1
|
Liao S, Liu Z, Lv W, Li S, Tian T, Wang Y, Wu H, Zhao ZH, Lin Y. Efficient Delivery of siRNA via Tetrahedral Framework Nucleic Acids: Inflammation Attenuation and Matrix Regeneration in Temporomandibular Joint Osteoarthritis. ACS APPLIED MATERIALS & INTERFACES 2024; 16:53499-53514. [PMID: 39330704 DOI: 10.1021/acsami.4c11089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/28/2024]
Abstract
Temporomandibular joint osteoarthritis (TMJOA) is the most common and severe subtype of temporomandibular disease characterized by inflammation and cartilage matrix degradation. Compared with traditional conservative treatment, small interfering RNAs (siRNAs) have emerged as a more efficient gene-targeted therapeutic tool for TMJOA treatment. Nuclear factor kappaB (NF-κB) is a transcription factor orchestrating the inflammatory processes in the pathogenesis of TMJOA. Employing siRNA-NF-κB could theoretically control the development of TMJOA. However, the clinical applications of siRNA-NF-κB are limited by its structural instability, poor cellular uptake, and short TMJ retention. To overcome these shortcomings, we developed a tetrahedral framework nucleic acid (tFNA) system carrying siRNA-NF-κB, named Tsi. The results indicated that Tsi exhibited excellent structural stability and excellent cellular uptake efficiency. It also demonstrated a superior NF-κB silencing effect over siRNA alone, attenuating the activation of NF-κB and upregulating the NRF2/HO-1 pathway. This system effectively reduced the release of inflammatory factors and reactive oxygen species (ROS), inhibiting cellular oxidative stress and apoptosis. In vivo, Tsi displayed enhanced TMJ retention capacity in comparison to siRNA alone and offered significant protective effects on both the cartilage matrix and subchondral bone, presenting a promising approach for TMJOA treatment.
Collapse
Affiliation(s)
- Shengnan Liao
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
- Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Zhiqiang Liu
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Weitong Lv
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Songhang Li
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Taoran Tian
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Yifan Wang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
- Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Haoyan Wu
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Zhi-He Zhao
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
- Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Yunfeng Lin
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
- Sichuan Provincial Engineering Research Center of Oral Biomaterials, Chengdu 610041, Sichuan, China
- National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
2
|
Zhao H, Wang T, Fang X, Xu T, Li J, Jing S, Chen G, Liu Y, Sheng G. 2D MXene Nanosheets with ROS Scavenging Ability Effectively Delay Osteoarthritis Progression. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:1572. [PMID: 39404298 PMCID: PMC11478061 DOI: 10.3390/nano14191572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 08/29/2024] [Accepted: 09/23/2024] [Indexed: 10/19/2024]
Abstract
MXenes nanosheets with high conductivity, hydrophilicity, and excellent reactive oxygen species (ROS) scavenging ability have shown promise in treating various degenerative diseases correlated with abnormal ROS accumulation. Herein, the therapeutic potential of Ti3C2Tx nanosheets, which is the most widely investigated MXene material, in delaying osteoarthritis (OA) progression is demonstrated. In vitro experiments indicate the strong ROS scavenging capacity of Ti3C2Tx nanosheets and their acceptable biocompatibility. Ti3C2Tx nanosheets effectively protect chondrocytes from cell death induced by oxidative stress. In addition, Ti3C2Tx nanosheets demonstrate a prominent anti-inflammatory effect and the ability to restore homeostasis between anabolic activities and catabolic activities in chondrocytes. Furthermore, RNA sequencing reveals the potential mechanism underlying the Ti3C2Tx nanosheet-mediated therapeutic effect. Finally, the in vivo curative effect of Ti3C2Tx nanosheets is verified using a rat OA model. Histological staining and immunohistochemical analyses indicate that Ti3C2Tx nanosheets effectively ameliorate OA progression. Conclusively, the in vitro and in vivo experiments suggest that Ti3C2Tx nanosheets could be a promising and effective option for OA treatment.
Collapse
Affiliation(s)
- Hongqi Zhao
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Tianqi Wang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xuan Fang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Tao Xu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jian Li
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences Tongji Shanxi Hospital, Taiyuan 030032, China
| | - Shaoze Jing
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences Tongji Shanxi Hospital, Taiyuan 030032, China
| | - Guangzi Chen
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yang Liu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Gaohong Sheng
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
3
|
Fang X, Zhao H, Xu T, Wu H, Sheng G. Anti-Inflammatory and Antioxidant Effects of Irigenen Alleviate Osteoarthritis Progression through Nrf2/HO-1 Pathway. Pharmaceuticals (Basel) 2024; 17:1268. [PMID: 39458910 PMCID: PMC11510601 DOI: 10.3390/ph17101268] [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: 08/05/2024] [Revised: 09/07/2024] [Accepted: 09/18/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND/OBJECTIVES Osteoarthritis (OA) is a prevalent degenerative disease globally, characterized by cartilage degradation and joint dysfunction. Current treatments are insufficient for halting OA progression. Irigenin (IRI), a flavonoid extracted from natural plants with anti-inflammatory and antioxidant properties, has demonstrated potential in mitigating inflammation and oxidative stress in various diseases; however, its effects on OA remain unexplored. This study aims to evaluate the therapeutic effects of IRI on OA through in vivo and in vitro experiments and to elucidate the underlying molecular mechanisms. METHODS In vitro, chondrocytes were exposed to hydrogen peroxide (H2O2) to induce an oxidative stress environment and were then treated with IRI. Western blotting, RT-qPCR, immunofluorescence staining assays, flow cytometry, and apoptosis assays were employed to assess the effects of IRI on chondrocyte matrix homeostasis, inflammatory response, and apoptosis. In vivo, an OA rat model was treated with regular IRI injections, and therapeutic effects were evaluated using micro-CT, histological staining, and immunohistochemistry assays. RESULTS IRI treatment restored matrix homeostasis in chondrocytes and effectively suppressed H2O2-induced inflammation and apoptosis. Subsequent studies further revealed that IRI exerts its therapeutic effects by activating the Nrf2/HO-1 pathway. Inhibition of Nrf2 expression in chondrocytes partially blocked the anti-inflammatory and antioxidant effects of IRI. In the OA rat model, regular IRI injections effectively ameliorated cartilage degeneration. CONCLUSIONS This study identifies IRI as a promising strategy for OA treatment by modulating inflammation and apoptosis through the Nrf2/HO-1 pathway.
