1
|
Mitsou E, Klein J. Liposome-Based Interventions in Knee Osteoarthritis. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2410060. [PMID: 40143645 PMCID: PMC12036560 DOI: 10.1002/smll.202410060] [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: 01/21/2025] [Indexed: 03/28/2025]
Abstract
Osteoarthritis (OA) is the most common degenerative disease of the joints, causing significant disability and socio-economic burden in the aging population. Simultaneously, however, it is a common occurrence in younger individuals, initiated by joint injuries or obesity alongside other factors. Intravenous and oral pharmaceutical OA management have both been associated with systemic adverse effects, thereby resulting in a growing interest in intra-articular (IA) treatment. IA-administered drugs circumvent the requirement for high dosage, offering immediate access to the site of interest while minimizing any unfavorable effects. Nonetheless, IA-injected drugs, administered in their free form, present low retention time in the knee joint raising the need for multiple injection dosage regimens, while their capability to target the cartilage or specific cell populations is limited. Liposomes, due to their unique characteristics and tunable nature, have proven to be excellent candidates for the management of knee OA. This review explores the last decade's research on the efficacy of various IA liposomal formulations, investigating their multifaceted properties as pharmaceutical carriers, lubricating agents, and a basis for combinatorial approaches paving the way to novel treatment solutions for OA.
Collapse
Affiliation(s)
- Evgenia Mitsou
- Department of Molecular Chemistry and Materials ScienceWeizmann Institute of ScienceHertzl Street 234Rehovot7610001Israel
- Present address:
Institute of Chemical BiologyNational Hellenic Research Foundation48, Vassileos Constantinou Ave.Athens11635Greece
| | - Jacob Klein
- Department of Molecular Chemistry and Materials ScienceWeizmann Institute of ScienceHertzl Street 234Rehovot7610001Israel
| |
Collapse
|
2
|
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
|
3
|
Wang Y, Guo X, Zeng H, Chen Z, Yue Y, Jin X. M2 Macrophage Polarization and Tissue Remodeling in Autologous Fat Grafting for Diabetic Skin Defects. J Craniofac Surg 2024; 35:2194-2199. [PMID: 38838369 DOI: 10.1097/scs.0000000000010386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 05/08/2024] [Indexed: 06/07/2024] Open
Abstract
Autologous adipose tissue was recognized as a promising therapeutic option for soft tissue defects owing to its regenerative potential and ability to facilitate tissue reconstruction. However, the mechanisms by which autologous fat grafting (AFG) promotes healing remain unclear, hindering its potential applications. This study aimed to investigate the distribution and phenotypic transition of infiltrating macrophages in transplanted adipose tissue, as well as their correlation with diabetic skin defect remodeling. Streptozotocin-induced diabetic rats with full-thickness dorsal skin defects were included in this study. The transplanted adipose tissue at the skin defects was collected and analyzed using flow cytometry to determine macrophage proportion and phenotype. The healing of skin defects was evaluated, and treatment was continued until day 14 as the designated endpoint of healing, followed by histopathologic examinations. Immunostaining with CD31 and lymphatic vessel endothelial receptor-1 was performed on wound tissues to analyze angiogenesis and lymphangiogenesis, respectively. Western blot and quantitative polymerase chain reaction analyses were used to assess the expression of the representative genes involved in the healing process. The results showed early polarization of M2 macrophages in the transplanted adipose tissue, concomitant with the upregulation of growth factors and downregulation of inflammatory factors. In vivo experiments revealed that AFG significantly promoted macrophage infiltration and M2 transformation in diabetic skin defects compared to the control groups, thereby promoting tissue extracellular matrix remodeling and lymphatic and vascular regeneration. However, the beneficial effects of AFG were inhibited by macrophage depletion. This study further demonstrated the potential of AFG for treating diabetic skin defects.
Collapse
Affiliation(s)
- Yu Wang
- Department of Craniomaxillofacial Surgery, Plastic Surgery Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | | | | | | | | | | |
Collapse
|
4
|
Chu B, Chen D, Ma S, Yang Y, Shang F, Lv W, Li Y. Novel poly(lactic-co-glycolic acid) nanoliposome-encapsulated diclofenac sodium and celecoxib enable long-lasting synergistic treatment of osteoarthritis. J Biomater Appl 2024; 39:221-234. [PMID: 38820587 DOI: 10.1177/08853282241258311] [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: 06/02/2024]
Abstract
BACKGROUND Diclofenac sodium (DS) and celecoxib (CEL) are primary anti-inflammatory agents used in the treatment of osteoarthritis (OA). Formulating these drugs into extended-release versions can effectively address the issue of multiple daily doses. In this study, we designed and synthesized a novel poly(lactic-co-glycolic acid) (PLGA) nanoliposome as a dual-drug delivery sustained-release formulation (PPLs-DS-CEL) to achieve long-lasting synergistic treatment of OA with both DS and CEL. METHODS PPLs-DS-CEL was synthesized by the reverse evaporation method and evaluated for its physicochemical properties, encapsulation efficiency, drug release kinetics and biological properties. A rat OA model was established to assess the therapeutic efficacy and biosafety of PPLs-DS-CEL. RESULTS The particle size of PPLs-DS-CEL was 218.36 ± 6.27 nm, with a potential of 32.56 ± 3.28 mv, indicating a homogeneous vesicle size. The encapsulation of DS and CEL by PPLs-DS-CEL was 95.18 ± 4.43% and 93.63 ± 5.11%, with drug loading of 9.56 ± 0.32% and 9.68 ± 0.34%, respectively. PPLs-DS-CEL exhibited low cytotoxicity and hemolysis, and was able to achieve long-lasting synergistic analgesic and anti-inflammatory therapeutic effects in OA through slow release of DS and CEL, demonstrating good biosafety properties. CONCLUSION This study developed a novel sustained-release nanoliposomes formulation capable of co-loading two drugs for the long-acting synergistic treatment of OA. It offers a new and effective therapeutic strategy for OA treatment in the clinic settings and presents a promising approach for drug delivery systems.
