1
|
Sun H, Zhan M, Zou Y, Ma J, Liang J, Tang G, Laurent R, Mignani S, Majoral JP, Shi X, Shen M. Bioactive phosphorus dendrimers deliver protein/drug to tackle osteoarthritis via cooperative macrophage reprogramming. Biomaterials 2025; 316:122999. [PMID: 39647219 DOI: 10.1016/j.biomaterials.2024.122999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 11/22/2024] [Accepted: 12/04/2024] [Indexed: 12/10/2024]
Abstract
Reprogramming imbalanced synovial macrophages and shaping an immune microenvironment conducive to bone and cartilage growth is crucial for efficient tackling of osteoarthritis (OA). Herein, we present a co-delivery nanosystem based on generation 2 (G2) hydroxyl-terminated bioactive phosphorus dendrimers (G2-OH24) that were loaded with both catalase (CAT) and quercetin (Que). The created G2-OH24/CAT@Que complexes exhibit a uniformly distributed spherical morphology with a size of 138.8 nm, possess robust stability, and induce macrophage reprogramming toward anti-inflammatory M2 phenotype polarization and antioxidation through cooperative CAT-catalyzed oxygen generation, Que-mediated mitochondrial homeostasis restoration, and inherent immunomodulatory activity of dendrimer. Such macrophage reprogramming leads to chondrocyte apoptosis inhibition and osteogenic differentiation of bone mesenchymal stem cells. Administration of G2-OH24/CAT@Que to an OA mouse model results in attenuation of pathological features such as cartilage degeneration, bone erosion, and synovitis through oxidative stress alleviation and inflammatory factor downregulation in inflamed joints. Excitingly, the G2-OH24/CAT@Que also polarized macrophages in adherent effusion monocytes (AEMs) extracted from joint cavity effusions of OA patients to M2 phenotype and downregulated reactive oxygen species levels in AEMs. This study suggests a promising nanomedicine formulation of phosphorus dendrimer-based co-delivery system to effectively tackle OA through the benefits of full-active ingredients of dendrimer, drug, and protein.
Collapse
Affiliation(s)
- Huxiao Sun
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, China
| | - Mengsi Zhan
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, China
| | - Yu Zou
- Laboratoire de Chimie de Coordination du CNRS, 205 Route de Narbonne, CEDEX 4, 31077, Toulouse, France; Université Toulouse, 118 Route de Narbonne, CEDEX 4, 31077, Toulouse, France
| | - Jie Ma
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, China
| | - Jiajia Liang
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, China
| | - Guo Tang
- Songjiang Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 201600, China
| | - Regis Laurent
- Laboratoire de Chimie de Coordination du CNRS, 205 Route de Narbonne, CEDEX 4, 31077, Toulouse, France; Université Toulouse, 118 Route de Narbonne, CEDEX 4, 31077, Toulouse, France
| | - Serge Mignani
- CQM-Centro de Química da Madeira, Universidade da Madeira, Campus Universitário da Penteada, 9020-105, Funchal, Portugal
| | - Jean-Pierre Majoral
- Laboratoire de Chimie de Coordination du CNRS, 205 Route de Narbonne, CEDEX 4, 31077, Toulouse, France; Université Toulouse, 118 Route de Narbonne, CEDEX 4, 31077, Toulouse, France
| | - Xiangyang Shi
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, China; CQM-Centro de Química da Madeira, Universidade da Madeira, Campus Universitário da Penteada, 9020-105, Funchal, Portugal.
| | - Mingwu Shen
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, China.
| |
Collapse
|
2
|
Tan S, Wu W, Chen Y, Gao H. High glucose induces senescence in synovial mesenchymal stem cells through mitochondrial dysfunction. BMC Oral Health 2025; 25:569. [PMID: 40234847 PMCID: PMC11998213 DOI: 10.1186/s12903-025-05938-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 04/02/2025] [Indexed: 04/17/2025] Open
Abstract
PURPOSE To investigate the impact of high glucose on the senescence of synovial mesenchymal stem cells (SMSCs) and to elucidate the role of mitochondrial dysfunction in this process. METHODS SMSCs were treated with medium containing high glucose (25 mmol/L) or low glucose (5.5 mmol/L) concentrations. The effects of high glucose concentrations on the proliferation, senescence, mitochondrial reactive oxygen species (ROS) levels, mitochondrial fission, and mitophagy of SMSCs were investigated. First, the impact of 24-hour high glucose treatment on SMSCs was investigated. After this initial 24-hour exposure, the medium was subsequently changed to low glucose, and the cells were cultivated for an additional 24 h; this was then compared with the effects of continuous 48-hour high-glucose exposure and continuous 48-hour low-glucose exposure. RESULTS High glucose concentrations did not promote the proliferation of SMSCs but rather accelerated their senescence by upregulating the mRNA expression of senescence-associated secretory phenotype (SASP) genes and increasing the number of senescence-associated β-galactosidase (SA-β-gal)-positive cells. Additionally, high glucose concentrations elevated ROS levels in mitochondria and facilitated mitochondrial fission; they also inhibited the mitophagy of SMSCs by suppressing the expression of mitophagy-related proteins (PINK1, PARKIN, and LC3B). High glucose-induced suppression of mRNA (Il-6, Cxcl1, Dnm1, Pink1, Prkn, Lc3b) and protein (P21) expression, along with increased SA-β-gal-positive cell numbers and elevated MitoSOX intensity, can be reversed by terminating the high glucose treatment. CONCLUSION High glucose concentrations induce senescence in SMSCs via mitochondrial dysfunction, manifested as ROS accumulation, excessive fission, and mitophagy suppression. Glucose normalization reversed senescence phenotypes, accompanied by restored mitophagy and reduced oxidative stress. Mitochondrial dysfunction may be one of the key mechanisms underlying high glucose-induced senescence in SMSCs.
Collapse
Affiliation(s)
- Shuyi Tan
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, China
| | - Wangxi Wu
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, China
| | - Yifan Chen
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, China
| | - Hai Gao
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, China.
| |
Collapse
|
3
|
Feng K, Liu J, Gong L, Ye T, Chen Z, Wang Y, Li Q, Xie X. Engineered MSC-sEVs as a Versatile Nanoplatform for Enhanced Osteoarthritis Treatment via Targeted Elimination of Senescent Chondrocytes and Maintenance of Cartilage Matrix Metabolic Homeostasis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2413759. [PMID: 39755936 PMCID: PMC11848604 DOI: 10.1002/advs.202413759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Revised: 12/17/2024] [Indexed: 01/06/2025]
Abstract
Chondrocyte senescence is an important pathogenic factor causing osteoarthritis (OA) progression through persistently producing pro-inflammatory factors. Mesenchymal stem cells-derived small extracellular vesicles (MSC-sEVs) have shown anti-inflammatory effects in OA models, while persistent existence of senescent chondrocytes still promotes cartilage destruction. Therefore, improving the targeted elimination ability on senescent chondrocytes is required to facilitate the translation of MSC-sEVs in OA treatment. In this study, versatile engineered MSC-sEVs are developed to targetedly clear senescent chondrocytes and maintain cartilage metabolic homeostasis. Specifically, MSC-sEVs are loaded with siRNA mouse double minute 2 homologue (siMDM2) and modified with cartilage-targeting peptide WYRGRL-PEG2K-DSPE (WPD), named WPD-sEVssiMDM2. The results demonstrate versatile modification improves the cellular uptake of MSC-sEVs in chondrocytes, and thus improves the antiaging effects. Importantly, multifunctional modification enhances cartilage penetration ability and extends joint retention time of MSC-sEVs. In both post-traumatic OA mice and naturally aged mice, WPD-sEVssiMDM2 more effectively eliminates senescent chondrocytes and maintained matrix metabolic homeostasis. By using the P53 phosphorylation inhibitor, the essential role MDM2-P53 pathway in the antiaging function of WPD-sEVssiMDM2 on chondrocytes is verified. In ex vivo cultured human OA cartilage explants, it is confirmed that WPD-sEVssiMDM2 alleviates senescent phenotype. Altogether, the findings suggest that WPD-sEVssiMDM2 have promising translational potential for OA treatment.
Collapse
Affiliation(s)
- Kai Feng
- Institute of Microsurgery on ExtremitiesDepartment of Orthopedic SurgeryShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghai200233China
| | - Jiashuo Liu
- Institute of Microsurgery on ExtremitiesDepartment of Orthopedic SurgeryShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghai200233China
| | - Liangzhi Gong
- Institute of Microsurgery on ExtremitiesDepartment of Orthopedic SurgeryShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghai200233China
| | - Teng Ye
- Institute of Microsurgery on ExtremitiesDepartment of Orthopedic SurgeryShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghai200233China
| | - Zhengsheng Chen
- Institute of Microsurgery on ExtremitiesDepartment of Orthopedic SurgeryShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghai200233China
| | - Yang Wang
- Institute of Microsurgery on ExtremitiesDepartment of Orthopedic SurgeryShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghai200233China
| | - Qing Li
- Institute of Microsurgery on ExtremitiesDepartment of Orthopedic SurgeryShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghai200233China
| | - Xuetao Xie
- Institute of Microsurgery on ExtremitiesDepartment of Orthopedic SurgeryShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghai200233China
| |
Collapse
|
4
|
Liu Y, Wang Y, Kulkarni RA, Wegiel LA, Lee B, Bedingfield SK, Weitz D. Syringable Microcapsules for Sustained, Localized, and Controllable siRNA Delivery. ACS APPLIED MATERIALS & INTERFACES 2025; 17:187-196. [PMID: 39705128 DOI: 10.1021/acsami.4c12805] [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: 12/22/2024]
Abstract
The clinical use of small interfering RNA (siRNA) and antisense oligonucleotides often requires invasive routes of administration, including intrathecal or intraocular injection. Additionally, these treatments often necessitate repeated injections. While nanoparticle formulation and chemical modifications have extended siRNA therapeutic durability, challenges persist, such as the side effects of bolus injections with high toxicity and maximum exposure in the acute phase. We present a microcapsule-based method to extend the activity of cholesterol-conjugated siRNA locally. Using microfluidics, microcapsules with well-defined size distribution and shell thickness are fabricated with poly(lactic-co-glycolic acid) (PLGA) with varying molecular weights and compositions. The microcapsules show a remarkably high drug encapsulation efficiency of nearly 100% and a high loading capacity (8900 μg siRNA/1 mg polymer). Additionally, microcapsules with an average diameter of 40 μm show superior syringeability when tested with needles ranging from gauge sizes of 27 to 32 G. This makes them suitable for various injection routes. Two sustained-release formulations were selected based on a 3-month in vitro release test. Subsequently, these formulations were injected subcutaneously into mice to verify their in vivo release profiles. The findings demonstrate that the microcapsules effectively shield the siRNAs from being cleared and enable them to be released constantly over 3 months. In contrast, unencapsulated siRNAs are rapidly cleared.
