1
|
Rao S, Kiick KL. Recent advances in extracellular matrix-inspired nanocarriers. Expert Opin Drug Deliv 2025:1-19. [PMID: 40503764 DOI: 10.1080/17425247.2025.2519809] [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: 03/15/2025] [Accepted: 06/10/2025] [Indexed: 06/19/2025]
Abstract
INTRODUCTION ECM-inspired nanocarriers have emerged as a promising platform for drug delivery due to their unique advantages over traditional nanocarrier systems. The ECM is a complex three-dimensional network comprising proteins and polysaccharides that play a key role in maintaining tissue function and homeostasis. Recent advances in the design and synthesis of ECM-inspired nanocarriers have resulted in superior efficacy, targeting, and responsive delivery systems. AREAS COVERED This review covers ECM-inspired nanocarriers, focusing on their design and fabrication methods, and applications in drug delivery, tissue engineering, and regenerative medicine. Specific focus is placed on nanocarriers derived from elastin, collagen, hyaluronic acid, and their combinations, creating 'conjugate nanoparticles' published in the last 5 years. This review also discusses the benefits of mimicking ECM structure and function, the advantages of each nanoparticle type, challenges associated with large-scale synthesis, and immunogenicity. EXPERT OPINION ECM-inspired nanocarriers are a novel avenue for the delivery of therapeutics with recent emphasis placed on complex, responsive systems. While substantial progress has been made in the design and application of these nanocarriers in pre-clinical studies, significant challenges remain, particularly concerning immunogenicity, scalability, and the need for more robust clinical data, before these innovations can be widely translated into clinical practice.
Collapse
Affiliation(s)
- Sanjna Rao
- Department of Biomedical Engineering, University of Delaware, Newark, DE, USA
| | - Kristi L Kiick
- Department of Biomedical Engineering, University of Delaware, Newark, DE, USA
- Department of Materials Science and Engineering, University of Delaware, Newark, DE, USA
| |
Collapse
|
2
|
Wang Y, Deng M, Wu Y, Zheng C, Zhang F, Guo C, Zhang B, Hu C, Kong Q, Wang Y. A multifunctional mitochondria-protective gene delivery platform promote intervertebral disc regeneration. Biomaterials 2025; 317:123067. [PMID: 39742837 DOI: 10.1016/j.biomaterials.2024.123067] [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: 08/12/2024] [Revised: 11/09/2024] [Accepted: 12/27/2024] [Indexed: 01/04/2025]
Abstract
Intervertebral disc degeneration (IDD) is a deleterious condition driven by localized inflammation and the associated disruption of the normal homeostatic balance between anabolism and catabolism, contributing to progressive functional abnormalities within the nucleus pulposus (NP). Despite our prior evidence demonstrating that a miR-21 inhibitor can have regenerative effects that counteract the progression of IDD, its application for IDD treatment remains limited by the inadequacy of current local delivery systems. Here, an injectable tannic acid (TA)-loaded hydrogel gene delivery system was developed and used for the encapsulation of a multifunctional mitochondria-protecting gene nanocarrier (PHs). This engineered platform was designed for the sustained on-demand delivery of both miR-21 inhibitor and ss-31 (mitochondrial-targeted peptide) constructs to the NP. This prepared hydrogel could be implanted into the intervertebral disc using a minimally invasive approach whereupon it was able to rapidly release TA. Sustained PHs release was then achieved as appropriate through a mechanism mediated by the activity of MMP-2. Following the targeted uptake of PHs by degenerated NP cells, the subsequent release of encapsulated miR-21 inhibitor suppressed apoptotic cell death and modulated the metabolism of the extracellular matrix (ECM) by targeting the Spry1 gene. At the same time, ss-31 was able to target damaged mitochondria and alleviate inflammatory activity via the suppression of mitochondrial ROS-NLRP3-IL-1β/Caspase1 pathway activity. Synergistic ECM regeneration and anti-inflammatory effects were sufficient to provide therapeutic benefits in an in vivo model of IDD. Together, these results thus highlight this hydrogel-based gene delivery platform as a promising novel approach to the treatment of IDD.
Collapse
Affiliation(s)
- Yu Wang
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Mingyan Deng
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ye Wu
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Cheng Zheng
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan, China
| | - Fanjun Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan, China
| | - Chuan Guo
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Bo Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan, China
| | - Cheng Hu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan, China
| | - Qingquan Kong
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| | - Yunbing Wang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
3
|
Huang B, Zeng R, Liu X, Pan L, Bai H, Liao J, Xu W, Fu H, Nan K, Lin S. Drug-free hyaluronic acid-microneedle with unexpected inhibition activity on benzalkonium chloride-induced corneal inflammation and stromal scarring. Mater Today Bio 2025; 32:101722. [PMID: 40236813 PMCID: PMC11997401 DOI: 10.1016/j.mtbio.2025.101722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Revised: 03/26/2025] [Accepted: 03/29/2025] [Indexed: 04/17/2025] Open
Abstract
Topical instilling of commercial artificial tears (cAT, containing 0.1 % hyaluronic acid) is widely employed to alleviate clinical manifestations of mild dry eye disease (DED) by preventing the pathological change of corneal epithelium. However, it showed limited therapy effectiveness on heavy DED which has further involved corneal stroma, due to its low stroma-available for hyaluronic acid (HA) resulting from the barrier of corneal epithelium. The present study developed a new microneedle-dosage form of cAT (cAT-MN). This cAT-MN can overstride the corneal epithelium and act as a long-lasting protective agent. Compared to cAT dosing (4 times/day), cAT-MN with one treatment exerted significantly higher therapeutic effects on curbing benzalkonium chloride (BAC)-induced corneal stroma scaring as well as alleviating the DED symptoms in the first 5-day BAC exposure; whereas, showed limited effects in a 10-day BAC exposure. To expand the therapy effects, MNs containing various amounts of HA were prepared. Thereinto, HA(6 %)-MN recovered corneal damage to healthy levels, which could be attributed to adding stroma-available for HA both by increasing the amounts of HA-delivery and enhancing HA-permeation. This study explores a new drug-free microneedle-dosage form of cAT to cure corneal stroma disorders which has expanded its indication, promising a wide clinical use in ophthalmology.
Collapse
Affiliation(s)
- Baoshan Huang
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
- School of Biomedical Engineering, Wenzhou Medical University, Wenzhou, 325027, China
| | - Rui Zeng
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Xiao Liu
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Lu Pan
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Haitong Bai
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Jiachen Liao
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Wenkai Xu
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Hong Fu
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Kaihui Nan
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
- School of Biomedical Engineering, Wenzhou Medical University, Wenzhou, 325027, China
| | - Sen Lin
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
- School of Biomedical Engineering, Wenzhou Medical University, Wenzhou, 325027, China
| |
Collapse
|
4
|
Chen Y, Jiang H, Zhu H, He J, Chen L. Theranostics of osteoarthritis: Applications and prospects of precision targeting nanotechnology. Int J Pharm 2025; 676:125548. [PMID: 40216040 DOI: 10.1016/j.ijpharm.2025.125548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2025] [Revised: 03/22/2025] [Accepted: 03/30/2025] [Indexed: 04/19/2025]
Abstract
Osteoarthritis (OA), a complex degenerative joint disease driven by cartilage degeneration, synovial inflammation, and subchondral bone remodeling, lacks effective disease-modifying therapies. Precision-targeted nanotechnology has emerged as a breakthrough strategy, offering enhanced drug delivery, reduced toxicity, and synergistic diagnostic-therapeutic capabilities. This review summarizes OA pathogenesis, focusing on dysregulated immune networks and self-perpetuating synovial microenvironmental interactions. We discuss advanced nanomedicine approaches, which leverage OA-specific pathological cues for localized treatment. Innovations in cytokine modulation, photothermal therapy, and integrated theranostics (photoacoustic/fluorescence imaging) are highlighted as transformative tools for real-time diagnosis and personalized intervention. Despite progress, challenges such as biocompatibility optimization, clinical translation barriers, OA heterogeneity necessitate further development of multifunctional nanocarriers and rationaldesigns. This work underscores the potential of nanotechnology to advance OA therapeutics, bridging preclinical innovation with clinical applicability in pharmaceutical sciences.
Collapse
Affiliation(s)
- Yujing Chen
- Department of Orthopedics, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Hongyi Jiang
- Department of Orthopedics, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Haoran Zhu
- Department of Orthopedics, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jinyan He
- Department of Orthopedics, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Liang Chen
- Department of Orthopedics, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
5
|
Chu PC, Birhan YS, Zhao MH, Syu WJ, Chen PY, Lin YT, Lai PS. Size-controlled immunomodulatory and vaccine adjuvant potentials of self-assembled hyaluronic acid nanoparticles: Activation and recruitment of immune cells. Int J Biol Macromol 2025; 314:144265. [PMID: 40381765 DOI: 10.1016/j.ijbiomac.2025.144265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 05/02/2025] [Accepted: 05/14/2025] [Indexed: 05/20/2025]
Abstract
Immunotherapy has paramount importance in treating chronic immune diseases, and vaccine development. Hyaluronic acid (HA) has been shown to elicit molecular weight-related distinct immune responses. Nonetheless, the particle size-dependent immunomodulatory effects of hyaluronic acid nanoparticles (HA-NPs) remain blurred. Thus, the present study aimed at investigating the effect of polymer configuration and particle size of HA-NPs assembled from HA36K-ODA and HA360k-ODA analogs in modulating macrophage-related immune activities. In the in vitro experiments, HA-NPs of HA36k-ODA analogs garnered significantly enhanced nitrite production with low interleukin 6 (IL-6) and tumor necrosis factor α (TNF-α) secretion in macrophages. Furthermore, smaller (< 200 nm) HA36k-ODA NPs activated CD11b+ cells whereas a ∼ 130 nm HA36k-ODA15 NPs preferentially induced CD11c+ cells in vivo, indicating the influence of particle size on the antigen-presenting cells (APCs) recruitment. Interestingly, self-assembled HA36k-ODA15 NPs without ovalbumin (OVA) activated immature dendritic cells (DCs) and augmented their migration toward the lymph nodes (LNs). Notably, HA36k-ODA15-activated macrophages behaved like M1 phenotype macrophages. Overall, our findings garnered valuable insights about the design and application of HA-NPs for modulating the immune responses in nanoscale materials-based immunotherapy.
Collapse
Affiliation(s)
- Po-Cheng Chu
- Department of Chemistry, National Chung Hsing University, Taichung 402, Taiwan; Basic Research and Development Department, Powin Biomedical Co. Ltd., Taichung 428, Taiwan
| | - Yihenew Simegniew Birhan
- Department of Chemistry, National Chung Hsing University, Taichung 402, Taiwan; Department of Chemistry, College of Natural and Computational Sciences, Debre Markos University, P.O. Box 269, Debre Markos, Ethiopia
| | - Min-Han Zhao
- Department of Chemistry, National Chung Hsing University, Taichung 402, Taiwan
| | - Wei-Jhe Syu
- Basic Research and Development Department, Powin Biomedical Co. Ltd., Taichung 428, Taiwan
| | - Po-Yen Chen
- Department of Chemical and Biomolecular Engineering, University of Maryland, College Park, MD 20742, United States
| | - Yu-Tsen Lin
- Department of Chemistry, National Chung Hsing University, Taichung 402, Taiwan
| | - Ping-Shan Lai
- Department of Chemistry, National Chung Hsing University, Taichung 402, Taiwan; Ph.D. Program in Tissue Engineering and Regenerative Medicine, National Chung Hsing University, Taichung 402, Taiwan.
| |
Collapse
|
6
|
Guo Q, Nan R, Du Y, Wang R, Xie M, Li X, Li K, Xiang T, Zhou S. Hyaluronic acid composite hydrogel with enhanced lubrication and controllable drug release for the mitigation of osteoarthritis. Int J Biol Macromol 2025; 308:142677. [PMID: 40164244 DOI: 10.1016/j.ijbiomac.2025.142677] [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: 11/05/2024] [Revised: 02/28/2025] [Accepted: 03/28/2025] [Indexed: 04/02/2025]
Abstract
Excessive inflammation, overexpressed reactive oxygen species (ROS), degradation of the extracellular matrix, and high friction aggravate osteoarthritis (OA). Intra-articular injection for local drug delivery in the joint cavity enhances drug retention duration for OA treatment. Nevertheless, it remains a challenge to attenuate OA by thoroughly modulating the joint microenvironment, achieving controlled drug release, and enhancing lubrication. This study develops hyaluronic acid (HA) composite hydrogels, infused with gelatin microspheres containing a drug, utilizing dynamic interactions between phenylboronic acid and catechol to modulate the microenvironment by scavenging ROS, facilitating controlled drug release, and improving lubrication to mitigate OA. The composite hydrogels can be injected by intra-articular injection due to the shear-thinning properties, demonstrating broad ROS scavenging capacity and matrix metalloproteinase-9 responsive drug release. Both the in vitro and in vivo experiments prove the protective efficacy of the composite hydrogels against the degradation of cartilage matrix. Additionally, the hydrogels can offer efficient lubrication and effectively attenuate OA. Thus, the injectable HA composite hydrogels demonstrated potential in the management of OA.
Collapse
Affiliation(s)
- Qianru Guo
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, China; School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Rui Nan
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, China; School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Yuxiao Du
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, China; School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Rui Wang
- State Key Laboratory of Solid Lubrication, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences, Lanzhou 730000, China
| | - Meiming Xie
- Affiliated Hospital of Southwest Jiaotong University, The General Hospital of Western Theater Command, Chengdu 610031, China
| | - Xilin Li
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Kezhou Li
- Division of Pancreatic Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Tao Xiang
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, China; School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China.
| | - Shaobing Zhou
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, China; School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China.
| |
Collapse
|
7
|
Xu X, Huang X, Lin S, Yu G, Xiao J, Wang J, Song C, Khalilov F, Issaro N, Xu N, Gao S. Hyaluronic acid/ε-polylysine hydrogel enriched with Saussurea involucrata polysaccharide for improved skin dryness induced by ultraviolet radiation. Int J Biol Macromol 2025; 308:142718. [PMID: 40174445 DOI: 10.1016/j.ijbiomac.2025.142718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2025] [Revised: 03/21/2025] [Accepted: 03/30/2025] [Indexed: 04/04/2025]
Abstract
Ultraviolet B (UVB) radiation exposure is a major contributor to skin photoaging, ultimately resulting in dryness. Conventional treatments for this condition frequently exhibit limited efficacy due to poor skin penetration and are associated with significant side effects. Saussurea involucrate polysaccharide (SIP) has been shown to alleviate photoaging-induced dry skin; however, its effectiveness is limited due to low drug utilization and the need for frequent dosing. In this study, we developed hyaluronic acid (HA) and ε-polylysine (ε-PLL) hydrogels designed to enhance skin hydration and enable sustained drug release. The optimal formulation, designated A5E3, contained 5 % HA plus 3 % ε-PLL. The sustained release of SIP from A5E3 demonstrated superior therapeutic effects in a mouse model of skin photoaging, including reduced inflammation and apoptosis, as well as enhanced keratinocyte differentiation and lipid production. The A5E3 hydrogel not only enhanced the drug utilization of SIP but also provided a sustained moisturizing environment, promoting faster skin repair. These findings support the development of skincare products with superior moisturizing and reparative properties, underscoring the potential of hydrogels as effective carriers for treating UVB-induced skin dryness and damage.
Collapse
Affiliation(s)
- Xiashun Xu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China; College of Life and Environmental Sciences, Wenzhou University, Wenzhou, China
| | - Xulong Huang
- College of Life and Environmental Sciences, Wenzhou University, Wenzhou, China
| | - Shisheng Lin
- College of Life and Environmental Sciences, Wenzhou University, Wenzhou, China
| | - Guangdong Yu
- College of Life and Environmental Sciences, Wenzhou University, Wenzhou, China
| | - Jiali Xiao
- College of Life and Environmental Sciences, Wenzhou University, Wenzhou, China
| | - Junchao Wang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Chengyang Song
- College of Life and Environmental Sciences, Wenzhou University, Wenzhou, China
| | - Farid Khalilov
- College of Life and Environmental Sciences, Wenzhou University, Wenzhou, China
| | - Nipatha Issaro
- College of Life and Environmental Sciences, Wenzhou University, Wenzhou, China
| | - Nuo Xu
- College of Life and Environmental Sciences, Wenzhou University, Wenzhou, China.
| | - Shuang Gao
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China.
| |
Collapse
|
8
|
Rodella G, Préat V, Gallez B, Malfanti A. Design Strategies for Hyaluronic Acid-based Drug Delivery Systems in Cancer Immunotherapy. J Control Release 2025; 383:113784. [PMID: 40294800 DOI: 10.1016/j.jconrel.2025.113784] [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: 03/25/2025] [Revised: 04/22/2025] [Accepted: 04/24/2025] [Indexed: 04/30/2025]
Abstract
Despite its robust therapeutic potential, cancer immunotherapy has provided little progress towards improved survival rates for patients bearing immunologically refractory tumors. The implementation of advanced drug delivery systems represents a powerful means of improving cancer immunotherapy by relieving immunosuppression and promoting immune response; however, the overall impact of these systems on immunotherapy currently remains modest. Hyaluronic acid represents a widely used polymer in drug delivery; meanwhile, recent studies linking hyaluronic acid to the immune system make this polymer an attractive component in the design of next-generation cancer immunotherapies. Herein, we review our current understanding of the immunological properties of hyaluronic acid and discuss them in the context of bioactive functions and immune-related interactions with receptors, immune, and cancer cells. We analyze the potential of hyaluronic acid as a component in advanced drug delivery systems, highlighting strategies for the design of more effective vaccines and cancer chemo-immunotherapies. Finally, we discuss critical considerations to facilitate design and clinical translation to overcome existing challenges and maximize therapeutic potential.
