J.E.); (J.J.); (D.L.G.); (M.T.E.)
| | - Jumar Etkins
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (J.E.); (J.J.); (D.L.G.); (M.T.E.)
| | - Joshua Jun
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (J.E.); (J.J.); (D.L.G.); (M.T.E.)
| | - Matthew Miller
- College of Human Ecology, Cornell University, Ithaca, NY 14850, USA;
| | - Gerald C. So
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (J.E.); (J.J.); (D.L.G.); (M.T.E.)
| | - Debora L. Gisch
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (J.E.); (J.J.); (D.L.G.); (M.T.E.)
| | - Michael T. Eadon
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (J.E.); (J.J.); (D.L.G.); (M.T.E.)
| |
Collapse
|
10
|
Zhai X, Wang Y. Physical modulation and peripheral nerve regeneration: a literature review. CELL REGENERATION (LONDON, ENGLAND) 2024; 13:32. [PMID: 39710804 DOI: 10.1186/s13619-024-00215-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 11/28/2024] [Accepted: 12/08/2024] [Indexed: 12/24/2024]
Abstract
Peripheral nerve injury (PNI) usually causes severe motor, sensory and autonomic dysfunction. In addition to direct surgical repair, rehabilitation exercises, and traditional physical stimuli, for example, electrical stimulation, have been applied in promoting the clinical recovery of PNI for a long time but showed low efficiency. Recently, significant progress has been made in new physical modulation to promote peripheral nerve regeneration. We hereby review current progress on the mechanism of peripheral nerve regeneration after injury and summarize the new findings and evidence for the application of physical modulation, including electrical stimulation, light, ultrasound, magnetic stimulation, and mechanical stretching in experimental studies and the clinical treatment of patients with PNI.
Collapse
Affiliation(s)
- Xiangwen Zhai
- College of Rehabilitation Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, China
| | - Yuzhong Wang
- Department of Neurology, Affiliated Hospital of Jining Medical University, 89 Guhuai Road, Jining, 272029, Shandong Province, China.
- Medical Research Centre, Affiliated Hospital of Jining Medical University, Jining, Shandong Province, China.
| |
Collapse
|
11
|
Li B, Chen H, Hang R. Osseointegration-Related Exosomes for Surface Functionalization of Titanium Implants. Biomater Res 2024; 28:0124. [PMID: 39711824 PMCID: PMC11661649 DOI: 10.34133/bmr.0124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 11/05/2024] [Accepted: 11/27/2024] [Indexed: 12/24/2024] Open
Abstract
Despite that the clinical application of titanium-based implants has achieved great success, patients' own diseases and/or unhealthy lifestyle habits often lead to implant failure. Many studies have been carried out to modify titanium implants to promote osseointegration and implant success. Recent studies showed that exosomes, proactively secreted extracellular vesicles by mammalian cells, could selectively target and modulate the functions of recipient cells such as macrophages, nerve cells, endothelial cells, and bone marrow mesenchymal stem cells that are closely involved in implant osseointegration. Accordingly, using exosomes to functionalize titanium implants has been deemed as a novel and effective way to improve their osseointegration ability. Herein, recent advances pertaining to surface functionalization of titanium implants with exosomes are analyzed and discussed, with focus on the role of exosomes in regulating the functions of osseointegration-related cells, and their immobilization strategies as well as resultant impact on osseointegration ability.
Collapse
Affiliation(s)
- Boqiong Li
- Department of Materials Science and Engineering,
Jinzhong University, Jinzhong 030619, China
| | - Huanming Chen
- Shanxi Key Laboratory of Biomedical Metal Materials, College of Materials Science and Engineering,
Taiyuan University of Technology, Taiyuan 030024, China
| | - Ruiqiang Hang
- Shanxi Key Laboratory of Biomedical Metal Materials, College of Materials Science and Engineering,
Taiyuan University of Technology, Taiyuan 030024, China
| |
Collapse
|
12
|
Koyama S, Weber EL, Heinbockel T. Possible Combinatorial Utilization of Phytochemicals and Extracellular Vesicles for Wound Healing and Regeneration. Int J Mol Sci 2024; 25:10353. [PMID: 39408681 PMCID: PMC11476926 DOI: 10.3390/ijms251910353] [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: 07/15/2024] [Revised: 09/16/2024] [Accepted: 09/17/2024] [Indexed: 10/20/2024] Open
Abstract
Organ and tissue damage can result from injury and disease. How to facilitate regeneration from damage has been a topic for centuries, and still, we are trying to find agents to use for treatments. Two groups of biological substances are known to facilitate wound healing. Phytochemicals with bioactive properties form one group. Many phytochemicals have anti-inflammatory effects and enhance wound healing. Recent studies have described their effects at the gene and protein expression levels, highlighting the receptors and signaling pathways involved. The extremely large number of phytochemicals and the multiple types of receptors they activate suggest a broad range of applicability for their clinical use. The hydrophobic nature of many phytochemicals and the difficulty with chemical stabilization have been a problem. Recent developments in biotechnology and nanotechnology methods are enabling researchers to overcome these problems. The other group of biological substances is extracellular vesicles (EVs), which are now known to have important biological functions, including the improvement of wound healing. The proteins and nanoparticles contained in mammalian EVs as well as the specificity of the targets of microRNAs included in the EVs are becoming clear. Plant-derived EVs have been found to contain phytochemicals. The overlap in the wound-healing capabilities of both phytochemicals and EVs and the differences in their nature suggest the possibility of a combinatorial use of the two groups, which may enhance their effects.
Collapse
Affiliation(s)
- Sachiko Koyama
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Erin L. Weber
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
| | - Thomas Heinbockel
- Department of Anatomy, College of Medicine, Howard University, Washington, DC 20059, USA
| |
Collapse
|
13
|
Putthanbut N, Lee JY, Borlongan CV. Extracellular vesicle therapy in neurological disorders. J Biomed Sci 2024; 31:85. [PMID: 39183263 PMCID: PMC11346291 DOI: 10.1186/s12929-024-01075-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: 06/17/2024] [Accepted: 08/16/2024] [Indexed: 08/27/2024] Open
Abstract
Extracellular vesicles (EVs) are vital for cell-to-cell communication, transferring proteins, lipids, and nucleic acids in various physiological and pathological processes. They play crucial roles in immune modulation and tissue regeneration but are also involved in pathogenic conditions like inflammation and degenerative disorders. EVs have heterogeneous populations and cargo, with numerous subpopulations currently under investigations. EV therapy shows promise in stimulating tissue repair and serving as a drug delivery vehicle, offering advantages over cell therapy, such as ease of engineering and minimal risk of tumorigenesis. However, challenges remain, including inconsistent nomenclature, complex characterization, and underdeveloped large-scale production protocols. This review highlights the recent advances and significance of EVs heterogeneity, emphasizing the need for a better understanding of their roles in disease pathologies to develop tailored EV therapies for clinical applications in neurological disorders.
Collapse
Affiliation(s)
- Napasiri Putthanbut
- Department of Neurosurgery, Center of Aging and Brain Repair, University of South Florida, Tampa, USA
- Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Salaya, Thailand
| | - Jea Young Lee
- Department of Neurosurgery, Center of Aging and Brain Repair, University of South Florida, Tampa, USA
| | - Cesario V Borlongan
- Department of Neurosurgery, Center of Aging and Brain Repair, University of South Florida, Tampa, USA.
| |
Collapse
|
14
|
Krsek A, Jagodic A, Baticic L. Nanomedicine in Neuroprotection, Neuroregeneration, and Blood-Brain Barrier Modulation: A Narrative Review. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1384. [PMID: 39336425 PMCID: PMC11433843 DOI: 10.3390/medicina60091384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 08/19/2024] [Accepted: 08/22/2024] [Indexed: 09/30/2024]
Abstract
Nanomedicine is a newer, promising approach to promote neuroprotection, neuroregeneration, and modulation of the blood-brain barrier. This review includes the integration of various nanomaterials in neurological disorders. In addition, gelatin-based hydrogels, which have huge potential due to biocompatibility, maintenance of porosity, and enhanced neural process outgrowth, are reviewed. Chemical modification of these hydrogels, especially with guanidine moieties, has shown improved neuron viability and underscores tailored biomaterial design in neural applications. This review further discusses strategies to modulate the blood-brain barrier-a factor critically associated with the effective delivery of drugs to the central nervous system. These advances bring supportive solutions to the solving of neurological conditions and innovative therapies for their treatment. Nanomedicine, as applied to neuroscience, presents a significant leap forward in new therapeutic strategies that might help raise the treatment and management of neurological disorders to much better levels. Our aim was to summarize the current state-of-knowledge in this field.
Collapse
Affiliation(s)
- Antea Krsek
- Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia;
| | - Ana Jagodic
- Department of Family Medicine, Community Health Center Krapina, 49000 Krapina, Croatia;
| | - Lara Baticic
- Department of Medical Chemistry, Biochemistry and Clinical Chemistry, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
| |
Collapse
|
15
|
Yu L, Bennett CJ, Lin CH, Yan S, Yang J. Scaffold design considerations for peripheral nerve regeneration. J Neural Eng 2024; 21:10.1088/1741-2552/ad628d. [PMID: 38996412 PMCID: PMC11883895 DOI: 10.1088/1741-2552/ad628d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 07/12/2024] [Indexed: 07/14/2024]
Abstract
Peripheral nerve injury (PNI) represents a serious clinical and public health problem due to its high incurrence and poor spontaneous recovery. Compared to autograft, which is still the best current practice for long-gap peripheral nerve defects in clinics, the use of polymer-based biodegradable nerve guidance conduits (NGCs) has been gaining momentum as an alternative to guide the repair of severe PNI without the need of secondary surgery and donor nerve tissue. However, simple hollow cylindrical tubes can barely outperform autograft in terms of the regenerative efficiency especially in critical sized PNI. With the rapid development of tissue engineering technology and materials science, various functionalized NGCs have emerged to enhance nerve regeneration over the past decades. From the aspect of scaffold design considerations, with a specific focus on biodegradable polymers, this review aims to summarize the recent advances in NGCs by addressing the onerous demands of biomaterial selections, structural designs, and manufacturing techniques that contributes to the biocompatibility, degradation rate, mechanical properties, drug encapsulation and release efficiency, immunomodulation, angiogenesis, and the overall nerve regeneration potential of NGCs. In addition, several commercially available NGCs along with their regulation pathways and clinical applications are compared and discussed. Lastly, we discuss the current challenges and future directions attempting to provide inspiration for the future design of ideal NGCs that can completely cure long-gap peripheral nerve defects.
Collapse
Affiliation(s)
- Le Yu
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, United States of America
| | - Carly Jane Bennett
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, United States of America
| | - Chung-Hsun Lin
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, United States of America
| | - Su Yan
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, United States of America
| | - Jian Yang
- Biomedical Engineering Program, Westlake University, Hangzhou, Zhejiang 310030, People’s Republic of China
- Research Center for Industries of the Future, Westlake University, Hangzhou, Zhejiang 310030, People’s Republic of China
| |
Collapse
|
16
|
Yi Y, Zhang S, Dai J, Zheng H, Peng X, Cheng L, Chen H, Hu Y. MiR-23b-3p Improves Brain Damage after Status Epilepticus by Reducing the Formation of Pathological High-Frequency Oscillations via Inhibition of cx43 in Rat Hippocampus. ACS Chem Neurosci 2024; 15:2633-2642. [PMID: 38967483 DOI: 10.1021/acschemneuro.4c00112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/06/2024] Open
Abstract
In order to investigate the effectiveness and safety of miR-23b-3p in anti-seizure activity and to elucidate the regulatory relationship between miR-23b-3p and Cx43 in the nervous system, we have established a lithium chloride-pilocarpine (PILO) status epilepticus (SE) model. Rats were randomly divided into the following groups: seizure control (PILO), valproate sodium (VPA+PILO), recombinant miR-23b-3p overexpression (miR+PILO), miR-23b-3p sponges (Sponges+PILO), and scramble sequence negative control (Scramble+PILO) (n = 6/group). After experiments, we got the following results. In the acute phase, the time required for rats to reach stage IV after PILO injection was significantly longer in VPA+PILO and miR+PILO. In the chronic phase after SE, the frequency of spontaneous recurrent seizures (SRSs) in VPA+PILO and miR+PILO was significantly reduced. At 10 min before seizure cessation, the average energy expression of fast ripples (FRs) in VPA+PILO and miR+PILO was significantly lower than in PILO. After 28 days of seizure, Cx43 expression in PILO was significantly increased, and Beclin1expression in all groups was significantly increased. After 28 days of SE,the number of synapses in the CA1 region of the hippocampus was significantly higher in the VPA+PILO and miR+PILO groups compared to that in the PILO group. After 28 days of SE ,hippocampal necrotic cells in the CA3 region were significantly lower in the VPA+PILO and miR+PILO groups compared to those in the PILO group. There were no significant differences in biochemical indicators among the experimental group rats 28 days after SE compared to the seizure control group. Based on the previous facts, we can reach the conclusion that MiR-23b-3p targets and blocks the expression of hippocampal Cx43 which can reduce the formation of pathological FRs, thereby alleviating the severity of seizures, improving seizure-induced brain damage.
