1
|
Zheng K, Wang S, Deng M, Luo Y, Li W, Zeng L, Wang Y. Mechanisms and Therapeutic Strategies of Macrophage Polarization in Intervertebral Disc Degeneration. JOR Spine 2025; 8:e70065. [PMID: 40371270 PMCID: PMC12077540 DOI: 10.1002/jsp2.70065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 03/03/2025] [Accepted: 03/26/2025] [Indexed: 05/16/2025] Open
Abstract
Background Intervertebral disc degeneration (IVDD) is a leading cause of low back pain (LBP), contributing significantly to global disability and productivity loss. Its pathogenesis involves complex processes, including inflammation, cellular senescence, angiogenesis, fibrosis, neural ingrowth, and sensitization. Emerging evidence highlights macrophages as central immune regulators infiltrating degenerated discs, with macrophage polarization implicated in IVDD progression. However, the mechanisms linking macrophage polarization to IVDD pathology remain poorly elucidated. Methods A comprehensive literature review was conducted by searching major databases (PubMed, Web of Science, and Scopus) for studies published in the last decade (2014-2024). Keywords included "intervertebral disc degeneration," "macrophage polarization," "inflammation," "senescence," and "therapeutic strategies." Relevant articles were selected, analyzed, and synthesized to evaluate the role of macrophage polarization in IVDD. Results Macrophage polarization dynamically influences IVDD through multiple pathways. Pro-inflammatory M1 macrophages exacerbate disc degeneration by amplifying inflammatory cytokines (e.g., TNF-α, IL-1β), promoting cellular senescence, and stimulating abnormal angiogenesis and neural ingrowth. In contrast, anti-inflammatory M2 macrophages may mitigate degeneration by suppressing inflammation and enhancing tissue repair. Therapeutic strategies targeting macrophage polarization include pharmacological agents (e.g., cytokines, small-molecule inhibitors), biologic therapies, gene editing, and physical interventions. Challenges persist, such as incomplete understanding of polarization triggers, lack of targeted delivery systems, and limited translational success in preclinical models. Conclusion Macrophage polarization is a pivotal regulator of IVDD pathology, offering promising therapeutic targets. Future research should focus on elucidating polarization mechanisms, optimizing spatiotemporal control of macrophage phenotypes, and developing personalized therapies. Addressing these challenges may advance innovative strategies to halt or reverse IVDD progression, ultimately improving clinical outcomes for LBP patients.
Collapse
Affiliation(s)
- Kaiyuan Zheng
- Department of Rehabilitation Medicine, Intensive Care MedicineAffiliated Hospital of North Sichuan Medical CollegeNanchongChina
| | - Siyu Wang
- Department of Rehabilitation Medicine, Intensive Care MedicineAffiliated Hospital of North Sichuan Medical CollegeNanchongChina
| | - Meng Deng
- Department of Clinical LaboratoryThe First People's Hospital of GuangyuanGuangyuanChina
| | - Yaomin Luo
- Department of Rehabilitation Medicine, Intensive Care MedicineAffiliated Hospital of North Sichuan Medical CollegeNanchongChina
| | - Wen Li
- Department of Rehabilitation Medicine, Intensive Care MedicineAffiliated Hospital of North Sichuan Medical CollegeNanchongChina
| | - Lianlin Zeng
- Department of Rehabilitation MedicineSuining Central HospitalSuiningChina
| | - Yinxu Wang
- Department of Rehabilitation Medicine, Intensive Care MedicineAffiliated Hospital of North Sichuan Medical CollegeNanchongChina
| |
Collapse
|
2
|
Li K, Lin H, Yu Y, Liu Y, Yang W, Chen S, Xu L, Huang W, Wang H, Meng C, Shao Z, Wei Y, Zhao L, Peng Y. Nucleus pulposus cell-mimicking nanoparticles for cell-specific HIF1A editing to modulate SASP-mediated disc inflammation via autophagy activation. Acta Biomater 2025; 197:357-373. [PMID: 40087134 DOI: 10.1016/j.actbio.2025.02.060] [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: 10/28/2024] [Revised: 02/20/2025] [Accepted: 02/26/2025] [Indexed: 03/16/2025]
Abstract
Traditional methods of plasmid delivery, including viral vectors, lipofection, and electroporation, are widely used for gene editing but have limitations, such as cellular toxicity, limited transfection efficiency in primary cells, and nonspecific side effects. Here, we report the development of nucleus pulposus cell (NPC)-mimicking nanoparticles (HIF1A@NNP) with an NPC membrane as the shell and pcDNA3.1+-rHIF1A encapsulated in the core via extrusion. HIF1A@NNP exhibited a protein expression pattern similar to that of the NPC membrane and displayed a typical vesicle profile. Compared to liposomes and lentiviruses, HIF1A@NNP overexpressed HIF1A in NPCs while improving cell viability. HIF1A@NNP was more readily internalized by NPCs than by other cell types, with fewer effects on vascularization, nerve growth, and macrophage polarization than HIF1A overexpression using lipo3000. HIF1A@NNP reduced the apoptotic rate and inhibited the senescent phenotype, as evidenced by reduced DNA damage, lower levels of senescence-related proteins, and fewer SA-β-Gal-positive cells. HIF1A@NNP induced a senescence-associated secretory phenotype (SASP), which enhanced macrophage migration and M1 polarization. Additionally, HIF1A@NNP activated autophagy in NPCs. In summary, HIF1A@NNP demonstrated satisfactory biocompatibility, alleviated the SASP, and inhibited SASP-mediated macrophage recruitment and inflammatory polarization, leading to reduced disc degeneration and providing a promising strategy for combating intervertebral disc degeneration. STATEMENT OF SIGNIFICANCE: Conventional plasmid delivery methods like viral vectors, lipofection, and electroporation struggle with cellular toxicity and inefficiency in primary cells. Non-cell-specific HIF1A activation via these methods may exacerbate inflammation and pain, as HIF1A drives angiogenesis and dendritic ingrowth into the disc. Thus, a cell-specific delivery strategy could circumvent such adverse effects. Our study introduces HIF1A@NNP, a nanoparticle mimicking nucleus pulposus cells (NPCs), with an NPC membrane shell encapsulating pcDNA3.1+-rHIF1A. It preferentially targets NPCs, achieving superior HIF1A overexpression and cell viability compared to liposomes and lentiviruses. This represents a highly promising and potentially transformative approach against intervertebral disc degeneration.
Collapse
Affiliation(s)
- Kanglu Li
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Hui Lin
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yihan Yu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yiran Liu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Wenbo Yang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Songfeng Chen
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Li Xu
- Department of Emergency, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Wei Huang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Hong Wang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Chunqing Meng
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zengwu Shao
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yulong Wei
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Lei Zhao
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Yizhong Peng
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
3
|
Jin Y, Wu O, Chen Z, Chen L, Zhang K, Chen Q, Tian H, Wang X, Jones M, Kwan KYH, Li YM, Makvandi P, Wang X, Hai X, Zhang J, Wu A. Exploring extracellular vesicles as novel therapeutic agents for intervertebral disc degeneration: delivery, applications, and mechanisms. Stem Cell Res Ther 2025; 16:221. [PMID: 40312404 PMCID: PMC12044939 DOI: 10.1186/s13287-025-04299-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 04/01/2025] [Indexed: 05/03/2025] Open
Abstract
Intervertebral disc degeneration is a multifactorial degenerative disease that poses a significant threat to the health of the elderly population. Current treatments primarily focus on physical therapy, medication, and surgery to alleviate symptoms associated with disc compression but do not address the progression of degeneration. Therefore, this review aimed to explore the potential of extracellular vesicle therapy as a novel preventive strategy to delay degeneration and enhance tissue repair in intervertebral discs. We cover the pathogenic mechanisms underlying intervertebral disc degeneration, including inflammation, apoptosis, pyroptosis, ferroptosis, autophagy dysregulation, and the roles of non-coding RNAs. Subsequently, we discussed the therapeutic potential of extracellular vesicles and their molecular components, such as proteins, RNAs, and lipids, in modulating these pathways to counter intervertebral disc degeneration. We provides a comprehensive review of the significant role of extracellular vesicle cargo in mediating repair mechanisms. It discusses the functional enhancement advantages exhibited by extracellular vesicles under current bioengineering modifications and drug loading. The challenges and future prospects of utilizing extracellular vesicle therapy to treat this degenerative condition are also summarized.
