1
|
Zhang C, Li CM, Jiang Q. Role of oxidative stress in intervertebral disc degeneration: mechanisms, pathogenesis, and therapeutic strategies. Mol Biol Rep 2025; 52:444. [PMID: 40314795 DOI: 10.1007/s11033-025-10538-8] [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: 03/08/2025] [Accepted: 04/23/2025] [Indexed: 05/03/2025]
Abstract
Intervertebral disc degeneration (IDD) is a prevalent and debilitating spinal condition, characterised by the progressive degradation of disc structure and function, often accompanied by pain. Despite our increasing understanding of IDD, the precise mechanisms underlying its development and potential therapeutic targets remain incompletely understood. Recent research has highlighted that oxidative stress, along with immune abnormalities, mechanical loading imbalances, and metabolic disruptions, play a pivotal role in IDD initiation and progression. Oxidative stress in IDD results from an overproduction of reactive oxygen species (ROS) and a compromised ability to eliminate them, disrupting the redox homeostasis within the intervertebral disc. This disturbance in redox balance leads to extracellular matrix degradation (ECM), induces cellular apoptosis, and worsens the damage to disc tissues. This review provides a comprehensive overview of the pathophysiological processes of IDD, with a particular focus on the role of oxidative stress. Additionally, we explore current advancements in therapeutic strategies targeting oxidative stress, including antioxidant drugs, biomaterials, and stem cell-based approaches, offering promising avenues for the management and treatment of IDD.
Collapse
Affiliation(s)
- Cong Zhang
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Affiliated Hospital of Medical School, Nanjing Drum Tower Hospital, Nanjing University, Nanjing, 210008, Jiangsu, PR China
- Department of Spine Surgery, School of Medicine, Zhongda Hospital, Southeast University, Nanjing, 210008, Jiangsu, PR China
| | - Cheng-Ming Li
- Department of Spine Surgery, School of Medicine, Zhongda Hospital, Southeast University, Nanjing, 210008, Jiangsu, PR China
| | - Qing Jiang
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Affiliated Hospital of Medical School, Nanjing Drum Tower Hospital, Nanjing University, Nanjing, 210008, Jiangsu, PR China.
| |
Collapse
|
2
|
Li Z, Cheng W, Gao K, Liang S, Ke L, Wang M, Fan J, Li D, Zhang P, Xu Z, Li N. Pyroptosis: A spoiler of peaceful coexistence between cells in degenerative bone and joint diseases. J Adv Res 2025; 71:227-262. [PMID: 38876191 DOI: 10.1016/j.jare.2024.06.010] [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: 02/17/2024] [Revised: 05/23/2024] [Accepted: 06/07/2024] [Indexed: 06/16/2024] Open
Abstract
BACKGROUND As people age, degenerative bone and joint diseases (DBJDs) become more prevalent. When middle-aged and elderly people are diagnosed with one or more disorders such as osteoporosis (OP), osteoarthritis (OA), and intervertebral disc degeneration (IVDD), it often signals the onset of prolonged pain and reduced functionality. Chronic inflammation has been identified as the underlying cause of various degenerative diseases, including DBJDs. Recently, excessive activation of pyroptosis, a form of programed cell death (PCD) mediated by inflammasomes, has emerged as a primary driver of harmful chronic inflammation. Consequently, pyroptosis has become a potential target for preventing and treating DBJDs. AIM OF REVIEW This review explored the physiological and pathological roles of the pyroptosis pathway in bone and joint development and its relation to DBJDs. Meanwhile, it elaborated the molecular mechanisms of pyroptosis within individual cell types in the bone marrow and joints, as well as the interplay among different cell types in the context of DBJDs. Furthermore, this review presented the latest compelling evidence supporting the idea of regulating the pyroptosis pathway for DBJDs treatment, and discussed the potential, limitations, and challenges of various therapeutic strategies involving pyroptosis regulation. KEY SCIENTIFIC CONCEPTS OF REVIEW In summary, an interesting identity for the unregulated pyroptosis pathway in the context of DBJDs was proposed in this review, which was undertaken as a spoiler of peaceful coexistence between cells in a degenerative environment. Over the extended course of DBJDs, pyroptosis pathway perpetuated its activity through crosstalk among pyroptosis cascades in different cell types, thus exacerbating the inflammatory environment throughout the entire bone marrow and joint degeneration environment. Correspondingly, pyroptosis regulation therapy emerged as a promising option for clinical treatment of DBJDs.
Collapse
Affiliation(s)
- Zhichao Li
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250014, China; Department of Orthopedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China; Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Wenxiang Cheng
- Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Kuanhui Gao
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Songlin Liang
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250014, China; Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Liqing Ke
- Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Mengjie Wang
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Jilin Fan
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Dandan Li
- College of Integrated Traditional Chinese and Western Medicine, Hebei University of Chinese Medicine, Shijiazhuang 050011, China
| | - Peng Zhang
- Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; Faculty of Biomedical Engineering, Shenzhen University of Advanced Technology, Shenzhen 518000, China; Key Laboratory of Biomedical Imaging Science and System, Chinese Academy of Sciences, Shenzhen, 518000 China; Shandong Zhongke Advanced Technology Co., Ltd., Jinan, 250300 China.
| | - Zhanwang Xu
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250014, China; Department of Orthopedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China.
| | - Nianhu Li
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250014, China; Department of Orthopedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, 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
|
Gao X, Jia S, Gao L, Chen S, Zhang Y, Liang X, Zhang L, Zhang B, Meng C. MSC-derived exosomes alleviate oxidative stress-induced lysosomal membrane permeabilization damage in degenerated nucleus pulposus cells via promoting m6A demethylation of Nrf2. Free Radic Biol Med 2025; 235:213-230. [PMID: 40316061 DOI: 10.1016/j.freeradbiomed.2025.04.051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2025] [Revised: 04/26/2025] [Accepted: 04/29/2025] [Indexed: 05/04/2025]
Abstract
Lysosomal membrane permeabilization (LMP) is a specific feature of lysosomal dysfunction; however, its specific role and underlying mechanisms involved in intervertebral disc degeneration (IVDD) remain elusive. Although the therapeutic potential of mesenchymal stem cell-derived exosomes (MSC-Exo) in ameliorating IVDD has been verified, it remains unclear whether their protective effects are referred to LMP damage. This work revealed that oxidative stress induced-LMP damage directly mediated the pathological process of human IVDD, which aggravated nucleus pulposus cells (NPCs) senescence by disrupting lysosomal autophagy function. Conversely, umbilical cord derived MSC-Exo inhibited LMP damage in degenerated NPCs by activating Nrf2-medaited anti-oxidative stress effects. Specifically, MSC-Exo facilitated H3K27ac modification in the demethylase FTO promoter by promoting histone acetyltransferase activity of p300/CBP, resulting in the enhanced FTO transcription. This process inhibited the elevation of N6-methyladenosine (m6A) modification of Nrf2 in degenerated NPCs, resulting in less recognition of YTHDF2 and enhanced stability of Nrf2 expression. Here, our finding demonstrates oxidative stress induced-LMP damage potentially establishing pathological conditions conducive to the progression of IVDD, and providing epigenetic regulatory targets for MSC-Exo in the treatment of IVDD.
Collapse
Affiliation(s)
- Xu Gao
- Department of Spine Surgery, Affiliated Hospital of Jining Medical University, Jining, Shandong, 272000, PR China
| | - Shu Jia
- Clinical Research Team of Spine & Spinal Cord Diseases, Medical Research Center, Affiliated Hospital of Jining Medical University, Jining, Shandong, 272000, PR China
| | - Longfei Gao
- Department of Spine Surgery, Affiliated Hospital of Jining Medical University, Jining, Shandong, 272000, PR China
| | - Shang Chen
- Clinical Research Team of Spine & Spinal Cord Diseases, Medical Research Center, Affiliated Hospital of Jining Medical University, Jining, Shandong, 272000, PR China
| | - Yanan Zhang
- Department of obstetrics, Affiliated Hospital of Jining Medical University, Jining, Shandong, 272000, PR China
| | - Xiao Liang
- Department of Spine Surgery, Affiliated Hospital of Jining Medical University, Jining, Shandong, 272000, PR China
| | - Lu Zhang
- Department of Spine Surgery, Affiliated Hospital of Jining Medical University, Jining, Shandong, 272000, PR China
| | - Bin Zhang
- Department of Laboratory Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, 272000, PR China.
| | - Chunyang Meng
- Department of Spine Surgery, Affiliated Hospital of Jining Medical University, Jining, Shandong, 272000, PR China.
| |
Collapse
|
5
|
Li ZP, Li H, Ruan YH, Wang P, Zhu MT, Fu WP, Wang RB, Tang XD, Zhang Q, Li SL, Yin H, Li CJ, Tian YG, Han RN, Wang YB, Zhang CJ. Stem cell therapy for intervertebral disc degeneration: Clinical progress with exosomes and gene vectors. World J Stem Cells 2025; 17:102945. [PMID: 40308883 PMCID: PMC12038459 DOI: 10.4252/wjsc.v17.i4.102945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 01/25/2025] [Accepted: 03/10/2025] [Indexed: 04/23/2025] Open
Abstract
Intervertebral disc degeneration is a leading cause of lower back pain and is characterized by pathological processes such as nucleus pulposus cell apoptosis, extracellular matrix imbalance, and annulus fibrosus rupture. These pathological changes result in disc height loss and functional decline, potentially leading to disc herniation. This comprehensive review aimed to address the current challenges in intervertebral disc degeneration treatment by evaluating the regenerative potential of stem cell-based therapies, with a particular focus on emerging technologies such as exosomes and gene vector systems. Through mechanisms such as differentiation, paracrine effects, and immunomodulation, stem cells facilitate extracellular matrix repair and reduce nucleus pulposus cell apoptosis. Despite recent advancements, clinical applications are hindered by challenges such as hypoxic disc environments and immune rejection. By analyzing recent preclinical and clinical findings, this review provided insights into optimizing stem cell therapy to overcome these obstacles and highlighted future directions in the field.
Collapse
Affiliation(s)
- Zhi-Peng Li
- Second Department of Orthopedics, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
- Tianjian Advanced Biomedical Laboratory, Zhengzhou University, Zhengzhou 450001, Henan Province, China
| | - Han Li
- Department of Orthopedics, Affiliated Dongyang Hospital of Wenzhou Medical University, Jinhua 322100, Zhejiang Province, China
| | - Yu-Hua Ruan
- Second Department of Orthopedics, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Peng Wang
- Second Department of Orthopedics, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Meng-Ting Zhu
- Department of Neurology, Union Medical College Hospital of Fujian Medical University, Fuzhou 350001, Fujian Province, China
| | - Wei-Ping Fu
- Second Department of Orthopedics, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Rui-Bo Wang
- Second Department of Orthopedics, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Xiao-Dong Tang
- Second Department of Orthopedics, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Qi Zhang
- Second Department of Orthopedics, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Sen-Li Li
- Second Department of Orthopedics, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - He Yin
- Second Department of Orthopedics, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Cheng-Jin Li
- Second Department of Orthopedics, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Yi-Gong Tian
- Third Department of Orthopedics, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Rui-Ning Han
- Third Department of Orthopedics, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Yao-Bin Wang
- Second Department of Orthopedics, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
- Henan Key Laboratory for Helicobacter pylori and Digestive Tract Microecology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Chang-Jiang Zhang
- Second Department of Orthopedics, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China.
| |
Collapse
|
6
|
Li H, Kelley J, Ye Y, Ye ZW, Townsend DM, Zhang J, Wu Y. REDOX Imbalance and Oxidative Stress in the Intervertebral Disc: The Effect of Mechanical Stress and Cigarette Smoking on ER Stress and Mitochondrial Dysfunction. Cells 2025; 14:613. [PMID: 40277939 PMCID: PMC12025608 DOI: 10.3390/cells14080613] [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: 03/11/2025] [Revised: 04/15/2025] [Accepted: 04/17/2025] [Indexed: 04/26/2025] Open
Abstract
Low back pain is a widespread condition that significantly impacts quality of life, with intervertebral disc degeneration (IDD) being a major contributing factor. However, the underlying mechanisms of IDD remain poorly understood, necessitating further investigation. Environmental risk factors, such as mechanical stress and cigarette smoke, elevate reactive oxygen species levels from both endogenous and exogenous sources, leading to redox imbalance and oxidative stress. The endoplasmic reticulum (ER) and mitochondria, two key organelles responsible for protein folding and energy production, respectively, are particularly vulnerable to oxidative stress. Under oxidative stress conditions, ER stress and mitochondrial dysfunction occur, resulting in unfolded protein response activation, impaired biosynthetic processes, and disruptions in the tricarboxylic acid cycle and electron transport chain, ultimately compromising energy metabolism. Prolonged and excessive ER stress can further trigger apoptosis through ER-mitochondrial crosstalk. Given the unique microenvironment of the intervertebral disc (IVD)-characterized by hypoxia, glucose starvation, and region-specific cellular heterogeneity-the differential effects of environmental stressors on distinct IVD cell populations require further investigation. This review explores the potential mechanisms through which environmental risk factors alter IVD cell activities, contributing to IDD progression, and discusses future therapeutic strategies aimed at mitigating disc degeneration.
Collapse
Affiliation(s)
- Hui Li
- Department of Bioengineering, Clemson University, Charleston, SC 29425, USA; (H.L.); (J.K.)
| | - Joshua Kelley
- Department of Bioengineering, Clemson University, Charleston, SC 29425, USA; (H.L.); (J.K.)
| | - Yiqing Ye
- Department of Orthopaedics and Physical Medicine & Rehabilitation, Medical University of South Carolina, Charleston, SC 29425, USA
- Academic Magnet High School, North Charleston, SC 29405, USA
| | - Zhi-Wei Ye
- Department of Pharmacology and Immunology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Danyelle M. Townsend
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Jie Zhang
- Department of Pharmacology and Immunology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Yongren Wu
- Department of Bioengineering, Clemson University, Charleston, SC 29425, USA; (H.L.); (J.K.)
- Department of Orthopaedics and Physical Medicine & Rehabilitation, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|
7
|
Keshavarz S, Alavi CE, Aghayan H, Jafari-Shakib R, Vojoudi E. Advancements in Degenerative Disc Disease Treatment: A Regenerative Medicine Approach. Stem Cell Rev Rep 2025:10.1007/s12015-025-10882-z. [PMID: 40232618 DOI: 10.1007/s12015-025-10882-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/07/2025] [Indexed: 04/16/2025]
Abstract
Regenerative medicine represents a transformative approach to treating nucleus pulposus degeneration and offers hope for patients suffering from chronic low back pain due to disc degeneration. By focusing on restoring the natural structure and function of the nucleus pulposus rather than merely alleviating symptoms, these innovative therapies hold the potential to significantly improve patient outcomes. As research continues to advance in this field, we may soon witness a paradigm shift in how we approach spinal health and degenerative disc disease. The main purpose of this review is to provide an overview of the various regenerative approaches that target the restoration of the nucleus pulposus, a primary site for initiation of intervertebral disc degeneration.
Collapse
Affiliation(s)
- Samaneh Keshavarz
- School of Paramedicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Cyrus Emir Alavi
- Department of Anesthesiology, Neuroscience Research Center, Avicenna University Hospital, Guilan University of Medical Sciences, Rasht, Iran
| | - Hamidreza Aghayan
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular- Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Jafari-Shakib
- Department of Immunology, School of Medicine, Guilan University of Medical Sciences, P.O.Box 41635 - 3363, Rasht, Iran.
| | - Elham Vojoudi
- Regenerative Medicine, Organ Procurement and Transplantation Multidisciplinary Center, School of Medicine, Razi Hospital, Guilan University of Medical Sciences, Rasht, Iran.
| |
Collapse
|
8
|
Zhao W, Liu Y, Hu Y, Zhang G. SOX4 accelerates intervertebral disc degeneration via EZH2/NRF2 pathway in response to mitochondrial ROS-dependent NLRP3 inflammasome activation in nucleus pulposus cells. J Transl Med 2025; 23:395. [PMID: 40181390 PMCID: PMC11969779 DOI: 10.1186/s12967-024-05913-1] [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: 11/13/2023] [Accepted: 11/22/2024] [Indexed: 04/05/2025] Open
Abstract
OBJECTIVE The transcription factor SRY-related HMG-box 4 (SOX4) has been implicated in intervertebral disc diseases. This study aimed to investigate the role of SOX4 in intervertebral disc degeneration (IDD) and explore the underlying molecular mechanisms. METHODS We established an IDD rat model via surgery and analyzed SOX4 expression using qRT-PCR and Western blotting. Histological evaluation, immunohistochemistry, and Safranin O staining assessed IDD progression. In vitro, an IDD cellular model was constructed using IL-1β-stimulated nucleus pulposus (NP) cells. SOX4 knockdown and overexpression experiments in NP cells examined SOX4 effects on ECM degradation, NLRP3-mediated pyroptosis, and mitochondrial ROS-dependent NLRP3 inflammasome activation. The involvement of the EZH2/NRF2 pathway in SOX4-mediated NLRP3 activation was also examined. RESULTS SOX4 expression was significantly increased in IDD rats and promoted IDD progression. Knockdown of SOX4 inhibited ECM degradation and NLRP3-mediated pyroptosis in NP cells. In vitro experiments showed that SOX4 promoted ECM degradation by upregulating MMPs and ADAMTS-5 expression, and suppressed collagen II and aggrecan synthesis. SOX4 knockdown inhibited NLRP3-mediated pyroptosis, while overexpression accelerated it in NP cells. Additionally, SOX4 was found to exacerbate mitochondrial ROS-dependent NLRP3 inflammasome activation in NP cells. Further investigation revealed that SOX4 enhanced NLRP3 inflammasome activation by upregulating EZH2 expression and modulating the EZH2/NRF2 pathway, with EZH2 inhibition attenuating SOX4-induced NLRP3 activation. CONCLUSION Our findings suggest that SOX4 accelerates IDD progression by promoting NLRP3 inflammasome activation via modulating the EZH2/NRF2 pathway, leading to NP cell pyroptosis and ECM degradation. Targeting SOX4 may represent a potential therapeutic strategy for treating IDD.
