1
|
Liu X, Liu P, Li H, Cen Y, Jiang G, Zhang W, Tian K, Wang X. Application of kartogenin for the treatment of cartilage defects: current practice and future directions. RSC Adv 2024; 14:33206-33222. [PMID: 39434994 PMCID: PMC11492430 DOI: 10.1039/d4ra06558a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 10/14/2024] [Indexed: 10/23/2024] Open
Abstract
Osteoarthritis and sports injuries often lead to cartilage defects. How to promote its repair and rebuild the smooth cartilage surface has been a hot spot of research in recent years. Kartogenin (KGN), a small molecule discovered in recent years, has been shown to promote the proliferation and chondrogenic differentiation of mesenchymal stem cells (MSCs). As more and more studies have been conducted on KGN, its mechanism of action has been gradually revealed. However, KGN is insoluble in water and therefore easily removed by body fluids. In order to address such issues, a number of systems for efficient intra-articular delivery of KGN have been developed. In addition, due to the complex pathology of cartilage repair, KGN is often used in combination with other drugs to target different stages. In addition, with the rapid development of tissue engineering, scholars have combined KGN with various scaffolds by physical or chemical methods. In this paper, we firstly introduce the general properties of KGN followed by a review of the latest advances in the intra-articular delivery modes of KGN. Finally, we discuss the prospects for the application of KGN in cartilage regeneration, which is aimed at providing a new idea and target for the treatment of cartilage defects.
Collapse
Affiliation(s)
- Xuemiao Liu
- First Affiliated Hospital of Dalian Medical University Dalian 116001 China
- Beijing National Laboratory for Molecular Sciences State Key Laboratory of Polymer Physics and Chemistry Institute of Chemistry Chinese Academy of Sciences Beijing 100190 China
| | - Pengfei Liu
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University Beijing 100191 China
| | - Han Li
- Xiongan Xuanwu Hospital Hebei 071700 China
| | - Ying Cen
- First Affiliated Hospital of Dalian Medical University Dalian 116001 China
| | - Guichun Jiang
- Liaoning Cancer Hospital & Institute, Clinical Skills Training Center Shenyang 110042 China
| | - Weiguo Zhang
- First Affiliated Hospital of Dalian Medical University Dalian 116001 China
| | - Kang Tian
- First Affiliated Hospital of Dalian Medical University Dalian 116001 China
| | - Xing Wang
- Beijing National Laboratory for Molecular Sciences State Key Laboratory of Polymer Physics and Chemistry Institute of Chemistry Chinese Academy of Sciences Beijing 100190 China
| |
Collapse
|
2
|
Zhang H, Yang Y, Gao M, Peng J, Li D, Zhu J. Bibliometric analysis of chondrocyte apoptosis in knee osteoarthritis. Medicine (Baltimore) 2024; 103:e40000. [PMID: 39465698 PMCID: PMC11460941 DOI: 10.1097/md.0000000000040000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Indexed: 10/29/2024] Open
Abstract
BACKGROUND Apoptosis, a form of programmed cell death, plays a significant role in osteoarthritis; however, bibliometric studies in this field remain scarce. Bibliometrics provides a visual representation of research outcomes and trends, guiding future investigations. METHOD Journal data from January 1, 2013, to December 31, 2023, in this field were obtained from the Web of Science (WOS) core database. Analysis was conducted using VOSviewer and CiteSpace. RESULTS Analysis revealed that over the past decade, 794 articles were published in 299 journals by 4447 authors from 49 countries and 877 institutions. The top contributors were China, the United States, and the United Kingdom. Zhuang Chao emerged as the most prolific author, and "osteoarthritis and cartilage" ranked as the most frequently cited journal. Keyword clustering focused on mechanisms, inflammation, and cartilage. The most-cited article was "chondrocyte apoptosis in the pathogenesis of osteoarthritis" in the "International Journal of Molecular Sciences." Burst word analysis highlighted extracellular matrix, circular RNA, micro RNA, indicating current research hotspots. CONCLUSION Utilizing bibliometrics and visual analysis, we explored the hotspots and trends in the field of chondrocyte apoptosis in osteoarthritis. Extracellular matrix, Circular RNA, Micro RNA, among others, are likely to become future research focal points and frontiers.
Collapse
Affiliation(s)
- Hongxing Zhang
- Department of Second Clinical Medical College, Anhui University of Chinese Medicine, Hefei, Anhui Province, China
| | - Yao Yang
- Department of Second Clinical Medical College, Anhui University of Chinese Medicine, Hefei, Anhui Province, China
| | - Minglei Gao
- Department of Second Clinical Medical College, Anhui University of Chinese Medicine, Hefei, Anhui Province, China
| | - Jiafeng Peng
- Department of Second Clinical Medical College, Anhui University of Chinese Medicine, Hefei, Anhui Province, China
| | - Danyang Li
- Department of Second Clinical Medical College, Anhui University of Chinese Medicine, Hefei, Anhui Province, China
| | - Junchen Zhu
- Department of Orthopaedics, Second Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui Province, China
| |
Collapse
|
3
|
Dhanabalan KM, Padhan B, Dravid AA, Agarwal S, Pancheri NM, Lin A, Willet NJ, Padmanabhan AK, Agarwal R. Nordihydroguaiaretic acid microparticles are effective in the treatment of osteoarthritis. J Mater Chem B 2024. [PMID: 39356214 DOI: 10.1039/d4tb01342e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/03/2024]
Abstract
Several disease-modifying osteoarthritis (OA) drugs have emerged, but none have been approved for clinical use due to their systemic side effects, short half-life, and rapid clearance from the joints. Nordihydroguaiaretic acid (NDGA), a reactive oxygen species (ROS) scavenger and autophagy inducer, could be a potential treatment for OA. In this report, we show for the first time that sustained delivery of NDGA through polymeric microparticles maintains therapeutic concentrations of drug in the joint and ameliorates post-traumatic OA (PTOA) in a mouse model. In vitro treatment of oxidatively stressed primary chondrocytes from OA patients using NDGA-loaded poly(lactic-co-glycolic acid) (PLGA) microparticles (NDGA-MP) inhibited 15-lipoxygenase, induced autophagy, prevented chondrosenescence, and sustained matrix production. In vivo intra-articular delivery of NDGA-MP was non-toxic and had prolonged retention time (up to 35 days) in murine knee joints. Intra-articular therapy using NDGA-MP effectively reduced cartilage damage and reduced pain in the surgery-induced PTOA mouse model. Our studies open new avenues to modulate the immune environment and treat post-traumatic OA using ROS quenchers and autophagy inducers.
Collapse
Affiliation(s)
- Kaamini M Dhanabalan
- Department of Bioengineering, Indian Institute of Science, Bengaluru, India, 560012.
| | - Bhagyashree Padhan
- Department of Bioengineering, Indian Institute of Science, Bengaluru, India, 560012.
| | - Ameya A Dravid
- Department of Bioengineering, Indian Institute of Science, Bengaluru, India, 560012.
| | - Smriti Agarwal
- Department of Bioengineering, Indian Institute of Science, Bengaluru, India, 560012.
| | - Nicholas M Pancheri
- Phil and Penny Knight Campus for Accelerating Scientific Impact, Department of Bioengineering, University of Oregon, Eugene, OR, USA, 97403
| | - Angela Lin
- Phil and Penny Knight Campus for Accelerating Scientific Impact, Department of Bioengineering, University of Oregon, Eugene, OR, USA, 97403
| | - Nick J Willet
- Phil and Penny Knight Campus for Accelerating Scientific Impact, Department of Bioengineering, University of Oregon, Eugene, OR, USA, 97403
| | | | - Rachit Agarwal
- Department of Bioengineering, Indian Institute of Science, Bengaluru, India, 560012.
| |
Collapse
|
4
|
Li Y, Yang Y, Zhu L, Xie S, Guo L, Zhang Z, Zhe C, Li W, Liu F. Angelica sinensis polysaccharide facilitates chondrogenic differentiation of adipose-derived stem cells via MDK-PI3K/AKT signaling cascade. Biomed Pharmacother 2024; 179:117349. [PMID: 39191028 DOI: 10.1016/j.biopha.2024.117349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 08/21/2024] [Accepted: 08/22/2024] [Indexed: 08/29/2024] Open
Abstract
OBJECT Adipose-derived mesenchymal stem cells (ADSCs) have received significant attention in the field of cartilage tissue repair. Angelica sinensis polysaccharide (ASP) can enhance both the proliferation and differentiation of mesenchymal stem cells. Therefore, we intend to explore the effect of ASP on chondrogenic differentiation of ADSCs in vitro, and elucidate the underlying mechanisms. METHOD ADSCs were treated with different concentrations of ASP to determine the optimal concentration. The chondrogenic differentiation of ADSCs was evaluated using Alcian blue staining, qRT-PCR, western blot, and IF staining. Transcriptome sequencing was performed to identify the expression profiles of ADSCs before and after ASP treatment, followed by bioinformatic analyses including differential expression analysis, enrichment analysis, and construction of PPI networks to identify differentially expressed genes (DEGs) associated with ASP and chondrogenic differentiation. RESULT Surface markers of isolated rat-derived ADSCs were identified by CD44+CD90+CD45-CD106-, and exhibited the capacity for lipogenic, osteogenic, and chondrogenic differentiation. With increasing concentration of ASP treatment, there was an upregulation in the activity and acidic mucosubstance of ADSCs. The levels of Aggrecan, COL2A1, and Sox9 showed an increase in ADSCs after 28 days of 80 µg/ml ASP treatment. Transcriptome sequencing revealed that ASP-associated DEGs regulate extracellular matrix synthesis, immune response, inflammatory response, and cell cycle, and are involved in the NF-κB, AGE-RAGE, and calcium pathways. Moreover, Edn1, Frzb, Mdk, Nog, and Sulf1 are hub genes in DEGs. Notably, ASP upregulated MDK levels in ADSCs, while knockdown of MDK mitigated ASP-induced elevations in acidic mucosubstance, chondrogenic differentiation-related markers (Aggrecan, COL2A1, and Sox9), and the activity of the PI3K/AKT pathway. CONCLUSION ASP enhances the proliferation and chondrogenic differentiation of ADSCs by activating the MDK-mediated PI3K/AKT pathway.
Collapse
Affiliation(s)
- Yangjie Li
- Qujing No.1 Hospital, Affiliated Qujing Hospital of Kunming Medical University, No. 1 Yuanlin Road, Qujing City, Yunnan Province, China
| | - Yongqiang Yang
- Qujing No.1 Hospital, Affiliated Qujing Hospital of Kunming Medical University, No. 1 Yuanlin Road, Qujing City, Yunnan Province, China
| | - Lina Zhu
- Qujing No.1 Hospital, Affiliated Qujing Hospital of Kunming Medical University, No. 1 Yuanlin Road, Qujing City, Yunnan Province, China
| | - Shukang Xie
- Qujing No.1 Hospital, Affiliated Qujing Hospital of Kunming Medical University, No. 1 Yuanlin Road, Qujing City, Yunnan Province, China
| | - Ling Guo
- Qujing No.1 Hospital, Affiliated Qujing Hospital of Kunming Medical University, No. 1 Yuanlin Road, Qujing City, Yunnan Province, China
| | - Zhiming Zhang
- Qujing No.1 Hospital, Affiliated Qujing Hospital of Kunming Medical University, No. 1 Yuanlin Road, Qujing City, Yunnan Province, China
| | - Chunyang Zhe
- Qujing No.1 Hospital, Affiliated Qujing Hospital of Kunming Medical University, No. 1 Yuanlin Road, Qujing City, Yunnan Province, China
| | - Wenhui Li
- Kunming Institute of Zoology, Chinese Academy of Sciences, No.17 Longxin Road, Kunming City, Yunnan Province, China
| | - Feng Liu
- Qujing No.1 Hospital, Affiliated Qujing Hospital of Kunming Medical University, No. 1 Yuanlin Road, Qujing City, Yunnan Province, China.
| |
Collapse
|
5
|
Lee SH, Shin MK, Sung JS. Tamarixetin Protects Chondrocytes against IL-1β-Induced Osteoarthritis Phenotype by Inhibiting NF-κB and Activating Nrf2 Signaling. Antioxidants (Basel) 2024; 13:1166. [PMID: 39456419 PMCID: PMC11505541 DOI: 10.3390/antiox13101166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 09/17/2024] [Accepted: 09/23/2024] [Indexed: 10/28/2024] Open
Abstract
Osteoarthritis (OA) is a degenerative joint disease characterized by cartilage breakdown and chronic inflammation in joints. As the most prevalent form of arthritis, OA affects around 600 million people globally. Despite the increasing number of individuals with OA risk factors, such as aging and obesity, there is currently no effective cure for the disease. In this context, this study investigated the therapeutic effects of tamarixetin, a flavonoid with antioxidative and anti-inflammatory properties, against OA pathology and elucidated the underlying molecular mechanism. In interleukin-1β (IL-1β)-treated chondrocytes, tamarixetin inhibited the OA phenotypes, restoring cell viability and chondrogenic properties while reducing hypertrophic differentiation and dedifferentiation. Tamarixetin alleviated oxidative stress via the nuclear factor erythroid 2-related factor 2 (Nrf2) pathway activation and inhibited mitogen-activated protein kinase and nuclear factor-κB (NF-κB). Furthermore, tamarixetin attenuated pyroptosis, a programmed cell death caused by excessive inflammation, by suppressing inflammasome activation. We confirmed that the chondroprotective effects of tamarixetin are mediated by the concurrent upregulation of Nrf2 signaling and downregulation of NF-κB signaling, which are key players in balancing antioxidative and inflammatory responses. Overall, our study demonstrated that tamarixetin possesses chondroprotective properties by alleviating IL-1β-induced cellular stress in chondrocytes, suggesting its therapeutic potential to relieve OA phenotype.
Collapse
Affiliation(s)
| | | | - Jung-Suk Sung
- Department of Life Science, Dongguk University-Seoul, Goyang 10326, Republic of Korea; (S.-H.L.); (M.K.S.)
| |
Collapse
|
6
|
Villagrán-Andrade KM, Núñez-Carro C, Blanco FJ, de Andrés MC. Nutritional Epigenomics: Bioactive Dietary Compounds in the Epigenetic Regulation of Osteoarthritis. Pharmaceuticals (Basel) 2024; 17:1148. [PMID: 39338311 PMCID: PMC11434976 DOI: 10.3390/ph17091148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 08/24/2024] [Accepted: 08/27/2024] [Indexed: 09/30/2024] Open
Abstract
Nutritional epigenomics is exceptionally important because it describes the complex interactions among food compounds and epigenome modifications. Phytonutrients or bioactive compounds, which are secondary metabolites of plants, can protect against osteoarthritis by suppressing the expression of inflammatory and catabolic mediators, modulating epigenetic changes in DNA methylation, and the histone or chromatin remodelling of key inflammatory genes and noncoding RNAs. The combination of natural epigenetic modulators is crucial because of their additive and synergistic effects, safety and therapeutic efficacy, and lower adverse effects than conventional pharmacology in the treatment of osteoarthritis. In this review, we have summarized the chondroprotective properties of bioactive compounds used for the management, treatment, or prevention of osteoarthritis in both human and animal studies. However, further research is needed into bioactive compounds used as epigenetic modulators in osteoarthritis, in order to determine their potential value for future clinical applications in osteoarthritic patients as well as their relation with the genomic and nutritional environment, in order to personalize food and nutrition together with disease prevention.
Collapse
Affiliation(s)
- Karla Mariuxi Villagrán-Andrade
- Unidad de Epigenética, Grupo de Investigación en Reumatología (GIR), Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario, de A Coruña (CHUAC), Sergas, 15006 A Coruña, Spain
| | - Carmen Núñez-Carro
- Unidad de Epigenética, Grupo de Investigación en Reumatología (GIR), Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario, de A Coruña (CHUAC), Sergas, 15006 A Coruña, Spain
| | - Francisco J Blanco
- Unidad de Epigenética, Grupo de Investigación en Reumatología (GIR), Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario, de A Coruña (CHUAC), Sergas, 15006 A Coruña, Spain
- Grupo de Investigación en Reumatología y Salud, Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Facultad de Fisioterapia, Campus de Oza, Universidade da Coruña (UDC), 15008 A Coruña, Spain
| | - María C de Andrés
- Unidad de Epigenética, Grupo de Investigación en Reumatología (GIR), Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario, de A Coruña (CHUAC), Sergas, 15006 A Coruña, Spain
| |
Collapse
|
7
|
Li Z, Lu H, Fan L, Ma X, Duan Z, Zhang Y, Fu Y, Wang S, Guan Y, Yang D, Chen Q, Xu T, Yang Y. Microneedle-Delivered PDA@Exo for Multifaceted Osteoarthritis Treatment via PI3K-Akt-mTOR Pathway. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024:e2406942. [PMID: 39206714 DOI: 10.1002/advs.202406942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Indexed: 09/04/2024]
Abstract
Osteoarthritis (OA) is marked by cartilage deterioration, subchondral bone changes, and an inflammatory microenvironment. The study introduces the Microneedle-Delivered Polydopamine-Exosome (PDA@Exo MN), a therapeutic that not only preserves cartilage and promotes bone regeneration but also improves localized drug delivery through enhanced penetration capabilities. PDA@Exo MN shows strong reactive oxygen species (ROS) scavenging abilities and high biocompatibility, fostering osteogenesis and balancing anabolic and catabolic processes in cartilage. It directs macrophage polarization from M0 to the anti-inflammatory M2 phenotype. RNA sequencing of treated chondrocytes demonstrates restored cellular function and activated antioxidant responses, with modulated inflammatory pathways. The PI3K-AKT-mTOR pathway's activation, essential for PDA@Exo's effects, is confirmed via bioinformatics and Western blot. In vivo assessments robustly validate that PDA@Exo MN prevents cartilage degradation and OA progression, supported by histological assessments and micro-CT analysis, highlighting its disease-modifying impact. The excellent biocompatibility of PDA@Exo MN, verified through histological (H&E) and blood tests showing no organ damage, underscores its safety and efficacy for OA therapy, making it a novel and multifunctional nanomedical approach in orthopedics, characterized by organ-friendliness and biosecurity.