Collapse
Affiliation(s)
- Giulia Rodella
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier 73 B1.73.12, 1200 Brussels, Belgium; UCLouvain, Louvain Drug Research Institute, Biomedical Magnetic Resonance, Avenue Mounier 73 B1.73.08, 1200 Brussels, Belgium
| | - Véronique Préat
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier 73 B1.73.12, 1200 Brussels, Belgium
| | - Bernard Gallez
- UCLouvain, Louvain Drug Research Institute, Biomedical Magnetic Resonance, Avenue Mounier 73 B1.73.08, 1200 Brussels, Belgium.
| | - Alessio Malfanti
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier 73 B1.73.12, 1200 Brussels, Belgium; Departement of Pharmaceutical and Pharmacological Sciences, University of Padova, Via F. Marzolo, 5, 35131 Padova, Italy.
| |
Collapse
|
9
|
Gao Q, Yao D, Yin Z, Yu G, Shi B, Wang J. Comprehensive multi-omics approach reveals potential therapeutic targets and agents for osteoarthritis. Postgrad Med J 2025; 101:464-474. [PMID: 39665162 DOI: 10.1093/postmj/qgae176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Revised: 10/14/2024] [Accepted: 11/25/2024] [Indexed: 12/13/2024]
Abstract
BACKGROUND The mechanisms underlying osteoarthritis (OA) remain unclear, and effective treatments are lacking. This study aims to identify OA-related genes and explore their potential in drug repositioning for OA treatment. METHODS Transcriptome-wide association studies (TWAS) were performed using genome-wide association studies summary data and expression quantitative trait loci data from the Genotype-Tissue Expression project. Differentially expressed genes between OA patients and healthy controls were identified using four datasets from the Gene Expression Omnibus database. Gene ontology and pathway enrichment analyses identified potential hub genes associated with OA. A network-based drug repositioning approach was applied to discover potential therapeutic drugs for OA. RESULTS Through TWAS and mRNA expression profiling, 7 and 167 OA-related genes were identified, respectively. From these, 128 OA-related genes were selected based on common biological processes. Using the maximal clique centrality algorithm, 10 core-related genes (JUN, VEGFA, FN1, CD44, PTGS2, STAT1, MAP 2K7, GRB2, EP300, and PXN) were identified for network-based drug repositioning. Consequently, 24 drugs were identified based on 128 OA-related genes and 23 drugs based on 10 core OA-related genes. Some identified drugs, such as dexamethasone, menadione, and hyaluronic acid, have been previously reported for OA and/or rheumatoid arthritis treatment. Network analysis also indicated that spironolactone, lovastatin, and atorvastatin may have potential in OA treatment. CONCLUSION This study identified potential OA-related genes and explored their roles in drug repositioning, suggesting the repurposing of existing drugs and the development of new therapeutic options for OA patients. Key message What is already known on this topic The exact pathogenesis of osteoarthritis (OA) remains unclear, and currently, there are no approved drugs that can prevent, halt, or inhibit the progression of OA. What this study adds We identified 128 OA-related genes and 10 core-related genes based on common biological processes revealed by TWAS and mRNA expression profiling. Using these genes, we discovered potential drugs for OA through the Network-based drug repositioning method. How this study might affect research, practice, or policy This study provides recommendations for repositioning existing drugs and developing new treatment options for patients with OA.
Collapse
Affiliation(s)
- Qingxia Gao
- Neck-Shoulder and Lumbocrural Pain Hospital of Shandong First Medical University, Shandong First Medical University (Shandong Academy of Medical Sciences), No. 18877, Jing 10 Road, Jinan 250000, Shandong, China
| | - Dawei Yao
- Endocrine and Metabolic Disease Hospital of Shandong First Medical University, No. 18877, Jing 10 Road, Jinan 250000, Shandong, China
| | - Zuozhen Yin
- Neck-Shoulder and Lumbocrural Pain Hospital of Shandong First Medical University, Shandong First Medical University (Shandong Academy of Medical Sciences), No. 18877, Jing 10 Road, Jinan 250000, Shandong, China
| | - Gongchang Yu
- Neck-Shoulder and Lumbocrural Pain Hospital of Shandong First Medical University, Shandong First Medical University (Shandong Academy of Medical Sciences), No. 18877, Jing 10 Road, Jinan 250000, Shandong, China
| | - Bin Shi
- Neck-Shoulder and Lumbocrural Pain Hospital of Shandong First Medical University, Shandong First Medical University (Shandong Academy of Medical Sciences), No. 18877, Jing 10 Road, Jinan 250000, Shandong, China
| | - Jiaying Wang
- Neck-Shoulder and Lumbocrural Pain Hospital of Shandong First Medical University, Shandong First Medical University (Shandong Academy of Medical Sciences), No. 18877, Jing 10 Road, Jinan 250000, Shandong, China
| |
Collapse
|
10
|
Chudzińska-Skorupinska J, Wawrzyńczak A, Feliczak-Guzik A. Carbohydrate-based polymer nanocarriers for environmentally friendly applications. Adv Colloid Interface Sci 2025; 338:103415. [PMID: 39884112 DOI: 10.1016/j.cis.2025.103415] [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: 07/15/2024] [Revised: 12/20/2024] [Accepted: 01/23/2025] [Indexed: 02/01/2025]
Abstract
Effective delivery of active substances and drugs is an important part of treatment. In order for a drug to work at the right place in the body, it must be transported there in the right way. For this reason, new carriers are being sought for active substances and drugs that can effectively deliver drugs to the target site without causing additional side effects. These include nanoparticles, microneedles, cubosomes and nanogels, among others. Recently, carriers based on biodegradable polymers such as hyaluronic acid or chitosan are becoming popular. In addition, modern carriers are designed to release the active ingredient in response to a specific agent. This paper reviews the literature from the past 5 years on novel delivery systems with medical, agricultural, food and cosmetic applications, with a special emphasis on the use of carbohydrate-based nanocarriers.
Collapse
Affiliation(s)
| | - Agata Wawrzyńczak
- Adam Mickiewicz University in Poznań, Faculty of Chemistry, Uniwersytetu Poznańskiego 8, 61-614 Poznań, Poland
| | - Agnieszka Feliczak-Guzik
- Adam Mickiewicz University in Poznań, Faculty of Chemistry, Uniwersytetu Poznańskiego 8, 61-614 Poznań, Poland.
| |
Collapse
|
11
|
Chen J, Wu G, Wu J, Jiao Z. Sodium alginate microspheres loaded with Quercetin/Mg nanoparticles as novel drug delivery systems for osteoarthritis therapy. J Orthop Surg Res 2025; 20:300. [PMID: 40108592 PMCID: PMC11924703 DOI: 10.1186/s13018-025-05698-z] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 03/09/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND Osteoarthritis (OA) is the most prevalent arthritic disease characterized by cartilage degradation and low-grade inflammation, for which there remains a lack of efficacious therapeutic interventions. Notably, mitigating the impact of oxidative stress (OS) and inflammatory factors could help alleviate or hinder the advancement of OA. Given the benefits of both quercetin (Que) and Magnesium ion (Mg2+) in OA treatment, coupled with the structural properties of Que, we have innovatively developed the Que-Mg2+ nanoparticles (NPs), aiming to deliver both Que and Mg2+ simultaneously and achieve enhanced therapeutic outcomes for OA. Moreover, to avoid the adverse reactions linked to frequent injections, sodium alginate (SA) microspheres encapsulating Que-Mg2+ NPs (Que-Mg@SA) were designed to treat the H2O2-induced OA cell model. METHODS Que-Mg@SA microspheres were synthesized using the ionotropic gelation technique, with calcium chloride acting as the cross-linking agent. Comprehensive characterization of the Que-Mg@SA was conducted through transmission electron microscope (TEM), dynamic light scattering (DLS), optical microscope, and scanning electron microscope (SEM), which provided detailed insights into their size, zeta potential, morphology, and micromorphology. Additionally, the microsphere swelling rate and Que release were evaluated. The biocompatibility of Que-Mg@SA microspheres, along with their impact on chondrocyte viability, were detected through CCK-8 assay and live/dead cell staining. Furthermore, the antioxidant and anti-inflammatory properties of Que-Mg@SA were evaluated by examining the ROS scavenging ability and pro-inflammatory factors levels, respectively. Finally, the regulatory influence of Que-Mg@SA microspheres on extracellular matrix (ECM) metabolism in OA was assessed by immunofluorescence staining and Western blot. RESULTS Characterization results revealed that Que-Mg NPs exhibit nanoscale diameter, exceptional stability, and good dispersibility, while Que-Mg@SA possesses high entrapment efficiency (EE%) and loading efficiency (LE%), pronounced hygroscopic properties, and sustained drug-release capabilities. Additionally, in vitro cellular assays revealed that the biocompatible Que-Mg@SA microspheres significantly restored chondrocyte viability, scavenged H2O2-induced excessive ROS, reduced the levels of inflammatory cytokines, upregulated cartilage anabolic gene expression, downregulated cartilage catabolic protease gene expression, and maintained the metabolic balance of cartilage tissue. CONCLUSION The functionalized Que-Mg@SA microspheres developed in our study hold great promise as a drug delivery system for OA and potentially other biomedical applications. CLINICAL TRIAL NUMBER Not applicable.
Collapse
Affiliation(s)
- Jun Chen
- Department of Orthopedics, Xianning Central Hospital, The First Affiliated Hospital of Hubei University of Science and Technology, NO. 228 Jingui Road, Xian'an District, Xianning, Hubei, 437100, China
| | - Guoya Wu
- Department of Orthopedics, Xianning Central Hospital, The First Affiliated Hospital of Hubei University of Science and Technology, NO. 228 Jingui Road, Xian'an District, Xianning, Hubei, 437100, China
| | - Jian Wu
- Department of Orthopedics, Xianning Central Hospital, The First Affiliated Hospital of Hubei University of Science and Technology, NO. 228 Jingui Road, Xian'an District, Xianning, Hubei, 437100, China
| | - Zhijian Jiao
- Department of Orthopedics, Xianning Central Hospital, The First Affiliated Hospital of Hubei University of Science and Technology, NO. 228 Jingui Road, Xian'an District, Xianning, Hubei, 437100, China.
| |
Collapse
|
12
|
Chen W, Ye Q, Zhang M, Xie R, Xu C. Lubrication for Osteoarthritis: From Single-Function to Multifunctional Lubricants. Int J Mol Sci 2025; 26:1856. [PMID: 40076486 PMCID: PMC11900089 DOI: 10.3390/ijms26051856] [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: 12/17/2024] [Revised: 02/14/2025] [Accepted: 02/17/2025] [Indexed: 03/14/2025] Open
Abstract
Osteoarthritis (OA) is a common degenerative joint disease that progressively destroys articular cartilage, leading to increased joint friction and severe pain. Therefore, OA can be treated by restoring the lubricating properties of cartilage. In this study, recent advances in lubricants for the treatment of OA are reviewed for both single-function and multifunctional lubricants. Single-function lubricants mainly include glycosaminoglycans, lubricin, and phospholipids, whereas multifunctional lubricants are composed of lubricating and anti-inflammatory bifunctional hydrogels, stem cell-loaded lubricating hydrogels, and drug-loaded lubricating nanoparticles. This review emphasizes the importance of restoring joint lubrication capacity for the treatment of OA and explores the structural features, lubrication properties, and role of these lubricants in modulating intracellular inflammatory responses and metabolism. Current challenges and future research directions in this field are also discussed, with the aim of providing a scientific basis and new ideas for the clinical treatment of OA.
Collapse
Affiliation(s)
- Wen Chen
- School of Rehabilitation Medicine, Gannan Medical University, Ganzhou 341000, China;
- Jiangxi Provincial Key Laboratory of Tissue Engineering (2024SSY06291), Gannan Medical University, Ganzhou 341000, China; (Q.Y.); (M.Z.)
| | - Qianwen Ye
- Jiangxi Provincial Key Laboratory of Tissue Engineering (2024SSY06291), Gannan Medical University, Ganzhou 341000, China; (Q.Y.); (M.Z.)
- School of Medical Information Engineering, Gannan Medical University, Ganzhou 341000, China
| | - Mingshuo Zhang
- Jiangxi Provincial Key Laboratory of Tissue Engineering (2024SSY06291), Gannan Medical University, Ganzhou 341000, China; (Q.Y.); (M.Z.)
- School of Medical Information Engineering, Gannan Medical University, Ganzhou 341000, China
| | - Renjian Xie
- Jiangxi Provincial Key Laboratory of Tissue Engineering (2024SSY06291), Gannan Medical University, Ganzhou 341000, China; (Q.Y.); (M.Z.)
- School of Medical Information Engineering, Gannan Medical University, Ganzhou 341000, China
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases (Ministry of Education), Gannan Medical University, Ganzhou 341000, China
| | - Chunming Xu
- School of Medical Information Engineering, Gannan Medical University, Ganzhou 341000, China
- School of Basic Medicine, Gannan Medical University, Ganzhou 341000, China
| |
Collapse
|
13
|
Chen M, Liu Y, Cao Y, Zhao C, Liu Q, Li N, Liu Y, Cui X, Liu P, Liang J, Fan Y, Wang Q, Zhang X. Remodeling the Proinflammatory Microenvironment in Osteoarthritis through Interleukin-1 Beta Tailored Exosome Cargo for Inflammatory Regulation and Cartilage Regeneration. ACS NANO 2025; 19:4924-4941. [PMID: 39848926 DOI: 10.1021/acsnano.4c16785] [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: 01/25/2025]
Abstract
Osteoarthritis (OA) presents a significant therapeutic challenge, with few options for preserving joint cartilage and repairing associated tissue damage. Inflammation is a pivotal factor in OA-induced cartilage deterioration and synovial inflammation. Recently, exosomes derived from human umbilical cord mesenchymal stem cells (HucMSCs) have gained recognition as a promising noncellular therapeutic modality, but their use is hindered by the challenge of harvesting a sufficient number of exosomes with effective therapeutic efficacy. Given that HucMSCs are highly sensitive to microenvironmental signals, we hypothesized that priming HucMSCs within a proinflammatory environment would increase the number of exosomes secreted with enhanced anti-inflammatory properties. Subsequent miRNA profiling and pathway analysis confirmed that interleukin-1 beta (IL-1β)-induced exosomes (C-Exos) exert positive effects through miRNA regulation and signaling pathway modulation. In vitro experiments revealed that C-Exos enhance chondrocyte functionality and cartilage matrix production, as well as macrophage polarization, thereby enhancing cartilage repair. C-Exos were encapsulated in hyaluronic acid hydrogel microspheres (HMs) to ensure sustained release, leading to substantial improvements in the inflammatory microenvironment and cartilage regeneration in a rat OA model. This study outlines a strategy to tailor exosome cargo for anti-inflammatory and cartilage regenerative purposes, with the functionalized HMs demonstrating potential for OA treatment.