Collapse
Affiliation(s)
- Wenzhi Zhao
- Department of Traumatic Orthopedics, The Second Affiliated Hospital, Dalian Medical University, Dalian, 116011, China
| | - Yadong Liu
- Department of Spinal Surgery, Dalian Municipal Central Hospital, Dalian, 116033, China
| | - Yunxiang Hu
- Department of Spinal Surgery, Dalian Municipal Central Hospital, Dalian, 116033, China
| | - Guiqi Zhang
- Department of Spinal Surgery, Dalian Municipal Central Hospital, Dalian, 116033, China.
| |
Collapse
|
9
|
Xu M, Ren M, Zhang X, Peng W, Li H, Liao W, Xie J, Zhang X. Mesenchymal stem cell-derived small extracellular vesicles restored nasal barrier function in allergic rhinitis via miR-143-GSK3B in human nasal epithelial cells. J Allergy Clin Immunol 2025; 155:1236-1249.e5. [PMID: 39542141 DOI: 10.1016/j.jaci.2024.10.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 08/05/2024] [Accepted: 10/21/2024] [Indexed: 11/17/2024]
Abstract
BACKGROUND The nasal epithelial barrier is the first line of defense against the deep entry of pathogens or aeroallergens and is more critical in allergic rhinitis (AR). Restoring epithelial barrier dysfunction might be a promising strategy for AR. Recent studies reported that mesenchymal stem cell-derived small extracellular vesicles (MSC-sEV) potentially inhibit the inflammation response and promote tissue regeneration. However, their effect on nasal epithelial cells remains unknown. OBJECTIVES This study sought to describe the therapeutic effect of MSC-sEV on AR, particularly focusing their effect on nasal epithelial cells and underlying molecular mechanisms. METHODS We utilized an ovalbumin-induced mouse model to study AR. Both primary and immortalized human nasal epithelial cells (HNEpC) were used to further validate the therapeutic effects of MSC-sEV on epithelial cell function. Then we constructed microRNA (miR)-143 overexpressing and low-expressing HNEpC and MSC-sEV to elucidate molecular mechanisms. Transcriptome analysis was performed to identify the downstream pathways involved. RESULTS MSC-sEV successfully maintained nasal barrier integrity in AR mouse model. The MSC-sEV therapeutic effect on the nasal barrier was substantiated in HNEpC. Mechanistically, miR-143 was a candidate mediator of the above effects. Subsequently, transfecting HNEpC with miR-143 partially mimicked the restoring effect of MSC-sEV. MSC-sEV overexpressing miR-143 exerted more therapeutic effects on tight junctions and barrier integrity. Moreover, miR-143 regulated the glycogen synthase kinase-3β (GSK3B) pathway. CONCLUSIONS Our results indicated that MSC-sEV mitigated AR and restored nasal epithelial barrier dysfunction through the miR-143-GSK3B axis, which suggested that MSC-sEV have the remarkable ability to treat AR.
Collapse
Affiliation(s)
- Meiqian Xu
- The State Key Laboratory of Respiratory Disease, Department of Otolaryngology, Head and Neck Surgery, Laboratory of Otolaryngology-Head and Neck Surgery, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China; Innovation and Transformation Platform of Upper Airway Disease in Guangdong Province, Guangzhou, China
| | - Mei Ren
- The State Key Laboratory of Respiratory Disease, Department of Otolaryngology, Head and Neck Surgery, Laboratory of Otolaryngology-Head and Neck Surgery, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China; Innovation and Transformation Platform of Upper Airway Disease in Guangdong Province, Guangzhou, China
| | - Xinyin Zhang
- The State Key Laboratory of Respiratory Disease, Department of Otolaryngology, Head and Neck Surgery, Laboratory of Otolaryngology-Head and Neck Surgery, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China; Innovation and Transformation Platform of Upper Airway Disease in Guangdong Province, Guangzhou, China
| | - Wenxu Peng
- The State Key Laboratory of Respiratory Disease, Department of Otolaryngology, Head and Neck Surgery, Laboratory of Otolaryngology-Head and Neck Surgery, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China; Innovation and Transformation Platform of Upper Airway Disease in Guangdong Province, Guangzhou, China
| | - Hao Li
- The State Key Laboratory of Respiratory Disease, Department of Otolaryngology, Head and Neck Surgery, Laboratory of Otolaryngology-Head and Neck Surgery, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China; Innovation and Transformation Platform of Upper Airway Disease in Guangdong Province, Guangzhou, China
| | - Wenjing Liao
- The State Key Laboratory of Respiratory Disease, Department of Otolaryngology, Head and Neck Surgery, Laboratory of Otolaryngology-Head and Neck Surgery, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China; Innovation and Transformation Platform of Upper Airway Disease in Guangdong Province, Guangzhou, China.
| | - Jianlei Xie
- The State Key Laboratory of Respiratory Disease, Department of Otolaryngology, Head and Neck Surgery, Laboratory of Otolaryngology-Head and Neck Surgery, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China; Innovation and Transformation Platform of Upper Airway Disease in Guangdong Province, Guangzhou, China.
| | - Xiaowen Zhang
- The State Key Laboratory of Respiratory Disease, Department of Otolaryngology, Head and Neck Surgery, Laboratory of Otolaryngology-Head and Neck Surgery, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China; Innovation and Transformation Platform of Upper Airway Disease in Guangdong Province, Guangzhou, China.
| |
Collapse
|
10
|
Feng C, Hu Z, Zhao M, Leng C, Li G, Yang F, Fan X. Region-specific mitophagy in nucleus pulposus, annulus fibrosus, and cartilage endplate of intervertebral disc degeneration: mechanisms and therapeutic strategies. Front Pharmacol 2025; 16:1579507. [PMID: 40248091 PMCID: PMC12003974 DOI: 10.3389/fphar.2025.1579507] [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: 02/20/2025] [Accepted: 03/24/2025] [Indexed: 04/19/2025] Open
Abstract
Intervertebral disc degeneration (IVDD) is a prevalent condition contributing to various spinal disorders, posing a significant global health burden. Mitophagy plays a crucial role in maintaining mitochondrial quantity and quality and is closely associated with the onset and progression of IVDD. Well-documented region-specific mitophagy mechanisms in IVDD are guiding the development of therapeutic strategies. In the nucleus pulposus (NP), impaired mitochondria lead to apoptosis, oxidative stress, senescence, extracellular matrix degradation and synthesis, excessive autophagy, inflammation, mitochondrial instability, and pyroptosis, with key regulatory targets including AMPK, PGC-1α, SIRT1, SIRT3, Progerin, p65, Mfn2, FOXO3, NDUFA4L2, SLC39A7, ITGα5/β1, Nrf2, and NLRP3 inflammasome. In the annulus fibrosus (AF), mitochondrial damage induces apoptosis and oxidative stress mediated by PGC-1α, while in the cartilage endplate (CEP), mitochondrial dysfunction similarly triggers apoptosis and oxidative stress. These mechanistic insights highlight therapeutic strategies such as activating Parkin-dependent and Ub-independent mitophagy pathways for NP, enhancing Parkin-dependent mitophagy for AF, and targeting Parkin-mediated mitophagy for CEP. These strategies include the use of natural ingredients, hormonal modulation, gene editing technologies, targeted compounds, and manipulation of related proteins. This review summarizes the mechanisms of mitophagy in different regions of the intervertebral disc and highlights therapeutic approaches using mitophagy modulators to ameliorate IVDD. It discusses the complex mechanisms of mitophagy and underscores its potential as a therapeutic target. The objective is to provide valuable insights and a scientific basis for the development of mitochondrial-targeted drugs for anti-IVDD.
Collapse
Affiliation(s)
- Chaoqun Feng
- Department of Orthopedics, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ziang Hu
- Department of Orthopedics, The TCM Hospital of Longquanyi District, Chengdu, China
| | - Min Zhao
- International Ward (Gynecology), Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Chuan Leng
- Department of Orthopedics, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Guangye Li
- Department of Orthopedics, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Fei Yang
- Department of Orthopedics, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaohong Fan
- Department of Orthopedics, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
11
|
Ma C, Fang J, Xu R, Ma L, Zhang G, Yang X, Peng Z, Lu S. Causal Relationship Between Mitochondrial DNA Copy Number and Intervertebral Disc Degeneration: A Bidirectional 2-Sample Mendelian Randomization Study. World Neurosurg 2025; 196:123732. [PMID: 39947317 DOI: 10.1016/j.wneu.2025.123732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 01/21/2025] [Accepted: 01/22/2025] [Indexed: 03/15/2025]
Abstract
OBJECTIVE The causal relationship between mitochondrial DNA copy number (mtDNA-CN) and intervertebral disc degeneration (IVDD) remains unclear. The study aimed to investigate the causal relationship between mtDNA-CN and IVDD using Mendelian randomization (MR) analyses. METHODS The causal relationship between mtDNA-CN and IVDD was estimated using a bidirectional 2-sample MR method. The inverse-variance weighted method was employed as the main MR method. Sensitivity analyses were conducted to validate the robustness and reliability of the MR results. RESULTS The MR results by inverse-variance weighted revealed that genetically predicted mtDNA-CN was not associated with IVDD (odds ratio = 0.91; 95% confidence interval = 0.79-1.04; P = 0.1731). Similar results were observed in other 4 MR methods (P > 0.05). Heterogeneity was found in the analysis of IVDD on mtDNA-CN (P < 0.05), while no horizontal pleiotropy was observed (P > 0.05). Furthermore, the leave-one-out analysis indicated the robustness of MR results not biased by a single nucleotide polymorphism. Moreover, the reverse MR analysis did not reveal any significant causal association of IVDD on mtDNA-CN. CONCLUSIONS The study revealed that there is no causal relationship of mtDNA-CN on the risk of IVDD, nor does IVDD have a significant causal effect on mtDNA-CN. Although the study did not find a significant causal relationship, it provides valuable insights into the complex interplay between genetic factors and IVDD. Further research is needed to explore to better understand the underlying mechanisms of this condition.
Collapse
Affiliation(s)
- Chengjie Ma
- Department of Orthopedics, the First People's Hospital of Yunnan Province & the Affiliated Hospital of Kunming University of Science and Technology, the Key Laboratory of Digital Orthopaedics of Yunnan Province, the Clinical Medicine Center of Spinal and Spinal Cord Disorders of Yunnan Province, Kunming, China
| | - Junhua Fang
- Department of Orthopedics, the First People's Hospital of Yunnan Province & the Affiliated Hospital of Kunming University of Science and Technology, the Key Laboratory of Digital Orthopaedics of Yunnan Province, the Clinical Medicine Center of Spinal and Spinal Cord Disorders of Yunnan Province, Kunming, China
| | - Rui Xu
- Department of Orthopedics, the First People's Hospital of Yunnan Province & the Affiliated Hospital of Kunming University of Science and Technology, the Key Laboratory of Digital Orthopaedics of Yunnan Province, the Clinical Medicine Center of Spinal and Spinal Cord Disorders of Yunnan Province, Kunming, China
| | - Langtao Ma
- Department of Orthopedics, the First People's Hospital of Yunnan Province & the Affiliated Hospital of Kunming University of Science and Technology, the Key Laboratory of Digital Orthopaedics of Yunnan Province, the Clinical Medicine Center of Spinal and Spinal Cord Disorders of Yunnan Province, Kunming, China
| | - Guiqian Zhang
- Department of Orthopedics, the First People's Hospital of Yunnan Province & the Affiliated Hospital of Kunming University of Science and Technology, the Key Laboratory of Digital Orthopaedics of Yunnan Province, the Clinical Medicine Center of Spinal and Spinal Cord Disorders of Yunnan Province, Kunming, China
| | - Xionggang Yang
- Department of Orthopedics, the First People's Hospital of Yunnan Province & the Affiliated Hospital of Kunming University of Science and Technology, the Key Laboratory of Digital Orthopaedics of Yunnan Province, the Clinical Medicine Center of Spinal and Spinal Cord Disorders of Yunnan Province, Kunming, China
| | - Zhi Peng
- Department of Orthopedics, the First People's Hospital of Yunnan Province & the Affiliated Hospital of Kunming University of Science and Technology, the Key Laboratory of Digital Orthopaedics of Yunnan Province, the Clinical Medicine Center of Spinal and Spinal Cord Disorders of Yunnan Province, Kunming, China
| | - Sheng Lu
- Department of Orthopedics, the First People's Hospital of Yunnan Province & the Affiliated Hospital of Kunming University of Science and Technology, the Key Laboratory of Digital Orthopaedics of Yunnan Province, the Clinical Medicine Center of Spinal and Spinal Cord Disorders of Yunnan Province, Kunming, China.
| |
Collapse
|
12
|
Gowtham A, Kaundal RK. Exploring the ncRNA landscape in exosomes: Insights into wound healing mechanisms and therapeutic applications. Int J Biol Macromol 2025; 292:139206. [PMID: 39732230 DOI: 10.1016/j.ijbiomac.2024.139206] [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: 10/26/2024] [Revised: 12/16/2024] [Accepted: 12/24/2024] [Indexed: 12/30/2024]
Abstract
Exosomal non-coding RNAs (ncRNAs), including miRNAs, lncRNAs, and circRNAs, have emerged as crucial modulators in cellular signaling, influencing wound healing processes. Stem cell-derived exosomes, which serve as vehicles for these ncRNAs, show remarkable therapeutic potential due to their ability to modulate wound healing stages, from initial inflammation to collagen formation. These ncRNAs act as molecular signals, regulating gene expression and protein synthesis necessary for cellular responses in healing. Wound healing is a complex, staged process involving inflammation, hemostasis, fibroblast proliferation, angiogenesis, and tissue remodeling. Stem cell-derived exosomal ncRNAs enhance these stages by reducing excessive inflammation, promoting anti-inflammatory responses, guiding fibroblast and keratinocyte maturation, enhancing vascularization, and ensuring organized collagen deposition. Their molecular cargo, particularly ncRNAs, specifically targets pathways to aid chronic wound repair and support scarless regeneration. This review delves into the unique composition and signaling roles of Stem cell-derived exosomes and ncRNAs, highlighting their impact across wound healing stages and their potential as innovative therapeutics. Understanding the interaction between exosomal ncRNAs and cellular signaling pathways opens new avenues in regenerative medicine, positioning Stem cell-derived exosomes and their ncRNAs as promising molecular-level interventions in wound healing.
Collapse
Affiliation(s)
- A Gowtham
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Raebareli (NIPER-R), Transit Campus, Bijnor-Sisendi Road, Sarojini Nagar, Near CRPF Base Camp, Lucknow, UP 226002, India
| | - Ravinder K Kaundal
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Raebareli (NIPER-R), Transit Campus, Bijnor-Sisendi Road, Sarojini Nagar, Near CRPF Base Camp, Lucknow, UP 226002, India.
| |
Collapse
|
13
|
Mai Y, Wu S, Zhang P, Chen N, Wu J, Wei F. The anti-oxidation related bioactive materials for intervertebral disc degeneration regeneration and repair. Bioact Mater 2025; 45:19-40. [PMID: 39588482 PMCID: PMC11585838 DOI: 10.1016/j.bioactmat.2024.10.012] [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: 08/06/2024] [Revised: 10/11/2024] [Accepted: 10/13/2024] [Indexed: 11/27/2024] Open
Abstract
Intervertebral disc degeneration (IVDD) is a prevalent chronic spinal condition characterized by the deterioration of the intervertebral discs (IVD), leading to structural damage and associated pain. This degenerative process is closely linked to oxidative stress injury, which plays a pivotal role in its onset and progression. Oxidative stress in IVDD results from the excessive production of reactive oxygen species (ROS) and impaired ROS clearance mechanisms, disrupting the redox balance within the intervertebral disc. Consequently, oxidative stress contributes to the degradation of the extracellular matrix (ECM), promotes cell apoptosis, and exacerbates disc tissue damage. Current treatment options for IVDD face significant challenges in effectively alleviating the oxidative stress-induced damage and facilitating disc tissue repair. However, recent advancements in biomaterials have opened new avenues of hope for IVDD treatment by addressing oxidative stress. In this review, we first provide an overview of the pathophysiological process of IVDD and explore the mechanisms and pathways associated with oxidative stress injury. Then, we delve into the current research on antioxidant biomaterials employed in the treatment of IVDD, and outline the advantages and limitations of hydrogel, nanomaterials, polyphenol and inorganic materials. Finally, we propose the future research direction of antioxidant biomaterials in IVDD treatment. The main idea of this review is shown in Scheme 1.