Collapse
Affiliation(s)
| | | | | | | | - Gaohong Sheng
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China; (X.F.); (H.Z.); (T.X.); (H.W.)
| |
Collapse
|
4
|
Zou Y, Liu C, Wang Z, Li G, Xiao J. Neural and immune roles in osteoarthritis pain: Mechanisms and intervention strategies. J Orthop Translat 2024; 48:123-132. [PMID: 39220678 PMCID: PMC11363721 DOI: 10.1016/j.jot.2024.07.010] [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: 04/14/2024] [Revised: 07/18/2024] [Accepted: 07/24/2024] [Indexed: 09/04/2024] Open
Abstract
Pain is the leading symptom for most individuals with osteoarthritis (OA), a complex condition marked by joint discomfort. Recently, the dynamic interplay between the nervous and immune systems has become a focal point for understanding pain regulation. Despite this, there is still a substantial gap in our comprehensive understanding of the neuroimmune interactions and their effects on pain in OA. This review examines the bidirectional influences between immune cells and nerves in OA progression. It explores current approaches that target neuroimmune pathways, including promoting M2 macrophage polarization and specific neuronal receptor targeting, for effective pain reduction. Translational potential statement This review provides a comprehensive overview of the mechanisms underlying the interplay between the immune system and nervous system during the progression of OA, as well as their contributions to pain. Additionally, it compiles existing intervention strategies targeting neuroimmunity for the treatment of OA pain. This information offers valuable insights for researchers seeking to address the challenge of OA pain.
Collapse
Affiliation(s)
- Yi Zou
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, Hubei, 430030, China
| | - Changyu Liu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, Hubei, 430030, China
| | - Zhenggang Wang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, Hubei, 430030, China
| | - Guanghui Li
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, Hubei, 430030, China
| | - Jun Xiao
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, Hubei, 430030, China
| |
Collapse
|
5
|
Li Z, Cheng Q, Lin L, Fu X, Wang Y. Plasma Membrane-Derived Biomimetic Apoptotic Nanovesicles Targeting Inflammation and Cartilage Degeneration for Osteoarthritis. SMALL METHODS 2024:e2400660. [PMID: 39036830 DOI: 10.1002/smtd.202400660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 06/14/2024] [Indexed: 07/23/2024]
Abstract
Osteoarthritis (OA) is a degenerative whole-joint disease in which the synovium and joint cartilage become inflamed and damaged. The essential role of inflammation in the development of OA has been recognized recently. Accordingly, simultaneous regulation of local inflammation and tissue degeneration is proposed as a promising therapeutic strategy. Herein, multifunctional biomimetic apoptotic nanovesicles (Apo-NVs) are constructed with plasma membrane derived from apoptotic T cells. The anti-inflammatory microRNA-124 is further encapsulated into Apo-NVs in the hope of achieving an enhanced immunomodulatory effect. It is found that apoptotic nanovesicles, including Apo-NVs and Apo-NVs-miR-124, both efficiently promote the M2 repolarization of M1 macrophages and inhibit the degenerative phenotype of chondrocytes. Further in vivo studies show that Apo-NVs and Apo-NVs-miR-124 alleviate synovial inflammation and protect cartilage tissue from degeneration in OA mice. The study highlights the potential of Apo-NVs in treating OA and other inflammation-related diseases.
Collapse
Affiliation(s)
- Zongyi Li
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510641, China
- National Engineering Research Center for Tissue Restoration and Reconstruction and Innovation Center for Tissue Restoration and Reconstruction, Guangzhou, 510006, China
- Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou, 510006, China
| | - Quhan Cheng
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Luoyao Lin
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510641, China
- National Engineering Research Center for Tissue Restoration and Reconstruction and Innovation Center for Tissue Restoration and Reconstruction, Guangzhou, 510006, China
- Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou, 510006, China
| | - Xiaoling Fu
- National Engineering Research Center for Tissue Restoration and Reconstruction and Innovation Center for Tissue Restoration and Reconstruction, Guangzhou, 510006, China
- Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou, 510006, China
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, China
| | - Yingjun Wang
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510641, China
- National Engineering Research Center for Tissue Restoration and Reconstruction and Innovation Center for Tissue Restoration and Reconstruction, Guangzhou, 510006, China
- Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou, 510006, China
| |
Collapse
|
6
|
Yi X, Leng P, Wang S, Liu L, Xie B. Functional Nanomaterials for the Treatment of Osteoarthritis. Int J Nanomedicine 2024; 19:6731-6756. [PMID: 38979531 PMCID: PMC11230134 DOI: 10.2147/ijn.s465243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 06/15/2024] [Indexed: 07/10/2024] Open
Abstract
Osteoarthritis (OA) is the most common degenerative joint disease, affecting more than 595 million people worldwide. Nanomaterials possess superior physicochemical properties and can influence pathological processes due to their unique structural features, such as size, surface interface, and photoelectromagnetic thermal effects. Unlike traditional OA treatments, which suffer from short half-life, low stability, poor bioavailability, and high systemic toxicity, nanotherapeutic strategies for OA offer longer half-life, enhanced targeting, improved bioavailability, and reduced systemic toxicity. These advantages effectively address the limitations of traditional therapies. This review aims to inspire researchers to develop more multifunctional nanomaterials and promote their practical application in OA treatment.
Collapse
Affiliation(s)
- Xinyue Yi
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, People’s Republic of China
- Clinical Medical College, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, People’s Republic of China
| | - Pengyuan Leng
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, People’s Republic of China
| | - Supeng Wang
- Clinical Medical College, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, People’s Republic of China
| | - Liangle Liu
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, People’s Republic of China
| | - Bingju Xie
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, People’s Republic of China
| |
Collapse
|
7
|
Mashayekhi K, Khazaie K, Faubion WA, Kim GB. Biomaterial-enhanced treg cell immunotherapy: A promising approach for transplant medicine and autoimmune disease treatment. Bioact Mater 2024; 37:269-298. [PMID: 38694761 PMCID: PMC11061617 DOI: 10.1016/j.bioactmat.2024.03.030] [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: 02/27/2024] [Revised: 03/20/2024] [Accepted: 03/25/2024] [Indexed: 05/04/2024] Open
Abstract
Regulatory T cells (Tregs) are crucial for preserving tolerance in the body, rendering Treg immunotherapy a promising treatment option for both organ transplants and autoimmune diseases. Presently, organ transplant recipients must undergo lifelong immunosuppression to prevent allograft rejection, while autoimmune disorders lack definitive cures. In the last years, there has been notable advancement in comprehending the biology of both antigen-specific and polyclonal Tregs. Clinical trials involving Tregs have demonstrated their safety and effectiveness. To maximize the efficacy of Treg immunotherapy, it is essential for these cells to migrate to specific target tissues, maintain stability within local organs, bolster their suppressive capabilities, and ensure their intended function's longevity. In pursuit of these goals, the utilization of biomaterials emerges as an attractive supportive strategy for Treg immunotherapy in addressing these challenges. As a result, the prospect of employing biomaterial-enhanced Treg immunotherapy holds tremendous promise as a treatment option for organ transplant recipients and individuals grappling with autoimmune diseases in the near future. This paper introduces strategies based on biomaterial-assisted Treg immunotherapy to enhance transplant medicine and autoimmune treatments.