Collapse
Affiliation(s)
- Zihua Li
- Department of Orthopedics, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, P. R. China
| | - Hengli Lu
- Department of Orthopedics, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, P. R. China
| | - Limin Fan
- School of Medicine, Tongji University, Shanghai, 200092, P. R. China
| | - Xiaoyi Ma
- School of Medicine, Tongji University, Shanghai, 200092, P. R. China
| | - Zhengwei Duan
- Department of Orthopedics, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, P. R. China
| | - Yiwei Zhang
- Department of Orthopedics, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, P. R. China
| | - Yuesong Fu
- Department of Orthopedics, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, P. R. China
| | - Sen Wang
- Department of Orthopedics, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, P. R. China
| | - Yonghao Guan
- Department of Orthopedics, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, P. R. China
| | - Dong Yang
- Department of Orthopedics, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, P. R. China
| | - Qingjing Chen
- Southern Medical University, Guangzhou, 510515, P. R. China
| | - Tianyang Xu
- Department of Orthopedics, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, P. R. China
| | - Yunfeng Yang
- Department of Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, P. R. China
| |
Collapse
|
8
|
Tong Y, Yuan J, Li Z, Deng C, Cheng Y. Drug-Loaded Bioscaffolds for Osteochondral Regeneration. Pharmaceutics 2024; 16:1095. [PMID: 39204440 PMCID: PMC11360256 DOI: 10.3390/pharmaceutics16081095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 07/20/2024] [Accepted: 07/25/2024] [Indexed: 09/04/2024] Open
Abstract
Osteochondral defect is a complex tissue loss disease caused by arthritis, high-energy trauma, and many other reasons. Due to the unique structural characteristics of osteochondral tissue, the repair process is sophisticated and involves the regeneration of both hyaline cartilage and subchondral bone. However, the current clinical treatments often fall short of achieving the desired outcomes. Tissue engineering bioscaffolds, especially those created via three-dimensional (3D) printing, offer promising solutions for osteochondral defects due to their precisely controllable 3D structures. The microstructure of 3D-printed bioscaffolds provides an excellent physical environment for cell adhesion and proliferation, as well as nutrient transport. Traditional 3D-printed bioscaffolds offer mere physical stimulation, while drug-loaded 3D bioscaffolds accelerate the tissue repair process by synergistically combining drug therapy with physical stimulation. In this review, the physiological characteristics of osteochondral tissue and current treatments of osteochondral defect were reviewed. Subsequently, the latest progress in drug-loaded bioscaffolds was discussed and highlighted in terms of classification, characteristics, and applications. The perspectives of scaffold design, drug control release, and biosafety were also discussed. We hope this article will serve as a valuable reference for the design and development of osteochondral regenerative bioscaffolds and pave the way for the use of drug-loaded bioscaffolds in clinical therapy.
Collapse
Affiliation(s)
| | | | | | - Cuijun Deng
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-like Intelligence, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai 200434, China; (Y.T.); (J.Y.); (Z.L.)
| | - Yu Cheng
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-like Intelligence, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai 200434, China; (Y.T.); (J.Y.); (Z.L.)
| |
Collapse
|
9
|
Guastaldi FPS, Matheus HR, Hadad H, Randolph MA, Redmond RW. A regenerative approach for temporomandibular joint repair: An in vitro and ex vivo study. J Oral Rehabil 2024; 51:1521-1529. [PMID: 38717007 DOI: 10.1111/joor.13728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 03/19/2024] [Accepted: 04/26/2024] [Indexed: 07/14/2024]
Abstract
BACKGROUND Current clinical approaches to regenerate temporomandibular joint (TMJ) articulating cartilage defects only treat the symptoms (i.e. pain and dysfunction) and do not seek to restore joint integrity for long-term relief. Therefore, we investigated a novel self-assembling tissue-engineered cartilage to overcome this significant clinical issue for TMJ regenerative purposes. OBJECTIVES Examine the maturation of dynamic self-regenerating cartilage (dSRC) using auricular chondrocytes and evaluate a novel combinatorial approach with fractional laser treatment and dSRC implantation for TMJ cartilage repair. MATERIALS AND METHODS A suspension of 107 freshly harvested rabbit ear chondrocytes was cultured under a continuous reciprocating motion to form the dSRC. After 2, 4 and 8 weeks of culture, dSRC samples were stained with H&E, Safranin-O and Toluidine Blue. Immunohistochemistry (IHC) was performed for collagens type I and II. Channels (300-500 μm diameter and 1.2-1.5 mm depth) were created in six freshly harvested condyles using a fractional Erbium laser. Two groups were tested: dSRC in a laser-ablated lesion (experimental) and an empty laser-ablated channel (control). TMJ condyles were cultured for up to 8 weeks and analysed as described above. RESULTS H&E staining showed a high cell density in dSRC compared to native cartilage. All dSRC groups demonstrated intense Safranin-O staining, indicating high glycosaminoglycan (GAG) production and intense Toluidine Blue staining showed high proteoglycan content. IHC confirmed that dSRC consisted predominantly of collagen type II. The experimental group showed improved cartilage repair at both time points compared to the empty channels. CONCLUSION dSRC viability and successful matrix formation were demonstrated in vitro. The combination of fractional laser ablation and dSRC implantation enhanced cartilage repair.
Collapse
Affiliation(s)
- Fernando P S Guastaldi
- Department of Oral and Maxillofacial Surgery, Massachusetts General Hospital, Harvard School of Dental Medicine, Boston, Massachusetts, USA
| | - Henrique R Matheus
- Department of Oral and Maxillofacial Surgery, Massachusetts General Hospital, Harvard School of Dental Medicine, Boston, Massachusetts, USA
- Department of Diagnosis and Surgery, Division of Periodontics, São Paulo State University (UNESP), School of Dentistry, Araçatuba, SP, Brazil
| | - Henrique Hadad
- Department of Oral and Maxillofacial Surgery, Massachusetts General Hospital, Harvard School of Dental Medicine, Boston, Massachusetts, USA
- Department of Diagnosis and Surgery, Oral & Maxillofacial Surgery Division, São Paulo State University (UNESP), School of Dentistry, Araçatuba, SP, Brazil
| | - Mark A Randolph
- Division of Plastic and Reconstructive Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Robert W Redmond
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
10
|
Cao M, Sheng R, Sun Y, Cao Y, Wang H, Zhang M, Pu Y, Gao Y, Zhang Y, Lu P, Teng G, Wang Q, Rui Y. Delivering Microrobots in the Musculoskeletal System. NANO-MICRO LETTERS 2024; 16:251. [PMID: 39037551 PMCID: PMC11263536 DOI: 10.1007/s40820-024-01464-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 06/16/2024] [Indexed: 07/23/2024]
Abstract
Disorders of the musculoskeletal system are the major contributors to the global burden of disease and current treatments show limited efficacy. Patients often suffer chronic pain and might eventually have to undergo end-stage surgery. Therefore, future treatments should focus on early detection and intervention of regional lesions. Microrobots have been gradually used in organisms due to their advantages of intelligent, precise and minimally invasive targeted delivery. Through the combination of control and imaging systems, microrobots with good biosafety can be delivered to the desired area for treatment. In the musculoskeletal system, microrobots are mainly utilized to transport stem cells/drugs or to remove hazardous substances from the body. Compared to traditional biomaterial and tissue engineering strategies, active motion improves the efficiency and penetration of local targeting of cells/drugs. This review discusses the frontier applications of microrobotic systems in different tissues of the musculoskeletal system. We summarize the challenges and barriers that hinder clinical translation by evaluating the characteristics of different microrobots and finally point out the future direction of microrobots in the musculoskeletal system.
Collapse
Affiliation(s)
- Mumin Cao
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, People's Republic of China
- School of Medicine, Southeast University, Nanjing, 210009, People's Republic of China
- Orthopaedic Trauma Institute (OTI), Southeast University, Nanjing, 210009, People's Republic of China
| | - Renwang Sheng
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, People's Republic of China
- School of Medicine, Southeast University, Nanjing, 210009, People's Republic of China
- Orthopaedic Trauma Institute (OTI), Southeast University, Nanjing, 210009, People's Republic of China
| | - Yimin Sun
- Jiangsu Key Laboratory for Design and Manufacture of Micro-Nano Biomedical Instruments, School of Mechanical Engineering, Southeast University, Nanjing, 210009, People's Republic of China
| | - Ying Cao
- Jiangsu Key Laboratory for Design and Manufacture of Micro-Nano Biomedical Instruments, School of Mechanical Engineering, Southeast University, Nanjing, 210009, People's Republic of China
| | - Hao Wang
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, People's Republic of China
- School of Medicine, Southeast University, Nanjing, 210009, People's Republic of China
- Orthopaedic Trauma Institute (OTI), Southeast University, Nanjing, 210009, People's Republic of China
| | - Ming Zhang
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, People's Republic of China
- School of Medicine, Southeast University, Nanjing, 210009, People's Republic of China
- Orthopaedic Trauma Institute (OTI), Southeast University, Nanjing, 210009, People's Republic of China
| | - Yunmeng Pu
- School of Medicine, Southeast University, Nanjing, 210009, People's Republic of China
| | - Yucheng Gao
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, People's Republic of China
- School of Medicine, Southeast University, Nanjing, 210009, People's Republic of China
- Orthopaedic Trauma Institute (OTI), Southeast University, Nanjing, 210009, People's Republic of China
| | - Yuanwei Zhang
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, People's Republic of China
- School of Medicine, Southeast University, Nanjing, 210009, People's Republic of China
- Orthopaedic Trauma Institute (OTI), Southeast University, Nanjing, 210009, People's Republic of China
| | - Panpan Lu
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, People's Republic of China
- School of Medicine, Southeast University, Nanjing, 210009, People's Republic of China
- Orthopaedic Trauma Institute (OTI), Southeast University, Nanjing, 210009, People's Republic of China
| | - Gaojun Teng
- Center of Interventional Radiology and Vascular Surgery, Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, People's Republic of China.
| | - Qianqian Wang
- Jiangsu Key Laboratory for Design and Manufacture of Micro-Nano Biomedical Instruments, School of Mechanical Engineering, Southeast University, Nanjing, 210009, People's Republic of China.
| | - Yunfeng Rui
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, People's Republic of China.
- School of Medicine, Southeast University, Nanjing, 210009, People's Republic of China.
- Orthopaedic Trauma Institute (OTI), Southeast University, Nanjing, 210009, People's Republic of China.
| |
Collapse
|
11
|
Demmer W, Schinacher J, Wiggenhauser PS, Giunta RE. Use of Acellular Matrices as Scaffolds in Cartilage Regeneration: A Systematic Review. Adv Wound Care (New Rochelle) 2024. [PMID: 38775424 DOI: 10.1089/wound.2024.0065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/11/2024] Open
Abstract
Significance: Cartilage regeneration remains a significant challenge in the field of regenerative medicine. Acellular matrix (AM)-based cartilage tissue regeneration offers an innovative approach to repairing cartilage defects by providing a scaffold for new tissue growth. Its significance lies in its potential to restore joint function, mitigate pain, and improve the quality of life for patients suffering from cartilage-related injuries and conditions. Recent Advances: Recent advances in AM-based cartilage regeneration have focused on enhancing scaffold properties for improved cell adhesion, proliferation, and differentiation. Moreover, several scaffold techniques such as combining acellular dermal matrix (ADM) and acellular cartilage matrix (ACM) with cartilage tissue, as well as biphasic scaffolding, enjoy rising research activity. Incorporating bioactive factors and advanced manufacturing techniques holds promise for producing more biomimetic scaffolds, advancing efficient cartilage repair and regeneration. Critical Issues: Obstacles in AM-based cartilage regeneration include achieving proper integration with the surrounding tissue and ensuring long-term durability of the regenerated cartilage. Furthermore, issues such as high costs and limited availability of suitable cells for scaffold seeding must be considered. The heterogeneity and limited regenerative capabilities of cartilage need to be addressed for successful clinical translation. Future Directions: Research should focus on exploring advanced biomaterials and developing new techniques, regarding easily reproducible scaffolds, ideally constructed from clinically validated and readily available commercial products. Findings underline the potential of AM-based approaches, especially the rising exploration of tissue-derived ADM and ACM. In future, the primary objective should not only be the regeneration of small cartilage defects but rather focus on fully regenerating a joint or larger cartilage defect.
Collapse
|
12
|
Jeyaraman M, Jeyaraman N, Ramasubramanian S, Ranjan R, Jha SK, Gupta A. Bone Marrow Aspirate Concentrate for Treatment of Primary Knee Osteoarthritis: A Prospective, Single-Center, Non-randomized Study with 2-Year Follow-Up. Indian J Orthop 2024; 58:894-904. [PMID: 38948370 PMCID: PMC11208343 DOI: 10.1007/s43465-024-01168-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Accepted: 04/23/2024] [Indexed: 07/02/2024]
Abstract
Introduction Knee osteoarthritis (OA) is a widespread, disabling condition with no intervention to fully restore cartilage or halt progression. Bone marrow aspirate concentrate (BMAC), an autologous product from bone marrow aspiration, has shown promise as a regenerative therapy due to its cell composition and chondrogenic effects. Our study aims to assess the functional outcomes, including pain, function, satisfaction, and complications post-BMAC injection in knee OA patients. Materials and Methods In this prospective, single-center study, 63 patients with grade II-III knee OA (Kellgren-Lawrence (K-L) scale) unresponsive to conservative management underwent BMAC injection. The procedure involved bone marrow aspiration from the anterior iliac crest, processing to obtain a concentrate, followed by intra-articular injection. Patients were followed for 24 months, assessing outcomes using the Visual Analog Scale (VAS), International Knee Documentation Committee (IKDC) score, and MOCART 2.0 score. Results The cohort, with a slight female predominance and predominantly aged 41-50 years, majorly comprised K-L grade III OA patients. BMAC treatment resulted in significant improvements in VAS pain scores, IKDC functional scores, and MOCART 2.0 scores over the 24-month follow-up. Conclusion BMAC injection provides significant improvement in both pain and functional outcomes at mid-term follow-up in patients with mild-to-moderate OA of the knee. Further high-quality, adequately powered, multi-center, prospective, double-blinded, randomized controlled trials with longer follow-up are necessary to justify the routine clinical use of BMAC for treatment of patients suffering with knee OA. Graphical Abstract
Collapse
Affiliation(s)
- Madhan Jeyaraman
- Department of Orthopaedics, ACS Medical College and Hospital, Dr MGR Educational and Research Institute, Chennai, Tamil Nadu 600077 India
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida, Uttar Pradesh 201310 India
- Clinical Research Associate, Virginia Tech India, Dr MGR Educational and Research Institute, Chennai, Tamil Nadu 600095 India
- South Texas Orthopaedic Research Institute (STORI Inc.), Laredo, TX 78045 USA
| | - Naveen Jeyaraman
- Department of Orthopaedics, ACS Medical College and Hospital, Dr MGR Educational and Research Institute, Chennai, Tamil Nadu 600077 India
| | - Swaminathan Ramasubramanian
- Department of Orthopaedics, Government Medical College, Omandurar Government Estate, Chennai, Tamil Nadu 600002 India
| | - Rajni Ranjan
- Department of Orthopaedics, School of Medical Sciences and Research, Sharda University, Greater Noida, Uttar Pradesh 201306 India
| | - Saurabh Kumar Jha
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida, Uttar Pradesh 201310 India
- Department of Zoology, Kalindi College, University of Delhi, New Delhi, 110008 India
| | - Ashim Gupta
- South Texas Orthopaedic Research Institute (STORI Inc.), Laredo, TX 78045 USA
- Regenerative Orthopaedics, Noida, Uttar Pradesh 201301 India
- Future Biologics, Lawrenceville, GA 30043 USA
- BioIntegrate, Lawrenceville, GA 30043 USA
| |
Collapse
|
13
|
Kalairaj MS, Pradhan R, Saleem W, Smith MM, Gaharwar AK. Intra-Articular Injectable Biomaterials for Cartilage Repair and Regeneration. Adv Healthc Mater 2024; 13:e2303794. [PMID: 38324655 PMCID: PMC11468459 DOI: 10.1002/adhm.202303794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 12/29/2023] [Indexed: 02/09/2024]
Abstract
Osteoarthritis is a degenerative joint disease characterized by cartilage deterioration and subsequent inflammatory changes in the underlying bone. Injectable hydrogels have emerged as a promising approach for controlled drug delivery in cartilage therapies. This review focuses on the latest developments in utilizing injectable hydrogels as vehicles for targeted drug delivery to promote cartilage repair and regeneration. The pathogenesis of osteoarthritis is discussed to provide a comprehensive understanding of the disease progression. Subsequently, the various types of injectable hydrogels used for intra-articular delivery are discussed. Specifically, physically and chemically crosslinked injectable hydrogels are critically analyzed, with an emphasis on their fabrication strategies and their capacity to encapsulate and release therapeutic agents in a controlled manner. Furthermore, the potential of incorporating growth factors, anti-inflammatory drugs, and cells within these injectable hydrogels are discussed. Overall, this review offers a comprehensive guide to navigating the landscape of hydrogel-based therapeutics in osteoarthritis.
Collapse
Affiliation(s)
| | - Ridhi Pradhan
- Department of Biomedical EngineeringCollege of EngineeringTexas A&M UniversityCollege StationTX77843USA
| | - Waqas Saleem
- Department of Biomedical EngineeringCollege of EngineeringTexas A&M UniversityCollege StationTX77843USA
| | - Morgan M. Smith
- Department of Veterinary Integrative BiosciencesSchool of Veterinary Medicine and Biomedical SciencesTexas A&M UniversityCollege StationTX77843USA
| | - Akhilesh K. Gaharwar
- Department of Biomedical EngineeringCollege of EngineeringTexas A&M UniversityCollege StationTX77843USA
- Department of Material Science and EngineeringCollege of EngineeringTexas A&M UniversityCollege StationTX77843USA
- Genetics and Genomics Interdisciplinary ProgramTexas A&M UniversityCollege StationTX77843USA
| |
Collapse
|
14
|
Fazio A, Di Martino A, Brunello M, Traina F, Marvi MV, Mazzotti A, Faldini C, Manzoli L, Evangelisti C, Ratti S. The involvement of signaling pathways in the pathogenesis of osteoarthritis: An update. J Orthop Translat 2024; 47:116-124. [PMID: 39021400 PMCID: PMC11254498 DOI: 10.1016/j.jot.2024.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 04/09/2024] [Accepted: 06/02/2024] [Indexed: 07/20/2024] Open
Abstract
Osteoarthritis (OA) is one of the most common disabling pathologies, characterized by joint pain and reduced function, significantly worsening the quality of life. Even if important progresses have been made in OA research, little is yet known about the precise cellular and molecular mechanisms underlying OA. Understanding dysregulated signaling networks and their crosstalk in OA may offer a strong opportunity for the development of combined targeted therapies. Hence, this review highlights the recent findings on the main pathways involved in OA development, including Wnt, Notch, Hedgehog, MAPK, AMPK, and JAK/STAT, providing insights on current targeted therapies in OA patients' management. The translational potential of this article The identification of key signaling pathways involved in OA development and the investigation of their signaling crosstalk could pave the way for more effective treatments and improved management of OA patients in the future.