Collapse
Affiliation(s)
- Yingjie Mai
- Department of Orthopaedics, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong Province, 518107, China
| | - Siying Wu
- Bioscience and Biomedical Engineering Thrust, The Hong Kong University of Science & Technology (Guangzhou), Nansha, Guangzhou, Guangdong Province, 511400, China
| | - Penghui Zhang
- Department of Orthopaedics, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong Province, 518107, China
| | - Ningning Chen
- Department of Orthopaedics, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong Province, 518107, China
| | - Jun Wu
- Bioscience and Biomedical Engineering Thrust, The Hong Kong University of Science & Technology (Guangzhou), Nansha, Guangzhou, Guangdong Province, 511400, China
- Division of Life Science, The Hong Kong University of Science & Technology, Hong Kong SAR, 999077, China
| | - Fuxin Wei
- Department of Orthopaedics, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong Province, 518107, China
| |
Collapse
|
14
|
Ding Y, Li F, Wang Y, Pan W, Fu X, Tan S. Nanomedicine Approaches for Intervertebral Disc Regeneration: From Bench to Bedside. Pharmaceutics 2025; 17:313. [PMID: 40142977 PMCID: PMC11944988 DOI: 10.3390/pharmaceutics17030313] [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: 01/27/2025] [Revised: 02/18/2025] [Accepted: 02/27/2025] [Indexed: 03/28/2025] Open
Abstract
Intervertebral disc degeneration (IDD) is a leading cause of low back pain (LBP) and neurological dysfunction, contributing significantly to disability-adjusted life years globally. The progression of IDD is driven by excessive oxidative stress, inflammation, apoptosis, and fibrosis, which disrupt the balance between anabolic and catabolic processes, leading to extracellular matrix (ECM) degradation and IDD. Current treatment options, such as conservative therapy and surgical intervention, are limited in halting the disease progression and often exacerbate degeneration in adjacent discs. This review highlights the challenges in treating IDD, particularly due to the limited drug delivery efficiency to the intervertebral disc (IVD). It explores the potential of nanobiomedicine and various nanomaterial-based delivery systems, including nanoparticles, microspheres, gene-nanocomplexes, fullerene, exosomes, and nanomaterial-composite hydrogels. These advanced delivery systems can enhance targeted drug delivery, improve local drug concentration, and sustain drug retention within the IVD, offering promising therapeutic strategies to address IDD. The review also examines the therapeutic effects of these nanomaterials on IDD, focusing on their impact on metabolism, inflammation, apoptosis, fibrosis, and stem cell migration and differentiation, aiming to provide innovative strategies for intervertebral disc regeneration.
Collapse
Affiliation(s)
- Yifan Ding
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.D.); (F.L.)
| | - Fan Li
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.D.); (F.L.)
| | - Yunyun Wang
- Department of Cardiology, the Fifth Hospital of Wuhan, Jianghan University, Wuhan 430030, China;
| | - Weizhen Pan
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China;
| | - Xiangning Fu
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.D.); (F.L.)
| | - Songwei Tan
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China;
| |
Collapse
|
15
|
Li H, Zhang P, Lin M, Li K, Zhang C, He X, Gao K. Pyroptosis: candidate key targets for mesenchymal stem cell-derived exosomes for the treatment of bone-related diseases. Stem Cell Res Ther 2025; 16:68. [PMID: 39940049 PMCID: PMC11816542 DOI: 10.1186/s13287-025-04167-y] [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: 10/21/2024] [Accepted: 01/21/2025] [Indexed: 02/14/2025] Open
Abstract
Bone-related diseases impact a large portion of the global population and, due to their high disability rates and limited treatment options, pose significant medical and economic challenges. Mesenchymal stem cells (MSCs) can differentiate into multiple cell types and offer strong regenerative potential, making them promising for treating various diseases. However, issues with the immune response and cell survival limit the effectiveness of cell transplantation. This has led to increased interest in cell-free stem cell therapy, particularly the use of exosomes, which is the most studied form of this approach. Exosomes are extracellular vesicles that contain proteins, lipids, and nucleic acids and play a key role in cell communication and material exchange. Pyroptosis, a form of cell death involved in innate immunity, is also associated with many diseases. Studies have shown that MSC-derived exosomes have therapeutic potential for treating a range of conditions by regulating inflammation and pyroptosis. This study explored the role of MSC-derived exosomes in modulating pyroptosis to improve the treatment of bone-related diseases.
Collapse
Affiliation(s)
- Haiming Li
- Shandong University of Traditional Chinese Medicine, Jinan, CN, China
| | - Peng Zhang
- Department of Orthopaedics, Jining No. 1 People's Hospital, Jining, 272011, People's Republic of China
| | - Minghui Lin
- Shandong University of Traditional Chinese Medicine, Jinan, CN, China
| | - Kang Li
- Department of Spine Surgery, Jining No. 1 People's Hospital, Jining, 272011, People's Republic of China
| | - Cunxin Zhang
- Department of Spine Surgery, Jining No. 1 People's Hospital, Jining, 272011, People's Republic of China.
| | - Xiao He
- Department of Orthopaedics, Jining No. 1 People's Hospital, Jining, 272011, People's Republic of China.
| | - Kai Gao
- Shandong University of Traditional Chinese Medicine, Jinan, CN, China.
- Department of Orthopaedics, Jining No. 1 People's Hospital, Jining, 272011, People's Republic of China.
| |
Collapse
|
16
|
Ganesh BH, Padinjarathil H, Rajendran RL, Ramani P, Gangadaran P, Ahn BC. The Role of Extracellular Vesicles in Aging and Age-Related Disorders. Antioxidants (Basel) 2025; 14:177. [PMID: 40002364 PMCID: PMC11851802 DOI: 10.3390/antiox14020177] [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: 12/24/2024] [Revised: 01/18/2025] [Accepted: 01/31/2025] [Indexed: 02/27/2025] Open
Abstract
A variety of molecular and cellular changes distinguish the multifaceted biological process of aging. Recent studies in this decade have demonstrated the essential role of extracellular vesicles (EVs) in the aging process. Mitochondrial malfunction and increased oxidative stress are major contributors for the aging process. This review investigates the role of EVs in intercellular communication, tissue regeneration, and inflammation in the context of aging. We also discuss the exosome and its utility to reduce oxidative stress, which is a key part of aging, as well as the possibility of using the exosomes (EVs) as anti-aging drugs. Changes in cargo composition can influence the aging phenotype and impact the functionality of cells and tissues. Additionally, the role of EVs in oxidative stress during the aging process addresses potential treatment strategies and the development of biomarkers for age-associated disorders. The review also highlighted the role of exosomes in providing antioxidant properties, which help reduce excessive reactive oxygen species (ROS) and strengthen cellular defenses against oxidative stress. Additionally, it emphasized the role of extracellular vesicles (EVs) in age-related pathologies, such as neurodegenerative diseases, cardiovascular disorders, and immunosenescence, offering insights into targeted interventions for promoting healthy aging. This article provides a comprehensive analysis of the current body of knowledge regarding the therapeutic effects of EVs on aging, with a particular emphasis on the implications of this emerging field of research and its relationship to oxidative stress.
Collapse
Affiliation(s)
- Bharathi Hassan Ganesh
- Dhanvanthri Laboratory, Department of Chemistry, Amrita School of Physical Sciences, Amrita Vishwa Vidyapeetham, Coimbatore 641112, India; (B.H.G.); (H.P.)
- Center of Excellence in Advanced Materials and Green Technologies (CoE-AMGT), Amrita School of Engineering, Amrita Vishwa Vidyapeetham, Coimbatore 641112, India
| | - Himabindu Padinjarathil
- Dhanvanthri Laboratory, Department of Chemistry, Amrita School of Physical Sciences, Amrita Vishwa Vidyapeetham, Coimbatore 641112, India; (B.H.G.); (H.P.)
- Center of Excellence in Advanced Materials and Green Technologies (CoE-AMGT), Amrita School of Engineering, Amrita Vishwa Vidyapeetham, Coimbatore 641112, India
| | - Ramya Lakshmi Rajendran
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea; (R.L.R.); (P.G.)
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
- Cardiovascular Research Institute, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Prasanna Ramani
- Dhanvanthri Laboratory, Department of Chemistry, Amrita School of Physical Sciences, Amrita Vishwa Vidyapeetham, Coimbatore 641112, India; (B.H.G.); (H.P.)
- Center of Excellence in Advanced Materials and Green Technologies (CoE-AMGT), Amrita School of Engineering, Amrita Vishwa Vidyapeetham, Coimbatore 641112, India
| | - Prakash Gangadaran
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea; (R.L.R.); (P.G.)
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
- Cardiovascular Research Institute, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Byeong-Cheol Ahn
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea; (R.L.R.); (P.G.)
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
- Cardiovascular Research Institute, Kyungpook National University, Daegu 41944, Republic of Korea
| |
Collapse
|
17
|
Wang X, Huang Y, Yang Y, Tian X, Jin Y, Jiang W, He H, Xu Y, Liu Y. Polysaccharide-based biomaterials for regenerative therapy in intervertebral disc degeneration. Mater Today Bio 2025; 30:101395. [PMID: 39759846 PMCID: PMC11699348 DOI: 10.1016/j.mtbio.2024.101395] [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: 09/29/2024] [Revised: 11/27/2024] [Accepted: 12/08/2024] [Indexed: 01/07/2025] Open
Abstract
Intervertebral disc (IVD) degeneration represents a significant cause of chronic back pain and disability, with a substantial impact on the quality of life. Conventional therapeutic modalities frequently address the symptoms rather than the underlying etiology, underscoring the necessity for regenerative therapies that restore disc function. Polysaccharide-based materials, such as hyaluronic acid, alginate, chitosan, and chondroitin sulfate, have emerged as promising candidates for intervertebral disc degeneration (IVDD) therapy due to their biocompatibility, biodegradability, and ability to mimic the native extracellular matrix (ECM) of the nucleus pulposus (NP). These materials have demonstrated the capacity to support cell viability, facilitate matrix production, and alleviate inflammation in vitro and in vivo, thus supporting tissue regeneration and restoring disc function in comparison to conventional treatment. Furthermore, polysaccharide-based hydrogels have demonstrated the potential to deliver bioactive molecules, including growth factors, cytokines and anti-inflammatory drugs, directly to the degenerated disc environment, thereby enhancing therapeutic outcomes. Therefore, polysaccharide-based materials provide structural support and facilitate the regeneration of native tissue, representing a versatile and effective approach for the treatment of IVDD. Despite their promise, challenges such as limited long-term stability, potential immunogenicity, and the difficulty in scaling up production for clinical use remain. This review delineates the potential of various polysaccharides during the fabrication of hydrogels and scaffolds for disc regeneration, guiding and inspiring future research to focus on optimizing these materials for clinical translation for IVDD repair and regeneration.
Collapse
Affiliation(s)
- Xin Wang
- Department of Orthopaedic Surgery, The Fourth Affiliated Hospital of Soochow University, Suzhou Medical College, Soochow University, Suzhou, 215000, China
| | - Yixue Huang
- Department of Orthopedic Surgery, The First Affiliated Hospital, Orthopedic Institute, MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College, Soochow University, Suzhou, 215000, Jiangsu, China
| | - Yilin Yang
- Department of Orthopedic Surgery, The First Affiliated Hospital, Orthopedic Institute, MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College, Soochow University, Suzhou, 215000, Jiangsu, China
| | - Xin Tian
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Centre for Bone and Arthritis Research at the Sahlgrenska Academy, University of Gothenburg, Gothenburg, 41346, Sweden
| | - Yesheng Jin
- Department of Orthopedic Surgery, The First Affiliated Hospital, Orthopedic Institute, MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College, Soochow University, Suzhou, 215000, Jiangsu, China
| | - Weimin Jiang
- Department of Orthopaedic Surgery, The Fourth Affiliated Hospital of Soochow University, Suzhou Medical College, Soochow University, Suzhou, 215000, China
| | - Hanliang He
- Department of Orthopaedic Surgery, The Fourth Affiliated Hospital of Soochow University, Suzhou Medical College, Soochow University, Suzhou, 215000, China
| | - Yong Xu
- Department of Orthopedic Surgery, The First Affiliated Hospital, Orthopedic Institute, MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College, Soochow University, Suzhou, 215000, Jiangsu, China
| | - Yijie Liu
- Department of Orthopaedic Surgery, The Fourth Affiliated Hospital of Soochow University, Suzhou Medical College, Soochow University, Suzhou, 215000, China
| |
Collapse
|
18
|
Tork MAB, Fotouhi S, Roozi P, Negah SS. Targeting NLRP3 Inflammasomes: A Trojan Horse Strategy for Intervention in Neurological Disorders. Mol Neurobiol 2025; 62:1840-1881. [PMID: 39042218 DOI: 10.1007/s12035-024-04359-2] [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: 02/05/2024] [Accepted: 07/09/2024] [Indexed: 07/24/2024]
Abstract
Recently, a growing focus has been on identifying critical mechanisms in neurological diseases that trigger a cascade of events, making it easier to target them effectively. One such mechanism is the inflammasome, an essential component of the immune response system that plays a crucial role in disease progression. The NLRP3 (nucleotide-binding oligomerization domain, leucine-rich repeat, and pyrin domain containing 3) inflammasome is a subcellular multiprotein complex that is widely expressed in the central nervous system (CNS) and can be activated by a variety of external and internal stimuli. When activated, the NLRP3 inflammasome triggers the production of proinflammatory cytokines interleukin-1β (IL-1β) and interleukin-18 (IL-18) and facilitates rapid cell death by assembling the inflammasome. These cytokines initiate inflammatory responses through various downstream signaling pathways, leading to damage to neurons. Therefore, the NLRP3 inflammasome is considered a significant contributor to the development of neuroinflammation. To counter the damage caused by NLRP3 inflammasome activation, researchers have investigated various interventions such as small molecules, antibodies, and cellular and gene therapy to regulate inflammasome activity. For instance, recent studies indicate that substances like micro-RNAs (e.g., miR-29c and mR-190) and drugs such as melatonin can reduce neuronal damage and suppress neuroinflammation through NLRP3. Furthermore, the transplantation of bone marrow mesenchymal stem cells resulted in a significant reduction in the levels of pyroptosis-related proteins NLRP3, caspase-1, IL-1β, and IL-18. However, it would benefit future research to have an in-depth review of the pharmacological and biological interventions targeting inflammasome activity. Therefore, our review of current evidence demonstrates that targeting NLRP3 inflammasomes could be a pivotal approach for intervention in neurological disorders.
Collapse
Affiliation(s)
- Mohammad Amin Bayat Tork
- Clinical Research Development Unit, Imam Reza Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Soroush Fotouhi
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Parvin Roozi
- Department of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Sajad Sahab Negah
- Clinical Research Development Unit, Imam Reza Hospital, Mashhad University of Medical Sciences, Mashhad, Iran.
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran.
- Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Pardis Campus, Azadi Square, Kalantari Blvd., Mashhad, Iran.
| |
Collapse
|
19
|
Martinez-Zalbidea I, Wagner G, Bergendahl N, Mesfin A, Puvanesarajah V, Hitzl W, Schulze S, Wuertz-Kozak K. CRISPR-dCas9 Activation of TSG-6 in MSCs Modulates the Cargo of MSC-Derived Extracellular Vesicles and Attenuates Inflammatory Responses in Human Intervertebral Disc Cells In Vitro. Cell Mol Bioeng 2025; 18:83-98. [PMID: 39949490 PMCID: PMC11813855 DOI: 10.1007/s12195-025-00843-4] [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: 10/21/2024] [Accepted: 01/16/2025] [Indexed: 02/16/2025] Open
Abstract
Purpose The purpose of this study was to boost the therapeutic effect of mesenchymal stem cell (MSC)-derived extracellular vesicles (EVs) by overexpressing the gene TSG-6 through CRISPR activation, and assess the biological activity of EVs from these modified MSCs in vitro on human intervertebral disc (IVD) cells. Methods An immortalized human MSC line was transduced with a CRISPR activation lentivirus system targeting TSG-6. MSC-EVs were harvested by ultracentrifugation and particle number/size distribution was determined by nanoparticle tracking analysis. The efficiency of transduction activation was assessed by analyzing gene and protein expression. EV proteomic contents were analyzed by mass spectrometry. Human IVD cells from patients undergoing spinal surgery were isolated, expanded, exposed to IL-1β pre-stimulation and co-treated with MSC-EVs. Results MSC-EVs presented size distribution, morphology, and molecular markers consistent with common EV characteristics. The expression level of TSG-6 was significantly higher (> 800 fold) in transduced MSCs relative to controls. Protein analysis of MSCs and EVs showed higher protein expression of TSG-6 in CRISPR activated samples than controls. Proteomics of EVs identified 35 proteins (including TSG-6) that were differentially expressed in TSG-6 activated EVs vs control EVs. EV co-Treatment of IL-1β pre-Stimulated IVD cells resulted in a significant downregulation of IL-8 and COX-2. Conclusions We successfully generated an MSC line overexpressing TSG-6. Furthermore, we show that EVs isolated from these modified MSCs have the potential to attenuate the pro-inflammatory gene expression in IVD cells. This genomic engineering approach hence holds promise for boosting the therapeutic effects of EVs. Supplementary Information The online version contains supplementary material available at 10.1007/s12195-025-00843-4.