Collapse
Affiliation(s)
- Kazem Mashayekhi
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
- Immunology of Infectious Diseases Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | | | - William A. Faubion
- Department of Immunology, Mayo Clinic, Scottsdale, AZ, USA
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Gloria B. Kim
- Department of Immunology, Mayo Clinic, Scottsdale, AZ, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Scottsdale, AZ, USA
| |
Collapse
|
8
|
Han J, Rindone AN, Elisseeff JH. Immunoengineering Biomaterials for Musculoskeletal Tissue Repair across Lifespan. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2311646. [PMID: 38416061 PMCID: PMC11239302 DOI: 10.1002/adma.202311646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 01/23/2024] [Indexed: 02/29/2024]
Abstract
Musculoskeletal diseases and injuries are among the leading causes of pain and morbidity worldwide. Broad efforts have focused on developing pro-regenerative biomaterials to treat musculoskeletal conditions; however, these approaches have yet to make a significant clinical impact. Recent studies have demonstrated that the immune system is central in orchestrating tissue repair and that targeting pro-regenerative immune responses can improve biomaterial therapeutic outcomes. However, aging is a critical factor negatively affecting musculoskeletal tissue repair and immune function. Hence, understanding how age affects the response to biomaterials is essential for improving musculoskeletal biomaterial therapies. This review focuses on the intersection of the immune system and aging in response to biomaterials for musculoskeletal tissue repair. The article introduces the general impacts of aging on tissue physiology, the immune system, and the response to biomaterials. Then, it explains how the adaptive immune system guides the response to injury and biomaterial implants in cartilage, muscle, and bone and discusses how aging impacts these processes in each tissue type. The review concludes by highlighting future directions for the development and translation of personalized immunomodulatory biomaterials for musculoskeletal tissue repair.
Collapse
Affiliation(s)
- Jin Han
- Translational Tissue Engineering Center, Wilmer Eye Institute and Department of Biomedical Engineering, Johns Hopkins University; Baltimore, MD 21231, USA
| | - Alexandra N. Rindone
- Translational Tissue Engineering Center, Wilmer Eye Institute and Department of Biomedical Engineering, Johns Hopkins University; Baltimore, MD 21231, USA
| | - Jennifer H. Elisseeff
- Translational Tissue Engineering Center, Wilmer Eye Institute and Department of Biomedical Engineering, Johns Hopkins University; Baltimore, MD 21231, USA
- Bloomberg~Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine; Baltimore, MD 21231, USA
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University; Baltimore, MD 21231, USA
| |
Collapse
|
9
|
Cui G, Yu X, He M, Huang S, Liu K, Li Y, Li J, Shao X, Lv Q, Li X, Tan M. Biological activity, limitations and steady-state delivery of functional substances for precision nutrition. ADVANCES IN FOOD AND NUTRITION RESEARCH 2024; 112:1-50. [PMID: 39218500 DOI: 10.1016/bs.afnr.2024.05.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Food-related functional substances with biological activity serve as a crucial material foundation for achieving precision nutrition, which has gained increasing attraction in regulating physiological functions, preventing chronic diseases, and maintaining human health. Nutritional substances typically include bioactive proteins, peptides, polysaccharides, polyphenols, functional lipids, carotenoids, probiotics, vitamins, saponins, and terpenes. These functional substances play an essential role in precise nutrition. This chapter introduces and summarizes typical functional substances to demonstrate the challenges in precision nutrition for their stability, solubility, and bioavailability. The current status of delivery systems of functional substances is described to give an insight into the development of desirable characteristics, such as food grade status, high loading capacity, site targeting, and controlled release capacity. Finally, the applications of food-borne delivery systems of functional substances for precision nutrition are emphasized to meet the requirement for precision nutrition during nutritional intervention for chronic diseases.
Collapse
Affiliation(s)
- Guoxin Cui
- Academy of Food Interdisciplinary Science, School of Food Science and Technology, Dalian Polytechnic University, Dalian, P.R. China; National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian, P.R. China; Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, P.R. China; State Key Laboratory of Marine Food Processing and Safety Control, Dalian Polytechnic University, Dalian, P.R. China
| | - Xiaoting Yu
- Academy of Food Interdisciplinary Science, School of Food Science and Technology, Dalian Polytechnic University, Dalian, P.R. China; National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian, P.R. China; Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, P.R. China; State Key Laboratory of Marine Food Processing and Safety Control, Dalian Polytechnic University, Dalian, P.R. China
| | - Ming He
- Academy of Food Interdisciplinary Science, School of Food Science and Technology, Dalian Polytechnic University, Dalian, P.R. China; National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian, P.R. China; Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, P.R. China; State Key Laboratory of Marine Food Processing and Safety Control, Dalian Polytechnic University, Dalian, P.R. China
| | - Shasha Huang
- Academy of Food Interdisciplinary Science, School of Food Science and Technology, Dalian Polytechnic University, Dalian, P.R. China; National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian, P.R. China; Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, P.R. China; State Key Laboratory of Marine Food Processing and Safety Control, Dalian Polytechnic University, Dalian, P.R. China
| | - Kangjing Liu
- Academy of Food Interdisciplinary Science, School of Food Science and Technology, Dalian Polytechnic University, Dalian, P.R. China; National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian, P.R. China; Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, P.R. China; State Key Laboratory of Marine Food Processing and Safety Control, Dalian Polytechnic University, Dalian, P.R. China
| | - Yu Li
- Academy of Food Interdisciplinary Science, School of Food Science and Technology, Dalian Polytechnic University, Dalian, P.R. China; National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian, P.R. China; Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, P.R. China; State Key Laboratory of Marine Food Processing and Safety Control, Dalian Polytechnic University, Dalian, P.R. China
| | - Jiaxuan Li
- Academy of Food Interdisciplinary Science, School of Food Science and Technology, Dalian Polytechnic University, Dalian, P.R. China; National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian, P.R. China; Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, P.R. China; State Key Laboratory of Marine Food Processing and Safety Control, Dalian Polytechnic University, Dalian, P.R. China
| | - Xiaoyang Shao
- Academy of Food Interdisciplinary Science, School of Food Science and Technology, Dalian Polytechnic University, Dalian, P.R. China; National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian, P.R. China; Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, P.R. China; State Key Laboratory of Marine Food Processing and Safety Control, Dalian Polytechnic University, Dalian, P.R. China
| | - Qiyan Lv
- Academy of Food Interdisciplinary Science, School of Food Science and Technology, Dalian Polytechnic University, Dalian, P.R. China; National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian, P.R. China; Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, P.R. China; State Key Laboratory of Marine Food Processing and Safety Control, Dalian Polytechnic University, Dalian, P.R. China
| | - Xueqian Li
- Academy of Food Interdisciplinary Science, School of Food Science and Technology, Dalian Polytechnic University, Dalian, P.R. China; National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian, P.R. China; Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, P.R. China; State Key Laboratory of Marine Food Processing and Safety Control, Dalian Polytechnic University, Dalian, P.R. China
| | - Mingqian Tan
- Academy of Food Interdisciplinary Science, School of Food Science and Technology, Dalian Polytechnic University, Dalian, P.R. China; National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian, P.R. China; Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, P.R. China; State Key Laboratory of Marine Food Processing and Safety Control, Dalian Polytechnic University, Dalian, P.R. China.