Collapse
Affiliation(s)
- Yanjun Yi
- Department of Neurology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing 400014, China
- Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Shimin Zhang
- Department of Neurology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing 400014, China
- Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Jiali Dai
- Department of Neurology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing 400014, China
- Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Hao Zheng
- Department of Neurology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing 400014, China
- Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Xiaoling Peng
- Guangdong Provincial Key Laboratory of Interdisciplinary Research and Application for Data Science, BNU-HKBU United International College, Zhuhai 519087, China
| | - Li Cheng
- Department of Neurology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing 400014, China
- Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Hengsheng Chen
- Department of Neurology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing 400014, China
- Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Yue Hu
- Department of Neurology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing 400014, China
| |
Collapse
|
17
|
Wang Z, Dong S, Zhou W. Pancreatic stellate cells: Key players in pancreatic health and diseases (Review). Mol Med Rep 2024; 30:109. [PMID: 38695254 PMCID: PMC11082724 DOI: 10.3892/mmr.2024.13233] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 04/09/2024] [Indexed: 05/12/2024] Open
Abstract
As a pluripotent cell, activated pancreatic stellate cells (PSCs) can differentiate into various pancreatic parenchymal cells and participate in the secretion of extracellular matrix and the repair of pancreatic damage. Additionally, PSCs characteristics allow them to contribute to pancreatic inflammation and carcinogenesis. Moreover, a detailed study of the pathogenesis of activated PSCs in pancreatic disease can offer promise for the development of innovative therapeutic strategies and improved patient prognoses. Therefore, the present study review aimed to examine the involvement of activated PSCs in pancreatic diseases and elucidate the underlying mechanisms to provide a viable therapeutic strategy for the management of pancreas‑related diseases.
Collapse
Affiliation(s)
- Zhengfeng Wang
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Shi Dong
- The Second School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Wence Zhou
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, Gansu 730000, P.R. China
| |
Collapse
|
18
|
Tang Y, Wu J, Liu C, Gan L, Chen H, Sun YL, Liu J, Tao YX, Zhu T, Chen C. Schwann cell-derived extracellular vesicles promote memory impairment associated with chronic neuropathic pain. J Neuroinflammation 2024; 21:99. [PMID: 38632655 PMCID: PMC11025217 DOI: 10.1186/s12974-024-03081-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 03/30/2024] [Indexed: 04/19/2024] Open
Abstract
BACKGROUND The pathogenesis of memory impairment, a common complication of chronic neuropathic pain (CNP), has not been fully elucidated. Schwann cell (SC)-derived extracellular vesicles (EVs) contribute to remote organ injury. Here, we showed that SC-EVs may mediate pathological communication between SCs and hippocampal neurons in the context of CNP. METHODS We used an adeno-associated virus harboring the SC-specific promoter Mpz and expressing the CD63-GFP gene to track SC-EVs transport. microRNA (miRNA) expression profiles of EVs and gain-of-function and loss-of-function regulatory experiments revealed that miR-142-5p was the main cargo of SC-EVs. Next, luciferase reporter gene and phenotyping experiments confirmed the direct targets of miR-142-5p. RESULTS The contents and granule sizes of plasma EVs were significantly greater in rats with chronic sciatic nerve constriction injury (CCI)than in sham rats. Administration of the EV biogenesis inhibitor GW4869 ameliorated memory impairment in CCI rats and reversed CCI-associated dendritic spine damage. Notably, during CCI stress, SC-EVs could be transferred into the brain through the circulation and accumulate in the hippocampal CA1-CA3 regions. miR-142-5p was the main cargo wrapped in SC-EVs and mediated the development of CCI-associated memory impairment. Furthermore, α-actinin-4 (ACTN4), ELAV-like protein 4 (ELAVL4) and ubiquitin-specific peptidase 9 X-linked (USP9X) were demonstrated to be important downstream target genes for miR-142-5p-mediated regulation of dendritic spine damage in hippocampal neurons from CCI rats. CONCLUSION Together, these findings suggest that SCs-EVs and/or their cargo miR-142-5p may be potential therapeutic targets for memory impairment associated with CNP.
Collapse
Affiliation(s)
- Yidan Tang
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Jiahui Wu
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Changliang Liu
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Lu Gan
- Research Laboratory of Emergency Medicine, West China Hospital, Emergency Medicine and National Clinical Research Center for Geriatrics, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Hai Chen
- Department of Respiratory and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Ya-Lan Sun
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Jin Liu
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yuan-Xiang Tao
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, 07103, USA.
| | - Tao Zhu
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China.
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China.
| | - Chan Chen
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China.
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China.
| |
Collapse
|
19
|
Li YY, Ji SF, Fu XB, Jiang YF, Sun XY. Biomaterial-based mechanical regulation facilitates scarless wound healing with functional skin appendage regeneration. Mil Med Res 2024; 11:13. [PMID: 38369464 PMCID: PMC10874556 DOI: 10.1186/s40779-024-00519-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 01/30/2024] [Indexed: 02/20/2024] Open
Abstract
Scar formation resulting from burns or severe trauma can significantly compromise the structural integrity of skin and lead to permanent loss of skin appendages, ultimately impairing its normal physiological function. Accumulating evidence underscores the potential of targeted modulation of mechanical cues to enhance skin regeneration, promoting scarless repair by influencing the extracellular microenvironment and driving the phenotypic transitions. The field of skin repair and skin appendage regeneration has witnessed remarkable advancements in the utilization of biomaterials with distinct physical properties. However, a comprehensive understanding of the underlying mechanisms remains somewhat elusive, limiting the broader application of these innovations. In this review, we present two promising biomaterial-based mechanical approaches aimed at bolstering the regenerative capacity of compromised skin. The first approach involves leveraging biomaterials with specific biophysical properties to create an optimal scarless environment that supports cellular activities essential for regeneration. The second approach centers on harnessing mechanical forces exerted by biomaterials to enhance cellular plasticity, facilitating efficient cellular reprogramming and, consequently, promoting the regeneration of skin appendages. In summary, the manipulation of mechanical cues using biomaterial-based strategies holds significant promise as a supplementary approach for achieving scarless wound healing, coupled with the restoration of multiple skin appendage functions.
Collapse
Affiliation(s)
- Ying-Ying Li
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Department, Chinese PLA General Hospital and PLA Medical College; PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing, 100048, China
| | - Shuai-Fei Ji
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Department, Chinese PLA General Hospital and PLA Medical College; PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing, 100048, China
| | - Xiao-Bing Fu
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Department, Chinese PLA General Hospital and PLA Medical College; PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing, 100048, China.
| | - Yu-Feng Jiang
- Department of Tissue Regeneration and Wound Repair, Chinese PLA General Hospital, Beijing, 100853, China.
| | - Xiao-Yan Sun
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Department, Chinese PLA General Hospital and PLA Medical College; PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing, 100048, China.
| |
Collapse
|
20
|
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: 3] [Impact Index Per Article: 3.0] [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
|
21
|
Li S, Gao X, Zheng Y, Yang Y, Gao J, Geng D, Guo L, Ma T, Hao Y, Wei B, Huang L, Wei Y, Xia B, Luo Z, Huang J. Hydralazine represses Fpn ubiquitination to rescue injured neurons via competitive binding to UBA52. J Pharm Anal 2024; 14:86-99. [PMID: 38352945 PMCID: PMC10859533 DOI: 10.1016/j.jpha.2023.08.006] [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: 05/07/2023] [Revised: 07/24/2023] [Accepted: 08/08/2023] [Indexed: 02/16/2024] Open
Abstract
A major impedance to neuronal regeneration after peripheral nerve injury (PNI) is the activation of various programmed cell death mechanisms in the dorsal root ganglion. Ferroptosis is a form of programmed cell death distinguished by imbalance in iron and thiol metabolism, leading to lethal lipid peroxidation. However, the molecular mechanisms of ferroptosis in the context of PNI and nerve regeneration remain unclear. Ferroportin (Fpn), the only known mammalian nonheme iron export protein, plays a pivotal part in inhibiting ferroptosis by maintaining intracellular iron homeostasis. Here, we explored in vitro and in vivo the involvement of Fpn in neuronal ferroptosis. We first delineated that reactive oxygen species at the injury site induces neuronal ferroptosis by increasing intracellular iron via accelerated UBA52-driven ubiquitination and degradation of Fpn, and stimulation of lipid peroxidation. Early administration of the potent arterial vasodilator, hydralazine (HYD), decreases the ubiquitination of Fpn after PNI by binding to UBA52, leading to suppression of neuronal cell death and significant acceleration of axon regeneration and motor function recovery. HYD targeting of ferroptosis is a promising strategy for clinical management of PNI.
Collapse
Affiliation(s)
| | | | | | - Yujie Yang
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Jianbo Gao
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Dan Geng
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Lingli Guo
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Teng Ma
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Yiming Hao
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Bin Wei
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Liangliang Huang
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Yitao Wei
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Bing Xia
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Zhuojing Luo
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Jinghui Huang
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| |
Collapse
|
22
|
Bolandghamat S, Behnam‐Rassouli M. Iron role paradox in nerve degeneration and regeneration. Physiol Rep 2024; 12:e15908. [PMID: 38176709 PMCID: PMC10766496 DOI: 10.14814/phy2.15908] [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: 10/07/2023] [Revised: 12/02/2023] [Accepted: 12/14/2023] [Indexed: 01/06/2024] Open
Abstract
Iron accumulates in the neural tissue during peripheral nerve degeneration. Some studies have already been suggested that iron facilitates Wallerian degeneration (WD) events such as Schwann cell de-differentiation. On the other hand, intracellular iron levels remain elevated during nerve regeneration and gradually decrease. Iron enhances Schwann cell differentiation and axonal outgrowth. Therefore, there seems to be a paradox in the role of iron during nerve degeneration and regeneration. We explain this contradiction by suggesting that the increase in intracellular iron concentration during peripheral nerve degeneration is likely to prepare neural cells for the initiation of regeneration. Changes in iron levels are the result of changes in the expression of iron homeostasis proteins. In this review, we will first discuss the changes in the iron/iron homeostasis protein levels during peripheral nerve degeneration and regeneration and then explain how iron is related to nerve regeneration. This data may help better understand the mechanisms of peripheral nerve repair and find a solution to prevent or slow the progression of peripheral neuropathies.
Collapse
Affiliation(s)
- Samira Bolandghamat
- Department of Biology, Faculty of ScienceFerdowsi University of MashhadMashhadIran
| | | |
Collapse
|
23
|
Xia B, Gao X, Qian J, Li S, Yu B, Hao Y, Wei B, Ma T, Wu H, Yang S, Zheng Y, Gao X, Guo L, Gao J, Yang Y, Zhang Y, Wei Y, Xue B, Jin Y, Luo Z, Zhang J, Huang J. A Novel Superparamagnetic Multifunctional Nerve Scaffold: A Remote Actuation Strategy to Boost In Situ Extracellular Vesicles Production for Enhanced Peripheral Nerve Repair. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2305374. [PMID: 37652460 DOI: 10.1002/adma.202305374] [Citation(s) in RCA: 26] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 08/03/2023] [Indexed: 09/02/2023]
Abstract
Extracellular vesicles (EVs) have inherent advantages over cell-based therapies in regenerative medicine because of their cargos of abundant bioactive cues. Several strategies are proposed to tune EVs production in vitro. However, it remains a challenge for manipulation of EVs production in vivo, which poses significant difficulties for EVs-based therapies that aim to promote tissue regeneration, particularly for long-term treatment of diseases like peripheral neuropathy. Herein, a superparamagnetic nanocomposite scaffold capable of controlling EVs production on-demand is constructed by incorporating polyethyleneglycol/polyethyleneimine modified superparamagnetic nanoparticles into a polyacrylamide/hyaluronic acid double-network hydrogel (Mag-gel). The Mag-gel is highly sensitive to a rotating magnetic field (RMF), and can act as mechano-stimulative platform to exert micro/nanoscale forces on encapsulated Schwann cells (SCs), an essential glial cell in supporting nerve regeneration. By switching the ON/OFF state of the RMF, the Mag-gel can scale up local production of SCs-derived EVs (SCs-EVs) both in vitro and in vivo. Further transcriptome sequencing indicates an enrichment of transcripts favorable in axon growth, angiogenesis, and inflammatory regulation of SCs-EVs in the Mag-gel with RMF, which ultimately results in optimized nerve repair in vivo. Overall, this research provides a noninvasive and remotely time-scheduled method for fine-tuning EVs-based therapies to accelerate tissue regeneration, including that of peripheral nerves.