Collapse
Affiliation(s)
- Iker Martinez-Zalbidea
- Department of Biomedical Engineering, Rochester Institute of Technology (RIT), Rochester, NY USA
| | - Gabbie Wagner
- Department of Biomedical Engineering, Rochester Institute of Technology (RIT), Rochester, NY USA
| | - Nea Bergendahl
- Department of Biomedical Engineering, Rochester Institute of Technology (RIT), Rochester, NY USA
| | - Addisu Mesfin
- Medstar Orthopaedic Institute, Georgetown University School of Medicine Washington, Washington, DC USA
| | - Varun Puvanesarajah
- Department of Orthopedics and Rehabilitation, University of Rochester Medical Center, Rochester, NY USA
| | - Wolfgang Hitzl
- Research and Innovation Management (RIM), Paracelsus Medical University, Salzburg, Austria
- Department of Ophthalmology and Optometry, Paracelsus Medical University, Salzburg, Austria
- Research Program Experimental Ophthalmology and Glaucoma Research, Paracelsus Medical University, Salzburg, Austria
| | - Stefan Schulze
- Thomas H. Gosnell School of Life Sciences, Rochester Institute of Technology, Rochester, NY USA
| | - Karin Wuertz-Kozak
- Department of Biomedical Engineering, Rochester Institute of Technology (RIT), Rochester, NY USA
- Schön Clinic Munich Harlaching, Spine Center, Academic Teaching Hospital and Spine Research Institute of the Paracelsus Medical University Salzburg (Austria), Munich, Germany
| |
Collapse
|
20
|
Han J, Xu K, Xu T, Song Q, Duan T, Yang J. The functional regulation between extracellular vesicles and the DNA damage responses. MUTATION RESEARCH. REVIEWS IN MUTATION RESEARCH 2025; 795:108532. [PMID: 39828141 DOI: 10.1016/j.mrrev.2025.108532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 01/04/2025] [Accepted: 01/14/2025] [Indexed: 01/22/2025]
Abstract
The DNA damage response (DDR) is a crucial regulatory mechanism for the survival of organisms, and irregularity of DDR may contribute to the development of various diseases, including tumors, making it is a prominent topic in therapeutic research. Extracellular vesicles (EVs), as important mediators of intercellular communication, have been extensively studied in recent years. Notably, an increasing number of studies have revealed a strong connection between DDR and EVs. On one hand, DNA damage affects the release of EVs and their compositional content; on the other hand, EVs can dictate cell survival or death by modulating DDR in both the parental and the recipient cells. This review outlines current progress in the inter-regulatory relationship between EVs and DDR, with special emphasis on the effects of EVs derived from various sources on DDR in recipient cells. In addition, the potential applications of EVs in research and tumor therapy are discussed.
Collapse
Affiliation(s)
- Jinyi Han
- Department of Nutrition and Toxicology, Hangzhou Normal University School of Public Health, Hangzhou, China
| | - Kexin Xu
- Department of Nutrition and Toxicology, Hangzhou Normal University School of Public Health, Hangzhou, China
| | - Ting Xu
- Department of Nutrition and Toxicology, Hangzhou Normal University School of Public Health, Hangzhou, China
| | - Qin Song
- Department of Nutrition and Toxicology, Hangzhou Normal University School of Public Health, Hangzhou, China
| | - Ting Duan
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China.
| | - Jun Yang
- Department of Nutrition and Toxicology, Hangzhou Normal University School of Public Health, Hangzhou, China; Zhejiang Provincial Center for Uterine Cancer Diagnosis and Therapy Research, The Affiliated Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
21
|
Chen W, Yang W, Meng B, Wang X, Duan H, Xu Q, Li H. Validation and the role of PDK4 relevant to ferroptosis in degenerative lumbar disc disease. J Orthop Surg Res 2025; 20:30. [PMID: 39794775 PMCID: PMC11724506 DOI: 10.1186/s13018-024-05293-8] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 11/18/2024] [Indexed: 01/30/2025] Open
Abstract
BACKGROUND Ferroptosis was involved in the pathogenesis of intervertebral disc degeneration (IVDD). However, the exact mechanism of IVDD associated with ferroptosis still required deeper studies. METHOD The differentially expressed genes (DEGs) in rat lumbar disc tissue between the control and IVDD group treated with IL-1β were detected by RNA sequencing (RNA-seq). Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis were performed on DEGs. We further screened the differential expressed ferroptosis-related genes (DEFRGs). Besides, a protein-protein interaction (PPI) network of DEFRGs was constructed by STRING database. The Cytoscape database identified significant modules and the hub genes. The loss function of PDK4 by siRNA inference was investigated in NPCs by CCK8 assay, ELISA assay, and the analysis of ferroptosis indicators. RESULT DEGs were identified using RNA-seq. KEGG pathway analysis showed that these genes were mainly involved in Parkinson's disease, oxytocin signaling pathway, calcium ion signaling pathway, AMPK signaling pathway, and glucagon signaling pathway. Eight hub genes (including LDHA, PKM, EP300, EGFR, EGLN1, SCD, PDK4, and FABP4) were found by the PPI network and Cytoscape on a total of 25 ferroptosis-related genes that were identified in rat lumbar disc tissue after IVDD treatment. PDK4 silencing promoted NPCS proliferation, decreased the levels of the proinflammatory factors, and suppressed ferroptosis. CONCLUSION The study suggested the potential roles of ferroptosis-related genes in IVDD and further revealed the role of PDK4 in the progression of IVDD.
Collapse
Affiliation(s)
- Wenhao Chen
- Department of Orthopaedics, Qilu Hospital of Shandong University, No.107, Wenhuaxi Road, Lixia District, Jinan, Shandong Province, 250012, China
- Cheeloo College of Medicine, Shandong University, No.107, Wenhuaxi Road, Lixia District, Jinan, Shandong Province, 250012, China
| | - Wanliang Yang
- Department of Orthopaedics, Qilu Hospital of Shandong University, No.107, Wenhuaxi Road, Lixia District, Jinan, Shandong Province, 250012, China
- Cheeloo College of Medicine, Shandong University, No.107, Wenhuaxi Road, Lixia District, Jinan, Shandong Province, 250012, China
| | - Bin Meng
- Department of Orthopaedics, Qilu Hospital of Shandong University, No.107, Wenhuaxi Road, Lixia District, Jinan, Shandong Province, 250012, China
- Cheeloo College of Medicine, Shandong University, No.107, Wenhuaxi Road, Lixia District, Jinan, Shandong Province, 250012, China
| | - Xingkun Wang
- Department of Orthopaedics, Qilu Hospital of Shandong University, No.107, Wenhuaxi Road, Lixia District, Jinan, Shandong Province, 250012, China
- Cheeloo College of Medicine, Shandong University, No.107, Wenhuaxi Road, Lixia District, Jinan, Shandong Province, 250012, China
| | - Heng Duan
- Department of Orthopaedics, Qilu Hospital of Shandong University, No.107, Wenhuaxi Road, Lixia District, Jinan, Shandong Province, 250012, China
- Cheeloo College of Medicine, Shandong University, No.107, Wenhuaxi Road, Lixia District, Jinan, Shandong Province, 250012, China
| | - Qian Xu
- Department of Orthopaedics, Qilu Hospital of Shandong University, No.107, Wenhuaxi Road, Lixia District, Jinan, Shandong Province, 250012, China
- Cheeloo College of Medicine, Shandong University, No.107, Wenhuaxi Road, Lixia District, Jinan, Shandong Province, 250012, China
| | - Hao Li
- Department of Orthopaedics, Qilu Hospital of Shandong University, No.107, Wenhuaxi Road, Lixia District, Jinan, Shandong Province, 250012, China.
| |
Collapse
|
22
|
Wang T, Wang J, Sun T, Zhang R, Li Y, Hu T. PU.1 regulates osteoarthritis progression via CSF1R in synovial cells. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167525. [PMID: 39313038 DOI: 10.1016/j.bbadis.2024.167525] [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: 10/15/2023] [Revised: 08/24/2024] [Accepted: 09/16/2024] [Indexed: 09/25/2024]
Abstract
This study elucidates the molecular mechanisms driving osteoarthritis (OA) by focusing on the transcription factor PU.1's role in synovial cells, specifically macrophages and fibroblast-like synoviocytes (FLS). Analyzing OA-related synovial gene expression from the GEO database highlighted immune regulation pathways in OA. Using protein-protein interaction and the JASPAR database, we pinpointed essential genes in OA development. Synovial tissues from OA patients and controls revealed pronounced PU.1 and its target CSF1R presence. In a surgically induced OA mouse model with PU.1 and CSF1R knockdown, ChIP assays confirmed PU.1's binding to the CSF1R promoter. Dual luciferase reporter assays and immunohistochemistry validated PU.1's regulatory impact on CSF1R transcription. Combined analysis of microarrays GSE55235 and GSE206848 showed heightened PU.1 expression in OA, associated with immune regulation in macrophages. In vitro findings aligned with in vivo results, emphasizing PU.1's influence on macrophage polarization and FLS-induced inflammation. PU.1's direct activation of CSF1R transcription underpins its key role in OA progression. This research offers insights into OA's molecular basis, suggesting potential therapeutic targets.
Collapse
Affiliation(s)
- Tingting Wang
- Department of Gerontology, The First Hospital of China Medical University, Shenyang 110001, PR China
| | - Jiakai Wang
- Department of Rheumatology and Immunology, The First Hospital of China Medical University, Shenyang 110001, PR China
| | - Tao Sun
- Department of Rheumatology and Immunology, The First Hospital of China Medical University, Shenyang 110001, PR China
| | - Rong Zhang
- Department of Rheumatology and Immunology, The First Hospital of China Medical University, Shenyang 110001, PR China
| | - Yishuo Li
- Department of Rheumatology and Immunology, The First Hospital of China Medical University, Shenyang 110001, PR China.
| | - Tianyu Hu
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang 110001, PR China.
| |
Collapse
|
23
|
Chen S, Dou Y, Zhang Y, Sun X, Liu X, Yang Q. Innovating intervertebral disc degeneration therapy: Harnessing the power of extracellular vesicles. J Orthop Translat 2025; 50:44-55. [PMID: 39868351 PMCID: PMC11761297 DOI: 10.1016/j.jot.2024.09.014] [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: 07/02/2024] [Revised: 09/11/2024] [Accepted: 09/26/2024] [Indexed: 01/28/2025] Open
Abstract
Intervertebral disc degeneration is the leading cause of low back pain, imposing significant burdens on patients, societies, and economies. Advancements in regenerative medicine have spotlighted extracellular vesicles as promising nanoparticles for intervertebral disc degeneration treatment. Extracellular vesicles retain the potential of cell therapy and serve as carriers to deliver their cargo to target cells, thereby regulating cell activity. This review summarizes the biogenesis and molecular composition of extracellular vesicles and explores their therapeutic roles in intervertebral disc degeneration treatment through various mechanisms. These mechanisms include mitigating cell loss and senescence, delaying extracellular matrix degeneration, and modulating the inflammatory microenvironment. Additionally, it highlights recent efforts in engineering extracellular vesicles to enhance their targeting and therapeutic efficacy. The integration of extracellular vesicle-based acellular therapy is anticipated to drive significant advancements in disc regenerative medicine. The translational potential of this article Existing clinical treatment strategies often fail to effectively address the challenges associated with regenerating degenerated intervertebral discs. As a new regenerative medicine strategy, the extracellular vesicle strategy avoids the risks associated with cell transplantation and shows great promise in treating intervertebral disc degeneration by carrying therapeutic cargo. This review comprehensively examines the latest research, underlying mechanisms, and therapeutic potential of extracellular vesicles, offering a promising new strategy for intervertebral disc degeneration treatment.
Collapse
Affiliation(s)
- Shanfeng Chen
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin, China
- Clinical School of Orthopedics, Tianjin Medical University, Tianjin, China
| | - Yiming Dou
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin, China
| | - Yiming Zhang
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin, China
- Clinical School of Orthopedics, Tianjin Medical University, Tianjin, China
| | - Xun Sun
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin, China
| | - Xinyu Liu
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Qiang Yang
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin, China
| |
Collapse
|
24
|
Lan T, Yu M, Ming T, Wang H, Deng J, Cheng S, Shen Z, Kong D. A novel cytoprotective organ perfusion platform for reconstructing homeostasis of DCD liver while alleviating IRI injury. Bioeng Transl Med 2025; 10:e10724. [PMID: 39801755 PMCID: PMC11711209 DOI: 10.1002/btm2.10724] [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: 01/31/2024] [Revised: 07/21/2024] [Accepted: 09/06/2024] [Indexed: 01/16/2025] Open
Abstract
Pump is a vital component for expelling the perfusate in small animal isolated organ normothermic machine perfusion (NMP) systems whose flexible structure and rhythmic contraction play a crucial role in maintaining perfusion system homeostasis. However, the continuous extrusion forming with the rigid stationary shaft of the peristaltic pumps can damage cells, leading to metabolic disorders and eventual dysfunction of transplanted organs. Here, we developed a novel biomimetic blood-gas system (BBGs) for preventing cell damage. This system mimics the cardiac cycle and features an adjustable inspiratory-to-expiratory (IE) ratio to mitigate acidosis caused by continuous oxygen inhalation. In our study, adipose stem cells (ADSCs) were cultured within the circulatory system for 10 min, 2, and 4 h. Compared to the peristaltic pump, the BBGs significantly reduced cell apoptosis and morphological injury while enhancing cell proliferation and adhesion. Additionally, when the supernatant from ADSCs was introduced to LPS-induced macrophages for 24 h, the BBGs group demonstrated a more pronounced anti-inflammatory effect, characterized by reduced M1 macrophage expression. Besides, with isolated rat livers from donation after circulatory death (DCD) perfusion with ADSCs for 6 h by the BBGs, we detected fewer apoptotic cells and a reduced inflammatory response, evidenced by down-regulated TNF-α expression. The development of BBGs demonstrates the feasibility of recreating physiological liquid-gas circulation in vitro, offering an alternative platform for isolated organ perfusion, especially for applications involving cell therapy.
Collapse
Affiliation(s)
- Tingting Lan
- Research Institute of Transplant Medicine, Tianjin First Central Hospital, School of Medicine, Nankai UniversityTianjinChina
| | - Mingxing Yu
- State Key Laboratory of Medicinal Chemical BiologyCollege of Life Science, Nankai UniversityTianjinChina
| | - Tao Ming
- Research Institute of Transplant Medicine, Tianjin First Central Hospital, School of Medicine, Nankai UniversityTianjinChina
| | - Hong Wang
- Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinChina
| | - Juan Deng
- Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinChina
| | - Shuhan Cheng
- State Key Laboratory of Medicinal Chemical BiologyCollege of Life Science, Nankai UniversityTianjinChina
| | - Zhongyang Shen
- Research Institute of Transplant Medicine, Tianjin First Central Hospital, School of Medicine, Nankai UniversityTianjinChina
| | - Deling Kong
- Research Institute of Transplant Medicine, Tianjin First Central Hospital, School of Medicine, Nankai UniversityTianjinChina
- State Key Laboratory of Medicinal Chemical BiologyCollege of Life Science, Nankai UniversityTianjinChina
| |
Collapse
|
25
|
Wang J, Zhang Y, Wang S, Wang X, Jing Y, Su J. Bone aging and extracellular vesicles. Sci Bull (Beijing) 2024; 69:3978-3999. [PMID: 39455324 DOI: 10.1016/j.scib.2024.10.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 08/01/2024] [Accepted: 10/14/2024] [Indexed: 10/28/2024]
Abstract
Bone aging, a major global health concern, is the natural decline in bone mass and strength. Concurrently, extracellular vesicles (EVs), tiny membrane-bound particles produced by cells, have gained recognition for their roles in various physiological processes and age-related diseases. The interaction between EVs and bone aging is of growing interest, particularly their effects on bone metabolism, which become increasingly critical with advancing age. In this review, we explored the biology, types, and functions of EVs and emphasized their regulatory roles in bone aging. We examined the effects of EVs on bone metabolism and highlighted their potential as biomarkers for monitoring bone aging progression. Furthermore, we discussed the therapeutic applications of EVs, including targeted drug delivery and bone regeneration, and addressed the challenges associated with EV-based therapies, including the technical complexities and regulatory issues. We summarized the current research and clinical trials investigating the role of EVs in bone aging and suggested future research directions. These include the potential for personalized medicine using EVs and the integration of EV research with advanced technologies to enhance the management of age-related bone health. This analysis emphasized the transformative potential of EVs in understanding and managing bone aging, thereby marking a significant advancement in skeletal health research.