| |
Collapse
|
10
|
Wang Y, Liu C, Ren Y, Song J, Fan K, Gao L, Ji X, Chen X, Zhao H. Nanomaterial-Based Strategies for Attenuating T-Cell-Mediated Immunodepression in Stroke Patients: Advancing Research Perspectives. Int J Nanomedicine 2024; 19:5793-5812. [PMID: 38882535 PMCID: PMC11180442 DOI: 10.2147/ijn.s456632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Accepted: 05/14/2024] [Indexed: 06/18/2024] Open
Abstract
This review article discusses the potential of nanomaterials in targeted therapy and immunomodulation for stroke-induced immunosuppression. Although nanomaterials have been extensively studied in various biomedical applications, their specific use in studying and addressing immunosuppression after stroke remains limited. Stroke-induced neuroinflammation is characterized by T-cell-mediated immunodepression, which leads to increased morbidity and mortality. Key observations related to immunodepression after stroke, including lymphopenia, T-cell dysfunction, regulatory T-cell imbalance, and cytokine dysregulation, are discussed. Nanomaterials, such as liposomes, micelles, polymeric nanoparticles, and dendrimers, offer advantages in the precise delivery of drugs to T cells, enabling enhanced targeting and controlled release of immunomodulatory agents. These nanomaterials have the potential to modulate T-cell function, promote neuroregeneration, and restore immune responses, providing new avenues for stroke treatment. However, challenges related to biocompatibility, stability, scalability, and clinical translation need to be addressed. Future research efforts should focus on comprehensive studies to validate the efficacy and safety of nanomaterial-based interventions targeting T cells in stroke-induced immunosuppression. Collaborative interdisciplinary approaches are necessary to advance the field and translate these innovative strategies into clinical practice, ultimately improving stroke outcomes and patient care.
Collapse
Grants
- This work was supported by the National Natural Science Foundation of China (Grant number 82001248), National University of Singapore (NUHSRO/2020/133/Startup/08, NUHSRO/2023/008/NUSMed/TCE/LOA, NUHSRO/2021/034/TRP/09/Nanomedicine, NUHSRO/2021/044/Kickstart/09/LOA, 23-0173-A0001), National Medical Research Council (MOH-001388-00, CG21APR1005, OFIRG23jul-0047), Singapore Ministry of Education (MOE-000387-00), and National Research Foundation (NRF-000352-00)
Collapse
Affiliation(s)
- Yan Wang
- Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, People’s Republic of China
| | - Cuiying Liu
- School of Nursing, Capital Medical University, Beijing, People’s Republic of China
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, People’s Republic of China
| | - Yanhong Ren
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, People’s Republic of China
| | - Jibin Song
- State Key Laboratory of Chemical Resource Engineering, College of Chemistry, Beijing University of Chemical Technology, Beijing, People’s Republic of China
| | - Kelong Fan
- CAS Engineering Laboratory for Nanozyme, Institute of Biophysics Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Lizeng Gao
- CAS Engineering Laboratory for Nanozyme, Institute of Biophysics Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Xunming Ji
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, People’s Republic of China
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Theranostics Center of Excellence (TCE), Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), Singapore
| | - Heng Zhao
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, People’s Republic of China
| |
Collapse
|
11
|
Pang WW, Cai YS, Cao C, Zhang FR, Zeng Q, Liu DY, Wang N, Qu XC, Chen XD, Deng HW, Tan LJ. Mendelian randomization and transcriptome analysis identified immune-related biomarkers for osteoarthritis. Front Immunol 2024; 15:1334479. [PMID: 38680491 PMCID: PMC11045931 DOI: 10.3389/fimmu.2024.1334479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 03/27/2024] [Indexed: 05/01/2024] Open
Abstract
Background The immune microenvironment assumes a significant role in the pathogenesis of osteoarthritis (OA). However, the current biomarkers for the diagnosis and treatment of OA are not satisfactory. Our study aims to identify new OA immune-related biomarkers to direct the prevention and treatment of OA using multi-omics data. Methods The discovery dataset integrated the GSE89408 and GSE143514 datasets to identify biomarkers that were significantly associated with the OA immune microenvironment through multiple machine learning methods and weighted gene co-expression network analysis (WGCNA). The identified signature genes were confirmed using two independent validation datasets. We also performed a two-sample mendelian randomization (MR) study to generate causal relationships between biomarkers and OA using OA genome-wide association study (GWAS) summary data (cases n = 24,955, controls n = 378,169). Inverse-variance weighting (IVW) method was used as the main method of causal estimates. Sensitivity analyses were performed to assess the robustness and reliability of the IVW results. Results Three signature genes (FCER1G, HLA-DMB, and HHLA-DPA1) associated with the OA immune microenvironment were identified as having good diagnostic performances, which can be used as biomarkers. MR results showed increased levels of FCER1G (OR = 1.118, 95% CI 1.031-1.212, P = 0.041), HLA-DMB (OR = 1.057, 95% CI 1.045 -1.069, P = 1.11E-21) and HLA-DPA1 (OR = 1.030, 95% CI 1.005-1.056, P = 0.017) were causally and positively associated with the risk of developing OA. Conclusion The present study identified the 3 potential immune-related biomarkers for OA, providing new perspectives for the prevention and treatment of OA. The MR study provides genetic support for the causal effects of the 3 biomarkers with OA and may provide new insights into the molecular mechanisms leading to the development of OA.
