1
|
Fan Y, Niu Z, Yin L, Yao L, Ding S, Tong Y, Wang J, Hong Z, Chen J, Zhang Q, Ji L, Chen J, Xia C, Bi Q. Membrane biomimetic nanoenzyme-incorporated hybrid glycyrrhizic acid hydrogel for precise mitochondrial ROS scavenging for osteoarthritis treatment. Mater Today Bio 2025; 32:101778. [PMID: 40290887 PMCID: PMC12032948 DOI: 10.1016/j.mtbio.2025.101778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 04/13/2025] [Accepted: 04/17/2025] [Indexed: 04/30/2025] Open
Abstract
Osteoarthritis (OA) is a progressive degenerative disorder which severely threatens the quality of life of older individuals. OA progression is closely related to heightened levels of mitochondrial reactive oxygen species (mtROS). Although nanozymes have a good ROS-scavenging effect, they cannot precisely scavenge mtROS because of the immune rejection of cell membranes, lysosomal escape, and the inability of conventional nanozymes to directly target mitochondria. Dual-target nanozymes were engineered to precisely scavenge mtROS in chondrocytes. We used chondrocyte membrane-camouflaged TPP-modified hollow Prussian blue nanozymes and subsequently encapsulated these nanozymes in a hybrid glycyrrhizic acid hydrogel. The therapeutic efficacy and underlying mechanisms were assessed in vitro and in vivo. The novel nanozymes enhanced cell selectivity, immune evasion capabilities, and mitochondrial targeting. The dual-targeted nanozymes exerted a pronounced therapeutic impact on inflammatory chondrocytes, mitigated mtDNA leakage by precisely scavenging mtROS, dampened cGAS-STING-NF-κB signaling, and enhanced chondrocyte function. The hybrid hydrogels also exhibited improved therapeutic outcomes. We confirmed the beneficial effects of the nanozyme-hydrogel combination on OA progression in mice. The nanozyme-hydrogel combination can reduce precisely scavenge mtROS in chondrocytes, avoiding the leakage of mtDNA and suppressing the cGAS-STING-NF-κB signaling pathway, thereby decreasing inflammatory responses and alleviate OA progression.
Collapse
Affiliation(s)
- Yong Fan
- Department of Sports Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310000, China
- Postgraduate training base Alliance of Wenzhou Medical University, Wenzhou, Zhejiang Province, 325000, China
- Center for Rehabilitation Medicine, Department of Orthopedics, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310000, China
- Institute of Sports Medicine and Osteoarthropathy of Hangzhou Medical College, Hangzhou, Zhejiang, 310000, China
| | - Zexuan Niu
- Department of Sports Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310000, China
- Postgraduate training base Alliance of Wenzhou Medical University, Wenzhou, Zhejiang Province, 325000, China
- Center for Rehabilitation Medicine, Department of Orthopedics, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310000, China
- Institute of Sports Medicine and Osteoarthropathy of Hangzhou Medical College, Hangzhou, Zhejiang, 310000, China
| | - Li Yin
- Department of Sports Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310000, China
- Postgraduate training base Alliance of Wenzhou Medical University, Wenzhou, Zhejiang Province, 325000, China
- Center for Rehabilitation Medicine, Department of Orthopedics, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310000, China
- Institute of Sports Medicine and Osteoarthropathy of Hangzhou Medical College, Hangzhou, Zhejiang, 310000, China
| | - Longtao Yao
- Department of Sports Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310000, China
- Postgraduate training base Alliance of Wenzhou Medical University, Wenzhou, Zhejiang Province, 325000, China
- Center for Rehabilitation Medicine, Department of Orthopedics, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310000, China
- Institute of Sports Medicine and Osteoarthropathy of Hangzhou Medical College, Hangzhou, Zhejiang, 310000, China
| | - Sheyuan Ding
- Department of Sports Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310000, China
- Center for Rehabilitation Medicine, Department of Orthopedics, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310000, China
- Institute of Sports Medicine and Osteoarthropathy of Hangzhou Medical College, Hangzhou, Zhejiang, 310000, China
| | - Yu Tong
- Department of Sports Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310000, China
- Center for Rehabilitation Medicine, Department of Orthopedics, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310000, China
- Institute of Sports Medicine and Osteoarthropathy of Hangzhou Medical College, Hangzhou, Zhejiang, 310000, China
| | - Jiao Wang
- Department of Sports Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310000, China
- Center for Rehabilitation Medicine, Department of Orthopedics, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310000, China
- Institute of Sports Medicine and Osteoarthropathy of Hangzhou Medical College, Hangzhou, Zhejiang, 310000, China
| | - Zheping Hong
- Department of Sports Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310000, China
- Center for Rehabilitation Medicine, Department of Orthopedics, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310000, China
- Institute of Sports Medicine and Osteoarthropathy of Hangzhou Medical College, Hangzhou, Zhejiang, 310000, China
| | - Jihang Chen
- Department of Sports Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310000, China
- Center for Rehabilitation Medicine, Department of Orthopedics, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310000, China
- Institute of Sports Medicine and Osteoarthropathy of Hangzhou Medical College, Hangzhou, Zhejiang, 310000, China
| | - Qiong Zhang
- Department of Nursing, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310000, China
| | - Lichen Ji
- Department of Joint Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200092, China
| | - Jiaxin Chen
- Center for Plastic & Reconstructive Surgery, Department of Plastic & Reconstructive Surgery, Zhejiang Provincial People's Hospital ( Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310000, China
| | - Chen Xia
- Department of Sports Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310000, China
- Center for Rehabilitation Medicine, Department of Orthopedics, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310000, China
- Institute of Sports Medicine and Osteoarthropathy of Hangzhou Medical College, Hangzhou, Zhejiang, 310000, China
| | - Qing Bi
- Department of Sports Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310000, China
- Postgraduate training base Alliance of Wenzhou Medical University, Wenzhou, Zhejiang Province, 325000, China
- Center for Rehabilitation Medicine, Department of Orthopedics, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310000, China
- Institute of Sports Medicine and Osteoarthropathy of Hangzhou Medical College, Hangzhou, Zhejiang, 310000, China
| |
Collapse
|
2
|
Chen P, Zeng L, Wang T, He J, Xiong S, Chen G, Wang Q, Chen H, Xie J. The communication role of extracellular vesicles in the osteoarthritis microenvironment. Front Immunol 2025; 16:1549833. [PMID: 40165965 PMCID: PMC11955493 DOI: 10.3389/fimmu.2025.1549833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Accepted: 02/26/2025] [Indexed: 04/02/2025] Open
Abstract
Osteoarthritis (OA) is the most common degenerative joint disease worldwide, characterized by synovial inflammation, cartilage loss, and reactive hyperplasia of subchondral bone, affecting the quality of life of hundreds of millions of people. However, the molecular mechanisms underlying the occurrence and progression of OA remain unclear, and there is no therapy can substantially interrupt or reverse the destructive process of OA. More insight into the pathogenesis of OA may result in innovative therapeutics. The OA microenvironment plays a pivotal role in the development and progression of OA, which encompasses chondrocytes, adipocytes, synovial fibroblasts, endothelial cells, and immune cells. Extracellular vesicles (EVs) have emerged as a novel form of intercellular communication, mediating the transfer of a range of bioactive molecules to create a specific microenvironment. Recent studies have reported that the cargos of EVs play a crucial role in the pathogenesis of OA, including noncoding RNAs (ncRNAs), proteins, and lipids. This review systematically analyzes and summarizes the biological characteristics and functionalities of EVs derived from diverse cellular sources, especially how EVs mediate communication between different cells in the OA microenvironment, with a view to providing new insights into the pathogenesis of OA.
Collapse
Affiliation(s)
- Pu Chen
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Lingfeng Zeng
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
- Guangdong Provincial Key Laboratory of Chinese Medicine for Prevention and Treatment of Refractory Chronic Diseases, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
- Bone and Joint Research Team of Degeneration and Injury, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China
| | - Ting Wang
- Department of Operating Room, Ji’an County Traditional Chinese Medicine Hospital, Ji’an, Jiangxi, China
| | - Jianbo He
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Shuai Xiong
- School of Anesthesiology, Shandong Second Medical University, Weifang, Shandong, China
| | - Gang Chen
- Department of Orthopedic Surgery, Jiangxi University of Traditional Chinese Medicine Affiliated Hospital, Nanchang, Jiangxi, China
| | - Qingfu Wang
- Department of Orthopedic Surgery, Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing, China
| | - Haiyun Chen
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
- Guangdong Provincial Hospital of Chinese Medicine, Zhuhai Hospital, Zhuhai, China
| | - Jiewei Xie
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| |
Collapse
|
3
|
Longfei H, Wenyuan H, Weihua F, Peng P, Sun L, Kun L, Mincong H, Fan Y, Wei H, Qiushi W. Exosomes in cartilage microenvironment regulation and cartilage repair. Front Cell Dev Biol 2025; 13:1460416. [PMID: 40109360 PMCID: PMC11919854 DOI: 10.3389/fcell.2025.1460416] [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: 07/06/2024] [Accepted: 02/17/2025] [Indexed: 03/22/2025] Open
Abstract
Osteoarthritis (OA) is a debilitating disease that predominantly impacts the hip, hand, and knee joints. Its pathology is defined by the progressive degradation of articular cartilage, formation of bone spurs, and synovial inflammation, resulting in pain, joint function limitations, and substantial societal and familial burdens. Current treatment strategies primarily target pain alleviation, yet improved interventions addressing the underlying disease pathology are scarce. Recently, exosomes have emerged as a subject of growing interest in OA therapy. Numerous studies have investigated exosomes to offer promising therapeutic approaches for OA through diverse in vivo and in vitro models, elucidating the mechanisms by which exosomes from various cell sources modulate the cartilage microenvironment and promote cartilage repair. Preclinical investigations have demonstrated the regulatory effects of exosomes originating from human cells, including mesenchymal stem cells (MSC), synovial fibroblasts, chondrocytes, macrophages, and exosomes derived from Chinese herbal medicines, on the modulation of the cartilage microenvironment and cartilage repair through diverse signaling pathways. Additionally, therapeutic mechanisms encompass cartilage inflammation, degradation of the cartilage matrix, proliferation and migration of chondrocytes, autophagy, apoptosis, and mitigation of oxidative stress. An increasing number of exosome carrier scaffolds are under development. Our review adopts a multidimensional approach to enhance comprehension of the pivotal therapeutic functions exerted by exosomes sourced from diverse cell types in OA. Ultimately, our aim is to pinpoint therapeutic targets capable of regulating the cartilage microenvironment and facilitating cartilage repair in OA.
Collapse
Affiliation(s)
- Han Longfei
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Hou Wenyuan
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Fang Weihua
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Peng Peng
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Lu Sun
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Lin Kun
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - He Mincong
- Traumatology and Orthopedics Institute of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Department of Orthopaedics, The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Yang Fan
- Traumatology and Orthopedics Institute of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Department of Orthopaedics, The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - He Wei
- Traumatology and Orthopedics Institute of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Department of Orthopaedics, The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Wei Qiushi
- Traumatology and Orthopedics Institute of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Department of Orthopaedics, The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- State Key Laboratory of Traditional Chinese Medicine Syndrome/Orthopaedic, The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| |
Collapse
|
4
|
Guan Z, Liu Y, Luo L, Jin X, Guan Z, Yang J, Liu S, Tao K, Pan J. Sympathetic innervation induces exosomal miR-125 transfer from osteoarthritic chondrocytes, disrupting subchondral bone homeostasis and aggravating cartilage damage in aging mice. J Adv Res 2025; 69:245-260. [PMID: 38554999 PMCID: PMC11954818 DOI: 10.1016/j.jare.2024.03.022] [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: 10/12/2023] [Revised: 03/16/2024] [Accepted: 03/27/2024] [Indexed: 04/02/2024] Open
Abstract
INTRODUCTION Osteoarthritis (OA) is a progressive disease that poses a significant threat to human health, particularly in aging individuals: Although sympathetic activation has been implicated in bone metabolism, its role in the development of OA related to aging remains poorly understood. Therefore, this study aimed to investigate how sympathetic regulation impacts aging-related OA through experiments conducted both in vivo and in vitro. METHODS To analyze the effect of sympathetic regulation on aging-related OA, we conducted experiments using various mouse models. These models included a natural aging model, a medial meniscus instability model, and a load-induced model, which were used to examine the involvement of sympathetic nerves. In order to evaluate the expression levels of β1-adrenergic receptor (Adrβ1) and sirtuin-6 (Sirt6) in chondrocytes of naturally aging OA mouse models, we performed assessments. Additionally, we investigated the influence of β1-adrenergic receptor knockout or treatment with a β1-adrenergic receptor blocker on the progression of OA in aging mice and detected exosome release and detected downstream signaling expression by inhibiting exosome release. Furthermore, we explored the impact of sympathetic depletion through tyrosine hydroxylase (TH) on OA progression in aging mice. Moreover, we studied the effects of norepinephrine(NE)-induced activation of the β1-adrenergic receptor signaling pathway on the release of exosomes and miR-125 from chondrocytes, subsequently affecting osteoblast differentiation in subchondral bone. RESULTS Our findings demonstrated a significant increase in sympathetic activity, such as NE levels, in various mouse models of OA including natural aging, medial meniscus instability, and load-induced models. Notably, we observed alterations in the expression levels of β1-adrenergic receptor and Sirt6 in chondrocytes in OA mouse models associated with natural aging, leading to an improvement in the progression of OA. Critically, we found that the knockout of β1-adrenergic receptor or treatment with a β1-adrenergic receptor blocker attenuated OA progression in aging mice and the degraded cartilage explants produced more exosome than the nondegraded ones, Moreover, sympathetic depletion through TH was shown to ameliorate OA progression in aging mice. Additionally, we discovered that NE-induced activation of the β1-adrenergic receptor signaling pathway facilitated the release of exosomes and miR-125 from chondrocytes, promoting osteoblast differentiation in subchondral bone. CONCLUSION In conclusion, our study highlights the role of sympathetic innervation in facilitating the transfer of exosomal miR-125 from osteoarthritic chondrocytes, ultimately disrupting subchondral bone homeostasis and exacerbating cartilage damage in aging mice. These findings provide valuable insights into the potential contribution of sympathetic regulation to the pathogenesis of aging-related OA.