Collapse
Affiliation(s)
- Manyu Chen
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, 29 Wangjiang Road, Chengdu 610064, P. R. China
| | - Yuhan Liu
- The Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121000, China
| | - Yi Cao
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, 29 Wangjiang Road, Chengdu 610064, P. R. China
| | - Chengkun Zhao
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, 29 Wangjiang Road, Chengdu 610064, P. R. China
| | - Quanying Liu
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Na Li
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, 29 Wangjiang Road, Chengdu 610064, P. R. China
- Sichuan Testing Center for Biomaterials and Medical Devices, Sichuan University, 29 Wangjiang Road, Chengdu 610000, China
| | - Yuan Liu
- Orthopedics Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu 610041, P. R. China
| | - Xiaolin Cui
- Cardiac and Osteochondral Tissue Engineering (COTE) Group, School of medicine, the Chinese University of Hong Kong, Shenzhen 518172, China
- Department of Orthopedic Surgery & Musculoskeletal Medicine, Centre for Bioengineering & Nanomedicine, University of Otago, Christchurch 8011, New Zealand
| | - Pengcheng Liu
- Chengdu Xiangyakanglin Biotechnology Co., Ltd, Chengdu 610213, China
| | - Jie Liang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, 29 Wangjiang Road, Chengdu 610064, P. R. China
- Sichuan Testing Center for Biomaterials and Medical Devices, Sichuan University, 29 Wangjiang Road, Chengdu 610000, China
| | - Yujiang Fan
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, 29 Wangjiang Road, Chengdu 610064, P. R. China
| | - Qiguang Wang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, 29 Wangjiang Road, Chengdu 610064, P. R. China
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, 29 Wangjiang Road, Chengdu 610064, P. R. China
| |
Collapse
|
14
|
Sheng W, Liao S, Cao S, Wei Y, Zhao J, Yue Y, Qin H, Qi T, Qian J, Lin J, Weng J, Chen Y, Wang D, Yu F, Liu P, Zeng H. Reactive oxygen species-sensitive fenofibrate-loaded dextran nanoparticles in alleviation of osteoarthritis. Carbohydr Polym 2025; 347:122768. [PMID: 39486995 DOI: 10.1016/j.carbpol.2024.122768] [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: 05/20/2024] [Revised: 09/12/2024] [Accepted: 09/17/2024] [Indexed: 11/04/2024]
Abstract
Osteoarthritis (OA) stands as a prevalent chronic joint pathology, emerging as a leading cause of disability on a global scale. However, the current therapeutic efficacy in OA treatment remains unsatisfactory. Chondrocyte ferroptosis has become to a critical target for OA treatment, while the fabrication of nanomedicines emerges as a promising strategy for OA treatment. Nevertheless, there exists a paucity of reported nanomedicine systems designed to combat chondrocyte ferroptosis for OA alleviation. In light of this, our study introduced a reactive oxygen species (ROS)-sensitive fenofibrate-loaded targeted nanoparticle (FN-CNPs) as a means of alleviating OA by suppressing chondrocyte ferroptosis. In vitro investigations demonstrated the FN-CNPs can achieve this through the reduction of lipid peroxidation and ROS levels, as well as the elevation of anti-ferroptosis markers (GPX4, FSP1, and ACSL3). Consequently, FN-CNPs exhibited significant anti-inflammatory effects and downregulated the expression of key catabolic mediators in vitro. Furthermore, in vivo studies underscored the ability of FN-CNPs to alleviate OA progression and protect cartilage. Collectively, these findings highlight the efficacy of FN-CNPs in mitigating OA progression by suppressing chondrocyte ferroptosis via regulating ROS levels, antioxidant systems and lipid metabolism of chondrocytes.
Collapse
Affiliation(s)
- Weibei Sheng
- Department of Bone & Joint Surgery, National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Shuai Liao
- Department of Bone & Joint Surgery, National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Siyang Cao
- Department of Bone & Joint Surgery, National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Yihao Wei
- Department of Bone & Joint Surgery, National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Jin Zhao
- Department of Bone & Joint Surgery, National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Yaohang Yue
- Department of Bone & Joint Surgery, National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Haotian Qin
- Department of Bone & Joint Surgery, National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Tiantian Qi
- Department of Bone & Joint Surgery, National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Junyu Qian
- Department of Bone & Joint Surgery, National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Jianjing Lin
- Department of Sports Medicine and Rehabilitation, Peking University Shenzhen Hospital, Shenzhen 518035, China
| | - Jian Weng
- Department of Bone & Joint Surgery, National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Yingqi Chen
- Department of Bone & Joint Surgery, National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Deli Wang
- Department of Bone & Joint Surgery, National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Fei Yu
- Department of Bone & Joint Surgery, National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen 518036, China.
| | - Peng Liu
- Department of Bone & Joint Surgery, National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen 518036, China.
| | - Hui Zeng
- Department of Bone & Joint Surgery, National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen 518036, China; Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen 518035, China.
| |
Collapse
|
15
|
Takagi Y, Kage M. Hyaluronan Tetrasaccharides Penetrate into the Skin by Passive Diffusion and Contribute to Skin Health. Chem Pharm Bull (Tokyo) 2025; 73:284-290. [PMID: 40175107 DOI: 10.1248/cpb.c23-00909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2025]
Abstract
Hyaluronan (HA) is a commonly used material in cosmetics and pharmaceuticals because of its various pharmacological activities. However, because of its large molecular weight, HA penetrates the skin very poorly and most of it remains on the skin surface. Thus, topically applied HA could not be expected to function biologically in the skin. However, we have confirmed that HA tetrasaccharides (HA4), which is the smallest unit of HA, penetrate into the skin by passive diffusion and affect epidermal metabolism. Topical treatment of HA4 rescues the epidermal damage caused by long-term UVA irradiation. Furthermore, various biological functions of HA4 to maintain healthy skin was observed in cell culture studies. This review describes the skin permeability of HA4 and how it contributes to healthy skin.
Collapse
Affiliation(s)
- Yutaka Takagi
- Laboratory of Dermatological Physiology, Faculty of Pharmacy and Pharmaceutical Sciences, Josai University
| | - Madoka Kage
- Laboratory of Dermatological Physiology, Faculty of Pharmacy and Pharmaceutical Sciences, Josai University
| |
Collapse
|
16
|
Lim MJ, Oh H, Jeon J, Cho C, Lee JS, Hwang Y, Kim SJ, Mo JS, Son P, Kang HC, Choi WI, Yang S. An intra articular injectable Mitocelle recovers dysfunctional mitochondria in cellular organelle disorders. Bioact Mater 2025; 43:305-318. [PMID: 39399840 PMCID: PMC11467566 DOI: 10.1016/j.bioactmat.2024.09.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 08/18/2024] [Accepted: 09/17/2024] [Indexed: 10/15/2024] Open
Abstract
Mitochondrial dysfunction increases ROS production and is closely related to many degenerative cellular organelle diseases. The NOX4-p22phox axis is a major contributor to ROS production and its dysregulation is expected to disrupt mitochondrial function. However, the field lacks a competitive inhibitor of the NOX4-p22phox interaction. Here, we created a povidone micelle-based Prussian blue nanozyme that we named "Mitocelle" to target the NOX4-p22phox axis, and characterized its impact on the major degenerative cellular organelle disease, osteoarthritis (OA). Mitocelle is composed of FDA-approved and biocompatible materials, has a regular spherical shape, and is approximately 88 nm in diameter. Mitocelle competitively inhibits the NOX4-p22phox interaction, and its uptake by chondrocytes can protect against mitochondrial malfunction. Upon intra-articular injection to an OA mouse model, Mitocelle shows long-term stability, effective uptake into the cartilage matrix, and the ability to attenuate joint degradation. Collectively, our findings suggest that Mitocelle, which functions as a competitive inhibitor of NOX4-p22phox, may be suitable for translational research as a therapeutic for OA and cellular organelle diseases related to dysfunctional mitochondria.
Collapse
Affiliation(s)
- Min Ju Lim
- Department of Biological Sciences, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Department of Biomedical Sciences, Graduate School, Ajou University School of Medicine, Suwon, 16499, Republic of Korea
| | - Hyeryeon Oh
- Center for Bio-Healthcare Materials, Bio-Convergence Materials R&D Division, Korea Institute of Ceramic Engineering and Technology, 202, Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju, Chungbuk, 28160, Republic of Korea
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology, 123, Cheomdan-gwagiro, Buk-gu, Gwangju, 61005, Republic of Korea
| | - Jimin Jeon
- Department of Biological Sciences, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Chanmi Cho
- Department of Biological Sciences, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Jin Sil Lee
- Center for Bio-Healthcare Materials, Bio-Convergence Materials R&D Division, Korea Institute of Ceramic Engineering and Technology, 202, Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju, Chungbuk, 28160, Republic of Korea
- Drug Manufacturing Center, Daegu-Gyeongbuk Medical Innovation Foundation (K-MEDI Hub), Daegu, 41061, Republic of Korea
| | - Yiseul Hwang
- Department of Biomedical Sciences, Graduate School, Ajou University School of Medicine, Suwon, 16499, Republic of Korea
- Department of Physiology, Ajou University School of Medicine, Suwon, Gyeonggi, 16499, Republic of Korea
| | - Seok Jung Kim
- Department of Orthopedic Surgery, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Jung-Soon Mo
- Department of Biomedical Sciences, Graduate School, Ajou University School of Medicine, Suwon, 16499, Republic of Korea
- Institute of Medical Science, Ajou University School of Medicine, Suwon, 16499, Republic of Korea
| | - Panmo Son
- Center for Bio-Healthcare Materials, Bio-Convergence Materials R&D Division, Korea Institute of Ceramic Engineering and Technology, 202, Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju, Chungbuk, 28160, Republic of Korea
- Department of Applied Bioengineering, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, 08826, Republic of Korea
| | - Ho Chul Kang
- Department of Biomedical Sciences, Graduate School, Ajou University School of Medicine, Suwon, 16499, Republic of Korea
- Department of Physiology, Ajou University School of Medicine, Suwon, Gyeonggi, 16499, Republic of Korea
| | - Won Il Choi
- Center for Bio-Healthcare Materials, Bio-Convergence Materials R&D Division, Korea Institute of Ceramic Engineering and Technology, 202, Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju, Chungbuk, 28160, Republic of Korea
| | - Siyoung Yang
- Department of Biological Sciences, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| |
Collapse
|
17
|
Wu D, Yang S, Gong Z, Zhu X, Hong J, Wang H, Xu W, Lai J, Wang X, Lu J, Fang X, Jiang G, Zhu J. Enhanced therapeutic potential of a self-healing hyaluronic acid hydrogel for early intervention in osteoarthritis. Mater Today Bio 2024; 29:101353. [PMID: 39687801 PMCID: PMC11647215 DOI: 10.1016/j.mtbio.2024.101353] [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: 05/23/2024] [Revised: 11/09/2024] [Accepted: 11/18/2024] [Indexed: 12/18/2024] Open
Abstract
Osteoarthritis (OA) is characterized by symptoms such as abnormal lubrication function of synovial fluid and heightened friction on the cartilage surface in its early stages, prior to evident cartilage damage. Current early intervention strategies employing lubricated hydrogels to shield cartilage from friction often overlook the significance of hydrogel-cartilage adhesion and enhancement of the cartilage extracellular matrix (ECM). Herein, we constructed a hydrogel based on dihydrazide-modified hyaluronic acid (HA) (AHA) and catechol-conjugated aldehyde-modified HA (CHA), which not only adheres to the cartilage surface as an effective lubricant but also improves the extracellular environment of chondrocytes in OA. Material characterization experiments on AHA/CHA hydrogels with varying concentrations validated their exceptional self-healing capabilities, superior injectability and viscoelasticity, sustained adhesion strength to cartilage, and a low friction coefficient. Chondrocytes exhibited robust adhesion and proliferation on the AHA/CHA hydrogel surface, with the upregulation of cartilage matrix protein expression. Intra-articular injection of AHA/CHA hydrogels was performed following destabilization of the medial meniscus (DMM) surgery in mice to assess its protective effect on cartilage. The AHA/CHA hydrogel effectively attenuated the degree of cartilage wear, facilitated chondrocytes' anabolic metabolism, and restored the ECM of cartilage. Therefore, the AHA/CHA hydrogel emerges as a promising therapeutic approach in clinical practices of OA treatment.
Collapse
Affiliation(s)
- Dongze Wu
- Department of Spinal Surgery, The First Affiliated Hospital of Ningbo University, Ningbo, 315000, Zhejiang, China
| | - Shuhui Yang
- School of Materials Science and Engineering, Zhejiang-Mauritius Joint Research Center for Biomaterials and Tissue Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, Zhejiang, China
| | - Zhe Gong
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine & Zhejiang Key Laboratory of Mechanism Research and Precision Repair of Orthopaedic Trauma and Aging Diseases, Hangzhou, 310016, Zhejiang, China
| | - Xinxin Zhu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, 310000, Zhejiang, China
| | - Juncong Hong
- Department of Anesthesiology, The First People's Hospital of Linping District, Hangzhou, 311100, Zhejiang, China
| | - Haitao Wang
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine & Zhejiang Key Laboratory of Mechanism Research and Precision Repair of Orthopaedic Trauma and Aging Diseases, Hangzhou, 310016, Zhejiang, China
| | - Wenbin Xu
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine & Zhejiang Key Laboratory of Mechanism Research and Precision Repair of Orthopaedic Trauma and Aging Diseases, Hangzhou, 310016, Zhejiang, China
| | - Juncheng Lai
- School of Materials Science and Engineering, Zhejiang-Mauritius Joint Research Center for Biomaterials and Tissue Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, Zhejiang, China
| | - Xiumei Wang
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, China
| | - Jiye Lu
- Department of Spinal Surgery, The First Affiliated Hospital of Ningbo University, Ningbo, 315000, Zhejiang, China
| | - Xiangqian Fang
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine & Zhejiang Key Laboratory of Mechanism Research and Precision Repair of Orthopaedic Trauma and Aging Diseases, Hangzhou, 310016, Zhejiang, China
| | - Guoqiang Jiang
- Department of Spinal Surgery, The First Affiliated Hospital of Ningbo University, Ningbo, 315000, Zhejiang, China
| | - Jinjin Zhu
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine & Zhejiang Key Laboratory of Mechanism Research and Precision Repair of Orthopaedic Trauma and Aging Diseases, Hangzhou, 310016, Zhejiang, China
| |
Collapse
|
18
|
Wang X, Zeng J, Gan D, Ling K, He M, Li J, Lu Y. Recent Strategies and Advances in Hydrogel-Based Delivery Platforms for Bone Regeneration. NANO-MICRO LETTERS 2024; 17:73. [PMID: 39601916 PMCID: PMC11602938 DOI: 10.1007/s40820-024-01557-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 10/01/2024] [Indexed: 11/29/2024]
Abstract
Bioactive molecules have shown great promise for effectively regulating various bone formation processes, rendering them attractive therapeutics for bone regeneration. However, the widespread application of bioactive molecules is limited by their low accumulation and short half-lives in vivo. Hydrogels have emerged as ideal carriers to address these challenges, offering the potential to prolong retention times at lesion sites, extend half-lives in vivo and mitigate side effects, avoid burst release, and promote adsorption under physiological conditions. This review systematically summarizes the recent advances in the development of bioactive molecule-loaded hydrogels for bone regeneration, encompassing applications in cranial defect repair, femoral defect repair, periodontal bone regeneration, and bone regeneration with underlying diseases. Additionally, this review discusses the current strategies aimed at improving the release profiles of bioactive molecules through stimuli-responsive delivery, carrier-assisted delivery, and sequential delivery. Finally, this review elucidates the existing challenges and future directions of hydrogel encapsulated bioactive molecules in the field of bone regeneration.
Collapse
Affiliation(s)
- Xiao Wang
- Scientific and Technological Innovation Center for Biomedical Materials and Clinical Research, Guangyuan Key Laboratory of Multifunctional Medical Hydrogel, Guangyuan Central Hospital, Guangyuan, 628000, People's Republic of China
| | - Jia Zeng
- Scientific and Technological Innovation Center for Biomedical Materials and Clinical Research, Guangyuan Key Laboratory of Multifunctional Medical Hydrogel, Guangyuan Central Hospital, Guangyuan, 628000, People's Republic of China
| | - Donglin Gan
- Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, Jiangsu Key Laboratory of Bio-Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing, 210023, People's Republic of China
| | - Kun Ling
- Scientific and Technological Innovation Center for Biomedical Materials and Clinical Research, Guangyuan Key Laboratory of Multifunctional Medical Hydrogel, Guangyuan Central Hospital, Guangyuan, 628000, People's Republic of China
| | - Mingfang He
- Scientific and Technological Innovation Center for Biomedical Materials and Clinical Research, Guangyuan Key Laboratory of Multifunctional Medical Hydrogel, Guangyuan Central Hospital, Guangyuan, 628000, People's Republic of China.
| | - Jianshu Li
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, People's Republic of China.
| | - Yongping Lu
- Scientific and Technological Innovation Center for Biomedical Materials and Clinical Research, Guangyuan Key Laboratory of Multifunctional Medical Hydrogel, Guangyuan Central Hospital, Guangyuan, 628000, People's Republic of China.
| |
Collapse
|
19
|
Cardoza JV, Ali Z, Simon S, Thakkar D, George SS, Isaac SP. The Role of Nanoparticles in Accelerating Tissue Recovery and Inflammation Control in Physiotherapy Practices. Cureus 2024; 16:e73540. [PMID: 39669817 PMCID: PMC11636964 DOI: 10.7759/cureus.73540] [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] [Accepted: 11/12/2024] [Indexed: 12/14/2024] Open
Abstract
Physiotherapy has significantly evolved since its inception in the late 19th century, expanding into various specializations such as sports, neurology, and wound care. Its primary goal is to restore or enhance bodily functions through therapeutic interventions, aiding in conditions ranging from injuries to chronic pain. Tissue recovery, which involves repair and regeneration, is a critical aspect of physiotherapy. This natural process is influenced by factors like inflammation and injury severity. Nanotechnology, a relatively recent advancement, has transformed medicine, including wound care, through innovations in drug delivery, diagnostics, and anti-inflammatory treatments. Nanoparticles, owing to their small size and enhanced bioavailability, play a crucial role in improving drug delivery, increasing the efficacy of treatments, and promoting faster recovery. In the context of tissue healing, nanoparticles aid in cell proliferation, inflammation control, and scar reduction, among other therapeutic benefits. They are increasingly used in physiotherapy applications, to support tissue regeneration and inflammation management. This review examines the role of nanoparticles in physiotherapy, with a focus on their application in wound healing, muscle recovery, and inflammation control. It discusses various in-vitro and in-vivo studies that have explored the therapeutic potential of nanoparticles in these domains, providing insights into their mechanisms of action and effectiveness in promoting tissue regeneration and managing inflammation in physiotherapy settings.