Collapse
Affiliation(s)
- Wei-Wei Pang
- Laboratory of Molecular and Statistical Genetics, College of Life Sciences, Hunan Normal University, Changsha, Hunan, China
| | - Yi-Sheng Cai
- Laboratory of Molecular and Statistical Genetics, College of Life Sciences, Hunan Normal University, Changsha, Hunan, China
| | - Chong Cao
- Laboratory of Molecular and Statistical Genetics, College of Life Sciences, Hunan Normal University, Changsha, Hunan, China
| | - Fu-Rong Zhang
- Laboratory of Molecular and Statistical Genetics, College of Life Sciences, Hunan Normal University, Changsha, Hunan, China
| | - Qin Zeng
- Laboratory of Molecular and Statistical Genetics, College of Life Sciences, Hunan Normal University, Changsha, Hunan, China
| | - Dan-Yang Liu
- Laboratory of Molecular and Statistical Genetics, College of Life Sciences, Hunan Normal University, Changsha, Hunan, China
| | - Ning Wang
- Laboratory of Molecular and Statistical Genetics, College of Life Sciences, Hunan Normal University, Changsha, Hunan, China
| | - Xiao-Chao Qu
- Laboratory of Molecular and Statistical Genetics, College of Life Sciences, Hunan Normal University, Changsha, Hunan, China
| | - Xiang-Ding Chen
- Laboratory of Molecular and Statistical Genetics, College of Life Sciences, Hunan Normal University, Changsha, Hunan, China
| | - Hong-Wen Deng
- Tulane Center of Biomedical Informatics and Genomics, Deming Department of Medicine, School of Medicine, Tulane University, New Orleans, LA, United States
| | - Li-Jun Tan
- Laboratory of Molecular and Statistical Genetics, College of Life Sciences, Hunan Normal University, Changsha, Hunan, China
| |
Collapse
|
12
|
Lee S, Yoo J, Bae G, Thangam R, Heo J, Park JY, Choi H, Kim C, An J, Kim J, Mun KR, Shin S, Zhang K, Zhao P, Kim Y, Kang N, Han SB, Kim D, Yoon J, Kang M, Kim J, Yang L, Karamikamkar S, Kim J, Zhu Y, Najafabadi AH, Song G, Kim DH, Lee KB, Oh SJ, Jung HD, Song HC, Jang WY, Bian L, Chu Z, Yoon J, Kim JS, Zhang YS, Kim Y, Jang HS, Kim S, Kang H. Photonic control of ligand nanospacing in self-assembly regulates stem cell fate. Bioact Mater 2024; 34:164-180. [PMID: 38343773 PMCID: PMC10859239 DOI: 10.1016/j.bioactmat.2023.12.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 11/30/2023] [Accepted: 12/14/2023] [Indexed: 10/28/2024] Open
Abstract
Extracellular matrix (ECM) undergoes dynamic inflation that dynamically changes ligand nanospacing but has not been explored. Here we utilize ECM-mimicking photocontrolled supramolecular ligand-tunable Azo+ self-assembly composed of azobenzene derivatives (Azo+) stacked via cation-π interactions and stabilized with RGD ligand-bearing poly(acrylic acid). Near-infrared-upconverted-ultraviolet light induces cis-Azo+-mediated inflation that suppresses cation-π interactions, thereby inflating liganded self-assembly. This inflation increases nanospacing of "closely nanospaced" ligands from 1.8 nm to 2.6 nm and the surface area of liganded self-assembly that facilitate stem cell adhesion, mechanosensing, and differentiation both in vitro and in vivo, including the release of loaded molecules by destabilizing water bridges and hydrogen bonds between the Azo+ molecules and loaded molecules. Conversely, visible light induces trans-Azo+ formation that facilitates cation-π interactions, thereby deflating self-assembly with "closely nanospaced" ligands that inhibits stem cell adhesion, mechanosensing, and differentiation. In stark contrast, when ligand nanospacing increases from 8.7 nm to 12.2 nm via the inflation of self-assembly, the surface area of "distantly nanospaced" ligands increases, thereby suppressing stem cell adhesion, mechanosensing, and differentiation. Long-term in vivo stability of self-assembly via real-time tracking and upconversion are verified. This tuning of ligand nanospacing can unravel dynamic ligand-cell interactions for stem cell-regulated tissue regeneration.
Collapse
Affiliation(s)
- Sungkyu Lee
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Jounghyun Yoo
- Chemical and Biological Integrative Research Center, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Gunhyu Bae
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Ramar Thangam
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Jeongyun Heo
- Chemical and Biological Integrative Research Center, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Jung Yeon Park
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea
| | - Honghwan Choi
- Chemical and Biological Integrative Research Center, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea
| | - Chowon Kim
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Jusung An
- Department of Chemistry, Korea University, Seoul, 02841, Republic of Korea
| | - Jungryun Kim
- Department of Chemistry, Korea University, Seoul, 02841, Republic of Korea
| | - Kwang Rok Mun
- Materials Architecturing Research Center, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Seungyong Shin
- Materials Architecturing Research Center, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Kunyu Zhang
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, China
| | - Pengchao Zhao
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, China
| | - Yuri Kim
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Nayeon Kang
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Seong-Beom Han
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea
| | - Dahee Kim
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Jiwon Yoon
- Chemical and Biological Integrative Research Center, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Misun Kang
- Materials Architecturing Research Center, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Jihwan Kim
- Chemical and Biological Integrative Research Center, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea
| | - Letao Yang
- Department of Chemistry and Chemical Biology, Rutgers University, Piscataway, NJ, 08854, USA
| | | | - Jinjoo Kim
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA, 90064, USA
| | - Yangzhi Zhu
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA, 90064, USA
| | | | - Guosheng Song
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, China
| | - Dong-Hwee Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea
| | - Ki-Bum Lee
- Department of Chemistry and Chemical Biology, Rutgers University, Piscataway, NJ, 08854, USA
| | - Soong Ju Oh
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Hyun-Do Jung
- Department of Biomedical-Chemical Engineering, The Catholic University of Korea, Gyeonggi-do, 14662, Republic of Korea
- Division of Materials Science and Engineering, Hanyang University, Seoul, 04763, Republic of Korea
| | - Hyun-Cheol Song
- Electronic Materials Research Center, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
- KIST-SKKU Carbon-Neutral Research Center, Sungkyunkwan University (SKKU), Suwon, 16419, Republic of Korea
| | - Woo Young Jang
- Department of Orthopedic Surgery, Korea University Anam Hospital, Seoul, 02841, Republic of Korea
| | - Liming Bian
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, China
| | - Zhiqin Chu
- Department of Electrical and Electronic Engineering and Joint Appointment with School of Biomedical Sciences, The University of Hong Kong, Hong Kong, 518057, China
| | - Juyoung Yoon
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Jong Seung Kim
- Department of Chemistry, Korea University, Seoul, 02841, Republic of Korea
| | - Yu Shrike Zhang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital Harvard Medical School, Cambridge, MA, 02139, USA
| | - Yongju Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea
| | - Ho Seong Jang
- Materials Architecturing Research Center, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
- Division of Nano & Information Technology, KIST School, Korea University of Science and Technology (UST), Seoul, 02792, Republic of Korea