Collapse
Affiliation(s)
- Jian Wang
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Trauma Orthopedics Center, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Institute of Musculoskeletal Injury and Translational Medicine of Organoids, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Institute of Translational Medicine, Shanghai University, Shanghai 200444, China; School of Medicine, Shanghai University, Shanghai 200444, China; National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai 200444, China
| | - Yuanwei Zhang
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Trauma Orthopedics Center, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Institute of Musculoskeletal Injury and Translational Medicine of Organoids, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Institute of Translational Medicine, Shanghai University, Shanghai 200444, China; National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai 200444, China
| | - Sicheng Wang
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China; National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai 200444, China; Department of Orthopedics, Shanghai Zhongye Hospital, Shanghai 200941, China
| | - Xinglong Wang
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, AZ 85721, USA.
| | - Yingying Jing
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China; National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai 200444, China.
| | - Jiacan Su
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Trauma Orthopedics Center, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Institute of Musculoskeletal Injury and Translational Medicine of Organoids, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Institute of Translational Medicine, Shanghai University, Shanghai 200444, China; National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai 200444, China.
| |
Collapse
|
26
|
Taylor W, Erwin WM. Intervertebral Disc Degeneration and Regeneration: New Molecular Mechanisms and Therapeutics: Obstacles and Potential Breakthrough Technologies. Cells 2024; 13:2103. [PMID: 39768194 PMCID: PMC11674193 DOI: 10.3390/cells13242103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 12/09/2024] [Accepted: 12/16/2024] [Indexed: 01/11/2025] Open
Abstract
Pain and disability secondary to degenerative disc disease continue to burden the healthcare system, creating an urgent need for effective, disease-modifying therapies. Contemporary research has identified potential therapies that include protein-, cellular- and/or matrix-related approaches; however, none have yet achieved a meaningful clinical impact. The tissue-specific realities of the intervertebral disc create considerable therapeutic challenges due to the disc's location, compartmentalization, hypovascularization and delicate physiological environment. Furthermore, the imaging modalities currently used in practice are largely unable to accurately identify sources of pain ostensibly discogenic in origin. These obstacles are considerable; however, recent research has begun to shed light on possible breakthrough technologies. Such breakthroughs include revolutionary imaging to better identify tissue sources of pain. Furthermore, novel molecular therapies have been shown to be able to mediate the progression of degenerative disc disease in some large animal studies, and even provide some insight into suppressing the development of tissue sources of discogenic pain. These potential breakthrough technologies have yet to be translated for clinical use.
Collapse
Affiliation(s)
- William Taylor
- Department of Surgery, Division of Neurosurgery, University of California at San Diego, 9350 Campus Point Dr., La Jolla, CA 92037, USA;
| | - William Mark Erwin
- Department of Surgery, Divisions of Orthopaedic and Neurosurgery, University of Toronto, 661 University Ave., Suite 13-1387, Toronto, ON M5G 0B7, Canada
| |
Collapse
|
27
|
Sun H, Guo J, Xiong Z, Zhuang Y, Ning X, Liu M. Targeting nucleus pulposus cell death in the treatment of intervertebral disc degeneration. JOR Spine 2024; 7:e70011. [PMID: 39703198 PMCID: PMC11655182 DOI: 10.1002/jsp2.70011] [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: 03/28/2024] [Revised: 08/27/2024] [Accepted: 10/10/2024] [Indexed: 12/21/2024] Open
Abstract
Background Intervertebral disc degeneration (IDD) is a progressive age-related disorder characterized by the reduction in the number of nucleus pulposus cells (NPCs) and degradation of extracellular matrix (ECM), thereby leading to chronic pain and disability. The pathogenesis of IDD is multifaceted, and current therapeutic strategies remain limited. The nucleus pulposus (NP), primarily composed of NPCs, proteoglycans, and type II collagen, constitutes essential components for maintaining intervertebral disc (IVD) function and spinal motion. The disturbed homeostasis of NPCs is closely associated with IDD. Accumulating evidence increasingly suggests the crucial role of programmed cell death (PCD) in regulating the homeostasis of NPCs. Aims This review aimed to elucidate various forms of PCD and their respective roles in IDD, and investigate diverse strategies targeting the cell death of NPCs for IDD treatment. Materials & Methods We collected the relevant literature regarding PCD and their roles in the development of IDD. Subsequently, we comprehensively summarized the intricate association between PCD and IDD, and also explored the potential and application of cell therapy and traditional Chinese medicine (TCM) in the prevention and treatment of IDD. Results Current literature indicated that the PCD of NPCs was closely associated with the pathogenesis of IDD. Additionally, the development of targeted pharmaceuticals based on the mechanisms of PCD could effectively impede the loss of NPCs. Conclusion This review demonstrated that targeting the PCD of NPCs may be a promising strategy for the treatment of IDD.
Collapse
Affiliation(s)
- Hong Sun
- Department of OrthopaedicsAffiliated Hospital of Guizhou Medical UniversityGuiyangChina
| | - Jiajie Guo
- Department of OrthopaedicsAffiliated Hospital of Guizhou Medical UniversityGuiyangChina
- School of Clinical Medicine, Guizhou Medical UniversityGuiyangChina
| | - Zhilin Xiong
- Department of OrthopaedicsAffiliated Hospital of Guizhou Medical UniversityGuiyangChina
- School of Clinical Medicine, Guizhou Medical UniversityGuiyangChina
| | - Yong Zhuang
- Department of OrthopaedicsAffiliated Hospital of Guizhou Medical UniversityGuiyangChina
| | - Xu Ning
- Department of OrthopaedicsAffiliated Hospital of Guizhou Medical UniversityGuiyangChina
| | - Miao Liu
- Department of OrthopaedicsAffiliated Hospital of Guizhou Medical UniversityGuiyangChina
| |
Collapse
|
28
|
Zhou H, Wu C, Jin Y, Wu O, Chen L, Guo Z, Wang X, Chen Q, Kwan KYH, Li YM, Xia D, Chen T, Wu A. Role of oxidative stress in mitochondrial dysfunction and their implications in intervertebral disc degeneration: Mechanisms and therapeutic strategies. J Orthop Translat 2024; 49:181-206. [PMID: 39483126 PMCID: PMC11526088 DOI: 10.1016/j.jot.2024.08.016] [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: 06/21/2024] [Revised: 08/03/2024] [Accepted: 08/22/2024] [Indexed: 11/03/2024] Open
Abstract
Background Intervertebral disc degeneration (IVDD) is widely recognized as one of the leading causes of low back pain. Intervertebral disc cells are the main components of the intervertebral disc (IVD), and their functions include synthesizing and secreting collagen and proteoglycans to maintain the structural and functional stability of the IVD. In addition, IVD cells are involved in several physiological processes. They help maintain nutrient metabolism balance in the IVD. They also have antioxidant and anti-inflammatory effects. Because of these roles, IVD cells are crucial in IVDD. When IVD cells are subjected to oxidative stress, mitochondria may become damaged, affecting normal cell function and accelerating degenerative changes. Mitochondria are the energy source of the cell and regulate important intracellular processes. As a key site for redox reactions, excessive oxidative stress and reactive oxygen species can damage mitochondria, leading to inflammation, DNA damage, and apoptosis, thus accelerating disc degeneration. Aim of review Describes the core knowledge of IVDD and oxidative stress. Comprehensively examines the complex relationship and potential mechanistic pathways between oxidative stress, mitochondrial dysfunction and IVDD. Highlights potential therapeutic targets and frontier therapeutic concepts. Draws researchers' attention and discussion on the future research of all three. Key scientific concepts of review Origin, development and consequences of IVDD, molecular mechanisms of oxidative stress acting on mitochondria, mechanisms of oxidative stress damage to IVD cells, therapeutic potential of targeting mitochondria to alleviate oxidative stress in IVDD. The translational potential of this article Targeted therapeutic strategies for oxidative stress and mitochondrial dysfunction are particularly critical in the treatment of IVDD. Using antioxidants and specific mitochondrial therapeutic agents can help reduce symptoms and pain. This approach is expected to significantly improve the quality of life for patients. Individualized therapeutic approaches, on the other hand, are based on an in-depth assessment of the patient's degree of oxidative stress and mitochondrial functional status to develop a targeted treatment plan for more precise and effective IVDD management. Additionally, we suggest preventive measures like customized lifestyle changes and medications. These are based on understanding how IVDD develops. The aim is to slow down the disease and reduce the chances of it coming back. Actively promoting clinical trials and evaluating the safety and efficacy of new therapies helps translate cutting-edge treatment concepts into clinical practice. These measures not only improve patient outcomes and quality of life but also reduce the consumption of healthcare resources and the socio-economic burden, thus having a positive impact on the advancement of the IVDD treatment field.
Collapse
Affiliation(s)
- Hao Zhou
- Department of Orthopaedics, Key Laboratory of Structural Malformations in Children of Zhejiang Province, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, 325000, China
- The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang Province, 315010, China
| | - Chenyu Wu
- The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang Province, 315010, China
| | - Yuxin Jin
- Department of Orthopaedics, Key Laboratory of Structural Malformations in Children of Zhejiang Province, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, 325000, China
| | - Ouqiang Wu
- Department of Orthopaedics, Key Laboratory of Structural Malformations in Children of Zhejiang Province, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, 325000, China
| | - Linjie Chen
- Department of Orthopaedics, Key Laboratory of Structural Malformations in Children of Zhejiang Province, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, 325000, China
| | - Zhenyu Guo
- Department of Orthopaedics, Key Laboratory of Structural Malformations in Children of Zhejiang Province, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, 325000, China
| | - Xinzhou Wang
- Department of Orthopaedics, Key Laboratory of Structural Malformations in Children of Zhejiang Province, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, 325000, China
| | - Qizhu Chen
- Department of Clinic of Spine Center, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200082, China
| | - 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, China
| | - Yan Michael Li
- Minimally Invasive Brain and Spine Institute, Upstate Medical University 475 Irving Ave, #402 Syracuse, NY, 13210, USA
| | - Dongdong Xia
- The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang Province, 315010, China
| | - Tao Chen
- Department of Orthopaedics, Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Tongji Hospital, Tongji University School of Medicine, School of Life Science and Technology, Tongji University, Shanghai, 200065, China
| | - Aimin Wu
- Department of Orthopaedics, Key Laboratory of Structural Malformations in Children of Zhejiang Province, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, 325000, China
| |
Collapse
|
29
|
Rivero-Pino F, Marquez-Paradas E, Montserrat-de la Paz S. Food-derived vesicles as immunomodulatory drivers: Current knowledge, gaps, and perspectives. Food Chem 2024; 457:140168. [PMID: 38908244 DOI: 10.1016/j.foodchem.2024.140168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 06/18/2024] [Accepted: 06/19/2024] [Indexed: 06/24/2024]
Abstract
Extracellular vesicles (EVs) are lipid-bound membrane vesicles released from cells, containing active compounds, which can be found in different foods. In this review, the role of food-derived vesicles (FDVs) as immunomodulatory drivers is summarized, with a focus on sources, isolation techniques and yields, as well as bioavailability and potential health implications. In addition, gaps and perspectives detected in this research field have been highlighted. FDVs have been efficiently extracted from different sources, and differential ultracentrifugation seems to be the most adequate isolation technique, with yields ranging from 108 to 1014 EV particles/mL. Animal studies show promising results in how these FDVs might regulate different pathways related to inflammation. Further investigation on the production of stable components in a cost-effective way, as well as human studies demonstrating safety and health-promoting properties, since scarce information has been reported until now, in the context of modulating the immune system are needed.
Collapse
Affiliation(s)
- Fernando Rivero-Pino
- Department of Medical Biochemistry, Molecular Biology and Immunology, School of Medicine, University of Seville, Av. Sanchez Pizjuan s/n, 41009, Seville, Spain; Instituto de Biomedicina de Sevilla, IBiS, Hospital Universitario Virgen del Rocio/CSIC/University of Seville, 41013 Seville, Spain.
| | - Elvira Marquez-Paradas
- Department of Medical Biochemistry, Molecular Biology and Immunology, School of Medicine, University of Seville, Av. Sanchez Pizjuan s/n, 41009, Seville, Spain; Instituto de Biomedicina de Sevilla, IBiS, Hospital Universitario Virgen del Rocio/CSIC/University of Seville, 41013 Seville, Spain.
| | - Sergio Montserrat-de la Paz
- Department of Medical Biochemistry, Molecular Biology and Immunology, School of Medicine, University of Seville, Av. Sanchez Pizjuan s/n, 41009, Seville, Spain; Instituto de Biomedicina de Sevilla, IBiS, Hospital Universitario Virgen del Rocio/CSIC/University of Seville, 41013 Seville, Spain.
| |
Collapse
|
30
|
Iorio R, Petricca S, Di Emidio G, Falone S, Tatone C. Mitochondrial Extracellular Vesicles (mitoEVs): Emerging mediators of cell-to-cell communication in health, aging and age-related diseases. Ageing Res Rev 2024; 101:102522. [PMID: 39369800 DOI: 10.1016/j.arr.2024.102522] [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: 07/03/2024] [Revised: 08/17/2024] [Accepted: 09/23/2024] [Indexed: 10/08/2024]
Abstract
Mitochondria are metabolic and signalling hubs that integrate a plethora of interconnected processes to maintain cell homeostasis. They are also dormant mediators of inflammation and cell death, and with aging damages affecting mitochondria gradually accumulate, resulting in the manifestation of age-associated disorders. In addition to coordinate multiple intracellular functions, mitochondria mediate intercellular and inter-organ cross talk in different physiological and stress conditions. To fulfil this task, mitochondrial signalling has evolved distinct and complex conventional and unconventional routes of horizontal/vertical mitochondrial transfer. In this regard, great interest has been focused on the ability of extracellular vesicles (EVs), such as exosomes and microvesicles, to carry selected mitochondrial cargoes to target cells, in response to internal and external cues. Over the past years, the field of mitochondrial EVs (mitoEVs) has grown exponentially, revealing unexpected heterogeneity of these structures associated with an ever-expanding mitochondrial function, though the full extent of the underlying mechanisms is far from being elucidated. Therefore, emerging subsets of EVs encompass exophers, migrasomes, mitophers, mitovesicles, and mitolysosomes that can act locally or over long-distances to restore mitochondrial homeostasis and cell functionality, or to amplify disease. This review provides a comprehensive overview of our current understanding of the biology and trafficking of MitoEVs in different physiological and pathological conditions. Additionally, a specific focus on the role of mitoEVs in aging and the onset and progression of different age-related diseases is discussed.
Collapse
Affiliation(s)
- Roberto Iorio
- Dept. of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Via Vetoio, L'Aquila 67100, Italy.
| | - Sabrina Petricca
- Dept. of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Via Vetoio, L'Aquila 67100, Italy
| | - Giovanna Di Emidio
- Dept. of Life, Health and Environmental Sciences, University of L'Aquila, Via Vetoio, L'Aquila 67100, Italy
| | - Stefano Falone
- Dept. of Life, Health and Environmental Sciences, University of L'Aquila, Via Vetoio, L'Aquila 67100, Italy
| | - Carla Tatone
- Dept. of Life, Health and Environmental Sciences, University of L'Aquila, Via Vetoio, L'Aquila 67100, Italy
| |
Collapse
|
31
|
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
|
32
|
Luo H, Lai Y, Tang W, Wang G, Shen J, Liu H. Mitochondrial transplantation: a promising strategy for treating degenerative joint diseases. J Transl Med 2024; 22:941. [PMID: 39407249 PMCID: PMC11475785 DOI: 10.1186/s12967-024-05752-0] [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: 08/19/2024] [Accepted: 10/08/2024] [Indexed: 10/20/2024] Open
Abstract
The prevalence of age-related degenerative joint diseases, particularly intervertebral disc degeneration and osteoarthritis, is increasing, thereby posing significant challenges for the elderly population. Mitochondrial dysfunction is a critical factor in the etiology and progression of these disorders. Therapeutic interventions that incorporate mitochondrial transplantation exhibit considerable promise by increasing mitochondrial numbers and improving their functionality. Existing evidence suggests that exogenous mitochondrial therapy improves clinical outcomes for patients with degenerative joint diseases. This review elucidates the mitochondrial abnormalities associated with degenerative joint diseases and examines the mechanisms of mitochondrial intercellular transfer and artificial mitochondrial transplantation. Furthermore, therapeutic strategies for mitochondrial transplantation in degenerative joint diseases are synthesized, and the concept of engineered mitochondrial transplantation is proposed.
Collapse
Affiliation(s)
- Hong Luo
- Department of Orthopedics, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Yue Lai
- Department of Orthopedics, Affiliated Hospital of Guangdong medical University, zhanjiang, 524000, China
| | - Weili Tang
- Department of Orthopedics, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Guoyou Wang
- Department of Orthopedics, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China.
| | - Jianlin Shen
- Central Laboratory, Affiliated Hospital of Putian University, Putian, 351100, Fujian, China.
- Department of Orthopedics, Affiliated Hospital of Putian University, Putian, 351100, Fujian, China.
| | - Huan Liu
- Department of Orthopedics, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China.
| |
Collapse
|
33
|
Peng S, Liu X, Chang L, Liu B, Zhang M, Mao Y, Shen X. Exosomes Derived from Rejuvenated Stem Cells Inactivate NLRP3 Inflammasome and Pyroptosis of Nucleus Pulposus Cells via the Transfer of Antioxidants. Tissue Eng Regen Med 2024; 21:1061-1077. [PMID: 39060654 PMCID: PMC11416441 DOI: 10.1007/s13770-024-00663-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Revised: 06/15/2024] [Accepted: 07/05/2024] [Indexed: 07/28/2024] Open
Abstract
BACKGROUND Accumulating evidence supports the potential of exosomes as a promising therapeutic approach for intervertebral disc degeneration (IDD). Nevertheless, enhancing the efficiency of exosome treatment remains an urgent concern. This study investigated the impact of quercetin on the characteristics of mesenchymal stem cells (MSCs) and their released exosomes. METHODS Exosomes were obtained from quercetin pre-treated MSCs and quantified for the production based on nanoparticle tracking and western blot analysis. The molecules involved in the secretion and cargo sorting of exosomes were investigated using western blot and immunofluorescence analysis. Based on the in vitro biological analysis and in vivo histological analysis, the effects of exosomes derived from conventional or quercetin-treated MSCs on nucleus pulposus (NP) cells were compared. RESULTS A significant enhancement in the production and transportation efficiency of exosomes was observed in quercetin-treated MSCs. Moreover, the exosomes derived from quercetin-treated MSCs exhibited a greater abundance of antioxidant proteins, specifically superoxide dismutase 1 (SOD1), which inhibit the activation of NOD-like receptor thermal protein domain associated protein 3 (NLRP3) inflammasome in NP cells. Through in vitro and in vivo experiments, it was elucidated that exosomes derived from quercetin-treated MSCs possessed enhanced anti-inflammatory and antioxidant properties. CONCLUSION Collectively, our research underscores an optimized therapeutic strategy for IDD utilizing MSC-derived exosomes, thereby augmenting the efficacy of exosomes in intervertebral disc regeneration.