Collapse
Affiliation(s)
- Yuxin Jin
- Department of Orthopaedics, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Ouqiang Wu
- Department of Orthopaedics, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Zhihua Chen
- Department of Orthopaedics, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Linjie Chen
- Department of Orthopaedics, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Kai Zhang
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedics, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qizhu Chen
- Department of Clinic of Spine Center, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200082, China
| | - Haijun Tian
- Department of Orthopaedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xinzhou Wang
- Department of Orthopaedics, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Morgan Jones
- Spine Unit, The Royal Orthopaedic Hospital, Bristol Road South, Northfield, Birmingham, B31 2AP, UK
| | - Kenny Yat Hong Kwan
- Department of Orthopaedics and Traumatology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 5/F Professorial Block, Queen Mary Hospital, 102 Pokfulam Road, Pokfulam, Hong Kong, China
| | - Yan Michael Li
- Department of Neurosurgery, University of Rochester Medical Center, 601 Elm-Wood Ave, Rochester, NY, 14642, USA
| | - Pooyan Makvandi
- University Centre for Research & Development, Chandigarh University, Mohali, 140413, Punjab, India
- Centre for Research Impact & Outcome, Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
| | - Xiangyang Wang
- Department of Orthopaedics, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Xiang Hai
- Ecological-Environment & Health College (EEHC), Zhejiang A & F University, Hangzhou, 311300, Zhejiang, China.
| | - Jun Zhang
- Department of Orthopedics, Zhejiang Provincial People's Hospital Bijie Hospital, Bijie, Guizhou, China.
| | - Aimin Wu
- Department of Orthopaedics, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.
| |
Collapse
|
4
|
Li L, Al‐Jallad H, Sun A, Georgiopoulos M, Bokhari R, Ouellet J, Jarzem P, Cherif H, Haglund L. The proteomic landscape of extracellular vesicles derived from human intervertebral disc cells. JOR Spine 2024; 7:e70007. [PMID: 39507593 PMCID: PMC11538033 DOI: 10.1002/jsp2.70007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 09/17/2024] [Accepted: 10/05/2024] [Indexed: 11/08/2024] Open
Abstract
Background Extracellular vesicles (EVs) function as biomarkers and are crucial in cell communication and regulation, with therapeutic potential for intervertebral disc (IVD)-related low back pain (LBP). EV cargo is often affected by tissue health, which may affect the therapeutic potential. There is currently limited knowledge of how the cargo of IVD cell-derived EVs varies with tissue health and how differences in proteomic profile affect the predicted biological functions. Methods Our study purified EVs from human IVD cell conditioned media by size-exclusion chromatography. Nanoparticle tracking analysis was conducted to measure EV size and concentration. Transmission electron microscopy and Western blot were performed to examine EV structure and markers. Tandem mass tag-mass spectrometry was conducted to determine protein cargo. Results Most EVs were exosomes and intermediate microvesicles with an increasing amount linked to disease progression. Of the proteins detected, 88.6% were shared across the non-degenerate, mildly-degenerate, and degenerate samples. GO and KEGG analyses revealed that cargo from the mildly-degenerate samples was the most distinct, with the proteins in high abundance strongly associated with extracellular matrix (ECM) organization and structure. Shared proteins, highly expressed in the non-degenerate and degenerate samples, showed strong associations with cell adhesion, ECM-receptor interaction, and vesicle-mediated transport, respectively. Conclusions Our findings indicate that EVs from IVD cells from tissue with different degrees of degeneration share a majority of the cargo proteins. However, the level of expression differs with degeneration grade. Cargo from the mildly-degenerate samples exhibits the most differences. A better understanding of changes in EV cargo in the degenerative process may provide novel information related to molecular mechanisms underlying IVD degeneration and suggest new potential treatment modalities for IVD-related LBP.
Collapse
Affiliation(s)
- Li Li
- Department of Surgery, Division of OrthopaedicsMcGill UniversityMontrealQuebecCanada
- The McGill Scoliosis and Spine Group, McGill University Health CentreMontrealQuebecCanada
- The Orthopaedic Research Laboratory, Research Institute of the McGill University Health CentreMontrealQuebecCanada
| | | | - Aiwei Sun
- Department of Anatomy and Cell BiologyMcGill UniversityMontrealQuebecCanada
| | - Miltiadis Georgiopoulos
- Department of Surgery, Division of OrthopaedicsMcGill UniversityMontrealQuebecCanada
- The McGill Scoliosis and Spine Group, McGill University Health CentreMontrealQuebecCanada
| | - Rakan Bokhari
- Department of Surgery, Division of OrthopaedicsMcGill UniversityMontrealQuebecCanada
- Department of Surgery, Division of NeurosurgeryFaculty of Medicine, King Abdulaziz UniversityJeddahSaudi Arabia
| | - Jean Ouellet
- Department of Surgery, Division of OrthopaedicsMcGill UniversityMontrealQuebecCanada
- The McGill Scoliosis and Spine Group, McGill University Health CentreMontrealQuebecCanada
- Shriners Hospital for ChildrenMontrealQuebecCanada
| | - Peter Jarzem
- Department of Surgery, Division of OrthopaedicsMcGill UniversityMontrealQuebecCanada
- The McGill Scoliosis and Spine Group, McGill University Health CentreMontrealQuebecCanada
| | - Hosni Cherif
- Department of Surgery, Division of OrthopaedicsMcGill UniversityMontrealQuebecCanada
- The McGill Scoliosis and Spine Group, McGill University Health CentreMontrealQuebecCanada
- The Orthopaedic Research Laboratory, Research Institute of the McGill University Health CentreMontrealQuebecCanada
| | - Lisbet Haglund
- Department of Surgery, Division of OrthopaedicsMcGill UniversityMontrealQuebecCanada
- The McGill Scoliosis and Spine Group, McGill University Health CentreMontrealQuebecCanada
- The Orthopaedic Research Laboratory, Research Institute of the McGill University Health CentreMontrealQuebecCanada
- Shriners Hospital for ChildrenMontrealQuebecCanada
| |
Collapse
|
5
|
Sheng L, Xu H, Wang Y, Ni J, Xiang T, Xu H, Zhou X, Wei K, Dai J. Systematic analysis of lysine lactylation in nucleus pulposus cells. iScience 2024; 27:111157. [PMID: 39524337 PMCID: PMC11546124 DOI: 10.1016/j.isci.2024.111157] [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: 12/13/2023] [Revised: 05/28/2024] [Accepted: 10/09/2024] [Indexed: 11/16/2024] Open
Abstract
Nucleus pulposus (NP) resides in hypoxic microenvironment and NP cells (NPCs), primarily reply on glycolysis and producing high levels of lactate. Intracellular lactate drives lysine lactylation (Kla) as a newly epigenetic modification. However, the impact of Kla on NPCs remains unknown. Here, single-cell RNA sequencing (scRNA-seq) data suggested an altered balance between glycolysis and aerobic oxidation in intervertebral disc degeneration (IDD). Liquid chromatography-tandem mass spectrometry (LC-MS/MS) analysis displayed 3510 lactylation sites on 1052 non-histone proteins of NPCs isolated from rat cultured in normoxia and hypoxia. Moreover, there are 18 proteins with 129 Kla sites and 117 Kla sites in 27 proteins exclusively detected in normoxia and hypoxia group, respectively. Bioinformatics analysis displayed that these lactylated proteins are tightly related to ribosome, spliceosome and the VEGFA-VEGFA2 signaling pathway. Together, our study reveals that Kla may play an important role in regulating cellular metabolism of NPCs.