Collapse
Affiliation(s)
- Zhiyuan Guan
- Department of Orthopedics, The Shanghai Tenth People's Hospital of Tongji University, Shanghai, China; Science and Technology Center, Fenyang College of Shanxi Medical University, Shanxi 032200, China
| | - Yanbin Liu
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiaotong University, No. 100 Haining Road, Shanghai 200080, China
| | - Liying Luo
- Department of Ophthalmology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiao Jin
- Department of Rheumatology and Immunology, Xuzhou Municipal Hospital Affiliated with Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
| | - Zhiqiang Guan
- Department of Dermatology, Xuzhou Municipal Hospital Affiliated with Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
| | - Jianjun Yang
- Department of Orthopedics, The Shanghai Tenth People's Hospital of Tongji University, Shanghai, China
| | - Shengfu Liu
- Department of Orthopedics, The Shanghai Tenth People's Hospital of Tongji University, Shanghai, China
| | - Kun Tao
- Department of Orthopedics, The Shanghai Tenth People's Hospital of Tongji University, Shanghai, China
| | - Jianfeng Pan
- Science and Technology Center, Fenyang College of Shanxi Medical University, Shanxi 032200, China.
| |
Collapse
|
5
|
Ye T, Yan J, Kan T, Xie G, Zhang Z, Yin W, Zhao B, Yu Z, Chu L. Articular cartilage degeneration and aberrant osteocyte perilacunar/canalicular remodeling in subchondral bone of patients with developmental dysplasia of the hip. BMC Musculoskelet Disord 2025; 26:165. [PMID: 39966795 PMCID: PMC11837434 DOI: 10.1186/s12891-025-08419-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 02/12/2025] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND Developmental dysplasia of the hip (DDH) is a congenital musculoskeletal disease that impairs the hip joint and exacerbates hip osteoarthritis. This study aims to investigate the alterations of osteocytic characteristics including apoptosis, lacuna-canalicular network, and perilacunar/canalicular remodeling (PLR) activity in subchondral bone from DDH patients, and potential relationship of these alterations between the cartilage degeneration and DDH progression. METHODS The femoral head specimens were acquired from 16 patients with hip fractures who received total hip arthroplasty operation, 24 patients with primary hip OA and 25 patients with DDH. The femoral head were scanned by a micro-computed tomography and the volume of interest was used for a micro-finite element analysis. Histological and immunohistochemical staining was used to observe chondrocytes in cartilage and osteocytes in subchondral bone. Terminal deoxynucleotidyl transferase dUTP nick end labeling staining was used to investigate the apoptotic osteocytes in subchondral bone. Ploton silver staining was used to visualize lacunocanalicular network and picrosirius red staining was to visualize collagen fiber orientation in subchondral bone. RESULTS The DDH group showed the highest apoptosis rate of osteocytes and increased PLR activity among the three groups. The micro-finite-element analysis revealed that DDH group had deteriorative microstructural and biomechanical properties of subchondral bone. The histological and immunohistochemical analyses showed that the cartilage degeneration in DDH group was the most severe. Linear regression analysis revealed a significant correlation between osteocytic activity in subchondral bone and cartilage degeneration in DDH. CONCLUSIONS Our findings indicate that the abnormal osteocyte activity in subchondral bone might contribute to the deterioration of subchondral bone structure, which accelerates cartilage degeneration and DDH progression. Targeting subchondral bone remodeling could offer a promising therapeutic strategy for DDH.
Collapse
Affiliation(s)
- Teng Ye
- Department of Sports Medicine, National Center for Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiren Yan
- Department of Sports Medicine, National Center for Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tianyou Kan
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200127, China
| | - Guoming Xie
- Department of Sports Medicine, National Center for Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhichang Zhang
- Department of Sports Medicine, National Center for Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenjing Yin
- Department of Sports Medicine, National Center for Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bizeng Zhao
- Department of Sports Medicine, National Center for Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhifeng Yu
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Linyang Chu
- Department of Sports Medicine, National Center for Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
6
|
Li X, Zhang Z, Jiang W, Ju Y, Guo W, Huang Z. Dipeptidyl Peptidase 4 (DPP4) Exacerbates Osteoarthritis Progression in an Enzyme-Independent Manner. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410525. [PMID: 39680708 PMCID: PMC11809337 DOI: 10.1002/advs.202410525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 11/25/2024] [Indexed: 12/18/2024]
Abstract
Chondrocyte senescence is a key driver of osteoarthritis (OA). Mitochondrial dysfunction and oxidative stress can induce chondrocyte senescence. However, the specific mechanisms by which senescence contributes to OA progression are not fully understood. Here, it is attested that Dipeptidyl peptidase 4 (DPP4) is significantly upregulated in osteoarthritic chondrocytes in both humans and mice. DPP4 promotes oxidative stress and cellular senescence in chondrocytes through excessive mitochondrial fission in an enzyme-independent manner. Intra-articular injection of adeno-associated virus 2 to upregulate DPP4 in chondrocytes promotes post-traumatic and aging-induced OA in mice in an enzyme-independent manner. Mechanistically, DPP4 competitively binds to Myosin heavy chain 9 (MYH9), interfering with its E3 ubiquitin ligase Carboxyl terminus of Hsc70-interacting protein (CHIP), and thereby upregulates MYH9 expression. Finally, a small molecule, 4,5-Dicaffeoylquinic acid is identified, which disrupts the interaction between DPP4 and MYH9, thereby ameliorating post-traumatic and aging-induced OA in mice caused by DPP4 upregulation. The study indicates that the non-enzymatic activity of DPP4 is a promising target for OA treatment.
Collapse
Affiliation(s)
- Xinyu Li
- Department of Orthopaedic SurgeryOrthopaedic Research InstituteWest China HospitalWest China Medical SchoolSichuan UniversityChengdu610041China
| | - Zhao Zhang
- Department of Orthopaedic SurgeryOrthopaedic Research InstituteWest China HospitalWest China Medical SchoolSichuan UniversityChengdu610041China
| | - Wenyu Jiang
- Department of Orthopaedic SurgeryOrthopaedic Research InstituteWest China HospitalWest China Medical SchoolSichuan UniversityChengdu610041China
| | - Yucan Ju
- Department of Orthopaedic SurgeryOrthopaedic Research InstituteWest China HospitalWest China Medical SchoolSichuan UniversityChengdu610041China
| | - Weihua Guo
- Department of Immuno‐OncologyBeckman Research Institute at City of HopeNational Medical CenterDuarte91010USA
| | - Zeyu Huang
- Department of Orthopaedic SurgeryOrthopaedic Research InstituteWest China HospitalWest China Medical SchoolSichuan UniversityChengdu610041China
| |
Collapse
|
7
|
Huang C, Xiao Y, Qing L, Tang J, Wu P. Exosomal non-coding RNAs in the regulation of bone metabolism homeostasis: Molecular mechanism and therapeutic potential. Heliyon 2025; 11:e41632. [PMID: 39911437 PMCID: PMC11795052 DOI: 10.1016/j.heliyon.2025.e41632] [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: 07/04/2024] [Revised: 12/24/2024] [Accepted: 01/01/2025] [Indexed: 02/07/2025] Open
Abstract
Bone metabolism is a dynamic balance between bone formation and absorption regulated by osteoblasts/osteoclasts. Bone metabolic disorders can lead to metabolic bone disease. Osteoporosis (OP), osteoarthritis (OA) and femoral head necrosis (ONFH) are common metabolic bone diseases. At present, the treatment of metabolic bone disease is still mainly to relieve pain and improve joint function. However, surgical treatment does not apply to the vast majority of high-risk groups, including postmenopausal women, patients with diabetes, cirrhosis, etc. Exosomes (Exos) are nanoscale membrane vesicles that are released by almost all cells. Exos are rich in a variety of bioactive substances, such as non-coding RNAs, nucleic acids, proteins and lipids. In view of the structure of Exos, it can protect the biologically active molecules can be smoothly delivered to the target cells and involved in the regulation of cell function. In this review, we focus on the regulation mechanism and function of bone homeostasis mediated by exosomal ncRNAs (Exos-ncRNAs), including macrophage polarization, autophagy, angiogenesis, signal transduction and competing endogenous RNA (ceRNA). We summarized the therapeutic strategies and potential drugs of Exos-ncRNAs in metabolic bone disease. Moreover, we discussed the shortcomings and potential research directions of Exos as carrier to deliver ncRNAs to play a role.
Collapse
Affiliation(s)
- Chengxiong Huang
- Department of Orthopedics, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
- National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
| | - Yu Xiao
- Department of Orthopedics, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
- National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
| | - Liming Qing
- Department of Orthopedics, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
- National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
| | - Juyu Tang
- Department of Orthopedics, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
- National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
| | - Panfeng Wu
- Department of Orthopedics, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
- National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
| |
Collapse
|
8
|
Piñeiro-Ramil M, Gómez-Seoane I, Rodríguez-Cendal AI, Sanjurjo-Rodríguez C, Riva-Mendoza S, Fuentes-Boquete I, De Toro-Santos J, Señarís-Rodríguez J, Díaz-Prado S. Disease-Associated Signatures Persist in Extracellular Vesicles from Reprogrammed Cells of Osteoarthritis Patients. Int J Mol Sci 2025; 26:870. [PMID: 39940641 PMCID: PMC11816895 DOI: 10.3390/ijms26030870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 01/07/2025] [Accepted: 01/15/2025] [Indexed: 02/16/2025] Open
Abstract
Osteoarthritis (OA) is a prevalent joint disorder that lacks effective therapies to halt cartilage degeneration. Mesenchymal stromal cell (MSC)-derived small extracellular vesicles (sEVs) are being investigated as promising chondroprotective agents. Compared to primary MSCs, induced pluripotent stem cell (iPSC)-derived MSCs (MLCs) offer superior scalability and enhanced paracrine activity. The aim of this study was to explore the feasibility of using autologous MLC-derived sEVs as a potential therapeutic strategy for OA through the analysis of their protein cargo. iPSCs from an OA patient and a healthy donor were differentiated into MLCs. sEVs were isolated from these MLCs and characterized, with a particular focus on their protein cargo. Both iPSC lines were successfully differentiated into MLCs, which secreted sEVs with comparable size distributions and yields. The analysis of differentially expressed proteins revealed a high abundance of proteins associated with OA pathology and cartilage degradation in sEVs from OA MLCs compared to those from healthy MLCs. The persistence of OA-associated protein signatures in autologous MLC-derived sEVs may limit their therapeutic efficacy. These findings underscore the importance of carefully evaluating disease-specific protein profiles in sEVs for regenerative applications.
Collapse
Affiliation(s)
- María Piñeiro-Ramil
- Grupo de Investigación en Terapia Celular y Medicina Regenerativa, Instituto de Investigación Biomédica de A Coruña (INIBIC), Fundación Pública Gallega de Investigación Biomédica INIBIC, Complexo Hospitalario Universitario de A Coruña (CHUAC), Servizo Galego de Saúde (SERGAS), 15006 A Coruña, Spain; (M.P.-R.); (I.G.-S.); (A.I.R.-C.); (I.F.-B.); (J.D.T.-S.); (J.S.-R.)
- Grupo de Investigación en Terapia Celular y Medicina Regenerativa, Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidade da Coruña (UDC), 15006 A Coruña, Spain
- Centro Interdisciplinar de Química y Biología (CICA), Universidade da Coruña (UDC), 15008 A Coruña, Spain
| | - Iván Gómez-Seoane
- Grupo de Investigación en Terapia Celular y Medicina Regenerativa, Instituto de Investigación Biomédica de A Coruña (INIBIC), Fundación Pública Gallega de Investigación Biomédica INIBIC, Complexo Hospitalario Universitario de A Coruña (CHUAC), Servizo Galego de Saúde (SERGAS), 15006 A Coruña, Spain; (M.P.-R.); (I.G.-S.); (A.I.R.-C.); (I.F.-B.); (J.D.T.-S.); (J.S.-R.)
| | - Ana Isabel Rodríguez-Cendal
- Grupo de Investigación en Terapia Celular y Medicina Regenerativa, Instituto de Investigación Biomédica de A Coruña (INIBIC), Fundación Pública Gallega de Investigación Biomédica INIBIC, Complexo Hospitalario Universitario de A Coruña (CHUAC), Servizo Galego de Saúde (SERGAS), 15006 A Coruña, Spain; (M.P.-R.); (I.G.-S.); (A.I.R.-C.); (I.F.-B.); (J.D.T.-S.); (J.S.-R.)
- Grupo de Investigación en Terapia Celular y Medicina Regenerativa, Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidade da Coruña (UDC), 15006 A Coruña, Spain
- Centro Interdisciplinar de Química y Biología (CICA), Universidade da Coruña (UDC), 15008 A Coruña, Spain
| | - Clara Sanjurjo-Rodríguez
- Grupo de Investigación en Terapia Celular y Medicina Regenerativa, Instituto de Investigación Biomédica de A Coruña (INIBIC), Fundación Pública Gallega de Investigación Biomédica INIBIC, Complexo Hospitalario Universitario de A Coruña (CHUAC), Servizo Galego de Saúde (SERGAS), 15006 A Coruña, Spain; (M.P.-R.); (I.G.-S.); (A.I.R.-C.); (I.F.-B.); (J.D.T.-S.); (J.S.-R.)