Collapse
Affiliation(s)
| | - Zeeshan Ali
- Physiology, Krupanidhi College of Physiotherapy, Bengaluru, IND
| | - Simi Simon
- Biochemistry, Krupanidhi College of Physiotherapy, Bengaluru, IND
| | - Darshni Thakkar
- Physiotherapy, Krupanidhi college of physiotherapy, Bengaluru, IND
| | - Sudhan S George
- Physiotherapy, Krupanidhi College of Physiotherapy, Bengaluru, IND
| | | |
Collapse
|
20
|
Yu P, Peng X, Sun H, Xin Q, Kang H, Wang P, Zhao Y, Xu X, Zhou G, Xie J, Li J. Inspired by lubricin: a tailored cartilage-armor with durable lubricity and autophagy-activated antioxidation for targeted therapy of osteoarthritis. MATERIALS HORIZONS 2024; 11:5352-5365. [PMID: 39143938 DOI: 10.1039/d4mh00812j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/16/2024]
Abstract
Osteoarthritis (OA), which disables articular cartilage, affects millions of people. The self-healing capacity is inhibited by internal oxidative stress and external lubrication deficiency and enzymatic degradation. To overcome these challenges, a tailored cartilage-armor is designed to ameliorate the inflamed cartilage, which is implemented by a novel collagen type II (Col II)-binding peptide conjugated zwitterionic polymer (PSB-b-PColBP, PSP). By mimicking natural lubricin, PSP specifically targets the cartilage surface and forms an in situ hydration armor. This engineered cartilage-armor can prevent enzymatic cartilage degradation (nearly 100% resistance to catabolic enzymes) and provide durable lubrication properties (COF < 0.013 for 500 cycles). An autophagy-activation process, absent in previous biomimetic lubricants, enhances the enzymatic activity of the tailored cartilage-armor, offering effective anti-oxidant properties to suppress oxidative stress. By inhibiting the PI3K-Akt/NF-κB signaling pathway, chondrocytes protected by the tailored armor can secrete a cartilage matrix even in inflammatory microenvironments. In OA rat models, osteophyte formation and the inflammatory response have been inhibited by the cartilage-armor, demonstrating a therapeutic effect comparable to most drug-loaded systems. This study underscores the potential of tailoring cartilage-armor with the cartilage targeting and autophagy-activating properties in integrating offensive-defensive mechanisms for cartilage remodeling. This represents an alternative strategy for clinical OA therapy.
Collapse
Affiliation(s)
- Peng Yu
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Med-X Center for Materials, Sichuan University, Chengdu 610065, P. R. China.
| | - Xu Peng
- Experimental and Research Animal Institute, Sichuan University, Chengdu 610207, P. R. China
| | - Hui Sun
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Med-X Center for Materials, Sichuan University, Chengdu 610065, P. R. China.
| | - Qiangwei Xin
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Med-X Center for Materials, Sichuan University, Chengdu 610065, P. R. China.
| | - Han Kang
- Life Science Core Facilities, College of Life Sciences, Sichuan University, Chengdu 610065, P. R. China
| | - Peng Wang
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Med-X Center for Materials, Sichuan University, Chengdu 610065, P. R. China.
| | - Yao Zhao
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Med-X Center for Materials, Sichuan University, Chengdu 610065, P. R. China.
| | - Xinyuan Xu
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Med-X Center for Materials, Sichuan University, Chengdu 610065, P. R. China.
| | - Guangwu Zhou
- School of Aeronautics and Astronautics, Sichuan University, Chengdu 610207, P. R. China
| | - Jing Xie
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Med-X Center for Materials, Sichuan University, Chengdu 610065, P. R. China.
| | - Jianshu Li
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Med-X Center for Materials, Sichuan University, Chengdu 610065, P. R. China.
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, P. R. China
| |
Collapse
|
21
|
Chen K, Wang J, Cao J, Liu F, Fang J, Zheng W, Liu S, Zhao Y, Shuai X, Huang J, Chen B. Enzyme-responsive microgel with controlled drug release, lubrication and adhesion capability for osteoarthritis attenuation. Acta Biomater 2024:S1742-7061(24)00618-4. [PMID: 39427765 DOI: 10.1016/j.actbio.2024.10.026] [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: 04/16/2024] [Revised: 10/15/2024] [Accepted: 10/17/2024] [Indexed: 10/22/2024]
Abstract
The treatment of osteoarthritis (OA) remains challenging due to the narrow therapeutic window and rapid clearance of therapeutic agents, even with intra-articular administration, resulting in a low treatment index. Recent advancements in local drug delivery systems have yet to overcome the issues of uncontrolled burst release and short retention time, leading to suboptimal OA treatment outcome. Herein, we developed a methacrylate-crosslinking hyaluronic acid (HA) microgel (abbreviated as CXB-HA-CBP) that covalently conjugates the anti-inflammatory drug celecoxib (CXB) via a metalloproteinase-2 (MMP-2)-responsive peptide linker (GGPLGLAGGC) and a collagen II binding peptide (WYRGRLC). The GGPLGLAGGC linker is specifically cleaved by the overexpressed MMP-2 enzyme within the OA joint, enabling the sustained and on-demand release of CXB entity. The synergistic action of CXB and HA effectively inhibited macrophage activation and reduced the production of pro-inflammatory cytokines, protecting chondrocytes from damage. Furthermore, the collagen II peptide introduced on the microgel surface enabled a cartilage-binding function to form an artificial lubrication microgel layer on the cartilage surface to reduce cartilage wear. The CXB-HA-CBP microgel showed an extended retention time of up to 18 days in the affected joint, leading to an effective OA treatment in rats. This sophistically designed microgel, characterized by the prolonged retention time, sustained drug delivery, and enhanced lubrication, presents a promising biomedicine for OA treatment. STATEMENT OF SIGNIFICANCE: A new methacrylate-crosslinking hyaluronic acid (HA) microgel, covalently conjugated with the celecoxib (CXB)-GGPLGLAGGC and the collagen II binding peptide (CBP, peptide sequence: WYRGRLC), was developed. The overexpressed MMP-2 in OA joint cleaved the GGPLGLAGGC linker to trigger the CXB moiety release. Besides, the CBP on the surface of microgels enabled a cartilage-attaching ability, resulting in a prolonged retention time and an improved lubrication property in joint. This advanced drug-loading microgel remarkably reduced macrophage activation and pro-inflammation cytokine production, while protecting the chondrocytes via a dual action of CXB and HA. This study demonstrated that the enzyme-responsive drug-loading microgel could serve as an platform to efficiently attenuate osteoarthritis.
Collapse
Affiliation(s)
- Keyu Chen
- Division of Orthopaedic Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jiachen Wang
- PCFM Lab of Ministry of Education, School of Materials Science and Engineering, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Jue Cao
- Division of Orthopaedic Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Fei Liu
- Division of Orthopaedic Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jintao Fang
- Department of Microsurgery, Trauma and Hand Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Weixin Zheng
- Division of Orthopaedic Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Shubo Liu
- Division of Orthopaedic Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yuexin Zhao
- Division of Orthopaedic Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Xintao Shuai
- Nanomedicine Research Center, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China; PCFM Lab of Ministry of Education, School of Materials Science and Engineering, Sun Yat-Sen University, Guangzhou, Guangdong, China.
| | - Jinsheng Huang
- Department of Urology, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong, China.
| | - Bin Chen
- Division of Orthopaedic Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
22
|
Sim YE, Kim CL, Kim DH, Hong JA, Lee IJ, Kwak JY, Kang LJ, Mo JS. Rosmarinic acid promotes cartilage regeneration through Sox9 induction via NF-κB pathway inhibition in mouse osteoarthritis progression. Heliyon 2024; 10:e38936. [PMID: 39444399 PMCID: PMC11497390 DOI: 10.1016/j.heliyon.2024.e38936] [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: 03/24/2024] [Revised: 10/02/2024] [Accepted: 10/02/2024] [Indexed: 10/25/2024] Open
Abstract
Background The natural polyphenolic compound known as Rosmarinic acid (RosA) can be found in various plants. Although its potential health benefits have been extensively studied, its effect on osteoarthritis (OA) progression and cartilage regeneration function still needs to be fully elucidated in OA animal models. This study elucidated the effect of RosA on OA progression and cartilage regeneration. Methods In vitro assessments were conducted using RT-PCR, qRT-PCR, Western blotting, and ELISA to measure the effects of RosA. The molecular mechanisms of RosA were determined by analyzing the translocation of p65 into the nucleus using immunocytochemistry (ICC). Histological analysis of cartilage explant was performed using alcian blue staining and immunohistochemistry (IHC). For in vivo analysis, the destabilization of the medial meniscus (DMM)-induced OA mouse model was utilized to evaluate cartilage destruction through Safranin-O staining. The expression of catabolic and anabolic factors in mice knee joints was quantified by immunohistochemistry. Results The expression of catabolic factors in chondrocytes was significantly impeded by RosA. It also suppressed the NF-κB signaling pathway by decreasing phosphorylation of p65 and reducing degradation of IκB protein. In ex vivo experiments, RosA protected sulfated proteoglycan erosion triggered by IL-1β and suppressed the catabolic factors in cartilage explant. RosA treatment in animal models resulted in preventing cartilage destruction and reducing catabolic factors in the cartilage. RosA was also found to promote the expression of Sox9, Col2a1, and Acan in vitro, ex vivo, and in vivo analyses. Conclusions RosA attenuated the OA progression by suppressing the catabolic factors expression. These effects were facilitated through the suppression of the NF-κB signaling pathway. Additionally, it promotes cartilage regeneration by inducing anabolic factors. Therefore, RosA shows potential as an effective therapeutic agent for treating OA.
Collapse
Affiliation(s)
- Ye Eun Sim
- Department of Biomedical Sciences, Graduate School, Ajou University School of Medicine, Suwon, 16499, South Korea
| | - Cho-Long Kim
- Department of Biomedical Sciences, Graduate School, Ajou University School of Medicine, Suwon, 16499, South Korea
| | - Dong Hyun Kim
- Department of Biomedical Sciences, Graduate School, Ajou University School of Medicine, Suwon, 16499, South Korea
| | - Ji-Ae Hong
- Jeonnam Bioindustry Foundation, Jeonnam Institute of Natural Resources Research, Jeollanamdo, 59338, South Korea
| | - In-Jeong Lee
- Three-Dimensional Immune System Imaging Core Facility, Ajou University, Suwon, 16499, South Korea
| | - Jong-Young Kwak
- Three-Dimensional Immune System Imaging Core Facility, Ajou University, Suwon, 16499, South Korea
- Department of Pharmacology, Ajou University School of Medicine, Suwon, 16499, South Korea
| | - Li-Jung Kang
- Three-Dimensional Immune System Imaging Core Facility, Ajou University, Suwon, 16499, South Korea
- Institute of Medical Science, Ajou University School of Medicine, Suwon, 16499, South Korea
| | - Jung-Soon Mo
- Institute of Medical Science, Ajou University School of Medicine, Suwon, 16499, South Korea
| |
Collapse
|
23
|
Feng H, Ang K, Guan P, Li J, Meng H, Yang J, Fan L, Sun Y. Application of adhesives in the treatment of cartilage repair. INTERDISCIPLINARY MEDICINE 2024; 2. [DOI: 10.1002/inmd.20240015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Accepted: 05/08/2024] [Indexed: 01/04/2025]
Abstract
AbstractFrom degeneration causing intervertebral disc issues to trauma‐induced meniscus tears, diverse factors can injure the different types of cartilage. This review highlights adhesives as a promising and rapidly implemented repair strategy. Compared to traditional techniques such as sutures and wires, adhesives offer several advantages. Importantly, they seamlessly connect with the injured tissue, deliver bioactive substances directly to the repair site, and potentially alleviate secondary problems like inflammation or degeneration. This review delves into the cutting‐edge advancements in adhesive technology, specifically focusing on their effectiveness in cartilage injury treatment and their underlying mechanisms. We begin by exploring the material characteristics of adhesives used in cartilage tissue, focusing on essential aspects like adhesion, biocompatibility, and degradability. Subsequently, we investigate the various types of adhesives currently employed in this context. Our discussion then moves to the unique role adhesives play in addressing different cartilage injuries. Finally, we acknowledge the challenges currently faced by this promising technology.
Collapse
Affiliation(s)
- Haoyang Feng
- Department of Pediatric Orthopedics The Third Affiliated Hospital of Southern Medical University Guangzhou China
| | - Kai Ang
- Department of Pediatric Orthopedics The Third Affiliated Hospital of Southern Medical University Guangzhou China
| | - Pengfei Guan
- Department of Pediatric Orthopedics The Third Affiliated Hospital of Southern Medical University Guangzhou China
| | - Junji Li
- Department of Pediatric Orthopedics The Third Affiliated Hospital of Southern Medical University Guangzhou China
| | - Huan Meng
- Postdoc Cartilage Biology AO Research Institute Davos Davos Platz Wellington Switzerland
| | - Jian Yang
- Biomedical Engineering Program School of Engineering Westlake University Hangzhou China
| | - Lei Fan
- Department of Orthopedic Surgery Nanfang Hospital Southern Medical University Guangzhou China
| | - Yongjian Sun
- Department of Pediatric Orthopedics The Third Affiliated Hospital of Southern Medical University Guangzhou China
| |
Collapse
|
24
|
Chen TY, Chen KC, Zhang YH, Lin CA, Hsu WY, Lin NY, Lai PS. Development of a dexamethasone-hyaluronic acid conjugate with selective targeting effect for acute lung injury therapy. Int J Biol Macromol 2024; 280:136149. [PMID: 39353517 DOI: 10.1016/j.ijbiomac.2024.136149] [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: 05/28/2024] [Revised: 09/22/2024] [Accepted: 09/28/2024] [Indexed: 10/04/2024]
Abstract
Acute lung injury (ALI), a critical complication of COVID-19, is characterized by widespread inflammation and severe pulmonary damage, necessitating intensive care for those affected. Although glucocorticoids (GCs), such as dexamethasone (Dex), have been employed clinically to lower mortality, their nonspecific systemic distribution has led to significant side effects, limiting their use in ALI treatment. In this study, we explored the conjugation of Dex to hyaluronic acid (HA) to achieve targeted delivery to inflamed lung tissues. We achieved a conjugation efficiency exceeding 98 % using a cosolvent system, with subsequent ester bond cleavage releasing the active Dex, as verified by liquid chromatography. Biodistribution and cellular uptake studies indicated the potential of the HA conjugate for cluster of differentiation 44 (CD44)-mediated targeting and accumulation. In a lipopolysaccharide-induced ALI mouse model, intravenous (IV) HA-Dex administration showed superior anti-inflammatory effects compared to free Dex administration. Flow cytometry analysis suggested that the HA conjugate preferentially accumulated in lung macrophages, suggesting the possibility of reducing clinical Dex dosages through this targeted delivery approach.