Collapse
Affiliation(s)
- Bo Chu
- Orthopaedics, Wuxi Xishan People's Hospital, Wuxi, China
| | - Dagui Chen
- Institute of Translational Medicine, Shanghai University, Shanghai, China
| | - Senlin Ma
- Department of Emergency Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Yong Yang
- Orthopaedics, Wuxi Xishan People's Hospital, Wuxi, China
| | - Fusheng Shang
- Institute of Translational Medicine, Shanghai University, Shanghai, China
| | - Wei Lv
- Orthopaedics, Wuxi Xishan People's Hospital, Wuxi, China
| | - Yinghua Li
- Institute of Translational Medicine, Shanghai University, Shanghai, China
| |
Collapse
|
5
|
Zhou J, Ning E, Lu L, Zhang H, Yang X, Hao Y. Effectiveness of low-intensity pulsed ultrasound on osteoarthritis: molecular mechanism and tissue engineering. Front Med (Lausanne) 2024; 11:1292473. [PMID: 38695024 PMCID: PMC11061361 DOI: 10.3389/fmed.2024.1292473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 04/05/2024] [Indexed: 05/04/2024] Open
Abstract
Osteoarthritis (OA) is distinguished by pathological alterations in the synovial membrane, articular cartilage, and subchondral bone, resulting in physical symptoms such as pain, deformity, and impaired mobility. Numerous research studies have validated the effectiveness of low-intensity pulsed ultrasound (LIPUS) in OA treatment. The periodic mechanical waves generated by LIPUS can mitigate cellular ischemia and hypoxia, induce vibration and collision, produce notable thermal and non-thermal effects, alter cellular metabolism, expedite tissue repair, improve nutrient delivery, and accelerate the healing process of damaged tissues. The efficacy and specific mechanism of LIPUS is currently under investigation. This review provides an overview of LIPUS's potential role in the treatment of OA, considering various perspectives such as the synovial membrane, cartilage, subchondral bone, and tissue engineering. It aims to facilitate interdisciplinary scientific research and further exploration of LIPUS as a complementary technique to existing methods or surgery. Ongoing research is focused on determining the optimal dosage, frequency, timing, and treatment strategy of LIPUS for OA. Additional research is required to clarify the precise mechanism of action and potential impacts on cellular, animal, and human systems prior to its integration into therapeutic applications.
Collapse
Affiliation(s)
- Jing Zhou
- Orthopedics and Sports Medicine Center, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
- Gusu School, Nanjing Medical University, Suzhou, China
| | - Eryu Ning
- Orthopedics and Sports Medicine Center, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
- Gusu School, Nanjing Medical University, Suzhou, China
| | - Lingfeng Lu
- Orthopedics and Sports Medicine Center, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
- Gusu School, Nanjing Medical University, Suzhou, China
| | - Huili Zhang
- Orthopedics and Sports Medicine Center, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
- Gusu School, Nanjing Medical University, Suzhou, China
| | - Xing Yang
- Orthopedics and Sports Medicine Center, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
- Gusu School, Nanjing Medical University, Suzhou, China
| | - Yuefeng Hao
- Orthopedics and Sports Medicine Center, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
- Gusu School, Nanjing Medical University, Suzhou, China
| |
Collapse
|
6
|
Wu YC, Su MC, Wu CS, Chen PY, Chen IF, Lin FH, Kuo SM. Ameliorative Effects of Cumin Extract-Encapsulated Chitosan Nanoparticles on Skeletal Muscle Atrophy and Grip Strength in Streptozotocin-Induced Diabetic Rats. Antioxidants (Basel) 2023; 13:6. [PMID: 38275626 PMCID: PMC10812640 DOI: 10.3390/antiox13010006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 12/13/2023] [Accepted: 12/15/2023] [Indexed: 01/27/2024] Open
Abstract
Skeletal muscle atrophy is a disorder characterized by reductions in muscle size and strength. Cumin extract (CE) possesses anti-inflammatory, antioxidant, and hypoglycemic properties. Its pharmaceutical applications are hindered by its low water solubility and by its cytotoxicity when administered at high doses. In this study, we have developed a simplified water distillation method using a rotary evaporator to isolate the active components in cumin seeds. The anti-inflammatory effects of CE and its potential to ameliorate skeletal muscle atrophy in rats with streptozotocin (STZ)-induced diabetes were evaluated. The half-maximal inhibitory concentration (IC50) of CE for cells was 80 μM. By encapsulating CE in chitosan nanoparticles (CECNs), an encapsulation efficacy of 87.1% was achieved with a slow release of 90% of CE after 24 h of culturing, resulting in CECNs with significantly reduced cytotoxicity (IC50, 1.2 mM). Both CE and CECNs significantly reduced the inflammatory response in interleukin (IL)-6 and IL-1β assays. STZ-induced diabetic rats exhibited sustained high blood glucose levels (>16.5 mmol/L), small and damaged pancreatic β islets, and skeletal muscle atrophy. CE and CECN treatments ameliorated skeletal muscle atrophy, recovered muscle fiber striated appearance, increased grip strength, and decreased IL-6 level. Furthermore, CE and CECNs led to a reduction of damage to the pancreas, restoring its morphological phenotype, increasing serum insulin levels, and lowering blood glucose levels in STZ-induced diabetic rats. Taken together, treatment with CECNs over a 6-week period yielded positive ameliorative effects in STZ-induced rats of muscle atrophy.
Collapse
Affiliation(s)
- Yu-Chiuan Wu
- Republic of China Military Academy, Kaohsiung 830208, Taiwan;
- Kaohsiung Armed Forces General Hospital, Kaohsiung 802301, Taiwan
| | - Min-Chien Su
- Department of Biomedical Engineering, I-Shou University, Kaohsiung 84001, Taiwan; (M.-C.S.); (P.-Y.C.); (I.-F.C.)
| | - Chun-Shien Wu
- Center of General Education, I-Shou University, Kaohsiung 84001, Taiwan;
| | - Pin-Yu Chen
- Department of Biomedical Engineering, I-Shou University, Kaohsiung 84001, Taiwan; (M.-C.S.); (P.-Y.C.); (I.-F.C.)
| | - I-Fen Chen
- Department of Biomedical Engineering, I-Shou University, Kaohsiung 84001, Taiwan; (M.-C.S.); (P.-Y.C.); (I.-F.C.)
| | - Feng-Huei Lin
- Department of Biomedical Engineering, National Taiwan University, Taipei 10617, Taiwan;
| | - Shyh-Ming Kuo
- Department of Biomedical Engineering, I-Shou University, Kaohsiung 84001, Taiwan; (M.-C.S.); (P.-Y.C.); (I.-F.C.)