Collapse
Affiliation(s)
- Yan Liu
- School of Engineering and Applied Sciences (SEAS), Harvard University, Cambridge, Massachusetts 02138, United States
| | - Yang Wang
- School of Engineering and Applied Sciences (SEAS), Harvard University, Cambridge, Massachusetts 02138, United States
| | | | | | - Byungkook Lee
- Eli Lilly & Company, Indianapolis, Indiana 46285, United States
| | | | - David Weitz
- School of Engineering and Applied Sciences (SEAS), Harvard University, Cambridge, Massachusetts 02138, United States
- Department of Physics, Harvard University, Cambridge, Massachusetts 02138, United States
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, Massachusetts 02138, United States
| |
Collapse
|
5
|
Chen M, Wang J, Liu Y, Cui X, Liang J, Lei N, Xiao Y, Wang Q, Fan Y, Zhang X. Injectable biomimetic microcarriers harness synergistic effects of paracrine factors and cellular membranes to alleviate osteoarthritis. CHEMICAL ENGINEERING JOURNAL 2025; 503:158451. [DOI: 10.1016/j.cej.2024.158451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
6
|
Wang D, Liu W, Venkatesan JK, Madry H, Cucchiarini M. Therapeutic Controlled Release Strategies for Human Osteoarthritis. Adv Healthc Mater 2025; 14:e2402737. [PMID: 39506433 PMCID: PMC11730424 DOI: 10.1002/adhm.202402737] [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: 07/24/2024] [Revised: 10/15/2024] [Indexed: 11/08/2024]
Abstract
Osteoarthritis is a progressive, irreversible debilitating whole joint disease that affects millions of people worldwide. Despite the availability of various options (non-pharmacological and pharmacological treatments and therapy, orthobiologics, and surgical interventions), none of them can definitively cure osteoarthritis in patients. Strategies based on the controlled release of therapeutic compounds via biocompatible materials may provide powerful tools to enhance the spatiotemporal delivery, expression, and activities of the candidate agents as a means to durably manage the pathological progression of osteoarthritis in the affected joints upon convenient intra-articular (injectable) delivery while reducing their clearance, dissemination, or side effects. The goal of this review is to describe the current knowledge and advancements of controlled release to treat osteoarthritis, from basic principles to applications in vivo using therapeutic recombinant molecules and drugs and more innovatively gene sequences, providing a degree of confidence to manage the disease in patients in a close future.
Collapse
Affiliation(s)
- Dan Wang
- Center of Experimental OrthopaedicsSaarland University and Saarland University Medical CenterKirrbergerstr. Bldg 37D‐66421Homburg/SaarGermany
| | - Wei Liu
- Center of Experimental OrthopaedicsSaarland University and Saarland University Medical CenterKirrbergerstr. Bldg 37D‐66421Homburg/SaarGermany
| | - Jagadeesh K. Venkatesan
- Center of Experimental OrthopaedicsSaarland University and Saarland University Medical CenterKirrbergerstr. Bldg 37D‐66421Homburg/SaarGermany
| | - Henning Madry
- Center of Experimental OrthopaedicsSaarland University and Saarland University Medical CenterKirrbergerstr. Bldg 37D‐66421Homburg/SaarGermany
| | - Magali Cucchiarini
- Center of Experimental OrthopaedicsSaarland University and Saarland University Medical CenterKirrbergerstr. Bldg 37D‐66421Homburg/SaarGermany
| |
Collapse
|
7
|
Kim JY, Rhim WK, Lee SY, Park JM, Song DH, Cha SG, Lee SH, Hwang DY, Kim BJ, Rho S, Ahn TK, Park CG, Han DK. Hybrid Nanoparticle Engineered with Transforming Growth Factor -β1-Overexpressed Extracellular Vesicle and Cartilage-Targeted Anti-Inflammatory Liposome for Osteoarthritis. ACS NANO 2024; 18:33937-33952. [PMID: 39648484 DOI: 10.1021/acsnano.4c07992] [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: 12/10/2024]
Abstract
Extracellular vesicles (EVs) possess the characteristics of their parent cells, based on which various studies have actively investigated treatments for diseases using mesenchymal stem cell-derived EVs due to their regenerative activity. Furthermore, in recent years, there have been significant efforts to engineer EVs to improve their native activities and integrate additional functions. Although both endogenous and exogenous methods are used for engineering EVs, endogenous methods may pose the problem of administering substances to cells undergoing metabolic changes, which can cause potential side effects. In addition, exogenous methods may have the limitation of losing beneficial factors inside EVs due to membrane disruption during engineering processes. Surface modification of EVs may also impair efficiency due to the presence of proteins on the EV surface. Therefore, in this study, a stable and efficient engineering method was achieved through the ethanol-mediated hybridization of EVs and functionalized lipid nanoparticles (LNPs) with a fusogenic lipid component. During hybridization, the internal bioactive factors and targeting moiety were maintained to possess the characteristics of both LNPs and EVs. The Ab-Hybrid, which was successfully synthesized through hybridization with nicotinamide-encapsulated and Col2A1 antibody-modified liposome and Transforming growth factor-β1 (TGF-β1)-overexpressed EVs, was administered to osteoarthritis (OA)-induced rats undergoing the destabilization of the medial meniscus surgery. Ultimately, the Ab-Hybrid demonstrated excellent chondroprotective and anti-inflammatory effects with targeting and long-lasting properties in OA lesions. We anticipate that this approach for manufacturing hybrid particles will serve as a valuable EV engineering method and a versatile platform technology applicable to various diseases.
Collapse
Affiliation(s)
- Jun Yong Kim
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do 13488, Republic of Korea
- Department of Biomedical Engineering, Sungkyunkwan University (SKKU), 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyeonggi-do 16419, Republic of Korea
- Intelligent Precision of Healthcare Convergence, SKKU Institute for Convergence, Sungkyunkwan University (SKKU), 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyeonggi-do 16419, Republic of Korea
| | - Won-Kyu Rhim
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do 13488, Republic of Korea
- Department of Ophthalmology, CHA Bundang Medical Center, CHA University, Seongnam-si 13496, Republic of Korea
| | - Seung Yeon Lee
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do 13488, Republic of Korea
| | - Jung Min Park
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do 13488, Republic of Korea
| | - Duck Hyun Song
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do 13488, Republic of Korea
| | - Seung-Gyu Cha
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do 13488, Republic of Korea
| | - Sang-Hyuk Lee
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do 13488, Republic of Korea
| | - Dong-Youn Hwang
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do 13488, Republic of Korea
| | - Byoung Ju Kim
- ATEMs, Jeongui-ro 8-gil, Songpa-gu, Seoul-si 05836, Republic of Korea
| | - Seungsoo Rho
- Department of Ophthalmology, CHA Bundang Medical Center, CHA University, Seongnam-si 13496, Republic of Korea
| | - Tae-Keun Ahn
- Department of Orthopedic Surgery, CHA Bundang Medical Center CHA University, Seongnam-si 13496, Republic of Korea
| | - Chun Gwon Park
- Department of Biomedical Engineering, Sungkyunkwan University (SKKU), 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyeonggi-do 16419, Republic of Korea
- Intelligent Precision of Healthcare Convergence, SKKU Institute for Convergence, Sungkyunkwan University (SKKU), 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyeonggi-do 16419, Republic of Korea
| | - Dong Keun Han
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do 13488, Republic of Korea
| |
Collapse
|
8
|
Raza IGA, Snelling SJB, Mimpen JY. Defining the extracellular matrix in non-cartilage soft-tissues in osteoarthritis: a systematic review. Bone Joint Res 2024; 13:703-715. [PMID: 39622273 PMCID: PMC11611391 DOI: 10.1302/2046-3758.1312.bjr-2024-0020.r1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/06/2024] Open
Abstract
Aims Extracellular matrix (ECM) is a critical determinant of tissue mechanobiology, yet remains poorly characterized in joint tissues beyond cartilage in osteoarthritis (OA). This review aimed to define the composition and architecture of non-cartilage soft joint tissue structural ECM in human OA, and to compare the changes observed in humans with those seen in animal models of the disease. Methods A systematic search strategy, devised using relevant matrix, tissue, and disease nomenclature, was run through the MEDLINE, Embase, and Scopus databases. Demographic, clinical, and biological data were extracted from eligible studies. Bias analysis was performed. Results A total of 161 studies were included, which covered capsule, ligaments, meniscus, skeletal muscle, synovium, and tendon in both humans and animals, and fat pad and intervertebral disc in humans only. These studies covered a wide variety of ECM features, including individual ECM components (i.e. collagens, proteoglycans, and glycoproteins), ECM architecture (i.e. collagen fibre organization and diameter), and viscoelastic properties (i.e. elastic and compressive modulus). Some ECM changes, notably calcification and the loss of collagen fibre organization, have been extensively studied across osteoarthritic tissues. However, most ECM features were only studied by one or a few papers in each tissue. When comparisons were possible, the results from animal experiments largely concurred with those from human studies, although some findings were contradictory. Conclusion Changes in ECM composition and architecture occur throughout non-cartilage soft tissues in the osteoarthritic joint, but most of these remain poorly defined due to the low number of studies and lack of healthy comparator groups.
Collapse
Affiliation(s)
| | - Sarah J. B. Snelling
- Botnar Institute for Musculoskeletal Sciences, Nuffield Department of Orthopaedics Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Jolet Y. Mimpen
- Botnar Institute for Musculoskeletal Sciences, Nuffield Department of Orthopaedics Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| |
Collapse
|
9
|
Scanzello CR, Hasty KA, Chung CB, Griffin TM, Willet NJ, Krug H, Chu CQ, Ewart D, Jerban S, Baker JF, Duvall CL, Brunger JM, Burdick JA, Spindler KP, Drissi H. Teaming up to overcome challenges toward translation of new therapeutics for osteoarthritis. J Orthop Res 2024; 42:2659-2672. [PMID: 39103981 DOI: 10.1002/jor.25944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 06/18/2024] [Accepted: 07/09/2024] [Indexed: 08/07/2024]
Abstract
As a leading global cause of musculoskeletal-related disability, osteoarthritis (OA) represents a public health urgency. Understanding of disease pathogenesis has advanced substantially in the past decade, yet no disease-modifying therapeutics have advanced to the clinic. To address this challenge, the CARE-AP (Cartilage Repair strategies to alleviate Arthritis Pain) collaborative research team was convened to bring together relevant multidisciplinary expertise and perspectives from across the VA research community nationwide. The first CARE-AP Annual Research Symposium took place (virtually) in February 2022 with roughly 90 participants. A number of innovative and therapeutic strategies were discussed, including siRNA approaches coupled with novel nanoparticle-based delivery systems, cellular engineering approaches to develop reparative cells that can probe the joint environment and respond to disease-specific cues, and novel biofabrication techniques to improve tissue engineering and effect "biological joint replacement." In addition, challenges and advances in rehabilitation approaches, imaging outcomes, and clinical studies were presented, which were integrated into a framework of recommendations for running "preclinical trials" to improve successful clinical translation.