Collapse
Affiliation(s)
- Bing Xia
- Department of Orthopaedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, P. R. China
- Research and Development Center for Tissue Engineering, School of Stomatology, Fourth Military Medical University, Xi'an, 710032, P. R. China
| | - Xue Gao
- Department of Orthopaedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, P. R. China
| | - Jiaqi Qian
- College of Chemical Engineering, Fuzhou University, Xueyuan Road, Fuzhou, 350108, P. R. China
| | - Shengyou Li
- Department of Orthopaedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, P. R. China
| | - Beibei Yu
- Department of Neurosurgery, The Second Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, 710032, P. R. China
| | - Yiming Hao
- Department of Orthopaedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, P. R. China
| | - Bin Wei
- Department of Orthopaedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, P. R. China
| | - Teng Ma
- Department of Orthopaedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, P. R. China
| | - Haining Wu
- Department of Orthopaedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, P. R. China
| | - Shijie Yang
- Department of Neurosurgery, The Second Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, 710032, P. R. China
| | - Yi Zheng
- Department of Orthopaedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, P. R. China
| | - Xueli Gao
- School of Ecology and Environment, Northwestern Polytechnical University, Xi'an, 710072, P. R. China
| | - Lingli Guo
- Department of Orthopaedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, P. R. China
| | - Jianbo Gao
- Department of Orthopaedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, P. R. China
| | - Yujie Yang
- Department of Orthopaedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, P. R. China
| | - Yongfeng Zhang
- Department of Neurosurgery, The Second Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, 710032, P. R. China
| | - Yitao Wei
- Department of Orthopaedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, P. R. China
| | - Borui Xue
- Department of Orthopaedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, P. R. China
| | - Yan Jin
- Research and Development Center for Tissue Engineering, School of Stomatology, Fourth Military Medical University, Xi'an, 710032, P. R. China
| | - Zhuojing Luo
- Department of Orthopaedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, P. R. China
| | - Jin Zhang
- College of Chemical Engineering, Fuzhou University, Xueyuan Road, Fuzhou, 350108, P. R. China
| | - Jinghui Huang
- Department of Orthopaedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, P. R. China
| |
Collapse
|
24
|
Wang YY, Cheng J, Liu YD, Wang YP, Yang QW, Zhou N. Exosome-based regenerative rehabilitation: A novel ice breaker for neurological disorders. Biomed Pharmacother 2023; 169:115920. [PMID: 37995565 DOI: 10.1016/j.biopha.2023.115920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 11/11/2023] [Accepted: 11/20/2023] [Indexed: 11/25/2023] Open
Abstract
Neurological disorders affect a large population, often leading to different levels of disability and resulting in decreased quality of life. Due to the limited recovery obtained from surgical procedures and other medical approaches, a large number of patients with prolonged dysfunction receive neurorehabilitation protocols to improve their neural plasticity and regeneration. However, the poor neural regeneration ability cannot effectively rebuild the tissue integrity and neural functional networks; consequently, the prognoses of neurorehabilitation remain undetermined. To increase the chances of neural regeneration and functional recovery for patients with neurological disorders, regenerative rehabilitation was introduced with combined regenerative medicine and neurorehabilitation protocols to repair neural tissue damage and create an optimized biophysical microenvironment for neural regeneration potential. With the deepening of exosome research, an increasing number of studies have found that the systemic therapeutic effects of neurorehabilitation approaches are mediated by exosomes released by physically stimulated cells, which provides new insight into rehabilitative mechanisms. Meanwhile, exosome therapy also serves as an alternative cell-free therapy of regenerative medicine that is applied in partnership with neurorehabilitation approaches and formulates exosome-based neurological regenerative rehabilitation. In this study, we review the current state of exosome-associated neurorehabilitation. On the one hand, we focus on presenting the varied mediating effects of exosomes in neurorehabilitation protocols of specific neurological pathologies; on the other hand, we discuss the diverse combinations of exosome therapies and neurorehabilitation approaches in the field of neurological regenerative rehabilitation, aiming to increase the awareness of exosome research and applications in the rehabilitation of neurological disorders.
Collapse
Affiliation(s)
- Yuan-Yi Wang
- Department of Spine Surgery, The First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Jin Cheng
- Department of Sport Medicine, Peking University Third Hospital, Beijing, China
| | - Ya-Dong Liu
- Department of Spine Surgery, The First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Yi-Peng Wang
- Department of Orthopedics, Peking Union Medical College Hospital, Beijing, China.
| | - Qi-Wei Yang
- Medical Research Center, The Second Hospital of Jilin University, Changchun, Jilin Province, China.
| | - Nan Zhou
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Henan Province, China.
| |
Collapse
|
25
|
Ghosh M, Pearse DD. Schwann Cell-Derived Exosomal Vesicles: A Promising Therapy for the Injured Spinal Cord. Int J Mol Sci 2023; 24:17317. [PMID: 38139147 PMCID: PMC10743801 DOI: 10.3390/ijms242417317] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 12/02/2023] [Accepted: 12/06/2023] [Indexed: 12/24/2023] Open
Abstract
Exosomes are nanoscale-sized membrane vesicles released by cells into their extracellular milieu. Within these nanovesicles reside a multitude of bioactive molecules, which orchestrate essential biological processes, including cell differentiation, proliferation, and survival, in the recipient cells. These bioactive properties of exosomes render them a promising choice for therapeutic use in the realm of tissue regeneration and repair. Exosomes possess notable positive attributes, including a high bioavailability, inherent safety, and stability, as well as the capacity to be functionalized so that drugs or biological agents can be encapsulated within them or to have their surface modified with ligands and receptors to imbue them with selective cell or tissue targeting. Remarkably, their small size and capacity for receptor-mediated transcytosis enable exosomes to cross the blood-brain barrier (BBB) and access the central nervous system (CNS). Unlike cell-based therapies, exosomes present fewer ethical constraints in their collection and direct use as a therapeutic approach in the human body. These advantageous qualities underscore the vast potential of exosomes as a treatment option for neurological injuries and diseases, setting them apart from other cell-based biological agents. Considering the therapeutic potential of exosomes, the current review seeks to specifically examine an area of investigation that encompasses the development of Schwann cell (SC)-derived exosomal vesicles (SCEVs) as an approach to spinal cord injury (SCI) protection and repair. SCs, the myelinating glia of the peripheral nervous system, have a long history of demonstrated benefit in repair of the injured spinal cord and peripheral nerves when transplanted, including their recent advancement to clinical investigations for feasibility and safety in humans. This review delves into the potential of utilizing SCEVs as a therapy for SCI, explores promising engineering strategies to customize SCEVs for specific actions, and examines how SCEVs may offer unique clinical advantages over SC transplantation for repair of the injured spinal cord.
Collapse
Affiliation(s)
- Mousumi Ghosh
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA;
- The Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Veterans Affairs, Veterans Affairs Medical Center, Miami, FL 33136, USA
| | - Damien D. Pearse
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA;
- The Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Veterans Affairs, Veterans Affairs Medical Center, Miami, FL 33136, USA
- The Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- The Neuroscience Program, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
26
|
Xu H, Gao Z, Wang Z, Wu W, Li H, Liu Y, Jia S, Hao D, Zhu L. Electrospun PCL Nerve Conduit Filled with GelMA Gel for CNTF and IGF-1 Delivery in Promoting Sciatic Nerve Regeneration in Rat. ACS Biomater Sci Eng 2023; 9:6309-6321. [PMID: 37919884 DOI: 10.1021/acsbiomaterials.3c01048] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2023]
Abstract
Neural tissue engineering is an essential strategy to repair long-segment peripheral nerve defects. Modification of the nerve conduit is an effective way to improve the local microenvironment of the injury site and facilitate nerve regeneration. However, the concurrent release of multiple growth cues that regulate the activity of Schwann cells and neurons remains a challenge. The present study involved the fabrication of a composite hydrogel, specifically methacrylate-anhydride gelatin-ciliary neurotrophic factor/insulin-like growth factor-1 (GelMA-CNTF/IGF-1), with the aim of providing a sustained release of CNTF and IGF-1. The GelMA-CNTF/IGF-1 hydrogels exhibited a swelling rate of 10.2% following a 24 h incubation in vitro. In vitro, GelMA hydrogels demonstrated a high degree of efficiency in the sustained release of CNTF and IGF-1 proteins, with a release rate of 85.9% for CNTF and 90.9% for IGF-1 shown at day 28. In addition, the GelMA-CNTF/IGF-1 composite hydrogel promoted the proliferation of Schwann cells and the production of nerve growth factor (NGF), connective tissue growth factor (CTGF), fibronectin, and laminin and also considerably promoted the axonal growth of neurons. Furthermore, GelMA-CNTF/IGF-1 hydrogels were loaded into PCL electrospun nerve conduits to repair 15 mm sciatic nerve defects in rats. In vivo studies indicated that PCL-GelMA-CNTF/IGF-1 could efficiently accelerate the regeneration of the rat sciatic nerve, promote the formation of the myelin sheath of new axons, promote the electrophysiological function of regenerated nerves, and eventually improve the recovery of motor function in rats. Overall, the PCL-GelMA-CNTF/IGF-1 scaffold presents an attractive new approach for generating an optimal therapeutic alternative for peripheral nerve restoration.
Collapse
Affiliation(s)
- Hailiang Xu
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Youyi East Road No.555, Beilin District, Xi'an, Shaanxi 710054, China
- Shaanxi Key Laboratory of Spine Bionic Treatment, Youyi East Road No.555, Beilin District, Xi'an, Shaanxi 710054, China
| | - Ziheng Gao
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Youyi East Road No.555, Beilin District, Xi'an, Shaanxi 710054, China
- Shaanxi Key Laboratory of Spine Bionic Treatment, Youyi East Road No.555, Beilin District, Xi'an, Shaanxi 710054, China
| | - Zhiyuan Wang
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Youyi East Road No.555, Beilin District, Xi'an, Shaanxi 710054, China
- Shaanxi Key Laboratory of Spine Bionic Treatment, Youyi East Road No.555, Beilin District, Xi'an, Shaanxi 710054, China
| | - Weidong Wu
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Youyi East Road No.555, Beilin District, Xi'an, Shaanxi 710054, China
- Shaanxi Key Laboratory of Spine Bionic Treatment, Youyi East Road No.555, Beilin District, Xi'an, Shaanxi 710054, China
| | - Hui Li
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Youyi East Road No.555, Beilin District, Xi'an, Shaanxi 710054, China
- Shaanxi Key Laboratory of Spine Bionic Treatment, Youyi East Road No.555, Beilin District, Xi'an, Shaanxi 710054, China
| | - Youjun Liu
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Youyi East Road No.555, Beilin District, Xi'an, Shaanxi 710054, China
- Shaanxi Key Laboratory of Spine Bionic Treatment, Youyi East Road No.555, Beilin District, Xi'an, Shaanxi 710054, China
| | - Shuaijun Jia
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Youyi East Road No.555, Beilin District, Xi'an, Shaanxi 710054, China
- Shaanxi Key Laboratory of Spine Bionic Treatment, Youyi East Road No.555, Beilin District, Xi'an, Shaanxi 710054, China
| | - Dingjun Hao
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Youyi East Road No.555, Beilin District, Xi'an, Shaanxi 710054, China
- Shaanxi Key Laboratory of Spine Bionic Treatment, Youyi East Road No.555, Beilin District, Xi'an, Shaanxi 710054, China
| | - Lei Zhu
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Youyi East Road No.555, Beilin District, Xi'an, Shaanxi 710054, China
- Shaanxi Key Laboratory of Spine Bionic Treatment, Youyi East Road No.555, Beilin District, Xi'an, Shaanxi 710054, China
| |
Collapse
|
27
|
Gao X, Li S, Yang Y, Yang S, Yu B, Zhu Z, Ma T, Zheng Y, Wei B, Hao Y, Wu H, Zhang Y, Guo L, Gao X, Wei Y, Xue B, Li J, Feng X, Lu L, Xia B, Huang J. A Novel Magnetic Responsive miR-26a@SPIONs-OECs for Spinal Cord Injury: Triggering Neural Regeneration Program and Orienting Axon Guidance in Inhibitory Astrocytic Environment. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2304487. [PMID: 37789583 PMCID: PMC10646239 DOI: 10.1002/advs.202304487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 09/12/2023] [Indexed: 10/05/2023]
Abstract
Addressing the challenge of promoting directional axonal regeneration in a hostile astrocytic scar, which often impedes recovery following spinal cord injury (SCI), remains a daunting task. Cell transplantation is a promising strategy to facilitate nerve restoration in SCI. In this research, a pro-regeneration system is developed, namely miR-26a@SPIONs-OECs, for olfactory ensheathing cells (OECs), a preferred choice for promoting nerve regeneration in SCI patients. These entities show high responsiveness to external magnetic fields (MF), leading to synergistic multimodal cues to enhance nerve regeneration. First, an MF stimulates miR-26a@SPIONs-OECs to release extracellular vesicles (EVs) rich in miR-26a. This encourages axon growth by inhibiting PTEN and GSK-3β signaling pathways in neurons. Second, miR-26a@SPIONs-OECs exhibit a tendency to migrate and orientate along the direction of the MF, thereby potentially facilitating neuronal reconnection through directional neurite elongation. Third, miR-26a-enriched EVs from miR-26a@SPIONs-OECs can interact with host astrocytes, thereby diminishing inhibitory cues for neurite growth. In a rat model of SCI, the miR-26a@SPIONs-OECs system led to significantly improved morphological and motor function recovery. In summary, the miR-26a@SPIONS-OECs pro-regeneration system offers innovative insights into engineering exogenous cells with multiple additional cues, augmenting their efficacy for stimulating and guiding nerve regeneration within a hostile astrocytic scar in SCI.