Collapse
Affiliation(s)
- Tzu-Yang Chen
- Department of Chemistry, National Chung Hsing University, Taichung 40227, Taiwan; Basic Research Division, Holy Stone Healthcare Co., Ltd., 114 Taipei, Taiwan
| | - Ke-Cheng Chen
- Department of Surgery, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Yu-Han Zhang
- Department of Chemistry, National Chung Hsing University, Taichung 40227, Taiwan
| | - Chih-An Lin
- Ph.D. Program of Tissue Engineering and Regenerative Medicine, National Chung Hsing University, Taichung 40227, Taiwan
| | - Wan-Yun Hsu
- Department of Chemistry, National Chung Hsing University, Taichung 40227, Taiwan
| | - Neng-Yu Lin
- Graduate Institute of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ping-Shan Lai
- Department of Chemistry, National Chung Hsing University, Taichung 40227, Taiwan; Ph.D. Program of Tissue Engineering and Regenerative Medicine, National Chung Hsing University, Taichung 40227, Taiwan.
| |
Collapse
|
25
|
Donati L, Valicenti ML, Giannoni S, Morena F, Martino S. Biomaterials Mimicking Mechanobiology: A Specific Design for a Specific Biological Application. Int J Mol Sci 2024; 25:10386. [PMID: 39408716 PMCID: PMC11476540 DOI: 10.3390/ijms251910386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 09/24/2024] [Accepted: 09/25/2024] [Indexed: 10/20/2024] Open
Abstract
Mechanosensing and mechanotransduction pathways between the Extracellular Matrix (ECM) and cells form the essential crosstalk that regulates cell homeostasis, tissue development, morphology, maintenance, and function. Understanding these mechanisms involves creating an appropriate cell support that elicits signals to guide cellular functions. In this context, polymers can serve as ideal molecules for producing biomaterials designed to mimic the characteristics of the ECM, thereby triggering responsive mechanisms that closely resemble those induced by a natural physiological system. The generated specific stimuli depend on the different natural or synthetic origins of the polymers, the chemical composition, the assembly structure, and the physical and surface properties of biomaterials. This review discusses the most widely used polymers and their customization to develop biomaterials with tailored properties. It examines how the characteristics of biomaterials-based polymers can be harnessed to replicate the functions of biological cells, making them suitable for biomedical and biotechnological applications.
Collapse
Affiliation(s)
- Leonardo Donati
- Department of Chemistry, Biology and Biotechnology, Biochemical and Biotechnological Sciences, University of Perugia, 06122 Perugia, Italy
| | - Maria Luisa Valicenti
- Department of Chemistry, Biology and Biotechnology, Biochemical and Biotechnological Sciences, University of Perugia, 06122 Perugia, Italy
| | - Samuele Giannoni
- Department of Chemistry, Biology and Biotechnology, Biochemical and Biotechnological Sciences, University of Perugia, 06122 Perugia, Italy
| | - Francesco Morena
- Department of Chemistry, Biology and Biotechnology, Biochemical and Biotechnological Sciences, University of Perugia, 06122 Perugia, Italy
| | - Sabata Martino
- Department of Chemistry, Biology and Biotechnology, Biochemical and Biotechnological Sciences, University of Perugia, 06122 Perugia, Italy
- Centro di Eccellenza Materiali Innovativi Nanostrutturati per Applicazioni Chimiche Fisiche e Biomediche (CEMIN), University of Perugia, 06123 Perugia, Italy
| |
Collapse
|
26
|
Senobari F, Abolmaali SS, Farahavr G, Tamaddon AM. Targeting inflammation with hyaluronic acid-based micro- and nanotechnology: A disease-oriented review. Int J Biol Macromol 2024; 280:135923. [PMID: 39322155 DOI: 10.1016/j.ijbiomac.2024.135923] [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: 12/03/2023] [Revised: 08/29/2024] [Accepted: 09/20/2024] [Indexed: 09/27/2024]
Abstract
Inflammation is a pivotal immune response in numerous diseases and presents therapeutic challenges. Traditional anti-inflammatory drugs and emerging cytokine inhibitors encounter obstacles such as limited bioavailability, poor tissue distribution, and adverse effects. Hyaluronic acid (HA), a versatile biopolymer, is widely employed to deliver therapeutic agents, including anti-inflammatory drugs, genes, and cell therapies owing to its unique properties, such as hydrophilicity, biodegradability, and safety. HA interacts with cell receptors to initiate processes such as angiogenesis, cell proliferation, and immune regulation. HA-based drug delivery systems offer dual strategies for effective inflammation management, capitalizing on passive and active mechanisms. This synergy permits the mitigation of inflammation by lowering the doses of anti-inflammatory drugs and their off-target adverse effects. A diverse array of micro- and nanotechnology techniques enable the fabrication of tailored HA-engineered systems, including hydrogels, microgels, nanogels, microneedles, nanofibers, and 3D-printed scaffolds, for diverse formulations and administration routes. This review explores recent insights into HA pharmacology in inflammatory conditions, material design, and fabrication methods, as well as its applications across a spectrum of inflammatory diseases, such as atherosclerosis, psoriasis, dermatitis, wound healing, rheumatoid arthritis, osteoarthritis, inflammatory bowel disease, and colitis, highlighting its potential for clinical translation.
Collapse
Affiliation(s)
- Fatemeh Senobari
- Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz 71345, Iran
| | - Samira Sadat Abolmaali
- Associate Professor, Pharmaceutical Nanotechnology Department and Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz 71345, Iran
| | - Ghazal Farahavr
- Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz 71345, Iran
| | - Ali Mohammad Tamaddon
- Professor, Pharmaceutics and Pharmaceutical Nanotechnology Department and Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz 71345, Iran.
| |
Collapse
|
27
|
Ye H, Zhang R, Zhang C, Xia Y, Jin L. Advances in hyaluronic acid: Bioactivity, complexed biomaterials and biological application: A review. Asian J Surg 2024:S1015-9584(24)01841-4. [PMID: 39217010 DOI: 10.1016/j.asjsur.2024.08.100] [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: 04/01/2024] [Revised: 08/02/2024] [Accepted: 08/15/2024] [Indexed: 09/04/2024] Open
Abstract
Hyaluronic acid (HA) is a natural glycosaminoglycan found in the human body, particularly in the extracellular matrix of body fluids and tissues. It plays a critical role in cellular processes of living organisms by maintaining tissue hydration, cell proliferation, differentiation, and inflammatory response. HA exhibits significant biological activity in skin care, aesthetic anti-aging, medical orthopedic repair, gynecological cancer monitoring, and other pathological conditions. Due to its exceptional biocompatibility, biodegradability, lack of toxicity, non-immunogenicity, and its capacity to bond with other substances, various HA-based biomedical products like hydrogels, microneedles, and microspheres have been developed. These innovations have also been applied in various medical and health fields, such as bone and tissue regeneration, gels for medical aesthetic fillers, and gynecology-related cancer treatment, utilizing the HA drug delivery pathway. The interest in HA and its products is increasing due to their biological functions. Therefore, this review aimed to summarize the biological properties of HA and to focus on its applications in the bone tissue engineering and healthcare, for HA has some practical applications of HA-based complexes in biomedical materials, tissue repair, medical aesthetics, and gynecology. Through this review, we seek to offer theoretical research assistance for the development of HA-based bioproducts in the healthcare domain and provide innovative insights for human health.
Collapse
Affiliation(s)
- Huijun Ye
- The Second Affiliated Hospital of Zhejiang Chinese Medical University, No.318 Chaowang Road, Hangzhou, 310005, Zhejiang, China
| | - Ruijuan Zhang
- Center for Peak of Excellence on Biological Science and Food Engineering, National University of Singapore (Suzhou) Research Institute, Suzhou, 215004, Jiangsu, China
| | - Chunye Zhang
- Center for Peak of Excellence on Biological Science and Food Engineering, National University of Singapore (Suzhou) Research Institute, Suzhou, 215004, Jiangsu, China
| | - Yujie Xia
- Center for Peak of Excellence on Biological Science and Food Engineering, National University of Singapore (Suzhou) Research Institute, Suzhou, 215004, Jiangsu, China.
| | - Lihua Jin
- The Second Affiliated Hospital of Zhejiang Chinese Medical University, No.318 Chaowang Road, Hangzhou, 310005, Zhejiang, China.
| |
Collapse
|
28
|
Yang L, Li W, Zhao Y, Wang Y, Shang L. Stem cell recruitment polypeptide hydrogel microcarriers with exosome delivery for osteoarthritis treatment. J Nanobiotechnology 2024; 22:512. [PMID: 39192268 PMCID: PMC11348651 DOI: 10.1186/s12951-024-02765-1] [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: 03/10/2024] [Accepted: 08/13/2024] [Indexed: 08/29/2024] Open
Abstract
With the accelerated aging tendency, osteoarthritis (OA) has become an intractable global public health challenge. Stem cells and their derivative exosome (Exo) have shown great potential in OA treatment. Research in this area tends to develop functional microcarriers for stem cell and Exo delivery to improve the therapeutic effect. Herein, we develop a novel system of Exo-encapsulated stem cell-recruitment hydrogel microcarriers from liquid nitrogen-assisted microfluidic electrospray for OA treatment. Benefited from the advanced droplet generation capability of microfluidics and mild cryogelation procedure, the resultant particles show uniform size dispersion and excellent biocompatibility. Moreover, acryloylated stem cell recruitment peptides SKPPGTSS are directly crosslinked within the particles by ultraviolet irradiation, thus simplifying the peptide coupling process and preventing its premature release. The SKPPGTSS-modified particles can recruit endogenous stem cells to promote cartilage repair and the released Exo from the particles further enhances the cartilage repair performance through synergistic effects. These features suggest that the proposed hydrogel microcarrier delivery system is a promising candidate for OA treatment.
Collapse
Affiliation(s)
- Lei Yang
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325001, China
| | - Wenzhao Li
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325001, China
| | - Yuanjin Zhao
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China.
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325001, China.
| | - Yongxiang Wang
- Department of Orthopedics, Northern Jiangsu People's Hospital, Clinical Teaching Hospital of Medical School, Nanjing University, Yangzhou, 225001, China.
| | - Luoran Shang
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China.
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, and The Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
29
|
Huang K, Liu X, Qin H, Li Y, Zhu J, Yin B, Zheng Q, Zuo C, Cao H, Tong Z, Sun Z. FGF18 encoding circular mRNA-LNP based on glycerolipid engineering of mesenchymal stem cells for efficient amelioration of osteoarthritis. Biomater Sci 2024; 12:4427-4439. [PMID: 39037353 DOI: 10.1039/d4bm00668b] [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: 07/23/2024]
Abstract
Mesenchymal stem cells (MSCs) exhibit substantial potential for osteoarthritis (OA) therapy through cartilage regeneration, yet the realization of optimal therapeutic outcomes is hampered by their limited intrinsic reparative capacities. Herein, MSCs are engineered with circular mRNA (cmRNA) encoding fibroblast growth factor 18 (FGF18) encapsulated within lipid nanoparticles (LNP) derived from a glycerolipid to facilitate OA healing. A proprietary biodegradable and ionizable glycerolipid, TG6A, with branched tails and five ester bonds, forms LNP exhibiting above 9-fold and 41-fold higher EGFP protein expression in MSCs than commercial LNP from DLin-MC3-DMA and ALC-0315, respectively. The introduction of FGF18 not only augmented the proliferative capacity of MSCs but also upregulated the expression of chondrogenic genes and glycosaminoglycan (GAG) content. Additionally, FGF18 enhanced the production of proteoglycans and type II collagen in chondrocyte pellet cultures in a three-dimensional culture. In an OA rat model, transplantation with FGF18-engineered MSCs remarkably preserved cartilage integrity and facilitated functional repair of cartilage lesions, as evidenced by thicker cartilage layers, reduced histopathological scores, maintenance of zone structure, and incremental type II collagen and extracellular matrix (ECM) deposition. Taken together, our findings suggest that TG6A-based LNP loading with cmRNA for engineering MSCs present an innovative strategy to overcome the current limitations in OA treatment.
Collapse
Affiliation(s)
- Ke Huang
- Department of Chemical and Biological Engineering, Monash University, Clayton, VIC 3800, Australia
- Suzhou Industrial Park Monash Research Institute of Science and Technology, Suzhou, 215000, China
| | - Xiaoyun Liu
- Jiangsu Purecell Biopharma Technology Co., Ltd, Suzhou 215125, China.
| | - Haitang Qin
- Jiangsu Purecell Biopharma Technology Co., Ltd, Suzhou 215125, China.
| | - Yingwen Li
- Suzhou CureMed Biopharma Technology Co., Ltd, Suzhou 215125, China.
| | - Jiafeng Zhu
- Suzhou CureMed Biopharma Technology Co., Ltd, Suzhou 215125, China.
| | - Bo Yin
- National University of Singapore (Suzhou) Research Institute, Suzhou, 215123, China.
| | - Qijun Zheng
- Department of Chemical and Biological Engineering, Monash University, Clayton, VIC 3800, Australia
- Suzhou Industrial Park Monash Research Institute of Science and Technology, Suzhou, 215000, China
| | - Chijian Zuo
- Suzhou CureMed Biopharma Technology Co., Ltd, Suzhou 215125, China.
| | - Hui Cao
- Jiangsu Purecell Biopharma Technology Co., Ltd, Suzhou 215125, China.
| | - Zhenbo Tong
- Southeast University-Monash University Joint Research Institute, Suzhou 215125, China
| | - Zhenhua Sun
- Suzhou CureMed Biopharma Technology Co., Ltd, Suzhou 215125, China.
| |
Collapse
|
30
|
Matějková N, Korecká L, Šálek P, Kočková O, Pavlova E, Kašparová J, Obořilová R, Farka Z, Frolich K, Adam M, Carrillo A, Šinkorová Z, Bílková Z. Hyaluronic Acid Nanoparticles with Parameters Required for In Vivo Applications: From Synthesis to Parametrization. Biomacromolecules 2024; 25:4934-4945. [PMID: 38943654 PMCID: PMC11323013 DOI: 10.1021/acs.biomac.4c00370] [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: 03/18/2024] [Revised: 06/14/2024] [Accepted: 06/14/2024] [Indexed: 07/01/2024]
Abstract
Hyaluronic acid is an excellent biocompatible material for in vivo applications. Its ability to bind CD44, a cell receptor involved in numerous biological processes, predetermines HA-based nanomaterials as unique carrier for therapeutic and theranostic applications. Although numerous methods for the synthesis of hyaluronic acid nanoparticles (HANPs) are available today, their low reproducibility and wide size distribution hinder the precise assessment of the effect on the organism. A robust and reproducible approach for producing HANPs that meet strict criteria for in vivo applications (e.g., to lung parenchyma) remains challenging. We designed and evaluated four protocols for the preparation of HANPs with those required parameters. The HA molecule was cross-linked by novel combinations of carbodiimide, and four different amine-containing compounds resulted in monodisperse HANPs with a low polydispersity index. By a complex postsynthetic characterization, we confirmed that the prepared HANPs meet the criteria for inhaled therapeutic delivery and other in vivo applications.
Collapse
Affiliation(s)
- Nikola Matějková
- Department
of Biological and Biochemical Sciences, Faculty of Chemical Technology, University of Pardubice, Studentská 573, Pardubice 532 10, Czech Republic
| | - Lucie Korecká
- Department
of Biological and Biochemical Sciences, Faculty of Chemical Technology, University of Pardubice, Studentská 573, Pardubice 532 10, Czech Republic
| | - Petr Šálek
- Institute
of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovského nám. 2, Praha 6 162 00, Czech Republic
| | - Olga Kočková
- Institute
of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovského nám. 2, Praha 6 162 00, Czech Republic
| | - Ewa Pavlova
- Institute
of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovského nám. 2, Praha 6 162 00, Czech Republic
| | - Jitka Kašparová
- Department
of Biological and Biochemical Sciences, Faculty of Chemical Technology, University of Pardubice, Studentská 573, Pardubice 532 10, Czech Republic
| | - Radka Obořilová
- Central
European Institute of Technology, Masaryk University, Kamenice 5, Brno 625 00, Czech Republic
- Department
of Biochemistry, Faculty of Science, Masaryk
University, Kamenice
5, Brno 625 00, Czech Republic
| | - Zdeněk Farka
- Central
European Institute of Technology, Masaryk University, Kamenice 5, Brno 625 00, Czech Republic
- Department
of Biochemistry, Faculty of Science, Masaryk
University, Kamenice
5, Brno 625 00, Czech Republic
| | - Karel Frolich
- Department
of Physical Chemistry, Faculty of Chemical Technology, University of Pardubice, Studentská 573, Pardubice 532 10, Czech Republic
| | - Martin Adam
- Department
of Analytical Chemistry, Faculty of Chemical
Technology, University of Pardubice, Studentská 573, Pardubice 532 10, Czech Republic
| | - Anna Carrillo
- Department
of Radiobiology, Faculty of Military Health
Sciences, University of Defence, Třebešská 1575, Hradec Králové 500 01, Czech Republic
| | - Zuzana Šinkorová
- Department
of Radiobiology, Faculty of Military Health
Sciences, University of Defence, Třebešská 1575, Hradec Králové 500 01, Czech Republic
| | - Zuzana Bílková
- Department
of Biological and Biochemical Sciences, Faculty of Chemical Technology, University of Pardubice, Studentská 573, Pardubice 532 10, Czech Republic
| |
Collapse
|
31
|
Zhu Y, Chen M, Yang C, Lu G, Huang S, Chen M, Wang Y, Ban J. Revealing Changes in Celecoxib Nanostructured Lipid Carrier's Bioavailability Using Hyaluronic Acid as an Enhancer by HPLC-MS/MS. Drug Des Devel Ther 2024; 18:3315-3327. [PMID: 39100220 PMCID: PMC11296516 DOI: 10.2147/dddt.s461969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 06/18/2024] [Indexed: 08/06/2024] Open
Abstract
Purpose Oral drug administration is the most common and convenient route, offering good patient compliance but drug solubility limits oral applications. Celecoxib, an insoluble drug, requires continuous high-dose oral administration, which may increase cardiovascular risk. The nanostructured lipid carriers prepared from drugs and lipid excipients can effectively improve drug bioavailability, reduce drug dosage, and lower the risk of adverse reactions. Methods In this study, we prepared hyaluronic acid-modified celecoxib nanostructured lipid carriers (HA-NLCs) to improve the bioavailability of celecoxib and reduce or prevent adverse drug reactions. Meanwhile, we successfully constructed a set of FDA-compliant biological sample test methods to investigate the pharmacokinetics of HA-NLCs in rats. Results The pharmacokinetic analysis confirmed that HA-NLCs significantly enhanced drug absorption, resulting in an AUC0-t 1.54 times higher than the reference formulation (Celebrex®). Moreover, compared with unmodified nanostructured lipid carriers (CXB-NLCs), HA-NLCs enhance the retention time and improve the drug's half-life in vivo. Conclusion HA-NLCs significantly increased the bioavailability of celecoxib. The addition of hyaluronic acid prolonged the drug's in vivo duration of action and reduced the risk of cardiovascular adverse effects associated with the frequent administration of oral celecoxib.