Collapse
Affiliation(s)
- Carla R Scanzello
- Translational Musculoskeletal Research Center, Corp. Michael J. Crescenz VA Medical Center, Philadelphia, Pennsylvania, USA
- Division of Rheumatology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Karen A Hasty
- Research Service 151, Lt. Col. Luke Weathers, Jr. VA Medical Center, Memphis, Tennessee, USA
- Department of Orthopaedic Surgery and Biomedical Engineering, Campbell Clinic/University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Christine B Chung
- Radiology Service, Veterans Affairs San Diego Healthcare System, San Diego, California, USA
- Department of Radiology, University of California San Diego, La Jolla, California, USA
| | - Timothy M Griffin
- Oklahoma City VA Health Care System, Oklahoma City, Oklahoma, USA
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Nick J Willet
- Veterans Affairs Portland Health Care System, Portland, Oregon, USA
- Phil and Penny Knight Campus for Accelerating Scientific Impact, Department of Bioengineering, University of Oregon, Eugene, Oregon, USA
| | - Hollis Krug
- Rheumatology Section, Minneapolis Veterans Affairs Medical Center, Minneapolis, Minnesota, USA
- Division of Rheumatology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Cong-Qiu Chu
- Veterans Affairs Portland Health Care System, Portland, Oregon, USA
- Division of Arthritis and Rheumatic Diseases, Oregon Health Sciences University, Portland, Oregon, USA
| | - David Ewart
- Rheumatology Section, Minneapolis Veterans Affairs Medical Center, Minneapolis, Minnesota, USA
- Division of Rheumatology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Saeed Jerban
- Radiology Service, Veterans Affairs San Diego Healthcare System, San Diego, California, USA
- Department of Radiology, University of California San Diego, La Jolla, California, USA
| | - Joshua F Baker
- Translational Musculoskeletal Research Center, Corp. Michael J. Crescenz VA Medical Center, Philadelphia, Pennsylvania, USA
- Division of Rheumatology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Craig L Duvall
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, USA
| | - Jonathan M Brunger
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, USA
- Center for Stem Cell Biology, Vanderbilt University, Nashville, Tennessee, USA
| | - Jason A Burdick
- BioFrontiers Institute and Department of Chemical and Biological Engineering, University of Colorado, Boulder, Colorado, USA
| | - Kurt P Spindler
- Department of Orthopaedic Surgery, Sports Medicine, Cleveland Clinic Florida, Coral Springs, Florida, USA
| | - Hicham Drissi
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, Georgia, USA
- Atlanta VA Medical Center, Decatur, Georgia, USA
| |
Collapse
|
10
|
Hridayanka KSN, Duttaroy AK, Basak S. Bioactive Compounds and Their Chondroprotective Effects for Osteoarthritis Amelioration: A Focus on Nanotherapeutic Strategies, Epigenetic Modifications, and Gut Microbiota. Nutrients 2024; 16:3587. [PMID: 39519419 PMCID: PMC11547880 DOI: 10.3390/nu16213587] [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: 09/24/2024] [Revised: 10/20/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024] Open
Abstract
In degenerative joint disease like osteoarthritis (OA), bioactive compounds like resveratrol, epigallocatechin gallate, curcumin, and other polyphenols often target various signalling pathways, including NFκB, TGFβ, and Wnt/β-catenin by executing epigenetic-modifying activities. Epigenetic modulation can target genes of disease pathophysiology via histone modification, promoter DNA methylation, and non-coding RNA expression, some of which are directly involved in OA but have been less explored. OA patients often seek options that can improve the quality of their life in addition to existing treatment with nonsteroidal anti-inflammatory drugs (NSAIDs). Although bioactive and natural compounds exhibit therapeutic potential against OA, several disadvantages loom, like insolubility and poor bioavailability. Nanoformulated bioactive compounds promise a better way to alleviate OA since they also control systemic events, including metabolic, immunological, and inflammatory responses, by modulating host gut microbiota that can regulate OA pathogenesis. Recent data suggest gut dysbiosis in OA. However, limited evidence is available on the role of bioactive compounds as epigenetic and gut modulators in ameliorating OA. Moreover, it is not known whether the effects of polyphenolic bioactive compounds on gut microbial response are mediated by epigenetic modulatory activities in OA. This narrative review highlights the nanotherapeutic strategies utilizing bioactive compounds, reporting their effects on chondrocyte growth, metabolism, and epigenetic modifications in osteoarthritis amelioration.
Collapse
Affiliation(s)
- Kota Sri Naga Hridayanka
- Molecular Biology Division, National Institute of Nutrition, Indian Council of Medical Research, Hyderabad 500007, India;
| | - Asim K. Duttaroy
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, 0317 Oslo, Norway;
| | - Sanjay Basak
- Molecular Biology Division, National Institute of Nutrition, Indian Council of Medical Research, Hyderabad 500007, India;
| |
Collapse
|
11
|
Fragassi A, Greco A, Palomba R. Lubricant Strategies in Osteoarthritis Treatment: Transitioning from Natural Lubricants to Drug Delivery Particles with Lubricant Properties. J Xenobiot 2024; 14:1268-1292. [PMID: 39311151 PMCID: PMC11417909 DOI: 10.3390/jox14030072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 09/06/2024] [Accepted: 09/13/2024] [Indexed: 09/26/2024] Open
Abstract
Osteoarthritis (OA) is a debilitating joint disease characterized by cartilage degradation, leading to pain and functional impairment. A key contributor to OA progression is the decline in cartilage lubrication. In physiological conditions, synovial fluid (SF) macromolecules like hyaluronic acid (HA), phospholipids, and lubricin play a crucial role in the boundary lubrication of articular cartilage. In early OA, cartilage damage triggers inflammation, altering SF composition and compromising the lubrication layer. This increases friction between mating interfaces, worsening cartilage degradation and local inflammation. Therefore, early-stage restoration of lubrication (by injecting in the joint different classes of compounds and formulations) could alleviate, and potentially reverse, OA progression. In the light of this, a broad variety of lubricants have been investigated for their ability to reduce friction in OA joints and promote cartilage repair in clinical and preclinical studies. This review examines recent advancements in lubricant-based therapy for OA, focusing on natural, bioinspired, and alternative products. Starting from the currently applied therapy, mainly based on natural lubricants as HA, we will present their modified versions, either in hydrogel form or with specific biomimetic moieties with the aim of reducing their clearance from the joint and of enhancing their lubricating properties. Finally, the most advanced and recent formulation, represented by alternative strategies, will be proposed. Particular emphasis will be placed on those ones involving new types of hydrogels, microparticles, nanoparticles, and liposomes, which are currently under investigation in preclinical studies. The potential application of particles and liposomes could foster the transition from natural lubricants to Drug Delivery Systems (DDSs) with lubricant features; transition which could provide more complete OA treatments, by simultaneously providing lubrication replacement and sustained release of different payloads and active agents directly at the joint level. Within each category, we will examine relevant preclinical studies, highlighting challenges and future prospects.
Collapse
Affiliation(s)
- Agnese Fragassi
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via F. Marzolo 5, 35131 Padova, Italy
| | - Antonietta Greco
- Department of Medicine and Surgery, NanoMedicine Center (NANOMIB), University of Milano-Bicocca, Via Follereau 3, 20854 Vedano al Lambro, Italy
| | - Roberto Palomba
- Laboratory of Nanotechnology for Precision Medicine, Italian Institute of Technology, Via Morego 30, 16163 Genova, Italy
| |
Collapse
|
12
|
Zhou D, Wei Y, Sheng S, Wang M, Lv J, Zhao B, Chen X, Xu K, Bai L, Wu Y, Song P, Cao L, Zhou F, Zhang H, Shi Z, Su J. MMP13-targeted siRNA-loaded micelles for diagnosis and treatment of posttraumatic osteoarthritis. Bioact Mater 2024; 37:378-392. [PMID: 38689658 PMCID: PMC11059470 DOI: 10.1016/j.bioactmat.2024.04.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 04/09/2024] [Accepted: 04/10/2024] [Indexed: 05/02/2024] Open
Abstract
Posttraumatic osteoarthritis (PTOA) patients are often diagnosed by X-ray imaging at a middle-late stage when drug interventions are less effective. Early PTOA is characterized by overexpressed matrix metalloprotease 13 (MMP13). Herein, we constructed an integrated diagnosis and treatment micelle modified with MMP13 enzyme-detachable, cyanine 5 (Cy5)-containing PEG, black hole quencher-3 (BHQ3), and cRGD ligands and loaded with siRNA silencing MMP13 (siM13), namely ERMs@siM13. ERMs@siM13 could be cleaved by MMP13 in the diseased cartilage tissues to detach the PEG shell, causing cRGD exposure. Accordingly, the ligand exposure promoted micelle uptake by the diseased chondrocytes by binding to cell surface αvβ3 integrin, increasing intracellular siM13 delivery for on-demand MMP13 downregulation. Meanwhile, the Cy5 fluorescence was restored by detaching from the BHQ3-containing micelle, precisely reflecting the diseased cartilage state. In particular, the intensity of Cy5 fluorescence generated by ERMs@siM13 that hinged on the MMP13 levels could reflect the PTOA severity, enabling the physicians to adjust the therapeutic regimen. Finally, in the murine PTOA model, ERMs@siM13 could diagnose the early-stage PTOA, perform timely interventions, and monitor the OA progression level during treatment through a real-time detection of MMP13. Therefore, ERMs@siM13 represents an appealing approach for early-stage PTOA theranostics.