Collapse
Affiliation(s)
- Shuai Peng
- Department of Spine Surgery, Hunan Provincial People's Hospital, 61 Jiefang West Road, Furong District, Changsha, 410005, Hunan, China
| | - Xiangyang Liu
- Department of Spine Surgery, Hunan Provincial People's Hospital, 61 Jiefang West Road, Furong District, Changsha, 410005, Hunan, China
| | - Lei Chang
- Department of Spine Surgery, Hunan Provincial People's Hospital, 61 Jiefang West Road, Furong District, Changsha, 410005, Hunan, China
| | - Bin Liu
- Department of Spine Surgery, Hunan Provincial People's Hospital, 61 Jiefang West Road, Furong District, Changsha, 410005, Hunan, China
| | - Mingyan Zhang
- Department of Spine Surgery, Hunan Provincial People's Hospital, 61 Jiefang West Road, Furong District, Changsha, 410005, Hunan, China
| | - Yan Mao
- Department of Ophthalmology, Hunan Provincial People's Hospital, Changsha, 410005, Hunan, China
| | - Xiongjie Shen
- Department of Spine Surgery, Hunan Provincial People's Hospital, 61 Jiefang West Road, Furong District, Changsha, 410005, Hunan, China.
| |
Collapse
|
34
|
Hou Q, Ouyang S, Xie Z, He Y, Deng Y, Guo J, Yu P, Tan X, Ma W, Li P, Yu J, Mo Q, Zhang Z, Chen D, Lin X, Liu Z, Chen X, Peng T, Li L, Xie W. Exosome is a Fancy Mobile Sower of Ferroptosis. J Cardiovasc Transl Res 2024; 17:1067-1082. [PMID: 38776048 DOI: 10.1007/s12265-024-10508-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 04/01/2024] [Indexed: 10/29/2024]
Abstract
Exosomes, nano-sized small extracellular vesicles, have been shown to serve as mediators between intercellular communications by transferring bioactive molecules, such as non-coding RNA, proteins, and lipids from secretory to recipient cells, modulating a variety of physiological and pathophysiological processes. Recent studies have gradually demonstrated that altered exosome charges may represent a key mechanism driving the pathological process of ferroptosis. This review summarizes the potential mechanisms and signal pathways relevant to ferroptosis and then discusses the roles of exosome in ferroptosis. As well as transporting iron, exosomes may also indirectly convey factors related to ferroptosis. Furthermore, ferroptosis may be transmitted to adjacent cells through exosomes, resulting in cascading effects. It is expected that further research on exosomes will be conducted to explore their potential in ferroptosis and will lead to the creation of new therapeutic avenues for clinical diseases.
Collapse
Affiliation(s)
- Qin Hou
- Institute of Cardiovascular Research, Key Laboratory for Atherosclerology of Hunan Province, Medical Research Center, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Siyu Ouyang
- Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Zhongcheng Xie
- Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Yinling He
- Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Yunong Deng
- Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Jiamin Guo
- Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Panpan Yu
- Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Xiaoqian Tan
- Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Wentao Ma
- Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Pin Li
- Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Jiang Yu
- Class of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Qinger Mo
- Class of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Zhixia Zhang
- Class of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Dandan Chen
- Class of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Xiaoyan Lin
- Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Zhiyang Liu
- Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Xi Chen
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Tianhong Peng
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.
| | - Liang Li
- Department of Physiology, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.
| | - Wei Xie
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.
| |
Collapse
|
35
|
Ramzan F, Salim A, Hussain A, Khan I. Unleashing the Healing Power of Mesenchymal Stem Cells for Osteochondral Abnormalities. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2024. [DOI: 10.1007/s40883-024-00356-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 07/25/2024] [Accepted: 08/31/2024] [Indexed: 01/11/2025]
|
36
|
Zhao Y, Xia Q, Zhu L, Xia J, Xiang S, Mao Q, Dong H, Weng Z, Liao W, Xin Z. Mapping knowledge structure and themes trends of non-surgical treatment in intervertebral disc degeneration. Heliyon 2024; 10:e36509. [PMID: 39286189 PMCID: PMC11402762 DOI: 10.1016/j.heliyon.2024.e36509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 03/23/2024] [Accepted: 08/16/2024] [Indexed: 09/19/2024] Open
Abstract
Background Intervertebral disc degeneration (IDD) is a chronic disabling disease caused by degeneration of nucleus pulposus cells, decreased activity and the number of nucleus pulposus cells, decreased extracellular matrix, and infiltration of inflammatory factors, resulting in low back and leg pain. Recent studies have shown that non-surgical treatment is of great significance in reversing the progression of degenerative disc disease, and there are more relevant literature reports. However, there is no bibliometric analysis in this area. This study aimed to describe the knowledge structure and thematic trends of non-surgical treatment methods for IDD through bibliometrics. Methods Articles and reviews on non-surgical treatment of disc degeneration from 1998 to 2022 were collected on the Web of Science. VOSviewer 1.6.18, CiteSpace 6.1.R3, R package "bibliometrix" and two online analysis platforms were used for bibliometric and visual literature analysis. Results 961 articles were screened for inclusion, including 821 articles and 140 reviews. The analysis of our study shows that publications in the non-surgical treatment of disc degeneration are increasing annually, with publications coming mainly from North America and Asia, with China and the United States dominating. Huazhong Univ Sci & Technol and Wang K are the most prolific institutions and authors, respectively, and Le Maitre CL is the most co-cited author. However, there is less collaboration between institutions in different countries. Spine is both the most published and the most cited journal. According to the co-citation and co-occurrence analysis results, "mesenchymal stem cells," "exosomes," "medication," and "tissue engineering" are the current research hotspots in this field. Conclusions This study employs bibliometric analysis to explore the knowledge structure and trends of non-surgical treatments for IDD from 2013 to 2022. Key research hotspots include mesenchymal stem cells, exosomes, medication, and tissue engineering. The number of publications, especially from China and the USA, has increased significantly, though international collaboration needs improvement. Influential contributors include Wang K and the journal Spine. These findings provide a comprehensive overview and highlight important future directions for the field.
Collapse
Affiliation(s)
- Yan Zhao
- Department of Orthopedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, Guizhou, China
| | - Qiuqiu Xia
- Department of Orthopedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, Guizhou, China
| | - Lu Zhu
- Department of Orthopedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, Guizhou, China
| | - Jiyue Xia
- Department of Orthopedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, Guizhou, China
| | - Shaojie Xiang
- Department of Orthopedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, Guizhou, China
| | - Qiming Mao
- Department of Orthopedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, Guizhou, China
| | - Huaize Dong
- Department of Orthopedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, Guizhou, China
| | - Zijing Weng
- Department of Orthopedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, Guizhou, China
| | - Wenbo Liao
- Department of Orthopedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, Guizhou, China
| | - Zhijun Xin
- Department of Orthopedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, Guizhou, China
- Institut Curie, PSL Research University, CNRS UMR3244, Dynamics of Genetic Information, Sorbonne Université, 75005, Paris, France
| |
Collapse
|
37
|
Jia B, Xue R, Li J, Xu G, Li X, Wang W, Li Z, Liu J. Molecular mechanisms of EGCG-CSH/n-HA/CMC in promoting osteogenic differentiation and macrophage polarization. Bioorg Chem 2024; 150:107493. [PMID: 38870703 DOI: 10.1016/j.bioorg.2024.107493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 05/22/2024] [Indexed: 06/15/2024]
Abstract
2. This research investigates the impact of the EGCG-CSH/n-HA/CMC composite material on bone defect repair, emphasizing its influence on macrophage polarization and osteogenic differentiation of BMSCs. Comprehensive evaluations of the composite's physical and chemical characteristics were performed. BMSC response to the material was tested in vitro for proliferation, migration, and osteogenic potential. An SD rat model was employed for in vivo assessments of bone repair efficacy. Both transcriptional and proteomic analyses were utilized to delineate the mechanisms influencing macrophage behavior and stem cell differentiation. The material maintained excellent structural integrity and significantly promoted BMSC functions critical to bone healing. In vivo results confirmed accelerated bone repair, and molecular analysis highlighted the role of macrophage M2 polarization, particularly through changes in the SIRPA gene and protein expression. EGCG-CSH/n-HA/CMC plays a significant role in enhancing bone repair, with implications for macrophage and BMSC function. Our findings suggest that targeting SIRPA may offer new therapeutic opportunities for bone regeneration.
Collapse
Affiliation(s)
- Bei Jia
- Nosocomial Infection Management Department, Hebei Medical University First Hospital, Shijiazhuang 050000, China
| | - Rui Xue
- Department of Orthopaedic Surgery, Hebei Medical University Third Hospital, Shijiazhuang 050051, China
| | - Jia Li
- Department of Orthopaedic Surgery, Hebei Medical University Third Hospital, Shijiazhuang 050051, China
| | - Guohui Xu
- Department of Orthopaedic Surgery, Hebei Medical University Third Hospital, Shijiazhuang 050051, China
| | - Xu Li
- Department of Orthopaedic Surgery, Hebei Medical University Third Hospital, Shijiazhuang 050051, China
| | - Wei Wang
- Department of Orthopaedic Surgery, Hebei Medical University Third Hospital, Shijiazhuang 050051, China
| | - Zhiyong Li
- Department of Orthopaedic Surgery, Hebei Medical University Third Hospital, Shijiazhuang 050051, China
| | - Jianning Liu
- Department of Orthopaedic Surgery, Hebei Medical University Third Hospital, Shijiazhuang 050051, China.
| |
Collapse
|
38
|
Wu Q, Du J, Bae EJ, Choi Y. Pyroptosis in Skeleton Diseases: A Potential Therapeutic Target Based on Inflammatory Cell Death. Int J Mol Sci 2024; 25:9068. [PMID: 39201755 PMCID: PMC11354934 DOI: 10.3390/ijms25169068] [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: 06/25/2024] [Revised: 08/15/2024] [Accepted: 08/20/2024] [Indexed: 09/03/2024] Open
Abstract
Skeletal disorders, including fractures, osteoporosis, osteoarthritis, rheumatoid arthritis, and spinal degenerative conditions, along with associated spinal cord injuries, significantly impair daily life and impose a substantial burden. Many of these conditions are notably linked to inflammation, with some classified as inflammatory diseases. Pyroptosis, a newly recognized form of inflammatory cell death, is primarily triggered by inflammasomes and executed by caspases, leading to inflammation and cell death through gasdermin proteins. Emerging research underscores the pivotal role of pyroptosis in skeletal disorders. This review explores the pyroptosis signaling pathways and their involvement in skeletal diseases, the modulation of pyroptosis by other signals in these conditions, and the current evidence supporting the therapeutic potential of targeting pyroptosis in treating skeletal disorders, aiming to offer novel insights for their management.
Collapse
Affiliation(s)
- Qian Wu
- Department of Biochemistry and Molecular Biology, Jeonbuk National University Medical School, Jeonju 54896, Republic of Korea (J.D.)
| | - Jiacheng Du
- Department of Biochemistry and Molecular Biology, Jeonbuk National University Medical School, Jeonju 54896, Republic of Korea (J.D.)
| | - Eun Ju Bae
- School of Pharmacy, Jeonbuk National University, Jeonju 54896, Republic of Korea
| | - Yunjung Choi
- Division of Rheumatology, Department of Internal Medicine, Jeonbuk National University Medical School, Jeonju 54896, Republic of Korea
- Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju 54907, Republic of Korea
| |
Collapse
|
39
|
Yu XJ, Bai XF, Qu YK, Wang SX, Zhang J, Yang W, Wang S, Yang Y, Wang YG, Hao DJ, Zhao YT. Unveiling the Therapeutic Potential of hUCMSC-Derived EVs in Intervertebral Disc Degeneration through MALAT1/ miR-138-5p/SLC7A11 Coexpression Regulation. ACS Biomater Sci Eng 2024; 10:4839-4854. [PMID: 39079050 DOI: 10.1021/acsbiomaterials.3c01944] [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: 08/13/2024]
Abstract
Intervertebral disc degeneration (IVDD) is a prevalent chronic condition causing spinal pain and functional impairment. This study investigates the role of extracellular vesicles (EVs) derived from human umbilical cord mesenchymal stem cells (hUCMSCs) in regulating IVDD. Using RNA-seq, we analyzed differential expressions of lncRNA and miRNA in nucleus pulposus tissues from various mouse groups. We identified key regulatory molecules, MALAT1 and miRNA-138-5p, which contribute to IVDD. Further experiments demonstrated that MALAT1 can up-regulate SLC7A11 expression by competitively binding to miR-138-5p, forming a MALAT1/miR-138-5p/SLC7A11 coexpression regulatory network. This study elucidates the molecular mechanism by which hUCMSC-derived EVs regulate IVDD and could help develop novel therapeutic strategies for treating this condition. Our findings demonstrate that hUCMSCs-EVs inhibit ferroptosis in nucleus pulposus cells, thereby improving IVDD. These results highlight the therapeutic potential of hUCMSCs-EVs in ameliorating the development of IVDD, offering significant scientific and clinical implications for new treatments.
Collapse
Affiliation(s)
- Xiao-Jun Yu
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an 710054, Shaanxi, China
- Shaanxi Key Laboratory of Spine Bionic Treatment, Xi'an 710054, Shaanxi, China
| | - Xiao-Fan Bai
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an 710054, Shaanxi, China
- Shaanxi Key Laboratory of Spine Bionic Treatment, Xi'an 710054, Shaanxi, China
| | - Yun-Kun Qu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Shan-Xi Wang
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an 710054, Shaanxi, China
- Shaanxi Key Laboratory of Spine Bionic Treatment, Xi'an 710054, Shaanxi, China
| | - Jianwei Zhang
- Department of Orthopedics, the First People's Hospital of Tianshui City, Tianshui 741000, Gansu Province, China
| | - Wenlong Yang
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an 710054, Shaanxi, China
- Shaanxi Key Laboratory of Spine Bionic Treatment, Xi'an 710054, Shaanxi, China
| | - Sibo Wang
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an 710054, Shaanxi, China
- Shaanxi Key Laboratory of Spine Bionic Treatment, Xi'an 710054, Shaanxi, China
| | - Yuli Yang
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an 710054, Shaanxi, China
| | - Ying-Guang Wang
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an 710054, Shaanxi, China
- Shaanxi Key Laboratory of Spine Bionic Treatment, Xi'an 710054, Shaanxi, China
| | - Ding-Jun Hao
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an 710054, Shaanxi, China
- Shaanxi Key Laboratory of Spine Bionic Treatment, Xi'an 710054, Shaanxi, China
| | - Yuan-Ting Zhao
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an 710054, Shaanxi, China
- Shaanxi Key Laboratory of Spine Bionic Treatment, Xi'an 710054, Shaanxi, China
| |
Collapse
|
40
|
Wu S, Yang T, Ma M, Fan L, Ren L, Liu G, Wang Y, Cheng B, Xia J, Hao Z. Extracellular vesicles meet mitochondria: Potential roles in regenerative medicine. Pharmacol Res 2024; 206:107307. [PMID: 39004243 DOI: 10.1016/j.phrs.2024.107307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 07/09/2024] [Accepted: 07/11/2024] [Indexed: 07/16/2024]
Abstract
Extracellular vesicles (EVs), secreted by most cells, act as natural cell-derived carriers for delivering proteins, nucleic acids, and organelles between cells. Mitochondria are highly dynamic organelles responsible for energy production and cellular physiological processes. Recent evidence has highlighted the pivotal role of EVs in intercellular mitochondrial content transfer, including mitochondrial DNA (mtDNA), proteins, and intact mitochondria. Intriguingly, mitochondria are crucial mediators of EVs release, suggesting an interplay between EVs and mitochondria and their potential implications in physiology and pathology. However, in this expanding field, much remains unknown regarding the function and mechanism of crosstalk between EVs and mitochondria and the transport of mitochondrial EVs. Herein, we shed light on the physiological and pathological functions of EVs and mitochondria, potential mechanisms underlying their interactions, delivery of mitochondria-rich EVs, and their clinical applications in regenerative medicine.