Collapse
Affiliation(s)
- Antonietta Fazio
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126, Bologna, Italy
| | - Alberto Di Martino
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126, Bologna, Italy
- Ist Orthopedic Department, IRCCS Istituto Ortopedico Rizzoli, 40136, Bologna, Italy
| | - Matteo Brunello
- Ist Orthopedic Department, IRCCS Istituto Ortopedico Rizzoli, 40136, Bologna, Italy
| | - Francesco Traina
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126, Bologna, Italy
- Ortopedia-Traumatologia e Chirurgia Protesica e dei Reimpianti d'anca e di Ginocchio, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
| | - Maria Vittoria Marvi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126, Bologna, Italy
| | - Antonio Mazzotti
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126, Bologna, Italy
- Ist Orthopedic Department, IRCCS Istituto Ortopedico Rizzoli, 40136, Bologna, Italy
| | - Cesare Faldini
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126, Bologna, Italy
- Ist Orthopedic Department, IRCCS Istituto Ortopedico Rizzoli, 40136, Bologna, Italy
| | - Lucia Manzoli
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126, Bologna, Italy
| | - Camilla Evangelisti
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126, Bologna, Italy
| | - Stefano Ratti
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126, Bologna, Italy
| |
Collapse
|
15
|
Saeedi T, Prokopovich P. Screening of poly-beta amino ester coated emulsion of ketorolac for cartilage delivery. J Mater Chem B 2024; 12:5930-5939. [PMID: 38804519 PMCID: PMC11186506 DOI: 10.1039/d4tb00313f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 05/13/2024] [Indexed: 05/29/2024]
Abstract
Osteoarthritis (OA) is a prevalent chronic health condition necessitating effective treatment strategies. Globally, there were 86 million people with incident knee osteoarthritis in 2020. Pain management remains the primary approach to OA as the nature of cartilage poses challenges for drug delivery. An emulsion-based delivery system, using a class of positively charged and hydrolysable polymers (poly-beta-amino-esters) to coat oil droplets containing drugs, has been shown to enhance and prolong drug localization in ex vivo cartilage models. As the properties of the polymers used in this technology strongly depend on the monomers used in the synthesis, this study presents the screening of a wide range of PBAEs as droplet coating agents and using ketorolac as a model of nonsteroidal anti-inflammatory drugs. The emulsions prepared with this PBAE library were characterized, and drug localisation and retention were evaluated in both native and glycosaminoglycan (GAG) depleted cartilage ex vivo models. Optimal candidates were identified and tested in an ex vivo model showing the ability to protect chondrocyte cell viability and increase both GAG and collagen contents in cartilage exposed to cytokine (IL-1α) simulating acute cartilage damage. This work demonstrates the potential of PBAE coated emulsion as a delivery system for effective drug delivery in OA treatment.
Collapse
Affiliation(s)
- Tahani Saeedi
- Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff, UK.
| | | |
Collapse
|
16
|
He Q, Lin Y, Chen B, Chen C, Zeng J, Dou X, Cai D, Zhou C, Wang H. Vitamin K2 ameliorates osteoarthritis by suppressing ferroptosis and extracellular matrix degradation through activation GPX4's dual functions. Biomed Pharmacother 2024; 175:116697. [PMID: 38759289 DOI: 10.1016/j.biopha.2024.116697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 04/23/2024] [Accepted: 05/01/2024] [Indexed: 05/19/2024] Open
Abstract
Vitamin K2 (VK2) is an effective compound for anti-ferroptosis and anti-osteoporosis, and Semen sojae praeparatum (Dandouchi in Chinese) is the main source of VK2. Chondrocyte ferroptosis and extracellular matrix (ECM) degradation playing a role in the pathogenesis of osteoarthritis (OA). Glutathione peroxidase 4 (GPX4) is the intersection of two mechanisms in regulating OA progression. But no studies have elucidated the therapeutic effects and mechanisms of VK2 on OA. This study utilized an in vivo rat OA model created via anterior cruciate ligament transection (ACLT) and an in vitro chondrocyte oxidative damage model induced by TBHP to investigate the protective effects and mechanisms of action of VK2 in OA. Knee joint pain in mice was evaluated using the Von Frey test. Micro-CT and Safranin O-Fast Green staining were employed to observe the extent of damage to the tibial cartilage and subchondral bone, while immunohistochemistry and PCR were used to examine GPX4 levels in joint cartilage. The effects of VK2 on rat chondrocyte viability were assessed using CCK-8 and flow cytometry assays, and chondrocyte morphology was observed with toluidine blue and alcian blue staining. The impact of VK2 on intracellular ferroptosis-related markers was observed using fluorescent staining and flow cytometry. Protein expression changes were detected by immunofluorescence and Western blot analysis. Furthermore, specific protein inhibitors were applied to confirm the dual-regulatory effects of VK2 on GPX4. VK2 can increase bone mass and cartilage thickness in the subchondral bone of the tibia, and reduce pain and the OARSI score induced by OA. Immunohistochemistry results indicate that VK2 exerts its anti-OA effects by regulating GPX4 to delay ECM degradation. VK2 can inhibit the activation of the MAPK/NFκB signaling pathway caused by reduced expression of intracellular GPX4, thereby decreasing ECM degradation. Additionally, VK2 can reverse the inhibitory effect of RSL3 on GPX4, increase intracellular GSH content and the GSH/GSSG ratio, reduce MDA content, and rescue chondrocyte ferroptosis. The protective mechanism of VK2 may involve its dual-target regulation of GPX4, reducing chondrocyte ferroptosis and inhibiting the MAPK/NFκB signaling pathway to decelerate the degradation of the chondrocyte extracellular matrix.
Collapse
Affiliation(s)
- Qi He
- First School of Clinical Medicine, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun Area, Guangzhou 510405, PR China; The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China; Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yuewei Lin
- First School of Clinical Medicine, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun Area, Guangzhou 510405, PR China; The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China
| | - Baihao Chen
- First School of Clinical Medicine, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun Area, Guangzhou 510405, PR China; The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China
| | - Chuyi Chen
- First School of Clinical Medicine, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun Area, Guangzhou 510405, PR China; The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China
| | - Jiaxu Zeng
- First School of Clinical Medicine, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun Area, Guangzhou 510405, PR China; The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China
| | - Xiangyun Dou
- First School of Clinical Medicine, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun Area, Guangzhou 510405, PR China; The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China
| | - Dongling Cai
- Department of Orthopedics, Panyu Hospital of Chinese Medicine, Guangzhou 510405, PR China.
| | - Chi Zhou
- Department of Orthopaedics, The First Affiliated Hospital, Guangzhou University of Chinese Medicine, 16 Jichang Road, Baiyun Area, Guangzhou 510405, PR China; Department of Orthopaedics, Maoming Hospital of Guangzhou University of Chinese Medicine, Maoming 525000, PR China.
| | - Haibin Wang
- Department of Orthopaedics, The First Affiliated Hospital, Guangzhou University of Chinese Medicine, 16 Jichang Road, Baiyun Area, Guangzhou 510405, PR China.
| |
Collapse
|
17
|
Krebs P, Nägele M, Fomina P, Virtanen V, Nippolainen E, Shaikh R, Afara I, Töyräs J, Usenov I, Sakharova T, Artyushenko V, Tafintseva V, Solheim J, Zimmermann B, Kohler A, König O, Saarakkala S, Mizaikoff B. Laser-irradiating infrared attenuated total reflection spectroscopy of articular cartilage: Potential and challenges for diagnosing osteoarthritis. OSTEOARTHRITIS AND CARTILAGE OPEN 2024; 6:100466. [PMID: 38623306 PMCID: PMC11016904 DOI: 10.1016/j.ocarto.2024.100466] [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: 09/20/2023] [Accepted: 03/26/2024] [Indexed: 04/17/2024] Open
Abstract
Objective A prototype infrared attenuated total reflection (IR-ATR) laser spectroscopic system designed for in vivo classification of human cartilage tissue according to its histological health status during arthroscopic surgery is presented. Prior to real-world in vivo applications, this so-called osteoarthritis (OA) scanner has been tested at in vitro conditions revealing the challenges associated with complex sample matrices and the accordingly obtained sparse spectral datasets. Methods In vitro studies on human knee cartilage samples at different contact pressures (i.e., 0.2-0.5 MPa) allowed recording cartilage degeneration characteristic IR signatures comparable to in vivo conditions with high temporal resolution. Afterwards, the cartilage samples were assessed based on the clinically acknowledged osteoarthritis cartilage histopathology assessment (OARSI) system and correlated with the obtained sparse IR data. Results Amide and carbohydrate signal behavior was observed to be almost identical between the obtained sparse IR data and previously measured FTIR data used for sparse partial least squares discriminant analysis (SPLSDA) to identify the spectral regions relevant to cartilage condition. Contact pressures between 0.3 and 0.4 MPa seem to provide the best sparse IR spectra for cylindrical (d = 3 mm) probe tips. Conclusion Laser-irradiating IR-ATR spectroscopy is a promising analytical technique for future arthroscopic applications to differentiate healthy and osteoarthritic cartilage tissue. However, this study also revealed that the flexible connection between the laser-based analyzer and the arthroscopic ATR-probe via IR-transparent fiberoptic cables may affect the robustness of the obtained IR data and requires further improvements.
Collapse
Affiliation(s)
- P. Krebs
- Institute of Analytical and Bioanalytical Chemistry, Ulm University, Ulm, Germany
| | | | - P. Fomina
- Institute of Analytical and Bioanalytical Chemistry, Ulm University, Ulm, Germany
| | - V. Virtanen
- Research Unit of Health Sciences and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland
| | - E. Nippolainen
- Department of Technical Physics, University of Eastern Finland, Kuopio, Finland
| | - R. Shaikh
- Department of Technical Physics, University of Eastern Finland, Kuopio, Finland
| | - I.O. Afara
- Department of Technical Physics, University of Eastern Finland, Kuopio, Finland
| | - J. Töyräs
- Department of Technical Physics, University of Eastern Finland, Kuopio, Finland
- Science Service Center, Kuopio University Hospital, Kuopio, Finland
- School of Electrical Engineering and Computer Science, The University of Queensland, Brisbane, Australia
| | | | | | | | - V. Tafintseva
- Faculty of Science and Technology, Norwegian University of Life Sciences, Ås, Norway
| | - J.H. Solheim
- Faculty of Science and Technology, Norwegian University of Life Sciences, Ås, Norway
| | - B. Zimmermann
- Faculty of Science and Technology, Norwegian University of Life Sciences, Ås, Norway
| | - A. Kohler
- Faculty of Science and Technology, Norwegian University of Life Sciences, Ås, Norway
| | - O. König
- Nanoplus Advanced Photonics Gerbrunn GmbH, Gerbrunn, Germany
| | - S. Saarakkala
- Research Unit of Health Sciences and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland
- Department of Diagnostic Radiology, Oulu University Hospital, Oulu, Finland
| | - B. Mizaikoff
- Institute of Analytical and Bioanalytical Chemistry, Ulm University, Ulm, Germany
- Hahn-Schickard, Ulm, Germany
| |
Collapse
|
18
|
Gracia-Ibáñez V, Rodríguez-Cervantes PJ, Granell P, Vergara M, Sancho-Bru JL. Using active or functional range of motion as a further indicator to detect hand osteoarthritis. A preliminary study. Disabil Rehabil 2024; 46:2269-2281. [PMID: 37273230 DOI: 10.1080/09638288.2023.2219068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 05/23/2023] [Indexed: 06/06/2023]
Abstract
PURPOSE Propose further indicators for helping to detect hand osteoarthritis (HOA) using either active or functional ranges of motion (AROM or FROM). METHOD Hand kinematics data of 16 hand joint angles from previous studies were used, taken from healthy participants and HOA patients with differently affected joints and distinct levels of compromise. Data consisted of: (i) AROM (extreme values and ranges); (ii) FROM while performing the Sollerman Hand Function Test (mean, extreme percentiles and ranges). Two linear discriminant analyses (stepwise method) were conducted, one per dataset (AROM and FROM), with condition (healthy/patient) as the grouping variable. The potential predictors were the data from the joints with significant differences between samples for each analysis (A-predictors and F-predictors). RESULTS Good sensitivity-specificity values of 85.2-90.9% and 93.8-93.9% for F-predictors and A-predictors, respectively. Sets of predictors corresponded to the joints more commonly affected by HOA. F-predictors: lower maximal flexion of carpometacarpal and interphalangeal thumb joints, higher maximal flexion of thumb metacarpal joint, lower flexion/extension range of ring proximal interphalangeal joint and higher maximal little finger adduction. A-predictors: narrower flexion/extension range of the thumb carpometacarpal joint, lesser extension of the ring metacarpophalangeal joint; lower flexion of the middle finger proximal interphalangeal joint along with a narrower palmar arch range. CONCLUSION Both sets of predictors provide discrimination capacity of HOA with good sensitivity-specificity, slightly better for A-predictors. The AROM measurement is technically less demanding and can be clinically applied even with manual goniometry.
Collapse
Affiliation(s)
- Verónica Gracia-Ibáñez
- Department of Mechanical Engineering and Construction, Universitat Jaume I, Castelló, Spain
| | | | - Pablo Granell
- Consorci Hospitalari Provincial de Castelló, Castelló, Spain
| | - Margarita Vergara
- Department of Mechanical Engineering and Construction, Universitat Jaume I, Castelló, Spain
| | | |
Collapse
|
19
|
Fu Y, Zhang C, Yang Y, Zhou B, Yang M, Zhu G, Zhu Y. Effect of umbilical cord blood-mononuclear cells on knee osteoarthritis in rabbits. J Orthop Surg Res 2024; 19:323. [PMID: 38811966 PMCID: PMC11138004 DOI: 10.1186/s13018-024-04815-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Accepted: 05/27/2024] [Indexed: 05/31/2024] Open
Abstract
BACKGROUND To investigate the effect and underlying mechanism of umbilical cord blood-mononuclear cells (UCB-MNCs) in treating knee osteoarthritis (KOA) in rabbits. METHODS A rabbit KOA model was prepared by anterior cruciate ligament transection (ACLT). Fifty New Zealand white rabbits were randomly divided into the control group, model group, sodium hyaluronate (SH) group, platelet-rich plasma (PRP) group and UCB-MNC group. Knee injections were performed once a week for five consecutive weeks. The gross view of the knee joint, morphology of knee cartilage and structural changes in the knee joint were observed on CT scans, and graded by the Lequesne MG behavioral score and the Mankin score. TNF-α and IL-1β levels in the synovial fluid of the knee were measured by the enzyme-linked immunosorbent assay (ELISA). Expression levels of MMP-13 and COL-II in the knee cartilage were detected by Western blotting and qRT-PCR. RESULTS The Lequesne MG behavioral score and the Mankin score were significantly higher in the model group than those in the control group (P < 0.05). Rabbits in the SH, PRP and UCB-MNC groups had sequentially lower scores than those in the model group. Imaging features of KOA were more pronounced in the model group than in the remaining groups. CB-MNC significantly relieved KOA, compared to SH and PRP. Significantly higher levels of TNF-α and IL-1β in the synovial fluid of the knee, and up-regulated MMP-13 and down-regulated COL-II in the knee cartilage were detected in the model group than in the control group. These changes were significantly reversed by the treatment with SH, PRP and UCB-MNCs, especially UCB-MNCs. CONCLUSION Injections of UCB-MNCs into knees protect the articular cartilage and hinder the progression of KOA in rabbits by improving the local microenvironment at knee joints.
Collapse
Affiliation(s)
- Yuhang Fu
- The Second School of Clinical Medicine of Binzhou Medical University, Yantai, 264199, Shandong Province, China
- Yantai Affiliated Hospital of Binzhou Medical University, Yantai, 264199, Shandong Province, China
| | - Chi Zhang
- Yantai Affiliated Hospital of Binzhou Medical University, Yantai, 264199, Shandong Province, China
| | - Yong Yang
- Yantai City Yantai Mountain Hospital, Yantai, 264008, Shandong Province, China
| | - Baisui Zhou
- Yantai Affiliated Hospital of Binzhou Medical University, Yantai, 264199, Shandong Province, China
| | - Meng Yang
- The Second School of Clinical Medicine of Binzhou Medical University, Yantai, 264199, Shandong Province, China
- Yantai Affiliated Hospital of Binzhou Medical University, Yantai, 264199, Shandong Province, China
| | - Guoshuai Zhu
- Yantai Affiliated Hospital of Binzhou Medical University, Yantai, 264199, Shandong Province, China
| | - Yonglin Zhu
- Yantai Affiliated Hospital of Binzhou Medical University, Yantai, 264199, Shandong Province, China.
| |
Collapse
|
20
|
Reyes Alcaraz V, Pattappa G, Miura S, Angele P, Blunk T, Rudert M, Hiraki Y, Shukunami C, Docheva D. A Narrative Review of the Roles of Chondromodulin-I (Cnmd) in Adult Cartilage Tissue. Int J Mol Sci 2024; 25:5839. [PMID: 38892027 PMCID: PMC11173128 DOI: 10.3390/ijms25115839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 05/13/2024] [Accepted: 05/16/2024] [Indexed: 06/21/2024] Open
Abstract
Articular cartilage is crucial for joint function but its avascularity limits intrinsic repair, leading to conditions like osteoarthritis (OA). Chondromodulin-I (Cnmd) has emerged as a key molecule in cartilage biology, with potential implications for OA therapy. Cnmd is primarily expressed in cartilage and plays an important role in chondrocyte proliferation, cartilage homeostasis, and the blocking of angiogenesis. In vivo and in vitro studies on Cnmd, also suggest an involvement in bone repair and in delaying OA progression. Its downregulation correlates with OA severity, indicating its potential as a therapeutic target. Further research is needed to fully understand the mode of action of Cnmd and its beneficial implications for managing OA. This comprehensive review aims to elucidate the molecular characteristics of Cnmd, from its expression pattern, role in cartilage maintenance, callus formation during bone repair and association with OA.