Collapse
Affiliation(s)
- Dongyang Zhou
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai), Shanghai University, Shanghai, 200444, China
- College of Medicine, Shanghai University, Shanghai, 200444, China
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, China
| | - Yan Wei
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai), Shanghai University, Shanghai, 200444, China
| | - Shihao Sheng
- Department of Orthopedic, Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Miaomiao Wang
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai), Shanghai University, Shanghai, 200444, China
- Department of Rehabilitation Medicine, Shanghai Zhongye Hospital, Shanghai, 200941, China
| | - Jiajing Lv
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai), Shanghai University, Shanghai, 200444, China
- College of Medicine, Shanghai University, Shanghai, 200444, China
| | - Bowen Zhao
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai), Shanghai University, Shanghai, 200444, China
- College of Medicine, Shanghai University, Shanghai, 200444, China
| | - Xiao Chen
- Department of Orthopedic, Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Ke Xu
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai), Shanghai University, Shanghai, 200444, China
| | - Long Bai
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai), Shanghai University, Shanghai, 200444, China
| | - Yan Wu
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai), Shanghai University, Shanghai, 200444, China
| | - Peiran Song
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai), Shanghai University, Shanghai, 200444, China
| | - Liehu Cao
- Department of Orthopedics, Shanghai Baoshan Luodian Hospital, Baoshan District, Shanghai, 201908, China
| | - Fengjin Zhou
- Department of Orthopaedics, Honghui Hospital, Xi'an Jiao Tong University, Xi'an, 710000, China
| | - Hao Zhang
- Department of Orthopedic, Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Zhongmin Shi
- Department of Orthopedics, Sixth People's Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, 200233, China
| | - Jiacan Su
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai), Shanghai University, Shanghai, 200444, China
- Department of Orthopedic, Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| |
Collapse
|
13
|
Li Y, Zhao J, Guo S, He D. siRNA therapy in osteoarthritis: targeting cellular pathways for advanced treatment approaches. Front Immunol 2024; 15:1382689. [PMID: 38895116 PMCID: PMC11184127 DOI: 10.3389/fimmu.2024.1382689] [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: 02/06/2024] [Accepted: 05/17/2024] [Indexed: 06/21/2024] Open
Abstract
Osteoarthritis (OA) is a common joint disorder characterized by the degeneration of cartilage and inflammation, affecting millions worldwide. The disease's complex pathogenesis involves various cell types, such as chondrocytes, synovial cells, osteoblasts, and immune cells, contributing to the intricate interplay of factors leading to tissue degradation and pain. RNA interference (RNAi) therapy, particularly through the use of small interfering RNA (siRNA), emerges as a promising avenue for OA treatment due to its capacity for specific gene silencing. siRNA molecules can modulate post-transcriptional gene expression, targeting key pathways involved in cellular proliferation, apoptosis, senescence, autophagy, biomolecule secretion, inflammation, and bone remodeling. This review delves into the mechanisms by which siRNA targets various cell populations within the OA milieu, offering a comprehensive overview of the potential therapeutic benefits and challenges in clinical application. By summarizing the current advancements in siRNA delivery systems and therapeutic targets, we provide a solid theoretical foundation for the future development of novel siRNA-based strategies for OA diagnosis and treatment, paving the way for innovative and more effective approaches to managing this debilitating disease.
Collapse
Affiliation(s)
- Yunshen Li
- Department of Rheumatology, Shanghai Guanghua Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Jianan Zhao
- Department of Rheumatology, Shanghai Guanghua Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Shicheng Guo
- Department of Rheumatology, Shanghai Guanghua Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Dongyi He
- Department of Rheumatology, Shanghai Guanghua Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
14
|
Yan W, Li Y, Xie S, Tao WA, Hu J, Liu H, Zhang G, Liu F, Nie Y, Chen X, Zhang X, Liu Y, Wei D, Ma C, Zhang H, Xu H, Wang S. Chondrocyte-Targeted Delivery System of Sortase A-Engineered Extracellular Vesicles Silencing MMP13 for Osteoarthritis Therapy. Adv Healthc Mater 2024; 13:e2303510. [PMID: 38545904 DOI: 10.1002/adhm.202303510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 03/21/2024] [Indexed: 04/09/2024]
Abstract
Targeted drug delivery and the reduction of off-target effects are crucial for the promising clinical application of nucleic acid drugs. To address this challenge, a new approach for treating osteoarthritis (OA) that accurately delivers antisense oligonucleotides (ASO) targeting matrix metalloproteinase-13 (ASO-MMP13) to chondrocytes, is developed. Small extracellular vesicles (exos) are ligated with chondrocyte affinity peptide (CAP) using Sortase A and subsequently incubated with cholesterol-modified ASO-MMP13 to construct a chondrocyte-targeted drug delivery exo (CAP-exoASO). Compared with exos without CAP (ExoASO), CAP-exoASOs attenuate IL-1β-induced chondrocyte damage and prolong the retention time of ASO-MMP13 in the joint without distribution in major organs following intra-articular injection. Notably, CAP-exoASOs decrease MMP13 expression (P < 0.001) and upregulate COL2A1 expression (P = 0.006), resulting in reorganization of the cartilage matrix and alleviation of progression in the OA model. Furthermore, the Osteoarthritis Research Society International (OARSI) score of articular cartilage tissues treated with CAP-exoASO is comparable with that of healthy rats (P = 0.148). A mechanistic study demonstrates that CAP-exoASO may reduce inflammation by suppressing the IL-17 and TNF signaling pathways. Based on the targeted delivery effect, CAP-exoASOs successfully accomplish cartilage repair and have considerable potential for development as a promising therapeutic modality for satisfactory OA therapy.
Collapse
Affiliation(s)
- Wenjing Yan
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, 210003, China
| | - Ying Li
- Center of Clinical Laboratory Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, 210009, China
- Department of Epidemiology, School of Public Health of Suzhou University, Suzhou, Jiangsu, 215127, China
| | - Shuqian Xie
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, 210003, China
| | - W Andy Tao
- Departments of Chemistry and Biochemistry, Purdue University, West Lafayette, IN, 47907, USA
| | - Jing Hu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, 210003, China
| | - Haohan Liu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, 210003, China
| | - Guiyuan Zhang
- State Key Laboratory of Bioelectronics, National Demonstration Center for Experimental Biomedical Engineering Education, Southeast University, Nanjing, Jiangsu, 210096, China
| | - Fengying Liu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, 210003, China
| | - Yamei Nie
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, 210003, China
| | - Xue Chen
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, 210003, China
| | - Xing Zhang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, 210003, China
| | - Yufeng Liu
- State Key Laboratory of Bioelectronics, National Demonstration Center for Experimental Biomedical Engineering Education, Southeast University, Nanjing, Jiangsu, 210096, China
| | - Dong Wei
- State Key Laboratory of Bioelectronics, National Demonstration Center for Experimental Biomedical Engineering Education, Southeast University, Nanjing, Jiangsu, 210096, China
| | - Changyan Ma
- Department of Medical Genetics, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Hao Zhang
- EVLiXiR Biotech Inc., Nanjing, Jiangsu, 210032, China
| | - Hongtao Xu
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Shizhi Wang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, 210003, China
| |
Collapse
|
15
|
Kim JY, Lee SY, Cha SG, Park JM, Song DH, Lee SH, Hwang DY, Kim BJ, Rho S, Park CG, Rhim WK, Han DK. Combinatory Nanovesicle with siRNA-Loaded Extracellular Vesicle and IGF-1 for Osteoarthritis Treatments. Int J Mol Sci 2024; 25:5242. [PMID: 38791285 PMCID: PMC11121733 DOI: 10.3390/ijms25105242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 05/07/2024] [Accepted: 05/09/2024] [Indexed: 05/26/2024] Open
Abstract
Extracellular vesicles (EVs) have been found to have the characteristics of their parent cells. Based on the characteristics of these EVs, various studies on disease treatment using mesenchymal stem cell (MSC)-derived EVs with regenerative activity have been actively conducted. The therapeutic nature of MSC-derived EVs has been shown in several studies, but in recent years, there have been many efforts to functionalize EVs to give them more potent therapeutic effects. Strategies for functionalizing EVs include endogenous and exogenous methods. In this study, human umbilical cord MSC (UCMSC)-derived EVs were selected for optimum OA treatments with expectation via bioinformatics analysis based on antibody array. And we created a novel nanovesicle system called the IGF-si-EV, which has the properties of both cartilage regeneration and long-term retention in the lesion site, attaching positively charged insulin-like growth factor-1 (IGF-1) to the surface of the UCMSC-derived Evs carrying siRNA, which inhibits MMP13. The downregulation of inflammation-related cytokine (MMP13, NF-kB, and IL-6) and the upregulation of cartilage-regeneration-related factors (Col2, Acan) were achieved with IGF-si-EV. Moreover, the ability of IGF-si-EV to remain in the lesion site for a long time has been proven through an ex vivo system. Collectively, the final constructed IGF-si-EV can be proposed as an effective OA treatment through its successful MMP13 inhibition, chondroprotective effect, and cartilage adhesion ability. We also believe that this EV-based nanoparticle-manufacturing technology can be applied as a platform technology for various diseases.
Collapse
Affiliation(s)
- Jun Yong Kim
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si 13488, Republic of Korea; (J.Y.K.); (S.Y.L.); (S.-G.C.); (J.M.P.); (D.H.S.); (S.-H.L.); (D.-Y.H.)
- Department of Biomedical Engineering, SKKU Institute for Convergence, Sungkyunkwan University (SKKU), 2066 Seobu-ro, Jangan-gu, Suwon-si 16419, Republic of Korea;
- Intelligent Precision of Healthcare Convergence, SKKU Institute for Convergence, Sungkyunkwan University (SKKU), 2066 Seobu-ro, Jangan-gu, Suwon-si 16419, Republic of Korea
| | - Seung Yeon Lee
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si 13488, Republic of Korea; (J.Y.K.); (S.Y.L.); (S.-G.C.); (J.M.P.); (D.H.S.); (S.-H.L.); (D.-Y.H.)
| | - Seung-Gyu Cha
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si 13488, Republic of Korea; (J.Y.K.); (S.Y.L.); (S.-G.C.); (J.M.P.); (D.H.S.); (S.-H.L.); (D.-Y.H.)
| | - Jung Min Park
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si 13488, Republic of Korea; (J.Y.K.); (S.Y.L.); (S.-G.C.); (J.M.P.); (D.H.S.); (S.-H.L.); (D.-Y.H.)
| | - Duck Hyun Song
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si 13488, Republic of Korea; (J.Y.K.); (S.Y.L.); (S.-G.C.); (J.M.P.); (D.H.S.); (S.-H.L.); (D.-Y.H.)
| | - Sang-Hyuk Lee
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si 13488, Republic of Korea; (J.Y.K.); (S.Y.L.); (S.-G.C.); (J.M.P.); (D.H.S.); (S.-H.L.); (D.-Y.H.)
| | - Dong-Youn Hwang
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si 13488, Republic of Korea; (J.Y.K.); (S.Y.L.); (S.-G.C.); (J.M.P.); (D.H.S.); (S.-H.L.); (D.-Y.H.)
| | - Byoung Ju Kim
- ATEMs, Jeongui-ro 8-gil, Songpa-gu, Seoul-si 05836, Republic of Korea;
| | - Seungsoo Rho
- Department of Ophthalmology, CHA Bundang Medical Center, CHA University, Seongnam-si 13496, Republic of Korea;
| | - Chun Gwon Park
- Department of Biomedical Engineering, SKKU Institute for Convergence, Sungkyunkwan University (SKKU), 2066 Seobu-ro, Jangan-gu, Suwon-si 16419, Republic of Korea;
- Intelligent Precision of Healthcare Convergence, SKKU Institute for Convergence, Sungkyunkwan University (SKKU), 2066 Seobu-ro, Jangan-gu, Suwon-si 16419, Republic of Korea
| | - Won-Kyu Rhim
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si 13488, Republic of Korea; (J.Y.K.); (S.Y.L.); (S.-G.C.); (J.M.P.); (D.H.S.); (S.-H.L.); (D.-Y.H.)