| |
Collapse
|
7
|
Bordon G, Berenbaum F, Distler O, Luciani P. Harnessing the multifunctionality of lipid-based drug delivery systems for the local treatment of osteoarthritis. Biomed Pharmacother 2023; 168:115819. [PMID: 37939613 DOI: 10.1016/j.biopha.2023.115819] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 10/29/2023] [Accepted: 10/31/2023] [Indexed: 11/10/2023] Open
Abstract
Osteoarthritis (OA) is a widespread joint condition affecting millions globally, presenting a growing socioeconomic burden thus making the development of more effective therapeutic strategies crucial. This review emphasizes recent advancements in lipid-based drug delivery systems (DDSs) for intra-articular administration of OA therapeutics, encompassing non-steroidal anti-inflammatory drugs, corticosteroids, small molecule disease-modifying OA drugs, and RNA therapeutics. Liposomes, lipid nanoparticles, lipidic mesophases, extracellular vesicles and composite systems exhibit enhanced stability, targeted delivery, and extended joint retention, which contribute to improved therapeutic outcomes and minimized systemic drug exposure. Although active targeting strategies hold promise, further research is needed to assess their targeting efficiency in physiologically relevant conditions. Simultaneously, multifunctional DDSs capable of delivering combinations of distinct therapeutic classes offer synergistic effects and superior OA treatment outcomes. The development of such long-acting systems that resist rapid clearance from the joint space is crucial, where particle size and targeting capabilities emerge as vital factors. Additionally, combining cartilage lubrication properties with sustained drug delivery has demonstrated potential in animal models, meriting further investigation in human clinical trials. This review highlights the crucial need for direct, head-to-head comparisons of novel DDSs with standard treatments, particularly within the same drug class. These comparisons are essential in accurately evaluating their effectiveness, safety, and clinical applicability, and are set to significantly shape the future of OA therapy.
Collapse
Affiliation(s)
- Gregor Bordon
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Bern, Switzerland
| | - Francis Berenbaum
- Sorbonne University, INSERM CRSA, AP-HP Saint-Antoine Hospital, Paris, France
| | - Oliver Distler
- Department of Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Paola Luciani
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Bern, Switzerland.
| |
Collapse
|
8
|
Min K, Sahu A, Jeon SH, Tae G. Emerging drug delivery systems with traditional routes - A roadmap to chronic inflammatory diseases. Adv Drug Deliv Rev 2023; 203:115119. [PMID: 37898338 DOI: 10.1016/j.addr.2023.115119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 07/17/2023] [Accepted: 10/23/2023] [Indexed: 10/30/2023]
Abstract
Inflammation is prevalent and inevitable in daily life but can generally be accommodated by the immune systems. However, incapable self-healing and persistent inflammation can progress to chronic inflammation, leading to prevalent or fatal chronic diseases. This review comprehensively covers the topic of emerging drug delivery systems (DDSs) for the treatment of chronic inflammatory diseases (CIDs). First, we introduce the basic biology of the chronic inflammatory process and provide an overview of the main CIDs of the major organs. Next, up-to-date information on various DDSs and the associated strategies for ensuring targeted delivery and stimuli-responsiveness applied to CIDs are discussed extensively. The implementation of traditional routes of drug administration to maximize their therapeutic effects against CIDs is then summarized. Finally, perspectives on future DDSs against CIDs are presented.
Collapse
Affiliation(s)
- Kiyoon Min
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Republic of Korea
| | - Abhishek Sahu
- Department of Biotechnology, National Institute of Pharmaceutical Education & Research (NIPER), Hajipur, 844102, India
| | - Sae Hyun Jeon
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Republic of Korea
| | - Giyoong Tae
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Republic of Korea.
| |
Collapse
|
9
|
Jia W, Zhou Z, Zhan W. Musculoskeletal Biomaterials: Stimulated and Synergized with Low Intensity Pulsed Ultrasound. J Funct Biomater 2023; 14:504. [PMID: 37888169 PMCID: PMC10607075 DOI: 10.3390/jfb14100504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 09/10/2023] [Accepted: 09/21/2023] [Indexed: 10/28/2023] Open
Abstract
Clinical biophysical stimulating strategies, which have significant effects on improving the function of organs or treating diseases by causing the salutary response of body, have shown many advantages, such as non-invasiveness, few side effects, and controllable treatment process. As a critical technique for stimulation, the low intensity pulsed ultrasound (LIPUS) has been explored in regulating osteogenesis, which has presented great promise in bone repair by delivering a combined effect with biomaterials. This review summarizes the musculoskeletal biomaterials that can be synergized with LIPUS for enhanced biomedical application, including bone regeneration, spinal fusion, osteonecrosis/osteolysis, cartilage repair, and nerve regeneration. Different types of biomaterials are categorized for summary and evaluation. In each subtype, the verified biological mechanisms are listed in a table or graphs to prove how LIPUS was effective in improving musculoskeletal tissue regeneration. Meanwhile, the acoustic excitation parameters of LIPUS that were promising to be effective for further musculoskeletal tissue engineering are discussed, as well as their limitations and some perspectives for future research. Overall, coupled with biomimetic scaffolds and platforms, LIPUS may be a powerful therapeutic approach to accelerate musculoskeletal tissue repair and even in other regenerative medicine applications.
Collapse
Affiliation(s)
- Wanru Jia
- Department of Ultrasound, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China;
| | - Zifei Zhou
- Department of Orthopedics, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Weiwei Zhan
- Department of Ultrasound, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China;
| |
Collapse
|
10
|
Wang L, Ding Y, Tang Q, Niu X. Preparation, Properties and Therapeutic Effect of a TPL Nanoparticle Thermosensitive Gel for Intra-Articular Injection. Molecules 2023; 28:4659. [PMID: 37375214 DOI: 10.3390/molecules28124659] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 06/05/2023] [Accepted: 06/07/2023] [Indexed: 06/29/2023] Open
Abstract
Most injectable preparations for the articular cavity are solution-type preparations that are frequently administered because of rapid elimination. In this study, triptolide (TPL), an effective ingredient in the treatment of rheumatoid arthritis (RA), was prepared in the form of a nanoparticle thermosensitive gel (TPL-NS-Gel). The particle size distribution and gel structure were investigated by TEM, laser particle size analysis and laser capture microdissection. The effect of the nanoparticle carrier material PLGA on the phase transition temperature was investigated by 1H variable temperature NMR and DSC. The tissue distribution, pharmacokinetic behavior, four inflammatory factors and therapeutic effect were determined in a rat RA model. The results suggested that PLGA increased the gel phase transition temperature. The drug concentration of the TPL-NS-Gel group in joint tissues was higher than that in other tissues at different time points, and the retention time was longer than that of the TPL-NS group. After 24 days of administration, TPL-NS-Gel significantly improved the joint swelling and stiffness of the rat models, and the improvement degree was better than that of the TPL-NS group. TPL-NS-Gel significantly decreased the levels of hs-CRP, IL-1, IL-6 and TNF-α in serum and joint fluid. There was a significant difference between the TPL-NS-Gel and TPL-NS groups on Day 24 (p < 0.05). Pathological section results showed that inflammatory cell infiltration was lower in the TPL-NS-Gel group, and no other obvious histological changes were observed. Upon articular injection, the TPL-NS-Gel prolonged drug release, reduced the drug concentration outside the articular tissue and improved the therapeutic effect in a rat RA model. The TPL-NS-Gel can be used as a new type of sustained-release preparation for articular injection.