Collapse
Affiliation(s)
- Xue Gao
- Department of OrthopaedicsXijing HospitalFourth Military Medical UniversityXi'an710032P. R. China
| | - Shengyou Li
- Department of OrthopaedicsXijing HospitalFourth Military Medical UniversityXi'an710032P. R. China
| | - Yujie Yang
- Department of OrthopaedicsXijing HospitalFourth Military Medical UniversityXi'an710032P. R. China
| | - Shijie Yang
- Department of NeurosurgeryThe Second Affiliated Hospital of Xi'an Jiao Tong UniversityXi'an710032P. R. China
| | - Beibei Yu
- Department of NeurosurgeryThe Second Affiliated Hospital of Xi'an Jiao Tong UniversityXi'an710032P. R. China
| | - Zhijie Zhu
- Department of OrthopaedicsXijing HospitalFourth Military Medical UniversityXi'an710032P. R. China
| | - Teng Ma
- Department of OrthopaedicsXijing HospitalFourth Military Medical UniversityXi'an710032P. R. China
| | - Yi Zheng
- Department of OrthopaedicsXijing HospitalFourth Military Medical UniversityXi'an710032P. R. China
| | - Bin Wei
- Department of OrthopaedicsXijing HospitalFourth Military Medical UniversityXi'an710032P. R. China
| | - Yiming Hao
- Department of OrthopaedicsXijing HospitalFourth Military Medical UniversityXi'an710032P. R. China
| | - Haining Wu
- Department of OrthopaedicsXijing HospitalFourth Military Medical UniversityXi'an710032P. R. China
| | - Yongfeng Zhang
- Department of NeurosurgeryThe Second Affiliated Hospital of Xi'an Jiao Tong UniversityXi'an710032P. R. China
| | - Lingli Guo
- Department of OrthopaedicsXijing HospitalFourth Military Medical UniversityXi'an710032P. R. China
| | - Xueli Gao
- School of Ecology and EnvironmentNorthwestern Polytechnical UniversityXi'an710072P. R. China
| | - Yitao Wei
- Department of OrthopaedicsXijing HospitalFourth Military Medical UniversityXi'an710032P. R. China
| | - Borui Xue
- Department of OrthopaedicsXijing HospitalFourth Military Medical UniversityXi'an710032P. R. China
| | - Jianzhong Li
- Department of Thoracic SurgeryThe Second Affiliated Hospital of Xi'an Jiao Tong UniversityXi'an710032P. R. China
| | - Xue Feng
- Department of Cell BiologySchool of MedicineNorthwest UniversityXi'an710032P. R. China
| | - Lei Lu
- State Key Laboratory of Military StomatologyNational Clinical Research Center for Oral DiseasesShaanxi International Joint Research Center for Oral DiseasesDepartment of Oral Anatomy and Physiology and TMDSchool of Stomatologythe Fourth Military Medical UniversityXi'an710032P. R. China
| | - Bing Xia
- Department of OrthopaedicsXijing HospitalFourth Military Medical UniversityXi'an710032P. R. China
| | - Jinghui Huang
- Department of OrthopaedicsXijing HospitalFourth Military Medical UniversityXi'an710032P. R. China
| |
Collapse
|
28
|
Ye K, Li Z, Yin Y, Zhou J, Li D, Gan Y, Peng D, Xiao M, Zhao L, Dai Y, Tang Y. LIPUS-SCs-Exo promotes peripheral nerve regeneration in cavernous nerve crush injury-induced ED rats via PI3K/Akt/FoxO signaling pathway. CNS Neurosci Ther 2023; 29:3239-3258. [PMID: 37157936 PMCID: PMC10580359 DOI: 10.1111/cns.14256] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 04/25/2023] [Accepted: 04/26/2023] [Indexed: 05/10/2023] Open
Abstract
OBJECTIVE Clinical treatment of erectile dysfunction (ED) caused by cavernous nerve (CN) injury during pelvic surgery is difficult. Low-intensity pulsed ultrasound (LIPUS) can be a potential strategy for neurogenic ED (NED). However, whether Schwann cells (SCs) can respond to LIPUS stimulation signals is unclear. This study aims to elucidate the signal transmission between SCs paracrine exosome (Exo) and neurons stimulated by LIPUS, as well as to analyze the role and potential mechanisms of exosomes in CN repair after injury. METHODS The major pelvic ganglion (MPG) neurons and MPG/CN explants were stimulated with LIPUS of different energy intensities to explore the appropriate LIPUS energy intensity. The exosomes were isolated and purified from LIPUS-stimulated SCs (LIPUS-SCs-Exo) and non-stimulated SCs (SCs-Exo). The effects of LIPUS-SCs-Exo on neurite outgrowth, erectile function, and cavernous penis histology were identified in bilateral cavernous nerve crush injury (BCNI)-induced ED rats. RESULTS LIPUS-SCs-Exo group can enhance the axon elongation of MPG/CN and MPG neurons compared to SCs-Exo group in vitro. Then, the LIPUS-SCs-Exo group showed a stronger ability to promote the injured CN regeneration and SCs proliferation compared to the SCs-Exo group in vivo. Furthermore, the LIPUS-SCs-Exo group increased the Max intracavernous pressure (ICP)/mean arterial pressure (MAP), lumen to parenchyma and smooth muscle to collagen ratios compared to the SCs-Exo group in vivo. Additionally, high-throughput sequencing combined with bioinformatics analysis revealed the differential expression of 1689 miRNAs between the SCs-Exo group and the LIPUS-SCs-Exo group. After LIPUS-SCs-Exo treatment, the phosphorylated levels of Phosphatidylinositol 3-kinase (PI3K), protein kinase B (Akt) and forkhead box O (FoxO) in MPG neurons increased significantly compared to negative control (NC) and SCs-Exo groups. CONCLUSION Our study revealed that LIPUS stimulation could regulate the gene of MPG neurons by changing miRNAs derived from SCs-Exo, then activating the PI3K-Akt-FoxO signal pathway to enhance nerve regeneration and restore erectile function. This study had important theoretical and practical significance for improving the NED treatment.
Collapse
Affiliation(s)
- Kun Ye
- Department of UrologyThe Fifth Affiliated Hospital of Sun Yat‐Sen UniversityZhuhaiGuangdongChina
- Guangdong Provincial Key Laboratory of Biomedical ImagingThe Fifth Affiliated Hospital, Sun Yat‐Sen UniversityZhuhaiGuangdongChina
| | - Zitaiyu Li
- Department of UrologyThe Fifth Affiliated Hospital of Sun Yat‐Sen UniversityZhuhaiGuangdongChina
- Guangdong Provincial Key Laboratory of Biomedical ImagingThe Fifth Affiliated Hospital, Sun Yat‐Sen UniversityZhuhaiGuangdongChina
| | - Yinghao Yin
- Department of UrologyThe Fifth Affiliated Hospital of Sun Yat‐Sen UniversityZhuhaiGuangdongChina
- Guangdong Provincial Key Laboratory of Biomedical ImagingThe Fifth Affiliated Hospital, Sun Yat‐Sen UniversityZhuhaiGuangdongChina
| | - Jun Zhou
- Department of UrologyThe Fifth Affiliated Hospital of Sun Yat‐Sen UniversityZhuhaiGuangdongChina
- Guangdong Provincial Key Laboratory of Biomedical ImagingThe Fifth Affiliated Hospital, Sun Yat‐Sen UniversityZhuhaiGuangdongChina
| | - Dongjie Li
- Department of UrologyXiangya Hospital, Central South UniversityChangshaChina
| | - Yu Gan
- Department of UrologyXiangya Hospital, Central South UniversityChangshaChina
| | - Dongyi Peng
- Department of UrologyThe Third Xiangya Hospital of Central South UniversityChangshaChina
| | - Ming Xiao
- Department of UrologyThe Fifth Affiliated Hospital of Sun Yat‐Sen UniversityZhuhaiGuangdongChina
- Guangdong Provincial Key Laboratory of Biomedical ImagingThe Fifth Affiliated Hospital, Sun Yat‐Sen UniversityZhuhaiGuangdongChina
| | - Liangyu Zhao
- Department of UrologyThe Fifth Affiliated Hospital of Sun Yat‐Sen UniversityZhuhaiGuangdongChina
- Guangdong Provincial Key Laboratory of Biomedical ImagingThe Fifth Affiliated Hospital, Sun Yat‐Sen UniversityZhuhaiGuangdongChina
| | - Yingbo Dai
- Department of UrologyThe Fifth Affiliated Hospital of Sun Yat‐Sen UniversityZhuhaiGuangdongChina
- Guangdong Provincial Key Laboratory of Biomedical ImagingThe Fifth Affiliated Hospital, Sun Yat‐Sen UniversityZhuhaiGuangdongChina
| | - Yuxin Tang
- Department of UrologyThe Fifth Affiliated Hospital of Sun Yat‐Sen UniversityZhuhaiGuangdongChina
- Guangdong Provincial Key Laboratory of Biomedical ImagingThe Fifth Affiliated Hospital, Sun Yat‐Sen UniversityZhuhaiGuangdongChina
| |
Collapse
|
29
|
Tai Y, Tonmoy TI, Win S, Brinkley NT, Park BH, Nam J. Enhanced peripheral nerve regeneration by mechano-electrical stimulation. NPJ Regen Med 2023; 8:57. [PMID: 37848428 PMCID: PMC10582163 DOI: 10.1038/s41536-023-00334-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 09/29/2023] [Indexed: 10/19/2023] Open
Abstract
To address limitations in current approaches for treating large peripheral nerve defects, the presented study evaluated the feasibility of functional material-mediated physical stimuli on peripheral nerve regeneration. Electrospun piezoelectric poly(vinylidene fluoride-trifluoroethylene) nanofibers were utilized to deliver mechanical actuation-activated electrical stimulation to nerve cells/tissues in a non-invasive manner. Using morphologically and piezoelectrically optimized nanofibers for neurite extension and Schwann cell maturation based on in vitro experiments, piezoelectric nerve conduits were synthesized and implanted in a rat sciatic nerve transection model to bridge a critical-sized sciatic nerve defect (15 mm). A therapeutic shockwave system was utilized to periodically activate the piezoelectric effect of the implanted nerve conduit on demand. The piezoelectric nerve conduit-mediated mechano-electrical stimulation (MES) induced enhanced peripheral nerve regeneration, resulting in full axon reconnection with myelin regeneration from the proximal to the distal ends over the critical-sized nerve gap. In comparison, a control group, in which the implanted piezoelectric conduits were not activated in vivo, failed to exhibit such nerve regeneration. In addition, at both proximal and distal ends of the implanted conduits, a decreased number of damaged myelination (ovoids), an increased number of myelinated nerves, and a larger axonal diameter were observed under the MES condition as compared to the control condition. Furthermore, unlike the control group, the MES condition exhibited a superior functional nerve recovery, assessed by walking track analysis and polarization-sensitive optical coherence tomography, demonstrating the significant potential of the piezoelectric conduit-based physical stimulation approach for the treatment of peripheral nerve injury.