- Grupo de Investigación en Terapia Celular y Medicina Regenerativa, Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidade da Coruña (UDC), 15006 A Coruña, Spain
- Centro Interdisciplinar de Química y Biología (CICA), Universidade da Coruña (UDC), 15008 A Coruña, Spain
| | - Selva Riva-Mendoza
- Grupo de Investigación en Reumatología (GIR), Instituto de Investigación Biomédica de A Coruña (INIBIC), Fundación Pública Gallega de Investigación Biomédica INIBIC, Complexo Hospitalario Universitario de A Coruña (CHUAC), Servizo Galego de Saúde (SERGAS), 15006 A Coruña, Spain;
| | - Isaac Fuentes-Boquete
- Grupo de Investigación en Terapia Celular y Medicina Regenerativa, Instituto de Investigación Biomédica de A Coruña (INIBIC), Fundación Pública Gallega de Investigación Biomédica INIBIC, Complexo Hospitalario Universitario de A Coruña (CHUAC), Servizo Galego de Saúde (SERGAS), 15006 A Coruña, Spain; (M.P.-R.); (I.G.-S.); (A.I.R.-C.); (I.F.-B.); (J.D.T.-S.); (J.S.-R.)
- Grupo de Investigación en Terapia Celular y Medicina Regenerativa, Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidade da Coruña (UDC), 15006 A Coruña, Spain
- Centro Interdisciplinar de Química y Biología (CICA), Universidade da Coruña (UDC), 15008 A Coruña, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid, Spain
| | - Javier De Toro-Santos
- Grupo de Investigación en Terapia Celular y Medicina Regenerativa, Instituto de Investigación Biomédica de A Coruña (INIBIC), Fundación Pública Gallega de Investigación Biomédica INIBIC, Complexo Hospitalario Universitario de A Coruña (CHUAC), Servizo Galego de Saúde (SERGAS), 15006 A Coruña, Spain; (M.P.-R.); (I.G.-S.); (A.I.R.-C.); (I.F.-B.); (J.D.T.-S.); (J.S.-R.)
- Grupo de Investigación en Terapia Celular y Medicina Regenerativa, Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidade da Coruña (UDC), 15006 A Coruña, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid, Spain
- Servicio de Reumatología, Complexo Hospitalario Universitario de A Coruña (CHUAC), Servizo Galego de Saúde (SERGAS), 15006 A Coruña, Spain
| | - José Señarís-Rodríguez
- Grupo de Investigación en Terapia Celular y Medicina Regenerativa, Instituto de Investigación Biomédica de A Coruña (INIBIC), Fundación Pública Gallega de Investigación Biomédica INIBIC, Complexo Hospitalario Universitario de A Coruña (CHUAC), Servizo Galego de Saúde (SERGAS), 15006 A Coruña, Spain; (M.P.-R.); (I.G.-S.); (A.I.R.-C.); (I.F.-B.); (J.D.T.-S.); (J.S.-R.)
- Grupo de Investigación en Terapia Celular y Medicina Regenerativa, Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidade da Coruña (UDC), 15006 A Coruña, Spain
- Centro Interdisciplinar de Química y Biología (CICA), Universidade da Coruña (UDC), 15008 A Coruña, Spain
- Servicio de Cirugía Ortopédica y Traumatología, Complexo Hospitalario Universitario de A Coruña (CHUAC), Servizo Galego de Saúde (SERGAS), 15006 A Coruña, Spain
| | - Silvia Díaz-Prado
- Grupo de Investigación en Terapia Celular y Medicina Regenerativa, Instituto de Investigación Biomédica de A Coruña (INIBIC), Fundación Pública Gallega de Investigación Biomédica INIBIC, Complexo Hospitalario Universitario de A Coruña (CHUAC), Servizo Galego de Saúde (SERGAS), 15006 A Coruña, Spain; (M.P.-R.); (I.G.-S.); (A.I.R.-C.); (I.F.-B.); (J.D.T.-S.); (J.S.-R.)
- Grupo de Investigación en Terapia Celular y Medicina Regenerativa, Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidade da Coruña (UDC), 15006 A Coruña, Spain
- Centro Interdisciplinar de Química y Biología (CICA), Universidade da Coruña (UDC), 15008 A Coruña, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid, Spain
| |
Collapse
|
9
|
Wang Y, Jin S, Guo Y, Zhu L, Lu Y, Li J, Heng BC, Liu Y, Deng X. Cordycepin-Loaded Dental Pulp Stem Cell-Derived Exosomes Promote Aged Bone Repair by Rejuvenating Senescent Mesenchymal Stem Cells and Endothelial Cells. Adv Healthc Mater 2025; 14:e2402909. [PMID: 39551987 DOI: 10.1002/adhm.202402909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 11/04/2024] [Indexed: 11/19/2024]
Abstract
Aging impairs bone marrow mesenchymal stem cell (BMSC) functions as well as associated angiogenesis which is critical for bone regeneration and repair. Hence, repairing bone defects in elderly patients poses a formidable challenge in regenerative medicine. Here, the engineered dental pulp stem cell-derived exosomes loaded with the natural derivative of adenosine Cordycepin (CY@D-exos) are fabricated by means of the intermittent ultrasonic shock, which dually rejuvenates both senescent BMSCs and endothelial cells and significantly improve bone regeneration and repair in aged animals. CY@D-exos can efficiently overcome the senescence of aged BMSCs and enhance their osteogenic differentiation by activating NRF2 signaling and maintaining heterochromatin stability. Importantly, CY@D-exos also potently overcomes the senescence of vascular endothelial cells and promotes angiogenesis. In vivo injectable gelatin methacryloyl (GelMA) hydrogels with sustained release of CY@D-exos can accelerate bone injury repair and promote new blood vessel formation in aged animals. Taken together, these results thus demonstrate that cordycepin-loaded dental pulp stem cell-derived exosomes display considerable potential to be developed as a next-generation therapeutic agent for promoting aged bone regeneration and repair.
Collapse
Affiliation(s)
- Yu Wang
- Department of Orthodontics, Peking University School and Hospital of Stomatology & National Center for Stomatology &National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing, 100081, China
| | - Shanshan Jin
- Department of Pediatric Dentistry, Peking University School and Hospital of Stomatology &National Center for Stomatology &National Clinical Research Center for Oral Diseases &National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing, 100081, China
| | - Yaru Guo
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental Materials, Beijing, 100081, China
| | - Lisha Zhu
- Department of Orthodontics, Peking University School and Hospital of Stomatology & National Center for Stomatology &National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing, 100081, China
| | - Yilong Lu
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental Materials, Beijing, 100081, China
| | - Jing Li
- Department of Orthodontics, Peking University School and Hospital of Stomatology & National Center for Stomatology &National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing, 100081, China
| | - Boon Chin Heng
- Central Laboratory, Peking University School and Hospital of Stomatology & National Center for Stomatology &National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing, 100081, China
| | - Yan Liu
- Department of Orthodontics, Peking University School and Hospital of Stomatology & National Center for Stomatology &National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing, 100081, China
| | - Xuliang Deng
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental Materials, Beijing, 100081, China
| |
Collapse
|
10
|
Peng Y, Wang Y, Bai R, Shi K, Zhou H, Chen C. Nanomaterials: Recent Advances in Knee Osteoarthritis Treatment. Adv Healthc Mater 2024; 13:e2400615. [PMID: 39308252 DOI: 10.1002/adhm.202400615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 08/16/2024] [Indexed: 12/28/2024]
Abstract
Osteoarthritis (OA) of the knee is the most prevalent degenerative joint condition that places a substantial financial and medical burden on society. However, due to drawbacks such as inefficiency, adverse effects, and brief duration of action, the clinical efficacy of the current major therapies for knee OA is largely restricted. Therefore, novel medication development is highly required to address these issues. Numerous studies in recent years have established that nanomaterials can be a potential and highly effective way to overcome these challenges. In this review, the anatomical distinctions between healthy and OA knee joints, as well as novel advances in the field of nanomaterials for the treatment of knee OA are summarized. The limits of the present therapeutic strategies for treating knee OA are also highlighted, as well as the potential prospects of nanomaterials in the future.
Collapse
Affiliation(s)
- Yufeng Peng
- Henan Institutes of Advanced Technology, Zhengzhou University, Zhengzhou, 450052, China
- New Cornerstone Science Laboratory, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology (NCNST), Beijing, 100190, China
| | - Ying Wang
- National Center for Orthopaedics, Department of Molecular Orthopaedics, Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China
| | - Ru Bai
- New Cornerstone Science Laboratory, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology (NCNST), Beijing, 100190, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- Research Unit of Nanoscience and Technology, Chinese Academy of Medical Sciences, Beijing, 100021, China
| | - Kejian Shi
- Department of Thoracic Surgery, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Huige Zhou
- New Cornerstone Science Laboratory, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology (NCNST), Beijing, 100190, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- Research Unit of Nanoscience and Technology, Chinese Academy of Medical Sciences, Beijing, 100021, China
| | - Chunying Chen
- Henan Institutes of Advanced Technology, Zhengzhou University, Zhengzhou, 450052, China
- New Cornerstone Science Laboratory, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology (NCNST), Beijing, 100190, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- Research Unit of Nanoscience and Technology, Chinese Academy of Medical Sciences, Beijing, 100021, China
| |
Collapse
|
11
|
Jin P, Liu H, Chen X, Liu W, Jiang T. From Bench to Bedside: The Role of Extracellular Vesicles in Cartilage Injury Treatment. Biomater Res 2024; 28:0110. [PMID: 39583872 PMCID: PMC11582190 DOI: 10.34133/bmr.0110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 09/20/2024] [Accepted: 10/26/2024] [Indexed: 11/26/2024] Open
Abstract
Cartilage repair is the key to the treatment of joint-related injury. However, because cartilage lacks vessels and nerves, its self-repair ability is extremely low. Extracellular vesicles (EVs) are bilayer nanovesicles with membranes mainly composed of ceramides, cholesterol, phosphoglycerides, and long-chain free fatty acids, containing DNA, RNA, and proteins (such as integrins and enzymes). For mediating intercellular communication and regulating mechanisms, EVs have been shown by multiple studies to be effective treatment options for cartilage repair. This review summarizes recent findings of different sources (mammals, plants, and bacteria) and uses of EVs in cartilage repair, mechanisms of EVs captured by injured chondrocytes, and quantification and storage of EVs, which may provide scientific guidance for promoting the development of EVs in the field of cartilage injury treatment.
Collapse
Affiliation(s)
- Pan Jin
- Health Science Center,
Yangtze University, Jingzhou 434023, Hubei, China
| | - Huan Liu
- Health Science Center,
Yangtze University, Jingzhou 434023, Hubei, China
| | - Xichi Chen
- Health Science Center,
Yangtze University, Jingzhou 434023, Hubei, China
| | - Wei Liu
- Health Science Center,
Yangtze University, Jingzhou 434023, Hubei, China
| | - Tongmeng Jiang
- Key Laboratory of Emergency and Trauma of Ministry of Education, Key Laboratory of Haikou Trauma, Key Laboratory of Hainan Trauma and Disaster Rescue, The First Affiliated Hospital of Hainan Medical University,
Hainan Medical University, Haikou 571199, China
- Engineering Research Center for Hainan Bio-Smart Materials and Bio-Medical Devices, Key Laboratory of Hainan Functional Materials and Molecular Imaging, College of Emergency and Trauma; Hainan Provincial Stem Cell Research Institute; Hainan Academy of Medical Sciences,
Hainan Medical University, Haikou 571199, China
| |
Collapse
|
12
|
Zhong C, Li N, Wang S, Li D, Yang Z, Du L, Huang G, Li H, Yeung WS, He S, Ma S, Wang Z, Jiang H, Zhang H, Li Z, Wen X, Xue S, Tao X, Li H, Xie D, Zhang Y, Chen Z, Wang J, Yan J, Liang Z, Zhang Z, Zhong Z, Wu Z, Wan C, Liang C, Wang L, Yu S, Ma Y, Yu Y, Li F, Chen Y, Zhang B, Lyu A, Ren F, Zhou H, Liu J, Zhang G. Targeting osteoblastic 11β-HSD1 to combat high-fat diet-induced bone loss and obesity. Nat Commun 2024; 15:8588. [PMID: 39362888 PMCID: PMC11449908 DOI: 10.1038/s41467-024-52965-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 09/27/2024] [Indexed: 10/05/2024] Open
Abstract
Excessive glucocorticoid (GC) action is linked to various metabolic disorders. Recent findings suggest that disrupting skeletal GC signaling prevents bone loss and alleviates metabolic disorders in high-fat diet (HFD)-fed obese mice, underpinning the neglected contribution of skeletal GC action to obesity and related bone loss. Here, we show that the elevated expression of 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1), the enzyme driving local GC activation, and GC signaling in osteoblasts, are associated with bone loss and obesity in HFD-fed male mice. Osteoblast-specific 11β-HSD1 knockout male mice exhibit resistance to HFD-induced bone loss and metabolic disorders. Mechanistically, elevated 11β-HSD1 restrains glucose uptake and osteogenic activity in osteoblast. Pharmacologically inhibiting osteoblastic 11β-HSD1 by using bone-targeted 11β-HSD1 inhibitor markedly promotes bone formation, ameliorates glucose handling and mitigated obesity in HFD-fed male mice. Taken together, our study demonstrates that osteoblastic 11β-HSD1 directly contributes to HFD-induced bone loss, glucose handling impairment and obesity.