Collapse
Affiliation(s)
- Lijuan Wang
- Department of Pharmacy, Chongqing Engineering Research Center of Pharmaceutical Sciences, Chongqing Medical and Pharmaceutical College, Chongqing 401331, China
| | - Yongliang Ding
- Department of Pharmacy, Chongqing Engineering Research Center of Pharmaceutical Sciences, Chongqing Medical and Pharmaceutical College, Chongqing 401331, China
| | - Qian Tang
- Department of Pharmacy, Chongqing Engineering Research Center of Pharmaceutical Sciences, Chongqing Medical and Pharmaceutical College, Chongqing 401331, China
| | - Xiaodong Niu
- Department of Pharmacy, Chongqing Engineering Research Center of Pharmaceutical Sciences, Chongqing Medical and Pharmaceutical College, Chongqing 401331, China
| |
Collapse
|
11
|
Zhuang H, Ren X, Zhang Y, Jiang F, Zhou P. Trimethylamine-N-oxide sensitizes chondrocytes to mechanical loading through the upregulation of Piezo1. Food Chem Toxicol 2023; 175:113726. [PMID: 36925039 DOI: 10.1016/j.fct.2023.113726] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 02/23/2023] [Accepted: 03/13/2023] [Indexed: 03/17/2023]
Abstract
BACKGROUND Mechanical strain plays a crucial role in chondrocyte apoptosis and osteoarthritis (OA) disease progression through Piezo1. Trimethylamine-N-oxide (TMAO) is a diet-derived metabolite that correlates positively with multiple chronic diseases. Herein, we explored the potential role of TMAO in sensitizing chondrocytes to Piezo1-mediated mechanotransduction. METHODS The cytotoxicity of TMAO on chondrocytes was assayed. Piezo1 expression was measured after TMAO intervention. Pathological mechanical loading or Yoda1 (a specific Piezo1 channel activator) was administered in chondrocytes. The calcium levels and cytoskeleton in chondrocytes were observed by fluorescence microscopy. Flow cytometry, western blotting, and mitochondrial membrane potential assays were utilized to evaluate apoptosis. A rat OA model was constructed by anterior cruciate ligament transection. Hematoxylin-eosin staining, Safranin-O/Fast Green staining, immunochemistry, and TUNEL were applied to estimate OA severity. RESULTS TMAO intervention alone did not affect chondrocyte viability up to 600 μM. TMAO significantly increased Piezo1 expression and up-regulated intracellular calcium levels, further leading to cytoskeletal damage. Mechanical strain or Yoda1 treatment significantly induced chondrocyte apoptosis. Notably, TMAO intervention further aggravated chondrocyte apoptosis and cartilage destruction under pathological mechanical loading. CONCLUSION TMAO significantly up-regulated Piezo1 expression and sensitized chondrocytes to mechanical loading, which may be closely related to the pathogenesis of OA.
Collapse
Affiliation(s)
- Huangming Zhuang
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xunshan Ren
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yuelong Zhang
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Fuze Jiang
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Panghu Zhou
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, China.
| |
Collapse
|
12
|
Effects and mechanotransduction pathways of therapeutic ultrasound on healthy and osteoarthritic chondrocytes: a systematic review of in vitro studies. Osteoarthritis Cartilage 2023; 31:317-339. [PMID: 36481451 DOI: 10.1016/j.joca.2022.07.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 07/11/2022] [Accepted: 07/12/2022] [Indexed: 12/12/2022]
Abstract
OBJECTIVE To investigate the effects and mechanotransduction pathways of therapeutic ultrasound on chondrocytes. METHOD PubMed, EMBASE and Web of Science databases were searched up to 19th September 2021 to identify in vitro studies exploring ultrasound to stimulate chondrocytes for osteoarthritis (OA) treatment. Study characteristics, ultrasound parameters, in vitro setup, and mechanotransduction pathways were collected. Risk of bias was judged using the Risk of Bias Assessment for Non-randomized Studies (RoBANS) tool. RESULTS Thirty-one studies were included comprising healthy and OA chondrocytes and explants. Most studies had high risk of performance, detection and pseudoreplication bias due to lack of temperature control, setup calibration, inadequate semi-quantitatively analyzes and independent experiments. Ultrasound was applied to the culture plate via acoustic gel, water bath or culture media. Regardless of the setup used, ultrasound stimulated the cartilage production and suppressed its degradation, although the effect size was nonsignificant. Ultrasound inhibited p38, c-Jun N-terminal kinases (JNK) and factor nuclear kappa B (NFκB) pathways in OA chondrocytes to reduce apoptosis, inflammation and matrix degradation, while triggered phosphoinositide-3-kinase/akt (PI3K/Akt), extracellular signal-regulated kinase (ERK), p38 and JNK pathways in healthy chondrocytes to promote matrix synthesis. CONCLUSION The included studies suggest that ultrasound application induces therapeutic effects on chondrocytes. However, these results should be interpreted with caution because high risk of performance, detection and pseudoreplication bias were identified. Future studies should explore the application of ultrasound on human OA chondrocytes cultures to potentiate the applicability of ultrasound towards cartilage regeneration of knee with OA.
Collapse
|
13
|
Amirsaadat S, Amirazad H, Hashemihesar R, Zarghami N. An update on the effect of intra-articular intervention strategies using nanomaterials in osteoarthritis: Possible clinical application. Front Bioeng Biotechnol 2023; 11:1128856. [PMID: 36873347 PMCID: PMC9978162 DOI: 10.3389/fbioe.2023.1128856] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 02/06/2023] [Indexed: 02/18/2023] Open
Abstract
Osteoarthritis (OA) is the most common progressive condition affecting joints. It mainly affects the knees and hips as predominant weight-bearing joints. Knee osteoarthritis (KOA) accounts for a large proportion of osteoarthritis and presents numerous symptoms that impair quality of life, such as stiffness, pain, dysfunction, and even deformity. For more than two decades, intra-articular (IA) treatment options for managing knee osteoarthritis have included analgesics, hyaluronic acid (HA), corticosteroids, and some unproven alternative therapies. Before effective disease-modifying treatments for knee osteoarthritis, treatments are primarily symptomatic, mainly including intra-articular corticosteroids and hyaluronic acid, so these agents represent the most frequently used class of drugs for managing knee osteoarthritis. But research suggests other factors, such as the placebo effect, have an essential role in the effectiveness of these drugs. Several novel intra-articular therapies are currently in the clinical trial processes, such as biological therapies, gene and cell therapies. Besides, it has been shown that the development of novel drug nanocarriers and delivery systems could improve the effectiveness of therapeutic agents in osteoarthritis. This review discusses the various treatment methods and delivery systems for knee osteoarthritis and the new agents that have been introduced or are in development.