Collapse
Affiliation(s)
- Lei Sheng
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, China
| | - Haoran Xu
- Department of Joint Surgery, Center for Orthopedic Surgery, Orthopedic Hospital of Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong 510000, China
| | - Yuexing Wang
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, China
| | - Jinhao Ni
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, China
| | - Taiyang Xiang
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, China
| | - Huanhuan Xu
- Department of Obstetrics and Gynecology, Wuhan Children’s Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430000, China
| | - Xiaozhong Zhou
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, China
| | - Kang Wei
- Department of Plastic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, China
| | - Jun Dai
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, China
| |
Collapse
|
6
|
Zhang S, Tong M, Li S, Zhang B, Zhang W, Wang R, Dong Z, Huang Y. The Role of Microvascular Variations in the Process of Intervertebral Disk Degeneration and Its Regulatory Mechanisms: A Literature Review. Orthop Surg 2024; 16:2587-2597. [PMID: 39205477 PMCID: PMC11541140 DOI: 10.1111/os.14209] [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: 03/11/2024] [Revised: 08/01/2024] [Accepted: 08/06/2024] [Indexed: 09/04/2024] Open
Abstract
Microvascular changes are considered key factors in the process of intervertebral disk degeneration (IDD). Microvascular invasion and growth into the nucleus pulposus (NP) and cartilaginous endplates are unfavorable factors that trigger IDD. In contrast, the rich distribution of microvessels in the bony endplates and outer layers of the annulus fibrosus is an important safeguard for the nutrient supply and metabolism of the intervertebral disk (IVD). In particular, the adequate supply of microvessels in the bony endplates is the main source of the nutritional supply for the entire IVD. Microvessels can affect the progression of IDD through a variety of pathways. Many studies have explored the effects of microvessel alterations in the NP, annulus fibrosus, cartilaginous endplates, and bony endplates on the local microenvironment through inflammation, apoptosis, and senescence. Studies also elucidated the important roles of microvessel alterations in the process of IDD, as well as conducted in-depth explorations of cytokines and biologics that can inhibit or promote the ingrowth of microvessels. Therefore, the present manuscript reviews the published literature on the effects of microvascular changes on IVD to summarize the roles of microvessels in IVD and elaborate on the mechanisms of action that promote or inhibit de novo microvessel formation in IVD.
Collapse
Affiliation(s)
- Si‐Ping Zhang
- Department of Spinal SurgeryTraditional Chinese Medicine Hospital affiliated to Xinjiang Medical UniversityUrumqiChina
- Xinjiang Uygur Autonomous Region Academy of Traditional Chinese MedicineUrumqiChina
| | - Min Tong
- Department of Spinal SurgeryTraditional Chinese Medicine Hospital affiliated to Xinjiang Medical UniversityUrumqiChina
- Xinjiang Uygur Autonomous Region Academy of Traditional Chinese MedicineUrumqiChina
| | - Shi‐Da Li
- Department of Spinal SurgeryTraditional Chinese Medicine Hospital affiliated to Xinjiang Medical UniversityUrumqiChina
- Xinjiang Uygur Autonomous Region Academy of Traditional Chinese MedicineUrumqiChina
| | - Bin Zhang
- Department of Spinal SurgeryTraditional Chinese Medicine Hospital affiliated to Xinjiang Medical UniversityUrumqiChina
- Xinjiang Uygur Autonomous Region Academy of Traditional Chinese MedicineUrumqiChina
| | - Wenhao Zhang
- Department of Spinal SurgeryTraditional Chinese Medicine Hospital affiliated to Xinjiang Medical UniversityUrumqiChina
- Xinjiang Uygur Autonomous Region Academy of Traditional Chinese MedicineUrumqiChina
| | - Rong Wang
- Department of Spinal SurgeryTraditional Chinese Medicine Hospital affiliated to Xinjiang Medical UniversityUrumqiChina
- Xinjiang Uygur Autonomous Region Academy of Traditional Chinese MedicineUrumqiChina
| | - Zhen‐Yu Dong
- Department of Spinal SurgeryTraditional Chinese Medicine Hospital affiliated to Xinjiang Medical UniversityUrumqiChina
- Xinjiang Uygur Autonomous Region Academy of Traditional Chinese MedicineUrumqiChina
| | - Yi‐Fei Huang
- Department of Spinal SurgeryTraditional Chinese Medicine Hospital affiliated to Xinjiang Medical UniversityUrumqiChina
- Xinjiang Uygur Autonomous Region Academy of Traditional Chinese MedicineUrumqiChina
| |
Collapse
|
7
|
Elmounedi N, Bahloul W, Keskes H. Current Therapeutic Strategies of Intervertebral Disc Regenerative Medicine. Mol Diagn Ther 2024; 28:745-775. [PMID: 39158834 DOI: 10.1007/s40291-024-00729-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/30/2024] [Indexed: 08/20/2024]
Abstract
Intervertebral disc degeneration (IDD) is one of the most frequent causes of low back pain. No treatment is currently available to delay the progression of IDD. Conservative treatment or surgical interventions is only used to target the symptoms of IDD rather than treat the underlying cause. Currently, numerous potential therapeutic strategies are available, including molecular therapy, gene therapy, and cell therapy. However, the hostile environment of degenerated discs is a major problem that has hindered the clinical applicability of such approaches. In this regard, the design of drugs using alternative delivery systems (macro-, micro-, and nano-sized particles) may resolve this problem. These can protect and deliver biomolecules along with helping to improve the therapeutic effect of drugs via concentrating, protecting, and prolonging their presence in the degenerated disc. This review summarizes the research progress of diagnosis and the current options for treating IDD.
Collapse
Affiliation(s)
- Najah Elmounedi
- Cell Therapy and Experimental Surgery of Musculoskeletal System LR18SP11 Lab, Sfax Faculty of Medicine, Majida Boulila Road, 3029, Sfax, Tunisia.
| | - Walid Bahloul
- Cell Therapy and Experimental Surgery of Musculoskeletal System LR18SP11 Lab, Sfax Faculty of Medicine, Majida Boulila Road, 3029, Sfax, Tunisia
- Department of Orthopedics and Traumatology, CHU Habib Bourguiba, Sfax, Tunisia
| | - Hassib Keskes
- Cell Therapy and Experimental Surgery of Musculoskeletal System LR18SP11 Lab, Sfax Faculty of Medicine, Majida Boulila Road, 3029, Sfax, Tunisia
- Department of Orthopedics and Traumatology, CHU Habib Bourguiba, Sfax, Tunisia
| |
Collapse
|
8
|
Moghassemi S, Dadashzadeh A, Sousa MJ, Vlieghe H, Yang J, León-Félix CM, Amorim CA. Extracellular vesicles in nanomedicine and regenerative medicine: A review over the last decade. Bioact Mater 2024; 36:126-156. [PMID: 38450204 PMCID: PMC10915394 DOI: 10.1016/j.bioactmat.2024.02.021] [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: 12/01/2023] [Revised: 02/15/2024] [Accepted: 02/19/2024] [Indexed: 03/08/2024] Open
Abstract
Small extracellular vesicles (sEVs) are known to be secreted by a vast majority of cells. These sEVs, specifically exosomes, induce specific cell-to-cell interactions and can activate signaling pathways in recipient cells through fusion or interaction. These nanovesicles possess several desirable properties, making them ideal for regenerative medicine and nanomedicine applications. These properties include exceptional stability, biocompatibility, wide biodistribution, and minimal immunogenicity. However, the practical utilization of sEVs, particularly in clinical settings and at a large scale, is hindered by the expensive procedures required for their isolation, limited circulation lifetime, and suboptimal targeting capacity. Despite these challenges, sEVs have demonstrated a remarkable ability to accommodate various cargoes and have found extensive applications in the biomedical sciences. To overcome the limitations of sEVs and broaden their potential applications, researchers should strive to deepen their understanding of current isolation, loading, and characterization techniques. Additionally, acquiring fundamental knowledge about sEVs origins and employing state-of-the-art methodologies in nanomedicine and regenerative medicine can expand the sEVs research scope. This review provides a comprehensive overview of state-of-the-art exosome-based strategies in diverse nanomedicine domains, encompassing cancer therapy, immunotherapy, and biomarker applications. Furthermore, we emphasize the immense potential of exosomes in regenerative medicine.