Collapse
Affiliation(s)
- Yi Zhu
- Guangdong Pharmaceutical University, Guangzhou, People’s Republic of China
- The Innovation Team for Integrating Pharmacy with Entrepreneurship, Guangdong Pharmaceutical University, Guangzhou, People’s Republic of China
| | - Meiling Chen
- Guangdong Laboratory Animals Monitoring Institute, Guangdong Provincial Key Laboratory of Laboratory Animals, Guangzhou, People’s Republic of China
| | - Chuangzan Yang
- Guangdong Pharmaceutical University, Guangzhou, People’s Republic of China
- The Innovation Team for Integrating Pharmacy with Entrepreneurship, Guangdong Pharmaceutical University, Guangzhou, People’s Republic of China
| | - Geng Lu
- Guangdong Pharmaceutical University, Guangzhou, People’s Republic of China
- The Innovation Team for Integrating Pharmacy with Entrepreneurship, Guangdong Pharmaceutical University, Guangzhou, People’s Republic of China
| | - Sa Huang
- Guangdong Pharmaceutical University, Guangzhou, People’s Republic of China
- The Innovation Team for Integrating Pharmacy with Entrepreneurship, Guangdong Pharmaceutical University, Guangzhou, People’s Republic of China
| | - Meili Chen
- Guangdong Laboratory Animals Monitoring Institute, Guangdong Provincial Key Laboratory of Laboratory Animals, Guangzhou, People’s Republic of China
| | - Yufei Wang
- Analytical and Testing Center of Guangzhou University, Guangzhou, People’s Republic of China
| | - Junfeng Ban
- Guangdong Pharmaceutical University, Guangzhou, People’s Republic of China
- The Innovation Team for Integrating Pharmacy with Entrepreneurship, Guangdong Pharmaceutical University, Guangzhou, People’s Republic of China
- Guangdong Laboratory Animals Monitoring Institute, Guangdong Provincial Key Laboratory of Laboratory Animals, Guangzhou, People’s Republic of China
| |
Collapse
|
32
|
Abbasifard M, Khorramdelazad H. Harmonizing hope: navigating the osteoarthritis melody through the CCL2/CCR2 axis for innovative therapeutic avenues. Front Immunol 2024; 15:1387651. [PMID: 39076996 PMCID: PMC11284107 DOI: 10.3389/fimmu.2024.1387651] [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: 02/18/2024] [Accepted: 07/01/2024] [Indexed: 07/31/2024] Open
Abstract
Osteoarthritis (OA) is characterized by a complex interplay of molecular signals orchestrated by the CCL2/CCR2 axis. The pathogenesis of OA has been revealed to be influenced by a multifaceted effect of CCL2/CCR2 signaling on inflammation, cartilage degradation, and joint homeostasis. The CCL2/CCR2 axis promotes immune cell recruitment and tips the balance toward degeneration by influencing chondrocyte behavior. Insights into these intricate pathways will offer novel therapeutic approaches, paving the way for targeted interventions that may redefine OA management in the future. This review article explores the molecular symphony through the lens of the CCL2/CCR2 axis, providing a harmonious blend of current knowledge and future directions on OA treatment. Furthermore, in this study, through a meticulous review of recent research, the key players and molecular mechanisms that amplify the catabolic cascade within the joint microenvironment are identified, and therapeutic approaches to targeting the CCL2/CCR axis are discussed.
Collapse
Affiliation(s)
- Mitra Abbasifard
- Department of Internal Medicine, School of Medicine, Ali-Ibn Abi-Talib Hospital, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Hossein Khorramdelazad
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| |
Collapse
|
33
|
Liao S, Jia S, Yue Y, Zeng H, Lin J, Liu P. Advancements in pH-Responsive nanoparticles for osteoarthritis treatment: Opportunities and challenges. Front Bioeng Biotechnol 2024; 12:1426794. [PMID: 39036562 PMCID: PMC11260422 DOI: 10.3389/fbioe.2024.1426794] [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: 05/02/2024] [Accepted: 06/06/2024] [Indexed: 07/23/2024] Open
Abstract
Osteoarthritis (OA) is a degenerative disease linked to aging and obesity. The global aging population has led to an increasing number of OA patients, imposing a significant economic burden on society. Traditional drugs treatment methods often fail to achieve satisfactory outcomes. With the rapid advancement of nanomaterial delivery systems, numerous studies have focused on utilizing nanomaterials as carriers to achieve efficient OA treatment by effectively loading and delivering bioactive ingredients (e.g., drugs, nucleic acids) tailored to the unique pathological conditions, such as the weakly acidic microenvironment of synovial fluid in OA patients. This review highlights the latest advancements in the use of pH-responsive nanoparticles for OA treatment, emphasizing the principle of targeted drug delivery leveraging the acidic microenvironment of inflamed joints. It further discusses the composition, synthesis, response mechanism, target selection, application, and recent research findings of nanoparticles, while also addressing the challenges and future directions in this promising field.
Collapse
Affiliation(s)
- Shuai Liao
- Department of Bone and Joint Surgery, Peking
University Shenzhen Hospital, Shenzhen,
China
- National and Local Joint Engineering Research Center of Orthopaedic
Biomaterials, Peking University Shenzhen
Hospital, Shenzhen,
China
- Shenzhen University School of Medicine,
Shenzhen, China
| | - Shicheng Jia
- Department of Sport Medicine, Peking
University Shenzhen Hospital, Shenzhen,
China
| | - Yaohang Yue
- Department of Bone and Joint Surgery, Peking
University Shenzhen Hospital, Shenzhen,
China
- National and Local Joint Engineering Research Center of Orthopaedic
Biomaterials, Peking University Shenzhen
Hospital, Shenzhen,
China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials
Research, Shenzhen,
China
| | - Hui Zeng
- National and Local Joint Engineering Research Center of Orthopaedic
Biomaterials, Peking University Shenzhen
Hospital, Shenzhen,
China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials
Research, Shenzhen,
China
| | - Jianjin Lin
- Department of Sport Medicine, Peking
University Shenzhen Hospital, Shenzhen,
China
| | - Peng Liu
- Department of Bone and Joint Surgery, Peking
University Shenzhen Hospital, Shenzhen,
China
- National and Local Joint Engineering Research Center of Orthopaedic
Biomaterials, Peking University Shenzhen
Hospital, Shenzhen,
China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials
Research, Shenzhen,
China
| |
Collapse
|
34
|
Ma B, Liu D, Wang Z, Zhang D, Jian Y, Zhang K, Zhou T, Gao Y, Fan Y, Ma J, Gao Y, Chen Y, Chen S, Liu J, Li X, Li L. A Top-Down Design Approach for Generating a Peptide PROTAC Drug Targeting Androgen Receptor for Androgenetic Alopecia Therapy. J Med Chem 2024; 67:10336-10349. [PMID: 38836467 DOI: 10.1021/acs.jmedchem.4c00828] [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: 06/06/2024]
Abstract
While large-scale artificial intelligence (AI) models for protein structure prediction and design are advancing rapidly, the translation of deep learning models for practical macromolecular drug development remains limited. This investigation aims to bridge this gap by combining cutting-edge methodologies to create a novel peptide-based PROTAC drug development paradigm. Using ProteinMPNN and RFdiffusion, we identified binding peptides for androgen receptor (AR) and Von Hippel-Lindau (VHL), followed by computational modeling with Alphafold2-multimer and ZDOCK to predict spatial interrelationships. Experimental validation confirmed the designed peptide's binding ability to AR and VHL. Transdermal microneedle patching technology was seamlessly integrated for the peptide PROTAC drug delivery in androgenic alopecia treatment. In summary, our approach provides a generic method for generating peptide PROTACs and offers a practical application for designing potential therapeutic drugs for androgenetic alopecia. This showcases the potential of interdisciplinary approaches in advancing drug development and personalized medicine.
Collapse
Affiliation(s)
- Bohan Ma
- Department of Urology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710049, China
| | - Donghua Liu
- Department of Urology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710049, China
| | - Zhe Wang
- Institute of Bioengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, Zhejiang 310000, China
| | - Dize Zhang
- Department of Urology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710049, China
| | - Yanlin Jian
- Department of Urology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710049, China
| | - Kun Zhang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710049, China
| | - Tianyang Zhou
- Department of Urology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710049, China
| | - Yibo Gao
- Department of Urology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710049, China
| | - Yizeng Fan
- Department of Urology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710049, China
| | - Jian Ma
- Department of Urology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710049, China
| | - Yang Gao
- Department of Urology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710049, China
| | - Yule Chen
- Department of Urology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710049, China
| | - Si Chen
- School of Medicine, Shanghai University, 99 Shangda Road, Shanghai 200444, China
| | - Jing Liu
- Department of Urology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710049, China
| | - Xiang Li
- School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China
| | - Lei Li
- Department of Urology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710049, China
| |
Collapse
|
35
|
Wang Q, Li H, Wu T, Yu B, Cong H, Shen Y. Nanodrugs based on co-delivery strategies to combat cisplatin resistance. J Control Release 2024; 370:14-42. [PMID: 38615892 DOI: 10.1016/j.jconrel.2024.04.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 03/24/2024] [Accepted: 04/09/2024] [Indexed: 04/16/2024]
Abstract
Cisplatin (CDDP), as a broad-spectrum anticancer drug, is able to bind to DNA and inhibit cell division. Despite the widespread use of cisplatin since its discovery, cisplatin resistance developed during prolonged chemotherapy, similar to other small molecule chemotherapeutic agents, severely limits its clinical application. Cisplatin resistance in cancer cells is mainly caused by three reasons: DNA repair, decreased cisplatin uptake/increased efflux, and cisplatin inactivation. In earlier combination therapies, the emergence of multidrug resistance (MDR) in cancer cells prevented the achievement of the desired therapeutic effect even with the accurate combination of two chemotherapeutic drugs. Therefore, combination therapy using nanocarriers for co-delivery of drugs is considered to be ideal for alleviating cisplatin resistance and reducing cisplatin-related toxicity in cancer cells. This article provides an overview of the design of cisplatin nano-drugs used to combat cancer cell resistance, elucidates the mechanisms of action of cisplatin and the pathways through which cancer cells develop resistance, and finally discusses the design of drugs and related carriers that can synergistically reduce cancer resistance when combined with cisplatin.
Collapse
Affiliation(s)
- Qiubo Wang
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao 266071, China
| | - Hui Li
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao 266071, China
| | - Taixia Wu
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao 266071, China
| | - Bing Yu
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao 266071, China; State Key Laboratory of Bio-Fibers and Eco-Textiles, Qingdao University, Qingdao 266071, China.
| | - Hailin Cong
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao 266071, China; State Key Laboratory of Bio-Fibers and Eco-Textiles, Qingdao University, Qingdao 266071, China; School of Materials Science and Engineering, Shandong University of Technology, Zibo 255000, China.
| | - Youqing Shen
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao 266071, China; Key Laboratory of Biomass Chemical Engineering of Ministry of Education, Center for Bio-nanoengineering, and Department of Chemical and Biological Engineering, Zhejiang University, Hangzhou, Zhejiang 310027, China
| |
Collapse
|
36
|
Chang W, Chen L, Chen K. The bioengineering application of hyaluronic acid in tissue regeneration and repair. Int J Biol Macromol 2024; 270:132454. [PMID: 38763255 DOI: 10.1016/j.ijbiomac.2024.132454] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 05/04/2024] [Accepted: 05/15/2024] [Indexed: 05/21/2024]
Abstract
The multifaceted role of hyaluronic acid (HA) across diverse biomedical disciplines underscores its versatility in tissue regeneration and repair. HA hydrogels employ different crosslinking including chemical (chitosan, collagen), photo- initiation (riboflavin, LAP), enzymatic (HRP/H2O2), and physical interactions (hydrogen bonds, metal coordination). In biophysics and biochemistry, HA's signaling pathways, primarily through CD44 and RHAMM receptors, modulate cell behavior (cell migration; internalization of HA), inflammation, and wound healing. Particularly, smaller HA fragments stimulate inflammatory responses through toll-like receptors, impacting macrophages and cytokine expression. HA's implications in oncology highlight its involvement in tumor progression, metastasis, and treatment. Elevated HA in tumor stroma impacts apoptosis resistance and promotes tumor growth, presenting potential therapeutic targets to halt tumor progression. In orthopedics, HA's presence in synovial fluid aids in osteoarthritis management, as its supplementation alleviates pain, enhances synovial fluid's viscoelastic properties, and promotes cartilage integrity. In ophthalmology, HA's application in dry eye syndrome addresses symptoms by moisturizing the eyes, replenishing tear film deficiencies, and facilitating wound healing. Intravitreal injections and hydrogel-based systems offer versatile approaches for drug delivery and vitreous humor replacement. For skin regeneration and wound healing, HA hydrogel dressings exhibit exceptional properties by promoting moist wound healing and facilitating tissue repair. Integration of advanced regenerative tools like stem cells and solubilized amnion membranes into HA-based systems accelerates wound closure and tissue recovery. Overall, HA's unique properties and interactions render it a promising candidate across diverse biomedical domains, showcasing immense potentials in tissue regeneration and therapeutic interventions. Nevertheless, many detailed cellular and molecular mechanisms of HA and its applications remain unexplored and warrant further investigation.
Collapse
Affiliation(s)
- WeiTing Chang
- Department of Obstetrics and Gynecology, Taipei Tzu-Chi Hospital, The Buddhist Tzu-Chi Medical Foundation, Taipei, Taiwan
| | - LiRu Chen
- Department of Physical Medicine and Rehabilitation, Mackay Memorial Hospital, Taipei, Taiwan; Department of Mechanical Engineering, National YangMing ChiaoTung University, Hsinchu, Taiwan
| | - KuoHu Chen
- Department of Obstetrics and Gynecology, Taipei Tzu-Chi Hospital, The Buddhist Tzu-Chi Medical Foundation, Taipei, Taiwan; School of Medicine, Tzu-Chi University, Hualien, Taiwan.
| |
Collapse
|
37
|
Lee CY, Chang YC, Yang KC, Lin YF, Wu ATH, Tseng CL. Development and functional evaluation of a hyaluronic acid coated nano-formulation with kaempferol as a novel intra-articular agent for Knee Osteoarthritis treatment. Biomed Pharmacother 2024; 175:116717. [PMID: 38749179 DOI: 10.1016/j.biopha.2024.116717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 04/29/2024] [Accepted: 05/06/2024] [Indexed: 06/03/2024] Open
Abstract
Knee osteoarthritis (OA) involves articular cartilage degradation driven mainly by inflammation. Kaempferol (KM), known for its anti-inflammatory property, holds potential for OA treatment. This study investigated the potential of hyaluronic acid (HA)-coated gelatin nanoparticles loaded with KM (HA-KM GNP) for treating knee OA. KM was encapsulated into gelatin nanoparticles (KM GNP) and then coated with HA to form HA-KM GNPs. Physical properties were characterized, and biocompatibility and cellular uptake were assessed in rat chondrocytes. Anti-inflammatory and chondrogenic properties were evaluated using IL-1β-stimulated rat chondrocytes, compared with HA-coated nanoparticles without KM (HA GNP) and KM alone. Preclinical efficacy was tested in an anterior cruciate ligament transection (ACLT)-induced knee OA rat model treated with intra-articular injection of HA-KM GNP. Results show spherical HA-KM GNPs (88.62 ± 3.90 nm) with positive surface charge. Encapsulation efficiency was 98.34 % with a sustained release rate of 18 % over 48 h. Non-toxic KM concentration was 2.5 μg/mL. In IL-1β-stimulated OA rat chondrocytes, HA-KM GNP significantly down-regulated RNA expression of IL-1β, TNF-α, COX-2, MMP-9, and MMP-13, while up-regulating SOX9 compared to HA GNP, and KM. In vivo imaging demonstrated significantly higher fluorescence intensity within rat knee joints for 3 hours post HA-KM GNP injection compared with KM GNP (185.2% ± 34.1% vs. 45.0% ± 16.7%). HA-KM GNP demonstrated significant effectiveness in reducing subchondral sclerosis, attenuating inflammation, inhibiting matrix degradation, restoring cartilage thickness, and reducing the severity of OA in the ACLT rat model. In conclusion, HA-KM GNP holds promise for knee OA therapy.