Collapse
Affiliation(s)
- Soumayeh Amirsaadat
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Halimeh Amirazad
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Medical Biotechnology, Faculty of Advanced Medical Science, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ramin Hashemihesar
- Department of Histology and Embryology, Faculty of Medicine, Altinbas University, Istanbul, Türkiye
| | - Nosratollah Zarghami
- Department of Medical Biochemistry, Faculty of Medicine, Istanbul Aydin University, Istanbul, Türkiye.,Department of Clinical Biochemistry and Laboratory Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
14
|
Lu J, Zhu D, Zhang X, Wang J, Cao H, Li L. The crucial role of LncRNA MIR210HG involved in the regulation of human cancer and other disease. Clin Transl Oncol 2023; 25:137-150. [PMID: 36088513 DOI: 10.1007/s12094-022-02943-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 08/30/2022] [Indexed: 01/07/2023]
Abstract
Long noncoding RNAs (lncRNAs) have evoked considerable interest in recent years due to their critical functions in the regulation of disease processes. Abnormal expression of lncRNAs is found in multiple diseases, and lncRNAs have been exploited for diverse medical applications. The lncRNA MIR210HG is a recently discovered lncRNA that is widely dysregulated in human disease. MIR210HG was described to have biological functions with potential roles in disease development, including cell proliferation, invasion, migration, and energy metabolism. And MIR210HG dysregulation was confirmed to have promising clinical values in disease diagnosis, treatment, and prognosis. In this review, we systematically summarize the expression profiles, roles, underlying mechanisms, and clinical applications of MIR210HG in human disease.
Collapse
Affiliation(s)
- Juan Lu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Shangcheng District, No. 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China
| | - Danhua Zhu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Shangcheng District, No. 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China
| | - Xiaoqian Zhang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Shangcheng District, No. 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China
| | - Jie Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Shangcheng District, No. 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China
| | - Hongcui Cao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Shangcheng District, No. 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China
| | - Lanjuan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Shangcheng District, No. 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China.
| |
Collapse
|
15
|
Bruno MC, Cristiano MC, Celia C, d'Avanzo N, Mancuso A, Paolino D, Wolfram J, Fresta M. Injectable Drug Delivery Systems for Osteoarthritis and Rheumatoid Arthritis. ACS NANO 2022; 16:19665-19690. [PMID: 36512378 DOI: 10.1021/acsnano.2c06393] [Citation(s) in RCA: 67] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Joint diseases are one of the most common causes of morbidity and disability worldwide. The main diseases that affect joint cartilage are osteoarthritis and rheumatoid arthritis, which require chronic treatment focused on symptomatic relief. Conventional drugs administered through systemic or intra-articular routes have low accumulation and/or retention in articular cartilage, causing dose-limiting toxicities and reduced efficacy. Therefore, there is an urgent need to develop improved strategies for drug delivery, in particular, the use of micro- and nanotechnology-based methods. Encapsulation of therapeutic agents in delivery systems reduces drug efflux from the joint and protects against rapid cellular and enzymatic clearance following intra-articular injection. Consequently, the use of drug delivery systems decreases side effects and increases therapeutic efficacy due to enhanced drug retention in the intra-articular space. Additionally, the frequency of intra-articular administration is reduced, as delivery systems enable sustained drug release. This review summarizes various advanced drug delivery systems, such as nano- and microcarriers, developed for articular cartilage diseases.
Collapse
Affiliation(s)
- Maria Chiara Bruno
- Department of Health Sciences, School of Pharmacy and Nutraceuticals, University "Magna Græcia" of Catanzaro, Campus Universitario "S. Venuta", Building of BioSciences, Viale S. Venuta, Germaneto-Catanzaro, I-88100, Italy
| | - Maria Chiara Cristiano
- Department of Experimental and Clinical Medicine, School of Pharmacy and Nutraceuticals, University "Magna Græcia" of Catanzaro, Campus Universitario "S. Venuta", Building of BioSciences, Viale S. Venuta, Germaneto-Catanzaro, I-88100, Italy
| | - Christian Celia
- Department of Pharmacy, University of Chieti - Pescara "G. d'Annunzio", Via dei Vestini 31, Chieti, I-66100, Italy
- Laboratory of Drug Targets Histopathology, Institute of Cardiology, Lithuanian University of Health Sciences, A. Mickeviciaus g. 9, LT-44307, Kaunas, Lithuania
| | - Nicola d'Avanzo
- Department of Health Sciences, School of Pharmacy and Nutraceuticals, University "Magna Græcia" of Catanzaro, Campus Universitario "S. Venuta", Building of BioSciences, Viale S. Venuta, Germaneto-Catanzaro, I-88100, Italy
- Department of Pharmacy, University of Chieti - Pescara "G. d'Annunzio", Via dei Vestini 31, Chieti, I-66100, Italy
| | - Antonia Mancuso
- Department of Experimental and Clinical Medicine, School of Pharmacy and Nutraceuticals, University "Magna Græcia" of Catanzaro, Campus Universitario "S. Venuta", Building of BioSciences, Viale S. Venuta, Germaneto-Catanzaro, I-88100, Italy
| | - Donatella Paolino
- Department of Experimental and Clinical Medicine, School of Pharmacy and Nutraceuticals, University "Magna Græcia" of Catanzaro, Campus Universitario "S. Venuta", Building of BioSciences, Viale S. Venuta, Germaneto-Catanzaro, I-88100, Italy
| | - Joy Wolfram
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
- School of Chemical Engineering, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Massimo Fresta
- Department of Health Sciences, School of Pharmacy and Nutraceuticals, University "Magna Græcia" of Catanzaro, Campus Universitario "S. Venuta", Building of BioSciences, Viale S. Venuta, Germaneto-Catanzaro, I-88100, Italy
| |
Collapse
|
16
|
Jin J, Liu Y, Jiang C, Shen Y, Chu G, Liu C, Jiang L, Huang G, Qin Y, Zhang Y, Zhang C, Wang Y. Arbutin-modified microspheres prevent osteoarthritis progression by mobilizing local anti-inflammatory and antioxidant responses. Mater Today Bio 2022; 16:100370. [PMID: 35937573 PMCID: PMC9352975 DOI: 10.1016/j.mtbio.2022.100370] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 06/29/2022] [Accepted: 07/14/2022] [Indexed: 12/03/2022] Open
Abstract
Osteoarthritis (OA) is a common degenerative joint disease worldwide and currently there is no effective strategy to stop its progression. It is known that oxidative stress and inflammation can promote the development of OA, and therapeutic strategies against these conditions may alleviate OA. Arbutin (ARB), a major ingredient of the Chinese medicinal herb cowberry leaf, exerts good antioxidant and anti-inflammatory activities yet has not been studied in OA. Here we developed ARB-loaded gelatine methacryloyl-Liposome (GM-Lipo@ARB) microspheres which showed long-term release of ARB and excellent cartilage-targeting effects. The ARB-loaded microspheres effectively reduced the inflammatory response in interleukin (IL)-1β-treated arthritic chondrocytes. Moreover, the synthesized GM-Lipo@ARB microspheres regulated cartilage extracellular matrix (ECM) homeostasis through anti-inflammation effect via inhibiting NF-κB signaling and anti-oxidative stress effect via activating Nrf2 pathway. Intra-articular use of GM-Lipo@ARB can effectively reduce inflammation and oxidative stress in the articular cartilage and thus, attenuating OA progression in a mouse model. The study proposed a novel ARB-laden functional microsphere, GM-Lipo@ARB, and demonstrated that this compound may be used as an alternative therapeutics for treating OA.