Collapse
Affiliation(s)
- Saeid Moghassemi
- Pôle de Recherche en Physiopathologie de La Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Arezoo Dadashzadeh
- Pôle de Recherche en Physiopathologie de La Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Maria João Sousa
- Pôle de Recherche en Physiopathologie de La Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Hanne Vlieghe
- Pôle de Recherche en Physiopathologie de La Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Jie Yang
- Pôle de Recherche en Physiopathologie de La Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Cecibel María León-Félix
- Pôle de Recherche en Physiopathologie de La Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Christiani A. Amorim
- Pôle de Recherche en Physiopathologie de La Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| |
Collapse
|
9
|
Yuan P, Wang ZH, Jiang H, Wang YH, Yang JY, Li LM, Wang WT, Chen J, Li DH, Long SY, Zhang W, He F, Wang WZ. Prevalence and plasma exosome-derive microRNA diagnostic biomarker screening of adolescent idiopathic scoliosis in Yunnan Province, China. Front Pediatr 2024; 12:1308931. [PMID: 38720947 PMCID: PMC11076730 DOI: 10.3389/fped.2024.1308931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 04/02/2024] [Indexed: 05/12/2024] Open
Abstract
Background Idiopathic scoliosis significantly affects the physical and mental health of children and adolescents, with varying prevalence rates in different regions. The occurrence of idiopathic scoliosis is associated with genetic regulation and biochemical factors, but the changes in exosome-derived miRNA profiles among idiopathic scoliosis patients remain unclear. This study aimed to determine the prevalence of idiopathic scoliosis in Yunnan Province, China, and identify key exosome-derived miRNAs in idiopathic scoliosis through a cohort study. Methods From January 2018 to December 2020, a cross-sectional study on idiopathic scoliosis in children and adolescents was conducted in Yunnan Province. A total of 84,460 students from 13 cities and counties in Yunnan Province participated in a scoliosis screening program, with ages ranging from 7 to 19 years. After confirmation through screening and imaging results, patients with severe idiopathic scoliosis and normal control individuals were selected using propensity matching. Subsequently, plasma exosome-derived miRNA sequencing and RT-qPCR validation were performed separately. Based on the validation results, diagnostic performance analysis and target gene prediction were conducted for differential plasma exosome-derived miRNAs. Results The overall prevalence of idiopathic scoliosis in children and adolescents in Yunnan Province was 1.10%, with a prevalence of 0.87% in males and 1.32% in females. The peak prevalence was observed at age 13. Among patients diagnosed with idiopathic scoliosis, approximately 12.8% had severe cases, and there were more cases of double curvature than of single curvature, with thoracolumbar curvature being the most common in the single-curvature group. Sequencing of plasma exosome-derived miRNAs associated with idiopathic scoliosis revealed 56 upregulated and 153 downregulated miRNAs. Further validation analysis confirmed that hsa-miR-27a-5p, hsa-miR-539-5p, and hsa-miR-1246 have potential diagnostic value. Conclusions We gained insights into the epidemiological characteristics of idiopathic scoliosis in Yunnan Province and conducted further analysis of plasma exosome-derived miRNA changes in patients with severe idiopathic scoliosis. This study has provided new insights for the prevention and diagnosis of idiopathic scoliosis, paving the way for exploring clinical biomarkers and molecular regulatory mechanisms. However, further validation and elucidation of the detailed biological mechanisms underlying these findings will be required in the future.
Collapse
Affiliation(s)
- Ping Yuan
- Department of Orthopedics, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
- The First Clinical College, Kunming Medical University, Kunming, Yunnan, China
| | - Zhi-Hua Wang
- Trauma Medicine Centre, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Hong Jiang
- Department of Medical Imaging, Kunming Children’s Hospital, Kunming Medical University, Kunming, Yunnan, China
| | - Yang-Hao Wang
- Department of Orthopedics, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
- The First Clinical College, Kunming Medical University, Kunming, Yunnan, China
- Department of Pathology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Jian-Yi Yang
- Department of Orthopaedics, Kunming Guandu District People’s Hospital, Kunming, Yunnan, China
| | - Lu-Ming Li
- Department of Orthopedics, Yunnan Sino-German Orthopedic Hospital, Kunming, Yunnan, China
| | - Wen-Tong Wang
- Department of Orthopedics, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
- The First Clinical College, Kunming Medical University, Kunming, Yunnan, China
| | - Jing Chen
- Department of Pathology and Pathophysiology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, Yunnan, China
| | - Deng-Hui Li
- Department of Orthopedics, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
- The First Clinical College, Kunming Medical University, Kunming, Yunnan, China
| | - Sheng-Yu Long
- The First Clinical College, Kunming Medical University, Kunming, Yunnan, China
- Department of Orthopedic, Qujing Affiliated Hospital of Kunming Medical University, Qujing, Yunnan, China
| | - Wan Zhang
- Department of Orthopedics, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
- The First Clinical College, Kunming Medical University, Kunming, Yunnan, China
| | - Fei He
- Department of Orthopedic, Qujing Affiliated Hospital of Kunming Medical University, Qujing, Yunnan, China
| | - Wei-Zhou Wang
- Department of Orthopedics, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
- The First Clinical College, Kunming Medical University, Kunming, Yunnan, China
| |
Collapse
|
10
|
Yue Y, Dai W, Wei Y, Cao S, Liao S, Li A, Liu P, Lin J, Zeng H. Unlocking the potential of exosomes: a breakthrough in the theranosis of degenerative orthopaedic diseases. Front Bioeng Biotechnol 2024; 12:1377142. [PMID: 38699435 PMCID: PMC11064847 DOI: 10.3389/fbioe.2024.1377142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 04/01/2024] [Indexed: 05/05/2024] Open
Abstract
Degenerative orthopaedic diseases pose a notable worldwide public health issue attributable to the global aging population. Conventional medical approaches, encompassing physical therapy, pharmaceutical interventions, and surgical methods, face obstacles in halting or reversing the degenerative process. In recent times, exosome-based therapy has gained widespread acceptance and popularity as an effective treatment for degenerative orthopaedic diseases. This therapeutic approach holds the potential for "cell-free" tissue regeneration. Exosomes, membranous vesicles resulting from the fusion of intracellular multivesicles with the cell membrane, are released into the extracellular matrix. Addressing challenges such as the rapid elimination of natural exosomes in vivo and the limitation of drug concentration can be effectively achieved through various strategies, including engineering modification, gene overexpression modification, and biomaterial binding. This review provides a concise overview of the source, classification, and preparation methods of exosomes, followed by an in-depth analysis of their functions and potential applications. Furthermore, the review explores various strategies for utilizing exosomes in the treatment of degenerative orthopaedic diseases, encompassing engineering modification, gene overexpression, and biomaterial binding. The primary objective is to provide a fresh viewpoint on the utilization of exosomes in addressing bone degenerative conditions and to support the practical application of exosomes in the theranosis of degenerative orthopaedic diseases.
Collapse
Affiliation(s)
- Yaohang Yue
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, China
- Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- National and Local Joint Engineering Research Centre of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Wei Dai
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, China
- Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- National and Local Joint Engineering Research Centre of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Department of Sports Medicine and Rehabilitation, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Yihao Wei
- Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- National and Local Joint Engineering Research Centre of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Siyang Cao
- Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- National and Local Joint Engineering Research Centre of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Shuai Liao
- Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- National and Local Joint Engineering Research Centre of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Aikang Li
- Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- National and Local Joint Engineering Research Centre of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Peng Liu
- Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- National and Local Joint Engineering Research Centre of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Jianjing Lin
- Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- National and Local Joint Engineering Research Centre of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Department of Sports Medicine and Rehabilitation, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Hui Zeng
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, China
- Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- National and Local Joint Engineering Research Centre of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Shenzhen Second People’s Hospital, Shenzhen, Guangdong, China
| |
Collapse
|
11
|
Zhu S, Wang J, Suo M, Huang H, Liu X, Wang J, Li Z. Can extracellular vesicles be considered as a potential frontier in the treatment of intervertebral disc disease? Ageing Res Rev 2023; 92:102094. [PMID: 37863436 DOI: 10.1016/j.arr.2023.102094] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 10/04/2023] [Accepted: 10/16/2023] [Indexed: 10/22/2023]
Abstract
As a global public health problem, low back pain (LBP) caused by intervertebral disc degeneration (IDD) seriously affects patients' quality of life. In addition, the prevalence of IDD tends to be younger, which brings a huge burden to individuals and society economically. Current treatments do not delay or reverse the progression of IDD. The emergence of biologic therapies has brought new hope for the treatment of IDD. Among them, extracellular vesicles (EVs), as nanoscale bioactive substances that mediate cellular communication, have now produced many surprising results in the research of the treatment of IDD. This article reviews the mechanisms and roles of EVs in delaying IDD and describes the prospects and challenges of EVs.