Collapse
Affiliation(s)
- Ching-Yu Lee
- International Ph.D. Program in Cell Therapy and Regenerative Medicine, College of Medicine, Taipei Medical University, Taipei 110301, Taiwan; Department of Orthopedics, Taipei Medical University Hospital, Taipei 110301, Taiwan; Department of Orthopaedics, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110301, Taiwan; Orthopedic Research Center, Taipei Medical University Hospital, Taipei 110301, Taiwan
| | - Yu-Chu Chang
- International Ph.D. Program in Cell Therapy and Regenerative Medicine, College of Medicine, Taipei Medical University, Taipei 110301, Taiwan; Department of Biochemistry and Molecular Cell Biology, School of Medicine, Taipei Medical University, Taipei 110301, Taiwan
| | - Kai-Chiang Yang
- School of Dental Technology, College of Oral Medicine, Taipei Medical University, Taipei 110301, Taiwan
| | - Yung-Fang Lin
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 110301, Taiwan; International Ph.D. Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 110301, Taiwan
| | - Alexander T H Wu
- The Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan; Clinical Research Center, Taipei Medical University Hospital, Taipei Medical University, Taipei 110301, Taiwan.
| | - Ching-Li Tseng
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 110301, Taiwan; International Ph.D. Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 110301, Taiwan.
| |
Collapse
|
38
|
Chapman JH, Ghosh D, Attari S, Ude CC, Laurencin CT. Animal Models of Osteoarthritis: Updated Models and Outcome Measures 2016-2023. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2024; 10:127-146. [PMID: 38983776 PMCID: PMC11233113 DOI: 10.1007/s40883-023-00309-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 05/19/2023] [Accepted: 06/06/2023] [Indexed: 07/11/2024]
Abstract
Purpose Osteoarthritis (OA) is a global musculoskeletal disorder that affects primarily the knee and hip joints without any FDA-approved disease-modifying therapies. Animal models are essential research tools in developing therapies for OA; many animal studies have provided data for the initiation of human clinical trials. Despite this, there is still a need for strategies to recapitulate the human experience using animal models to better develop treatments and understand pathogenesis. Since our last review on animal models of osteoarthritis in 2016, there have been exciting updates in OA research and models. The main purpose of this review is to update the latest animal models and key features of studies in OA research. Method We used our existing classification method and screened articles in PubMed and bibliographic search for animal OA models between 2016 and 2023. Relevant and high-cited articles were chosen for inclusion in this narrative review. Results Recent studies were analyzed and classified. We also identified ex vivo models as an area of ongoing research. Each animal model offers its own benefit in the study of OA and there are a full range of outcome measures that can be assessed. Despite the vast number of models, each has its drawbacks that have limited translating approved therapies for human use. Conclusion Depending on the outcome measures and objective of the study, researchers should pick the best model for their work. There have been several exciting studies since 2016 that have taken advantage of regenerative engineering techniques to develop therapies and better understand OA. Lay Summary Osteoarthritis (OA) is a chronic debilitating disease without any cure that affects mostly the knee and hip joints and often results in surgical joint replacement. Cartilage protects the joint from mechanical forces and degrades with age or in response to injury. The many contributing causes of OA are still being investigated, and animals are used for preclinical research and to test potential new treatments. A single consensus OA animal model for preclinical studies is non-existent. In this article, we review the many animal models for OA and provide a much-needed update on studies and model development since 2016.
Collapse
Affiliation(s)
- James H. Chapman
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut, 263 Farmington Avenue, Farmington, CT 06030-3711, USA
- Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, UConn Health, Farmington, CT 06030, USA
- Department of Orthopedic Surgery, UConn Health, Farmington, CT 06030, USA
| | - Debolina Ghosh
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut, 263 Farmington Avenue, Farmington, CT 06030-3711, USA
- Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, UConn Health, Farmington, CT 06030, USA
- Department of Orthopedic Surgery, UConn Health, Farmington, CT 06030, USA
| | - Seyyedmorteza Attari
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut, 263 Farmington Avenue, Farmington, CT 06030-3711, USA
- Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, UConn Health, Farmington, CT 06030, USA
- Department of Orthopedic Surgery, UConn Health, Farmington, CT 06030, USA
- Department of Materials Science and Engineering, University of Connecticut, Storrs, CT 06269, USA
| | - Chinedu C. Ude
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut, 263 Farmington Avenue, Farmington, CT 06030-3711, USA
- Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, UConn Health, Farmington, CT 06030, USA
- Department of Orthopedic Surgery, UConn Health, Farmington, CT 06030, USA
| | - Cato T. Laurencin
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut, 263 Farmington Avenue, Farmington, CT 06030-3711, USA
- Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, UConn Health, Farmington, CT 06030, USA
- Department of Orthopedic Surgery, UConn Health, Farmington, CT 06030, USA
- Department of Materials Science and Engineering, University of Connecticut, Storrs, CT 06269, USA
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269, USA
- Department of Chemical and Bimolecular Engineering, University of Connecticut, Storrs, CT 06269, USA
| |
Collapse
|
39
|
Zhang Y, Zhang D, Jiao X, Yue X, Cai B, Lu S, Xu R. Uncovering the shared neuro-immune-related regulatory mechanisms between spinal cord injury and osteoarthritis. Heliyon 2024; 10:e30336. [PMID: 38707272 PMCID: PMC11068815 DOI: 10.1016/j.heliyon.2024.e30336] [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: 05/10/2023] [Revised: 04/21/2024] [Accepted: 04/24/2024] [Indexed: 05/07/2024] Open
Abstract
Adults with spinal cord injury (SCI), a destructive neurological injury, have a significantly higher incidence of osteoarthritis (OA), a highly prevalent chronic joint disorder. This study aimed to dissect the neuroimmune-related regulatory mechanisms of SCI and OA using bioinformatics analysis. Using microarray data from the Gene Expression Omnibus database, differentially expressed genes (DEGs) were screened between SCI and sham samples and between OA and control samples. Common DEGs were used to construct a protein-protein interaction (PPI) network. Weighted gene co-expression network analysis (WGCNA) was used to mine SCI- and OA-related modules. Shared miRNAs were identified, and target genes were predicted using the Human MicroRNA Disease Database (HMDD) database. A miRNA-gene-pathway regulatory network was constructed with overlapping genes, miRNAs, and significantly enriched pathways. Finally, the expression of the identified genes and miRNAs was verified using RT-qPCR. In both the SCI and OA groups, 185 common DEGs were identified, and three hub clusters were obtained from the PPI network. WGCNA revealed three SCI-related modules and two OA-related modules. There were 43 overlapping genes between the PPI network clusters and the WGCNA network modules. Seventeen miRNAs shared between patients with SCI and OA were identified. A regulatory network consisting of five genes, six miRNAs, and six signaling pathways was constructed. Upregulation of CD44, TGFBR1, CCR5, and IGF1, while lower levels of miR-125b-5p, miR-130a-3p, miR-16-5p, miR-204-5p, and miR-204-3p in both SCI and OA were successfully verified using RT-qPCR. Our study suggests that a miRNA-gene-pathway network is implicated in the neuroimmune-related regulatory mechanisms of SCI and OA. CD44, TGFBR1, CCR5, and IGF1, and their related miRNAs (miR-125b-5p, miR-130a-3p, miR-16-5p, miR-204-5p, and miR-204-3p) may serve as promising biomarkers and candidate therapeutic targets for SCI and OA.
Collapse
Affiliation(s)
- Yuxin Zhang
- Department of Rehabilitation Medicine, Fengcheng branch, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, 200011, China
- Shanghai Key Laboratory of Orthopedic Implants, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Dahe Zhang
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, 200011, China
| | - Xin Jiao
- Shanghai Key Laboratory of Orthopedic Implants, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Xiaokun Yue
- Shanghai Key Laboratory of Orthopedic Implants, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Bin Cai
- Department of Rehabilitation Medicine, Fengcheng branch, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Shenji Lu
- Department of Rehabilitation Medicine, Fengcheng branch, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Renjie Xu
- Department of Rehabilitation Medicine, Kunshan Rehabilitation Hospital, Suzhou 210000, Jiangsu, China
| |
Collapse
|
40
|
Zhou H, Zhang Z, Mu Y, Yao H, Zhang Y, Wang DA. Harnessing Nanomedicine for Cartilage Repair: Design Considerations and Recent Advances in Biomaterials. ACS NANO 2024; 18:10667-10687. [PMID: 38592060 DOI: 10.1021/acsnano.4c00780] [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: 04/10/2024]
Abstract
Cartilage injuries are escalating worldwide, particularly in aging society. Given its limited self-healing ability, the repair and regeneration of damaged articular cartilage remain formidable challenges. To address this issue, nanomaterials are leveraged to achieve desirable repair outcomes by enhancing mechanical properties, optimizing drug loading and bioavailability, enabling site-specific and targeted delivery, and orchestrating cell activities at the nanoscale. This review presents a comprehensive survey of recent research in nanomedicine for cartilage repair, with a primary focus on biomaterial design considerations and recent advances. The review commences with an introductory overview of the intricate cartilage microenvironment and further delves into key biomaterial design parameters crucial for treating cartilage damage, including microstructure, surface charge, and active targeting. The focal point of this review lies in recent advances in nano drug delivery systems and nanotechnology-enabled 3D matrices for cartilage repair. We discuss the compositions and properties of these nanomaterials and elucidate how these materials impact the regeneration of damaged cartilage. This review underscores the pivotal role of nanotechnology in improving the efficacy of biomaterials utilized for the treatment of cartilage damage.
Collapse
Affiliation(s)
- Huiqun Zhou
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR 999077, China
| | - Zhen Zhang
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR 999077, China
| | - Yulei Mu
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR 999077, China
| | - Hang Yao
- School of Chemistry and Chemical Engineering, Yangzhou University, Yangzhou 225009, China
| | - Yi Zhang
- School of Integrated Circuit Science and Engineering, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Dong-An Wang
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR 999077, China
- Center for Neuromusculoskeletal Restorative Medicine, InnoHK, HKSTP, Sha Tin, Hong Kong SAR 999077, China
| |
Collapse
|
41
|
Cao H, Deng S, Chen X, Cui X, Yuan T, Liang J, Zhang X, Fan Y, Wang Q. An injectable cartilage-coating composite with long-term protection, effective lubrication and chondrocyte nourishment for osteoarthritis treatment. Acta Biomater 2024; 179:95-105. [PMID: 38513723 DOI: 10.1016/j.actbio.2024.03.015] [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: 11/28/2023] [Revised: 02/27/2024] [Accepted: 03/14/2024] [Indexed: 03/23/2024]
Abstract
The osteoarthritic (OA) environment within articular cartilage poses significant challenges, resulting in chondrocyte dysfunction and cartilage matrix degradation. While intra-articular injections of anti-inflammatory drugs, biomaterials, or bioactive agents have demonstrated some effectiveness, they primarily provide temporary relief from OA pain without arresting OA progression. This study presents an injectable cartilage-coating composite, comprising hyaluronic acid and decellularized cartilage matrix integrated with specific linker polymers. It enhances the material retention, protection, and lubrication on the cartilage surface, thereby providing an effective physical barrier against inflammatory factors and reducing the friction and shear force associated with OA joint movement. Moreover, the composite gradually releases nutrients, nourishing OA chondrocytes, aiding in the recovery of cellular function, promoting cartilage-specific matrix production, and mitigating OA progression in a rat model. Overall, this injectable cartilage-coating composite offers promising potential as an effective cell-free treatment for OA. STATEMENT OF SIGNIFICANCE: Osteoarthritis (OA) in the articular cartilage leads to chondrocyte dysfunction and cartilage matrix degradation. This study introduces an intra-articular injectable composite material (HDC), composed of decellularized cartilage matrix (dECMs), hyaluronan (HA), and specially designed linker polymers to provide an effective cell-free OA treatment. The linker polymers bind HA and dECMs to form an integrated HDC structure with an enhanced degradation rate, potentially reducing the need for frequent injections and associated trauma. They also enable HDC to specifically coat the cartilage surface, forming a protective and lubricating layer that enhances long-term retention, acts as a barrier against inflammatory factors, and reduces joint movement friction. Furthermore, HDC nourishes OA chondrocytes through gradual nutrient release, aiding cellular function recovery, promoting cartilage-specific matrix production, and mitigating OA progression.
Collapse
Affiliation(s)
- Hongfu Cao
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610065, China; College of Biomedical Engineering, Sichuan University, Chengdu, Sichuan 610065, China
| | - Siyan Deng
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610065, China; College of Biomedical Engineering, Sichuan University, Chengdu, Sichuan 610065, China
| | - Xi Chen
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610065, China; College of Biomedical Engineering, Sichuan University, Chengdu, Sichuan 610065, China
| | - Xiaolin Cui
- School of Medicine, the Chinese University of Hong Kong, Shenzhen 518172, China; Department of Orthopedic Surgery & Musculoskeletal Medicine, Centre for Bioengineering & Nanomedicine, University of Otago, Christchurch 8011, New Zealand
| | - Tun Yuan
- Sichuan Testing Center for Biomaterials and Medical Devices Co. Ltd, 29 Wangjiang Road, Chengdu, Sichuan, China
| | - Jie Liang
- Sichuan Testing Center for Biomaterials and Medical Devices Co. Ltd, 29 Wangjiang Road, Chengdu, Sichuan, China
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610065, China; College of Biomedical Engineering, Sichuan University, Chengdu, Sichuan 610065, China
| | - Yujiang Fan
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610065, China; College of Biomedical Engineering, Sichuan University, Chengdu, Sichuan 610065, China.
| | - Qiguang Wang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610065, China; College of Biomedical Engineering, Sichuan University, Chengdu, Sichuan 610065, China.
| |
Collapse
|
42
|
Chen Q, Jin Y, Chen T, Zhou H, Wang X, Wu O, Chen L, Zhang Z, Guo Z, Sun J, Wu A, Qian Q. Injectable nanocomposite hydrogels with enhanced lubrication and antioxidant properties for the treatment of osteoarthritis. Mater Today Bio 2024; 25:100993. [PMID: 38440110 PMCID: PMC10909650 DOI: 10.1016/j.mtbio.2024.100993] [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: 11/19/2023] [Revised: 01/21/2024] [Accepted: 02/03/2024] [Indexed: 03/06/2024] Open
Abstract
Osteoarthritis (OA) is a chronic inflammatory joint disease characterized by progressive cartilage degeneration, synovitis, and osteoid formation. In order to effectively treat OA, it is important to block the harmful feedback caused by reactive oxygen species (ROS) produced during joint wear. To address this challenge, we have developed injectable nanocomposite hydrogels composed of polygallate-Mn (PGA-Mn) nanoparticles, oxidized sodium alginate, and gelatin. The inclusion of PGA-Mn not only enhances the mechanical strength of the biohydrogel through a Schiff base reaction with gelatin but also ensures efficient ROS scavenging ability. Importantly, the nanocomposite hydrogel exhibits excellent biocompatibility, allowing it to effectively remove ROS from chondrocytes and reduce the expression of inflammatory factors within the joint. Additionally, the hygroscopic properties of the hydrogel contribute to reduced intra-articular friction and promote the production of cartilage-related proteins, supporting cartilage synthesis. In vivo experiments involving the injection of nanocomposite hydrogels into rat knee joints with an OA model have demonstrated successful reduction of osteophyte formation and protection of cartilage from wear, highlighting the therapeutic potential of this approach for treating OA.