Collapse
Affiliation(s)
- Youyi Tai
- Department of Bioengineering, University of California, Riverside, CA, 92521, USA
| | | | - Shwe Win
- Department of Bioengineering, University of California, Riverside, CA, 92521, USA
| | - Natasha T Brinkley
- Department of Bioengineering, University of California, Riverside, CA, 92521, USA
| | - B Hyle Park
- Department of Bioengineering, University of California, Riverside, CA, 92521, USA
| | - Jin Nam
- Department of Bioengineering, University of California, Riverside, CA, 92521, USA.
| |
Collapse
|
30
|
Oliveira JT, Yanick C, Wein N, Gomez Limia CE. Neuron-Schwann cell interactions in peripheral nervous system homeostasis, disease, and preclinical treatment. Front Cell Neurosci 2023; 17:1248922. [PMID: 37900588 PMCID: PMC10600466 DOI: 10.3389/fncel.2023.1248922] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 09/19/2023] [Indexed: 10/31/2023] Open
Abstract
Schwann cells (SCs) have a critical role in the peripheral nervous system. These cells are able to support axons during homeostasis and after injury. However, mutations in genes associated with the SCs repair program or myelination result in dysfunctional SCs. Several neuropathies such as Charcot-Marie-Tooth (CMT) disease, diabetic neuropathy and Guillain-Barré syndrome show abnormal SC functions and an impaired regeneration process. Thus, understanding SCs-axon interaction and the nerve environment in the context of homeostasis as well as post-injury and disease onset is necessary. Several neurotrophic factors, cytokines, and regulators of signaling pathways associated with proliferation, survival and regeneration are involved in this process. Preclinical studies have focused on the discovery of therapeutic targets for peripheral neuropathies and injuries. To study the effect of new therapeutic targets, modeling neuropathies and peripheral nerve injuries (PNIs) in vitro and in vivo are useful tools. Furthermore, several in vitro protocols have been designed using SCs and neuron cell lines to evaluate these targets in the regeneration process. SCs lines have been used to generate effective myelinating SCs without success. Alternative options have been investigated using direct conversion from somatic cells to SCs or SCs derived from pluripotent stem cells to generate functional SCs. This review will go over the advantages of these systems and the problems associated with them. In addition, there have been challenges in establishing adequate and reproducible protocols in vitro to recapitulate repair SC-neuron interactions observed in vivo. So, we also discuss the mechanisms of repair SCs-axon interactions in the context of peripheral neuropathies and nerve injury (PNI) in vitro and in vivo. Finally, we summarize current preclinical studies evaluating transgenes, drug, and novel compounds with translational potential into clinical studies.
Collapse
Affiliation(s)
| | | | - Nicolas Wein
- Center for Gene Therapy, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, United States
- Department of Pediatrics, The Ohio State University, Columbus, OH, United States
| | | |
Collapse
|
31
|
Shekari F, Alibhai FJ, Baharvand H, Börger V, Bruno S, Davies O, Giebel B, Gimona M, Salekdeh GH, Martin‐Jaular L, Mathivanan S, Nelissen I, Nolte‐’t Hoen E, O'Driscoll L, Perut F, Pluchino S, Pocsfalvi G, Salomon C, Soekmadji C, Staubach S, Torrecilhas AC, Shelke GV, Tertel T, Zhu D, Théry C, Witwer K, Nieuwland R. Cell culture-derived extracellular vesicles: Considerations for reporting cell culturing parameters. JOURNAL OF EXTRACELLULAR BIOLOGY 2023; 2:e115. [PMID: 38939735 PMCID: PMC11080896 DOI: 10.1002/jex2.115] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 08/18/2023] [Accepted: 09/17/2023] [Indexed: 06/29/2024]
Abstract
Cell culture-conditioned medium (CCM) is a valuable source of extracellular vesicles (EVs) for basic scientific, therapeutic and diagnostic applications. Cell culturing parameters affect the biochemical composition, release and possibly the function of CCM-derived EVs (CCM-EV). The CCM-EV task force of the Rigor and Standardization Subcommittee of the International Society for Extracellular Vesicles aims to identify relevant cell culturing parameters, describe their effects based on current knowledge, recommend reporting parameters and identify outstanding questions. While some recommendations are valid for all cell types, cell-specific recommendations may need to be established for non-mammalian sources, such as bacteria, yeast and plant cells. Current progress towards these goals is summarized in this perspective paper, along with a checklist to facilitate transparent reporting of cell culturing parameters to improve the reproducibility of CCM-EV research.
Collapse
Affiliation(s)
- Faezeh Shekari
- Department of Stem Cells and Developmental Biology, Cell Science Research CenterRoyan Institute for Stem Cell Biology and Technology, ACECRTehranIran
- Advanced Therapy Medicinal Product Technology Development Center (ATMP‐TDC), Cell Science Research CenterRoyan Institute for Stem Cell Biology and Technology, ACECRTehranIran
| | | | - Hossein Baharvand
- Department of Stem Cells and Developmental Biology, Cell Science Research CenterRoyan Institute for Stem Cell Biology and Technology, ACECRTehranIran
- Department of Developmental Biology, School of Basic Sciences and Advanced Technologies in BiologyUniversity of Science and CultureTehranIran
| | - Verena Börger
- Institute for Transfusion MedicineUniversity Hospital Essen, University of Duisburg‐EssenEssenGermany
| | - Stefania Bruno
- Department of Medical Sciences and Molecular Biotechnology CenterUniversity of TorinoTurinItaly
| | - Owen Davies
- School of Sport, Exercise and Health SciencesLoughborough UniversityLoughboroughUK
| | - Bernd Giebel
- Institute for Transfusion MedicineUniversity Hospital Essen, University of Duisburg‐EssenEssenGermany
| | - Mario Gimona
- GMP UnitSpinal Cord Injury & Tissue Regeneration Centre Salzburg (SCI‐TReCS) and Research Program “Nanovesicular Therapies” Paracelsus Medical UniversitySalzburgAustria
| | | | - Lorena Martin‐Jaular
- Institut Curie, INSERM U932 and Curie CoreTech Extracellular VesiclesPSL Research UniversityParisFrance
| | - Suresh Mathivanan
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular ScienceLa Trobe UniversityMelbourneVICAustralia
| | - Inge Nelissen
- VITO (Flemish Institute for Technological Research), Health departmentBoeretangBelgium
| | - Esther Nolte‐’t Hoen
- Department of Biomolecular Health Sciences, Faculty of Veterinary MedicineUtrecht UniversityUtrechtThe Netherlands
| | - Lorraine O'Driscoll
- School of Pharmacy and Pharmaceutical Sciences & Trinity Biomedical Sciences InstituteTrinity College DublinDublinIreland
| | - Francesca Perut
- Biomedical Science and Technologies and Nanobiotechnology LabIRCCS Istituto Ortopedico RizzoliBolognaItaly
| | - Stefano Pluchino
- Department of Clinical NeurosciencesUniversity of CambridgeCambridgeUK
| | - Gabriella Pocsfalvi
- Institute of Biosciences and BioResourcesNational Research CouncilNaplesItaly
| | - Carlos Salomon
- Translational Extracellular Vesicles in Obstetrics and Gynae‐Oncology Group, UQ Centre for Clinical Research, Royal Brisbane and Women's Hospital, Faculty of MedicineThe University of QueenslandBrisbaneAustralia
| | - Carolina Soekmadji
- School of Biomedical Sciences, Faculty of MedicineUniversity of QueenslandBrisbaneAustralia
| | | | - Ana Claudia Torrecilhas
- Laboratório de Imunologia Celular e Bioquímica de Fungos e Protozoários, Departamento de Ciências FarmacêuticasUniversidade Federal de São Paulo (UNIFESP)SPBrazil
| | - Ganesh Vilas Shelke
- Neurosciences and Cellular and Structural Biology Division, Eunice Kennedy Shriver National Institute of Child Health and Human DevelopmentNational Institutes of HealthBethesdaMarylandUSA
| | - Tobias Tertel
- Institute for Transfusion MedicineUniversity Hospital Essen, University of Duisburg‐EssenEssenGermany
| | - Dandan Zhu
- The Ritchie CentreHudson Institute of Medical ResearchClaytonVICAustralia
| | - Clotilde Théry
- Institut Curie, INSERM U932 and Curie CoreTech Extracellular VesiclesPSL Research UniversityParisFrance
| | - Kenneth Witwer
- Departments of Molecular and Comparative Pathobiology and Neurology and Richman Family Precision Medicine Center of Excellence in Alzheimer's DiseaseJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Rienk Nieuwland
- Laboratory of Experimental Clinical Chemistry, Department of Clinical Chemistry, Amsterdam University Medical CentersLocation AMC, University of AmsterdamAmsterdamThe Netherlands
- Amsterdam Vesicle Center, Amsterdam University Medical Centers, location AMCUniversity of AmsterdamAmsterdamThe Netherlands
| |
Collapse
|
32
|
Thompson W, Papoutsakis ET. Similar but distinct: The impact of biomechanical forces and culture age on the production, cargo loading, and biological efficacy of human megakaryocytic extracellular vesicles for applications in cell and gene therapies. Bioeng Transl Med 2023; 8:e10563. [PMID: 37693047 PMCID: PMC10486331 DOI: 10.1002/btm2.10563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 05/18/2023] [Accepted: 06/01/2023] [Indexed: 09/12/2023] Open
Abstract
Megakaryocytic extracellular vesicles (MkEVs) promote the growth and megakaryopoiesis of hematopoietic stem and progenitor cells (HSPCs) largely through endogenous miR-486-5p and miR-22-3p cargo. Here, we examine the impact of biomechanical force and culture age/differentiation on the formation, properties, and biological efficacy of MkEVs. We applied biomechanical force to Mks using two methods: shake flask cultures and a syringe pump system. Force increased MkEV production in a magnitude-dependent manner, with similar trends emerging regardless of whether flow cytometry or nanoparticle tracking analysis was used for MkEV counting. Both methods produced MkEVs that were relatively depleted of miR-486-5p and miR-22-3p cargo. However, while the shake flask-derived MkEVs were correspondingly less effective in promoting megakaryocytic differentiation of HSPCs, the syringe pump-derived MkEVs were more effective in doing so, suggesting the presence of unique, unidentified miRNA cargo components. Higher numbers of MkEVs were also produced by "older" Mk cultures, though miRNA cargo levels and MkEV bioactivity were unaffected by culture age. A reduction in MkEV production by Mks derived from late-differentiating HSPCs was also noted. Taken together, our results demonstrate that biomechanical force has an underappreciated and deeply influential role in MkEV biology, though that role may vary significantly depending on the nature of the force. Given the ubiquity of biomechanical force in vivo and in biomanufacturing, this phenomenon must be grappled with before MkEVs can attain clinical relevance.
Collapse
Affiliation(s)
- Will Thompson
- Department of Chemical and Biomolecular EngineeringUniversity of DelawareNewarkDelawareUSA
| | | |
Collapse
|
33
|
Fan B, Chopp M, Zhang Y, Wang X, Kemper A, Zhang ZG, Liu XS. Ablation of Argonaute 2 in Schwann cells accelerates the progression of diabetic peripheral neuropathy. Glia 2023; 71:2196-2209. [PMID: 37178056 PMCID: PMC11057225 DOI: 10.1002/glia.24387] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 04/25/2023] [Accepted: 05/05/2023] [Indexed: 05/15/2023]
Abstract
Schwann cells (SCs) form myelin and provide metabolic support for axons, and are essential for normal nerve function. Identification of key molecules specific to SCs and nerve fibers may provide new therapeutic targets for diabetic peripheral neuropathy (DPN). Argonaute2 (Ago2) is a key molecular player that mediates the activity of miRNA-guided mRNA cleavage and miRNA stability. Our study found that Ago2 knockout (Ago2-KO) in proteolipid protein (PLP) lineage SCs in mice resulted in a significant reduction of nerve conduction velocities and impairments of thermal and mechanical sensitivities. Histopathological data revealed that Ago2-KO significantly induced demyelination and neurodegeneration. When DPN was induced in both wild-type and Ago2-KO mice, Ago2-KO mice exhibited further decreased myelin thickness and exacerbated neurological outcomes compared with wild-type mice. Deep sequencing analysis of Ago2 immunoprecipitated complexes showed that deregulated miR-206 in Ago2-KO mice is highly related to mitochondrial function. In vitro data showed that knockdown of miR-200 induced mitochondrial dysfunction and apoptosis in SCs. Together, our data suggest that Ago2 in SCs is essential to maintain peripheral nerve function while ablation of Ago2 in SCs exacerbates SC dysfunction and neuronal degeneration in DPN. These findings provide new insight into the molecular mechanisms of DPN.