Collapse
Affiliation(s)
- Viviana Reyes Alcaraz
- Department of Musculoskeletal Tissue Regeneration, Orthopaedic Hospital König-Ludwig-Haus, University of Würzburg, 97070 Würzburg, Germany; (V.R.A.); (G.P.)
| | - Girish Pattappa
- Department of Musculoskeletal Tissue Regeneration, Orthopaedic Hospital König-Ludwig-Haus, University of Würzburg, 97070 Würzburg, Germany; (V.R.A.); (G.P.)
| | - Shigenori Miura
- Department of Molecular Biology and Biochemistry, Division of Dental Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8553, Japan; (S.M.); (C.S.)
| | - Peter Angele
- Laboratory of Experimental Trauma Surgery, Department of Trauma Surgery, University Hospital Regensburg, 93053 Regensburg, Germany;
| | - Torsten Blunk
- Department of Trauma, Hand, Plastic and Reconstructive Surgery, University Hospital Würzburg, 97080 Würzburg, Germany;
| | - Maximilian Rudert
- Department of Orthopaedics, Orthopaedic Hospital König-Ludwig-Haus, University of Würzburg, 97070 Würzburg, Germany;
| | - Yuji Hiraki
- Department of Cellular Differentiation, Institute for Frontier Medical Sciences, Kyoto University, Kyoto 606-8501, Japan;
| | - Chisa Shukunami
- Department of Molecular Biology and Biochemistry, Division of Dental Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8553, Japan; (S.M.); (C.S.)
| | - Denitsa Docheva
- Department of Musculoskeletal Tissue Regeneration, Orthopaedic Hospital König-Ludwig-Haus, University of Würzburg, 97070 Würzburg, Germany; (V.R.A.); (G.P.)
| |
Collapse
|
21
|
Reis IL, Lopes B, Sousa P, Sousa AC, Caseiro AR, Mendonça CM, Santos JM, Atayde LM, Alvites RD, Maurício AC. Equine Musculoskeletal Pathologies: Clinical Approaches and Therapeutical Perspectives-A Review. Vet Sci 2024; 11:190. [PMID: 38787162 PMCID: PMC11126110 DOI: 10.3390/vetsci11050190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Revised: 04/12/2024] [Accepted: 04/23/2024] [Indexed: 05/25/2024] Open
Abstract
Musculoskeletal injuries such as equine osteoarthritis, osteoarticular defects, tendonitis/desmitis, and muscular disorders are prevalent among sport horses, with a fair prognosis for returning to exercise or previous performance levels. The field of equine medicine has witnessed rapid and fruitful development, resulting in a diverse range of therapeutic options for musculoskeletal problems. Staying abreast of these advancements can be challenging, prompting the need for a comprehensive review of commonly used and recent treatments. The aim is to compile current therapeutic options for managing these injuries, spanning from simple to complex physiotherapy techniques, conservative treatments including steroidal and non-steroidal anti-inflammatory drugs, hyaluronic acid, polysulfated glycosaminoglycans, pentosan polysulfate, and polyacrylamides, to promising regenerative therapies such as hemoderivatives and stem cell-based therapies. Each therapeutic modality is scrutinized for its benefits, limitations, and potential synergistic actions to facilitate their most effective application for the intended healing/regeneration of the injured tissue/organ and subsequent patient recovery. While stem cell-based therapies have emerged as particularly promising for equine musculoskeletal injuries, a multidisciplinary approach is underscored throughout the discussion, emphasizing the importance of considering various therapeutic modalities in tandem.
Collapse
Affiliation(s)
- Inês L. Reis
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal; (I.L.R.); (B.L.); (P.S.); (A.C.S.); (C.M.M.); (J.M.S.); (L.M.A.); (R.D.A.)
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal;
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), 1300-477 Lisboa, Portugal
- Departamento de Ciências Veterinárias, Cooperativa de Ensino Superior Politécnico e Universitário (CESPU), Instituto Universitário de Ciências da Saúde (IUCS), Avenida Central de Gandra 1317, 4585-116 Gandra, Portugal
| | - Bruna Lopes
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal; (I.L.R.); (B.L.); (P.S.); (A.C.S.); (C.M.M.); (J.M.S.); (L.M.A.); (R.D.A.)
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal;
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), 1300-477 Lisboa, Portugal
| | - Patrícia Sousa
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal; (I.L.R.); (B.L.); (P.S.); (A.C.S.); (C.M.M.); (J.M.S.); (L.M.A.); (R.D.A.)
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal;
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), 1300-477 Lisboa, Portugal
| | - Ana C. Sousa
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal; (I.L.R.); (B.L.); (P.S.); (A.C.S.); (C.M.M.); (J.M.S.); (L.M.A.); (R.D.A.)
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal;
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), 1300-477 Lisboa, Portugal
| | - Ana R. Caseiro
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal;
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), 1300-477 Lisboa, Portugal
- Veterinary Sciences Department, University School Vasco da Gama (EUVG), Avenida José R. Sousa Fernandes, Lordemão, 3020-210 Coimbra, Portugal
- Vasco da Gama Research Center (CIVG), University School Vasco da Gama (EUVG), Avenida José R. Sousa Fernandes, Lordemão, 3020-210 Coimbra, Portugal
| | - Carla M. Mendonça
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal; (I.L.R.); (B.L.); (P.S.); (A.C.S.); (C.M.M.); (J.M.S.); (L.M.A.); (R.D.A.)
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal;
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), 1300-477 Lisboa, Portugal
- Campus Agrário de Vairão, Centro Clínico de Equinos de Vairão (CCEV), Rua da Braziela n° 100, 4485-144 Vairão, Portugal
| | - Jorge M. Santos
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal; (I.L.R.); (B.L.); (P.S.); (A.C.S.); (C.M.M.); (J.M.S.); (L.M.A.); (R.D.A.)
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal;
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), 1300-477 Lisboa, Portugal
| | - Luís M. Atayde
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal; (I.L.R.); (B.L.); (P.S.); (A.C.S.); (C.M.M.); (J.M.S.); (L.M.A.); (R.D.A.)
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal;
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), 1300-477 Lisboa, Portugal
- Campus Agrário de Vairão, Centro Clínico de Equinos de Vairão (CCEV), Rua da Braziela n° 100, 4485-144 Vairão, Portugal
| | - Rui D. Alvites
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal; (I.L.R.); (B.L.); (P.S.); (A.C.S.); (C.M.M.); (J.M.S.); (L.M.A.); (R.D.A.)
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal;
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), 1300-477 Lisboa, Portugal
- Departamento de Ciências Veterinárias, Cooperativa de Ensino Superior Politécnico e Universitário (CESPU), Instituto Universitário de Ciências da Saúde (IUCS), Avenida Central de Gandra 1317, 4585-116 Gandra, Portugal
| | - Ana C. Maurício
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal; (I.L.R.); (B.L.); (P.S.); (A.C.S.); (C.M.M.); (J.M.S.); (L.M.A.); (R.D.A.)
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal;
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), 1300-477 Lisboa, Portugal
- Campus Agrário de Vairão, Centro Clínico de Equinos de Vairão (CCEV), Rua da Braziela n° 100, 4485-144 Vairão, Portugal
| |
Collapse
|
22
|
Lu YC, Ho TC, Huang CH, Yeh SI, Chen SL, Tsao YP. PEDF peptide plus hyaluronic acid stimulates cartilage regeneration in osteoarthritis via STAT3-mediated chondrogenesis. Bone Joint Res 2024; 13:137-148. [PMID: 38555936 PMCID: PMC10981997 DOI: 10.1302/2046-3758.134.bjr-2023-0179.r2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/02/2024] Open
Abstract
Aims Pigment epithelium-derived factor (PEDF) is known to induce several types of tissue regeneration by activating tissue-specific stem cells. Here, we investigated the therapeutic potential of PEDF 29-mer peptide in the damaged articular cartilage (AC) in rat osteoarthritis (OA). Methods Mesenchymal stem/stromal cells (MSCs) were isolated from rat bone marrow (BM) and used to evaluate the impact of 29-mer on chondrogenic differentiation of BM-MSCs in culture. Knee OA was induced in rats by a single intra-articular injection of monosodium iodoacetate (MIA) in the right knees (set to day 0). The 29-mer dissolved in 5% hyaluronic acid (HA) was intra-articularly injected into right knees at day 8 and 12 after MIA injection. Subsequently, the therapeutic effect of the 29-mer/HA on OA was evaluated by the Osteoarthritis Research Society International (OARSI) histopathological scoring system and changes in hind paw weight distribution, respectively. The regeneration of chondrocytes in damaged AC was detected by dual-immunostaining of 5-bromo-2'-deoxyuridine (BrdU) and chondrogenic markers. Results The 29-mer promoted expansion and chondrogenic differentiation of BM-MSCs cultured in different defined media. MIA injection caused chondrocyte death throughout the AC, with cartilage degeneration thereafter. The 29-mer/HA treatment induced extensive chondrocyte regeneration in the damaged AC and suppressed MIA-induced synovitis, accompanied by the recovery of cartilage matrix. Pharmacological inhibitors of PEDF receptor (PEDFR) and signal transducer and activator of transcription 3 (STAT3) signalling substantially blocked the chondrogenic promoting activity of 29-mer on the cultured BM-MSCs and injured AC. Conclusion The 29-mer/HA formulation effectively induces chondrocyte regeneration and formation of cartilage matrix in the damaged AC.
Collapse
Affiliation(s)
- Yung-Chang Lu
- Department of Medical Research, Mackay Memorial Hospital, New Taipei City, Taiwan
- Department of Medicine, Mackay Medical College, New Taipei City, Taiwan
| | - Tsung-Chuan Ho
- Department of Medical Research, Mackay Memorial Hospital, New Taipei City, Taiwan
| | - Chang-Hung Huang
- Department of Medical Research, Mackay Memorial Hospital, New Taipei City, Taiwan
- Department of Medicine, Mackay Medical College, New Taipei City, Taiwan
- School of Dentistry, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Shu-I Yeh
- Department of Medical Research, Mackay Memorial Hospital, New Taipei City, Taiwan
- Department of Medicine, Mackay Medical College, New Taipei City, Taiwan
| | - Show-Li Chen
- Graduate Institute of Microbiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yeou-Ping Tsao
- Department of Medical Research, Mackay Memorial Hospital, New Taipei City, Taiwan
- Department of Medicine, Mackay Medical College, New Taipei City, Taiwan
| |
Collapse
|
23
|
Luo Z, Zeng H, Yang K, Wang Y. FOXQ1 inhibits the progression of osteoarthritis by regulating pyroptosis. Aging (Albany NY) 2024; 16:5077-5090. [PMID: 38503493 PMCID: PMC11006491 DOI: 10.18632/aging.205600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 01/17/2024] [Indexed: 03/21/2024]
Abstract
BACKGROUND Osteoarthritis (OA) is the most common age-related joint disease, and the NLRP3-induced pyroptosis has been demonstrated in its progression. The upstream molecules or specific mechanisms controlling NLRP3 and pyroptosis in OA remain unclear. METHODS Transcriptome sequencing was performed in the OA mice model, and the expression levels of differentially expressed genes were assessed by qRT-PCR. The cell model was constructed by IL-1β-induced ATDC5 cells. The cell proliferation was examined using CCK-8 assay, and apoptosis was tested using flow cytometry. Western blot was used in protein inspection, and ELISA was used in inflammatory response evaluation. RESULTS Compared with the control group, there were 229 up-regulated and 32 down-regulated genes in model group. We detected that FOXQ1 was down-regulated in the OA mice model, improved proliferation, and restrained apoptosis of chondrocytes. Over-expression of FOXQ1 could inhibit pyroptosis-related proteins and inflammatory cytokines, containing NLRP3, Caspase-1, GSDMD, IL-6, IL-18, and TNF-α, and in contrast, FOXQ1 silencing exerted the opposite trend. CONCLUSIONS FOXQ1 may inhibit OA progression via down-regulating NLRP3-induced pyroptosis in the present study.
Collapse
Affiliation(s)
- Zhihuan Luo
- Department of Sports Medicine, Ganzhou People’s Hospital, Ganzhou 341000, Jiangxi Province, China
| | - Hui Zeng
- Department of Sports Medicine, Ganzhou People’s Hospital, Ganzhou 341000, Jiangxi Province, China
| | - Kanghua Yang
- Department of Sports Medicine, Ganzhou People’s Hospital, Ganzhou 341000, Jiangxi Province, China
| | - Yihai Wang
- Department of Sports Medicine, Ganzhou People’s Hospital, Ganzhou 341000, Jiangxi Province, China
| |
Collapse
|
24
|
Zhou Z, Song P, Wu Y, Wang M, Shen C, Ma Z, Ren X, Wang X, Chen X, Hu Y, Li Z, Zhang Q, Li M, Geng Z, Su J. Dual-network DNA-silk fibroin hydrogels with controllable surface rigidity for regulating chondrogenic differentiation. MATERIALS HORIZONS 2024; 11:1465-1483. [PMID: 38221872 DOI: 10.1039/d3mh01581e] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2024]
Abstract
Osteoarthritis (OA) is a common joint disease known for cartilage degeneration, leading to a substantial burden on individuals and society due to its high disability rate. However, current clinical treatments for cartilage defects remain unsatisfactory due to the unclear mechanisms underlying cartilage regeneration. Tissue engineering hydrogels have emerged as an attractive approach in cartilage repair. Recent research studies have indicated that stem cells can sense the mechanical strength of hydrogels, thereby regulating their differentiation fate. In this study, we present the groundbreaking construction of dual-network DNA-silk fibroin (SF) hydrogels with controllable surface rigidity. The supramolecular networks, formed through DNA base-pairing, induce the development of β-sheet structures by constraining and aggregating SF molecules. Subsequently, SF was cross-linked via horseradish peroxidase (HRP)-mediated enzyme reactions to form the second network. Experimental results demonstrated a positive correlation between the surface rigidity of dual-network DNA-SF hydrogels and the DNA content. Interestingly, it was observed that dual-network DNA-SF hydrogels with moderate surface rigidity exhibited the highest effectiveness in facilitating the migration of bone marrow mesenchymal stem cells (BMSCs) and their chondrogenic differentiation. Transcriptome sequencing further confirmed that dual-network DNA-SF hydrogels primarily enhanced chondrogenic differentiation of BMSCs by upregulating the Wnt and TGF-β signaling pathways while accelerating collagen II synthesis. Furthermore, in vivo studies revealed that dual-network DNA-SF hydrogels with moderate surface rigidity significantly accelerated cartilage regeneration. In summary, the dual-network DNA-SF hydrogels represent a promising and novel therapeutic strategy for cartilage regeneration.
Collapse
Affiliation(s)
- Ziyang Zhou
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China.
- Organoid Research Center, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
- School of Medicine, Shanghai University, Shanghai, 200444, China
- School of Life Sciences, Shanghai University, Shanghai, 200444, China
| | - Peiran Song
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China.
- Organoid Research Center, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Yan Wu
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China.
- Organoid Research Center, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Miaomiao Wang
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China.
- Organoid Research Center, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Congyi Shen
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China.
- Organoid Research Center, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
- School of Medicine, Shanghai University, Shanghai, 200444, China
- School of Life Sciences, Shanghai University, Shanghai, 200444, China
| | - Zhixin Ma
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China.
- Organoid Research Center, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
- School of Medicine, Shanghai University, Shanghai, 200444, China
- School of Life Sciences, Shanghai University, Shanghai, 200444, China
| | - Xiaoxiang Ren
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China.
- Organoid Research Center, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Xiuhui Wang
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China.
- Organoid Research Center, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Xiao Chen
- Department of Orthopedics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Yan Hu
- Department of Orthopedics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Zuhao Li
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China.
- Organoid Research Center, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
- Department of Orthopedics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Qin Zhang
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China.
- Organoid Research Center, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Mengmeng Li
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China.
- Organoid Research Center, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Zhen Geng
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China.
- Organoid Research Center, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Jiacan Su
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China.
- Organoid Research Center, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
- Department of Orthopedics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| |
Collapse
|
25
|
Yammine KM, Abularach SM, Kim SY, Bikovtseva AA, Lilianty J, Butty VL, Schiavoni RP, Bateman JF, Lamandé SR, Shoulders MD. ER procollagen storage defect without coupled unfolded protein response drives precocious arthritis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.10.19.562780. [PMID: 37905055 PMCID: PMC10614947 DOI: 10.1101/2023.10.19.562780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
Collagenopathies are a group of clinically diverse disorders caused by defects in collagen folding and secretion. For example, mutations in the gene encoding collagen type-II, the primary collagen in cartilage, can lead to diverse chondrodysplasias. One example is the Gly1170Ser substitution in procollagen-II, which causes precocious osteoarthritis. Here, we biochemically and mechanistically characterize an induced pluripotent stem cell-based cartilage model of this disease, including both hetero- and homozygous genotypes. We show that Gly1170Ser procollagen-II is notably slow to fold and secrete. Instead, procollagen-II accumulates intracellularly, consistent with an endoplasmic reticulum (ER) storage disorder. Owing to unique features of the collagen triple helix, this accumulation is not recognized by the unfolded protein response. Gly1170Ser procollagen-II interacts to a greater extent than wild-type with specific proteostasis network components, consistent with its slow folding. These findings provide mechanistic elucidation into the etiology of this disease. Moreover, the cartilage model will enable rapid testing of therapeutic strategies to restore proteostasis in the collagenopathies.
Collapse
|
26
|
Liu Y, Hao R, Lv J, Yuan J, Wang X, Xu C, Ma D, Duan Z, Zhang B, Dai L, Cheng Y, Lu W, Zhang X. Targeted knockdown of PGAM5 in synovial macrophages efficiently alleviates osteoarthritis. Bone Res 2024; 12:15. [PMID: 38433252 PMCID: PMC10909856 DOI: 10.1038/s41413-024-00318-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 01/02/2024] [Accepted: 01/19/2024] [Indexed: 03/05/2024] Open
Abstract
Osteoarthritis (OA) is a common degenerative disease worldwide and new therapeutics that target inflammation and the crosstalk between immunocytes and chondrocytes are being developed to prevent and treat OA. These attempts involve repolarizing pro-inflammatory M1 macrophages into the anti-inflammatory M2 phenotype in synovium. In this study, we found that phosphoglycerate mutase 5 (PGAM5) significantly increased in macrophages in OA synovium compared to controls based on histology of human samples and single-cell RNA sequencing results of mice models. To address the role of PGAM5 in macrophages in OA, we found conditional knockout of PGAM5 in macrophages greatly alleviated OA symptoms and promoted anabolic metabolism of chondrocytes in vitro and in vivo. Mechanistically, we found that PGAM5 enhanced M1 polarization via AKT-mTOR/p38/ERK pathways, whereas inhibited M2 polarization via STAT6-PPARγ pathway in murine bone marrow-derived macrophages. Furthermore, we found that PGAM5 directly dephosphorylated Dishevelled Segment Polarity Protein 2 (DVL2) which resulted in the inhibition of β-catenin and repolarization of M2 macrophages into M1 macrophages. Conditional knockout of both PGAM5 and β-catenin in macrophages significantly exacerbated osteoarthritis compared to PGAM5-deficient mice. Motivated by these findings, we successfully designed mannose modified fluoropolymers combined with siPGAM5 to inhibit PGAM5 specifically in synovial macrophages via intra-articular injection, which possessed desired targeting abilities of synovial macrophages and greatly attenuated murine osteoarthritis. Collectively, these findings defined a key role for PGAM5 in orchestrating macrophage polarization and provides insights into novel macrophage-targeted strategy for treating OA.
Collapse
Affiliation(s)
- Yuhang Liu
- Department of Orthopedic Surgery, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, 200092, China
- National Facility for Translational Medicine, Shanghai, 200240, China
| | - Ruihan Hao
- Department of Orthopedic Surgery, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, 200092, China
- National Facility for Translational Medicine, Shanghai, 200240, China
| | - Jia Lv
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Jie Yuan
- Department of Orthopaedic Surgery, The Second Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Xuelei Wang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Churong Xu
- School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Ding Ma
- Department of Orthopedic Surgery, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, 200092, China
- National Facility for Translational Medicine, Shanghai, 200240, China
| | - Zhouyi Duan
- Department of Orthopedic Surgery, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, 200092, China
- National Facility for Translational Medicine, Shanghai, 200240, China
| | - Bingjun Zhang
- Department of Orthopedic Surgery, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, 200092, China
- National Facility for Translational Medicine, Shanghai, 200240, China
| | - Liming Dai
- Department of Orthopedic Surgery, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, 200092, China
- National Facility for Translational Medicine, Shanghai, 200240, China
| | - Yiyun Cheng
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai, 200241, China.
| | - Wei Lu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200031, China.
| | - Xiaoling Zhang
- Department of Orthopedic Surgery, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, 200092, China.