| | - Sehoon Kim
- Chemical and Biological Integrative Research Center, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea
| | - Heemin Kang
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
- College of Medicine, Korea University, Seoul, 02841, Republic of Korea
| |
Collapse
|
13
|
Deng R, Zhao R, Zhang Z, Chen Y, Yang M, Lin Y, Ye J, Li N, Qin H, Yan X, Shi J, Yuan F, Song S, Xu Z, Song Y, Fu J, Xu B, Nie G, Yu JK. Chondrocyte membrane-coated nanoparticles promote drug retention and halt cartilage damage in rat and canine osteoarthritis. Sci Transl Med 2024; 16:eadh9751. [PMID: 38381849 DOI: 10.1126/scitranslmed.adh9751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Accepted: 01/30/2024] [Indexed: 02/23/2024]
Abstract
Osteoarthritis (OA) is a chronic joint disease characterized by progressive degeneration of articular cartilage. A challenge in the development of disease-modifying drugs is effective delivery to chondrocytes. The unique structure of the joint promotes rapid clearance of drugs through synovial fluid, and the dense and avascular cartilage extracellular matrix (ECM) limits drug penetration. Here, we show that poly(lactide-co-glycolic acid) nanoparticles coated in chondrocyte membranes (CM-NPs) were preferentially taken up by rat chondrocytes ex vivo compared with uncoated nanoparticles. Internalization of the CM-NPs was mediated primarily by E-cadherin, clathrin-mediated endocytosis, and micropinocytosis. These CM-NPs adhered to the cartilage ECM in rat knee joints in vivo and penetrated deeply into the cartilage matrix with a residence time of more than 34 days. Simulated synovial fluid clearance studies showed that CM-NPs loaded with a Wnt pathway inhibitor, adavivint (CM-NPs-Ada), delayed the catabolic metabolism of rat and human chondrocytes and cartilage explants under inflammatory conditions. In a surgical model of rat OA, drug-loaded CM-NPs effectively restored gait, attenuated periarticular bone remodeling, and provided chondroprotection against cartilage degeneration. OA progression was also mitigated by CM-NPs-Ada in a canine model of anterior cruciate ligament transection. These results demonstrate the feasibility of using chondrocyte membrane-coated nanoparticles to improve the pharmacokinetics and efficacy of anti-OA drugs.
Collapse
Affiliation(s)
- Ronghui Deng
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing 100191, P. R. China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center of Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P. R. China
- Beijing Key Laboratory of Sports Injuries, Beijing 100191, P. R. China
| | - Ruifang Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center of Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Zining Zhang
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing 100191, P. R. China
- Beijing Key Laboratory of Sports Injuries, Beijing 100191, P. R. China
| | - Yang Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center of Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Meng Yang
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing 100191, P. R. China
- Beijing Key Laboratory of Sports Injuries, Beijing 100191, P. R. China
| | - Yixuan Lin
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center of Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Jing Ye
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing 100191, P. R. China
- Beijing Key Laboratory of Sports Injuries, Beijing 100191, P. R. China
| | - Nan Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center of Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Hao Qin
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center of Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Xin Yan
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing 100191, P. R. China
- Beijing Key Laboratory of Sports Injuries, Beijing 100191, P. R. China
| | - Jian Shi
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center of Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P. R. China
| | - Fuzhen Yuan
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing 100191, P. R. China
- Beijing Key Laboratory of Sports Injuries, Beijing 100191, P. R. China
| | - Shitang Song
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing 100191, P. R. China
- Beijing Key Laboratory of Sports Injuries, Beijing 100191, P. R. China
| | - Zijie Xu
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing 100191, P. R. China
- Beijing Key Laboratory of Sports Injuries, Beijing 100191, P. R. China
| | - Yifan Song
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing 100191, P. R. China
- Beijing Key Laboratory of Sports Injuries, Beijing 100191, P. R. China
| | - Jiangnan Fu
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing 100191, P. R. China
- Beijing Key Laboratory of Sports Injuries, Beijing 100191, P. R. China
| | - Bingbing Xu
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing 100191, P. R. China
- Beijing Key Laboratory of Sports Injuries, Beijing 100191, P. R. China
| | - Guangjun Nie
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center of Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Jia-Kuo Yu
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing 100191, P. R. China
- Beijing Key Laboratory of Sports Injuries, Beijing 100191, P. R. China
- Orthopedic Sports Medicine Center, Beijing Tsinghua Changgung Hospital, Affiliated Hospital of Tsinghua University, Beijing 102218, P. R. China
| |
Collapse
|
14
|
Prinz E, Schlupp L, Dyson G, Barrett M, Szymczak A, Velasco C, Izda V, Dunn CM, Jeffries MA. OA susceptibility in mice is partially mediated by the gut microbiome, is transferrable via microbiome transplantation and is associated with immunophenotype changes. Ann Rheum Dis 2024; 83:382-393. [PMID: 37979958 PMCID: PMC10922159 DOI: 10.1136/ard-2023-224907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 11/03/2023] [Indexed: 11/20/2023]
Abstract
OBJECTIVES The Murphy Roths Large (MRL)/MpJ 'superhealer' mouse strain is protected from post-traumatic osteoarthritis (OA), although no studies have evaluated the microbiome in the context of this protection. This study characterised microbiome differences between MRL and wild-type mice, evaluated microbiome transplantation and OA and investigated microbiome-associated immunophenotypes. METHODS Cecal material from mixed sex C57BL6/J (B6) or female MRL/MpJ (MRL) was transplanted into B6 and MRL mice, then OA was induced by disruption of the medial meniscus surgery (DMM). In other experiments, transplantation was performed after DMM and transplantation was performed into germ-free mice. Transplanted mice were bred through F2. OARSI, synovitis and osteophyte scores were determined blindly 8 weeks after DMM. 16S microbiome sequencing was performed and metagenomic function was imputed. Immunophenotypes were determined using mass cytometry. RESULTS MRL-into-B6 transplant prior to DMM showed reduced OA histopathology (OARSI score 70% lower transplant vs B6 control), synovitis (60% reduction) and osteophyte scores (30% reduction) 8 weeks after DMM. When performed 48 hours after DMM, MRL-into-B6 transplant improved OA outcomes but not when performed 1-2 weeks after DMM. Protection was seen in F1 (60% reduction) and F2 progeny (30% reduction). Several cecal microbiome clades were correlated with either better (eg, Lactobacillus, R=-0.32, p=0.02) or worse (eg, Rikenellaceae, R=0.43, p=0.001) OA outcomes. Baseline immunophenotypes associated with MRL-into-B6 transplants and MRL included reduced double-negative T cells and increased CD25+CD4+ T cells. CONCLUSION The gut microbiome is responsible in part for OA protection in MRL mice and is transferrable by microbiome transplantation. Transplantation induces resting systemic immunophenotyping changes that correlate with OA protection.