Collapse
|
17
|
Song P, Cui Z, Hu L. Applications and prospects of intra-articular drug delivery system in arthritis therapeutics. J Control Release 2022; 352:946-960. [PMID: 36375618 DOI: 10.1016/j.jconrel.2022.11.018] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 11/06/2022] [Accepted: 11/08/2022] [Indexed: 11/17/2022]
Abstract
Arthritis is a kind of chronic disease that affects joints and muscles with the symptoms of joint pain, inflammation and limited movement of joints. Among various clinical therapies, drug therapy has been extensively applied because of its accessibility, safety and effectiveness. In recent years, the intra-articular injection has dramatic therapeutic effects in treating arthritis with high patient compliance and low side effects. In this review, we will introduce pathology of arthritis, along with the accessible treatment and diagnosis methods, then we will summarize major advances of current hopeful intra-articular delivery systems such as microspheres, hydrogels, nanoparticles and liposomes. At last, some safety assessments in the preclinical work and the main challenges for the further development of intra-articular treatment were also discussed.
Collapse
Affiliation(s)
- Pengjin Song
- Key Laboratory of Pharmaceutical Quality Control of Hebei Province, School of Pharmaceutical Sciences, Hebei University, Baoding 071000, China
| | - Zhe Cui
- Key Laboratory of Pharmaceutical Quality Control of Hebei Province, School of Pharmaceutical Sciences, Hebei University, Baoding 071000, China.
| | - Liandong Hu
- Key Laboratory of Pharmaceutical Quality Control of Hebei Province, School of Pharmaceutical Sciences, Hebei University, Baoding 071000, China.
| |
Collapse
|
18
|
Chou HC, Lin SY, Chou LY, Ho ML, Chuang SC, Cheng TL, Kang L, Lin YS, Wang YH, Wei CW, Chen CH, Wang CZ. Ablation of Discoidin Domain Receptor 1 Provokes an Osteopenic Phenotype by Regulating Osteoblast/Osteocyte Autophagy and Apoptosis. Biomedicines 2022; 10:biomedicines10092173. [PMID: 36140274 PMCID: PMC9496360 DOI: 10.3390/biomedicines10092173] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 08/10/2022] [Accepted: 08/30/2022] [Indexed: 11/16/2022] Open
Abstract
Discoidin domain receptor 1 (DDR1) is a collagen receptor that belongs to the receptor tyrosine kinase family. We have previously shown that DDR1 plays a crucial role during bone development, resulting in dwarfism and a short stature in osteoblast-specific knockout mice (OKO mice). However, the detailed pathophysiological effects of DDR1 on bone development throughout adulthood have remained unclear. This study aims to identify how DDR1 regulates osteoblast and osteocyte functions in vivo and in vitro during bone development in adulthood. The metabolic changes in bone tissues were analyzed using Micro-CT and immunohistochemistry staining (IHC) in vivo; the role of DDR1 in regulating osteoblasts was examined in MC3T3-E1 cells in vitro. The Micro-CT analysis results demonstrated that OKO mice showed a 10% reduction in bone-related parameters from 10 to 14 weeks old and a significant reduction in cortical thickness and diameter compared with flox/flox control mice (FF) mice. These results indicated that DDR1 knockout in OKO mice exhibiting significant bone loss provokes an osteopenic phenotype. The IHC staining revealed a significant decrease in osteogenesis-related genes, including RUNX2, osteocalcin, and osterix. We noted that DDR1 knockout significantly induced osteoblast/osteocyte apoptosis and markedly decreased autophagy activity in vivo. Additionally, the results of the gain- and loss-of-function of the DDR1 assay in MC3T3-E1 cells indicated that DDR1 can regulate the osteoblast differentiation through activating autophagy by regulating the phosphorylation of the mechanistic target of rapamycin (p-mTOR), light chain 3 (LC3), and beclin-1. In conclusion, our study highlights that the ablation of DDR1 results in cancellous bone loss by regulating osteoblast/osteocyte autophagy. These results suggest that DDR1 can act as a potential therapeutic target for managing cancellous bone loss.
Collapse
Affiliation(s)
- Hsin-Chiao Chou
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Regeneration Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Sung-Yen Lin
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Regeneration Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Orthopedics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Departments of Orthopedics, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Orthopedics, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung City 80145, Taiwan
| | - Liang-Yin Chou
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Regeneration Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Mei-Ling Ho
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Regeneration Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Departments of Orthopedics, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Physiology, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 80424, Taiwan
| | - Shu-Chun Chuang
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Regeneration Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Tsung-Lin Cheng
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Regeneration Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Orthopedics, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung City 80145, Taiwan
| | - Lin Kang
- Department of Obstetrics and Gynecology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
| | - Yi-Shan Lin
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Regeneration Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Yan-Hsiung Wang
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Regeneration Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- School of Dentistry, College of Dental Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Chun-Wang Wei
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 80424, Taiwan
| | - Chung-Hwan Chen
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Regeneration Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Orthopedics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Departments of Orthopedics, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Orthopedics, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung City 80145, Taiwan
- Institute of Medical Science and Technology, National Sun Yat-Sen University, Kaohsiung 80424, Taiwan
- Department of Healthcare Administration and Medical Informatics, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Ph.D. Program in Biomedical Engineering, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Graduate Institute of Animal Vaccine Technology, College of Veterinary Medicine, National Pingtung University of Science and Technology, Pingtung 912301, Taiwan
- Correspondence: (C.-H.C.); (C.-Z.W.); Tel.: +886-7-3209209 (C.-H.C.); +886-7-3121101 (ext. 2140) (C.-Z.W.)