Collapse
Affiliation(s)
- Shujie Wu
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
| | - Tao Yang
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
| | - Meirui Ma
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
| | - Le Fan
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
| | - Lin Ren
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
| | - Gen Liu
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
| | - Yiqiao Wang
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
| | - Bin Cheng
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China.
| | - Juan Xia
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China.
| | - Zhichao Hao
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China.
| |
Collapse
|
41
|
Abdal Dayem A, Yan E, Do M, Kim Y, Lee Y, Cho SG, Kim DH. Engineering extracellular vesicles for ROS scavenging and tissue regeneration. NANO CONVERGENCE 2024; 11:24. [PMID: 38922501 PMCID: PMC11208369 DOI: 10.1186/s40580-024-00430-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 05/22/2024] [Indexed: 06/27/2024]
Abstract
Stem cell therapy holds promise for tissue regeneration, yet significant challenges persist. Emerging as a safer and potentially more effective alternative, extracellular vesicles (EVs) derived from stem cells exhibit remarkable abilities to activate critical signaling cascades, thereby facilitating tissue repair. EVs, nano-scale membrane vesicles, mediate intercellular communication by encapsulating a diverse cargo of proteins, lipids, and nucleic acids. Their therapeutic potential lies in delivering cargos, activating signaling pathways, and efficiently mitigating oxidative stress-an essential aspect of overcoming limitations in stem cell-based tissue repair. This review focuses on engineering and applying EVs in tissue regeneration, emphasizing their role in regulating reactive oxygen species (ROS) pathways. Additionally, we explore strategies to enhance EV therapeutic activity, including functionalization and incorporation of antioxidant defense proteins. Understanding these molecular mechanisms is crucial for optimizing EV-based regenerative therapies. Insights into EV and ROS signaling modulation pave the way for targeted and efficient regenerative therapies harnessing the potential of EVs.
Collapse
Affiliation(s)
- Ahmed Abdal Dayem
- Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center, Institute of Advanced Regenerative Science, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05029, Republic of Korea
| | - Ellie Yan
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Minjae Do
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Yoojung Kim
- Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center, Institute of Advanced Regenerative Science, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05029, Republic of Korea
| | - Yeongseo Lee
- Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center, Institute of Advanced Regenerative Science, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05029, Republic of Korea
| | - Ssang-Goo Cho
- Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center, Institute of Advanced Regenerative Science, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05029, Republic of Korea.
- R&D Team, StemExOne Co., Ltd., 307 KU Technology Innovation Bldg, 120, Neungdong-ro, Gwangjin- gu, Seoul, 05029, Republic of Korea.
| | - Deok-Ho Kim
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21205, USA.
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, 21205, USA.
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, 21205, USA.
- Center for Microphysiological Systems, Johns Hopkins University, Baltimore, MD, 21205, USA.
- Institute for NanoBiotechnology, Johns Hopkins University, Baltimore, MD, 21218, USA.
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| |
Collapse
|
42
|
Houerbi N, Kim J, Overbey EG, Batra R, Schweickart A, Patras L, Lucotti S, Ryon KA, Najjar D, Meydan C, Damle N, Chin C, Narayanan SA, Guarnieri JW, Widjaja G, Beheshti A, Tobias G, Vatter F, Hirschberg JW, Kleinman A, Afshin EE, MacKay M, Chen Q, Miller D, Gajadhar AS, Williamson L, Tandel P, Yang Q, Chu J, Benz R, Siddiqui A, Hornburg D, Gross S, Shirah B, Krumsiek J, Mateus J, Mao X, Matei I, Mason CE. Secretome profiling reveals acute changes in oxidative stress, brain homeostasis, and coagulation following short-duration spaceflight. Nat Commun 2024; 15:4862. [PMID: 38862464 PMCID: PMC11166969 DOI: 10.1038/s41467-024-48841-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 05/15/2024] [Indexed: 06/13/2024] Open
Abstract
As spaceflight becomes more common with commercial crews, blood-based measures of crew health can guide both astronaut biomedicine and countermeasures. By profiling plasma proteins, metabolites, and extracellular vesicles/particles (EVPs) from the SpaceX Inspiration4 crew, we generated "spaceflight secretome profiles," which showed significant differences in coagulation, oxidative stress, and brain-enriched proteins. While >93% of differentially abundant proteins (DAPs) in vesicles and metabolites recovered within six months, the majority (73%) of plasma DAPs were still perturbed post-flight. Moreover, these proteomic alterations correlated better with peripheral blood mononuclear cells than whole blood, suggesting that immune cells contribute more DAPs than erythrocytes. Finally, to discern possible mechanisms leading to brain-enriched protein detection and blood-brain barrier (BBB) disruption, we examined protein changes in dissected brains of spaceflight mice, which showed increases in PECAM-1, a marker of BBB integrity. These data highlight how even short-duration spaceflight can disrupt human and murine physiology and identify spaceflight biomarkers that can guide countermeasure development.
Collapse
Affiliation(s)
- Nadia Houerbi
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
| | - JangKeun Kim
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
| | - Eliah G Overbey
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
| | - Richa Batra
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
| | - Annalise Schweickart
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
- Tri-Institutional Biology and Medicine program, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Laura Patras
- Children's Cancer and Blood Foundation Laboratories, Departments of Pediatrics and Cell and Developmental Biology, Drukier Institute for Children's Health, Weill Cornell Medicine, New York, NY, USA
- Department of Molecular Biology and Biotechnology, Center of Systems Biology, Biodiversity and Bioresources, Faculty of Biology and Geology, Babes-Bolyai University, Cluj-Napoca, Romania
| | - Serena Lucotti
- Children's Cancer and Blood Foundation Laboratories, Departments of Pediatrics and Cell and Developmental Biology, Drukier Institute for Children's Health, Weill Cornell Medicine, New York, NY, USA
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
| | - Krista A Ryon
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
| | - Deena Najjar
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
| | - Cem Meydan
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
| | - Namita Damle
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
| | - Christopher Chin
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
| | - S Anand Narayanan
- Department of Nutrition & Integrative Physiology, Florida State University, Tallahassee, FL, USA
| | - Joseph W Guarnieri
- Center of Mitochondrial and Epigenomic Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Gabrielle Widjaja
- Center of Mitochondrial and Epigenomic Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Afshin Beheshti
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- KBR, Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, USA
| | - Gabriel Tobias
- Children's Cancer and Blood Foundation Laboratories, Departments of Pediatrics and Cell and Developmental Biology, Drukier Institute for Children's Health, Weill Cornell Medicine, New York, NY, USA
- Seer, Inc., Redwood City, CA, 94065, USA
| | - Fanny Vatter
- Children's Cancer and Blood Foundation Laboratories, Departments of Pediatrics and Cell and Developmental Biology, Drukier Institute for Children's Health, Weill Cornell Medicine, New York, NY, USA
- Seer, Inc., Redwood City, CA, 94065, USA
| | | | - Ashley Kleinman
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
| | - Evan E Afshin
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
| | - Matthew MacKay
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
| | - Qiuying Chen
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
| | - Dawson Miller
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
| | | | | | | | - Qiu Yang
- Seer, Inc., Redwood City, CA, 94065, USA
| | | | - Ryan Benz
- Seer, Inc., Redwood City, CA, 94065, USA
| | | | | | - Steven Gross
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
| | - Bader Shirah
- Department of Neuroscience, King Faisal Specialist Hospital & Research Centre, Jeddah, Saudi Arabia
| | - Jan Krumsiek
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
- Tri-Institutional Biology and Medicine program, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Jaime Mateus
- Space Exploration Technologies Corporation (SpaceX), Hawthorne, CA, USA
| | - Xiao Mao
- Department of Basic Sciences, Division of Biomedical Engineering Sciences (BMES), Loma Linda University Health, Loma Linda, CA, 92350, USA
| | - Irina Matei
- Children's Cancer and Blood Foundation Laboratories, Departments of Pediatrics and Cell and Developmental Biology, Drukier Institute for Children's Health, Weill Cornell Medicine, New York, NY, USA.
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY, 10065, USA.
| | - Christopher E Mason
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA.
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA.
- Tri-Institutional Biology and Medicine program, Weill Cornell Medicine, New York, NY, 10021, USA.
- The Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, 10021, USA.
- WorldQuant Initiative for Quantitative Prediction, Weill Cornell Medicine, New York, NY, 10021, USA.
| |
Collapse
|
43
|
Yang X, Zhang S, Lu J, Chen X, Zheng T, He R, Ye C, Xu J. Therapeutic potential of mesenchymal stem cell-derived exosomes in skeletal diseases. Front Mol Biosci 2024; 11:1268019. [PMID: 38903180 PMCID: PMC11187108 DOI: 10.3389/fmolb.2024.1268019] [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: 07/27/2023] [Accepted: 05/16/2024] [Indexed: 06/22/2024] Open
Abstract
Skeletal diseases impose a considerable burden on society. The clinical and tissue-engineering therapies applied to alleviate such diseases frequently result in complications and are inadequately effective. Research has shifted from conventional therapies based on mesenchymal stem cells (MSCs) to exosomes derived from MSCs. Exosomes are natural nanocarriers of endogenous DNA, RNA, proteins, and lipids and have a low immune clearance rate and good barrier penetration and allow targeted delivery of therapeutics. MSC-derived exosomes (MSC-exosomes) have the characteristics of both MSCs and exosomes, and so they can have both immunosuppressive and tissue-regenerative effects. Despite advances in our knowledge of MSC-exosomes, their regulatory mechanisms and functionalities are unclear. Here we review the therapeutic potential of MSC-exosomes for skeletal diseases.
Collapse
Affiliation(s)
- Xiaobo Yang
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, China
| | - Shaodian Zhang
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, China
| | - Jinwei Lu
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, China
| | - Xiaoling Chen
- Department of Plastic Surgery, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang, China
| | - Tian Zheng
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, China
| | - Rongxin He
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, China
| | - Chenyi Ye
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, China
| | - Jianbin Xu
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, China
| |
Collapse
|
44
|
Wang D, Zhang L, He D, Zhang Y, Zhao L, Miao Z, Cheng W, Zhu C, Shao Y, Ge G, Zhu H, Jin H, Zhang W, Pan H. A natural hydrogel complex improves intervertebral disc degeneration by correcting fatty acid metabolism and inhibiting nucleus pulposus cell pyroptosis. Mater Today Bio 2024; 26:101081. [PMID: 38741924 PMCID: PMC11089368 DOI: 10.1016/j.mtbio.2024.101081] [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: 02/14/2024] [Revised: 03/26/2024] [Accepted: 05/02/2024] [Indexed: 05/16/2024] Open
Abstract
The degeneration of intervertebral discs is strongly associated with the occurrence of pyroptosis in nucleus pulposus (NP) cells. This pyroptosis is characterized by abnormal metabolism of fatty acids in the degenerative pathological state, which is further exacerbated by the inflammatory microenvironment and degradation of the extracellular matrix. In order to address this issue, we have developed a fibrin hydrogel complex (FG@PEV). This intricate formulation amalgamates the beneficial attributes of platelet extravasation vesicles, contributing to tissue repair and regeneration. Furthermore, this complex showcases exceptional stability, gradual-release capabilities, and a high degree of biocompatibility. In order to substantiate the biological significance of FG@PEV in intervertebral disc degeneration (IVDD), we conducted a comprehensive investigation into its potential mechanism of action through the integration of RNA-seq sequencing and metabolomics analysis. Furthermore, these findings were subsequently validated through experimentation in both in vivo and in vitro models. The experimental results revealed that the FG@PEV intervention possesses the capability to reshape the inflammatory microenvironment within the disc. It also addresses the irregularities in fatty acid metabolism of nucleus pulposus cells, consequently hindering cellular pyroptosis and slowing down disc degeneration through the regulation of extracellular matrix synthesis and degradation. As a result, this injectable gel system represents a promising and innovative therapeutic approach for mitigating disc degeneration.
Collapse
Affiliation(s)
- Dong Wang
- Department of Orthopaedics, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University (Hangzhou Hospital of Traditional Chinese Medicine), Hangzhou, 310000, Zhejiang Province, PR China
- Department of Orthopaedics, Hangzhou Dingqiao Hospital, Huanding Road NO 1630, Hangzhou, 310021, Zhejiang Province, PR China
- Institute of Orthopaedics and Traumatology, Hangzhou Traditional Chinese Medicine Hospital Affiliated to Zhejiang Chinese Medical University, Tiyuchang Road NO 453, Hangzhou, 310007, Zhejiang Province, PR China
| | - Liangping Zhang
- Department of Orthopaedics, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University (Hangzhou Hospital of Traditional Chinese Medicine), Hangzhou, 310000, Zhejiang Province, PR China
| | - Du He
- Department of Orthopaedics, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University (Hangzhou Hospital of Traditional Chinese Medicine), Hangzhou, 310000, Zhejiang Province, PR China
| | - Yujun Zhang
- Department of Orthopaedics, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University (Hangzhou Hospital of Traditional Chinese Medicine), Hangzhou, 310000, Zhejiang Province, PR China
| | - Lan Zhao
- Department of Orthopaedics, Hangzhou Dingqiao Hospital, Huanding Road NO 1630, Hangzhou, 310021, Zhejiang Province, PR China
| | - Zhimin Miao
- Department of Orthopaedics, Hangzhou Dingqiao Hospital, Huanding Road NO 1630, Hangzhou, 310021, Zhejiang Province, PR China
| | - Wei Cheng
- Department of Orthopaedics, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University (Hangzhou Hospital of Traditional Chinese Medicine), Hangzhou, 310000, Zhejiang Province, PR China
- Department of Orthopaedics, Hangzhou Dingqiao Hospital, Huanding Road NO 1630, Hangzhou, 310021, Zhejiang Province, PR China
| | - Chengyue Zhu
- Department of Orthopaedics, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University (Hangzhou Hospital of Traditional Chinese Medicine), Hangzhou, 310000, Zhejiang Province, PR China
- Department of Orthopaedics, Hangzhou Dingqiao Hospital, Huanding Road NO 1630, Hangzhou, 310021, Zhejiang Province, PR China
- Institute of Orthopaedics and Traumatology, Hangzhou Traditional Chinese Medicine Hospital Affiliated to Zhejiang Chinese Medical University, Tiyuchang Road NO 453, Hangzhou, 310007, Zhejiang Province, PR China
| | - Yinyan Shao
- Department of Orthopaedics, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University (Hangzhou Hospital of Traditional Chinese Medicine), Hangzhou, 310000, Zhejiang Province, PR China
| | - Guofen Ge
- Department of Orthopaedics, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University (Hangzhou Hospital of Traditional Chinese Medicine), Hangzhou, 310000, Zhejiang Province, PR China
| | - Hang Zhu
- Department of Orthopaedics, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University (Hangzhou Hospital of Traditional Chinese Medicine), Hangzhou, 310000, Zhejiang Province, PR China
| | - HongTing Jin
- Department of Orthopaedics, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University (Hangzhou Hospital of Traditional Chinese Medicine), Hangzhou, 310000, Zhejiang Province, PR China
- Institute of Orthopaedics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, PR China
| | - Wei Zhang
- Department of Orthopaedics, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University (Hangzhou Hospital of Traditional Chinese Medicine), Hangzhou, 310000, Zhejiang Province, PR China
- Department of Orthopaedics, Hangzhou Dingqiao Hospital, Huanding Road NO 1630, Hangzhou, 310021, Zhejiang Province, PR China
- Institute of Orthopaedics and Traumatology, Hangzhou Traditional Chinese Medicine Hospital Affiliated to Zhejiang Chinese Medical University, Tiyuchang Road NO 453, Hangzhou, 310007, Zhejiang Province, PR China
| | - Hao Pan
- Department of Orthopaedics, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University (Hangzhou Hospital of Traditional Chinese Medicine), Hangzhou, 310000, Zhejiang Province, PR China
- Department of Orthopaedics, Hangzhou Dingqiao Hospital, Huanding Road NO 1630, Hangzhou, 310021, Zhejiang Province, PR China
- Institute of Orthopaedics and Traumatology, Hangzhou Traditional Chinese Medicine Hospital Affiliated to Zhejiang Chinese Medical University, Tiyuchang Road NO 453, Hangzhou, 310007, Zhejiang Province, PR China
| |
Collapse
|
45
|
Esfahani SK, Dehghani S, Hosseinzadeh H, Abnous K, Taghdisi SM, Ramezani M, Alibolandi M. An exosomal approach for oral delivery of resveratrol: Implications for inflammatory bowel disease treatment in rat model. Life Sci 2024; 346:122638. [PMID: 38614294 DOI: 10.1016/j.lfs.2024.122638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 03/24/2024] [Accepted: 04/10/2024] [Indexed: 04/15/2024]
Abstract
AIMS Resveratrol (RSV) is a polyphenolic substance found in numerous natural products. Despite the wide range of therapeutic activities, including antioxidant and anti-inflammatory effects, the poor pharmacokinetic characteristics decrease the RSV bioavailability following oral administration. Milk-derived exosomes (MEXOs), as a class of natural nanocarriers, are promising candidates for oral drug delivery approaches. MAIN METHODS The current study developed RSV-loaded MEXOs to enhance the RSV oral bioavailability, introducing a suitable exosomal formulation for suppressing colon inflammation in acetic acid-induced rat models. KEY FINDINGS The results showed a remarkable encapsulation efficiency of 83.33 %. The in vitro release profile demonstrated a good retaining capability in acidic conditions (pH 1.2) and a considerable release in a simulated duodenal environment (pH 6.8). According to the permeability study, encapsulation of RSV improved its transportation across the Caco-2 monolayer. Moreover, the in vivo and histological analysis results proved that the RSV-MEXOs formulation successfully alleviates the inflammation in colitis rat models and effectively relieves the colitis. SIGNIFICANCE Our findings suggest that MEXOs should be of great attention as promising oral drug delivery vehicles for further clinical evaluations.