Collapse
Affiliation(s)
- Baoyan Fan
- Department of Neurology, Henry Ford Health System, Detroit, Michigan, USA
| | - Michael Chopp
- Department of Neurology, Henry Ford Health System, Detroit, Michigan, USA
- Department of Physics, Oakland University, Rochester, Michigan, USA
| | - Yi Zhang
- Department of Neurology, Henry Ford Health System, Detroit, Michigan, USA
| | - Xinli Wang
- Department of Neurology, Henry Ford Health System, Detroit, Michigan, USA
| | - Amy Kemper
- Department of Pathology, Henry Ford Hospital, Detroit, Michigan, USA
| | - Zheng Gang Zhang
- Department of Neurology, Henry Ford Health System, Detroit, Michigan, USA
| | - Xian Shuang Liu
- Department of Neurology, Henry Ford Health System, Detroit, Michigan, USA
| |
Collapse
|
34
|
Zhiguo F, Ji W, Shenyuan C, Guoyou Z, Chen K, Hui Q, Wenrong X, Zhai X. A swift expanding trend of extracellular vesicles in spinal cord injury research: a bibliometric analysis. J Nanobiotechnology 2023; 21:289. [PMID: 37612689 PMCID: PMC10463993 DOI: 10.1186/s12951-023-02051-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 08/04/2023] [Indexed: 08/25/2023] Open
Abstract
Extracellular vesicles (EVs) in the field of spinal cord injury (SCI) have garnered significant attention for their potential applications in diagnosis and therapy. However, no bibliometric assessment has been conducted to evaluate the scientific progress in this area. A search of articles in Web of Science (WoS) from January 1, 1991, to May 1, 2023, yielded 359 papers that were analyzed using various online analysis tools. These articles have been cited 10,842 times with 30.2 times per paper. The number of publications experienced explosive growth starting in 2015. China and the United States led this research initiative. Keywords were divided into 3 clusters, including "Pathophysiology of SCI", "Bioactive components of EVs", and "Therapeutic effects of EVs in SCI". By integrating the average appearing year (AAY) of keywords in VoSviewer with the time zone map of the Citation Explosion in CiteSpace, the focal point of research has undergone a transformative shift. The emphasis has moved away from pathophysiological factors such as "axon", "vesicle", and "glial cell" to more mechanistic and applied domains such as "activation", "pathways", "hydrogels" and "therapy". In conclusions, institutions are expected to allocate more resources towards EVs-loaded hydrogel therapy and the utilization of innovative materials for injury mitigation.
Collapse
Affiliation(s)
- Fan Zhiguo
- Department of Orthopedics, Shanghai Changhai Hospital, Shanghai, 200433, China
| | - Wu Ji
- Department of Orthopedics, Shanghai Changhai Hospital, Shanghai, 200433, China
| | - Chen Shenyuan
- Key Laboratory of Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Zhang Guoyou
- Department of Orthopedics, Shanghai Changhai Hospital, Shanghai, 200433, China
| | - Kai Chen
- Department of Orthopedics, Shanghai Changhai Hospital, Shanghai, 200433, China.
| | - Qian Hui
- Key Laboratory of Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, China.
| | - Xu Wenrong
- Key Laboratory of Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, China.
| | - Xiao Zhai
- Department of Orthopedics, Shanghai Changhai Hospital, Shanghai, 200433, China.
| |
Collapse
|
35
|
Zhang L, Liu J, Zhou C. Current aspects of small extracellular vesicles in pain process and relief. Biomater Res 2023; 27:78. [PMID: 37563666 PMCID: PMC10416402 DOI: 10.1186/s40824-023-00417-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 08/04/2023] [Indexed: 08/12/2023] Open
Abstract
Small extracellular vesicles (sEVs) have been identified as a noteworthy paracrine mechanism of intercellular communication in diagnosing and managing neurological disorders. Current research suggests that sEVs play a pivotal role in the pathological progression of pain, emphasizing their critical function in the pathological progression of pain in acute and chronic pain models. By facilitating the transfer of diverse molecules, such as proteins, nucleic acids, and metabolites, sEVs can modulate pain signaling transmission in both the central and peripheral nervous systems. Furthermore, the unique molecules conveyed by sEVs in pain disorders indicate their potential as diagnostic biomarkers. The application of sEVs derived from mesenchymal stem cells (MSCs) in regenerative pain medicine has emerged as a promising strategy for pain management. Moreover, modified sEVs have garnered considerable attention in the investigation of pathological processes and therapeutic interventions. This review presents a comprehensive overview of the current knowledge regarding the involvement of sEVs in pain pathogenesis and treatment. Nevertheless, additional research is imperative to facilitate their clinical implementation. Schematic diagram of sEVs in the biogenesis, signal transmission, diagnosis, and treatment of pain disorders. Small extracellular vesicles (sEVs) are secreted by multiple cells, loading with various biomolecules, such as miRNAs, transmembrane proteins, and amino acids. They selectively target other cells and regulating pain signal transmission. The composition of sEVs can serve as valuable biomarkers for pain diagnosis. In particular, mesenchymal stem cell-derived sEVs have shown promise as regenerative medicine for managing multiple pain disorders. Furthermore, by modifying the structure or contents of sEVs, they could potentially be used as a potent analgesic method.
Collapse
Affiliation(s)
- Lanyu Zhang
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Anesthesia & Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital of Sichuan University, Chengdu, 610041, Sichuan, China
| | - Jin Liu
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
| | - Cheng Zhou
- Laboratory of Anesthesia & Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital of Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
36
|
Zheng B, Peng M, Gong J, Li C, Cheng H, Li Y, Tang Y. Circulating exosomal microRNA-4497 as a potential biomarker for metastasis and prognosis in non-small-cell lung cancer. Exp Biol Med (Maywood) 2023; 248:1403-1413. [PMID: 37551102 PMCID: PMC10657587 DOI: 10.1177/15353702231184223] [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: 10/09/2022] [Accepted: 04/12/2023] [Indexed: 08/09/2023] Open
Abstract
Circulating exosomal microRNAs (miRNAs) have shown great potential for the diagnosis, prognosis, and treatment monitoring of patients with non-small-cell lung cancer (NSCLC). Our main purpose was to determine the clinical value of serum exosomal miR-4497 as a new non-invasive biomarker for NSCLC. The exoRNeasy Kit (QIAGEN, Hilden, Germany) was used to isolate exosomes and exoRNA from the serum of 84 patients with NSCLC (NSCLC group), 30 patients with benign lung lesion (BLL group), and 47 healthy controls. Six serum exosomal miRNAs (Let-7b-5p, miR-122-5p, miR-155-5p, miR-223-3p, miR-320c, and miR-4497) were selected as candidate miRNAs and analyzed using real-time qPCR, among which miR-4497 displayed the most striking differences. Exosomal miR-4497 expressed significantly lower in NSCLC than in BLL patients and healthy controls (P < 0.001). Further investigation showed that miR-4497 was negatively correlated with the malignant characteristics of tumors (tumor size, tumor-node-metastasis [TNM] stage, and distant metastasis) and was an independent tumor suppressor (P < 0.05). According to receiver operating characteristic (ROC) analysis, exosomal miR-4497 independently exhibited excellent diagnostic efficacy, which could be improved by combining it with traditional markers (for identifying tumor size, the area under the curve [AUC] = 0.761; TNM stage, AUC = 0.878; distant metastasis, AUC = 0.895; all P < 0.001). Moreover, longitudinal analysis revealed that exosomal miR-4497 levels increased after chemoradiotherapy (P < 0.001). According to the survival analysis, poor overall survival (OS) and disease-free survival (DFS) were associated with low exosomal miR-4497 levels (P < 0.05). Moreover, exosomal miR-4497 was an independent protective factor affecting DFS (hazard ratio = 0.190, P = 0.009) in the Cox proportional hazards model. Therefore, serum exosomal miR-4497 can be used as a potential biomarker to identify NSCLC and healthy individuals or BLL patients for early screening or as a biomarker for staging and grading, prognosis, and monitoring recurrence, metastasis, and the therapeutic effects in patients with NSCLC.
Collapse
Affiliation(s)
- Bokun Zheng
- Department of Clinical Laboratory, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, China
- Wuhan Research Center for Infectious Diseases and Cancer, Chinese Academy of Medical Sciences, Wuhan, Hubei 430071, China
| | - Mingcheng Peng
- Department of Clinical Laboratory, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, China
- Wuhan Research Center for Infectious Diseases and Cancer, Chinese Academy of Medical Sciences, Wuhan, Hubei 430071, China
| | - Jun Gong
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, China
| | - Changsheng Li
- Department of Thoracic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, China
| | - Hongbing Cheng
- Department of Thoracic Surgery, Xiantao First People’s Hospital Affiliated to Yangtze University, Xiantao, Hubei 433099, China
| | - Yirong Li
- Department of Clinical Laboratory, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, China
- Wuhan Research Center for Infectious Diseases and Cancer, Chinese Academy of Medical Sciences, Wuhan, Hubei 430071, China
| | - Yueting Tang
- Department of Clinical Laboratory, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, China
- Wuhan Research Center for Infectious Diseases and Cancer, Chinese Academy of Medical Sciences, Wuhan, Hubei 430071, China
| |
Collapse
|
37
|
Freiría-Martínez L, Iglesias-Martínez-Almeida M, Rodríguez-Jamardo C, Rivera-Baltanás T, Comís-Tuche M, Rodrígues-Amorím D, Fernández-Palleiro P, Blanco-Formoso M, Diz-Chaves Y, González-Freiria N, Suárez-Albo M, Martín-Forero-Maestre M, Durán Fernández-Feijoo C, Fernández-Lorenzo JR, Concheiro Guisán A, Olivares JM, Spuch C. Human Breast Milk microRNAs, Potential Players in the Regulation of Nervous System. Nutrients 2023; 15:3284. [PMID: 37513702 PMCID: PMC10384760 DOI: 10.3390/nu15143284] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/12/2023] [Accepted: 07/15/2023] [Indexed: 07/30/2023] Open
Abstract
Human milk is the biological fluid with the highest exosome amount and is rich in microRNAs (miRNAs). These are key regulators of gene expression networks in both normal physiologic and disease contexts, miRNAs can influence many biological processes and have also shown promise as biomarkers for disease. One of the key aspects in the regeneration of the nervous system is that there are practically no molecules that can be used as potential drugs. In the first weeks of lactation, we know that human breast milk must contain the mechanisms to transmit molecular and biological information for brain development. For this reason, our objective is to identify new modulators of the nervous system that can be used to investigate neurodevelopmental functions based on miRNAs. To do this, we collected human breast milk samples according to the time of delivery and milk states: mature milk and colostrum at term; moderate and very preterm mature milk and colostrum; and late preterm mature milk. We extracted exosomes and miRNAs and realized the miRNA functional assays and target prediction. Our results demonstrate that miRNAs are abundant in human milk and likely play significant roles in neurodevelopment and normal function. We found 132 different miRNAs were identified across all samples. Sixty-nine miRNAs had significant differential expression after paired group comparison. These miRNAs are implicated in gene regulation of dopaminergic/glutamatergic synapses and neurotransmitter secretion and are related to the biological process that regulates neuron projection morphogenesis and synaptic vesicle transport. We observed differences according to the delivery time and with less clarity according to the milk type. Our data demonstrate that miRNAs are abundant in human milk and likely play significant roles in neurodevelopment and normal function.