Collapse
Affiliation(s)
- Emmaline Prinz
- Arthritis & Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
- Division of Rheumatology, Immunology, and Allergy, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Leoni Schlupp
- Arthritis & Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Gabby Dyson
- Arthritis & Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Montana Barrett
- Arthritis & Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Aleksander Szymczak
- Arthritis & Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Cassandra Velasco
- Division of Rheumatology, Immunology, and Allergy, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Vladislav Izda
- Arthritis & Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Christopher M Dunn
- Arthritis & Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
- Division of Rheumatology, Immunology, and Allergy, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Matlock A Jeffries
- Arthritis & Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
- Division of Rheumatology, Immunology, and Allergy, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| |
Collapse
|
15
|
Hao Y, Zhu G, Yu L, Ren Z, Zhou W, Zhang P, Lian X. FOXO3-Activated HOTTIP Sequesters miR-615-3p away from COL2A1 to Mitigate Intervertebral Disc Degeneration. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:280-295. [PMID: 37981220 DOI: 10.1016/j.ajpath.2023.10.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 10/15/2023] [Accepted: 10/31/2023] [Indexed: 11/21/2023]
Abstract
In this study, knockout of FOXO3 was found to impair intervertebral disc maturation and homeostasis in postnatal mice as well as facilitating extracellular matrix degradation. RNA sequencing can uncover disease-related gene expression and investigate disease pathophysiology. High-throughput transcriptome sequencing and experimental validations were used to identify the essential gene and mechanism involved in intervertebral disc degeneration (IDD). Nucleus pulposus (NP) tissue samples were collected from the mice with conditional knockout of FOXO3 (FOXO3 KO) for high-throughput sequencing, followed by screening of differentially expressed lncRNAs and mRNAs. The mRNAs were subjected to GO and KEGG enrichment analyses. Interactions among FOXO3, HOTTIP, miR-615-3p, and COL2A1 were analyzed. NP cells were subjected to a series of mimics, inhibitors, overexpression plasmids, and shRNAs to validate the mechanisms of FOXO3 in controlling HOTTIP/miR-615-3p/COL2A1 in IDD. Mechanistically, FOXO3 transcriptionally activated HOTTIP, facilitated the competitive HOTTIP binding to miR-615-3p, and increased the expression of the miR-615-3p target gene COL2A1. Thus, NP cell proliferation was induced, cell apoptosis was diminished, resulting in delayed development of IDD. Based on these data, the transcription factor FOXO3 may decrease miR-615-3p binding to COL2A1 and up-regulate COL2A1 expression by activating HOTTIP transcription, which in turn inhibits NP cell apoptosis and promotes its proliferation, to prevent the degradation of intervertebral disc matrix and maintain the normal physiological function of intervertebral disc, thereby preventing the occurrence and development of IDD.
Collapse
Affiliation(s)
- Yingjie Hao
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Guangduo Zhu
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Lei Yu
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhinan Ren
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Weiwei Zhou
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Panke Zhang
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xu Lian
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
16
|
Kapnick SM, Martin CA, Jewell CM. Engineering metabolism to modulate immunity. Adv Drug Deliv Rev 2024; 204:115122. [PMID: 37935318 PMCID: PMC10843796 DOI: 10.1016/j.addr.2023.115122] [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: 01/14/2023] [Revised: 07/19/2023] [Accepted: 10/25/2023] [Indexed: 11/09/2023]
Abstract
Metabolic programming and reprogramming have emerged as pivotal mechanisms for altering immune cell function. Thus, immunometabolism has become an attractive target area for treatment of immune-mediated disorders. Nonetheless, many hurdles to delivering metabolic cues persist. In this review, we consider how biomaterials are poised to transform manipulation of immune cell metabolism through integrated control of metabolic configurations to affect outcomes in autoimmunity, regeneration, transplant, and cancer. We emphasize the features of nanoparticles and other biomaterials that permit delivery of metabolic cues to the intracellular compartment of immune cells, or strategies for altering signals in the extracellular space. We then provide perspectives on the potential for reciprocal regulation of immunometabolism by the physical properties of materials themselves. Lastly, opportunities for clinical translation are highlighted. This discussion contributes to our understanding of immunometabolism, biomaterials-based strategies for altering metabolic configurations in immune cells, and emerging concepts in this evolving field.
Collapse
Affiliation(s)
- Senta M Kapnick
- Fischell Department of Bioengineering, University of Maryland, 8278 Paint Branch Drive, College Park, MD, USA; Department of Veterans Affairs, VA Maryland Health Care System, 10 N Green Street, Baltimore, MD, USA
| | - Corinne A Martin
- Fischell Department of Bioengineering, University of Maryland, 8278 Paint Branch Drive, College Park, MD, USA
| | - Christopher M Jewell
- Fischell Department of Bioengineering, University of Maryland, 8278 Paint Branch Drive, College Park, MD, USA; Department of Veterans Affairs, VA Maryland Health Care System, 10 N Green Street, Baltimore, MD, USA; Robert E. Fischell Institute for Biomedical Devices, 8278 Paint Branch Drive, College Park, MD, USA; Marlene and Stewart Greenebaum Comprehensive Cancer Center, 22 S Greene Street, Suite N9E17, Baltimore, MD, USA.
| |
Collapse
|
17
|
Xu R, Du Y, Li X, Mao X, Zheng L, Zhao M. Differences between soluble and insoluble undenatured type II collagen in improving osteoarthritis in rats and their potential mechanisms. Food Funct 2023; 14:10240-10251. [PMID: 37921641 DOI: 10.1039/d3fo02954a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2023]
Abstract
Our previous research showed that soluble (SC II) and insoluble (IC II) undenatured type II collagen had significant differences during gastrointestinal digestion in vitro, and SC II exposed more type II collagen with triple helix structure. However, the differences in their in vivo digestive characteristics, improvement on osteoarthritis (OA), and possible mechanisms have not been elucidated. The aim of this study was to explore these issues. After oral administration of SC II and IC II, the joint swelling of OA rats significantly reduced, and the weight bearing ratio of right hind limb significantly increased, especially in SC II group (raised to 48%). The Mankin and OARSI scores decreased by 35% and 48% in SC II group, respectively. SC II and IC II increased the mRNA expression of anti-inflammatory factors and the proportion of regulatory T cells (Treg). Importantly, type II collagen released by IC II during in vivo gastrointestinal digestion was far less than SC II, which explained the higher ability of SC II to induce immune tolerance in small intestine than IC II. Bioinformatics analysis showed that the differential genes between model and control were significantly enriched in PI3K/AKT, PPAR and AMPK signalling pathways, and 24 hub genes were analyzed. SC II significantly down-regulated the mRNA expression of Il6, Ccl7, NF-κB, AKT and up-regulated the mRNA expression of Scd1. These results showed that SC II was superior to IC II in improving OA by inducing immune tolerance and could regulate key biomarkers and signalling pathways in OA rats.