Collapse
Affiliation(s)
- Shengxu Zhu
- Department of Orthopedics, First Affiliated Hospital of Dalian Medical University, Dalian, the People's Republic of China; Key Laboratory of Molecular Mechanism for Repair and Remodeling of Orthopedic Diseases, Liaoning Province, the People's Republic of China
| | - Junlin Wang
- Department of Orthopedics, First Affiliated Hospital of Dalian Medical University, Dalian, the People's Republic of China
| | - Moran Suo
- Department of Orthopedics, First Affiliated Hospital of Dalian Medical University, Dalian, the People's Republic of China; Key Laboratory of Molecular Mechanism for Repair and Remodeling of Orthopedic Diseases, Liaoning Province, the People's Republic of China
| | - Huagui Huang
- Department of Orthopedics, First Affiliated Hospital of Dalian Medical University, Dalian, the People's Republic of China; Key Laboratory of Molecular Mechanism for Repair and Remodeling of Orthopedic Diseases, Liaoning Province, the People's Republic of China
| | - Xin Liu
- Department of Orthopedics, First Affiliated Hospital of Dalian Medical University, Dalian, the People's Republic of China; Key Laboratory of Molecular Mechanism for Repair and Remodeling of Orthopedic Diseases, Liaoning Province, the People's Republic of China
| | - Jinzuo Wang
- Department of Orthopedics, First Affiliated Hospital of Dalian Medical University, Dalian, the People's Republic of China; Key Laboratory of Molecular Mechanism for Repair and Remodeling of Orthopedic Diseases, Liaoning Province, the People's Republic of China
| | - Zhonghai Li
- Department of Orthopedics, First Affiliated Hospital of Dalian Medical University, Dalian, the People's Republic of China; Key Laboratory of Molecular Mechanism for Repair and Remodeling of Orthopedic Diseases, Liaoning Province, the People's Republic of China.
| |
Collapse
|
12
|
Huang S, Liu Y, Wang C, Xiang W, Wang N, Peng L, Jiang X, Zhang X, Fu Z. Strategies for Cartilage Repair in Osteoarthritis Based on Diverse Mesenchymal Stem Cells-Derived Extracellular Vesicles. Orthop Surg 2023; 15:2749-2765. [PMID: 37620876 PMCID: PMC10622303 DOI: 10.1111/os.13848] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 07/10/2023] [Accepted: 07/19/2023] [Indexed: 08/26/2023] Open
Abstract
Osteoarthritis (OA) causes disability and significant economic and social burden. Cartilage injury is one of the main pathological features of OA, and is often manifested by excessive chondrocyte death, inflammatory response, abnormal bone metabolism, imbalance of extracellular matrix (ECM) metabolism, and abnormal vascular or nerve growth. Regrettably, due to the avascular nature of cartilage, its capacity to repair is notably limited. Mesenchymal stem cells-derived extracellular vesicles (MSCs-EVs) play a pivotal role in intercellular communication, presenting promising potential not only as early diagnostic biomarkers in OA but also as efficacious therapeutic strategy. MSCs-EVs were confirmed to play a therapeutic role in the pathological process of cartilage injury mentioned above. This paper comprehensively provides the functions and mechanisms of MSCs-EVs in cartilage repair.
Collapse
Affiliation(s)
- Shanjun Huang
- Orthopedics DepartmentThe Affiliated Traditional Chinese Medicine Hospital of Southwest Medical UniversityLuzhouChina
| | - Yujiao Liu
- Orthopedics DepartmentThe Affiliated Traditional Chinese Medicine Hospital of Southwest Medical UniversityLuzhouChina
| | - Chenglong Wang
- Orthopedics DepartmentThe Affiliated Traditional Chinese Medicine Hospital of Southwest Medical UniversityLuzhouChina
| | - Wei Xiang
- Orthopedics DepartmentThe Affiliated Traditional Chinese Medicine Hospital of Southwest Medical UniversityLuzhouChina
| | - Nianwu Wang
- Orthopedics DepartmentThe Affiliated Traditional Chinese Medicine Hospital of Southwest Medical UniversityLuzhouChina
| | - Li Peng
- Orthopedics DepartmentThe Affiliated Traditional Chinese Medicine Hospital of Southwest Medical UniversityLuzhouChina
| | - Xuanang Jiang
- Orthopedics DepartmentThe Affiliated Traditional Chinese Medicine Hospital of Southwest Medical UniversityLuzhouChina
| | - Xiaomin Zhang
- Orthopedics DepartmentThe Affiliated Traditional Chinese Medicine Hospital of Southwest Medical UniversityLuzhouChina
| | - Zhijiang Fu
- Orthopedics DepartmentThe Affiliated Traditional Chinese Medicine Hospital of Southwest Medical UniversityLuzhouChina
| |
Collapse
|
13
|
Karchevskaya AE, Poluektov YM, Korolishin VA. Understanding Intervertebral Disc Degeneration: Background Factors and the Role of Initial Injury. Biomedicines 2023; 11:2714. [PMID: 37893088 PMCID: PMC10604877 DOI: 10.3390/biomedicines11102714] [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: 08/29/2023] [Revised: 09/21/2023] [Accepted: 09/22/2023] [Indexed: 10/29/2023] Open
Abstract
The etiology of intervertebral disc degeneration (IVDD) is complex and multifactorial, and it is still not fully understood. A better understanding of the pathogenesis of IVDD will help to improve treatment regimens and avoid unnecessary surgical aggression. In order to summarize recent research data on IVDD pathogenesis, including genetic and immune factors, a literature review was conducted. The pathogenesis of IVDD is a complex multifactorial process without an evident starting point. There are extensive data on the role of the different genetic factors affecting the course of the disease, such as mutations in structural proteins and enzymes involved in the immune response. However, these factors alone are not sufficient for the development of the disease. Nevertheless, like mechanical damage, they can also be considered risk factors for IVDD. In conclusion, currently, there is no consensus on a single concept for the pathogenesis of IVDD. We consider the intervertebral disc autoimmune damage hypothesis to be the most promising hypothesis for clinicians, because it can be extrapolated to all populations and does not counteract other factors. The genetic factors currently known do not allow for building effective predictive models; however, they can be used to stratify the risks of individual populations.
Collapse
Affiliation(s)
- Anna E. Karchevskaya
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, 5A Butlerova Str., 117485 Moscow, Russia;
- Medical Faculty, I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), 8-2 Trubetskaya Str., 119991 Moscow, Russia
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilov Str. 32, 119334 Moscow, Russia
| | - Yuri M. Poluektov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilov Str. 32, 119334 Moscow, Russia
- Department of Spinal Surgery, Burdenko Neurosurgical Institute, 4th-Tverskaya-Yamskaya Str. 16, 125047 Moscow, Russia
| | - Vasiliy A. Korolishin
- Russian Medical Academy of Postgraduate Education Studies, 2/1 Barrikadnaya Str., Building 1, 125993 Moscow, Russia;
| |
Collapse
|
14
|
Samanta A, Lufkin T, Kraus P. Intervertebral disc degeneration-Current therapeutic options and challenges. Front Public Health 2023; 11:1156749. [PMID: 37483952 PMCID: PMC10359191 DOI: 10.3389/fpubh.2023.1156749] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 06/12/2023] [Indexed: 07/25/2023] Open
Abstract
Degeneration of the intervertebral disc (IVD) is a normal part of aging. Due to the spine's declining function and the development of pain, it may affect one's physical health, mental health, and socioeconomic status. Most of the intervertebral disc degeneration (IVDD) therapies today focus on the symptoms of low back pain rather than the underlying etiology or mechanical function of the disc. The deteriorated disc is typically not restored by conservative or surgical therapies that largely focus on correcting symptoms and structural abnormalities. To enhance the clinical outcome and the quality of life of a patient, several therapeutic modalities have been created. In this review, we discuss genetic and environmental causes of IVDD and describe promising modern endogenous and exogenous therapeutic approaches including their applicability and relevance to the degeneration process.