| | - Chau-Zen Wang
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Regeneration Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Physiology, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
- College of Professional Studies, National Pingtung University of Science and Technology, Pingtung 912301, Taiwan
- Correspondence: (C.-H.C.); (C.-Z.W.); Tel.: +886-7-3209209 (C.-H.C.); +886-7-3121101 (ext. 2140) (C.-Z.W.)
| |
Collapse
|
19
|
Wang L, He C. Nrf2-mediated anti-inflammatory polarization of macrophages as therapeutic targets for osteoarthritis. Front Immunol 2022; 13:967193. [PMID: 36032081 PMCID: PMC9411667 DOI: 10.3389/fimmu.2022.967193] [Citation(s) in RCA: 81] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Accepted: 07/27/2022] [Indexed: 12/14/2022] Open
Abstract
Macrophages are the most abundant immune cells within the synovial joints, and also the main innate immune effector cells triggering the initial inflammatory responses in the pathological process of osteoarthritis (OA). The transition of synovial macrophages between pro-inflammatory and anti-inflammatory phenotypes can play a key role in building the intra-articular microenvironment. The pro-inflammatory cascade induced by TNF-α, IL-1β, and IL-6 is closely related to M1 macrophages, resulting in the production of pro-chondrolytic mediators. However, IL-10, IL1RA, CCL-18, IGF, and TGF are closely related to M2 macrophages, leading to the protection of cartilage and the promoted regeneration. The inhibition of NF-κB signaling pathway is central in OA treatment via controlling inflammatory responses in macrophages, while the nuclear factor erythroid 2-related factor 2 (Nrf2) signaling pathway appears not to attract widespread attention in the field. Nrf2 is a transcription factor encoding a large number of antioxidant enzymes. The activation of Nrf2 can have antioxidant and anti-inflammatory effects, which can also have complex crosstalk with NF-κB signaling pathway. The activation of Nrf2 can inhibit the M1 polarization and promote the M2 polarization through potential signaling transductions including TGF-β/SMAD, TLR/NF-κB, and JAK/STAT signaling pathways, with the regulation or cooperation of Notch, NLRP3, PI3K/Akt, and MAPK signaling. And the expression of heme oxygenase-1 (HO-1) and the negative regulation of Nrf2 for NF-κB can be the main mechanisms for promotion. Furthermore, the candidates of OA treatment by activating Nrf2 to promote M2 phenotype macrophages in OA are also reviewed in this work, such as itaconate and fumarate derivatives, curcumin, quercetin, melatonin, mesenchymal stem cells, and low-intensity pulsed ultrasound.
Collapse
Affiliation(s)
- Lin Wang
- Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China,Key Laboratory of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Chengqi He
- Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China,Key Laboratory of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China,*Correspondence: Chengqi He,
| |
Collapse
|
20
|
Yu C, Li L, Liang D, Wu A, Dong Q, Jia S, Li Y, Li Y, Guo X, Zang H. Glycosaminoglycan-based injectable hydrogels with multi-functions in the alleviation of osteoarthritis. Carbohydr Polym 2022; 290:119492. [DOI: 10.1016/j.carbpol.2022.119492] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 04/02/2022] [Accepted: 04/12/2022] [Indexed: 01/08/2023]
|
21
|
Nanomedicine and regenerative medicine approaches in osteoarthritis therapy. Aging Clin Exp Res 2022; 34:2305-2315. [PMID: 35867240 DOI: 10.1007/s40520-022-02199-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Accepted: 07/06/2022] [Indexed: 11/01/2022]
Abstract
Osteoarthritis (OA), the most common chronic joint disease, is a degenerative disease that affects 7% of the worldwide population, more than 500 million people all over the world. OA is the main factor of disability in elderly people which decreases the quality of life of patients. It is characterized by joint pain, low bone density, and deterioration of the joint structure. Despite ongoing novel advances in drug discovery and drug delivery, OA therapy is still a big challenge since there is no available effective treatment and the existing therapies mainly focus on pain and symptomatic management rather than improving and/or suppressing its progression. This review aims to summarize the currently available and novel emerging therapies for OA including regenerative medicine and nanotechnology-based materials and formulations at the clinical and experimental levels. Applications of regenerative medicine and novel technologies such as nanotechnology in OA treatments have opened a new window to support OA patients by offering treatments that could halt or delay OA progression satisfactorily or provide an effective cure in near future. Nanomedicine and regenerative medicine suggest novel alternatives in the regeneration of cartilage, repair of bone damage, and control of chronic pain in OA therapy.
Collapse
|
22
|
Pape E, Pinzano A, Henrionnet C, Scala-Bertola J, Gillet P, Gambier N. Rat synovial tissue and blood rapamycin pharmacokinetics after intra-articular injection of free solution or nanoparticles vs free rapamycin intravenous shot. Int J Pharm 2022; 624:122026. [PMID: 35863592 DOI: 10.1016/j.ijpharm.2022.122026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 07/09/2022] [Accepted: 07/13/2022] [Indexed: 10/17/2022]
Abstract
Intra-articular (IA) injection of a chondroprotective candidate may delay the osteoarthritis (OA) course, but its rapid absorption into systemic circulation may limit efficacy and produce untoward effects. We compared the pharmacokinetics (PK) of IA rapamycin injected as sustained release in nanoparticles (NPs) versus a free rapamycin suspension in the rat knee compared to an intravenous (IV) free rapamycin shot taken as a reference. Rats received either a single IV injection of free rapamycin (10 µM) or an IA of free or NPs-loaded rapamycin. After sequential exsanguination (15, 30, 60, 180, 360 min, D1, and D7), knee synovial tissue (ST) and cartilage histology were performed. Blood and ST concentrations (LC-MS/MS), PK parameters (area under the curve: AUC; mean residence time: MRT; elimination half-life: T1/2), and IA biocompatibility were assessed. AUCIV was significantly higher for IV than for both IA injections (AUCIA free and AUCIA NPs), with 4248 vs 28 and 74 µg.min.L-1. For ST parameters, we observed a significant difference between AUCIA free and AUCIA NPs with 3735 and 10513 µg.min.L-1 correspondingly. Articular T1/2 and MRT were higher after NPs than after free rapamycin injection: 57.8 and 5.0 h for T1/2 and 80.6 and 5.5 h for MRT, respectively. Histological analysis revealed no chondral injuries and slight transient synovitis only 3 h after the administration of NPs. In the rat knee, rapamycin-loaded NPs delivery via a single IA injection is biocompatible and prolongs synovium joint residency, diminishes blood levels, and reduces detrimental systemic exposure.