Collapse
Affiliation(s)
- Chuanxin Zhong
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
- Department of Materials Science and Engineering, Southern University of Science and Technology, Shenzhen, Guangdong, China
- Guangdong-Hong Kong-Macao Greater Bay Area International Research Platform for Aptamer-based Translational Medicine and Drug Discovery, Hong Kong SAR, China
- Institute of Systems Medicine and Health Sciences, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Nanxi Li
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
- Institute of Systems Medicine and Health Sciences, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Shengzheng Wang
- Department of Medicinal Chemistry, School of Pharmacy, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Dijie Li
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
- Guangxi Universities Key Laboratory of Stem cell and Biopharmaceutical Technology, College of Life Sciences, Guangxi Normal University, Gui Lin, China
| | - Zhihua Yang
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lin Du
- Sports Medicine Center, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Guangxin Huang
- Department of Joint Surgery, The Third Affiliated Hospital of Southern Medical University, The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Haitian Li
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Wing Sze Yeung
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Shan He
- Department of Materials Science and Engineering, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Shuting Ma
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Zhuqian Wang
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Hewen Jiang
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Huarui Zhang
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Zhanghao Li
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Xiaoxin Wen
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Song Xue
- Department of Sports Medicine and Rehabilitation, Peking University Shenzhen Hospital, Shenzhen, China
| | - Xiaohui Tao
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Haorui Li
- Sports Medicine Center, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Duoli Xie
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Yihao Zhang
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Zefeng Chen
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Junqin Wang
- Department of Materials Science and Engineering, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Jianfeng Yan
- Department of Materials Science and Engineering, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Zhengming Liang
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Zongkang Zhang
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Zhigang Zhong
- Sports Medicine Center, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Zeting Wu
- International Medical Service Center, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Chao Wan
- Key Laboratory of Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Chao Liang
- Department of Biology, Southern University of Science and Technology, Shenzhen, China
| | - Luyao Wang
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
- Guangdong-Hong Kong-Macao Greater Bay Area International Research Platform for Aptamer-based Translational Medicine and Drug Discovery, Hong Kong SAR, China
- Institute of Systems Medicine and Health Sciences, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Sifan Yu
- Guangdong-Hong Kong-Macao Greater Bay Area International Research Platform for Aptamer-based Translational Medicine and Drug Discovery, Hong Kong SAR, China
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yuan Ma
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
- Guangdong-Hong Kong-Macao Greater Bay Area International Research Platform for Aptamer-based Translational Medicine and Drug Discovery, Hong Kong SAR, China
| | - Yuanyuan Yu
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
- Guangdong-Hong Kong-Macao Greater Bay Area International Research Platform for Aptamer-based Translational Medicine and Drug Discovery, Hong Kong SAR, China
- Institute of Systems Medicine and Health Sciences, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Fangfei Li
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
- Guangdong-Hong Kong-Macao Greater Bay Area International Research Platform for Aptamer-based Translational Medicine and Drug Discovery, Hong Kong SAR, China
- Institute of Systems Medicine and Health Sciences, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Yang Chen
- Key Laboratory of Phytochemistry and Natural Medicines, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Baoting Zhang
- Guangdong-Hong Kong-Macao Greater Bay Area International Research Platform for Aptamer-based Translational Medicine and Drug Discovery, Hong Kong SAR, China
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Aiping Lyu
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China.
- Guangdong-Hong Kong-Macao Greater Bay Area International Research Platform for Aptamer-based Translational Medicine and Drug Discovery, Hong Kong SAR, China.
- Institute of Systems Medicine and Health Sciences, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China.
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Hong Kong, China.
| | - Fuzeng Ren
- Department of Materials Science and Engineering, Southern University of Science and Technology, Shenzhen, Guangdong, China.
| | - Hong Zhou
- Bone Research Program, ANZAC Research Institute, The University of Sydney, Sydney, Australia.
| | - Jin Liu
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China.
- Guangdong-Hong Kong-Macao Greater Bay Area International Research Platform for Aptamer-based Translational Medicine and Drug Discovery, Hong Kong SAR, China.
- Institute of Systems Medicine and Health Sciences, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China.
- Key Laboratory of Phytochemistry and Natural Medicines, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China.
| | - Ge Zhang
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China.
- Guangdong-Hong Kong-Macao Greater Bay Area International Research Platform for Aptamer-based Translational Medicine and Drug Discovery, Hong Kong SAR, China.
- Institute of Systems Medicine and Health Sciences, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China.
| |
Collapse
|
13
|
Zhang G, Kang Y, Dong J, Shi D, Xiang Y, Gao H, Lin Z, Wei X, Ding R, Fan B, Zhang H, Zhu T, Wang L, Yan X. Fluffy hybrid nanoadjuvants for reversing the imbalance of osteoclastic and osteogenic niches in osteoporosis. Bioact Mater 2024; 39:354-374. [PMID: 38846529 PMCID: PMC11153935 DOI: 10.1016/j.bioactmat.2024.05.037] [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: 02/22/2024] [Revised: 05/04/2024] [Accepted: 05/20/2024] [Indexed: 06/09/2024] Open
Abstract
Osteoporosis is majorly caused by an imbalance between osteoclastic and osteogenic niches. Despite the development of nationally recognized first-line anti-osteoporosis drugs, including alendronate (AL), their low bioavailability, poor uptake rate, and dose-related side effects present significant challenges in treatment. This calls for an urgent need for more effective bone-affinity drug delivery systems. In this study, we produced hybrid structures with bioactive components and stable fluffy topological morphology by cross-linking calcium and phosphorus precursors based on mesoporous silica to fabricate nanoadjuvants for AL delivery. The subsequent grafting of -PEG-DAsp8 ensured superior biocompatibility and bone targeting capacity. RNA sequencing revealed that these fluffy nanoadjuvants effectively activated adhesion pathways through CARD11 and CD34 molecular mechanisms, hence promoting cellular uptake and intracellular delivery of AL. Experiments showed that small-dose AL nanoadjuvants effectively suppress osteoclast formation and potentially promote osteogenesis. In vivo results restored the balance between osteogenic and osteoclastic niches against osteoporosis as well as the consequent significant recovery of bone mass. Therefore, this study constructed a drug nanoadjuvant with peculiar topological structures and high bone targeting capacities, efficient intracellular drug delivery as well as bone bioactivity. This provides a novel perspective on drug delivery for osteoporosis and treatment strategies for other bone diseases.
Collapse
Affiliation(s)
- Guoyang Zhang
- Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No. 600 Yishan Road, Shanghai, 200233, China
- Shanghai Jiao Tong University School of Medicine, No. 227 South Chongqing Road, Shanghai, 200025, China
- Regenerative Sports Medicine and Translational Youth Science and Technology Innovation Workroom, Shanghai Jiao Tong University School of Medicine, No. 227 South Chongqing Road, Shanghai, 200025, China
- Regenerative Sports Medicine Lab of the Institute of Microsurgery on Extremities, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No. 600 Yishan Road, Shanghai, 200233, China
| | - Yuhao Kang
- Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No. 600 Yishan Road, Shanghai, 200233, China
- Shanghai Jiao Tong University School of Medicine, No. 227 South Chongqing Road, Shanghai, 200025, China
- Regenerative Sports Medicine and Translational Youth Science and Technology Innovation Workroom, Shanghai Jiao Tong University School of Medicine, No. 227 South Chongqing Road, Shanghai, 200025, China
- Regenerative Sports Medicine Lab of the Institute of Microsurgery on Extremities, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No. 600 Yishan Road, Shanghai, 200233, China
| | - Jizhao Dong
- Multidisciplinary Centre for Advanced Materials, Institute for Frontier Medical Technology, School of Chemistry and Chemical Engineering, Shanghai University of Engineering Science, No.333 Longteng Road, Shanghai, 201620, China
| | - Dingyi Shi
- Shanghai Jiao Tong University School of Medicine, No. 227 South Chongqing Road, Shanghai, 200025, China
- Regenerative Sports Medicine and Translational Youth Science and Technology Innovation Workroom, Shanghai Jiao Tong University School of Medicine, No. 227 South Chongqing Road, Shanghai, 200025, China
| | - Yu Xiang
- Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No. 600 Yishan Road, Shanghai, 200233, China
- Shanghai Jiao Tong University School of Medicine, No. 227 South Chongqing Road, Shanghai, 200025, China
- Regenerative Sports Medicine Lab of the Institute of Microsurgery on Extremities, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No. 600 Yishan Road, Shanghai, 200233, China
| | - Haihan Gao
- Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No. 600 Yishan Road, Shanghai, 200233, China
- Shanghai Jiao Tong University School of Medicine, No. 227 South Chongqing Road, Shanghai, 200025, China
- Regenerative Sports Medicine Lab of the Institute of Microsurgery on Extremities, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No. 600 Yishan Road, Shanghai, 200233, China
| | - Zhiqi Lin
- Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No. 600 Yishan Road, Shanghai, 200233, China
- Shanghai Jiao Tong University School of Medicine, No. 227 South Chongqing Road, Shanghai, 200025, China
- Regenerative Sports Medicine Lab of the Institute of Microsurgery on Extremities, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No. 600 Yishan Road, Shanghai, 200233, China
| | - Xiaojuan Wei
- Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No. 600 Yishan Road, Shanghai, 200233, China
- Regenerative Sports Medicine Lab of the Institute of Microsurgery on Extremities, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No. 600 Yishan Road, Shanghai, 200233, China
| | - Ren Ding
- Shanghai Baoshan District Hospital of Integrated Traditional Chinese and Western Medicine, Department of Orthopedics, No. 181 Youyi Road, Shanghai, 201900, China
| | - Beibei Fan
- Shanghai Baoshan District Hospital of Integrated Traditional Chinese and Western Medicine, Department of Pharmacy, No. 181 Youyi Road, Shanghai, 201900, China
| | - Hongmei Zhang
- Multidisciplinary Centre for Advanced Materials, Institute for Frontier Medical Technology, School of Chemistry and Chemical Engineering, Shanghai University of Engineering Science, No.333 Longteng Road, Shanghai, 201620, China
| | - Tonghe Zhu
- Multidisciplinary Centre for Advanced Materials, Institute for Frontier Medical Technology, School of Chemistry and Chemical Engineering, Shanghai University of Engineering Science, No.333 Longteng Road, Shanghai, 201620, China
| | - Liren Wang
- Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No. 600 Yishan Road, Shanghai, 200233, China
- Shanghai Jiao Tong University School of Medicine, No. 227 South Chongqing Road, Shanghai, 200025, China
- Regenerative Sports Medicine and Translational Youth Science and Technology Innovation Workroom, Shanghai Jiao Tong University School of Medicine, No. 227 South Chongqing Road, Shanghai, 200025, China
- Regenerative Sports Medicine Lab of the Institute of Microsurgery on Extremities, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No. 600 Yishan Road, Shanghai, 200233, China
| | - Xiaoyu Yan
- Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No. 600 Yishan Road, Shanghai, 200233, China
- Shanghai Jiao Tong University School of Medicine, No. 227 South Chongqing Road, Shanghai, 200025, China
- Regenerative Sports Medicine Lab of the Institute of Microsurgery on Extremities, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No. 600 Yishan Road, Shanghai, 200233, China
| |
Collapse
|
14
|
Liu Q, Han M, Wu Z, Fu W, Ji J, Liang Q, Tan M, Zhai L, Gao J, Shi D, Jiang Q, Sun Z, Lai Y, Xu Q, Sun Y. DDX5 inhibits hyaline cartilage fibrosis and degradation in osteoarthritis via alternative splicing and G-quadruplex unwinding. NATURE AGING 2024; 4:664-680. [PMID: 38760576 PMCID: PMC11108786 DOI: 10.1038/s43587-024-00624-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 04/04/2024] [Indexed: 05/19/2024]
Abstract
Hyaline cartilage fibrosis is typically considered an end-stage pathology of osteoarthritis (OA), which results in changes to the extracellular matrix. However, the mechanism behind this is largely unclear. Here, we found that the RNA helicase DDX5 was dramatically downregulated during the progression of OA. DDX5 deficiency increased fibrosis phenotype by upregulating COL1 expression and downregulating COL2 expression. In addition, loss of DDX5 aggravated cartilage degradation by inducing the production of cartilage-degrading enzymes. Chondrocyte-specific deletion of Ddx5 led to more severe cartilage lesions in the mouse OA model. Mechanistically, weakened DDX5 resulted in abundance of the Fn1-AS-WT and Plod2-AS-WT transcripts, which promoted expression of fibrosis-related genes (Col1, Acta2) and extracellular matrix degradation genes (Mmp13, Nos2 and so on), respectively. Additionally, loss of DDX5 prevented the unfolding Col2 promoter G-quadruplex, thereby reducing COL2 production. Together, our data suggest that strategies aimed at the upregulation of DDX5 hold significant potential for the treatment of cartilage fibrosis and degradation in OA.
Collapse
Affiliation(s)
- Qianqian Liu
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, School of Life Sciences, Nanjing University, Nanjing, China
| | - Mingrui Han
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, School of Life Sciences, Nanjing University, Nanjing, China
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China
| | - Zhigui Wu
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Wenqiang Fu
- High Magnetic Field Laboratory, Chinese Academy of Sciences, Hefei, China
| | - Jun Ji
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Qingqing Liang
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Minjia Tan
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Linhui Zhai
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Jian Gao
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, School of Life Sciences, Nanjing University, Nanjing, China
| | - Dongquan Shi
- Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Qing Jiang
- Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Ziying Sun
- Department of Orthopaedics, Jinling Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Yuping Lai
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai, China
| | - Qiang Xu
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, School of Life Sciences, Nanjing University, Nanjing, China.