- National Facility for Translational Medicine, Shanghai, 200240, China.
| |
Collapse
|
27
|
Zhang Y, Li Z, Chen C, Wei W, Li Z, Huang H, Zhou H, He W, Xia J, Li B, Yang Y. S100A12 is involved in the pathology of osteoarthritis by promoting M1 macrophage polarization via the NF-κB pathway. Connect Tissue Res 2024; 65:133-145. [PMID: 38492210 DOI: 10.1080/03008207.2024.2310852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 01/23/2024] [Indexed: 03/18/2024]
Abstract
BACKGROUND Osteoarthritis (OA) is a degenerative joint disease that affects millions worldwide. Synovitis and macrophage polarization are important factors in the development of OA. However, the specific components of synovial fluid (SF) responsible for promoting macrophage polarization remain unclear. METHODS Semi-quantitative antibody arrays were used to outline the proteome of SF. Differential expression analysis and GO/KEGG were performed on the obtained data. Immunohistochemistry and ELISA were used to investigate the relationship between SF S100A12 levels and synovitis levels in clinalclinical samples. In vitro cell experiments were conducted to investigate the effect of S100A12 on macrophage polarization. Public databases were utilized to predict and construct an S100A12-centered lncRNA-miRNA-mRNA competing endogenous RNA network, which was preliminarily validated using GEO datasets. RESULTS The study outlines the protein profile in OA and non-OA SF. The results showed that the S100A12 level was significantly increased in OA SF and inflammatory chondrocytes. The OA synovium had more severe synovitis and higher levels of S100A12 than non-OA synovium. Exogenous S100A12 upregulated the levels of M1 markers and phosphorylated p65 and promoted p65 nuclear translocation, while pretreatment with BAY 11-7082 reversed these changes. It was also discovered that LINC00894 was upregulated in OA and significantly correlated with S100A12, potentially regulating S100A12 expression by acting as a miRNA sponge. CONCLUSIONS This study demonstrated that S100A12 promotes M1 macrophage polarization through the NF-κB pathway, and found that LINC00894 has the potential to regulate the expression of S100A12 as a therapeutic approach.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Orthopedics, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
- Department of Orthopedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zihua Li
- Department of Orthopedics, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
- Department of Orthopaedics, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Cheng Chen
- Department of Orthopedics, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
- Department of Orthopedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wang Wei
- The First Department of Orthopaedics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, No.157, Xiwu Road, Xi'an, Shaanxi, China
| | - Zhendong Li
- Department of Orthopedics, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Hui Huang
- Department of Orthopedics, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Haichao Zhou
- Department of Orthopedics, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Wenbao He
- Department of Orthopedics, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jiang Xia
- Department of Orthopedics, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Bing Li
- Department of Orthopedics, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
- Department of Orthopedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yunfeng Yang
- Department of Orthopedics, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
- Department of Orthopedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
28
|
Lv Z, Wang P, Li W, Xie Y, Sun W, Jin X, Jiang R, Fei Y, Liu Y, Shi T, Guo H, Sun Z, Lin J, Wang X, Tan G, Wu Y, Bao N, Shi D. Bifunctional TRPV1 Targeted Magnetothermal Switch to Attenuate Osteoarthritis Progression. RESEARCH (WASHINGTON, D.C.) 2024; 7:0316. [PMID: 38371274 PMCID: PMC10871150 DOI: 10.34133/research.0316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 01/20/2024] [Indexed: 02/20/2024]
Abstract
Transient receptor potential vanilloid family member 1 (TRPV1) has been revealed as a therapeutic target of osteoarthritis (OA), the most common deteriorating whole joint disease, by impeding macrophagic inflammation and chondrocytes ferroptosis. However, the clinical application for capsaicin as the TRPV1 agonist is largely limited by its chronic toxicity. To address this issue, we developed a bifunctional controllable magnetothermal switch targeting TRPV1 for the alleviation of OA progression by coupling of magnetic nanoparticles (MNPs) to TRPV1 monoclonal antibodies (MNPs-TRPV1). Under the alternating magnetic field (AMF) stimulation, MNPs-TRPV1 locally dissipated heat, which was sufficient to trigger the opening and activation of TRPV1, and effectively impeded macrophagic inflammation and chondrocyte ferroptosis. This magnetothermal modulation of TRPV1 simultaneously attenuated synovitis and cartilage degeneration in mice incurred by destabilization of medial meniscus surgery, indicating the delayed OA progression. Furthermore, MNPs-TRPV1 with AMF exposure remarkably reduced knee pain sensitivity, alleviated the crippled gait, and improved spontaneous ambulatory activity performance in the mice OA model. Overall, this work provides a potential pathogenesis-based precise OA therapy with temporally and spatially magnetothermal modulation of TRPV1 in a controllable manner.
Collapse
Affiliation(s)
- Zhongyang Lv
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School,
Nanjing University, Nanjing 210008, China
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical School,
Nanjing University, Nanjing 210002, China
| | - Peng Wang
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School,
Nanjing University, Nanjing 210008, China
| | - Weitong Li
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery,
Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, 321 Zhongshan Road, Nanjing 210008, Jiangsu, China
| | - Ya Xie
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery,
Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, 321 Zhongshan Road, Nanjing 210008, Jiangsu, China
| | - Wei Sun
- Department of orthopedic,
The Jiangyin Clinical College of Xuzhou Medical University, Jiangyin 214400, China
| | - Xiaoyu Jin
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery,
Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, 321 Zhongshan Road, Nanjing 210008, Jiangsu, China
| | - Ruiyang Jiang
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery,
Nanjing Drum Tower Hospital Clinical College of Xuzhou Medical University, 321 Zhongshan Road, Nanjing 210008, Jiangsu, China
| | - Yuxiang Fei
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School,
Nanjing University, Nanjing 210008, China
| | - Yuan Liu
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School,
Nanjing University, Nanjing 210008, China
| | - Tianshu Shi
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School,
Nanjing University, Nanjing 210008, China
| | - Hu Guo
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School,
Nanjing University, Nanjing 210008, China
| | - Ziying Sun
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School,
Nanjing University, Nanjing 210008, China
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical School,
Nanjing University, Nanjing 210002, China
| | - Jintao Lin
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical School,
Nanjing University, Nanjing 210002, China
| | - Xucai Wang
- Co-Innovation Center for Efficient Processing and Utilization of Forest Resources, College of Chemical Engineering,
Nanjing Forestry University, Nanjing, 210037, China
| | - Guihua Tan
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School,
Nanjing University, Nanjing 210008, China
| | - Yizhang Wu
- Department of Applied Physical Sciences,
The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Nirong Bao
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical School,
Nanjing University, Nanjing 210002, China
| | - Dongquan Shi
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School,
Nanjing University, Nanjing 210008, China
| |
Collapse
|
29
|
Mancini FE, Humphreys PEA, Woods S, Bates N, Cuvertino S, O'Flaherty J, Biant L, Domingos MAN, Kimber SJ. Effect of a retinoic acid analogue on BMP-driven pluripotent stem cell chondrogenesis. Sci Rep 2024; 14:2696. [PMID: 38302538 PMCID: PMC10834951 DOI: 10.1038/s41598-024-52362-3] [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: 06/21/2023] [Accepted: 01/17/2024] [Indexed: 02/03/2024] Open
Abstract
Osteoarthritis is the most common degenerative joint condition, leading to articular cartilage (AC) degradation, chronic pain and immobility. The lack of appropriate therapies that provide tissue restoration combined with the limited lifespan of joint-replacement implants indicate the need for alternative AC regeneration strategies. Differentiation of human pluripotent stem cells (hPSCs) into AC progenitors may provide a long-term regenerative solution but is still limited due to the continued reliance upon growth factors to recapitulate developmental signalling processes. Recently, TTNPB, a small molecule activator of retinoic acid receptors (RARs), has been shown to be sufficient to guide mesodermal specification and early chondrogenesis of hPSCs. Here, we modified our previous differentiation protocol, by supplementing cells with TTNPB and administering BMP2 at specific times to enhance early development (referred to as the RAPID-E protocol). Transcriptomic analyses indicated that activation of RAR signalling significantly upregulated genes related to limb and embryonic skeletal development in the early stages of the protocol and upregulated genes related to AC development in later stages. Chondroprogenitors obtained from RAPID-E could generate cartilaginous pellets that expressed AC-related matrix proteins such as Lubricin, Aggrecan, and Collagen II, but additionally expressed Collagen X, indicative of hypertrophy. This protocol could lay the foundations for cell therapy strategies for osteoarthritis and improve the understanding of AC development in humans.
Collapse
Affiliation(s)
- Fabrizio E Mancini
- Division of Cell Matrix and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, University of Manchester, Oxford Rd, Manchester, M13 9PT, UK
- Department of Solids and Structures, School of Engineering, Faculty of Science and Engineering, University of Manchester, Manchester, M13 9PL, UK
| | - Paul E A Humphreys
- Division of Cell Matrix and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, University of Manchester, Oxford Rd, Manchester, M13 9PT, UK
| | - Steven Woods
- Division of Cell Matrix and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, University of Manchester, Oxford Rd, Manchester, M13 9PT, UK
| | - Nicola Bates
- Division of Cell Matrix and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, University of Manchester, Oxford Rd, Manchester, M13 9PT, UK
| | - Sara Cuvertino
- Division of Evolution, Infection and Genomics, Faculty of Biology, Medicine and Health, The University of Manchester, Oxford Rd, Manchester, M13 9PT, UK
| | - Julieta O'Flaherty
- Division of Cell Matrix and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, University of Manchester, Oxford Rd, Manchester, M13 9PT, UK
| | - Leela Biant
- Division of Cell Matrix and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, University of Manchester, Oxford Rd, Manchester, M13 9PT, UK
| | - Marco A N Domingos
- Department of Solids and Structures, School of Engineering, Faculty of Science and Engineering, University of Manchester, Manchester, M13 9PL, UK
| | - Susan J Kimber
- Division of Cell Matrix and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, University of Manchester, Oxford Rd, Manchester, M13 9PT, UK.
| |
Collapse
|
30
|
Koshy J, Sangeetha D. Recent progress and treatment strategy of pectin polysaccharide based tissue engineering scaffolds in cancer therapy, wound healing and cartilage regeneration. Int J Biol Macromol 2024; 257:128594. [PMID: 38056744 DOI: 10.1016/j.ijbiomac.2023.128594] [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: 08/15/2023] [Revised: 11/12/2023] [Accepted: 12/02/2023] [Indexed: 12/08/2023]
Abstract
Natural polymers and its mixtures in the form of films, sponges and hydrogels are playing a major role in tissue engineering and regenerative medicine. Hydrogels have been extensively investigated as standalone materials for drug delivery purposes as they enable effective encapsulation and sustained release of drugs. Biopolymers are widely utilised in the fabrication of hydrogels due to their safety, biocompatibility, low toxicity, and regulated breakdown by human enzymes. Among all the biopolymers, polysaccharide-based polymer is well suited to overcome the limitations of traditional wound dressing materials. Pectin is a polysaccharide which can be extracted from different plant sources and is used in various pharmaceutical and biomedical applications including cartilage regeneration. Pectin itself cannot be employed as scaffolds for tissue engineering since it decomposes quickly. This article discusses recent research and developments on pectin polysaccharide, including its types, origins, applications, and potential demands for use in AI-mediated scaffolds. It also covers the materials-design process, strategy for implementation to material selection and fabrication methods for evaluation. Finally, we discuss unmet requirements and current obstacles in the development of optimal materials for wound healing and bone-tissue regeneration, as well as emerging strategies in the field.
Collapse
Affiliation(s)
- Jijo Koshy
- Department of Chemistry, School of Advanced Sciences, Vellore Institute of Technology, Vellore 632014, Tamil Nadu, India
| | - D Sangeetha
- Department of Chemistry, School of Advanced Sciences, Vellore Institute of Technology, Vellore 632014, Tamil Nadu, India.
| |
Collapse
|
31
|
Xu L, Wang Z, Wang G. Screening of Biomarkers Associated with Osteoarthritis Aging Genes and Immune Correlation Studies. Int J Gen Med 2024; 17:205-224. [PMID: 38268862 PMCID: PMC10807283 DOI: 10.2147/ijgm.s447035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 01/10/2024] [Indexed: 01/26/2024] Open
Abstract
Purpose Osteoarthritis (OA) is a joint disease with a long and slow course, which is one of the major causes of disability in middle and old-aged people. This study was dedicated to excavating the cellular senescence-associated biomarkers of OA. Methods The Gene Expression Omnibus (GEO) database was searched and five datasets pertaining to OA were obtained. After removing the batch effect, the GSE55235, GSE55457, GSE82107, and GSE12021 datasets were integrated together for screening of the candidate genes by differential analysis and weighted gene co-expression network analysis (WGCNA). Next, those genes were further filtered by machine learning algorithms to obtain cellular senescence-associated biomarkers of OA. Subsequently, enrichment analyses based on those biomarkers were conducted, and we profiled the infiltration levels of 22 types immune cells with the ERSORT algorithm. A lncRNA-miRNA-mRNA regulatory and drug-gene network were constructed. Finally, we validated the senescence-associated biomarkers at both in vivo and in vitro levels. Results Five genes (BCL6, MCL1, SLC16A7, PIM1, and EPHA3) were authenticated as cellular senescence-associated biomarkers in OA. ROC curves demonstrated the reliable capacity of the five genes as a whole to discriminate OA samples from normal samples. The nomogram diagnostic model based on 5 genes proved to be a reliable predictor of OA. Single-gene GSEA results pointed to the involvement of the five biomarkers in immune-related pathways and oxidative phosphorylation in the development of OA. Immune infiltration analysis manifested that the five genes were significantly correlated with differential immune cells. Subsequently, a lncRNA-miRNA-mRNA network and gene-drug network containing were generated based on five cellular senescence-associated biomarkers in OA. Conclusion A foundation for understanding the pathophysiology of OA and new insights into OA diagnosis and treatment were provided by the identification of five genes, namely BCL6, MCL1, SLC16A7, PIM1, and EPHA3, as biomarkers associated with cellular senescence in OA.
Collapse
Affiliation(s)
- Lanwei Xu
- Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People’s Republic of China
- Department of Hand and Foot Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, People’s Republic of China
| | - Zheng Wang
- Department of Neurosurgery, Liaocheng Traditional Chinese Medicine Hospital, Liaocheng, 252000, People’s Republic of China
| | - Gang Wang
- Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People’s Republic of China
| |
Collapse
|
32
|
Hannani MT, Thudium CS, Karsdal MA, Ladel C, Mobasheri A, Uebelhoer M, Larkin J, Bacardit J, Struglics A, Bay-Jensen AC. From biochemical markers to molecular endotypes of osteoarthritis: a review on validated biomarkers. Expert Rev Mol Diagn 2024; 24:23-38. [PMID: 38353446 DOI: 10.1080/14737159.2024.2315282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 02/02/2024] [Indexed: 02/22/2024]
Abstract
INTRODUCTION Osteoarthritis (OA) affects over 500 million people worldwide. OA patients are symptomatically treated, and current therapies exhibit marginal efficacy and frequently carry safety-risks associated with chronic use. No disease-modifying therapies have been approved to date leaving surgical joint replacement as a last resort. To enable effective patient care and successful drug development there is an urgent need to uncover the pathobiological drivers of OA and how these translate into disease endotypes. Endotypes provide a more precise and mechanistic definition of disease subgroups than observable phenotypes, and a panel of tissue- and pathology-specific biochemical markers may uncover treatable endotypes of OA. AREAS COVERED We have searched PubMed for full-text articles written in English to provide an in-depth narrative review of a panel of validated biochemical markers utilized for endotyping of OA and their association to key OA pathologies. EXPERT OPINION As utilized in IMI-APPROACH and validated in OAI-FNIH, a panel of biochemical markers may uncover disease subgroups and facilitate the enrichment of treatable molecular endotypes for recruitment in therapeutic clinical trials. Understanding the link between biochemical markers and patient-reported outcomes and treatable endotypes that may respond to given therapies will pave the way for new drug development in OA.
Collapse
Affiliation(s)
- Monica T Hannani
- ImmunoScience, Nordic Bioscience A/S, Herlev, Denmark
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | | | | | - Ali Mobasheri
- Research Unit of Health Sciences and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania
- Department of Joint Surgery, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- World Health Organization Collaborating Centre for Public Health Aspects of Musculoskeletal Health and Aging, Université de Liège, Liège, Belgium
| | | | - Jonathan Larkin
- Research Unit of Health Sciences and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland
- SynOA Therapeutics, Philadelphia, PA, USA
| | - Jaume Bacardit
- School of Computing, Newcastle University, Newcastle upon Tyne, UK
| | - André Struglics
- Department of Clinical Sciences, Orthopaedics, Lund University, Lund, Sweden
| | | |
Collapse
|
33
|
Fan SH, Chang Y, Xiong XY, Xiang M, Yuan WL, Yang XQ, Wei WH, Chen L, Cheng MN, Zhu FH, He SJ, Zuo JP, Lin ZM. Reversible SAHH inhibitor ameliorates MIA-induced osteoarthritis of rats through suppressing MEK/ERK pathway. Biomed Pharmacother 2024; 170:115975. [PMID: 38070246 DOI: 10.1016/j.biopha.2023.115975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 11/23/2023] [Accepted: 11/29/2023] [Indexed: 01/10/2024] Open
Abstract
Osteoarthritis (OA) is characterized by gradual articular cartilage degradation, accompanied by persistent low-grade joint inflammation, correlating with radiographic and pain-related progression. The latent therapeutic potential of DZ2002, a reversible inhibitor of S-adenosyl-L-homocysteine hydrolase (SAHH), holds promise for OA intervention. This study endeavored to examine the therapeutic efficacy of DZ2002 within the milieu of OA. The cytotoxicity of DZ2002 was evaluated using the MTT assay on bone marrow-derived macrophages. The inhibitory impact of DZ2002 during the process of osteoclastogenesis was assessed using TRAP staining, analysis of bone resorption pits, and F-actin ring formation. Mechanistic insights were derived from qPCR and Western blot analyses. Through the intra-articular injection of monosodium iodoacetate (MIA), an experimental rat model of OA was successfully instituted. This was subsequently accompanied by a series of assessments including Von Frey filament testing, analysis of weight-bearing behaviors, and micro-CT imaging, all aimed at assessing the effectiveness of DZ2002. The findings emphasized the effectiveness of DZ2002 in mitigating osteoclastogenesis induced by M-CSF/RANKL, evident through a reduction in TRAP-positive OCs and bone resorption. Moreover, DZ2002 modulated bone resorption-associated gene and protein expression (CTSK, CTR, Integrin β3) via the MEK/ERK pathway. Encouragingly, DZ2002 also alleviates MIA-induced pain, cartilage degradation, and bone loss. In conclusion, DZ2002 emerges as a potential therapeutic contender for OA, as evidenced by its capacity to hinder in vitro M-CSF/RANKL-induced osteoclastogenesis and mitigate in vivo osteoarthritis progression. This newfound perspective provides substantial support for considering DZ2002 as a compelling agent for osteoarthritis intervention.