- Department of Ophthalmology, CHA Bundang Medical Center, CHA University, Seongnam-si 13496, Republic of Korea;
| | - Dong Keun Han
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si 13488, Republic of Korea; (J.Y.K.); (S.Y.L.); (S.-G.C.); (J.M.P.); (D.H.S.); (S.-H.L.); (D.-Y.H.)
| |
Collapse
|
16
|
Zhang H, Yan W, Wang J, Xie S, Tao WA, Lee CW, Zhang X, Zhang G, Liu Y, Wei D, Hu J, Liu H, Liu F, Nie Y, Chen X, Xu H, Xia J, Wang S. Surface functionalization of exosomes for chondrocyte-targeted siRNA delivery and cartilage regeneration. J Control Release 2024; 369:493-505. [PMID: 38582335 DOI: 10.1016/j.jconrel.2024.04.009] [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: 12/07/2023] [Revised: 03/25/2024] [Accepted: 04/03/2024] [Indexed: 04/08/2024]
Abstract
Osteoarthritis (OA) is the most prevalent degenerative cartilage disease, but no effective treatment is currently available to ameliorate the dysregulation of cartilage catabolism. Cartilage degeneration is closely related to the change in the physiology of chondrocytes: for example, chondrocytes of the OA patients overexpress matrix metallopeptidase 13 (MMP13), a.k.a. collagenase 3, which damages the extracellular matrix (ECM) of the cartilage and deteriorate the disease progression. Inhibiting MMP13 has shown to be beneficial for OA treatments, but delivering therapeutics to the chondrocytes embedded in the dense cartilage is a challenge. Here, we engineered the exosome surface with the cartilage affinity peptide (CAP) through lipid insertion to give chondrocyte-targeting exosomes, CAP-Exo, which was then loaded with siRNA against MMP13 (siMMP13) in the interior to give CAP-Exo/siMMP13. Intra-articular administration of CAP-Exo/siMMP13 reduced the MMP13 level and increased collagen COL2A1 and proteoglycan in cartilage in a rat model of anterior cruciate ligament transection (ACLT)-induced OA. Proteomic analysis showed that CAP-Exo/siMMP13 treatment restored the altered protein levels in the IL-1β-treated chondrocytes. Taken together, a facile exosome engineering method enabled targeted delivery of siRNA to chondrocytes and chondrocyte-specific silencing of MMP13 to attenuate cartilage degeneration.
Collapse
Affiliation(s)
- Hao Zhang
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu, China; EVLiXiR Biotech Inc., Nanjing 210032, Jiangsu, China
| | - Wenjing Yan
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, China
| | - Jinhui Wang
- Xiamen Children's Hospital, Xiamen, Fujian, China
| | - Shuqian Xie
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, China
| | - W Andy Tao
- Departments of Chemistry and Biochemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Chien-Wei Lee
- Center for Translational Genomics & Regenerative Medicine Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Xing Zhang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, China
| | - Guiyuan Zhang
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu, China
| | - Yufeng Liu
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu, China
| | - Dong Wei
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu, China
| | - Jing Hu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, China
| | - Haohan Liu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, China
| | - Fengying Liu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, China
| | - Yamei Nie
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, China
| | - Xue Chen
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, China
| | - Hongtao Xu
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Jiang Xia
- Department of Chemistry, the Chinese University of Hong Kong, Shatin, Hong Kong SAR, China..
| | - Shizhi Wang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, China.
| |
Collapse
|
17
|
Chen Y, Le Y, Yang J, Yang Y, Feng X, Cai J, Shang Y, Sugiarto S, Wei Q, Kai D, Zheng L, Zhao J. 3D Bioprinted Xanthan Hydrogels with Dual Antioxidant and Chondrogenic Functions for Post-traumatic Cartilage Regeneration. ACS Biomater Sci Eng 2024; 10:1661-1675. [PMID: 38364815 DOI: 10.1021/acsbiomaterials.3c01636] [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: 02/18/2024]
Abstract
Intra-articular trauma typically initiates the overgeneration of reactive oxidative species (ROS), leading to post-traumatic osteoarthritis and cartilage degeneration. Xanthan gum (XG), a branched polysaccharide, has shown its potential in many biomedical fields, but some of its inherent properties, including undesirable viscosity and poor mechanical stability, limit its application in 3D printed scaffolds for cartilage regeneration. In this project, we developed 3D bioprinted XG hydrogels by modifying XG with methacrylic (MA) groups for post-traumatic cartilage therapy. Our results demonstrated that the chemical modification optimized the viscoelasticity of the bioink, improved printability, and enhanced the mechanical properties of the resulting scaffolds. The XG hydrogels also exhibit decent ROS scavenging capacities to protect stem cells from oxidative stress. Furthermore, XGMA(H) (5% MA substitution) exhibited superior chondrogenic potential in vitro and promoted cartilage regeneration in vivo. These dual-functional XGMA hydrogels may provide a new opportunity for cartilage tissue engineering.
Collapse
Affiliation(s)
- Yuting Chen
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Yiguan Le
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330008, China
| | - Junxu Yang
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
- Department of Orthopaedics Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Yifeng Yang
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Xianjing Feng
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Jinhong Cai
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Yifeng Shang
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Sigit Sugiarto
- Institute of Sustainability for Chemicals, Energy, and Environment (ISCE2), Agency for Science, Technology, and Research (A*STAR), 2 Fusionopolis Way, Innovis, #08-03, 138634 Republic of Singapore
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, Innovis #08-03, 138634 Republic of Singapore
| | - Qingjun Wei
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
- Department of Orthopaedics Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Dan Kai
- Institute of Sustainability for Chemicals, Energy, and Environment (ISCE2), Agency for Science, Technology, and Research (A*STAR), 2 Fusionopolis Way, Innovis, #08-03, 138634 Republic of Singapore
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, Innovis #08-03, 138634 Republic of Singapore
| | - Li Zheng
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
- Guangxi Key Laboratory of Regenerative Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Jinmin Zhao
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
- Department of Orthopaedics Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
- Guangxi Key Laboratory of Regenerative Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| |
Collapse
|
18
|
DeJulius CR, Walton BL, Colazo JM, d'Arcy R, Francini N, Brunger JM, Duvall CL. Engineering approaches for RNA-based and cell-based osteoarthritis therapies. Nat Rev Rheumatol 2024; 20:81-100. [PMID: 38253889 PMCID: PMC11129836 DOI: 10.1038/s41584-023-01067-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/07/2023] [Indexed: 01/24/2024]
Abstract
Osteoarthritis (OA) is a chronic, debilitating disease that substantially impairs the quality of life of affected individuals. The underlying mechanisms of OA are diverse and are becoming increasingly understood at the systemic, tissue, cellular and gene levels. However, the pharmacological therapies available remain limited, owing to drug delivery barriers, and consist mainly of broadly immunosuppressive regimens, such as corticosteroids, that provide only short-term palliative benefits and do not alter disease progression. Engineered RNA-based and cell-based therapies developed with synthetic chemistry and biology tools provide promise for future OA treatments with durable, efficacious mechanisms of action that can specifically target the underlying drivers of pathology. This Review highlights emerging classes of RNA-based technologies that hold potential for OA therapies, including small interfering RNA for gene silencing, microRNA and anti-microRNA for multi-gene regulation, mRNA for gene supplementation, and RNA-guided gene-editing platforms such as CRISPR-Cas9. Various cell-engineering strategies are also examined that potentiate disease-dependent, spatiotemporally regulated production of therapeutic molecules, and a conceptual framework is presented for their application as OA treatments. In summary, this Review highlights modern genetic medicines that have been clinically approved for other diseases, in addition to emerging genome and cellular engineering approaches, with the goal of emphasizing their potential as transformative OA treatments.
Collapse
Affiliation(s)
- Carlisle R DeJulius
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Bonnie L Walton
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Juan M Colazo
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Richard d'Arcy
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Nora Francini
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Jonathan M Brunger
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA.
| | - Craig L Duvall
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
19
|
Colazo JM, Hoogenboezem EN, Keech MC, Francini N, Shah V, Yu F, Lo JH, Sorets AG, McCune JT, Cho H, DeJulius CR, Michell DL, Maerz T, Vickers KC, Gibson-Corley KN, Hasty KA, Crofford L, Cook RS, Duvall CL. Albumin-binding RNAi Conjugate for Carrier Free Treatment of Arthritis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.31.542971. [PMID: 37333210 PMCID: PMC10274717 DOI: 10.1101/2023.05.31.542971] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Osteoarthritis (OA) and rheumatoid arthritis (RA) are joint diseases that are associated with pain and lost quality of life. No disease modifying OA drugs are currently available. RA treatments are better established but are not always effective and can cause immune suppression. Here, an MMP13-selective siRNA conjugate was developed that, when delivered intravenously, docks onto endogenous albumin and promotes preferential accumulation in articular cartilage and synovia of OA and RA joints. MMP13 expression was diminished upon intravenous delivery of MMP13 siRNA conjugates, consequently decreasing multiple histological and molecular markers of disease severity, while also reducing clinical manifestations such as swelling (RA) and joint pressure sensitivity (RA and OA). Importantly, MMP13 silencing provided more comprehensive OA treatment efficacy than standard of care (steroids) or experimental MMP inhibitors. These data demonstrate the utility of albumin 'hitchhiking' for drug delivery to arthritic joints, and establish the therapeutic utility of systemically delivered anti-MMP13 siRNA conjugates in OA and RA. Editorial summary Lipophilic siRNA conjugates optimized for albumin binding and "hitchhiking" can be leveraged to achieve preferential delivery to and gene silencing activity within arthritic joints. Chemical stabilization of the lipophilic siRNA enables intravenous siRNA delivery without lipid or polymer encapsulation. Using siRNA sequences targeting MMP13, a key driver of arthritis-related inflammation, albumin hitchhiking siRNA diminished MMP13, inflammation, and manifestations of osteoarthritis and rheumatoid arthritis at molecular, histological, and clinical levels, consistently outperforming clinical standards of care and small molecule MMP antagonists.