Collapse
Affiliation(s)
| | | | - Petra Kraus
- Department of Biology, Clarkson University, Potsdam, NY, United States
| |
Collapse
|
15
|
Zhang S, Wang P, Hu B, Lv X, Liu W, Chen S, Shao Z. Inhibiting Heat Shock Protein 90 Attenuates Nucleus Pulposus Fibrosis and Pathologic Angiogenesis Induced by Macrophages via Down-Regulating Cell Migration-Inducing Protein. THE AMERICAN JOURNAL OF PATHOLOGY 2023; 193:960-976. [PMID: 37088454 DOI: 10.1016/j.ajpath.2023.03.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 03/12/2023] [Accepted: 03/23/2023] [Indexed: 04/25/2023]
Abstract
Intervertebral disc (IVD) degeneration (IVDD) is usually accompanied by nucleus pulposus (NP) fibrosis and pathologic angiogenesis, which are possibly associated with macrophage infiltration. Previous research indicates a destructive role of macrophages and the protective effect of inhibiting heat shock protein 90 (HSP90) in IVDD. Herein, the effects of inhibiting HSP90 on NP fibrosis and pathologic angiogenesis induced by macrophages were investigated further. Single-cell RNA-sequencing analysis was used to classify fibrotic NP cell (NPC) clusters and healthy NPC clusters in human NP tissues. The fibrotic NPC clusters were possibly associated with angiogenesis-related biological processes. Immunostaining showed the spatial association between blood vessel ingrowth and macrophage infiltration, as well as elevated levels of cell migration-inducing protein (CEMIP) and vascular endothelial growth factor A in severely degenerated human IVD tissues. Particularly, HSP90 inhibitor tanespimycin (17-AAG) ameliorated macrophage-induced fibrotic phenotype of NPCs via inhibiting CEMIP. M2, but not M1, macrophages promoted the pro-angiogenic ability of endothelial cells, which was attenuated by 17-AAG or HSP90 siRNA. Reversing the fibrotic phenotype of NPCs by Cemip siRNA also mitigated the pro-angiogenic effects of M2-conditioned medium-treated NPCs. Moreover, the murine IVDD model supported the 17-AAG-induced amelioration of NP fibrosis and endothelial cell invasion in IVD tissues. In conclusion, inhibiting HSP90 attenuated two interrelated pathologic processes, NP fibrosis and pathologic angiogenesis, induced by macrophages via down-regulating CEMIP.
Collapse
Affiliation(s)
- Shuo Zhang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Peng Wang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Binwu Hu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiao Lv
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Weijian Liu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Songfeng Chen
- Department of Orthopaedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Zengwu Shao
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
16
|
Zhang A, Cheng Z, Chen Y, Shi P, Gan W, Zhang Y. Emerging tissue engineering strategies for annulus fibrosus therapy. Acta Biomater 2023:S1742-7061(23)00337-9. [PMID: 37330029 DOI: 10.1016/j.actbio.2023.06.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 05/31/2023] [Accepted: 06/12/2023] [Indexed: 06/19/2023]
Abstract
Low back pain is a major public health concern experienced by 80% of the world's population during their lifetime, which is closely associated with intervertebral disc (IVD) herniation. IVD herniation manifests as the nucleus pulposus (NP) protruding beyond the boundaries of the intervertebral disc due to disruption of the annulus fibrosus (AF). With a deepening understanding of the importance of the AF structure in the pathogenesis of intervertebral disc degeneration, numerous advanced therapeutic strategies for AF based on tissue engineering, cellular regeneration, and gene therapy have emerged. However, there is still no consensus concerning the optimal approach for AF regeneration. In this review, we summarized strategies in the field of AF repair and highlighted ideal cell types and pro-differentiation targeting approaches for AF repair, and discussed the prospects and difficulties of implant systems combining cells and biomaterials to guide future research directions. STATEMENT OF SIGNIFICANCE: Low back pain is a major public health concern experienced by 80% of the world's population during their lifetime, which is closely associated with intervertebral disc (IVD) herniation. However, there is still no consensus concerning the optimal approach for annulus fibrosus (AF) regeneration. In this review, we summarized strategies in the field of AF repair and highlighted ideal cell types and pro-differentiation targeting approaches for AF repair, and discussed the prospects and difficulties of implant systems combining cells and biomaterials to guide future research directions.
Collapse
Affiliation(s)
- Anran Zhang
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zhangrong Cheng
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yuhang Chen
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Pengzhi Shi
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Weikang Gan
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yukun Zhang
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
17
|
Multiple nano-drug delivery systems for intervertebral disc degeneration: Current status and future perspectives. Bioact Mater 2023; 23:274-299. [DOI: 10.1016/j.bioactmat.2022.11.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 10/16/2022] [Accepted: 11/14/2022] [Indexed: 11/21/2022] Open
|
18
|
Exosomes treating osteoarthritis: hope with challenge. Heliyon 2023; 9:e13152. [PMID: 36711315 PMCID: PMC9880404 DOI: 10.1016/j.heliyon.2023.e13152] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Revised: 01/18/2023] [Accepted: 01/19/2023] [Indexed: 01/23/2023] Open
Abstract
Osteoarthritis (OA) has been proven as the second primary cause of pain and disability in the elderly population, impact patients both physically and mentally, as well as imposing a heavy burden on the global healthcare system. Current treatment methods, whether conservative or surgical, that aim at relieving symptoms can not delay or reverse the degenerative process in the structure. Scientists and clinicians are facing a revolution in OA treatment strategies. The emergence of exosomes brings hope for OA treatment based on pathology, which is attributed to its full potential in protecting chondrocytes from excessive death, alleviating inflammation, maintaining cartilage matrix metabolism, and regulating angiogenesis and subchondral bone remodeling. Therefore, we summarized the recent studies of exosomes in OA, aiming to comprehensively understand the functions and mechanisms of exosomes in OA treatment, which may provide direction and theoretical support for formulating therapeutic strategies in the future.
Collapse
|
19
|
Liu Z, Fu C. Application of single and cooperative different delivery systems for the treatment of intervertebral disc degeneration. Front Bioeng Biotechnol 2022; 10:1058251. [PMID: 36452213 PMCID: PMC9702580 DOI: 10.3389/fbioe.2022.1058251] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 11/01/2022] [Indexed: 11/07/2023] Open
Abstract
Intervertebral disc (IVD) degeneration (IDD) is the most universal pathogenesis of low back pain (LBP), a prevalent and costly medical problem across the world. Persistent low back pain can seriously affect a patient's quality of life and even lead to disability. Furthermore, the corresponding medical expenses create a serious economic burden to both individuals and society. Intervertebral disc degeneration is commonly thought to be related to age, injury, obesity, genetic susceptibility, and other risk factors. Nonetheless, its specific pathological process has not been completely elucidated; the current mainstream view considers that this condition arises from the interaction of multiple mechanisms. With the development of medical concepts and technology, clinicians and scientists tend to intervene in the early or middle stages of intervertebral disc degeneration to avoid further aggravation. However, with the aid of modern delivery systems, it is now possible to intervene in the process of intervertebral disc at the cellular and molecular levels. This review aims to provide an overview of the main mechanisms associated with intervertebral disc degeneration and the delivery systems that can help us to improve the efficacy of intervertebral disc degeneration treatment.