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China.
| | - Yang Sun
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, School of Life Sciences, Nanjing University, Nanjing, China.
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China.
| |
Collapse
|
15
|
Dai J, Hu Z, Zeng F, Gong X, Tang H, Deng J, Li J, Dong S. Osteoclast-derived exosomal miR-212-3p suppressed the anabolism and accelerated the catabolism of chondrocytes in osteoarthritis by targeting TGF-β1/Smad2 signaling. Arch Biochem Biophys 2024; 751:109827. [PMID: 38000494 DOI: 10.1016/j.abb.2023.109827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 11/10/2023] [Accepted: 11/18/2023] [Indexed: 11/26/2023]
Abstract
Osteoarthritis (OA) is a common aging-related disease affecting entire joint structures, encompassing articular cartilage and subchondral bone. Although senescence and dysfunction of chondrocytes are considered crucial factors in the occurrence of OA, the exact pathogenesis remains to be investigated. In our study, chondrocytes were incubated with a conditioned medium obtained from osteoclasts at different differentiation stages, suggesting that osteoclasts and osteoclast precursors suppressed anabolism and promoted the catabolism of chondrocytes in vitro. In contrast, the function of osteoclasts was more significant than osteoclast precursors. Further blocking of osteoclast exosome secretion by using GW4869 abolished the effect of osteoclasts on chondrocytes. Functionally, exosomal transfer of osteoclast-derived miR-212-3p inhibited Smad2 to mediate chondrocyte dysfunction, thus accelerating cartilage matrix degradation in OA via TGF-β1/Smad2 signaling. The mechanism was also confirmed within the articular cartilage in OA patients and surgery-induced OA mice. Our study provides new information on intercellular interactions in the bone microenvironment within articular cartilage and subchondral bone during OA progression. The miR-212-3p/Smad2 axis is a potential target for the prevention and therapy of OA.
Collapse
Affiliation(s)
- Jingjin Dai
- Department of Biomedical Materials Science, Army Medical University, Chongqing, China
| | - Zhaoyang Hu
- Department of Burns, the 921th Hospital of Joint Logistic Support Force, Changsha, China
| | - Fanchun Zeng
- College of Bioengineering, Chongqing University, Chongqing, China
| | - Xiaoshan Gong
- Department of Biomedical Materials Science, Army Medical University, Chongqing, China
| | - Hao Tang
- Department of Biomedical Materials Science, Army Medical University, Chongqing, China
| | - Jiezhong Deng
- Department of Orthopedics, Southwest Hospital, Army Medical University, Chongqing, China
| | - Jianmei Li
- Department of Biomedical Materials Science, Army Medical University, Chongqing, China
| | - Shiwu Dong
- Department of Biomedical Materials Science, Army Medical University, Chongqing, China; State Key Laboratory of Trauma and Chemical Poisoning, Army Medical University, Chongqing, China.
| |
Collapse
|
16
|
Bakinowska E, Kiełbowski K, Pawlik A. The Role of Extracellular Vesicles in the Pathogenesis and Treatment of Rheumatoid Arthritis and Osteoarthritis. Cells 2023; 12:2716. [PMID: 38067147 PMCID: PMC10706487 DOI: 10.3390/cells12232716] [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: 10/26/2023] [Revised: 11/24/2023] [Accepted: 11/25/2023] [Indexed: 12/18/2023] Open
Abstract
Cells can communicate with each other through extracellular vesicles (EVs), which are membrane-bound structures that transport proteins, lipids and nucleic acids. These structures have been found to mediate cellular differentiation and proliferation apoptosis, as well as inflammatory responses and senescence, among others. The cargo of these vesicles may include immunomodulatory molecules, which can then contribute to the pathogenesis of various diseases. By contrast, EVs secreted by mesenchymal stem cells (MSCs) have shown important immunosuppressive and regenerative properties. Moreover, EVs can be modified and used as drug carriers to precisely deliver therapeutic agents. In this review, we aim to summarize the current evidence on the roles of EVs in the progression and treatment of rheumatoid arthritis (RA) and osteoarthritis (OA), which are important and prevalent joint diseases with a significant global burden.
Collapse
Affiliation(s)
| | | | - Andrzej Pawlik
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland; (E.B.); (K.K.)
| |
Collapse
|
17
|
Wu X, Chen M, Lin S, Chen S, Gu J, Wu Y, Qu M, Gong W, Yao Q, Li H, Zou X, Chen D, Xiao G. Loss of Pinch Proteins Causes Severe Degenerative Disc Disease-Like Lesions in Mice. Aging Dis 2023; 14:1818-1833. [PMID: 37196110 PMCID: PMC10529740 DOI: 10.14336/ad.2023.0212] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Accepted: 02/12/2023] [Indexed: 05/19/2023] Open
Abstract
Degenerative disc disease (DDD) is one of the most common skeletal disorders affecting aged populations. DDD is the leading cause of low back/neck pain, resulting in disability and huge socioeconomic burdens. However, the molecular mechanisms underlying DDD initiation and progression remain poorly understood. Pinch1 and Pinch2 are LIM-domain-containing proteins with crucial functions in mediating multiple fundamental biological processes, such as focal adhesion, cytoskeletal organization, cell proliferation, migration, and survival. In this study, we found that Pinch1 and Pinch2 were both highly expressed in healthy intervertebral discs (IVDs) and dramatically downregulated in degenerative IVDs in mice. Deleting Pinch1 in aggrecan-expressing cells and Pinch2 globally (AggrecanCreERT2; Pinch1fl/fl; Pinch2-/-) caused striking spontaneous DDD-like lesions in lumbar IVDs in mice. Pinch loss inhibited cell proliferation and promoted extracellular matrix (ECM) degradation and apoptosis in lumbar IVDs. Pinch loss markedly enhanced the production of pro-inflammatory cytokines, especially TNFα, in lumbar IVDs and exacerbated instability-induced DDD defects in mice. Pharmacological inhibition of TNFα signaling mitigated the DDD-like lesions caused by Pinch loss. In human degenerative NP samples, reduced expression of Pinch proteins was correlated with severe DDD progression and a markedly upregulated expression of TNFα. Collectively, we demonstrate the crucial role of Pinch proteins in maintaining IVD homeostasis and define a potential therapeutic target for DDD.
Collapse
Affiliation(s)
- Xiaohao Wu
- Department of Biochemistry, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, China.
| | - Mingjue Chen
- Department of Biochemistry, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, China.
| | - Sixiong Lin
- Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Department of Spine Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.
| | - Sheng Chen
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, China.
| | - Jingliang Gu
- Department of Orthopedics, Shanghai municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, China.
| | - Yuchen Wu
- Department of Endocrinology, Chongqing Traditional Chinese Medicine Hospital, Chongqing, China.
| | - Minghao Qu
- Department of Biochemistry, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, China.
| | - Weiyuan Gong
- Department of Biochemistry, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, China.
| | - Qing Yao
- Department of Biochemistry, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, China.
| | - Huiping Li
- Department of Respiratory and Critical Care Medicine, Shenzhen People’s Hospital, Southern University of Science and Technology, Shenzhen, China.
| | - Xuenong Zou
- Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Department of Spine Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.
| | - Di Chen
- Research Center for Human Tissues and Organs Degeneration, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.
| | - Guozhi Xiao
- Department of Biochemistry, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, China.
| |
Collapse
|
18
|
Lu J, Zhang Y, Yang X, Zhao H. Harnessing exosomes as cutting-edge drug delivery systems for revolutionary osteoarthritis therapy. Biomed Pharmacother 2023; 165:115135. [PMID: 37453195 DOI: 10.1016/j.biopha.2023.115135] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 06/30/2023] [Accepted: 07/07/2023] [Indexed: 07/18/2023] Open
Abstract
Exosomes, remarkable extracellular vesicles, have emerged as an advanced frontier in intercellular communication. This remarkable capacity positions them as promising contenders in drug delivery systems (DDSs) for osteoarthritis (OA) therapy, capitalizing on their inherent biocompatibility, stability, and minimal immunogenicity. In this comprehensive review, we summarize the emerging developments surrounding exosome-based DDSs for OA therapy. Focusing on exosome origins, we meticulously explore the diverse sources contributing to their production, including invaluable stem cells, immune cells, and an array of other cell types. In addition, we unravel the underlying mechanisms of action that govern these exosome-borne therapeutics, illuminating the intricate interplay between exosomes and recipient cells. In summary, this review highlights the present challenges that permeate exosome-based DDSs for OA therapy. Through an in-depth exploration of the intricacies within this emerging field, this review aims to shed light on the future direction of exosome-based DDSs in OA. It serves as a bridge for fostering collaboration and collective efforts in reshaping the treatment landscape of OA.
Collapse
Affiliation(s)
- Jun Lu
- Department of Foot and Ankle Surgery, Honghui Hospital of Xi'an Jiaotong University, Xi'an City, Shaanxi Province 710054, China
| | - Yan Zhang
- Department of Foot and Ankle Surgery, Honghui Hospital of Xi'an Jiaotong University, Xi'an City, Shaanxi Province 710054, China
| | - Xinquan Yang
- Department of Foot and Ankle Surgery, Honghui Hospital of Xi'an Jiaotong University, Xi'an City, Shaanxi Province 710054, China
| | - Hongmou Zhao
- Department of Foot and Ankle Surgery, Honghui Hospital of Xi'an Jiaotong University, Xi'an City, Shaanxi Province 710054, China.
| |
Collapse
|
19
|
Xu H, Wang W, Liu X, Huang W, Zhu C, Xu Y, Yang H, Bai J, Geng D. Targeting strategies for bone diseases: signaling pathways and clinical studies. Signal Transduct Target Ther 2023; 8:202. [PMID: 37198232 DOI: 10.1038/s41392-023-01467-8] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Revised: 04/02/2023] [Accepted: 04/19/2023] [Indexed: 05/19/2023] Open
Abstract
Since the proposal of Paul Ehrlich's magic bullet concept over 100 years ago, tremendous advances have occurred in targeted therapy. From the initial selective antibody, antitoxin to targeted drug delivery that emerged in the past decades, more precise therapeutic efficacy is realized in specific pathological sites of clinical diseases. As a highly pyknotic mineralized tissue with lessened blood flow, bone is characterized by a complex remodeling and homeostatic regulation mechanism, which makes drug therapy for skeletal diseases more challenging than other tissues. Bone-targeted therapy has been considered a promising therapeutic approach for handling such drawbacks. With the deepening understanding of bone biology, improvements in some established bone-targeted drugs and novel therapeutic targets for drugs and deliveries have emerged on the horizon. In this review, we provide a panoramic summary of recent advances in therapeutic strategies based on bone targeting. We highlight targeting strategies based on bone structure and remodeling biology. For bone-targeted therapeutic agents, in addition to improvements of the classic denosumab, romosozumab, and PTH1R ligands, potential regulation of the remodeling process targeting other key membrane expressions, cellular crosstalk, and gene expression, of all bone cells has been exploited. For bone-targeted drug delivery, different delivery strategies targeting bone matrix, bone marrow, and specific bone cells are summarized with a comparison between different targeting ligands. Ultimately, this review will summarize recent advances in the clinical translation of bone-targeted therapies and provide a perspective on the challenges for the application of bone-targeted therapy in the clinic and future trends in this area.
Collapse
Affiliation(s)
- Hao Xu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu, 215006, P. R. China
| | - Wentao Wang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu, 215006, P. R. China
| | - Xin Liu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu, 215006, P. R. China
| | - Wei Huang
- Department of Orthopedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230031, Anhui, China
| | - Chen Zhu
- Department of Orthopedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230031, Anhui, China
| | - Yaozeng Xu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu, 215006, P. R. China
| | - Huilin Yang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu, 215006, P. R. China.
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, 215006, Jiangsu, China.
| | - Jiaxiang Bai
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu, 215006, P. R. China.
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, 215006, Jiangsu, China.
| | - Dechun Geng
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu, 215006, P. R. China.
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, 215006, Jiangsu, China.
| |
Collapse
|
20
|
Yao Y, Wei G, Deng L, Cui W. Visualizable and Lubricating Hydrogel Microspheres Via NanoPOSS for Cartilage Regeneration. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2207438. [PMID: 36973540 DOI: 10.1002/advs.202207438] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Revised: 03/01/2023] [Indexed: 05/27/2023]
Abstract
The monitoring of tissue regeneration is particularly important. However, most materials do not allow direct observation of the regeneration process in the cartilage layer. Here, using sulfhydryl polyhedral oligomeric silsesquioxane (POSS-SH) as a nano-construction platform, poly(ethylene glycol) (PEG), Kartogenin (KGN), hydrogenated soya phosphatidylcholine (HSPC), and fluorescein are linked through the "click chemistry" method to construct nanomaterial with fluorescence visualization for cartilage repair: POSS linked with PEG, KGN, HSPC, and fluorescein (PPKHF). PPKHF nanoparticles are encapsulated with hyaluronic acid methacryloyl to prepare PPKHF-loaded microfluidic hyaluronic acid methacrylate spheres (MHS@PPKHF) for in situ injection into the joint cavity using microfluidic technology. MHS@PPKHF forms a buffer lubricant layer in the joint space to reduce friction between articular cartilages, while releasing encapsulated positively charged PPKHF to the deep cartilage through electromagnetic force, facilitating visualization of the location of the drug via fluorescence. Moreover, PPKHF facilitates differentiation of bone marrow mesenchymal stem cells into chondrocytes, which are located in the subchondral bone. In animal experiment, the material accelerates cartilage regeneration while allowing monitoring of cartilage layer repair progression via fluorescence signals. Thus, these POSS-based micro-nano hydrogel microspheres can be used for cartilage regeneration and monitoring and potentially for clinical osteoarthritis therapy.