Collapse
Affiliation(s)
- Shu-Hui Fan
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, Jiangsu Province 210029, China; Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yuan Chang
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, Jiangsu Province 210029, China; Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Xiao-Yu Xiong
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, Jiangsu Province 210029, China; Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Mai Xiang
- Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; School of Pharmacy, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wen-Long Yuan
- Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; School of Pharmacy, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiao-Qian Yang
- Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Wen-Hui Wei
- Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; School of Pharmacy, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Li Chen
- Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Meng-Nan Cheng
- Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Feng-Hua Zhu
- Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Shi-Jun He
- Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Jian-Ping Zuo
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, Jiangsu Province 210029, China; Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; School of Pharmacy, University of Chinese Academy of Sciences, Beijing 100049, China; Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Ze-Min Lin
- Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; School of Pharmacy, University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
34
|
Sun W, Lv Z, Li W, Lu J, Xie Y, Wang P, Jiang R, Dong J, Guo H, Liu Z, Fei Y, Tan G, Wang M, Ren K, Xu J, Sun H, Jiang X, Shi D. XJB-5-131 protects chondrocytes from ferroptosis to alleviate osteoarthritis progression via restoring Pebp1 expression. J Orthop Translat 2024; 44:114-124. [PMID: 38304614 PMCID: PMC10830431 DOI: 10.1016/j.jot.2023.12.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 12/30/2023] [Accepted: 12/30/2023] [Indexed: 02/03/2024] Open
Abstract
Background Osteoarthritis (OA) is the most common age-related musculoskeletal disease. However, there is still a lack of therapy that can modify OA progression due to the complex pathogenic mechanisms. The aim of the study was to explore the role and mechanism of XJB-5-131 inhibiting chondrocytes ferroptosis to alleviate OA progression. Methods We treated tert-butyl hydroperoxide (TBHP)-induced ferroptosis of mouse primary chondrocytes with XJB-5-131 in vitro. The intracellular ferroptotic hallmarks, cartilage anabolic and catabolic markers, ferroptosis regulatory genes and proteins were detected. Then we established a mouse OA model via destabilization of the medial meniscus (DMM) surgery. The OA mice were treated with intra-articular injection of XJB-5-131 regularly (2 μM, 3 times per week). After 4 and 8 weeks, we performed micro-CT and histological examination to evaluate the protection role of XJB-5-131 in mouse OA subjects. RNA sequencing analysis was performed to unveil the key downstream gene of XJB-5-131 exerting the anti-ferroptotic effect in OA. Results XJB-5-131 significantly suppressed TBHP-induced increases of ferroptotic hallmarks (ROS, lipid peroxidation, and Fe2+ accumulation), ferroptotic drivers (Ptgs2, Pgd, Tfrc, Atf3, Cdo1), while restored the expression of ferroptotic suppressors (Gpx4, Fth1). XJB-5-131 evidently promoted the expression of cartilage anabolic and decreased the expression of cartilage catabolic markers. Moreover, intra-articular injection of XJB-5-131 significantly inhibited the expression of Cox2 and Mmp13, while promoted the expression of Col2a1, Gpx4 and Fth1 in DMM-induced mouse articular cartilage. Further, we identified Pebp1 as a potential target of XJB-5-131 by RNA sequencing analysis. The anti-ferroptosis and chondroprotective effects of XJB-5-131 were significantly diminished by Locostatin, a specific antagonist of Pebp1. Conclusion XJB-5-131 significantly protects chondrocytes from ferroptosis in TBHP-induced mouse primary chondrocytes and DMM surgery-induced OA mice model via restoring the expression of Pebp1. XJB-5-131 is a potential therapeutic drug in the management of OA progression.
Collapse
Affiliation(s)
- Wei Sun
- Department of Orthopedics, Jiangyin People's Hospital Affiliated to Nantong University, 163 Shoushan Road, Jiangyin, 214400, Jiangsu, PR China
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Affiliated Drum Tower Hospital, Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Department of Orthopedics, The Jiangyin Clinical College of Xuzhou Medical University, 163 Shoushan Road, Jiangyin, 214400, Jiangsu, PR China
| | - Zhongyang Lv
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, PR China
| | - Weitong Li
- Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210008, Jiangsu, PR China
| | - Jun Lu
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Affiliated Drum Tower Hospital, Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
| | - Ya Xie
- Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210008, Jiangsu, PR China
| | - Peng Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Affiliated Drum Tower Hospital, Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
| | - Ruiyang Jiang
- Nanjing Drum Tower Hospital Clinical College of Xuzhou Medical University, Xuzhou Medical University, Nanjing, 210008, Jiangsu, PR China
| | - Jian Dong
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Affiliated Drum Tower Hospital, Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
| | - Hu Guo
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Affiliated Drum Tower Hospital, Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
| | - Zizheng Liu
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Affiliated Drum Tower Hospital, Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
| | - Yuxiang Fei
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Affiliated Drum Tower Hospital, Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
| | - Guihua Tan
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Affiliated Drum Tower Hospital, Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
| | - Maochun Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Affiliated Drum Tower Hospital, Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
| | - Kewei Ren
- Department of Orthopedics, Jiangyin People's Hospital Affiliated to Nantong University, 163 Shoushan Road, Jiangyin, 214400, Jiangsu, PR China
| | - Jun Xu
- Department of Orthopedics, Jiangyin People's Hospital Affiliated to Nantong University, 163 Shoushan Road, Jiangyin, 214400, Jiangsu, PR China
| | - Huiqing Sun
- Department of Orthopedics, Jiangyin People's Hospital Affiliated to Nantong University, 163 Shoushan Road, Jiangyin, 214400, Jiangsu, PR China
| | - Xuefeng Jiang
- Department of Orthopedics, Jiangyin People's Hospital Affiliated to Nantong University, 163 Shoushan Road, Jiangyin, 214400, Jiangsu, PR China
- Department of Orthopedics, The Jiangyin Clinical College of Xuzhou Medical University, 163 Shoushan Road, Jiangyin, 214400, Jiangsu, PR China
| | - Dongquan Shi
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Affiliated Drum Tower Hospital, Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210008, Jiangsu, PR China
- Nanjing Drum Tower Hospital Clinical College of Xuzhou Medical University, Xuzhou Medical University, Nanjing, 210008, Jiangsu, PR China
| |
Collapse
|
35
|
Liu Y, Liu M, Zhang C, Li X, Zheng S, Wen L, Liu P, Li P, Yang Z. The silencing of NREP aggravates OA cartilage damage through the TGF-β1/Smad2/3 pathway in chondrocytes. J Orthop Translat 2024; 44:26-34. [PMID: 38179126 PMCID: PMC10765488 DOI: 10.1016/j.jot.2023.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 11/02/2023] [Accepted: 11/06/2023] [Indexed: 01/06/2024] Open
Abstract
Background Osteoarthritis (OA) is a common chronic degenerative joint disease. Due to the limited understanding of its complex pathological mechanism, there is currently no effective treatment that can alleviate or even reverse cartilage damage associated with OA. With improvement in public databases, researchers have successfully identified the key factors involved in the occurrence and development of OA through bioinformatics analysis. The aim of this study was to screen for the differentially expressed genes (DEGs) between the normal and OA cartilage through bioinformatics, and validate the function of the TGF-β1/Smad2/3 pathway-related neuron regeneration related protein (NREP) in the articular cartilage. Methods The DEGs between the cartilage tissues of OA patients and healthy controls were screened by bioinformatics, and functionally annotated by Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses. The expression levels of the DEG in human and murine OA cartilage was verified by reverse transcription-quantitative PCR (RT-qPCR), Western blotting and immunohistochemistry (IHC). RT-qPCR, Western-blotting, Cell Counting Kit-8(CCK8) and EdU assays were used to evaluate the effects of knocking down NREP in normal chondrocytes, and the molecular mechanisms were investigated by RT-qPCR, Western blotting and IHC. Results In this study, we identified NREP as a DEG in OA through bioinformatics analysis, and found that NREP was downregulated in the damaged articular cartilage of OA patients and mouse model with surgically-induced OA. In addition, knockdown of NREP in normal chondrocytes reduced their proliferative capacity, which is the pathological basis of OA. At the molecular level, knock-down of NREP inactivated the TGF-β1/Smad2/3 pathway, resulting in the downregulation of the anabolic markers Col2a1 and Sox9, and an increase in the expression of the catabolic markers MMP3 and MMP13. Conclusion NREP plays a key role in the progression of OA by regulating the TGF-β1/Smad2/3 pathway in chondrocytes, and warrants further study as a potential therapeutic target.
Collapse
Affiliation(s)
- Yang Liu
- Department of Orthopedics, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, 030000, PR China
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Shanxi Medical University, Taiyuan, Shanxi, 030000, PR China
- Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Taiyuan, Shanxi, 030000, PR China
| | - Mengrou Liu
- Department of Orthopedics, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, 030000, PR China
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Shanxi Medical University, Taiyuan, Shanxi, 030000, PR China
- Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Taiyuan, Shanxi, 030000, PR China
| | - Chengming Zhang
- Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Taiyuan, Shanxi, 030000, PR China
| | - Xiaoke Li
- Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Taiyuan, Shanxi, 030000, PR China
| | - Siyu Zheng
- Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Taiyuan, Shanxi, 030000, PR China
| | - Le Wen
- Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Taiyuan, Shanxi, 030000, PR China
| | - Peidong Liu
- Department of Orthopedics, HongHui Hospital of Xi'an Jiao Tong University, Xi'an, Shannxi, 710000, PR China
| | - Pengcui Li
- Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Taiyuan, Shanxi, 030000, PR China
| | - Ziquan Yang
- Department of Orthopedics, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, 030000, PR China
- Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Taiyuan, Shanxi, 030000, PR China
| |
Collapse
|
36
|
Feng W, Zhu C, Miao R, Li D, Xiong X, Wang R, Liu G, Ma J. Comparative Efficacy of Endogenous Stem Cells Recruiting Hydrogels and Stem Cell-loaded Hydrogels in Knee Cartilage Regeneration: A Meta- analysis. Curr Stem Cell Res Ther 2024; 19:993-1008. [PMID: 37711133 DOI: 10.2174/1574888x19666230914123443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 07/10/2023] [Accepted: 07/24/2023] [Indexed: 09/16/2023]
Abstract
BACKGROUND Cartilage defects remain a challenge in diseases such as osteoarthritis (OA) and fractures. Scientists have explored the use of hydrogels in conjunction with stem cell technology as a tissue engineering method to treat cartilage defects in joints. In recent years, research into hydrogels containing stem cell technology for cartilage repair has mainly focused on two categories: stem cell-loaded hydrogels and endogenous stem cell recruiting hydrogels. The latter, utilizing cell-free products, represents a novel concept with several advantages, including easier dose standardization, wider sources, and simpler storage. This meta-analysis aims to assess and compare the therapeutic effects of endogenous stem cell recruiting hydrogels and stem cell-loaded hydrogels in promoting articular cartilage regeneration in animal models, with the goal of exploring endogenous stem cell recruiting hydrogels as a promising replacement therapy for knee cartilage regeneration in preclinical animal studies. METHODS We systematically searched PubMed, Web of Science, Cochrane Library, and Embase until January 2023 using key words related to stem cells, cartilage regeneration and hydrogel. A random-effects meta-analysis was performed to evaluate the therapeutic effect on newborn cartilage formation. Stratified analyses were also carried out by independently classifying trials according to similar characteristics. The level of evidence was determined using the GRADE method. RESULTS Twenty-eight studies satisfied the inclusion criteria. Comprehensive analyses revealed that the use of endogenous stem cell recruiting hydrogels significantly promoted the formation of new cartilage in the knee joint, as evidenced by the histological score (3.77, 95% CI 2.40, 5.15; p < 0.0001) and the International Cartilage Repair Society (ICRS) macroscopic score (3.00, 95% CI 1.83, 4.18; p = 0.04), compared with the control group. The stem cell-loaded hydrogels also increased cartilage regeneration in the knee with the histological score (3.13, 95% CI 2.22, 4.04; p = 0.02) and the ICRS macroscopic score (2.49, 95% CI 1.16, 3.82; p = 0.03) in comparison to the control. Significant heterogeneity between studies was observed, and further stratified and sensitivity analyses identified the transplant site and modelling method as the sources of heterogeneity. CONCLUSION The current study indicates that both endogenous stem cell recruiting hydrogels and stem cell loaded hydrogels can effectively promote knee joint cartilage regeneration in animal trials.
Collapse
Affiliation(s)
- Wanyun Feng
- Hebei Medical University-National University of Ireland Galway Stem Cell Research Center, Hebei Medical University, Hebei Province, 050017, China
- Hebei Research Center for Stem Cell Medical Translational Engineering, Hebei Province, 050017, China
| | - Chaohua Zhu
- The First Hospital of Hebei Medical University, Hebei Province, 050000, China
| | - Ruoxiang Miao
- Hebei Medical University-National University of Ireland Galway Stem Cell Research Center, Hebei Medical University, Hebei Province, 050017, China
- Hebei Research Center for Stem Cell Medical Translational Engineering, Hebei Province, 050017, China
| | - Danni Li
- Hebei Medical University-National University of Ireland Galway Stem Cell Research Center, Hebei Medical University, Hebei Province, 050017, China
- Hebei Research Center for Stem Cell Medical Translational Engineering, Hebei Province, 050017, China
| | - Xi Xiong
- Hebei Medical University-National University of Ireland Galway Stem Cell Research Center, Hebei Medical University, Hebei Province, 050017, China
- Hebei Research Center for Stem Cell Medical Translational Engineering, Hebei Province, 050017, China
| | - Ruyu Wang
- Hebei Medical University-National University of Ireland Galway Stem Cell Research Center, Hebei Medical University, Hebei Province, 050017, China
- Hebei Research Center for Stem Cell Medical Translational Engineering, Hebei Province, 050017, China
| | - Guobin Liu
- The First Hospital of Hebei Medical University, Hebei Province, 050000, China
| | - Jun Ma
- Hebei Medical University-National University of Ireland Galway Stem Cell Research Center, Hebei Medical University, Hebei Province, 050017, China
- Hebei Research Center for Stem Cell Medical Translational Engineering, Hebei Province, 050017, China
- Department of Human Anatomy, Hebei Medical University, Hebei Province, 050017, China
| |
Collapse
|
37
|
Li S, Li Y, Hou L, Tang L, Gao F. Forsythoside B alleviates osteoarthritis through the HMGB1/TLR4/NF-κB and Keap1/Nrf2/HO-1 pathways. J Biochem Mol Toxicol 2024; 38:e23569. [PMID: 37943572 DOI: 10.1002/jbt.23569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 05/05/2023] [Accepted: 10/18/2023] [Indexed: 11/10/2023]
Abstract
Osteoarthritis (OA) is a joint pain and dysfunction syndrome resulting from severe joint degeneration. Inflammation and degeneration of the articular cartilage are two main features of OA and have tight interactions during OA progression. Conventional treatment with nonsteroidal anti-inflammatory drugs has been widely utilized clinically, whereas the side effects have restricted their application. Forsythoside B has been found with anti-inflammatory effects and antiapoptosis in inflammatory diseases, whereas in OA it remains poorly understood. Interleukin (IL)-1β (10 ng/mL) was taken to induce an OA cell model on HC-A chondrocytes and an OA rat model was constructed for in vivo experiments. Forsythoside B was adopted to treat HC-A chondrocytes and OA rats. As shown by the data, Forsythoside B hampered IL-1β-elicited rat chondrocyte apoptosis, oxidative stress, and facilitated proliferation. The profiles of inflammatory factors, NOD-like receptor family pyrin domain containing 3 inflammasomes, Kelch-like epichlorohydrin-associated protein-1 (Keap1), and nuclear factor-κB (NF-κB) phosphorylation were suppressed by Forsythoside B, whereas the nuclear factor E2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1) levels were promoted. Further, Forsythoside B mitigated cartilage damage and degeneration. Moreover, the oxidative stress and inflammation mediators in the cartilage tissue of OA rats were remarkably abated. Collectively, Forsythoside B hinders the NF-κB and Keap1/Nrf2/HO-1 pathways to curb IL-1β-elicited OA rat oxidative stress and inflammation both in vivo and ex vivo, ameliorating OA development. All over, this study provides an underlying strategy for treating OA, which might help the clinical treatment of OA patients.
Collapse
Affiliation(s)
- Shujuan Li
- Neurology Department, Wuxi People Hosptial, Wuxi, China
| | - Yan Li
- Rehabilitation Medicine Department, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Li Hou
- Department of Geriatrics, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Li Tang
- Rehabilitation Medicine Department, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fang Gao
- Rehabilitation Medicine Department, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
38
|
Cao J, Wang D, Yuan J, Hu F, Wu Z. Exploration of the potential mechanism of Duhuo Jisheng Decoction in osteoarthritis treatment by using network pharmacology and molecular dynamics simulation. Comput Methods Biomech Biomed Engin 2024; 27:251-265. [PMID: 37830364 DOI: 10.1080/10255842.2023.2268232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 10/01/2023] [Indexed: 10/14/2023]
Abstract
In this study, the active ingredients of 15 Chinese herbal medicines of Duhuo Jisheng Decoction and their corresponding targets were obtained from the Traditional Chinese Medicine Systems Pharmacology (TCMSP) database. The microarray data of Osteoarthritis (OA) were obtained through the GEO database for differential analysis and then a drug target-OA-related gene protein-protein interaction (PPI) network was established. The potential targets of Duhuo Jisheng Decoction in the treatment of OA were acquired by intersecting the OA-associated genes with the target genes of active ingredients. Random walk with restart (RWR) analysis of PPI networks was performed using potential targets as seed, and the top 50 genes of affinity coefficients were used as key action genes of Duhuo Jisheng Decoction in the treatment of OA. A drug-active ingredient-gene interaction network was established. AKT1, a key target of Duhuo Jisheng Decoction in the treatment of OA, was obtained by topological analysis of the gene interaction network. Molecular docking and molecular dynamics verified the binding of AKT1 to its corresponding drug active ingredients. CETSA assay demonstrated that the combination of luteolin and AKT1 increased the stability of AKT1, and the combination efficiency was high. In conclusion, the molecular mechanism of Duhuo Jisheng Decoction in treating OA featured by multiple components, targets, and pathways had been further investigated in this study, which is of significance for discovering as well as developing new drugs for this disease. The findings can also offer personalized diagnosis and treatment strategies for patients with OA in clinical practice.