Collapse
Affiliation(s)
- Zongtai Liu
- Department of Orthopedics, Affiliated Hospital of Beihua University, Jilin, China
| | - Changfeng Fu
- Department of Spine Surgery, First Hospital of Jilin University, Changchun, China
| |
Collapse
|
20
|
Li Z, Wu Y, Tan G, Xu Z, Xue H. Exosomes and exosomal miRNAs: A new therapy for intervertebral disc degeneration. Front Pharmacol 2022; 13:992476. [PMID: 36160436 PMCID: PMC9492865 DOI: 10.3389/fphar.2022.992476] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 08/22/2022] [Indexed: 11/15/2022] Open
Abstract
Low back pain has been found as a major cause of global disease burden and disability. Intervertebral disc degeneration is recognized as the vital factor causing low back pain. Intervertebral disc degeneration has a complex mechanism and cannot be avoided. Traditional strategies for the treatment of intervertebral disc degeneration cannot meet the needs of intervertebral disc regeneration, so novel treatment methods are urgently required. Exosomes refer to extracellular vesicles that can be released by most cells, and play major roles in intercellular material transport and information transmission. MicroRNAs have been identified as essential components in exosomes, which can be selectively ingested by exosomes and delivered to receptor cells for the regulation of the physiological activities and functions of receptor cells. Existing studies have progressively focused on the role of exosomes and exosomal microRNAs in the treatment of intervertebral disc degeneration. The focus on this paper is placed on the changes of microenvironment during intervertebral disc degeneration and the biogenesis and mechanism of action of exosomes and exosomal microRNAs. The research results and deficiencies of exosomes and exosomal microRNAs in the regulation of apoptosis, extracellular matrix homeostasis, inflammatory response, oxidative stress, and angiogenesis in intervertebral disc degeneration are primarily investigated. The aim of this paper is to identify the latest research results, potential applications and challenges of this emerging treatment strategy.
Collapse
Affiliation(s)
- Zhichao Li
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yan Wu
- Department of Orthopedics, The First Affiliated Hospital of Shandong First Medcial Unversity, Jinan, China
| | - Guoqing Tan
- Department of Orthopedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Zhanwang Xu
- Department of Orthopedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Haipeng Xue
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
- Department of Orthopedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
- *Correspondence: Haipeng Xue,
| |
Collapse
|
21
|
Abstract
Tissue engineering and regenerative medicine (TERM) may be defined as a translational discipline focused on the development of novel techniques, devices, and materials to replace or repair injured or diseased tissue and organs. The main approaches typically use cells, scaffolds, and signaling molecules, either alone or in combination, to promote repair and regeneration. Although cells are required to create new functional tissue, the source of cells, either from an exogenous allogeneic or autologous source or through the recruitment of endogenous (autologous) cells, is technically challenging and risks the host rejection of new tissue. Regardless of the cell source, these approaches also require appropriate instruction for proliferation, differentiation, and in vivo spatial organization to create new functional tissue. Such instruction is supplied through the microenvironment where cells reside, environments which largely consist of the extracellular matrix (ECM). The specific components of the ECM, and broadly the extracellular space, responsible for promoting tissue regeneration and repair, are not fully understood, however extracellular vesicles (EVs) found in body fluids and solid phases of ECM have emerged as key mediators of tissue regeneration and repair. Additionally, these EVs might serve as potential cell-free tools in TERM to promote tissue repair and regeneration with minimal risk for host rejection and adverse sequelae. The past two decades have shown a substantial interest in understanding the therapeutic role of EVs and their applications in the context of TERM. Therefore, the purpose of this review is to highlight the fundamental characteristics of EVs, the current pre-clinical and clinical applications of EVs in TERM, and the future of EV-based strategies in TERM.
Collapse
|
22
|
MECHANISM OF MIR-25-3P CARRIED BY EXTRACELLULAR VESICLES DERIVED FROM PLATELET-RICH PLASMA IN IL-1β-INDUCED NUCLEUS PULPOSUS CELL DEGENERATION VIA THE SOX4/CXCR7 AXIS. Shock 2022; 58:56-67. [PMID: 35984761 DOI: 10.1097/shk.0000000000001947] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
ABSTRACT Objectives: Nucleus pulposus (NP) cell degeneration promotes the progression of intervertebral disc (IVD) degeneration. MicroRNAs (miRs) are associated with IVD degeneration. This study expounded the mechanism of microRNA (miR)-25-3p carried by extracellular vesicles (EVs) derived from platelet-rich plasma (PRP) in interleukin (IL)-1β-induced NP cell degeneration. Methods: Platelet-rich plasma from mouse blood was obtained, and EVs were isolated from PRP (EVs derived from PRP [PRP-EVs]) and identified. Nucleus pulposus cells were isolated from the mouse lumbar IVD and treated with IL-1β to induce NP cell degeneration. Extracellular vesicles derived from PRP were added into NP cell culture medium. Afterward, intracellular miR-25-3p, sex determining region Y-related high-mobility-group box 4 (SOX4), and CXC chemokine receptor 7 (CXCR7) levels were examined. Nucleus pulposus cell viability, apoptosis, and inflammation were detected. Extracellular vesicles derived from PRP were labeled by PKH67 to obverse the uptake of EVs by NP cells. The binding relations between SOX4 and miR-25-3p and CXCR7 were predicted and examined. Functional rescue experiments were performed to investigate the roles of miR-25-3p, SOX4, and CXCR7 in NP cell degeneration. Results: miR-25-3p was downregulated, whereas SOX4 and CXCR7 were upregulated in IL-1β-induced NP cells. Extracellular vesicles derived from PRP increased the cell viability, and decreased apoptosis and inflammation. miR-25-3p carried by PRP-EVs into NP cells alleviated NP cell degeneration. miR-25-3p inhibited SOX4 expression and limited CXCR7 transcription. Silencing miR-25-3p or overexpressing SOX4 or CXCR7 reversed the alleviating role of PRP-EVs in NP cell degeneration. Conclusion: miR-25-3p carried by PRP-EVs into NP cells elevated intracellular miR-25-3p expression, which suppressed SOX4 expression and further limited CXCR7 transcription, thus alleviating IL-1β-induced NP cell degeneration. Extracellular vesicles derived from PRP containing miR-25-3p may be a new method for IVD treatment.
Collapse
|
23
|
Ginini L, Billan S, Fridman E, Gil Z. Insight into Extracellular Vesicle-Cell Communication: From Cell Recognition to Intracellular Fate. Cells 2022; 11:1375. [PMID: 35563681 PMCID: PMC9101098 DOI: 10.3390/cells11091375] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Revised: 04/13/2022] [Accepted: 04/14/2022] [Indexed: 01/27/2023] Open
Abstract
Extracellular vesicles (EVs) are heterogamous lipid bilayer-enclosed membranous structures secreted by cells. They are comprised of apoptotic bodies, microvesicles, and exosomes, and carry a range of nucleic acids and proteins that are necessary for cell-to-cell communication via interaction on the cells surface. They initiate intracellular signaling pathways or the transference of cargo molecules, which elicit pleiotropic responses in recipient cells in physiological processes, as well as pathological processes, such as cancer. It is therefore important to understand the molecular means by which EVs are taken up into cells. Accordingly, this review summarizes the underlying mechanisms involved in EV targeting and uptake. The primary method of entry by EVs appears to be endocytosis, where clathrin-mediated, caveolae-dependent, macropinocytotic, phagocytotic, and lipid raft-mediated uptake have been variously described as being prevalent. EV uptake mechanisms may depend on proteins and lipids found on the surfaces of both vesicles and target cells. As EVs have been shown to contribute to cancer growth and progression, further exploration and targeting of the gateways utilized by EVs to internalize into tumor cells may assist in the prevention or deceleration of cancer pathogenesis.