Collapse
Affiliation(s)
- Luis Freiría-Martínez
- Translational Neuroscience Group, Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO (Servizo Galego de Saúde-Universidade de Vigo), 36312 Vigo, Spain
- Department of Functional Biology and Health Sciences, Campus Lagoas Marcosende, Universidade de Vigo, 36310 Vigo, Spain
| | - Marta Iglesias-Martínez-Almeida
- Translational Neuroscience Group, Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO (Servizo Galego de Saúde-Universidade de Vigo), 36312 Vigo, Spain
- Department of Functional Biology and Health Sciences, Campus Lagoas Marcosende, Universidade de Vigo, 36310 Vigo, Spain
| | - Cynthia Rodríguez-Jamardo
- Translational Neuroscience Group, Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO (Servizo Galego de Saúde-Universidade de Vigo), 36312 Vigo, Spain
- Department of Functional Biology and Health Sciences, Campus Lagoas Marcosende, Universidade de Vigo, 36310 Vigo, Spain
| | - Tania Rivera-Baltanás
- Translational Neuroscience Group, Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO (Servizo Galego de Saúde-Universidade de Vigo), 36312 Vigo, Spain
| | - María Comís-Tuche
- Translational Neuroscience Group, Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO (Servizo Galego de Saúde-Universidade de Vigo), 36312 Vigo, Spain
- Department of Functional Biology and Health Sciences, Campus Lagoas Marcosende, Universidade de Vigo, 36310 Vigo, Spain
| | - Daniela Rodrígues-Amorím
- Translational Neuroscience Group, Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO (Servizo Galego de Saúde-Universidade de Vigo), 36312 Vigo, Spain
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Patricia Fernández-Palleiro
- Translational Neuroscience Group, Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO (Servizo Galego de Saúde-Universidade de Vigo), 36312 Vigo, Spain
| | - María Blanco-Formoso
- Department of Physical Chemistry, Singular Center for Biomedical Research (CINBIO), Universidade de Vigo, 36310 Vigo, Spain
| | - Yolanda Diz-Chaves
- Laboratory of Endocrinology, Singular Center for Biomedical Research (CINBIO), Universidade de Vigo, 36310 Vigo, Spain
| | | | - María Suárez-Albo
- Neonatal Intensive Care Unit, Alvaro Cunqueiro Hospital, 36312 Vigo, Spain
| | | | | | | | | | - Jose Manuel Olivares
- Translational Neuroscience Group, Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO (Servizo Galego de Saúde-Universidade de Vigo), 36312 Vigo, Spain
- CIBERSAM (Network Biomedical Research Center on Mental Health), 28029 Madrid, Spain
| | - Carlos Spuch
- Translational Neuroscience Group, Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO (Servizo Galego de Saúde-Universidade de Vigo), 36312 Vigo, Spain
- CIBERSAM (Network Biomedical Research Center on Mental Health), 28029 Madrid, Spain
| |
Collapse
|
38
|
Wang C, Cai Z, Li Z, Liu R. The potential of targeting microRNA-23b-3p signaling in Alzheimer's disease: opinions on recent findings. Front Pharmacol 2023; 14:1245352. [PMID: 37502212 PMCID: PMC10368870 DOI: 10.3389/fphar.2023.1245352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 07/03/2023] [Indexed: 07/29/2023] Open
|
39
|
Hao Z, Ren L, Zhang Z, Yang Z, Wu S, Liu G, Cheng B, Wu J, Xia J. A multifunctional neuromodulation platform utilizing Schwann cell-derived exosomes orchestrates bone microenvironment via immunomodulation, angiogenesis and osteogenesis. Bioact Mater 2023; 23:206-222. [DOI: 10.1016/j.bioactmat.2022.10.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 10/17/2022] [Accepted: 10/18/2022] [Indexed: 11/15/2022] Open
|
40
|
Wang DR, Pan J. Extracellular vesicles: Emerged as a promising strategy for regenerative medicine. World J Stem Cells 2023; 15:165-181. [PMID: 37181006 PMCID: PMC10173817 DOI: 10.4252/wjsc.v15.i4.165] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/30/2023] [Accepted: 03/20/2023] [Indexed: 04/26/2023] Open
Abstract
Cell transplantation therapy has certain limitations including immune rejection and limited cell viability, which seriously hinder the transformation of stem cell-based tissue regeneration into clinical practice. Extracellular vesicles (EVs) not only possess the advantages of its derived cells, but also can avoid the risks of cell transplantation. EVs are intelligent and controllable biomaterials that can participate in a variety of physiological and pathological activities, tissue repair and regeneration by transmitting a variety of biological signals, showing great potential in cell-free tissue regeneration. In this review, we summarized the origins and characteristics of EVs, introduced the pivotal role of EVs in diverse tissues regeneration, discussed the underlying mechanisms, prospects, and challenges of EVs. We also pointed out the problems that need to be solved, application directions, and prospects of EVs in the future and shed new light on the novel cell-free strategy for using EVs in the field of regenerative medicine.
Collapse
Affiliation(s)
- Dian-Ri Wang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Jian Pan
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan Province, China
| |
Collapse
|
41
|
Thompson W, Papoutsakis ET. The role of biomechanical stress in extracellular vesicle formation, composition and activity. Biotechnol Adv 2023; 66:108158. [PMID: 37105240 DOI: 10.1016/j.biotechadv.2023.108158] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 04/23/2023] [Accepted: 04/24/2023] [Indexed: 04/29/2023]
Abstract
Extracellular vesicles (EVs) are cornerstones of intercellular communication with exciting fundamental, clinical, and more broadly biotechnological applications. However, variability in EV composition, which results from the culture conditions used to generate the EVs, poses significant fundamental and applied challenges and a hurdle for scalable bioprocessing. Thus, an understanding of the relationship between EV production (and for clinical applications, manufacturing) and EV composition is increasingly recognized as important and necessary. While chemical stimulation and culture conditions such as cell density are known to influence EV biology, the impact of biomechanical forces on the generation, properties, and biological activity of EVs remains poorly understood. Given the omnipresence of these forces in EV preparation and in biomanufacturing, expanding the understanding of their impact on EV composition-and thus, activity-is vital. Although several publications have examined EV preparation and bioprocessing and briefly discussed biomechanical stresses as variables of interest, this review represents the first comprehensive evaluation of the impact of such stresses on EV production, composition and biological activity. We review how EV biogenesis, cargo, efficacy, and uptake are uniquely affected by various types, magnitudes, and durations of biomechanical forces, identifying trends that emerge both generically and for individual cell types. We also describe implications for scalable bioprocessing, evaluating processes inherent in common EV production and isolation methods, and propose a path forward for rigorous EV quality control.
Collapse
Affiliation(s)
- Will Thompson
- Department of Chemical and Biomolecular Engineering, University of Delaware, 590 Avenue 1743, Newark, DE 19713, USA
| | - Eleftherios Terry Papoutsakis
- Department of Chemical and Biomolecular Engineering, University of Delaware, 590 Avenue 1743, Newark, DE 19713, USA.
| |
Collapse
|
42
|
Wang C, Ji Y, Zhang H, Ye Y, Zhang G, Zhang S, Zhao C, Wang Y. Increased level of exosomal miR-20b-5p derived from hypothermia-treated microglia promotes neurite outgrowth and synapse recovery after traumatic brain injury. Neurobiol Dis 2023; 179:106042. [PMID: 36804284 DOI: 10.1016/j.nbd.2023.106042] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 02/01/2023] [Accepted: 02/12/2023] [Indexed: 02/18/2023] Open
Abstract
Mild hypothermia has been proven to inhibit microglia activation after TBI. Exosomal microRNA derived from microglia played a critical role in promoting neurite outgrowth and synapse recovery. Here, we aimed to investigate the role of microRNAs in microglial exosomes after hypothermia treatment on neuronal regeneration after TBI. For in vitro study, stretch-injured neurons were co-cultured with microglial exosomes. For in vivo study, C57BL/6 mice were under controlled cortical impact and injected with microglial exosomes. The results showed that MG-LPS-EXOHT increased the number of dendrite branches and total length of dendrites both in vitro and in vivo, elevated the expression levels of PSD-95 and GluR1 in stretch-injured neurons, and increased spine density in the pericontusion region. Moreover, MG-LPS-EXOHT improved motor function and motor coordination. A high-throughput sequencing showed that miR-20b-5p was upregulated in MG-LPS-EXOHT. Elevating miR-20b-5p promoted neurite outgrowth and synapse recovery of injured neurons both in vitro and in vivo. Following mechanistic study demonstrated that miR-20b-5p might promote neurite outgrowth and synapse recovery by directly targeting PTEN and activating PI3K-AKT pathway. In conclusion, mild hypothermia could modify the microRNA prolife of exosomes derived from LPS activated BV2 cells. Furthermore, high level of microglial exosomal miR-20b-5p induced by mild hypothermia could transfer into injured neurons and promote neurite outgrowth and synapse recovery after TBI via activating the PI3K-AKT pathway by suppressing PTEN expression.
Collapse
Affiliation(s)
- Chuanfang Wang
- Department of Neurosurgery and Neurosurgical Disease Research Centre, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China; Department of Neurosurgery, the First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong, China
| | - Yunxiang Ji
- Department of Neurosurgery and Neurosurgical Disease Research Centre, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Huabin Zhang
- Department of Neurosurgery and Neurosurgical Disease Research Centre, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yongyi Ye
- Department of Neurosurgery and Neurosurgical Disease Research Centre, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Guilong Zhang
- Department of Neurosurgery and Neurosurgical Disease Research Centre, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Shizhen Zhang
- Department of Neurosurgery and Neurosurgical Disease Research Centre, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Chengcheng Zhao
- Department of Neurosurgery, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, Guangdong, China; Department of Neurosurgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China.
| | - Yezhong Wang
- Department of Neurosurgery and Neurosurgical Disease Research Centre, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
43
|
Pu P, Wu S, Zhang K, Xu H, Guan J, Jin Z, Sun W, Zhang H, Yan B. Mechanical force induces macrophage-derived exosomal UCHL3 promoting bone marrow mesenchymal stem cell osteogenesis by targeting SMAD1. J Nanobiotechnology 2023; 21:88. [PMID: 36915132 PMCID: PMC10012474 DOI: 10.1186/s12951-023-01836-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 03/02/2023] [Indexed: 03/16/2023] Open
Abstract
BACKGROUND Orthodontic tooth movement (OTM), a process of alveolar bone remodelling, is induced by mechanical force and regulated by local inflammation. Bone marrow-derived mesenchymal stem cells (BMSCs) play a fundamental role in osteogenesis during OTM. Macrophages are mechanosensitive cells that can regulate local inflammatory microenvironment and promote BMSCs osteogenesis by secreting diverse mediators. However, whether and how mechanical force regulates osteogenesis during OTM via macrophage-derived exosomes remains elusive. RESULTS Mechanical stimulation (MS) promoted bone marrow-derived macrophage (BMDM)-mediated BMSCs osteogenesis. Importantly, when exosomes from mechanically stimulated BMDMs (MS-BMDM-EXOs) were blocked, the pro-osteogenic effect was suppressed. Additionally, compared with exosomes derived from BMDMs (BMDM-EXOs), MS-BMDM-EXOs exhibited a stronger ability to enhance BMSCs osteogenesis. At in vivo, mechanical force-induced alveolar bone formation was impaired during OTM when exosomes were blocked, and MS-BMDM-EXOs were more effective in promoting alveolar bone formation than BMDM-EXOs. Further proteomic analysis revealed that ubiquitin carboxyl-terminal hydrolase isozyme L3 (UCHL3) was enriched in MS-BMDM-EXOs compared with BMDM-EXOs. We went on to show that BMSCs osteogenesis and mechanical force-induced bone formation were impaired when UCHL3 was inhibited. Furthermore, mothers against decapentaplegic homologue 1 (SMAD1) was identified as the target protein of UCHL3. At the mechanistic level, we showed that SMAD1 interacted with UCHL3 in BMSCs and was downregulated when UCHL3 was suppressed. Consistently, overexpression of SMAD1 rescued the adverse effect of inhibiting UCHL3 on BMSCs osteogenesis. CONCLUSIONS This study suggests that mechanical force-induced macrophage-derived exosomal UCHL3 promotes BMSCs osteogenesis by targeting SMAD1, thereby promoting alveolar bone formation during OTM.
Collapse
Affiliation(s)
- Panjun Pu
- Department of Orthodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, 210000, China
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, 210000, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, 210000, China
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Shengnan Wu
- Department of Orthodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, 210000, China
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, 210000, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, 210000, China
| | - Kejia Zhang
- Department of Orthodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, 210000, China
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, 210000, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, 210000, China
| | - Hao Xu
- Department of Orthodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, 210000, China
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, 210000, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, 210000, China
| | - Jiani Guan
- Department of Orthodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, 210000, China
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, 210000, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, 210000, China
| | - Zhichun Jin
- Department of Orthodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, 210000, China
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, 210000, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, 210000, China
| | - Wen Sun
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, 210000, China
| | - Hanwen Zhang
- School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 210000, China.
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, 210000, China.
| | - Bin Yan
- Department of Orthodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, 210000, China.
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, 210000, China.