Collapse
Affiliation(s)
- Jin Cao
- Department of Orthopedics, First People's Hospital of Linping District, Hangzhou, China
| | - Dayong Wang
- Department of Orthopedics, First People's Hospital of Linping District, Hangzhou, China
| | - Jianhua Yuan
- Department of Orthopedics, First People's Hospital of Linping District, Hangzhou, China
| | - Fenggen Hu
- Department of Orthopedics, First People's Hospital of Linping District, Hangzhou, China
| | - Zhen Wu
- Department of Orthopedics, Tongde Hospital of Zhejiang Province, Hangzhou, China
| |
Collapse
|
39
|
Zhu W, Yang X, Liu S, Wang Y, Li W, Zhong Q, Zhang L, Xu J. Lentivirus-based shRNA of Caspase-3 gene silencing inhibits chondrocyte apoptosis and delays the progression of surgically induced osteoarthritis. Biotechnol J 2024; 19:e2300031. [PMID: 37750185 DOI: 10.1002/biot.202300031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 09/20/2023] [Accepted: 09/21/2023] [Indexed: 09/27/2023]
Abstract
Chondrocyte apoptosis is an important pathological feature of osteoarthritis (OA). Excessive apoptosis of chondrocytes disrupts the dynamic balance of cell proliferation and apoptosis, with a marked reduction in chondrocytes and cartilage matrix disintegration, which represents the main pathology of OA. Caspases, especially Caspase-3, play a central role in cell apoptosis. In this study, a lentiviral vector was used to transduce caspase-3 short hairpin RNA (shRNA) into rat chondrocytes (RCs), and the apoptotic and phenotypic genes of RCs were analyzed using real-time PCR and western blotting in vitro. In addition, in vivo intra-articular injection of Caspase-3 shRNA lentivirus was performed in a surgically induced OA rat model. Our results showed that Caspase-3 gene silencing could down-regulate the TNF-α-mediated inflammatory gene expression of TNFR1, FADD, and IL-1β, apoptotic gene expression of APAF1, Caspase-3, and Caspase-9, thereby attenuating the apoptotic pathway in vitro. Caspase-3 gene silencing also attenuated TNF-α-mediated decreased gene expression of ACAN, Col1-a1, and Col2-a1. Furthermore, Caspase-3 gene silencing could effectively reduce the OARSI score, and gene expression of Caspase-3, Caspase-9, MMP13, and TNF-α in a surgically induced OA rat model. Caspase-3 gene silencing may serve as a novel therapeutic strategy for cartilage injury and OA.
Collapse
Affiliation(s)
- Weicong Zhu
- Guangzhou Institute of Traumatic Surgery, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, China
| | - Xiaohong Yang
- Guangzhou Institute of Traumatic Surgery, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, China
| | - Shaojie Liu
- Surgical Department, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, China
| | - Yiwen Wang
- Department of Pharmacy, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, China
| | - Wenxu Li
- Department of Orthopedics, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, China
| | - Qiguang Zhong
- Surgical Department, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, China
| | - Lihua Zhang
- Surgical Department, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, China
| | - Jiake Xu
- School of Biomedical Sciences, The University of Western Australia, Perth, WA, Australia
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| |
Collapse
|
40
|
Gao J, Pei H, Lv F, Niu X, You Y, He L, Hu S, Shah KM, Liu M, Chen Y, Du B, Xiong H, Luo J. JD-312 - A novel small molecule that facilitates cartilage repair and alleviates osteoarthritis progression. J Orthop Translat 2024; 44:60-71. [PMID: 38269355 PMCID: PMC10805627 DOI: 10.1016/j.jot.2023.11.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 08/12/2023] [Accepted: 11/21/2023] [Indexed: 01/26/2024] Open
Abstract
Background The chondrogenic differentiation of mesenchymal stem cells (MSCs) to enhance cartilage repair and regeneration is a promising strategy to alleviate osteoarthritis (OA) progression. Method The potency of JD-312 in inducing chondrogenic differentiation of MSCs was assessed and verified. The efficacy of JD-312-treated MSCs was evaluated using a Sprague-Dawley rat DMM model. Additionally, the capacity of JD-312 to successfully recruit bone marrow-derived mesenchymal stem cells (BMSCs) for the treatment of OA in vitro was confirmed via intra-articular injection. The repair status of the articular cartilage was analyzed in vivo through histological examination. Result In this study, we identify JD-312 as a novel non-toxic small molecule that can promote chondrogenic differentiation in human umbilical cord-derived MSCs (hUCMSCs) and human bone marrow MSCS (hBMSCs) in vitro. We also show that transient differentiation of MSCs with JD-312 prior to in vivo administration remarkably improves the regeneration of cartilage and promotes Col2a1 and Acan expression in rat models of DMM, in comparison to kartogenin (KGN) pre-treatment or MSCs alone. Furthermore, direct intra-articular injection of JD-312 in murine model of OA showed reduced loss of articular cartilage and improved pain parameters. Lastly, we identified that the effects of JD-312 are at least in part mediated via upregulation of genes associated with the focal adhesion, PI3K-Akt signaling and the ECM-receptor interaction pathways, and specifically cartilage oligomeric matrix protein (COMP) may play a vital role. Conclusion Our study demonstrated that JD-312 showed encouraging repair effects for OA in vivo. The translational potential of this article Together, our findings demonstrate that JD-312 is a promising new therapeutic molecule for cartilage regeneration with clinical potential.
Collapse
Affiliation(s)
- Jingduo Gao
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, PR China
| | - Haixiang Pei
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, PR China
- Institute for Advanced Study, Shenzhen University and Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen, PR China
| | - Fang Lv
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, PR China
| | - Xin Niu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, PR China
| | - Yu You
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, PR China
| | - Liang He
- Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Tongji University School of Medicine, Shanghai, PR China
| | - Shijia Hu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, PR China
| | - Karan M. Shah
- Department of Oncology and Metabolism, The Medical School, The University of Sheffield, Sheffield, United Kingdom
| | - Mingyao Liu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, PR China
| | - Yihua Chen
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, PR China
| | - Bing Du
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, PR China
| | - Hai Xiong
- Institute for Advanced Study, Shenzhen University and Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen, PR China
| | - Jian Luo
- Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Tongji University School of Medicine, Shanghai, PR China
| |
Collapse
|
41
|
Chen Y, Cheng RJ, Wu Y, Huang D, Li Y, Liu Y. Advances in Stem Cell-Based Therapies in the Treatment of Osteoarthritis. Int J Mol Sci 2023; 25:394. [PMID: 38203565 PMCID: PMC10779279 DOI: 10.3390/ijms25010394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 12/16/2023] [Accepted: 12/21/2023] [Indexed: 01/12/2024] Open
Abstract
Osteoarthritis (OA) is a chronic, degenerative joint disease presenting a significant global health threat. While current therapeutic approaches primarily target symptom relief, their efficacy in repairing joint damage remains limited. Recent research has highlighted mesenchymal stem cells (MSCs) as potential contributors to cartilage repair, anti-inflammatory modulation, and immune regulation in OA patients. Notably, MSCs from different sources and their derivatives exhibit variations in their effectiveness in treating OA. Moreover, pretreatment and gene editing techniques of MSCs can enhance their therapeutic outcomes in OA. Additionally, the combination of novel biomaterials with MSCs has shown promise in facilitating the repair of damaged cartilage. This review summarizes recent studies on the role of MSCs in the treatment of OA, delving into their advantages and exploring potential directions for development, with the aim of providing fresh insights for future research in this critical field.
Collapse
Affiliation(s)
- Ye Chen
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu 610041, China; (Y.C.); (R.-J.C.); (Y.W.); (D.H.)
- Rare Diseases Center, West China Hospital, Sichuan University, Chengdu 610041, China
- Institute of Immunology and Inflammation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Chengdu 610041, China
| | - Rui-Juan Cheng
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu 610041, China; (Y.C.); (R.-J.C.); (Y.W.); (D.H.)
- Rare Diseases Center, West China Hospital, Sichuan University, Chengdu 610041, China
- Institute of Immunology and Inflammation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Chengdu 610041, China
| | - Yinlan Wu
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu 610041, China; (Y.C.); (R.-J.C.); (Y.W.); (D.H.)
- Rare Diseases Center, West China Hospital, Sichuan University, Chengdu 610041, China
- Institute of Immunology and Inflammation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Chengdu 610041, China
| | - Deying Huang
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu 610041, China; (Y.C.); (R.-J.C.); (Y.W.); (D.H.)
- Rare Diseases Center, West China Hospital, Sichuan University, Chengdu 610041, China
- Institute of Immunology and Inflammation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Chengdu 610041, China
| | - Yanhong Li
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu 610041, China; (Y.C.); (R.-J.C.); (Y.W.); (D.H.)
- Rare Diseases Center, West China Hospital, Sichuan University, Chengdu 610041, China
- Institute of Immunology and Inflammation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Chengdu 610041, China
| | - Yi Liu
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu 610041, China; (Y.C.); (R.-J.C.); (Y.W.); (D.H.)
- Rare Diseases Center, West China Hospital, Sichuan University, Chengdu 610041, China
- Institute of Immunology and Inflammation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Chengdu 610041, China
| |
Collapse
|
42
|
Oh E, Jang H, Ok S, Eom J, Lee H, Kim SH, Kim JH, Jeong YM, Kim KJ, Yun SP, Kwon HJ, Lee IC, Park JY, Yang S. WGA-M001, a Mixture of Total Extracts of Tagetes erecta and Ocimum basilicum, Synergistically Alleviates Cartilage Destruction by Inhibiting ERK and NF-κB Signaling. Int J Mol Sci 2023; 24:17459. [PMID: 38139287 PMCID: PMC10743532 DOI: 10.3390/ijms242417459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 12/11/2023] [Accepted: 12/12/2023] [Indexed: 12/24/2023] Open
Abstract
Tagetes erecta and Ocimum basilicum are medicinal plants that exhibit anti-inflammatory effects against various diseases. However, their individual and combined effects on osteoarthritis (OA) are unknown. Herein, we aimed to demonstrate the effects of T. erecta, O. basilicum, and their mixture, WGA-M001, on OA pathogenesis. The administration of total extracts of T. erecta and O. basilicum reduced cartilage degradation and inflammation without causing cytotoxicity. Although WGA-M001 contained lower concentrations of the individual extracts, it strongly inhibited the expression of pathogenic factors. In vivo OA studies also supported that WGA-M001 had protective effects against cartilage destruction at lower doses than those of T. erecta and O. basilicum. Moreover, its effects were stronger than those observed using Boswellia and Perna canaliculus. WGA-M001 effectively inhibited the interleukin (IL)-1β-induced nuclear factor kappa-light-chain-enhancer of the activated B cell (NF-κB) pathway and ERK phosphorylation. Furthermore, RNA-sequence analysis also showed that WGA-M001 decreased the expression of genes related to the IL-1β-induced NF-κB and ERK signaling pathways. Therefore, WGA-M001 is more effective than the single total extracts of T. erecta and O. basilicum in attenuating OA progression by regulating ERK and NF-κB signaling. Our results open new possibilities for WGA-M001 as a potential therapeutic agent for OA treatment.
Collapse
Affiliation(s)
- Eunjeong Oh
- Department of Biological Sciences, Sungkyunkwan University, Suwon 16419, Republic of Korea; (E.O.); (H.J.); (S.O.); (J.E.)
- Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon 16499, Republic of Korea
| | - Hahyeong Jang
- Department of Biological Sciences, Sungkyunkwan University, Suwon 16419, Republic of Korea; (E.O.); (H.J.); (S.O.); (J.E.)
| | - Subin Ok
- Department of Biological Sciences, Sungkyunkwan University, Suwon 16419, Republic of Korea; (E.O.); (H.J.); (S.O.); (J.E.)
| | - Jiwon Eom
- Department of Biological Sciences, Sungkyunkwan University, Suwon 16419, Republic of Korea; (E.O.); (H.J.); (S.O.); (J.E.)
| | - Hyunyong Lee
- Wooree Green Science, Ansan 15409, Republic of Korea; (H.L.); (S.H.K.); (J.H.K.); (Y.M.J.); (K.J.K.)
| | - Sung Hun Kim
- Wooree Green Science, Ansan 15409, Republic of Korea; (H.L.); (S.H.K.); (J.H.K.); (Y.M.J.); (K.J.K.)
| | - Jong Hwa Kim
- Wooree Green Science, Ansan 15409, Republic of Korea; (H.L.); (S.H.K.); (J.H.K.); (Y.M.J.); (K.J.K.)
| | - Yu Mi Jeong
- Wooree Green Science, Ansan 15409, Republic of Korea; (H.L.); (S.H.K.); (J.H.K.); (Y.M.J.); (K.J.K.)
| | - Kyeong Jin Kim
- Wooree Green Science, Ansan 15409, Republic of Korea; (H.L.); (S.H.K.); (J.H.K.); (Y.M.J.); (K.J.K.)
- Department of Horticulture, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Seung Pil Yun
- Department of Pharmacology, Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju 52727, Republic of Korea;
| | - Hyung-Jun Kwon
- Center for Companion Animal New Drug Development, Jeonbuk Branch, Korea Institute of Toxicology, Jeongeup 53212, Republic of Korea; (H.-J.K.); (I.-C.L.); (J.-Y.P.)
| | - In-Chul Lee
- Center for Companion Animal New Drug Development, Jeonbuk Branch, Korea Institute of Toxicology, Jeongeup 53212, Republic of Korea; (H.-J.K.); (I.-C.L.); (J.-Y.P.)
| | - Ji-Young Park
- Center for Companion Animal New Drug Development, Jeonbuk Branch, Korea Institute of Toxicology, Jeongeup 53212, Republic of Korea; (H.-J.K.); (I.-C.L.); (J.-Y.P.)
| | - Siyoung Yang
- Department of Biological Sciences, Sungkyunkwan University, Suwon 16419, Republic of Korea; (E.O.); (H.J.); (S.O.); (J.E.)
| |
Collapse
|
43
|
Vyawahare A, Ansari MM, Kumar A, Ahmad A, Mishra RK, Jori C, Nadeem A, Siddiqui N, Raza SS, Khan R. Enzyme targeted delivery of sivelestat loaded nanomicelle inhibits arthritic severity in experimental arthritis. Life Sci 2023; 334:122206. [PMID: 37879159 DOI: 10.1016/j.lfs.2023.122206] [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: 08/10/2023] [Revised: 10/21/2023] [Accepted: 10/22/2023] [Indexed: 10/27/2023]
Abstract
AIMS Rheumatoid arthritis (RA) is chronic inflammatory disorder mainly affects the lining of articular cartilage of synovial joints characterized by severe inflammation and joint damage. The expression of proteolytic enzymes like MMP-2 and Neutrophil Elastase (NE) worsens the RA condition. To address this concern, we have synthesized dual enzyme targeted chlorotoxin conjugated nanomicelles loaded with sivelestat as broad spectrum treatment for RA. MATERIALS AND METHODS Conjugation of the chlorotoxin over nanomicelle and incorporation of sivelestat in nanomicelle provide it dual targeting potential. The sivelestat loaded nanomicelle (SLM) evaluated for the drug release and in-vitro cytocompatibility. Further, investigated its in-vivo anti-arthritic potential on collagen-induced arthritis in wistar rats. KEY FINDINGS The microscopic observation of SLM showed spherical ball like appearance with size ranging from 190 to 230 nm. SLM showed good drug loading and encapsulation efficiency along with no cytotoxicity against healthy cell lines. In-vivo therapeutic assessment on collagen induced arthritis rat model showed potential chondroprotection. The microscopic visualization of articular cartilage by staining showed that it restores the cartilage integrity and lowers the expression of pro-inflammatory enzymes showed by Immunohistochemistry and Immunofluorescence. We observed that, it restrain the mediators of synovial inflammation by simultaneous inhibition of the proteolytic enzymes involved in swelling, cartilage destruction and joint damage which provides strong chondroprotection. SIGNIFICANCE We report that significant alleviation of inflammation and inhibition of proteolytic enzymes together might provide enhanced potential for the treatment and management of RA.
Collapse
Affiliation(s)
- Akshay Vyawahare
- Department of Chemical Biology, Institute of Nano Science and Technology, Sector 81, Knowledge city, Sahibzada Ajit Singh Nagar, Mohali, Punjab 140306, India
| | - Md Meraj Ansari
- Centre for Pharmaceutical Nanotechnology, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, S.A.S Nagar, Sector 67, Mohali, Punjab 160062, India
| | - Ajay Kumar
- Department of Chemical Biology, Institute of Nano Science and Technology, Sector 81, Knowledge city, Sahibzada Ajit Singh Nagar, Mohali, Punjab 140306, India
| | - Anas Ahmad
- Julia McFarlane Diabetes Research Centre (JMDRC) and Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases and Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - Rakesh Kumar Mishra
- Department of Chemical Biology, Institute of Nano Science and Technology, Sector 81, Knowledge city, Sahibzada Ajit Singh Nagar, Mohali, Punjab 140306, India
| | - Chandrashekhar Jori
- Department of Chemical Biology, Institute of Nano Science and Technology, Sector 81, Knowledge city, Sahibzada Ajit Singh Nagar, Mohali, Punjab 140306, India
| | - Ahmed Nadeem
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Nahid Siddiqui
- Amity Institute of Biotechnology, Amity University, Noida 201303, India
| | - Syed Shadab Raza
- Laboratory for Stem Cell & Restorative Neurology, Department of Biotechnology, Era's Lucknow Medical College and Hospital, Sarfarazganj, Lucknow 226003, Uttar Pradesh, India
| | - Rehan Khan
- Department of Chemical Biology, Institute of Nano Science and Technology, Sector 81, Knowledge city, Sahibzada Ajit Singh Nagar, Mohali, Punjab 140306, India.
| |
Collapse
|
44
|
Xu W, Zhang B, Xi C, Qin Y, Lin X, Wang B, Kong P, Yan J. Ferroptosis Plays a Role in Human Chondrocyte of Osteoarthritis Induced by IL-1β In Vitro. Cartilage 2023; 14:455-466. [PMID: 36786219 PMCID: PMC10807732 DOI: 10.1177/19476035221142011] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 11/09/2022] [Accepted: 11/09/2022] [Indexed: 02/15/2023] Open
Abstract
OBJECTIVE Osteoarthritis (OA) is a common disease with complex and unclear pathogenesis. Ferroptosis is a new cell death mode, which is proved to be involved in different kinds of disease. We hypothesized that ferroptosis contributes to the progress of human OA. DESIGN Chondrocytes were extracted from waste cartilage of total knee arthroplasty, and stimulated with interleukin-1β (IL-1β). Then, we detected the morphology, proliferation, and viability, and levels of Fe3+, glutathione (GSH), reactive oxygen species (ROS), malondialdehyde (MDA), and 5 proteins related to ferroptosis with or without intervention of ferrostatin-1 (Fer-1). In addition, we compared the effect of Fer-1 and liproxstatin-1 (Lip-1) on ferroptosis and the protection of chondrocytes by detecting several markers of both ferroptosis and OA. RESULTS After stimulation of IL-1β, there were significant changes on the shape of chondrocyte, with lower viability and proliferation. There was accumulation of intracellular Fe3+, GSH, ROS, and MDA, with the changes of expression of 5 ferroptosis-related proteins. With the contribution of Fer-1, results above were reversed. Moreover, there was no significant difference in GPX4 and ACSL4 between Fer-1 and Lip-1 group. However, the expression of COLX, ADAMTS5, and MMP-13 are lower after the treatment of Fer-1 compared with Lip-1. CONCLUSIONS Ferroptosis plays an important role in human OA chondrocytes, which can be reversed by Fer-1, illustrating that inhibitor of ferroptosis may be a potential treatment of OA. Moreover, Lip-1 and Fer-1 can both alleviate the level of ferroptosis in OA chondrocytes, but Fer-1 had a more protective effect.