Collapse
Affiliation(s)
- Yubin Yao
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, P. R. China
- Department of Orthopaedics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325006, P. R. China
| | - Gang Wei
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, P. R. China
| | - Lianfu Deng
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, P. R. China
| | - Wenguo Cui
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, P. R. China
| |
Collapse
|
21
|
Tavasolian F, Inman RD. Biology and therapeutic potential of mesenchymal stem cell extracellular vesicles in axial spondyloarthritis. Commun Biol 2023; 6:413. [PMID: 37059822 PMCID: PMC10104809 DOI: 10.1038/s42003-023-04743-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 03/21/2023] [Indexed: 04/16/2023] Open
Abstract
Axial spondyloarthritis (AxSpA) is a chronic, inflammatory, autoimmune disease that predominantly affects the joints of the spine, causes chronic pain, and, in advanced stages, may result in spinal fusion. Recent developments in understanding the immunomodulatory and tissue-differentiating properties of mesenchymal stem cell (MSC) therapy have raised the possibility of applying such treatment to AxSpA. The therapeutic effectiveness of MSCs has been shown in numerous studies spanning a range of diseases. Several studies have been conducted examining acellular therapy based on MSC secretome. Extracellular vesicles (EVs) generated by MSCs have been proven to reproduce the impact of MSCs on target cells. These EVs are associated with immunological regulation, tissue remodeling, and cellular homeostasis. EVs' biological effects rely on their cargo, with microRNAs (miRNAs) integrated into EVs playing a particularly important role in gene expression regulation. In this article, we will discuss the impact of MSCs and EVs generated by MSCs on target cells and how these may be used as unique treatment strategies for AxSpA.
Collapse
Affiliation(s)
- Fataneh Tavasolian
- Spondylitis Program, Division of Rheumatology, Schroeder Arthritis Institute, University Health Network, Toronto, Ontario, Canada
| | - Robert D Inman
- Spondylitis Program, Division of Rheumatology, Schroeder Arthritis Institute, University Health Network, Toronto, Ontario, Canada.
- Krembil Research Institute, University Health Network, Toronto, Ontario, Canada.
- Departments of Medicine and Immunology, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
22
|
Kalluri R, McAndrews KM. The role of extracellular vesicles in cancer. Cell 2023; 186:1610-1626. [PMID: 37059067 PMCID: PMC10484374 DOI: 10.1016/j.cell.2023.03.010] [Citation(s) in RCA: 250] [Impact Index Per Article: 125.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 02/17/2023] [Accepted: 03/07/2023] [Indexed: 04/16/2023]
Abstract
Intercellular communication is a key feature of cancer progression and metastasis. Extracellular vesicles (EVs) are generated by all cells, including cancer cells, and recent studies have identified EVs as key mediators of cell-cell communication via packaging and transfer of bioactive constituents to impact the biology and function of cancer cells and cells of the tumor microenvironment. Here, we review recent advances in understanding the functional contribution of EVs to cancer progression and metastasis, as cancer biomarkers, and the development of cancer therapeutics.
Collapse
Affiliation(s)
- Raghu Kalluri
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA.
| | - Kathleen M McAndrews
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA.
| |
Collapse
|
23
|
Qu M, Chen M, Gong W, Huo S, Yan Q, Yao Q, Lai Y, Chen D, Wu X, Xiao G. Pip5k1c Loss in Chondrocytes Causes Spontaneous Osteoarthritic Lesions in Aged Mice. Aging Dis 2023; 14:502-514. [PMID: 37008048 PMCID: PMC10017150 DOI: 10.14336/ad.2022.0828] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 08/28/2022] [Indexed: 11/18/2022] Open
Abstract
Osteoarthritis (OA) is the most common degenerative joint disease affecting the older populations globally. Phosphatidylinositol-4-phosphate 5-kinase type-1 gamma (Pip5k1c), a lipid kinase catalyzing the synthesis of phospholipid phosphatidylinositol 4,5-bisphosphate (PIP2), is involved in various cellular processes, such as focal adhesion (FA) formation, cell migration, and cellular signal transduction. However, whether Pip5k1c plays a role in the pathogenesis of OA remains unclear. Here we show that inducible deletion of Pip5k1c in aggrecan-expressing chondrocytes (cKO) causes multiple spontaneous OA-like lesions, including cartilage degradation, surface fissures, subchondral sclerosis, meniscus deformation, synovial hyperplasia, and osteophyte formation in aged (15-month-old) mice, but not in adult (7-month-old) mice. Pip5k1c loss promotes extracellular matrix (ECM) degradation, chondrocyte hypertrophy and apoptosis, and inhibits chondrocyte proliferation in the articular cartilage of aged mice. Pip5k1c loss dramatically downregulates the expressions of several key FA proteins, including activated integrin β1, talin, and vinculin, and thus impairs the chondrocyte adhesion and spreading on ECM. Collectively, these findings suggest that Pip5k1c expression in chondrocytes plays a critical role in maintaining articular cartilage homeostasis and protecting against age-related OA.
Collapse
Affiliation(s)
- Minghao Qu
- Department of Biochemistry, School of Medicine, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, Shenzhen, China.
| | - Mingjue Chen
- Department of Biochemistry, School of Medicine, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, Shenzhen, China.
| | - Weiyuan Gong
- Department of Biochemistry, School of Medicine, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, Shenzhen, China.
| | - Shaochuan Huo
- Department of Biochemistry, School of Medicine, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, Shenzhen, China.
- Shenzhen Hospital (Futian) of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China.
| | - Qinnan Yan
- Department of Biochemistry, School of Medicine, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, Shenzhen, China.
| | - Qing Yao
- Department of Biochemistry, School of Medicine, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, Shenzhen, China.
| | - Yumei Lai
- Department of Orthopedic Surgery, Rush University Medical Center, Chicago, IL 60612, USA.
| | - Di Chen
- Research Center for Human Tissues and Organs Degeneration, Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.
| | - Xiaohao Wu
- Department of Biochemistry, School of Medicine, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, Shenzhen, China.
| | - Guozhi Xiao
- Department of Biochemistry, School of Medicine, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, Shenzhen, China.
| |
Collapse
|
24
|
Song X, Xu L, Zhang W. Biomimetic synthesis and optimization of extracellular vesicles for bone regeneration. J Control Release 2023; 355:18-41. [PMID: 36706840 DOI: 10.1016/j.jconrel.2023.01.057] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 01/18/2023] [Accepted: 01/20/2023] [Indexed: 01/29/2023]
Abstract
Critical-size bone defect repair is in high demand but is difficult to treat. Modern therapies, such as autograft and cell-based treatments, face limitations, including potential immunological rejection and tumorigenesis. Therefore, extracellular vesicle (EV)-based strategies have been proposed as a novel approach for tissue regeneration owing to EVs' complex composition of lipids, proteins, and nucleic acids, as well as their low immunogenicity and congenital cell-targeting features. Despite these remarkable features of EVs, biomimetic synthesis and optimization of natural EVs can lead to enhanced bioactivity, increased cellular uptake, and specific cell targeting, aiming to achieve optimal therapeutic efficacy. To maximize their function, these nanoparticles can be integrated into bone graft biomaterials for superior bone regeneration. Herein, we summarize the role of naturally occurring EVs from distinct cell types in bone regeneration, the current strategies for optimizing biomimetic synthetic EVs in bone regeneration, and discuss the recent advances in applying bone graft biomaterials for the delivery of EVs to bone defect repair. We focused on distinct strategies for optimizing EVs with different functions and the most recent research on achieving time-controlled release of nanoparticles from EV-loaded biomaterials. Furthermore, we thoroughly discuss several current challenges and proposed solutions, aiming to provide insight into current progress, inspiration for future development directions, and incentives for clinical application in this field.
Collapse
Affiliation(s)
- Xinyu Song
- Department of Prosthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China; College of Stomatology, Shanghai Jiao Tong University, Shanghai, China; National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Shanghai, China
| | - Ling Xu
- Department of Prosthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China; College of Stomatology, Shanghai Jiao Tong University, Shanghai, China; National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Shanghai, China.
| | - Wenjie Zhang
- Department of Prosthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China; College of Stomatology, Shanghai Jiao Tong University, Shanghai, China; National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Shanghai, China.
| |
Collapse
|
25
|
Yao Q, Wu X, Tao C, Gong W, Chen M, Qu M, Zhong Y, He T, Chen S, Xiao G. Osteoarthritis: pathogenic signaling pathways and therapeutic targets. Signal Transduct Target Ther 2023; 8:56. [PMID: 36737426 PMCID: PMC9898571 DOI: 10.1038/s41392-023-01330-w] [Citation(s) in RCA: 449] [Impact Index Per Article: 224.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 01/06/2023] [Accepted: 01/17/2023] [Indexed: 02/05/2023] Open
Abstract
Osteoarthritis (OA) is a chronic degenerative joint disorder that leads to disability and affects more than 500 million population worldwide. OA was believed to be caused by the wearing and tearing of articular cartilage, but it is now more commonly referred to as a chronic whole-joint disorder that is initiated with biochemical and cellular alterations in the synovial joint tissues, which leads to the histological and structural changes of the joint and ends up with the whole tissue dysfunction. Currently, there is no cure for OA, partly due to a lack of comprehensive understanding of the pathological mechanism of the initiation and progression of the disease. Therefore, a better understanding of pathological signaling pathways and key molecules involved in OA pathogenesis is crucial for therapeutic target design and drug development. In this review, we first summarize the epidemiology of OA, including its prevalence, incidence and burdens, and OA risk factors. We then focus on the roles and regulation of the pathological signaling pathways, such as Wnt/β-catenin, NF-κB, focal adhesion, HIFs, TGFβ/ΒΜP and FGF signaling pathways, and key regulators AMPK, mTOR, and RUNX2 in the onset and development of OA. In addition, the roles of factors associated with OA, including MMPs, ADAMTS/ADAMs, and PRG4, are discussed in detail. Finally, we provide updates on the current clinical therapies and clinical trials of biological treatments and drugs for OA. Research advances in basic knowledge of articular cartilage biology and OA pathogenesis will have a significant impact and translational value in developing OA therapeutic strategies.
Collapse
Affiliation(s)
- Qing Yao
- Department of Biochemistry, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, 518055, China.
| | - Xiaohao Wu
- Department of Biochemistry, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Chu Tao
- Department of Biochemistry, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Weiyuan Gong
- Department of Biochemistry, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Mingjue Chen
- Department of Biochemistry, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Minghao Qu
- Department of Biochemistry, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Yiming Zhong
- Department of Biochemistry, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Tailin He
- Department of Biochemistry, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Sheng Chen
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Guozhi Xiao
- Department of Biochemistry, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, 518055, China.
| |
Collapse
|
26
|
Wu X, Lin S, Liao R, Yao Q, Lin L, Zou X, Xiao G. Brief research report: Effects of Pinch deficiency on cartilage homeostasis in adult mice. Front Cell Dev Biol 2023; 11:1116128. [PMID: 36743414 PMCID: PMC9892552 DOI: 10.3389/fcell.2023.1116128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 01/11/2023] [Indexed: 01/20/2023] Open
Abstract
Pinch1 and Pinch2 are LIM domain-containing proteins with crucial functions in mediating focal adhesion formation. Our previous studies have demonstrated that Pinch1/2 expression is essential for cartilage and bone formation during skeletal development in mice. Loss of Pinch expression (Prx1Cre; Pinch1flox/flox; Pinch2-/-) inhibits chondrocyte proliferation and promotes chondrocyte apoptosis, resulting in severe chondrodysplasia and limb shortening. Based on these observations, we wonder if Pinch proteins have a role in adult cartilage and whether Pinch deficiency will compromise cartilage homeostasis and promote osteoarthritis (OA)-related defects in adult mice. To this end, we generated the AggrecanCreERT2; Pinch1flox/flox; Pinch2-/- mice, in which the Pinch1 gene can be inducibly deleted in aggrecan-expressing chondrocytes by tamoxifen and the Pinch2 gene is globally inactivated. Immunofluorescent staining confirmed that the expression of Pinch proteins was significantly decreased in articular cartilage in tamoxifen-treated adult AggrecanCreERT2; Pinch1flox/flox; Pinch2-/- mice. Unexpectedly, our results showed that Pinch loss did not induce marked abnormalities in articular cartilage and other joint tissues in the knee joints of either adult (10-month-old) mice or aged (17-month-old) mice. In a destabilization of the medial meniscus (DMM)-induced OA model, the surgically-induced OA lesions were comparable between Pinch-deficient mice and control mice. Given the fact that Pinch proteins are essential for chondrogenesis and cartilage formation during skeletal development, these findings suggest that Pinch expression is seemingly not indispensable for adult cartilage homeostasis in mice.