Collapse
Affiliation(s)
- Lana Ginini
- Rappaport Family Institute for Research in the Medical Sciences, Technion–Israel Institute of Technology, Haifa 31096, Israel; (L.G.); (E.F.)
| | - Salem Billan
- Head and Neck Institute, The Holy Family Hospital Nazareth, Nazareth 1641100, Israel;
- Medical Oncology and Radiation Therapy Program, Oncology Section, Rambam Health Care Campus, HaAliya HaShniya Street 8, Haifa 3109601, Israel
| | - Eran Fridman
- Rappaport Family Institute for Research in the Medical Sciences, Technion–Israel Institute of Technology, Haifa 31096, Israel; (L.G.); (E.F.)
| | - Ziv Gil
- Head and Neck Institute, The Holy Family Hospital Nazareth, Nazareth 1641100, Israel;
| |
Collapse
|
24
|
Exosomes: A promising therapeutic strategy for intervertebral disc degeneration. Exp Gerontol 2022; 163:111806. [DOI: 10.1016/j.exger.2022.111806] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 03/25/2022] [Accepted: 04/06/2022] [Indexed: 11/23/2022]
|
25
|
DiStefano TJ, Vaso K, Danias G, Chionuma HN, Weiser JR, Iatridis JC. Extracellular Vesicles as an Emerging Treatment Option for Intervertebral Disc Degeneration: Therapeutic Potential, Translational Pathways, and Regulatory Considerations. Adv Healthc Mater 2022; 11:e2100596. [PMID: 34297485 PMCID: PMC8783929 DOI: 10.1002/adhm.202100596] [Citation(s) in RCA: 77] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 07/08/2021] [Indexed: 12/14/2022]
Abstract
Emergent approaches in regenerative medicine look toward the use of extracellular vesicles (EVs) as a next-generation treatment strategy for intervertebral disc (IVD) degeneration (IVDD) because of their ability to attenuate chronic inflammation, reduce apoptosis, and stimulate proliferation in a number of tissue systems. Yet, there are no Food and Drug Administration (FDA)-approved EV therapeutics in the market with an indication for IVDD, which motivates this article to review the current state of the field and provide an IVD-specific framework to assess its efficacy. In this systematic review, 29 preclinical studies that investigate EVs in relation to the IVD are identified, and additionally, the regulatory approval process is reviewed in an effort to accelerate emerging EV-based therapeutics toward FDA submission and timeline-to-market. The majority of studies focus on nucleus pulposus responses to EV treatment, where the main findings show that stem cell-derived EVs can decelerate the progression of IVDD on the molecular, cellular, and organ level. The findings also highlight the importance of the EV parent cell's pathophysiological and differentiation state, which affects downstream treatment responses and therapeutic outcomes. This systematic review substantiates the use of EVs as a promising cell-free strategy to treat IVDD and enhance endogenous repair.
Collapse
Affiliation(s)
- Tyler J. DiStefano
- Leni and Peter W. May Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York NY, USA
| | - Keti Vaso
- Department of Chemical Engineering, The Cooper Union for the Advancement of Science and Art, New York NY, USA
| | - George Danias
- Leni and Peter W. May Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York NY, USA
| | - Henry N. Chionuma
- Leni and Peter W. May Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York NY, USA
| | - Jennifer R. Weiser
- Department of Chemical Engineering, The Cooper Union for the Advancement of Science and Art, New York NY, USA
| | - James C. Iatridis
- Leni and Peter W. May Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York NY, USA
| |
Collapse
|
26
|
Li W, Zhang S, Wang D, Zhang H, Shi Q, Zhang Y, Wang M, Ding Z, Xu S, Gao B, Yan M. Exosomes Immunity Strategy: A Novel Approach for Ameliorating Intervertebral Disc Degeneration. Front Cell Dev Biol 2022; 9:822149. [PMID: 35223870 PMCID: PMC8870130 DOI: 10.3389/fcell.2021.822149] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 12/21/2021] [Indexed: 12/11/2022] Open
Abstract
Low back pain (LBP), which is one of the most severe medical and social problems globally, has affected nearly 80% of the population worldwide, and intervertebral disc degeneration (IDD) is a common musculoskeletal disorder that happens to be the primary trigger of LBP. The pathology of IDD is based on the impaired homeostasis of catabolism and anabolism in the extracellular matrix (ECM), uncontrolled activation of immunologic cascades, dysfunction, and loss of nucleus pulposus (NP) cells in addition to dynamic cellular and biochemical alterations in the microenvironment of intervertebral disc (IVD). Currently, the main therapeutic approach regarding IDD is surgical intervention, but it could not considerably cure IDD. Exosomes, extracellular vesicles with a diameter of 30–150 nm, are secreted by various kinds of cell types like stem cells, tumor cells, immune cells, and endothelial cells; the lipid bilayer of the exosomes protects them from ribonuclease degradation and helps improve their biological efficiency in recipient cells. Increasing lines of evidence have reported the promising applications of exosomes in immunological diseases, and regarded exosomes as a potential therapeutic source for IDD. This review focuses on clarifying novel therapies based on exosomes derived from different cell sources and the essential roles of exosomes in regulating IDD, especially the immunologic strategy.
Collapse
Affiliation(s)
- Weihang Li
- Department of Orthopedic Surgery, Xijing Hospital, Air Force Medical University, Xi’an, China
| | - Shilei Zhang
- Department of Orthopedic Surgery, Xijing Hospital, Air Force Medical University, Xi’an, China
| | - Dong Wang
- Department of Orthopedic Surgery, Xijing Hospital, Air Force Medical University, Xi’an, China
- Department of Orthopaedics, Affiliated Hospital of Yanan University, Yanan, China
| | - Huan Zhang
- Department of Orthopedic Surgery, Xijing Hospital, Air Force Medical University, Xi’an, China
| | - Quan Shi
- Department of Orthopedic Surgery, Xijing Hospital, Air Force Medical University, Xi’an, China
| | - Yuyuan Zhang
- Department of Critical Care Medicine, Xijing Hospital, Air Force Medical University, Xi’an, China
| | - Mo Wang
- The First Brigade of Basic Medical College, Air Force Military Medical University, Xi’an, China
| | - Ziyi Ding
- Department of Orthopedic Surgery, Xijing Hospital, Air Force Medical University, Xi’an, China
| | - Songjie Xu
- Beijing Luhe Hospital, Capital Medical University, Beijing, China
- *Correspondence: Songjie Xu, ; Bo Gao, ; Ming Yan,
| | - Bo Gao
- Department of Orthopedic Surgery, Xijing Hospital, Air Force Medical University, Xi’an, China
- *Correspondence: Songjie Xu, ; Bo Gao, ; Ming Yan,
| | - Ming Yan
- Department of Orthopedic Surgery, Xijing Hospital, Air Force Medical University, Xi’an, China
- *Correspondence: Songjie Xu, ; Bo Gao, ; Ming Yan,
| |
Collapse
|
27
|
Yang H, Yang S, Shen H, Wu S, Ruan J, Lyu G. Construction of the amniotic fluid-derived exosomal ceRNA network associated with ventricular septal defect. Genomics 2021; 113:4293-4302. [PMID: 34758360 DOI: 10.1016/j.ygeno.2021.11.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 10/28/2021] [Accepted: 11/03/2021] [Indexed: 01/08/2023]
Abstract
Ventricular septal defect (VSD) is the most frequent congenital cardiac malformations. Amniotic fluid (AF) contains a higher abundance of biological compounds that could reflect fetal health information. The aims of our study were to construct a competitive endogenous RNA (ceRNA) network based on AF-derived exosomal ncRNAs. We conducted whole transcriptome profiling in six pairs of AF-derived exosomes from VSD fetuses and matched healthy controls. A total of 1252 differentially expressed (DE) mRNAs, 256 DE-miRNAs and 1090 DE-lncRNAs were found to be significantly altered in the VSD group. We constructed a ceRNA regulatory network including 46 mRNAs, 11 miRNAs and 47 lncRNAs. The expression level of 6 hub RNAs were validated using qRT-PCR. In conclusion, AF-derived exosomal VSD-related ceRNAs provide a basis for a better understanding of the role of ncRNAs in the pathogenesis and mechanisms of VSD, which may lead to the discovery of potential diagnostic biomarkers for fetal VSD.
Collapse
Affiliation(s)
- Hainan Yang
- Department of Ultrasound, the Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Shuping Yang
- Department of Ultrasound, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou, Fujian, China
| | - Haolin Shen
- Department of Ultrasound, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou, Fujian, China
| | - Shufen Wu
- Department of Ultrasound, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou, Fujian, China
| | - Junxian Ruan
- Department of Ultrasound, Quanzhou Women's and Children's Hospital, Quanzhou, Fujian, China
| | - Guorong Lyu
- Department of Ultrasound, the Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China; Collaborative Innovation Center for Maternal and Infant Health Service Application Technology of Education Ministry, Quanzhou Medical College, Quanzhou, Fujian, China.
| |
Collapse
|