Collapse
Affiliation(s)
- Wenbo Xu
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Bowen Zhang
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Chunyang Xi
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yong Qin
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xin Lin
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Bo Wang
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Pengyu Kong
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jinglong Yan
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
45
|
Chun JM, Nam H, Lee JH, Seo YH, Kim HS, Moon BC, Park JH. Chondroprotective effects of Protaetia brevitarsis seulensis larvae as an edible insect on osteoarthritis in mice. Food Sci Nutr 2023; 11:7887-7899. [PMID: 38107146 PMCID: PMC10724628 DOI: 10.1002/fsn3.3706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 09/05/2023] [Accepted: 09/08/2023] [Indexed: 12/19/2023] Open
Abstract
Osteoarthritis (OA) is a common chronic joint inflammatory disease characterized by progressive destruction of the articular cartilage, bone remodeling, and excessive chronic pain. Most therapeutic approaches do not rescue the progression of OA effectively or provide relief of symptoms. Protaetia brevitarsis seulensis larva (PBSL), which is attracting attention, is an edible insect with very high nutritional value and herbal medicine for the treatment of blood stasis, hepatic disease, and various inflammatory diseases. However, the effect of PBSL on OA has not yet been investigated. This study aimed to demonstrate the effects of PBSL water extract on the progression of OA using monosodium iodoacetate (MIA)-induced mice and SW1353 chondrocytes or murine macrophages. We injected MIA into the intraarticular area of mice following pretreatment with either saline or PBSL (200 mg/kg) for 2 weeks, and then locomotor activity, microcomputed tomography and histopathological analysis, quantitative reverse transcriptase-polymerase chain reaction analysis, and western blot analysis were performed. To determine the molecular effects of PBSL, we used interleukin-1β (IL-1β)-induced SW1353 chondrosarcoma or lipopolysaccharide (LPS)-stimulated macrophages. Pretreatment with PBSL diminished the symptoms of OA. Physical activity, articular cartilage damage, and the generation of microfractures were rescued by pretreatment with PBSL in the mouse model. Pretreatment with PBSL suppressed the progress of OA through the regulation of articular cartilage degradation genes and inflammation in both in vivo and in vitro models. Our results demonstrated that PBSL has value as edible insect that can be used in the development of functional foods for OA.
Collapse
Affiliation(s)
- Jin Mi Chun
- Herbal Medicine Resources Research CenterKorea Institute of Oriental MedicineNaju‐siRepublic of Korea
| | - Hyeon‐Hwa Nam
- Herbal Medicine Resources Research CenterKorea Institute of Oriental MedicineNaju‐siRepublic of Korea
| | - Ji Hye Lee
- Herbal Medicine Resources Research CenterKorea Institute of Oriental MedicineNaju‐siRepublic of Korea
- School of Korean MedicinePusan National UniversityBusan‐siRepublic of Korea
| | - Young Hye Seo
- Herbal Medicine Resources Research CenterKorea Institute of Oriental MedicineNaju‐siRepublic of Korea
| | - Hyo Seon Kim
- Herbal Medicine Resources Research CenterKorea Institute of Oriental MedicineNaju‐siRepublic of Korea
| | - Byeong Cheol Moon
- Herbal Medicine Resources Research CenterKorea Institute of Oriental MedicineNaju‐siRepublic of Korea
| | - Jun Hong Park
- Herbal Medicine Resources Research CenterKorea Institute of Oriental MedicineNaju‐siRepublic of Korea
| |
Collapse
|
46
|
Reina-Mahecha A, Beers MJ, van der Veen HC, Zuhorn IS, van Kooten TG, Sharma PK. A Review of the Role of Bioreactors for iPSCs-Based Tissue-Engineered Articular Cartilage. Tissue Eng Regen Med 2023; 20:1041-1052. [PMID: 37861960 PMCID: PMC10645985 DOI: 10.1007/s13770-023-00573-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 07/04/2023] [Accepted: 07/05/2023] [Indexed: 10/21/2023] Open
Abstract
BACKGROUND Osteoarthritis (OA) is the most common degenerative joint disease without an ultimate treatment. In a search for novel approaches, tissue engineering (TE) has shown great potential to be an effective way for hyaline cartilage regeneration and repair in advanced stages of OA. Recently, induced pluripotent stem cells (iPSCs) have been appointed to be essential stem cells for degenerative disease treatment because they allow a personalized medicine approach. For clinical translation, bioreactors in combination with iPSCs-engineerd cartilage could match patients needs, serve as platform for large-scale patient specific cartilage production, and be a tool for patient OA modelling and drug screening. Furthermore, to minimize in vivo experiments and improve cell differentiation and cartilage extracellular matrix (ECM) deposition, TE combines existing approaches with bioreactors. METHODS This review summarizes the current understanding of bioreactors and the necessary parameters when they are intended for cartilage TE, focusing on the potential use of iPSCs. RESULTS Bioreactors intended for cartilage TE must resemble the joint cavity niche. However, recreating human synovial joints is not trivial because the interactions between various stimuli are not entirely understood. CONCLUSION The use of mechanical and electrical stimulation to differentiate iPSCs, and maintain and test chondrocytes are key stimuli influencing hyaline cartilage homeostasis. Incorporating these stimuli to bioreactors can positively impact cartilage TE approaches and their possibility for posterior translation into the clinics.
Collapse
Affiliation(s)
- Alejandro Reina-Mahecha
- Department of Biomedical Engineering, University of Groningen, University Medical Center Groningen, FB40, Antonius Deusinglaan -1, 9713AV, Groningen, The Netherlands
| | - Martine J Beers
- Department of Orthopedics, University Medical Center Groningen, Groningen, The Netherlands
| | - Hugo C van der Veen
- Department of Orthopedics, University Medical Center Groningen, Groningen, The Netherlands
| | - Inge S Zuhorn
- Department of Biomedical Engineering, University of Groningen, University Medical Center Groningen, FB40, Antonius Deusinglaan -1, 9713AV, Groningen, The Netherlands
| | - Theo G van Kooten
- Department of Biomedical Engineering, University of Groningen, University Medical Center Groningen, FB40, Antonius Deusinglaan -1, 9713AV, Groningen, The Netherlands
| | - Prashant K Sharma
- Department of Biomedical Engineering, University of Groningen, University Medical Center Groningen, FB40, Antonius Deusinglaan -1, 9713AV, Groningen, The Netherlands.
| |
Collapse
|
47
|
Pontes AP, van der Wal S, Ranamalla SR, Roelofs K, Tomuta I, Creemers LB, Rip J. Cell uptake and intracellular trafficking of bioreducible poly(amidoamine) nanoparticles for efficient mRNA translation in chondrocytes. Front Bioeng Biotechnol 2023; 11:1290871. [PMID: 38026902 PMCID: PMC10668025 DOI: 10.3389/fbioe.2023.1290871] [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: 09/08/2023] [Accepted: 10/20/2023] [Indexed: 12/01/2023] Open
Abstract
Disulfide-containing poly(amidoamine) (PAA) is a cationic and bioreducible polymer, with potential use as a nanocarrier for mRNA delivery in the treatment of several diseases including osteoarthritis (OA). Successful transfection of joint cells with PAA-based nanoparticles (NPs) was shown previously, but cell uptake, endosomal escape and nanoparticle biodegradation were not studied in detail. In this study, C28/I2 human chondrocytes were transfected with NPs co-formulated with a PEG-polymer coating and loaded with EGFP mRNA for confocal imaging of intracellular trafficking and evaluation of transfection efficiency. Compared with uncoated NPs, PEG-coated NPs showed smaller particle size, neutral surface charge, higher colloidal stability and superior transfection efficiency. Furthermore, endosomal entrapment of these PEG-coated NPs decreased over time and mRNA release could be visualized both in vitro and in live cells. Importantly, cell treatment with modulators of the intracellular reducing environment showed that glutathione (GSH) concentrations affect translation of the mRNA payload. Finally, we applied a D-optimal experimental design to test different polymer-to-RNA loading ratios and dosages, thus obtaining an optimal formulation with up to ≈80% of GFP-positive cells and without toxic effects. Together, the biocompatibility and high transfection efficiency of this system may be a promising tool for intra-articular delivery of therapeutical mRNA in OA treatment.
Collapse
Affiliation(s)
| | | | - Saketh R. Ranamalla
- Department of Pharmaceutical Technology and Biopharmacy, University of Medicine and Pharmacy “Iuliu Hațieganu”, Cluj-Napoca, Romania
| | | | - Ioan Tomuta
- Department of Pharmaceutical Technology and Biopharmacy, University of Medicine and Pharmacy “Iuliu Hațieganu”, Cluj-Napoca, Romania
| | - Laura B. Creemers
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, Netherlands
| | - Jaap Rip
- 20Med Therapeutics BV, Leiden, Netherlands
| |
Collapse
|
48
|
Dong L, Zhao Y, Sun C, Ou Yang Z, Chen F, Hu W, Zhang H, Wang Y, Zhu R, Cheng Y, Chen Y, Li S, Wang K, Ding C, Zhou R, Hu W. ASIC1a-CMPK2-mediated M1 macrophage polarization exacerbates chondrocyte senescence in osteoarthritis through IL-18. Int Immunopharmacol 2023; 124:110878. [PMID: 37660594 DOI: 10.1016/j.intimp.2023.110878] [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: 01/30/2023] [Revised: 08/25/2023] [Accepted: 08/28/2023] [Indexed: 09/05/2023]
Abstract
PURPOSE Identification of a role for, and the mechanism of action of, the acid-sensing ion channel 1a (ASIC1a) in M1 macrophage polarization, which results in osteoarthritis (OA)-associated chondrocyte senescence. METHOD ASIC1a expression in synovial M1 macrophages of OA patients was assessed by immunofluorescence. A role for ASIC1a in M1 macrophage and chondrocyte senescence was assessed in a mouse OA model. RESULTS ASIC1a expression was found to be upregulated in synovial M1 macrophages of OA patients. Extracellular acidification (pH 6.0) promoted M1 polarization of bone marrow derived macrophages (BMDMs), which was reversed by PcTx-1 or ASIC1a-siRNA. RNA-seq transcriptome results demonstrated a downregulation of M1 macrophage-associated genes in BMDMs after PcTx-1 treatment. Mechanistically, a role for the ASIC1a-cytidine/uridine monophosphate kinase 2 (CMPK2) axis in M1 macrophage polarization was demonstrated. The concentration of IL-18 was elevated in synovial fluid and supernatants of acid-activated BMDMs. In vitro, IL-18 stimulation or co-culture with acid-activated macrophages promoted chondrocyte senescence. In vivo, intra-articular administration of PcTx-1 reduced articular cartilage destruction and chondrocytes senescence in OA mice, which related to reduced numbers of M1 macrophages and IL-18 in affected joints. CONCLUSION These results demonstrate a novel pathogenic process that results in OA cartilage damage, in which M1 macrophage derived IL-18 induces articular chondrocytes senescence. Further, the ASIC1a-CMPK2 axis was shown to positively regulate M1 macrophage polarization. Hence, ASIC1a is a promising treatment target for M1 macrophage-mediated diseases, such as OA.
Collapse
Affiliation(s)
- Lei Dong
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China; The Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Yingjie Zhao
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China; The Key Laboratory of Anti-inflammatory and Immune Medicine, Anhui Medical University, Ministry of Education, Hefei 230032, China
| | - Cheng Sun
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China; The Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Ziwei Ou Yang
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China; The Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Fan Chen
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China; The Key Laboratory of Anti-inflammatory and Immune Medicine, Anhui Medical University, Ministry of Education, Hefei 230032, China
| | - Weirong Hu
- The Key Laboratory of Anti-inflammatory and Immune Medicine, Anhui Medical University, Ministry of Education, Hefei 230032, China
| | - Hailin Zhang
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China; The Key Laboratory of Anti-inflammatory and Immune Medicine, Anhui Medical University, Ministry of Education, Hefei 230032, China
| | - Yan Wang
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China; The Key Laboratory of Anti-inflammatory and Immune Medicine, Anhui Medical University, Ministry of Education, Hefei 230032, China
| | - Rendi Zhu
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China; The Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Yuanzhi Cheng
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China; The Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Yong Chen
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Shufang Li
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Ke Wang
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China; The Key Laboratory of Anti-inflammatory and Immune Medicine, Anhui Medical University, Ministry of Education, Hefei 230032, China
| | - Changhai Ding
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China; Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Renpeng Zhou
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China; The Key Laboratory of Anti-inflammatory and Immune Medicine, Anhui Medical University, Ministry of Education, Hefei 230032, China.
| | - Wei Hu
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China; The Key Laboratory of Anti-inflammatory and Immune Medicine, Anhui Medical University, Ministry of Education, Hefei 230032, China; The Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China.
| |
Collapse
|
49
|
Xia W, Xiao J, Tong C, Lu J, Tu Y, Li S, Ni L, Shi Y, Luo P, Zhang X, Wang X. Orientin inhibits inflammation in chondrocytes and attenuates osteoarthritis through Nrf2/NF-κB and SIRT6/NF-κB pathway. J Orthop Res 2023; 41:2405-2417. [PMID: 37186383 DOI: 10.1002/jor.25573] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 03/04/2023] [Accepted: 04/17/2023] [Indexed: 05/17/2023]
Abstract
Effects of Orientin on murine chondrocytes treated with interleukin-1β (IL-1β) were evaluated using qPCR, western blot analysis, ELISA, and immunofluorescent staining in vitro. In vivo, We established a standard OA model by performing the destabilized medial meniscus (DMM) surgery on C57BL/6 mice, and assessed healing effect of Orientin by X-ray imaging, histopathological analysis, immunohistochemical staining. Osteoarthritis (OA) is the most common form of degenerative joint disease in clinic and the chondrocyte inflammation plays the most important role in OA development. The natural flavonoid compound (Orientin) has anti-inflammatory bioactive properties in the treatment of various diseases. But studies have not explored whether Orientin modulates OA progression. In this study, a significant suppression in IL-1β-mediated pro-inflammatory mediators and the degradation of cartilage extracellular matrix (ECM) was observed in vitro through qPCR, western blot analysis, ELISA, and immunofluorescent staining after the treatment with Orientin. In addition, Orientin abrogated DMM surgery induced cartilage degradation in mice, which was assessed by X-ray imaging, histopathological analysis, immunohistochemical staining. Mechanistic studies showed that Orientin suppressed OA development by downregulating activation of NF-κB by activating Nrf2/HO-1 axis and SIRT6 signaling pathway. These results provide evidence that Orientin serves as a potentially viable compound for the treatment of OA.
Collapse
Affiliation(s)
- Weiyi Xia
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Jian Xiao
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - ChengLin Tong
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Jiajie Lu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Yurong Tu
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Sunlong Li
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Libing Ni
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Yifeng Shi
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Peng Luo
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Xiaolei Zhang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Xiangyang Wang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
50
|
Ng JQ, Jafarov TH, Little CB, Wang T, Ali AM, Ma Y, Radford GA, Vrbanac L, Ichinose M, Whittle S, Hunter DJ, Lannagan TRM, Suzuki N, Goyne JM, Kobayashi H, Wang TC, Haynes DR, Menicanin D, Gronthos S, Worthley DL, Woods SL, Mukherjee S. Loss of Grem1-lineage chondrogenic progenitor cells causes osteoarthritis. Nat Commun 2023; 14:6909. [PMID: 37907525 PMCID: PMC10618187 DOI: 10.1038/s41467-023-42199-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Accepted: 10/03/2023] [Indexed: 11/02/2023] Open
Abstract
Osteoarthritis (OA) is characterised by an irreversible degeneration of articular cartilage. Here we show that the BMP-antagonist Gremlin 1 (Grem1) marks a bipotent chondrogenic and osteogenic progenitor cell population within the articular surface. Notably, these progenitors are depleted by injury-induced OA and increasing age. OA is also caused by ablation of Grem1 cells in mice. Transcriptomic and functional analysis in mice found that articular surface Grem1-lineage cells are dependent on Foxo1 and ablation of Foxo1 in Grem1-lineage cells caused OA. FGFR3 signalling was confirmed as a promising therapeutic pathway by administration of pathway activator, FGF18, resulting in Grem1-lineage chondrocyte progenitor cell proliferation, increased cartilage thickness and reduced OA. These findings suggest that OA, in part, is caused by mechanical, developmental or age-related attrition of Grem1 expressing articular cartilage progenitor cells. These cells, and the FGFR3 signalling pathway that sustains them, may be effective future targets for biological management of OA.
Collapse
Affiliation(s)
- Jia Q Ng
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Toghrul H Jafarov
- Department of Medicine, Columbia University Medical Center, New York, NY, USA
| | - Christopher B Little
- Raymond Purves Bone & Joint Research Laboratories, Kolling Institute, University of Sydney Faculty of Medicine and Health, Royal North Shore Hospital, St. Leonards, NSW, Australia
| | - Tongtong Wang
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
- Precision Cancer Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| | - Abdullah M Ali
- Department of Medicine, Columbia University Medical Center, New York, NY, USA
| | - Yan Ma
- Department of Medicine, Columbia University Medical Center, New York, NY, USA
| | - Georgette A Radford
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Laura Vrbanac
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Mari Ichinose
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Samuel Whittle
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
- Rheumatology Unit, The Queen Elizabeth Hospital, Woodville South, SA, Australia
| | - David J Hunter
- Northern Clinical School, University of Sydney, St. Leonards, Sydney, NSW, Australia
| | - Tamsin R M Lannagan
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Nobumi Suzuki
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Jarrad M Goyne
- Precision Cancer Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| | - Hiroki Kobayashi
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Timothy C Wang
- Department of Medicine and Irving Cancer Research Center, Columbia University, New York, NY, USA
| | - David R Haynes
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Danijela Menicanin
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Stan Gronthos
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
- School of Biomedicine, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Daniel L Worthley
- Precision Cancer Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia.
- Colonoscopy Clinic, Brisbane, QLD, Australia.
| | - Susan L Woods
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia.
- Precision Cancer Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia.
| | | |
Collapse
|