Collapse
Affiliation(s)
- Xiaohao Wu
- Department of Biochemistry, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Sixiong Lin
- Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Department of Spine Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Rongdong Liao
- Department of Orthopaedics, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Qing Yao
- Department of Biochemistry, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Lijun Lin
- Department of Orthopaedics, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xuenong Zou
- Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Department of Spine Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Guozhi Xiao
- Department of Biochemistry, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| |
Collapse
|
27
|
Cheung KCP, Jiao M, Xingxuan C, Wei J. Extracellular vesicles derived from host and gut microbiota as promising nanocarriers for targeted therapy in osteoporosis and osteoarthritis. Front Pharmacol 2023; 13:1051134. [PMID: 36686680 PMCID: PMC9859449 DOI: 10.3389/fphar.2022.1051134] [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: 09/22/2022] [Accepted: 12/21/2022] [Indexed: 01/08/2023] Open
Abstract
Osteoporosis (OP), a systemic bone disease that causes structural bone loss and bone mass loss, is often associated with fragility fractures. Extracellular vesicles (EVs) generated by mammalian and gut bacteria have recently been identified as important mediators in the intercellular signaling pathway that may play a crucial role in microbiota-host communication. EVs are tiny membrane-bound vesicles, which range in size from 20 to 400 nm. They carry a variety of biologically active substances across intra- and intercellular space. These EVs have developed as a promising research area for the treatment of OP because of their nanosized architecture, enhanced biocompatibility, reduced toxicity, drug loading capacity, ease of customization, and industrialization. This review describes the latest development of EVs derived from mammals and bacteria, including their internalization, isolation, biogenesis, classifications, topologies, and compositions. Additionally, breakthroughs in chemical sciences and the distinctive biological features of bacterial extracellular vesicles (BEVs) allow for the customization of modified BEVs for the therapy of OP. In conclusion, we give a thorough and in-depth summary of the main difficulties and potential future of EVs in the treatment of OP, as well as highlight innovative uses and choices for the treatment of osteoarthritis (OA).
Collapse
Affiliation(s)
- Kenneth Chat Pan Cheung
- Hong Kong Traditional Chinese Medicine Phenome Research Center, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Ma Jiao
- Hong Kong Traditional Chinese Medicine Phenome Research Center, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Chen Xingxuan
- Hong Kong Traditional Chinese Medicine Phenome Research Center, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Jia Wei
- Hong Kong Traditional Chinese Medicine Phenome Research Center, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
- Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| |
Collapse
|
28
|
Dilley JE, Bello MA, Roman N, McKinley T, Sankar U. Post-traumatic osteoarthritis: A review of pathogenic mechanisms and novel targets for mitigation. Bone Rep 2023. [DOI: 10.1016/j.bonr.2023.101658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
|
29
|
Kern F, Kuhn T, Ludwig N, Simon M, Gröger L, Fabis N, Aparicio-Puerta E, Salhab A, Fehlmann T, Hahn O, Engel A, Wagner V, Koch M, Winek K, Soreq H, Nazarenko I, Fuhrmann G, Wyss-Coray T, Meese E, Keller V, Laschke MW, Keller A. Ageing-associated small RNA cargo of extracellular vesicles. RNA Biol 2023; 20:482-494. [PMID: 37498213 PMCID: PMC10376918 DOI: 10.1080/15476286.2023.2234713] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 06/06/2023] [Accepted: 06/09/2023] [Indexed: 07/28/2023] Open
Abstract
Previous work on murine models and humans demonstrated global as well as tissue-specific molecular ageing trajectories of RNAs. Extracellular vesicles (EVs) are membrane vesicles mediating the horizontal transfer of genetic information between different tissues. We sequenced small regulatory RNAs (sncRNAs) in two mouse plasma fractions at five time points across the lifespan from 2-18 months: (1) sncRNAs that are free-circulating (fc-RNA) and (2) sncRNAs bound outside or inside EVs (EV-RNA). Different sncRNA classes exhibit unique ageing patterns that vary between the fcRNA and EV-RNA fractions. While tRNAs showed the highest correlation with ageing in both fractions, rRNAs exhibited inverse correlation trajectories between the EV- and fc-fractions. For miRNAs, the EV-RNA fraction was exceptionally strongly associated with ageing, especially the miR-29 family in adipose tissues. Sequencing of sncRNAs and coding genes in fat tissue of an independent cohort of aged mice up to 27 months highlighted the pivotal role of miR-29a-3p and miR-29b-3p in ageing-related gene regulation that we validated in a third cohort by RT-qPCR.
Collapse
Affiliation(s)
- Fabian Kern
- Chair for Clinical Bioinformatics, Saarland Informatics Campus, Saarland University, Saarbrücken, Germany
- Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz-Centre for Infection Research (HZI), Department for Clinical Bioinformatics, Saarbrücken, Germany
| | - Thomas Kuhn
- Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz-Centre for Infection Research (HZI), Biogenic Nanotherapeutics Group (BION), Saarbrücken, Germany
- Department of Pharmacy, Saarland University, Saarbrücken, Germany
| | - Nicole Ludwig
- Department of Human Genetics, Saarland University, Homburg, Germany
- Center for Human and Molecular Biology, Saarland University, Homburg, Germany
| | - Martin Simon
- Molecular Cell Biology and Microbiology, Wuppertal University, Wuppertal, Germany
| | - Laura Gröger
- Department of Human Genetics, Saarland University, Homburg, Germany
| | - Natalie Fabis
- Molecular Cell Biology and Microbiology, Wuppertal University, Wuppertal, Germany
| | - Ernesto Aparicio-Puerta
- Chair for Clinical Bioinformatics, Saarland Informatics Campus, Saarland University, Saarbrücken, Germany
| | - Abdulrahman Salhab
- Department of Genetics and Epigenetics, Saarland University, Saarbrücken, Germany
| | - Tobias Fehlmann
- Chair for Clinical Bioinformatics, Saarland Informatics Campus, Saarland University, Saarbrücken, Germany
| | - Oliver Hahn
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, USA
| | - Annika Engel
- Chair for Clinical Bioinformatics, Saarland Informatics Campus, Saarland University, Saarbrücken, Germany
| | - Viktoria Wagner
- Chair for Clinical Bioinformatics, Saarland Informatics Campus, Saarland University, Saarbrücken, Germany
| | - Marcus Koch
- INM – Leibniz Institute for New Materials, Saarbrücken, Germany
| | - Katarzyna Winek
- The Edmond and Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Hermona Soreq
- The Edmond and Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Irina Nazarenko
- Faculty of Medicine, Institute for Infection Prevention and Control; Medical Center - University of Freiburg, Freiburg, Germany
| | - Gregor Fuhrmann
- Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz-Centre for Infection Research (HZI), Biogenic Nanotherapeutics Group (BION), Saarbrücken, Germany
- Department of Pharmacy, Saarland University, Saarbrücken, Germany
| | - Tony Wyss-Coray
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, USA
| | - Eckart Meese
- Department of Human Genetics, Saarland University, Homburg, Germany
| | - Verena Keller
- Chair for Clinical Bioinformatics, Saarland Informatics Campus, Saarland University, Saarbrücken, Germany
| | - Matthias W. Laschke
- Institute for Clinical and Experimental Surgery, Saarland University, Homburg, Germany
| | - Andreas Keller
- Chair for Clinical Bioinformatics, Saarland Informatics Campus, Saarland University, Saarbrücken, Germany
- Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz-Centre for Infection Research (HZI), Department for Clinical Bioinformatics, Saarbrücken, Germany
- Center for Bioinformatics, Saarland University, Saarbrücken, Germany
| |
Collapse
|
30
|
Gu J, Rao W, Huo S, Fan T, Qiu M, Zhu H, Chen D, Sheng X. MicroRNAs and long non-coding RNAs in cartilage homeostasis and osteoarthritis. Front Cell Dev Biol 2022; 10:1092776. [PMID: 36582467 PMCID: PMC9793335 DOI: 10.3389/fcell.2022.1092776] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 11/29/2022] [Indexed: 12/14/2022] Open
Abstract
During the last decade, osteoarthritis (OA) has become one of the most prevalent musculoskeletal diseases worldwide. OA is characterized by progressive loss of articular cartilage, abnormal remodeling of subchondral bone, hyperplasia of synovial cells, and growth of osteophytes, which lead to chronic pain and disability. The pathological mechanisms underlying OA initiation and progression are still poorly understood. Non-coding RNAs (ncRNAs) constitute a large portion of the transcriptome that do not encode proteins but function in numerous biological processes. Cumulating evidence has revealed a strong association between the changes in expression levels of ncRNA and the disease progression of OA. Moreover, loss- and gain-of-function studies utilizing transgenic animal models have demonstrated that ncRNAs exert vital functions in regulating cartilage homeostasis, degeneration, and regeneration, and changes in ncRNA expression can promote or decelerate the progression of OA through distinct molecular mechanisms. Recent studies highlighted the potential of ncRNAs to serve as diagnostic biomarkers, prognostic indicators, and therapeutic targets for OA. MiRNAs and lncRNAs are two major classes of ncRNAs that have been the most widely studied in cartilage tissues. In this review, we focused on miRNAs and lncRNAs and provided a comprehensive understanding of their functional roles as well as molecular mechanisms in cartilage homeostasis and OA pathogenesis.
Collapse
Affiliation(s)
- Jingliang Gu
- Department of Orthopedics, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wu Rao
- Department of Orthopedics, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Shaochuan Huo
- Shenzhen Hospital of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Tianyou Fan
- Department of Orthopedics, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Minlei Qiu
- Department of Orthopedics, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Haixia Zhu
- Department of Orthopedics, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Deta Chen
- Department of Orthopedics, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiaoping Sheng
- Department of Orthopedics, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
31
|
Hirose K, Kuwahara M, Nakata E, Tetsunaga T, Yamada K, Saiga K, Takigawa M, Ozaki T, Kubota S, Hattori T. Elevated Expression of CCN3 in Articular Cartilage Induces Osteoarthritis in Hip Joints Irrespective of Age and Weight Bearing. Int J Mol Sci 2022; 23:15311. [PMID: 36499638 PMCID: PMC9738275 DOI: 10.3390/ijms232315311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 11/29/2022] [Accepted: 12/02/2022] [Indexed: 12/11/2022] Open
Abstract
Osteoarthritis (OA) occurs not only in the knee but also in peripheral joints throughout the whole body. Previously, we have shown that the expression of cellular communication network factor 3 (CCN3), a matricellular protein, increases with age in knee articular cartilage, and the misexpression of CCN3 in cartilage induces senescence-associated secretory phenotype (SASP) factors, indicating that CCN3 promotes cartilage senescence. Here, we investigated the correlation between CCN3 expression and OA degenerative changes, principally in human femoral head cartilage. Human femoral heads obtained from patients who received total hip arthroplasty were categorized into OA and femoral neck fracture (normal) groups without significant age differences. Gene expression analysis of RNA obtained from femoral head cartilage revealed that CCN3 and MMP-13 expression in the non-weight-bearing part was significantly higher in the OA group than in the normal group, whereas the weight-bearing OA parts and normal cartilage showed no significant differences in the expression of these genes. The expression of COL10A1, however, was significantly higher in weight-bearing OA parts compared with normal weight-bearing parts, and was also higher in weight-bearing parts compared with non-weight-bearing parts in the OA group. In contrast, OA primary chondrocytes from weight-bearing parts showed higher expression of CCN3, p16, ADAMTS4, and IL-1β than chondrocytes from the corresponding normal group, and higher ADAMTS4 and IL-1β in the non-weight-bearing part compared with the corresponding normal group. Acan expression was significantly lower in the non-weight-bearing group in OA primary chondrocytes than in the corresponding normal chondrocytes. The expression level of CCN3 did not show significant differences between the weight-bearing part and non-weight-bearing part in both OA and normal primary chondrocytes. Immunohistochemical analysis showed accumulated CCN3 and aggrecan neoepitope staining in both the weight-bearing part and non-weight-bearing part in the OA group compared with the normal group. The CCN3 expression level in cartilage had a positive correlation with the Mankin score. X-ray analysis of cartilage-specific CCN3 overexpression mice (Tg) revealed deformation of the femoral and humeral head in the early stage, and immunohistochemical analysis showed accumulated aggrecan neoepitope staining as well as CCN3 staining and the roughening of the joint surface in Tg femoral and humeral heads. Primary chondrocytes from the Tg femoral head showed enhanced expression of Ccn3, Adamts5, p16, Il-6, and Tnfα, and decreased expression of Col2a1 and -an. These findings indicate a correlation between OA degenerative changes and the expression of CCN3, irrespective of age and mechanical loading. Furthermore, the Mankin score indicates that the expression level of Ccn3 correlates with the progression of OA.