Collapse
Affiliation(s)
- Qizhu Chen
- Department of Orthopaedics, Key Laboratory of Structural Malformations in Children of Zhejiang Province, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Yuxin Jin
- Department of Orthopaedics, Key Laboratory of Structural Malformations in Children of Zhejiang Province, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Tao Chen
- Department of Orthopaedics, Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Tongji Hospital of Tongji University, Shanghai, 200065, China
| | - Hao Zhou
- Department of Orthopaedics, Key Laboratory of Structural Malformations in Children of Zhejiang Province, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Xinzhou Wang
- Department of Orthopaedics, Key Laboratory of Structural Malformations in Children of Zhejiang Province, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Ouqiang Wu
- Department of Orthopaedics, Key Laboratory of Structural Malformations in Children of Zhejiang Province, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Linjie Chen
- Department of Orthopaedics, Key Laboratory of Structural Malformations in Children of Zhejiang Province, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Zhiguang Zhang
- Department of Orthopaedics, Key Laboratory of Structural Malformations in Children of Zhejiang Province, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Zhengyu Guo
- Department of Orthopaedics, Key Laboratory of Structural Malformations in Children of Zhejiang Province, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Jin Sun
- Department of Orthopaedics, Key Laboratory of Structural Malformations in Children of Zhejiang Province, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Aimin Wu
- Department of Orthopaedics, Key Laboratory of Structural Malformations in Children of Zhejiang Province, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Qiuping Qian
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325000, China
| |
Collapse
|
43
|
Ma Y, Morozova SM, Kumacheva E. From Nature-Sourced Polysaccharide Particles to Advanced Functional Materials. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024:e2312707. [PMID: 38391153 DOI: 10.1002/adma.202312707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 02/15/2024] [Indexed: 02/24/2024]
Abstract
Polysaccharides constitute over 90% of the carbohydrate mass in nature, which makes them a promising feedstock for manufacturing sustainable materials. Polysaccharide particles (PSPs) are used as effective scavengers, carriers of chemical and biological cargos, and building blocks for the fabrication of macroscopic materials. The biocompatibility and degradability of PSPs are advantageous for their uses as biomaterials with more environmental friendliness. This review highlights the progresses in PSP applications as advanced functional materials, by describing PSP extraction, preparation, and surface functionalization with a variety of functional groups, polymers, nanoparticles, and biologically active species. This review also outlines the fabrication of PSP-derived macroscopic materials, as well as their applications in soft robotics, sensing, scavenging, water harvesting, drug delivery, and bioengineering. The paper is concluded with an outlook providing perspectives in the development and applications of PSP-derived materials.
Collapse
Affiliation(s)
- Yingshan Ma
- Department of Chemistry, University of Toronto, 80 St. George Street, Toronto, Ontario, M5S 3H6, Canada
| | - Sofia M Morozova
- Department of Chemistry, University of Toronto, 80 St. George Street, Toronto, Ontario, M5S 3H6, Canada
- Center of Fluid Physics and Soft Matter, N.E. Bauman Moscow State Technical University, 5/1 2-nd Baumanskaya street, Moscow, 105005, Russia
| | - Eugenia Kumacheva
- Department of Chemistry, University of Toronto, 80 St. George Street, Toronto, Ontario, M5S 3H6, Canada
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College Street, Toronto, Ontario, M5S 3E5, Canada
- The Institute of Biomaterials and Biomedical Engineering, University of Toronto, 4 Taddle Creek Road, Toronto, Ontario, M5S 3G9, Canada
| |
Collapse
|
44
|
Lee WH, Kim W. Self-assembled hyaluronic acid nanoparticles for the topical treatment of inflammatory skin diseases: Beyond drug carriers. J Control Release 2024; 366:114-127. [PMID: 38145664 DOI: 10.1016/j.jconrel.2023.12.026] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 12/10/2023] [Accepted: 12/17/2023] [Indexed: 12/27/2023]
Abstract
Inflammatory skin diseases represent a significant health concern, affecting approximately 20-25% of the global population. These conditions not only reduce an individual's quality of life but also impose a huge burden on both humanity and society. However, addressing these challenges is hindered by their chronic nature, insufficient therapeutic effectiveness, and the propensity for recurrence and adverse side effects. Hyaluronic acid (HA) has emerged as a potential solution to these barriers, owing to its excellent attributes such as biocompatibility, non-toxicity, and targeted drug delivery. However, its practical application has been limited because endogenous hyaluronidase (HYAL) rapidly degrades HA in inflamed skin thus reducing its ability to penetrate deep into the skin. Interestingly, recent research has expanded the role of self-assembled HA-nanoparticles (HA-NPs) beyond drug carriers; they are resistant to HYAL, thereby enabling deep skin penetration, and possess inherent anti-inflammatory properties. Moreover, these abilities can be fine-tuned depending on the conditions during particle synthesis. Additionally, their role as a drug delivery system holds potential for use as a multi-target drug or hybrid drug. In conclusion, this review aims to specifically introduce and highlight the emerging potential of HA-NPs as a topical treatment for inflammatory skin conditions.
Collapse
Affiliation(s)
- Wang Hee Lee
- Department of Molecular Science & Technology, Ajou University, Suwon 16499, Republic of Korea
| | - Wook Kim
- Department of Molecular Science & Technology, Ajou University, Suwon 16499, Republic of Korea.
| |
Collapse
|
45
|
Zhou D, Liu H, Zheng Z, Wu D. Design principles in mechanically adaptable biomaterials for repairing annulus fibrosus rupture: A review. Bioact Mater 2024; 31:422-439. [PMID: 37692911 PMCID: PMC10485601 DOI: 10.1016/j.bioactmat.2023.08.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 07/25/2023] [Accepted: 08/12/2023] [Indexed: 09/12/2023] Open
Abstract
Annulus fibrosus (AF) plays a crucial role in the biomechanical loading of intervertebral disc (IVD). AF is difficult to self-heal when the annulus tears develop, because AF has a unique intricate structure and biologic milieu in vivo. Tissue engineering is promising for repairing AF rupture, but construction of suitable mechanical matching devices or scaffolds is still a grand challenge. To deeply know the varied forces involved in the movement of the native annulus is highly beneficial for designing biomimetic scaffolds to recreate the AF function. In this review, we overview six freedom degrees of forces and adhesion strength on AF tissue. Then, we summarize the mechanical modalities to simulate related forces on AF and to assess the characteristics of biomaterials. We finally outline some current advanced techniques to develop mechanically adaptable biomaterials for AF rupture repair.
Collapse
Affiliation(s)
- Dan Zhou
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Hongmei Liu
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Zhaomin Zheng
- Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, China
- Pain Research Center, Sun Yat-Sen University, Guangzhou 510080, China
| | - Decheng Wu
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, 518055, China
| |
Collapse
|
46
|
Guo C, Liu Y, Zhao Z, Wu Y, Kong Q, Wang Y. Regulating inflammation and apoptosis: A smart microgel gene delivery system for repairing degenerative nucleus pulposus. J Control Release 2024; 365:1004-1018. [PMID: 38128882 DOI: 10.1016/j.jconrel.2023.12.029] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 12/15/2023] [Accepted: 12/18/2023] [Indexed: 12/23/2023]
Abstract
The progression of intervertebral disc degeneration (IDD) is attributed to the gradual exacerbation of cellular apoptosis and impaired extracellular matrix (ECM) synthesis, both of which are induced by progressive inflammation. Therefore, it is crucial to address the inflammatory microenvironment and rectify the excessive apoptosis of nucleus pulposus cells (NPCs) to achieve intervertebral disc (IVD) regeneration. In this study, we devised a smart microgel gene delivery system that incorporates functionalized gene nanoparticles (NPs) for the purpose of IVD regeneration. siGrem1 was loaded into the NPs to enhance their antiapoptotic ability and protective effects. Furthermore, the encapsulation of HADA further endows the NPs (referred to as HSGN) with targeted delivery and anti-inflammatory effects, as well as reactive oxygen species (ROS) scavenging capacities. To create an microenvironment-responsive microgel system, phenylboronic acid-functionalized microspheres (referred to as M.S.) were fabricated and dynamically loaded with the HSGN. This microgel system (MHSGN), which is highly biocompatible, enables the sustained release of siGrem1, effectively modulating inflammation, scavenging ROS, and alleviating apoptosis in NPCs. These multifunctional capabilities promote the restoration of metabolic homeostasis within the nucleus pulposus ECM, ultimately leading to delayed IDD.
Collapse
Affiliation(s)
- Chuan Guo
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, China, 610041
| | - Yuheng Liu
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, China, 610041
| | - Zhen Zhao
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, China, 610041
| | - Ye Wu
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, China, 610041
| | - Qingquan Kong
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, China, 610041.
| | - Yu Wang
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, China, 610041.
| |
Collapse
|
47
|
Li YX, Bao YT, Hu JB. Engineering of targeting antioxidant polypeptide nanopolyplexes for the treatment of acute lung injury. Int J Biol Macromol 2024; 254:127872. [PMID: 37939759 DOI: 10.1016/j.ijbiomac.2023.127872] [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: 08/08/2023] [Revised: 10/20/2023] [Accepted: 11/01/2023] [Indexed: 11/10/2023]
Abstract
The pathogenesis of acute lung injury (ALI) involves various mechanisms, such as oxidative stress, inflammation, and epithelial cell apoptosis. However, current drug therapies face limitations due to issues like systemic distribution, drug degradation in vivo, and hydrophobicity. To address these challenges, we developed a pH-responsive nano-drug delivery system for delivering antioxidant peptides to treat ALI. In this study, we utilized low molecular weight chitosan (LMWC) and hyaluronic acid (HA) as carrier materials. LMWC carries a positive charge, while HA carries a negative charge. By stirring the two together, the electrostatic adsorption between LMWC and HA yielded aggregated drug carriers. To specifically target the antioxidant drug WNWAD to lung lesions and enhance therapeutic outcomes for ALI, we created a targeted drug delivery system known as HA/LMWC@WNWAD (NPs) through a 12-h stirring process. In our research, we characterized the particle size and drug release of NPs. Additionally, we assessed the targeting ability of NPs. Lastly, we evaluated the improvement of lung injury at the cellular and animal levels to investigate the therapeutic mechanism of this drug targeting delivery system.
Collapse
Affiliation(s)
- Yi-Xuan Li
- Faculty of Materials Science and Chemical Engineering, Ningbo University, Ningbo 315211, China
| | - Ya-Ting Bao
- College of Medical, Ningbo University, Ningbo 315211, China
| | - Jing-Bo Hu
- Faculty of Materials Science and Chemical Engineering, Ningbo University, Ningbo 315211, China.
| |
Collapse
|
48
|
Ma C, Wang T, Jin X, Zhang W, Lv Q. Lineage-specific multifunctional double-layer scaffold accelerates the integrated regeneration of cartilage and subchondral bone. Mater Today Bio 2023; 23:100800. [PMID: 37766897 PMCID: PMC10520449 DOI: 10.1016/j.mtbio.2023.100800] [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: 05/08/2023] [Revised: 09/04/2023] [Accepted: 09/14/2023] [Indexed: 09/29/2023] Open
Abstract
Repairing cartilage/subchondral bone defects that involve subchondral bone is a major challenge in clinical practice. Overall, the integrated repair of the structure and function of the osteochondral (OC) unit is very important. Some studies have demonstrated that the differentiation of cartilage is significantly enhanced by reducing the intake of nutrients such as lipids. This study demonstrates that using starvation can effectively optimize the therapeutic effect of bone marrow mesenchymal stem cells (BMSCs)-derived extracellular vesicles (EVs). A hyaluronic acid (HA)-based hydrogel containing starved BMSCs-EVs displayed continuous release for more than 3 weeks and significantly promoted the proliferation and biosynthesis of chondrocytes around the defect regulated by the forkhead-box class O (FOXO) pathway. When combined with vascular inhibitors, the hydrogel inhibited cartilage hypertrophy and facilitated the regeneration of hyaline cartilage. A porous methacrylate gelatine (GelMA)-based hydrogel containing calcium salt loaded with thrombin rapidly promoted haematoma formation upon contact with the bone marrow cavity to quickly block the pores and prevent the blood vessels in the bone marrow cavity from invading the cartilage layer. Furthermore, the haematoma could be used as nutrients to accelerate bone survival. The in vivo experiments demonstrated that the multifunctional lineage-specific hydrogel promoted the integrated regeneration of cartilage/subchondral bone. Thus, this hydrogel may represent a new strategy for osteochondral regeneration and repair.
Collapse
Affiliation(s)
- Chunhui Ma
- Department of Orthopedic Surgery, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, 200080, China
| | - Tao Wang
- Department of Orthopedic Surgery, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, 200080, China
| | - Xinmeng Jin
- Department of Orthopedic Surgery, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, 200080, China
| | - Wanglin Zhang
- Department of Orthopaedics, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Qi Lv
- Department of Medical Imaging, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
| |
Collapse
|
49
|
Alsaikhan F. Hyaluronic acid-empowered nanotheranostics in breast and lung cancers therapy. ENVIRONMENTAL RESEARCH 2023; 237:116951. [PMID: 37633628 DOI: 10.1016/j.envres.2023.116951] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 08/15/2023] [Accepted: 08/21/2023] [Indexed: 08/28/2023]
Abstract
Nanomedicine application in cancer therapy is an urgency because of inability of current biological therapies for complete removal of tumor cells. The development of smart and novel nanoplatforms for treatment of cancer can provide new insight in tumor suppression. Hyaluronic acid is a biopolymer that can be employed for synthesis of smart nanostructures capable of selective targeting CD44-overexpressing tumor cells. The breast and lung cancers are among the most malignant and common tumors in both females and males that environmental factors, lifestyle and genomic alterations are among the risk factors for their pathogenesis and development. Since etiology of breast and lung tumors is not certain and multiple factors participate in their development, preventative measures have not been completely successful and studies have focused on developing new treatment strategies for them. The aim of current review is to provide a comprehensive discussion about application of hyaluronic acid-based nanostructures for treatment of breast and lung cancers. The main reason of using hyaluronic acid-based nanoparticles is their ability in targeting breast and lung cancers in a selective way due to upregulation of CD44 receptor on their surface. Moreover, nanocarriers developed from hyaluronic acid or functionalized with hyaluronic acid have high biocompatibility and their safety is appreciated. The drugs and genes used for treatment of breast and lung cancers lack specific accumulation at cancer site and their cytotoxicity is low, but hyaluronic acid-based nanostructures provide their targeted delivery to tumor site and by increasing internalization of drugs and genes in breast and lung tumor cells, they improve their therapeutic index. Furthermore, hyaluronic acid-based nanostructures can be used for phototherapy-mediated breast and lung cancers ablation. The stimuli-responsive and smart kinds of hyaluronic acid-based nanostructures such as pH- and light-responsive can increase selective targeting of breast and lung cancers.
Collapse
Affiliation(s)
- Fahad Alsaikhan
- College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj, Saudi Arabia.
| |
Collapse
|
50
|
Liang J, Liu L, Feng H, Yue Y, Zhang Y, Wang Q, Zhao H. Therapeutics of osteoarthritis and pharmacological mechanisms: A focus on RANK/RANKL signaling. Biomed Pharmacother 2023; 167:115646. [PMID: 37804812 DOI: 10.1016/j.biopha.2023.115646] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 09/15/2023] [Accepted: 10/03/2023] [Indexed: 10/09/2023] Open
Abstract
Osteoarthritis (OA) is a chronic degenerative disease afflicting millions globally. Despite the development of numerous pharmacological treatments for OA, a substantial unmet need for effective therapies persists. The RANK/RANKL signaling pathway has emerged as a promising therapeutic target for OA, owing to its pivotal role in regulating osteoclast differentiation and activity. In this comprehensive review, we aim to elucidate the relevant mechanisms of OA mediated by RANK/RANKL signaling, including bone remodeling, inflammation, cartilage degradation, osteophyte formation, and pain sensitization. Furthermore, we discuss and summarize the cutting-edge strategies targeting RANK/RANKL signaling for OA therapy, encompassing approaches such as gene-based interventions and biomaterials-aided pharmacotherapy. In addition, we highlight the prevailing challenges associated with pharmacological OA treatments and explore potential future directions, approached through a clinical-translational lens.
Collapse
Affiliation(s)
- Jingqi Liang
- Department of Foot and Ankle Surgery, Honghui Hospital of Xi'an Jiaotong University, China
| | - Liang Liu
- Department of Foot and Ankle Surgery, Honghui Hospital of Xi'an Jiaotong University, China
| | - Hui Feng
- Department of Foot and Ankle Surgery, Honghui Hospital of Xi'an Jiaotong University, China
| | - Yang Yue
- Department of Foot and Ankle Surgery, Honghui Hospital of Xi'an Jiaotong University, China
| | - Yan Zhang
- Department of Foot and Ankle Surgery, Honghui Hospital of Xi'an Jiaotong University, China
| | - Qiong Wang
- Department of Foot and Ankle Surgery, Honghui Hospital of Xi'an Jiaotong University, China
| | - Hongmou Zhao
- Department of Foot and Ankle Surgery, Honghui Hospital of Xi'an Jiaotong University, China.
| |
Collapse
|