Collapse
|
20
|
Fletcher RB, Stokes LD, Kelly IB, Henderson KM, Vallecillo-Viejo IC, Colazo JM, Wong BV, Yu F, d'Arcy R, Struthers MN, Evans BC, Ayers J, Castanon M, Weirich MJ, Reilly SK, Patel SS, Ivanova YI, Silvera Batista CA, Weiss SM, Gersbach CA, Brunger JM, Duvall CL. Nonviral In Vivo Delivery of CRISPR-Cas9 Using Protein-Agnostic, High-Loading Porous Silicon and Polymer Nanoparticles. ACS NANO 2023; 17:16412-16431. [PMID: 37582231 PMCID: PMC11129837 DOI: 10.1021/acsnano.2c12261] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/17/2023]
Abstract
The complexity of CRISPR machinery is a challenge to its application for nonviral in vivo therapeutic gene editing. Here, we demonstrate that proteins, regardless of size or charge, efficiently load into porous silicon nanoparticles (PSiNPs). Optimizing the loading strategy yields formulations that are ultrahigh loading─>40% cargo by volume─and highly active. Further tuning of a polymeric coating on the loaded PSiNPs yields nanocomposites that achieve colloidal stability under cryopreservation, endosome escape, and gene editing efficiencies twice that of the commercial standard Lipofectamine CRISPRMAX. In a mouse model of arthritis, PSiNPs edit cells in both the cartilage and synovium of knee joints, and achieve 60% reduction in expression of the therapeutically relevant MMP13 gene. Administered intramuscularly, they are active over a broad dose range, with the highest tested dose yielding nearly 100% muscle fiber editing at the injection site. The nanocomposite PSiNPs are also amenable to systemic delivery. Administered intravenously in a model that mimics muscular dystrophy, they edit sites of inflamed muscle. Collectively, the results demonstrate that the PSiNP nanocomposites are a versatile system that can achieve high loading of diverse cargoes and can be applied for gene editing in both local and systemic delivery applications.
Collapse
Affiliation(s)
- R Brock Fletcher
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235-1631, United States
| | - Larry D Stokes
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235-1631, United States
| | - Isom B Kelly
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235-1631, United States
| | - Katelyn M Henderson
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235-1631, United States
| | - Isabel C Vallecillo-Viejo
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235-1631, United States
| | - Juan M Colazo
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235-1631, United States
| | - Benjamin V Wong
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235-1631, United States
| | - Fang Yu
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235-1631, United States
| | - Richard d'Arcy
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235-1631, United States
| | - Morgan N Struthers
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235-1631, United States
| | - Brian C Evans
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235-1631, United States
| | - Jacob Ayers
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235-1631, United States
| | - Matthew Castanon
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235-1631, United States
| | - Michael J Weirich
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235-1631, United States
| | - Sarah K Reilly
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235-1631, United States
| | - Shrusti S Patel
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235-1631, United States
| | - Yoanna I Ivanova
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235-1631, United States
| | - Carlos A Silvera Batista
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, Tennessee 37235-1631, United States
| | - Sharon M Weiss
- Department of Electrical Engineering, Vanderbilt University, Nashville, Tennessee 37235-1631, United States
| | - Charles A Gersbach
- Department of Biomedical Engineering, Duke University, Durham, North Carolina 27708, United States
| | - Jonathan M Brunger
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235-1631, United States
| | - Craig L Duvall
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235-1631, United States
| |
Collapse
|
21
|
Chen H, Chen F, Hu F, Li Y, Zhang M, Zhou Q, Ding T, Tulufu N, Ye T, Wang F, Guo L. MicroRNA-224-5p nanoparticles balance homeostasis via inhibiting cartilage degeneration and synovial inflammation for synergistic alleviation of osteoarthritis. Acta Biomater 2023:S1742-7061(23)00336-7. [PMID: 37330028 DOI: 10.1016/j.actbio.2023.06.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 05/18/2023] [Accepted: 06/12/2023] [Indexed: 06/19/2023]
Abstract
MicroRNAs play a crucial role in regulating cartilage extracellular matrix (ECM) metabolism and are being explored as potential therapeutic targets for osteoarthritis (OA). The present study indicated that microRNA-224-5p (miR-224-5p) could balance the homeostasis of OA via regulating cartilage degradation and synovium inflammatory simultaneously. Multifunctional polyamidoamine dendrimer with amino acids used as efficient vector to deliver miR-224-5p. The vector could condense miR-224-5p into transfected nanoparticles, which showed higher cellular uptake and transfection efficiency compared to lipofectamine 3000, and also protected miR-224-5p from RNase degradation. After treatment with the nanoparticles, the chondrocytes showed an increase in autophagy rate and ECM anabolic components, as evidenced by the upregulation of autophagy-related proteins and OA-related anabolic mediators. This led to a corresponding inhibition of cell apoptosis and ECM catabolic proteases, ultimately resulting in the alleviation of ECM degradation. In addition, miR-224-5p also inhibited human umbilical vein endothelial cells angiogenesis and fibroblast-like synoviocytes inflammatory hyperplasia. Integrating the above synergistic effects of miR-224-5p in regulating homeostasis, intra-articular injection of nanoparticles performed outstanding therapeutic effect by reducing articular space width narrowing, osteophyte formation, subchondral bone sclerosis and inhibiting synovial hypertrophy and proliferation in the established mouse OA model. The present study provides a new therapy target and an efficient intra-articular delivery method for improving OA therapy. STATEMENT OF SIGNIFICANCE: Osteoarthritis (OA) is the most prevalent joint disease worldwide. Gene therapy, which involves delivering microRNAs, has the potential to treat OA. In this study, we demonstrated that miR-224-5p can simultaneously regulate cartilage degradation and synovium inflammation, thereby restoring homeostasis in OA gene therapy. Moreover, compared to traditional transfection reagents such as lipofectamine 3000, G5-AHP showed better efficacy in both microRNA transfection and protection against degradation due to its specific surface structure. In summary, G5-AHP/miR-224-5p was developed to meet the clinical needs of OA patients and the high requirement of gene transfection efficiency, providing a promising paradigm for the future application and development of gene therapy.
Collapse
Affiliation(s)
- Haoyi Chen
- Shanghai Key Laboratory for Bone and Joint Diseases, Shanghai Institute of Orthopaedics and Traumatology, department of orthopedics, Shanghai Ruijin Hospital, Shanghai Jiaotong University School of Medicine, China.
| | - Fangjing Chen
- Department of Orthopedic Surgery, Shanghai Tenth People's Hospital, Tongji University, Shanghai, China.
| | - Fangqiong Hu
- Shanghai Key Laboratory for Bone and Joint Diseases, Shanghai Institute of Orthopaedics and Traumatology, department of orthopedics, Shanghai Ruijin Hospital, Shanghai Jiaotong University School of Medicine, China
| | - Yifan Li
- Shanghai Key Laboratory for Bone and Joint Diseases, Shanghai Institute of Orthopaedics and Traumatology, department of orthopedics, Shanghai Ruijin Hospital, Shanghai Jiaotong University School of Medicine, China
| | - Meixing Zhang
- Shanghai Key Laboratory for Bone and Joint Diseases, Shanghai Institute of Orthopaedics and Traumatology, department of orthopedics, Shanghai Ruijin Hospital, Shanghai Jiaotong University School of Medicine, China
| | - Qi Zhou
- Shanghai Key Laboratory for Bone and Joint Diseases, Shanghai Institute of Orthopaedics and Traumatology, department of orthopedics, Shanghai Ruijin Hospital, Shanghai Jiaotong University School of Medicine, China
| | - Tao Ding
- Shanghai Key Laboratory for Bone and Joint Diseases, Shanghai Institute of Orthopaedics and Traumatology, department of orthopedics, Shanghai Ruijin Hospital, Shanghai Jiaotong University School of Medicine, China
| | - Nijiati Tulufu
- Shanghai Key Laboratory for Bone and Joint Diseases, Shanghai Institute of Orthopaedics and Traumatology, department of orthopedics, Shanghai Ruijin Hospital, Shanghai Jiaotong University School of Medicine, China
| | - Tianwen Ye
- Department of Orthopaedic Surgery, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China..
| | - Fei Wang
- Shanghai Key Laboratory for Bone and Joint Diseases, Shanghai Institute of Orthopaedics and Traumatology, department of orthopedics, Shanghai Ruijin Hospital, Shanghai Jiaotong University School of Medicine, China.
| | - Lei Guo
- Shanghai Key Laboratory for Bone and Joint Diseases, Shanghai Institute of Orthopaedics and Traumatology, department of orthopedics, Shanghai Ruijin Hospital, Shanghai Jiaotong University School of Medicine, China.
| |
Collapse
|
22
|
Zhou D, Zhou F, Sheng S, Wei Y, Chen X, Su J. Intra-articular nanodrug delivery strategies for treating osteoarthritis. Drug Discov Today 2023; 28:103482. [PMID: 36584875 DOI: 10.1016/j.drudis.2022.103482] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 12/09/2022] [Accepted: 12/22/2022] [Indexed: 12/28/2022]
Abstract
Osteoarthritis (OA) is characterized by progressive cartilage degeneration. Pharmaceutical intervention remains a main treatment approach. However, drug delivery via intra-articular administration (IA) can be restricted by rapid clearance, the dense and highly negatively charged extracellular matrix (ECM) of cartilage, and uneven distribution of diseased chondrocytes. Nanodrug delivery systems, such as liposomes, micelles, and nanoparticles (NPs), have shown great potential to prolong intra-articular residence, penetrate the ECM, and achieve diseased chondrocyte-specific delivery. In this review, we discuss the challenges associated with intra-articular drug delivery in OA and the nanodrug delivery strategies developed to overcome these challenges. It is anticipated that these nanodrug delivery strategies will advance IA of drugs into broader applications in OA treatment.
Collapse
Affiliation(s)
- Dongyang Zhou
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China; Institute of Advanced Interdisciplinary Materials Science, Shanghai University, Shanghai 200444, China; College of Medicine, Shanghai University, Shanghai 200444, China; Organoid Research Center, Shanghai University, Shanghai 200444, China
| | - Fengjin Zhou
- Department of Orthopaedics, Honghui Hospital, Xi'an Jiao Tong University, Xi'an 710000, China
| | - Shihao Sheng
- Department of Trauma Orthopedics, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Yan Wei
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China; Organoid Research Center, Shanghai University, Shanghai 200444, China.
| | - Xiao Chen
- Department of Trauma Orthopedics, Changhai Hospital, Naval Medical University, Shanghai 200433, China.
| | - Jiacan Su
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China; Institute of Advanced Interdisciplinary Materials Science, Shanghai University, Shanghai 200444, China; Organoid Research Center, Shanghai University, Shanghai 200444, China; Department of Trauma Orthopedics, Changhai Hospital, Naval Medical University, Shanghai 200433, China.
| |
Collapse
|
23
|
Tang X, Wang Z, Xie Y, Liu Y, Yang K, Li T, Shen H, Zhao M, Jin J, Xiao H, Liu H, Gu N. Radiation-Triggered Selenium-Engineered Mesoporous Silica Nanocapsules for RNAi Therapy in Radiotherapy-Resistant Glioblastoma. ACS NANO 2023; 17:4062-4076. [PMID: 36759969 DOI: 10.1021/acsnano.3c00269] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Radiotherapy-resistant glioblastoma (rrGBM) remains a significant clinical challenge because of high infiltrative growth characterized by activation of antiapoptotic signal transduction. Herein, we describe an efficiently biodegradable selenium-engineered mesoporous silica nanocapsule, initiated by high-energy X-ray irradiation and employed for at-site RNA interference (RNAi) to inhibit rrGBM invasion and achieve maximum therapeutic benefit. Our radiation-triggered RNAi nanocapsule showed high physiological stability, good blood-brain barrier transcytosis, and potent rrGBM accumulation. An intratumoral RNAi nanocapsule permitted low-dose X-ray radiation-triggered dissociation for cofilin-1 knockdown, inhibiting rrGBM infiltration. More importantly, tumor suppression was further amplified by electron-affinity aminoimidazole products converted from metronidazole polymers under X-ray radiation-exacerbated hypoxia, which sensitized cell apoptosis to ionizing radiation by fixing reactive oxygen species-induced DNA lesions. In vivo experiments confirmed that our RNAi nanocapsule reduced tumor growth and invasion, prolonging survival in an orthotopic rrGBM model. Generally, we present a promising radiosensitizer that would effectively improve rrGBM-patient outcomes with low-dose X-ray irradiation.