Collapse
Affiliation(s)
- Shaghayegh Kazemi Esfahani
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sadegh Dehghani
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hossein Hosseinzadeh
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Khalil Abnous
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Mohammad Taghdisi
- Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Ramezani
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Mona Alibolandi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
46
|
Théron A, Maumus M, Biron-Andreani C, Sirvent N, Jorgensen C, Noël D. What is the rationale for mesenchymal stromal cells based therapies in the management of hemophilic arthropathies? Osteoarthritis Cartilage 2024; 32:634-642. [PMID: 38160743 DOI: 10.1016/j.joca.2023.12.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 11/21/2023] [Accepted: 12/07/2023] [Indexed: 01/03/2024]
Abstract
Hemophilia A and B are rare X-linked genetic bleeding disorders due to a complete or partial deficiency in the coagulation factors VIII or IX, respectively. The main treatment for hemophilia is prophylactic and based on coagulation factor replacement therapies. These treatments have significantly reduced bleeding and improved the patients' quality of life. Nevertheless, repeated joint bleedings (hemarthroses), even subclinical hemarthroses, can lead to hemophilic arthropathy (HA). This disabling condition is characterized by chronic pain due to synovial inflammation, cartilage and bone destruction requiring ultimately joint replacement. HA resembles to rheumatoid arthritis because of synovitis but HA is considered as having similarities with osteoarthritis as illustrated by the migration of immune cells, production of inflammatory cytokines, synovial hypertrophy and cartilage damage. Various drugs have been evaluated for the management of HA with limited success. The objective of the review is to discuss new therapeutic approaches with a special focus on the studies that have investigated the potential of using mesenchymal stromal cells (MSCs) in the management of HA. A systematic review of the literature has been made. Most of the studies have focused on the interest of MSCs for the delivery of missing factors VIII or IX but in some studies, more insight on the effect of MSC injection on synovial inflammation or cartilage structure were provided and put in perspective for possible clinical applications.
Collapse
Affiliation(s)
- Alexandre Théron
- IRMB, University of Montpellier, INSERM, Montpellier, France; Resources and Competence Center for Hereditary Hemorrhagic Diseases, CHU Montpellier, Montpellier, France; Department of Pediatric Oncology and Hematology, CHU Montpellier, Montpellier, France
| | - Marie Maumus
- IRMB, University of Montpellier, INSERM, Montpellier, France
| | - Christine Biron-Andreani
- Resources and Competence Center for Hereditary Hemorrhagic Diseases, CHU Montpellier, Montpellier, France
| | - Nicolas Sirvent
- Department of Pediatric Oncology and Hematology, CHU Montpellier, Montpellier, France
| | - Christian Jorgensen
- IRMB, University of Montpellier, INSERM, Montpellier, France; Clinical Immunology and Osteoarticular Disease Therapeutic Unit, Department of Rheumatology, CHU Montpellier, Montpellier, France
| | - Danièle Noël
- IRMB, University of Montpellier, INSERM, Montpellier, France; Clinical Immunology and Osteoarticular Disease Therapeutic Unit, Department of Rheumatology, CHU Montpellier, Montpellier, France.
| |
Collapse
|
47
|
Ma S, Xue R, Zhu H, Han Y, Ji X, Zhang C, Wei N, Xu J, Li F. Selenomethionine preconditioned mesenchymal stem cells derived extracellular vesicles exert enhanced therapeutic efficacy in intervertebral disc degeneration. Int Immunopharmacol 2024; 132:112028. [PMID: 38593507 DOI: 10.1016/j.intimp.2024.112028] [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: 02/19/2024] [Revised: 03/28/2024] [Accepted: 04/04/2024] [Indexed: 04/11/2024]
Abstract
Extracellular vesicles (EVs) derived from Mesenchymal Stromal Cells (MSCs) have shown promising therapeutic potential for multiple diseases, including intervertebral disc degeneration (IDD). Nevertheless, the limited production and unstable quality of EVs hindered the clinical application of EVs in IDD. Selenomethionine (Se-Met), the major form of organic selenium present in the cereal diet, showed various beneficial effects, including antioxidant, immunomodulatory and anti-apoptotic effects. In the current study, Se-Met was employed to treat MSCs to investigate whether Se-Met can facilitate the secretion of EVs by MSCs and optimize their therapeutic effects on IDD. On the one hand, Se-Met promoted the production of EVs by enhancing the autophagy activity of MSCs. On the other hand, Se-Met pretreated MSC-derived EVs (Se-EVs) exhibited an enhanced protective effects on alleviating nucleus pulposus cells (NPCs) senescence and attenuating IDD compared with EVs isolated from control MSCs (C-EVs) in vitro and in vivo. Moreover, we performed a miRNA microarray sequencing analysis on EVs to explore the potential mechanism of the protective effects of EVs. The result indicated that miR-125a-5p is markedly enriched in Se-EVs compared to C-EVs. Further in vitro and in vivo experiments revealed that knockdown of miR-125a-5p in Se-EVs (miRKD-Se-EVs) impeded the protective effects of Se-EVs, while overexpression of miR-125a-5p (miROE-Se-EVs) boosted the protective effects. In conclusion, Se-Met facilitated the MSC-derived EVs production and increased miR-125a-5p delivery in Se-EVs, thereby improving the protective effects of MSC-derived EVs on alleviating NPCs senescence and attenuating IDD.
Collapse
Affiliation(s)
- Shengli Ma
- Department of Emergency, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China.
| | - Rui Xue
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China.
| | - Haiyang Zhu
- Department of Emergency, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China.
| | - Yu Han
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China.
| | - Xiang Ji
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China.
| | - Chaoyang Zhang
- Department of Emergency, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China.
| | - Na Wei
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China.
| | - Jingjing Xu
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China.
| | - Feng Li
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China.
| |
Collapse
|
48
|
Li Y, Huang R, Ye J, Han X, Meng T, Song D, Yin H. Identification of key eRNAs for intervertebral disc degeneration by integrated multinomial bioinformatics analysis. BMC Musculoskelet Disord 2024; 25:356. [PMID: 38704519 PMCID: PMC11069191 DOI: 10.1186/s12891-024-07438-6] [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: 07/10/2023] [Accepted: 04/12/2024] [Indexed: 05/06/2024] Open
Abstract
BACKGROUND Intervertebral disc degeneration (IVDD) is a common degenerative condition leading to abnormal stress distribution under load, causing intervertebral stenosis, facet joint degeneration, and foraminal stenosis. Very little is known about the molecular mechanism of eRNAs in IVDD. METHODS Gene expression profiles of 38 annulus disc samples composed of 27 less degenerated discs (LDs) and 11 more degenerated discs (MDs) were retrieved from the GEO database. Then, differentially expressed enhancer RNAs (DEeRNAs), differentially expressed target genes (DETGs), and differentially expressed transcription factors (DETFs), hallmark of cancer signalling pathways according to GSVA; the types and quantity of immune cells according to CIBERSORT; and immune gene sets according to ssGSEA were analysed to construct an IVDD-related eRNA network. Then, multidimensional validation was performed to explore the interactions among DEeRNAs, DETFs and DEGs in space. RESULTS A total of 53 components, 14 DETGs, 15 DEeRNAs, 3 DETFs, 5 immune cells, 9 hallmarks, and 7 immune gene sets, were selected to construct the regulatory network. After validation by online multidimensional databases, 21 interactive DEeRNA-DEG-DETF axes related to IVDD exacerbation were identified, among which the C1S-CTNNB1-CHD4 axis was the most significant. CONCLUSION Based upon the results of our study, we theorize that the C1S-CTNNB1-CHD4 axis plays a vital role in IVDD exacerbation. Specifically, C1S recruits CTNNB1 and upregulates the expression of CHD4 in IVDD, and subsequently, CHD4 suppresses glycolysis and activates oxidative phosphorylation, thus generating insoluble collagen fibre deposits and leading to the progression of IVDD. Overall, these DEeRNAs could comprise promising therapeutic targets for IVDD due to their high tissue specificity.
Collapse
Affiliation(s)
- Yongai Li
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Runzhi Huang
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Jianxin Ye
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaying Han
- Department of General Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Tong Meng
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Dianwen Song
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Huabin Yin
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
49
|
Jia S, Yang T, Gao S, Bai L, Zhu Z, Zhao S, Wang Y, Liang X, Li Y, Gao L, Zhang Z, Gao X, Li D, Chen S, Zhang B, Meng C. Exosomes from umbilical cord mesenchymal stem cells ameliorate intervertebral disc degeneration via repairing mitochondrial dysfunction. J Orthop Translat 2024; 46:103-115. [PMID: 38841339 PMCID: PMC11150913 DOI: 10.1016/j.jot.2023.10.004] [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: 05/25/2023] [Revised: 10/04/2023] [Accepted: 10/11/2023] [Indexed: 06/07/2024] Open
Abstract
Background Reactive oxygen species (ROS), predominantly generated by mitochondria, play a crucial role in the pathogenesis of intervertebral disc degeneration (IVDD). Reduction of ROS levels may be an effective strategy to delay IVDD. In this study, we assessed whether umbilical cord mesenchymal stem cell-exosomes (UCMSC-exos) can be used to treat IVDD by suppressing ROS production caused by mitochondrial dysfunction. Materials and methods Human UCMSC-exos were isolated and identified. Nucleus pulposus cells (NPCs) were stimulated with H2O2 in the presence or absence of exosomes. Then, 4D label free quantitative (4D-LFQ) proteomics were used to analyze the differentially expressed (DE) proteins. Mitochondrial membrane potential (MMP), mitochondrial ROS and protein levels were determined via immunofluorescence staining, flow cytometry and western blotting respectively. Additionally, high-throughput sequencing was performed to identify the DE miRNAs in NPCs. Finally, therapeutic effects of UCMSC-exos were investigated in a puncture-induced IVDD rat model. Degenerative grades of rat IVDs were assessed using magnetic resonance imaging and histochemical staining. Results UCMSC-exos effectively improved the viability of NPCs and restored the expression of the extracellular matrix (ECM) proteins, collagen type II alpha-1 (COL2A1) and matrix metalloproteinase-13 induced by H2O2. Additionally, UCMSC-exos not only reduced the total intracellular ROS and mitochondrial superoxide levels, but also increased MMP in pathological NPCs. 4D-LFQ proteomics and western blotting further revealed that UCMSC-exos up-regulated the levels of the mitochondrial protein, mitochondrial transcription factor A (TFAM), in H2O2-induced NPCs. High-throughput sequencing and qRT-PCR uncovered that UCMSC-exos down-regulated the levels of miR-194-5p, a potential negative regulator of TFAM, induced by H2O2. Finally, in vivo results showed that UCMSC-exos injection improved the histopathological structure and enhanced the expression levels of COL2A1 and TFAM in the rat IVDD model. Conclusions Our findings suggest that UCMSC-exos promote ECM synthesis, relieve mitochondrial oxidative stress, and attenuate mitochondrial dysfunction in vitro and in vivo, thereby effectively treating IVDD. The translational potential of this article This study provides solid experimental data support for the therapeutic effects of UCMSC-exos on IVDD, suggesting that UCMSC-exos will be a promising nanotherapy for IVDD.
Collapse
Affiliation(s)
- Shu Jia
- Clinical Research Team of Spine & Spinal Cord Diseases, Medical Research Center, Affiliated Hospital of Jining Medical University, 89 Guhuai Road, Jining, Shandong Province, 272000, China
- Postdoctoral Mobile Station, Shandong University of Traditional Chinese Medicine, 4655 Daxue Road, Jinan, Shandong Province, 250355, China
| | - Tao Yang
- Department of Spine Surgery, Affiliated Hospital of Jining Medical University, 89 Guhuai Road, Jining, Shandong Province, 272000, China
| | - Sheng Gao
- Department of Spine Surgery, Affiliated Hospital of Jining Medical University, 89 Guhuai Road, Jining, Shandong Province, 272000, China
| | - Luyue Bai
- Department of Spine Surgery, Affiliated Hospital of Jining Medical University, 89 Guhuai Road, Jining, Shandong Province, 272000, China
| | - Zhiguo Zhu
- Clinical Research Team of Spine & Spinal Cord Diseases, Medical Research Center, Affiliated Hospital of Jining Medical University, 89 Guhuai Road, Jining, Shandong Province, 272000, China
- Postdoctoral Mobile Station, Shandong University of Traditional Chinese Medicine, 4655 Daxue Road, Jinan, Shandong Province, 250355, China
| | - Siqi Zhao
- Department of Spine Surgery, Affiliated Hospital of Jining Medical University, 89 Guhuai Road, Jining, Shandong Province, 272000, China
| | - Yexin Wang
- Department of Spine Surgery, Affiliated Hospital of Jining Medical University, 89 Guhuai Road, Jining, Shandong Province, 272000, China
| | - Xiao Liang
- Department of Spine Surgery, Affiliated Hospital of Jining Medical University, 89 Guhuai Road, Jining, Shandong Province, 272000, China
| | - Yanpeng Li
- Department of Spine Surgery, Affiliated Hospital of Jining Medical University, 89 Guhuai Road, Jining, Shandong Province, 272000, China
| | - Longfei Gao
- Department of Spine Surgery, Affiliated Hospital of Jining Medical University, 89 Guhuai Road, Jining, Shandong Province, 272000, China
| | - Zifang Zhang
- Department of Spine Surgery, Affiliated Hospital of Jining Medical University, 89 Guhuai Road, Jining, Shandong Province, 272000, China
| | - Xu Gao
- Department of Medicine, Qingdao University, 38 Dengzhou Road, Qingdao, Shandong Province, 266021, China
| | - Dongru Li
- Department of Clinical Medical College, Jining Medical University, 45 Jianshe Road, Jining, Shandong Province, 272000, China
| | - Shang Chen
- Clinical Research Team of Spine & Spinal Cord Diseases, Medical Research Center, Affiliated Hospital of Jining Medical University, 89 Guhuai Road, Jining, Shandong Province, 272000, China
| | - Bin Zhang
- Department of Laboratory Medicine, Affiliated Hospital of Jining Medical University, 89 Guhuai Road, Jining, Shandong Province, 272000, China
| | - Chunyang Meng
- Clinical Research Team of Spine & Spinal Cord Diseases, Medical Research Center, Affiliated Hospital of Jining Medical University, 89 Guhuai Road, Jining, Shandong Province, 272000, China
- Department of Spine Surgery, Affiliated Hospital of Jining Medical University, 89 Guhuai Road, Jining, Shandong Province, 272000, China
| |
Collapse
|
50
|
Li B, Shen E, Wu Z, Qi H, Wu C, Liu D, Jiang X. BMSC-Derived Exosomes Attenuate Rat Osteoarthritis by Regulating Macrophage Polarization through PINK1/Parkin Signaling Pathway. Cartilage 2024:19476035241245805. [PMID: 38641989 PMCID: PMC11569690 DOI: 10.1177/19476035241245805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 03/15/2024] [Accepted: 03/22/2024] [Indexed: 04/21/2024] Open
Abstract
OBJECTIVE Exosomes derived from bone marrow mesenchymal stem cells (BMSC-Exos) may modulate the M1/M2 polarization of macrophages during osteoarthritis (OA). However, the underlying mechanisms of BMSC-Exos in this process still need to be elucidated. In this study, we explored the role of BMSC-Exos in the polarization of macrophages in vitro and the OA rats in vivo. METHODS The effects of BMSC-Exos on RAW264.7 cells were determined, including the production of reactive oxygen species (ROS) and the protein expression of Akt, PINK1, and Parkin. We prepared an OA model by resecting the anterior cruciate ligament and medial meniscus of Sprague-Dawley (SD) rats. Hematoxylin-eosin (H&E) and safranin O-fast green staining, immunohistochemistry and immunofluorescence analyses, and the examination of interleukin 6 (IL-6), interleukin 1β (IL-1β), tumor necrosis factor alpha (TNF-α), and interleukin 10 (IL-10) were performed to assess changes in cartilage and synovium. RESULTS BMSC-Exos inhibited mitochondrial membrane damage, ROS production, and the protein expression of PINK1 and Parkin. Akt phosphorylation was downregulated under lipopolysaccharide (LPS) induction but significantly recovered after treatment with BMSC-Exos. BMSC-Exos alleviated cartilage damage, inhibited M1 polarization, and promoted M2 polarization in the synovium in OA rats. The expression of PINK1 and Parkin in the synovium and the levels of IL-6, IL-1β, and TNF-α in the serum decreased, but the level of IL-10 increased when BMSC-Exos were used in OA rats. CONCLUSION BMSC-Exos ameliorate OA development by regulating synovial macrophage polarization, and one of the underlying mechanisms may be through inhibiting PINK1/Parkin signaling.
Collapse
Affiliation(s)
- Beibei Li
- Department of Orthopaedics, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Enpu Shen
- Shanghai Putuo District Central Hospital, Shanghai, China
| | - Zhiwen Wu
- Department of Orthopaedics, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Hui Qi
- Beijing Research Institute of Traumatology and Orthopaedics, Beijing, China
| | - Cheng’ai Wu
- Beijing Research Institute of Traumatology and Orthopaedics, Beijing, China
| | - Danping Liu
- Department of Orthopaedics, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Xu Jiang
- Department of Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|