Collapse
Affiliation(s)
- Rong Xu
- School of Food Science and Engineering, South China University of Technology, Guangzhou, 510640, China.
| | - Yulan Du
- Perfect (China) Co., Ltd, Zhongshan, 528400, China
- Perfect Life and Health Sciences Co., Ltd, 528451, China
| | - Xiaomin Li
- Perfect (China) Co., Ltd, Zhongshan, 528400, China
- Perfect Life and Health Sciences Co., Ltd, 528451, China
| | - Xinliang Mao
- Perfect (China) Co., Ltd, Zhongshan, 528400, China
- Perfect Life and Health Sciences Co., Ltd, 528451, China
| | - Lin Zheng
- School of Food Science and Engineering, South China University of Technology, Guangzhou, 510640, China.
| | - Mouming Zhao
- School of Food Science and Engineering, South China University of Technology, Guangzhou, 510640, China.
| |
Collapse
|
18
|
Lee ES, Ko H, Kim CH, Kim HC, Choi SK, Jeong SW, Lee SG, Lee SJ, Na HK, Park JH, Shin JM. Disease-microenvironment modulation by bare- or engineered-exosome for rheumatoid arthritis treatment. Biomater Res 2023; 27:81. [PMID: 37635253 PMCID: PMC10464174 DOI: 10.1186/s40824-023-00418-2] [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: 05/22/2023] [Accepted: 08/13/2023] [Indexed: 08/29/2023] Open
Abstract
BACKGROUND Exosomes are extracellular vesicles secreted by eukaryotic cells and have been extensively studied for their surface markers and internal cargo with unique functions. A deeper understanding of exosomes has allowed their application in various research areas, particularly in diagnostics and therapy. MAIN BODY Exosomes have great potential as biomarkers and delivery vehicles for encapsulating therapeutic cargo. However, the limitations of bare exosomes, such as rapid phagocytic clearance and non-specific biodistribution after injection, pose significant challenges to their application as drug delivery systems. This review focuses on exosome-based drug delivery for treating rheumatoid arthritis, emphasizing pre/post-engineering approaches to overcome these challenges. CONCLUSION This review will serve as an essential resource for future studies to develop novel exosome-based therapeutic approaches for rheumatoid arthritis. Overall, the review highlights the potential of exosomes as a promising therapeutic approach for rheumatoid arthritis treatment.
Collapse
Affiliation(s)
- Eun Sook Lee
- Safety Measurement Institute, Korea Research Institute of Standards and Science (KRISS), 267 Gajeong-Ro, Yuseong-Gu, Daejeon, 34113, Republic of Korea
| | - Hyewon Ko
- Bionanotechnology Research Center, Korea Research Institute of Bioscience & Biotechnology (KRIBB), Yuseong-Gu, Daejeon, 34141, Republic of Korea
| | - Chan Ho Kim
- School of Chemical Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Hyun-Chul Kim
- Division of Biotechnology, Convergence Research Institute, DGIST, 333 Techno Jungang-Daero, Daegu, 42988, Republic of Korea
| | - Seong-Kyoon Choi
- Division of Biotechnology, Convergence Research Institute, DGIST, 333 Techno Jungang-Daero, Daegu, 42988, Republic of Korea
| | - Sang Won Jeong
- Division of Biotechnology, Convergence Research Institute, DGIST, 333 Techno Jungang-Daero, Daegu, 42988, Republic of Korea
| | - Se-Guen Lee
- Division of Biotechnology, Convergence Research Institute, DGIST, 333 Techno Jungang-Daero, Daegu, 42988, Republic of Korea
| | - Sung-Jun Lee
- Division of Biotechnology, Convergence Research Institute, DGIST, 333 Techno Jungang-Daero, Daegu, 42988, Republic of Korea
| | - Hee-Kyung Na
- Safety Measurement Institute, Korea Research Institute of Standards and Science (KRISS), 267 Gajeong-Ro, Yuseong-Gu, Daejeon, 34113, Republic of Korea
| | - Jae Hyung Park
- School of Chemical Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Jung Min Shin
- Division of Biotechnology, Convergence Research Institute, DGIST, 333 Techno Jungang-Daero, Daegu, 42988, Republic of Korea.
- Department of Polymer Science and Engineering, Korea National University of Transportation, Chungju, 27469, Republic of Korea.
| |
Collapse
|
19
|
Dravid AA, Dhanabalan KM, Naskar S, Vashistha A, Agarwal S, Padhan B, Dewani M, Agarwal R. Sustained release resolvin D1 liposomes are effective in the treatment of osteoarthritis in obese mice. J Biomed Mater Res A 2023; 111:765-777. [PMID: 36773024 DOI: 10.1002/jbm.a.37512] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 01/24/2023] [Accepted: 01/31/2023] [Indexed: 02/12/2023]
Abstract
Osteoarthritis (OA) is the most common joint disorder and currently affects >500 million patients worldwide, with ~60% of them also suffering from obesity. There is no drug approved for human use that changes the course of OA progression. OA is one of the most common comorbidities of obesity, and obesity-related OA (ObOA) is a serious health concern because it shows heightened severity of tissue damage and also predominantly affects the working population. Unresolved inflammation is a major driver of ObOA, thus, resolving disease-associated inflammation is a viable strategy to treat ObOA. Resolvins are highly potent molecules that play a role in the resolution of inflammation and promote tissue healing. However, small molecules (like Resolvin D1; RvD1) have to be administered frequently or prior to injury because they lose their in vivo activity rapidly either by lymphatic clearance, or oxidation-mediated deactivation. In this study, we have encapsulated RvD1 in liposomes and established its efficacy in the mouse model of ObOA at much lower dosages than freely administered RvD1. Liposomal RvD1 (lipo-RvD1) acted as a source of the RvD1 molecules for ~11 days in vitro in synovial fluid derived from patients. When administered prophylactically or therapeutically, lipo-RvD1 suppressed cartilage damage in male C57BL/6 mice compared to untreated and free RvD1 treatments. This efficacy was achieved by increasing the proportion of the proresolution M2 macrophages over proinflammatory M1 macrophages in the synovial membrane. These results show the potential of lipo-RvD1 as an anti-OA agent.
Collapse
|
20
|
McHugh J. T reg cell-inducing nanoparticles show promise for treating OA. Nat Rev Rheumatol 2023; 19:62. [PMID: 36624262 DOI: 10.1038/s41584-023-00906-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|