Collapse
Affiliation(s)
- Xianglong Tang
- Department of Neuro-Psychiatric Institute, The Affiliated Brain Hospital with Nanjing Medical University, Nanjing 210029, China
- Department of Neurosurgery, The Affiliated Brain Hospital with Nanjing Medical University, Fourth Clinical College of Nanjing Medical University, Nanjing 210029, China
| | - Zhen Wang
- Department of Neurosurgery, The Affiliated Brain Hospital with Nanjing Medical University, Fourth Clinical College of Nanjing Medical University, Nanjing 210029, China
| | - Yandong Xie
- Department of Neurosurgery, The Affiliated Brain Hospital with Nanjing Medical University, Fourth Clinical College of Nanjing Medical University, Nanjing 210029, China
| | - Yuyang Liu
- Department of Neurosurgery, The Affiliated Brain Hospital with Nanjing Medical University, Fourth Clinical College of Nanjing Medical University, Nanjing 210029, China
| | - Kun Yang
- Department of Neurosurgery, The Affiliated Brain Hospital with Nanjing Medical University, Fourth Clinical College of Nanjing Medical University, Nanjing 210029, China
| | - Taiping Li
- Department of Neuro-Psychiatric Institute, The Affiliated Brain Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Hong Shen
- Department of Neuro-Psychiatric Institute, The Affiliated Brain Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Mengjie Zhao
- Department of Neuro-Psychiatric Institute, The Affiliated Brain Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Juan Jin
- School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing 211166, China
| | - Hong Xiao
- Department of Neuro-Psychiatric Institute, The Affiliated Brain Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Hongyi Liu
- Department of Neurosurgery, The Affiliated Brain Hospital with Nanjing Medical University, Fourth Clinical College of Nanjing Medical University, Nanjing 210029, China
| | - Ning Gu
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences and Medical Engineering, Southeast University, Nanjing 210096, China
- School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing 211166, China
| |
Collapse
|
24
|
Ozkan H, Di Francesco M, Willcockson H, Valdés-Fernández J, Di Francesco V, Granero-Moltó F, Prósper F, Decuzzi P, Longobardi L. Sustained inhibition of CC-chemokine receptor-2 via intraarticular deposition of polymeric microplates in post-traumatic osteoarthritis. Drug Deliv Transl Res 2023; 13:689-701. [PMID: 36109442 PMCID: PMC9794532 DOI: 10.1007/s13346-022-01235-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/31/2022] [Indexed: 12/31/2022]
Abstract
Posttraumatic osteoarthritis (PTOA) is mostly treated via corticosteroid administration, and total joint arthroplasty continues to be the sole effective intervention in severe conditions. To assess the therapeutic potential of CCR2 targeting in PTOA, we used biodegradable microplates (µPLs) to achieve a slow and sustained intraarticular release of the CCR2 inhibitor RS504393 into injured knees and followed joint damage during disease progression. RS504393-loaded µPLs (RS-µPLs) were fabricated via a template-replica molding technique. A mixture of poly(lactic-co-glycolic acid) (PLGA) and RS504393 was deposited into 20 × 10 μm (length × height) wells in a polyvinyl alcohol (PVA) square-patterned template. After physicochemical and toxicological characterizations, the RS504393 release profile from µPL was assessed in PBS buffer. C57BL/6 J male mice were subjected to destabilization of the medial meniscus (DMM)/sham surgery, and RS-µPLs (1 mg/kg) were administered intraarticularly 1 week postsurgery. Administrations were repeated at 4 and 7 weeks post-DMM. Drug free-µPLs (DF-µPLs) and saline injections were performed as controls. Mice were euthanized at 4 and 10 weeks post-DMM, corresponding to the early and severe PTOA stages, respectively. Knees were evaluated for cartilage structure score (ACS, H&E), matrix loss (safranin O score), osteophyte formation and maturation from cartilage to bone (cartilage quantification), and subchondral plate thickness. The RS-µPL architecture ensured the sustained release of CCR2 inhibitors over several weeks, with ~ 20% of RS504393 still available at 21 days. This prolonged release improved cartilage structure and reduced bone damage and synovial hyperplasia at both PTOA stages. Extracellular matrix loss was also attenuated, although with less efficacy. The results indicate that local sustained delivery is needed to optimize CCR2-targeted therapies.
Collapse
Affiliation(s)
- Huseyin Ozkan
- Division of Rheumatology, Allergy and Immunology and the Thurston Arthritis Research Center, University of North Carolina-, Chapel Hill, 3300 Thurston Bowels Bldg, Campus, Box 7280, Chapel Hill, NC 27599 USA
| | - Martina Di Francesco
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano Di Tecnologia, Genoa, Italy
| | - Helen Willcockson
- Division of Rheumatology, Allergy and Immunology and the Thurston Arthritis Research Center, University of North Carolina-, Chapel Hill, 3300 Thurston Bowels Bldg, Campus, Box 7280, Chapel Hill, NC 27599 USA
| | - José Valdés-Fernández
- Division of Rheumatology, Allergy and Immunology and the Thurston Arthritis Research Center, University of North Carolina-, Chapel Hill, 3300 Thurston Bowels Bldg, Campus, Box 7280, Chapel Hill, NC 27599 USA ,Cell Therapy Area, Clínica Universidad de Navarra, Pamplona, Spain
| | - Valentina Di Francesco
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano Di Tecnologia, Genoa, Italy
| | - Froilán Granero-Moltó
- Cell Therapy Area, Clínica Universidad de Navarra, Pamplona, Spain ,Department of Orthopedic Surgery and Traumatology, Clínica Universidad de Navarra, Pamplona, Spain ,Program of Regenerative Medicine, Center for Applied Medical Research (CIMA), Universidad de Navarra, Pamplona, Spain ,Instituto de Investigacion Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Felipe Prósper
- Cell Therapy Area, Clínica Universidad de Navarra, Pamplona, Spain ,Program of Regenerative Medicine, Center for Applied Medical Research (CIMA), Universidad de Navarra, Pamplona, Spain ,Instituto de Investigacion Sanitaria de Navarra (IdiSNA), Pamplona, Spain ,Department of Hematology, Clínica Universidad de Navarra, Pamplona, Spain ,Program of Hemato-Oncology, Center for Applied Medical Research (CIMA), Universidad de Navarra, Pamplona, Spain
| | - Paolo Decuzzi
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano Di Tecnologia, Genoa, Italy
| | - Lara Longobardi
- Division of Rheumatology, Allergy and Immunology and the Thurston Arthritis Research Center, University of North Carolina-, Chapel Hill, 3300 Thurston Bowels Bldg, Campus, Box 7280, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
25
|
Polymeric Nanoparticles for Drug Delivery in Osteoarthritis. Pharmaceutics 2022; 14:pharmaceutics14122639. [PMID: 36559133 PMCID: PMC9788411 DOI: 10.3390/pharmaceutics14122639] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 11/25/2022] [Accepted: 11/26/2022] [Indexed: 12/02/2022] Open
Abstract
Osteoarthritis (OA) is a degenerative musculoskeletal disorder affecting the whole synovial joint and globally impacts more than one in five individuals aged 40 and over, representing a huge socioeconomic burden. Drug penetration into and retention within the joints are major challenges in the development of regenerative therapies for OA. During the recent years, polymeric nanoparticles (PNPs) have emerged as promising drug carrier candidates due to their biodegradable properties, nanoscale structure, functional versatility, and reproducible manufacturing, which makes them particularly attractive for cartilage penetration and joint retention. In this review, we discuss the current development state of natural and synthetic PNPs for drug delivery and OA treatment. Evidence from in vitro and pre-clinical in vivo studies is used to show how disease pathology and key cellular pathways of joint inflammation are modulated by these nanoparticle-based therapies. Furthermore, we compare the biodegradability and surface modification of these nanocarriers in relation to the drug release profile and tissue targeting. Finally, the main challenges for nanoparticle delivery to the cartilage are discussed, as a function of disease state and physicochemical properties of PNPs such as size and surface charge.
Collapse
|
26
|
Wang Y, Liu L, Le Z, Tay A. Analysis of Nanomedicine Efficacy for Osteoarthritis. ADVANCED NANOBIOMED RESEARCH 2022. [DOI: 10.1002/anbr.202200085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Affiliation(s)
- Yuwen Wang
- Department of Biomedical Engineering National University of Singapore Singapore 117583 Singapore
| | - Ling Liu
- Institute of Health Innovation and Technology National University of Singapore Singapore 117599 Singapore
| | - Zhicheng Le
- Department of Biomedical Engineering National University of Singapore Singapore 117583 Singapore
| | - Andy Tay
- Department of Biomedical Engineering National University of Singapore Singapore 117583 Singapore
- Institute of Health Innovation and Technology National University of Singapore Singapore 117599 Singapore
- Tissue Engineering Programme National University of Singapore Singapore 117510 Singapore
| |
Collapse
|
27
|
Huang X, Pan X, Xiong X, Zhao Z, Cen X. Drug delivery systems for treatment of temporomandibular joint osteoarthritis. Front Pharmacol 2022; 13:1054703. [PMID: 36419625 PMCID: PMC9676453 DOI: 10.3389/fphar.2022.1054703] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 10/26/2022] [Indexed: 01/27/2025] Open
Abstract
The number of people suffering from temporomandibular joint osteoarthritis (TMJOA) has been increasing. TMJOA cause joint noise, pain on TMJ and/or masticatory muscles, and restricted mandibular movement, which disturb eating, laughing and conversation, and impose serious lifestyle impediments. Chondrocyte apoptosis, extracellular matrix degradation, synovitis, and subchondral bone remodeling are the main pathological features of TMJOA. Various drug delivery systems are developed to controlled release at specific activation sites with high bioactivity and inhibit rapid dilution to enable long-term therapeutic response, which present great potential for the treatment of TMJOA. This review focuses on recently developed drug delivery systems by different administration in the TMJOA treatment, and summarizes their effects, duration, safety, and limitations, which would pave the way for development of TMJOA therapy.