Collapse
Affiliation(s)
- Kazuki Hirose
- Department of Biochemistry and Molecular Dentistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
- Department of Orthopaedic Surgery, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, Japan
| | - Miho Kuwahara
- Department of Biochemistry and Molecular Dentistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Eiji Nakata
- Department of Orthopaedic Surgery, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, Japan
| | - Tomonori Tetsunaga
- Department of Orthopaedic Surgery, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, Japan
| | - Kazuki Yamada
- Department of Orthopaedic Surgery, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, Japan
| | - Kenta Saiga
- Department of Orthopaedic Surgery, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, Japan
| | - Masaharu Takigawa
- Advanced Research Center for Oral and Craniofacial Sciences, Okayama University Dental School/Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Toshifumi Ozaki
- Department of Orthopaedic Surgery, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, Japan
| | - Satoshi Kubota
- Department of Biochemistry and Molecular Dentistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Takako Hattori
- Department of Biochemistry and Molecular Dentistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| |
Collapse
|
32
|
Han S. Osteoarthritis year in review 2022: biology. Osteoarthritis Cartilage 2022; 30:1575-1582. [PMID: 36150676 DOI: 10.1016/j.joca.2022.09.003] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Revised: 09/07/2022] [Accepted: 09/08/2022] [Indexed: 02/02/2023]
Abstract
The field of osteoarthritis (OA) biology is rapidly evolving and brilliant progress has been made this year as well. Landmark studies of OA biology published in 2021 and early 2022 were selected through PubMed search by personal opinion. These papers were classified by their molecular mechanisms, and it was largely divided into the intracellular signaling mechanisms and the inter-compartment interaction in chondrocyte homeostasis and OA progression. The intracellular signaling mechanisms involving OA progression included (1) Piezo1/transient receptor potential channels of the vanilloid subtype (TRPV) 4-mediated calcium signaling, (2) mechanical load-F-box and WD repeat domain containing 7 (FBXW7) in chondrocyte senescence, (3) mechanical loading-primary cilia-hedgehog signaling, (4) low grade inflammation by toll-like receptor (TLR)-CD14-lipopolysaccharide-binding protein (LBP) complex and inhibitor of NF-κB kinase (IKK) β-nuclear factor kappa B (NF-κB) signaling, (5) selenium pathway and reactive oxygen species (ROS) production, (6) G protein-coupled receptor (GPCR) and cyclic adenosine monophosphate (cAMP) signaling, (7) peroxisome proliferator-activated receptor α (PPARα)-acyl-CoA thioesterase 12 (ACOT12)-mediated de novo lipogenesis and (8) hypoxia-disruptor of telomeric silencing 1-like (DOT1L)-H3-lysine 79 (H3K79) methylation pathway. The studies on inter-compartment or intercellular interaction in OA progression included the following subjects; (1) the anabolic role of lubricin, glycoprotein from superficial zone cells, (2) osteoclast-chondrocyte interaction via exosomal miRNA and sphingosine 1-phosphate (S1P), (3) senescent fibroblast-like synoviocyte and chondrocyte interaction, (4) synovial macrophage and chondrocyte interaction through Flightless I, (5) αV integrin-mediated transforming growth factor beta (TGFβ) activation by mechanical loading, and (6) osteocytic TGFβ in subchondral bone thickening. Despite the disastrous Covid-19 pandemic, many outstanding studies have expanded the boundary of OA biology. They provide both critical insight into the pathophysiology as well as clues for the treatment of OA.
Collapse
Affiliation(s)
- S Han
- Laboratory for for Arthritis and Cartilage Biology, Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, Republic of Korea; Department of Internal Medicine, School of Medicine, Kyungpook National University, Daegu, Republic of Korea.
| |
Collapse
|
33
|
Liu Q, Huang J, Xia J, Liang Y, Li G. Tracking tools of extracellular vesicles for biomedical research. Front Bioeng Biotechnol 2022; 10:943712. [PMID: 36466335 PMCID: PMC9716315 DOI: 10.3389/fbioe.2022.943712] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Accepted: 11/03/2022] [Indexed: 08/02/2023] Open
Abstract
Imaging of extracellular vesicles (EVs) will facilitate a better understanding of their biological functions and their potential as therapeutics and drug delivery vehicles. In order to clarify EV-mediated cellular communication in vitro and to track the bio-distribution of EV in vivo, various strategies have been developed to label and image EVs. In this review, we summarized recent advances in the tracking of EVs, demonstrating the methods for labeling and imaging of EVs, in which the labeling methods include direct and indirect labeling and the imaging modalities include fluorescent imaging, bioluminescent imaging, nuclear imaging, and nanoparticle-assisted imaging. These techniques help us better understand the mechanism of uptake, the bio-distribution, and the function of EVs. More importantly, we can evaluate the pharmacokinetic properties of EVs, which will help promote their further clinical application.
Collapse
Affiliation(s)
- Qisong Liu
- Shenzhen Key Laboratory of Musculoskeletal Tissue Reconstruction and Function Restoration, Department of Orthopaedic Surgery, Shenzhen People’s Hospital (The Second Clinical Medical College of Jinan University), Shenzhen, China
| | - Jianghong Huang
- Department of Orthopedics, Shenzhen Second People’s Hospital (First Affiliated Hospital of Shenzhen University, Health Science Center), Shenzhen, China
- Tsinghua University Shenzhen International Graduate School, Shenzhen, China
| | - Jiang Xia
- Department of Chemistry, The Chinese University of Hong Kong, Hong Kong, China
| | - Yujie Liang
- Department of Child and Adolescent Psychiatry, Shenzhen Kangning Hospital, Shenzhen Mental Health Center, Shenzhen, China
- Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, China
| | - Guangheng Li
- Shenzhen Key Laboratory of Musculoskeletal Tissue Reconstruction and Function Restoration, Department of Orthopaedic Surgery, Shenzhen People’s Hospital (The Second Clinical Medical College of Jinan University), Shenzhen, China
| |
Collapse
|
34
|
Wu Y, Li J, Zeng Y, Pu W, Mu X, Sun K, Peng Y, Shen B. Exosomes rewire the cartilage microenvironment in osteoarthritis: from intercellular communication to therapeutic strategies. Int J Oral Sci 2022; 14:40. [PMID: 35927232 PMCID: PMC9352673 DOI: 10.1038/s41368-022-00187-z] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 06/02/2022] [Accepted: 06/14/2022] [Indexed: 02/08/2023] Open
Abstract
Osteoarthritis (OA) is a prevalent degenerative joint disease characterized by cartilage loss and accounts for a major source of pain and disability worldwide. However, effective strategies for cartilage repair are lacking, and patients with advanced OA usually need joint replacement. Better comprehending OA pathogenesis may lead to transformative therapeutics. Recently studies have reported that exosomes act as a new means of cell-to-cell communication by delivering multiple bioactive molecules to create a particular microenvironment that tunes cartilage behavior. Specifically, exosome cargos, such as noncoding RNAs (ncRNAs) and proteins, play a crucial role in OA progression by regulating the proliferation, apoptosis, autophagy, and inflammatory response of joint cells, rendering them promising candidates for OA monitoring and treatment. This review systematically summarizes the current insight regarding the biogenesis and function of exosomes and their potential as therapeutic tools targeting cell-to-cell communication in OA, suggesting new realms to improve OA management.
Collapse
Affiliation(s)
- Yuangang Wu
- Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, China
| | - Jiao Li
- Laboratory of Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yi Zeng
- Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, China
| | - Wenchen Pu
- Laboratory of Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaoyu Mu
- Laboratory of Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Kaibo Sun
- Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, China
| | - Yong Peng
- Laboratory of Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China.
| | - Bin Shen
- Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
35
|
Dang L, Li N, Wu X, Li D, Zhang Z, Zhang BT, Lyu A, Chen L, Zhang G, Liu J. A Rapid Protocol for Direct Isolation of Osteoclast Lineage Cells from Mouse Bone Marrow. Bio Protoc 2022; 12:e4338. [PMID: 35592608 PMCID: PMC8918216 DOI: 10.21769/bioprotoc.4338] [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: 01/03/2022] [Revised: 09/30/2021] [Accepted: 01/12/2022] [Indexed: 12/29/2022] Open
Abstract
Osteoclast lineage cells (OLCs), including osteoclast precursors (OCPs) and mature osteoclasts (MOCs), participate in bone remodeling and mediate pathologic bone loss. Thus, it is essential to obtain OLCs for exploring their molecular features in both physiological and pathological conditions in vivo. However, the conventional protocols for obtaining OLCs ex vivo are not only time-consuming, but also unable to capture the cellular status of OLCs in vivo. In addition, the current antibody-based isolation approaches, such as fluorescence-/ magnetic-activated cell sorting, are not able to obtain pure osteoclasts because no unique surface antigen for osteoclasts has been identified. Here, we develop a rapid protocol for directly isolating OLCs from mouse bone marrow through magnetic-activated cell sorting (MACS). This protocol can rapidly enrich OCPs and MOCs, respectively, depending on the expression of the distinctive surface markers at their differentiation stages. It is optimized to isolate OLCs from four mice concurrently, of which sorting procedure could be completed within ~5 h.
Collapse
Affiliation(s)
- Lei Dang
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Nanxi Li
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Xiaohao Wu
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Dijie Li
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Zongkang Zhang
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Bao-Ting Zhang
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Aiping Lyu
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
- Institute of Integrated Bioinfomedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lin Chen
- Laboratory of Wound Repair and Rehabilitation, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Ge Zhang
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
- Institute of Integrated Bioinfomedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Jin Liu
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
36
|
Li M, Yin H, Yan Z, Li H, Wu J, Wang Y, Wei F, Tian G, Ning C, Li H, Gao C, Fu L, Jiang S, Chen M, Sui X, Liu S, Chen Z, Guo Q. The immune microenvironment in cartilage injury and repair. Acta Biomater 2022; 140:23-42. [PMID: 34896634 DOI: 10.1016/j.actbio.2021.12.006] [Citation(s) in RCA: 172] [Impact Index Per Article: 57.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Revised: 12/01/2021] [Accepted: 12/05/2021] [Indexed: 02/07/2023]
Abstract
The ability of articular cartilage to repair itself is limited because it lacks blood vessels, nerves, and lymph tissue. Once damaged, it can lead to joint swelling and pain, accelerating the progression of osteoarthritis. To date, complete regeneration of hyaline cartilage exhibiting mechanical properties remains an elusive goal, despite the many available technologies. The inflammatory milieu created by cartilage damage is critical for chondrocyte death and hypertrophy, extracellular matrix breakdown, ectopic bone formation, and progression of cartilage injury to osteoarthritis. In the inflammatory microenvironment, mesenchymal stem cells (MSCs) undergo aberrant differentiation, and chondrocytes begin to convert or dedifferentiate into cells with a fibroblast phenotype, thereby resulting in fibrocartilage with poor mechanical qualities. All these factors suggest that inflammatory problems may be a major stumbling block to cartilage repair. To produce a milieu conducive to cartilage repair, multi-dimensional management of the joint inflammatory microenvironment in place and time is required. Therefore, this calls for elucidation of the immune microenvironment of cartilage repair after injury. This review provides a brief overview of: (1) the pathogenesis of cartilage injury; (2) immune cells in cartilage injury and repair; (3) effects of inflammatory cytokines on cartilage repair; (4) clinical strategies for treating cartilage defects; and (5) strategies for targeted immunoregulation in cartilage repair. STATEMENT OF SIGNIFICANCE: Immune response is increasingly considered the key factor affecting cartilage repair. It has both negative and positive regulatory effects on the process of regeneration and repair. Proinflammatory factors are secreted in large numbers, and necrotic cartilage is removed. During the repair period, immune cells can secrete anti-inflammatory factors and chondrogenic cytokines, which can inhibit inflammation and promote cartilage repair. However, inflammatory factors persist, which accelerate the degradation of the cartilage matrix. Furthermore, in an inflammatory microenvironment, MSCs undergo abnormal differentiation, and chondrocytes begin to transform or dedifferentiate into fibroblast-like cells, forming fibrocartilage with poor mechanical properties. Consequently, cartilage regeneration requires multi-dimensional regulation of the joint inflammatory microenvironment in space and time to make it conducive to cartilage regeneration.
Collapse
|
37
|
Xiaomin W, Bian B, Lin Z, Wu C, Sun Y, Pan Y, Dai Y, Lui TH, Zhuang T, Pan X. Identification of exosomal mRNA, lncRNA and circRNA signatures in an osteoarthritis synovial fluid-exosomal study. Exp Cell Res 2021; 410:112881. [PMID: 34780784 DOI: 10.1016/j.yexcr.2021.112881] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 10/04/2021] [Accepted: 10/11/2021] [Indexed: 12/18/2022]
Abstract
AIMS Osteoarthritis (OA) is a common degenerative disease that is pathologically characterized by destruction of the joint matrix and reduction of articular chondrocytes, resulting in joint deformity and motor dysfunction. However, the molecular mechanisms governing this pathology have not been elucidated to date. METHODS In this study, we determined the expression levels of lncRNAs, circRNAs, and mRNAs extracted from synovial exosomes of OA and control patients. A network of circRNA/lncRNA-miRNA-mRNA interactions was established using MiRanda and TargetScan software to explore OA pathogenesis. The exosomal lncRNA, circRNA and mRNA expression profiles of the OA and control groups were analysed using LC human competing endogenous RNA (ceRNA) microarrays. The differentially expressed genes were analysed to determine their potential roles in the pathogenesis of OA by bioinformatic analysis. RESULTS There were 52 mRNAs, 196 lncRNAs and 98 circRNAs differentially expressed in synovial exosomes between osteoarthritis synovial and the control group. The final ceRNA network of lncRNAs and circRNAs exhibited a complex interaction between ncRNA and mRNA related to OA pathological mechanisms. An intersection analysis of the ceRNA network showed that 22 miRNAs, 45 lncRNAs, and 34 circRNAs enriched in the PI3K/Akt and autophagy pathways correlated with 7 mRNAs and may play important roles in OA pathological mechanisms. CONCLUSION Our work analysed mRNA/lncRNA/circRNA expression and displayed the ceRNA network of lncRNAs and circRNAs to profile the pathogenesis of OA in synovial exosomes. The results of this study may help to elucidate the pathogenesis of OA and may provide important references for further research attempting to identify more effective targets for the diagnosis and therapy of OA.
Collapse
|
38
|
Meulenbelt I, Ramos YFM, Baglio SR, Pegtel DM. Censoring exosomal crosstalk in osteoarthritis. NATURE AGING 2021; 1:332-334. [PMID: 37117594 DOI: 10.1038/s43587-021-00052-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2023]
Affiliation(s)
- Ingrid Meulenbelt
- Department of Biomedical Data Sciences, Section Molecular Epidemiology, Leiden University Medical Center, Leiden, the Netherlands.
| | - Yolande F M Ramos
- Department of Biomedical Data Sciences, Section Molecular Epidemiology, Leiden University Medical Center, Leiden, the Netherlands
| | - S Rubina Baglio
- Department of Pathology, Cancer Center Amsterdam, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - D Michiel Pegtel
- Department of Pathology, Cancer Center Amsterdam, Amsterdam University Medical Centers, Amsterdam, the Netherlands.
| |
Collapse
|