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, 210000, China.
| |
Collapse
|
44
|
Fan Z, Wen X, Ding X, Wang Q, Wang S, Yu W. Advances in biotechnology and clinical therapy in the field of peripheral nerve regeneration based on magnetism. Front Neurol 2023; 14:1079757. [PMID: 36970536 PMCID: PMC10036769 DOI: 10.3389/fneur.2023.1079757] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 02/07/2023] [Indexed: 03/12/2023] Open
Abstract
Peripheral nerve injury (PNI) is one of the most common neurological diseases. Recent studies on nerve cells have provided new ideas for the regeneration of peripheral nerves and treatment of physical trauma or degenerative disease-induced loss of sensory and motor neuron functions. Accumulating evidence suggested that magnetic fields might have a significant impact on the growth of nerve cells. Studies have investigated different magnetic field properties (static or pulsed magnetic field) and intensities, various magnetic nanoparticle-encapsulating cytokines based on superparamagnetism, magnetically functionalized nanofibers, and their relevant mechanisms and clinical applications. This review provides an overview of these aspects as well as their future developmental prospects in related fields.
Collapse
|
45
|
Wang Y, Zhang S, Zhi J, Huang M, Pei F. A bibliometric analysis: Current status and frontier trends of Schwann cells in neurosciences. Front Mol Neurosci 2023; 15:1087550. [PMID: 36710927 PMCID: PMC9877341 DOI: 10.3389/fnmol.2022.1087550] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 12/15/2022] [Indexed: 01/15/2023] Open
Abstract
Background This review aims to present a comprehensive bibliometric analysis related to Schwann cells (SCs) in neurosciences from 2012 to 2021. Methods We used the Web of Science core collection database to obtain publications on SCs in the field of neurosciences from 2012 to 2021. The obtained data were further visually analyzed by using CiteSpace, VOSviewer, and an online bibliometric platform. Results We retrieved a total of 1,923 publications related to SCs in neurosciences. The number of publications in this field fluctuates steadily each year, and the number of citations is increasing year by year. The United States is leading the field, with LERU and the University OF London as influential institutions, Jessen KR and Feltri ML as the most representative authors, and GLIA and JOURNAL OF NEUROSCIENCE as authoritative journals in the field. Meanwhile, we predict that a more in-depth study of autophagy and phagocytosis functions of SCs and the key regulator c-Jun will probably be a hot spot for future research. Conclusion This study summarizes the current research results and predicts research trends for further research, which will facilitate researchers in quickly understanding the current state of research in the field while referring to new research directions.
Collapse
Affiliation(s)
- Yan Wang
- Rehabilitation Center of the Second Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China,*Correspondence: Yan Wang,
| | - Shiwen Zhang
- Graduate School of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Jincao Zhi
- Graduate School of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Meiling Huang
- Graduate School of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Fei Pei
- Graduate School of Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
46
|
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: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [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
|
47
|
Liu Z, Wang S, Huo N, Yang S, Shi Q, Xu J. Extracellular vesicles: A potential future strategy for dental and maxillofacial tissue repair and regeneration. Front Physiol 2022; 13:1012241. [PMID: 36479350 PMCID: PMC9719951 DOI: 10.3389/fphys.2022.1012241] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 11/09/2022] [Indexed: 06/18/2024] Open
Abstract
Extracellular vesicles (EVs), nano-sized bilayer membrane structures containing lipids, proteins and nucleic acids, play key roles in intercellular communication. Compared to stem cells, EVs have lower tumorigenicity and immunogenicity, are easier to manage and cause fewer ethic problems. In recent years, EVs have emerged as a potential solution for tissue regeneration in stomatology through cell-free therapies. The present review focuses on the role of EVs in dental and maxillofacial tissue repair and regeneration, including in dental and periodontal tissue, maxilla and mandible bone, temporomandibular joint cartilage, peripheral nerve and soft tissue. We also make a brief overview on the mechanism of EVs performing functions. However, limitations and challenges in clinical application of EVs still exist and should be addressed in future researches.
Collapse
Affiliation(s)
- Ziwei Liu
- Medical School of Chinese PLA, Beijing, China
- Department of Stomatology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
- Orthopedic Laboratory of PLA General Hospital, Beijing, China
| | - Situo Wang
- Medical School of Chinese PLA, Beijing, China
- Department of Stomatology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
- Orthopedic Laboratory of PLA General Hospital, Beijing, China
| | - Na Huo
- Department of Stomatology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Shuo Yang
- Department of Stomatology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Quan Shi
- Department of Stomatology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Juan Xu
- Department of Stomatology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
48
|
Pan B, Guo D, Jing L, Li K, Li X, Li G, Gao X, Li ZW, Zhao W, Feng H, Cao MH. Long noncoding RNA Pvt1 promotes the proliferation and migration of Schwann cells by sponging microRNA-214 and targeting c-Jun following peripheral nerve injury. Neural Regen Res 2022; 18:1147-1153. [PMID: 36255005 PMCID: PMC9827779 DOI: 10.4103/1673-5374.353497] [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] [Indexed: 01/11/2023] Open
Abstract
Research has shown that long-chain noncoding RNAs (lncRNAs) are involved in the regulation of a variety of biological processes, including peripheral nerve regeneration, in part by acting as competing endogenous RNAs. c-Jun plays a key role in the repair of peripheral nerve injury. However, the precise underlying mechanism of c-Jun remains unclear. In this study, we performed microarray and bioinformatics analysis of mouse crush-injured sciatic nerves and found that the lncRNA Pvt1 was overexpressed in Schwann cells after peripheral nerve injury. Mechanistic studies revealed that Pvt1 increased c-Jun expression through sponging miRNA-214. We overexpressed Pvt1 in Schwann cells cultured in vitro and found that the proliferation and migration of Schwann cells were enhanced, and overexpression of miRNA-214 counteracted the effects of Pvt1 overexpression on Schwann cell proliferation and migration. We conducted in vivo analyses and injected Schwann cells overexpressing Pvt1 into injured sciatic nerves of mice. Schwann cells overexpressing Pvt1 enhanced the regeneration of injured sciatic nerves following peripheral nerve injury and the locomotor function of mice was improved. Our findings reveal the role of lncRNAs in the repair of peripheral nerve injury and highlight lncRNA Pvt1 as a novel potential treatment target for peripheral nerve injury.
Collapse
Affiliation(s)
- Bin Pan
- Department of Orthopedics, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Di Guo
- Department of Orthopedics, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Li Jing
- Department of Orthopedics, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Ke Li
- Department of Imaging, Xuzhou Central Hospital, Xuzhou, Jiangsu Province, China
| | - Xin Li
- Department of Orthopedics, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Gen Li
- Department of Orthopedics, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Xiao Gao
- Department of Orthopedics, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Zhi-Wen Li
- College of Extended Education, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Wei Zhao
- Department of Orthopedics, Kuitun Hospital, Yili Kazak Autonomous Prefecture, Xinjiang Uygur Autonomous Region, China
| | - Hu Feng
- Department of Orthopedics, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, China,Correspondence to: Meng-Han Cao, ; Hu Feng, .
| | - Meng-Han Cao
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, China,Correspondence to: Meng-Han Cao, ; Hu Feng, .
| |
Collapse
|
49
|
Qian T, Qiao P, Lu Y, Wang H. Transcription factor SS18L1 regulates the proliferation, migration and differentiation of Schwann cells in peripheral nerve injury. Front Vet Sci 2022; 9:936620. [PMID: 36046506 PMCID: PMC9420995 DOI: 10.3389/fvets.2022.936620] [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: 05/10/2022] [Accepted: 07/27/2022] [Indexed: 11/30/2022] Open
Abstract
Transcription factors bind to specific DNA sequences, modulate the transcription of target genes, and regulate various biological processes, including peripheral nerve regeneration. Our previous analysis showed that SS18L1, a gene encoding the transcription factor SS18-like protein 1, was differentially expressed in the distal sciatic nerve stumps after rat sciatic nerve transection injury, but its effect on peripheral nerve injury has not been reported. In the current study, we isolated and cultured primary Schwann cells, and examined the role of SS18L1 for the biological functions of the cells. Depletion of SS18L1 by siRNA in Schwann cells enhanced cell proliferation and inhibited cell migration, as determined by EdU assay and transwell migration assay, respectively. In addition, silencing of SS18L1 inhibited Schwann cell differentiation induced by HRG and cAMP. Bioinformatics analyses revealed an interaction network of SS18L1, including DF2, SMARCD1, SMARCA4, and SMARCE1, which may be implicated in the regulatory functions of SS18L1 on the proliferation, migration and differentiation of Schwann cells. In conclusion, our results revealed a temporal expression profile of SS18L1 in peripheral nerve injury and its potential roles during the process of nerve recovery.
Collapse
Affiliation(s)
- Tianmei Qian
- Suzhou Medical College of Soochow University, Suzhou, China
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| | - Pingping Qiao
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| | - Yingnan Lu
- School of Overseas Education, Changzhou University, Changzhou, China
| | - Hongkui Wang
- Suzhou Medical College of Soochow University, Suzhou, China
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| |
Collapse
|
50
|
Kong L, Gao X, Qian Y, Sun W, You Z, Fan C. Biomechanical microenvironment in peripheral nerve regeneration: from pathophysiological understanding to tissue engineering development. Am J Cancer Res 2022; 12:4993-5014. [PMID: 35836812 PMCID: PMC9274750 DOI: 10.7150/thno.74571] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Accepted: 06/11/2022] [Indexed: 01/12/2023] Open
Abstract
Peripheral nerve injury (PNI) caused by trauma, chronic disease and other factors may lead to partial or complete loss of sensory, motor and autonomic functions, as well as neuropathic pain. Biological activities are always accompanied by mechanical stimulation, and biomechanical microenvironmental homeostasis plays a complicated role in tissue repair and regeneration. Recent studies have focused on the effects of biomechanical microenvironment on peripheral nervous system development and function maintenance, as well as neural regrowth following PNI. For example, biomechanical factors-induced cluster gene expression changes contribute to formation of peripheral nerve structure and maintenance of physiological function. In addition, extracellular matrix and cell responses to biomechanical microenvironment alterations after PNI directly trigger a series of cascades for the well-organized peripheral nerve regeneration (PNR) process, where cell adhesion molecules, cytoskeletons and mechanically gated ion channels serve as mechanosensitive units, mechanical effector including focal adhesion kinase (FAK) and yes-associated protein (YAP)/transcriptional coactivator with PDZ-binding motif (TAZ) as mechanotransduction elements. With the rapid development of tissue engineering techniques, a substantial number of PNR strategies such as aligned nerve guidance conduits, three-dimensional topological designs and piezoelectric scaffolds emerge expected to improve the neural biomechanical microenvironment in case of PNI. These tissue engineering nerve grafts display optimized mechanical properties and outstanding mechanomodulatory effects, but a few bottlenecks restrict their application scenes. In this review, the current understanding in biomechanical microenvironment homeostasis associated with peripheral nerve function and PNR is integrated, where we proposed the importance of balances of mechanosensitive elements, cytoskeletal structures, mechanotransduction cascades, and extracellular matrix components; a wide variety of promising tissue engineering strategies based on biomechanical modulation are introduced with some suggestions and prospects for future directions.
Collapse
Affiliation(s)
- Lingchi Kong
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China.,Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai, 200233, China.,Youth Science and Technology Innovation Studio of Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Xin Gao
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Belt and Road Joint Laboratory of Advanced Fiber and Low-dimension Materials, College of Materials Science and Engineering, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, Donghua University, Shanghai, 201620, China
| | - Yun Qian
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China.,Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai, 200233, China.,Youth Science and Technology Innovation Studio of Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.,✉ Corresponding authors: Yun Qian, E-mail: ; Wei Sun, E-mail: ; Zhengwei You, E-mail: ; Cunyi Fan, E-mail:
| | - Wei Sun
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Belt and Road Joint Laboratory of Advanced Fiber and Low-dimension Materials, College of Materials Science and Engineering, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, Donghua University, Shanghai, 201620, China.,✉ Corresponding authors: Yun Qian, E-mail: ; Wei Sun, E-mail: ; Zhengwei You, E-mail: ; Cunyi Fan, E-mail:
| | - Zhengwei You
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Belt and Road Joint Laboratory of Advanced Fiber and Low-dimension Materials, College of Materials Science and Engineering, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, Donghua University, Shanghai, 201620, China.,✉ Corresponding authors: Yun Qian, E-mail: ; Wei Sun, E-mail: ; Zhengwei You, E-mail: ; Cunyi Fan, E-mail:
| | - Cunyi Fan
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China.,Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai, 200233, China.,Youth Science and Technology Innovation Studio of Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.,✉ Corresponding authors: Yun Qian, E-mail: ; Wei Sun, E-mail: ; Zhengwei You, E-mail: ; Cunyi Fan, E-mail:
| |
Collapse
|