Collapse
Affiliation(s)
- Xinqi Huang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xuefeng Pan
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xiner Xiong
- Hospital of Stomatology, Zunyi Medical University, Zunyi, China
| | - Zhihe Zhao
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xiao Cen
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
28
|
Xiao L, Cui J, Sun Z, Liu Y, Zheng J, Dong Y. Therapeutic potential of nanotechnology-based approaches in osteoarthritis. Front Pharmacol 2022; 13:920824. [PMID: 36003519 PMCID: PMC9394598 DOI: 10.3389/fphar.2022.920824] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 07/01/2022] [Indexed: 11/30/2022] Open
Abstract
Osteoarthritis (OA) is a multifactorial disease that affects the entire joint, often resulting in severe pain, disability, psychological distress, and a lower quality of life. Patient self-management is emphasized in OA clinical recommendations. Currently, the clinical treatment of OA mainly focuses on pain relief and the improvement of joint function, with few options for regenerating degenerative cartilage or slowing the progression of OA. Therefore, we first reviewed the current treatment of OA, and then summarized the research advances of nanotechnology in OA treatment, including nano drug delivery systems for small molecule drugs, nucleic acids and proteins, nano-scaffolds for cartilage regeneration, and nanoparticle lubricants. Finally, we discussed the opportunities and potential challenges of nanotechnology in OA treatment.
Collapse
Affiliation(s)
- Likang Xiao
- Department of Orthopedics, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Henan University People’s Hospital, Zhengzhou, China
| | - Jiarui Cui
- School of Rehabilitation and Health Preservation, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhuang Sun
- Department of Orthopedics, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Henan University People’s Hospital, Zhengzhou, China
| | - Yunke Liu
- Department of Orthopedics, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Henan University People’s Hospital, Zhengzhou, China
| | - Jia Zheng
- Department of Orthopedics, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Henan University People’s Hospital, Zhengzhou, China
- *Correspondence: Jia Zheng, ; Yonghui Dong,
| | - Yonghui Dong
- Department of Orthopedics, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Henan University People’s Hospital, Zhengzhou, China
- *Correspondence: Jia Zheng, ; Yonghui Dong,
| |
Collapse
|
29
|
Wang C, Zhang J, Li B, Zuo J, Li Y, Sun Y, Wang F, Liu K, Li J. High-Efficiency Treatment for Osteoarthritis via Self-Assembled Dual-Functionalized Nanobiologics. ACS Biomater Sci Eng 2022; 8:3320-3328. [PMID: 35801691 DOI: 10.1021/acsbiomaterials.2c00332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Osteoarthritis (OA) is a progressive joint disease that has a complex pathogenesis and lacks effective drugs. OA develops with cartilage degeneration and inflammation, thus synthesizing a drug with both anti-inflammatory properties and cartilage-repair capacity provides a promising treatment strategy. Therefore, in this study, we report self-assembled nanobiologics composed of an engineered recombinant IL-1 receptor antagonist (IL-1Ra) chimeric protein with chondroitin sulfate (CS). The nanobiologics, termed ICN, exhibit extraordinary biocompatibility, low immunogenicity, and good bioefficacy. Furthermore, our study revealed that ICN significantly reduced cartilage degradation, inhibited synovial inflammation, and suppressed osteophyte formation in OA rat models. The excellent therapeutic effects on OA can be attributed to the synergistic anti-inflammatory and cartilage-repair properties of ICN's constituents. Thus, our novel strategy offers insights into the development of drugs for OA treatment and research on nanobiomedicine, which can also be adapted for other diseases with similar pathologies.
Collapse
Affiliation(s)
- Chenyu Wang
- Department of Orthopedics, China-Japan Union Hospital of Jilin University, Changchun 130033, China.,State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Jinrui Zhang
- Department of Orthopedics, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Bo Li
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China.,Department of Chemistry, Tsinghua University, Beijing 100084, China
| | - Jianlin Zuo
- Department of Orthopedics, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Yuanxin Li
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Yao Sun
- Department of Chemistry, Tsinghua University, Beijing 100084, China
| | - Fan Wang
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Kai Liu
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China.,Department of Chemistry, Tsinghua University, Beijing 100084, China
| | - Jingjing Li
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| |
Collapse
|
30
|
Guo X, Lou J, Wang F, Fan D, Qin Z. Recent Advances in Nano-Therapeutic Strategies for Osteoarthritis. Front Pharmacol 2022; 13:924387. [PMID: 35800449 PMCID: PMC9253376 DOI: 10.3389/fphar.2022.924387] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 05/30/2022] [Indexed: 01/08/2023] Open
Abstract
Osteoarthritis (OA) is the most common type of arthritis and the leading cause of disability globally. It tends to occur in middle age or due to an injury or obesity. OA occurs with the onset of symptoms, including joint swelling, joint effusion, and limited movement at a late stage of the disease, which leads to teratogenesis and loss of joint function. During the pathogenesis of this degenerative joint lesion, several local inflammatory responses are activated, resulting in synovial proliferation and pannus formation that facilitates the destruction of the bone and the articular cartilage. The commonly used drugs for the clinical diagnosis and treatment of OA have limitations such as low bioavailability, short half-life, poor targeting, and high systemic toxicity. With the application of nanomaterials and intelligent nanomedicines, novel nanotherapeutic strategies have shown more specific targeting, prolonged half-life, refined bioavailability, and reduced systemic toxicity, compared to the existing medications. In this review, we summarized the recent advancements in new nanotherapeutic strategies for OA and provided suggestions for improving the treatment of OA.
Collapse
Affiliation(s)
- Xinjing Guo
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Jia Lou
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Fazhan Wang
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- *Correspondence: Daoyang Fan, ; Fazhan Wang, ; Zhihai Qin,
| | - Daoyang Fan
- Department of Orthopedic, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- *Correspondence: Daoyang Fan, ; Fazhan Wang, ; Zhihai Qin,
| | - Zhihai Qin
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
- *Correspondence: Daoyang Fan, ; Fazhan Wang, ; Zhihai Qin,
| |
Collapse
|
31
|
Di Francesco M, Fragassi A, Pannuzzo M, Ferreira M, Brahmachari S, Decuzzi P. Management of osteoarthritis: From drug molecules to nano/micromedicines. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2022; 14:e1780. [PMID: 35253405 PMCID: PMC9285805 DOI: 10.1002/wnan.1780] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 12/29/2021] [Accepted: 01/21/2022] [Indexed: 12/12/2022]
Abstract
With the change in lifestyle and aging of the population, osteoarthritis (OA) is emerging as a major medical burden globally. OA is a chronic inflammatory and degenerative disease initially manifesting with joint pain and eventually leading to permanent disability. To date, there are no drugs available for the definitive treatment of osteoarthritis and most therapies have been palliative in nature by alleviating symptoms rather than curing the disease. This coupled with the vague understanding of the early symptoms and methods of diagnosis so that the disease continues as a global problem and calls for concerted research efforts. A cascade of events regulates the onset and progression of osteoarthritis starting with the production of proinflammatory cytokines, including interleukin (IL)‐1β, IL‐6, tumor necrosis factor (TNF)‐α; catabolic enzymes, such as matrix metalloproteinases (MMPs)‐1, ‐3, and ‐13, culminating into cartilage breakdown, loss of lubrication, pain, and inability to load the joint. Although intra‐articular injections of small and macromolecules are often prescribed to alleviate symptoms, low residence times within the synovial cavity severely impair their efficacy. This review will briefly describe the factors dictating the onset and progression of the disease, present the current clinically approved methods for its treatment and diagnosis, and finally elaborate on the main challenges and opportunities for the application of nano/micromedicines in the treatment of osteoarthritis. Thus, future treatment regimens will benefit from simultaneous consideration of the mechanobiological, the inflammatory, and tissue degradation aspects of the disease. This article is categorized under:Nanotechnology Approaches to Biology > Nanoscale Systems in Biology Implantable Materials and Surgical Technologies > Nanotechnology in Tissue Repair and Replacement
Collapse
Affiliation(s)
- Martina Di Francesco
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Genoa, Italy
| | - Agnese Fragassi
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Genoa, Italy.,Department of Chemistry and Industrial Chemistry, University of Genova, Genoa, Italy
| | - Martina Pannuzzo
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Genoa, Italy
| | - Miguel Ferreira
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Genoa, Italy
| | - Sayanti Brahmachari
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Genoa, Italy
| | - Paolo Decuzzi
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Genoa, Italy
| |
Collapse
|
32
|
Huang H, Lou Z, Zheng S, Wu J, Yao Q, Chen R, Kou L, Chen D. Intra-articular drug delivery systems for osteoarthritis therapy: shifting from sustained release to enhancing penetration into cartilage. Drug Deliv 2022; 29:767-791. [PMID: 35261301 PMCID: PMC8920370 DOI: 10.1080/10717544.2022.2048130] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Osteoarthritis (OA) is a progressive chronic inflammation that leads to cartilage degeneration. OA Patients are commonly given pharmacological treatment, but the available treatments are not sufficiently effective. The development of sustained-release drug delivery systems (DDSs) for OA may be an attractive strategy to prevent rapid drug clearance and improve the half-life of a drug at the joint cavity. Such delivery systems will improve the therapeutic effects of anti-inflammatory effects in the joint cavity. Whereas, for disease-modifying OA drugs (DMOADs) which target chondrocytes or act on mesenchymal stem cells (MSCs), the cartilage-permeable DDSs are required to maximize their efficacy. This review provides an overview of joint structure in healthy and pathological conditions, introduces the advances of the sustained-release DDSs and the permeable DDSs, and discusses the rational design of the permeable DDSs for OA treatment. We hope that the ideas generated in this review will promote the development of effective OA drugs in the future.
Collapse
Affiliation(s)
- Huirong Huang
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zijian Lou
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,Department of Orthopedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Shimin Zheng
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jianing Wu
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Qing Yao
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Ruijie Chen
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Longfa Kou
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Daosen Chen
- Department of Orthopedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
33
|
Loughlin J. Translating osteoarthritis genetics research: challenging times ahead. Trends Mol Med 2022; 28:176-182. [PMID: 35033441 DOI: 10.1016/j.molmed.2021.12.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 12/21/2021] [Accepted: 12/21/2021] [Indexed: 12/15/2022]
Abstract
The ultimate goal of molecular genetic studies of human diseases is to translate the discoveries for patient benefit. For diseases that lack licensed disease-modifying therapeutics, such as osteoarthritis (OA), the need is acute. OA is polygenic and affects older individuals, with a recent genome-wide study of over 800 000 individuals adding 52 novel association signals to those already reported on for this common arthritis. Many of the predicted effector genes of these signals encode proteins that are targets of drugs for other indications, highlighting repurposing opportunities. Here, the potential for OA genetic data to translate is discussed, including whether the developmental origin of OA will limit the application of genetic risk data for disease-modification purposes.
Collapse
Affiliation(s)
- John Loughlin
- Newcastle University, Biosciences Institute, International Centre for Life, Newcastle upon Tyne, NE1 3BZ, UK.
| |
Collapse
|