Collapse
Affiliation(s)
- Elise Pape
- Université de Lorraine, CNRS, IMoPA, F-54000 Nancy, France; Laboratoire de Pharmacologie-Toxicologie, Pharmacovigilance & CEIPA, Bâtiment de Biologie Médicale et de Biopathologie, CHRU de Nancy-Brabois, 5 Rue du Morvan, F54511 Vandœuvre-Lès-Nancy, France.
| | - Astrid Pinzano
- Université de Lorraine, CNRS, IMoPA, F-54000 Nancy, France.
| | | | - Julien Scala-Bertola
- Université de Lorraine, CNRS, IMoPA, F-54000 Nancy, France; Laboratoire de Pharmacologie-Toxicologie, Pharmacovigilance & CEIPA, Bâtiment de Biologie Médicale et de Biopathologie, CHRU de Nancy-Brabois, 5 Rue du Morvan, F54511 Vandœuvre-Lès-Nancy, France.
| | - Pierre Gillet
- Université de Lorraine, CNRS, IMoPA, F-54000 Nancy, France; Laboratoire de Pharmacologie-Toxicologie, Pharmacovigilance & CEIPA, Bâtiment de Biologie Médicale et de Biopathologie, CHRU de Nancy-Brabois, 5 Rue du Morvan, F54511 Vandœuvre-Lès-Nancy, France.
| | - Nicolas Gambier
- Université de Lorraine, CNRS, IMoPA, F-54000 Nancy, France; Laboratoire de Pharmacologie-Toxicologie, Pharmacovigilance & CEIPA, Bâtiment de Biologie Médicale et de Biopathologie, CHRU de Nancy-Brabois, 5 Rue du Morvan, F54511 Vandœuvre-Lès-Nancy, France.
| |
Collapse
|
23
|
Veronesi F, Salamanna F, Martini L, Fini M. Naturally Occurring Osteoarthritis Features and Treatments: Systematic Review on the Aged Guinea Pig Model. Int J Mol Sci 2022; 23:ijms23137309. [PMID: 35806306 PMCID: PMC9266929 DOI: 10.3390/ijms23137309] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 06/27/2022] [Accepted: 06/29/2022] [Indexed: 12/09/2022] Open
Abstract
To date, several in vivo models have been used to reproduce the onset and monitor the progression of osteoarthritis (OA), and guinea pigs represent a standard model for studying naturally occurring, age-related OA. This systematic review aims to characterize the guinea pig for its employment in in vivo, naturally occurring OA studies and for the evaluation of specific disease-modifying agents. The search was performed in PubMed, Scopus, and Web of Knowledge in the last 10 years. Of the 233 records screened, 49 studies were included. Results showed that within a relatively short period of time, this model develops specific OA aspects, including cartilage degeneration, marginal osteophytes formation, and subchondral bone alterations. Disease severity increases with age, beginning at 3 months with mild OA and reaching moderate–severe OA at 18 months. Among the different strains, Dunkin Hartley develops OA at a relatively early age. Thus, disease-modifying agents have mainly been evaluated for this strain. As summarized herein, spontaneous development of OA in guinea pigs represents an excellent model for studying disease pathogenesis and for evaluating therapeutic interventions. In an ongoing effort at standardization, a detailed characterization of specific OA models is necessary, even considering the main purpose of these models, i.e., translatability to human OA.
Collapse
|
24
|
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
|
25
|
Ma L, Zheng X, Lin R, Sun AR, Song J, Ye Z, Liang D, Zhang M, Tian J, Zhou X, Cui L, Liu Y, Liu Y. Knee Osteoarthritis Therapy: Recent Advances in Intra-Articular Drug Delivery Systems. Drug Des Devel Ther 2022; 16:1311-1347. [PMID: 35547865 PMCID: PMC9081192 DOI: 10.2147/dddt.s357386] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 04/17/2022] [Indexed: 12/12/2022] Open
Abstract
Drug delivery for osteoarthritis (OA) treatment is a continuous challenge because of their poor bioavailability and rapid clearance in joints. Intra-articular (IA) drug delivery is a common strategy and its therapeutic effects depend mainly on the efficacy of the drug-delivery system used for OA therapy. Different types of IA drug-delivery systems, such as microspheres, nanoparticles, and hydrogels, have been rapidly developed over the past decade to improve their therapeutic effects. With the continuous advancement in OA mechanism research, new drugs targeting specific cell/signaling pathways in OA are rapidly evolving and effective drug delivery is critical for treating OA. In this review, recent advances in various IA drug-delivery systems for OA treatment, OA targeted strategies, and related signaling pathways in OA treatment are summarized and analyzed based on current publications.
Collapse
Affiliation(s)
- Luoyang Ma
- Guangdong Provincial Key Laboratory for Research and Development of Natural Drug, School of Pharmacy, Guangdong Medical University, Zhanjiang City, Guangdong Province, 524023, People’s Republic of China
- Marine Medical Research Institute of Zhanjiang, Zhanjiang City, Guangdong Province, 524023, People’s Republic of China
| | - Xiaoyan Zheng
- Guangdong Provincial Key Laboratory for Research and Development of Natural Drug, School of Pharmacy, Guangdong Medical University, Zhanjiang City, Guangdong Province, 524023, People’s Republic of China
- Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang city, Guangdong province, 524045, People's Republic of China
| | - Rui Lin
- Guangdong Provincial Key Laboratory for Research and Development of Natural Drug, School of Pharmacy, Guangdong Medical University, Zhanjiang City, Guangdong Province, 524023, People’s Republic of China
| | - Antonia RuJia Sun
- Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Science, Shenzhen City, Guangdong Province, 518055, People’s Republic of China
| | - Jintong Song
- Guangdong Provincial Key Laboratory for Research and Development of Natural Drug, School of Pharmacy, Guangdong Medical University, Zhanjiang City, Guangdong Province, 524023, People’s Republic of China
| | - Zhiqiang Ye
- Guangdong Provincial Key Laboratory for Research and Development of Natural Drug, School of Pharmacy, Guangdong Medical University, Zhanjiang City, Guangdong Province, 524023, People’s Republic of China
| | - Dahong Liang
- Guangdong Provincial Key Laboratory for Research and Development of Natural Drug, School of Pharmacy, Guangdong Medical University, Zhanjiang City, Guangdong Province, 524023, People’s Republic of China
| | - Min Zhang
- Guangdong Provincial Key Laboratory for Research and Development of Natural Drug, School of Pharmacy, Guangdong Medical University, Zhanjiang City, Guangdong Province, 524023, People’s Republic of China
| | - Jia Tian
- Guangdong Provincial Key Laboratory for Research and Development of Natural Drug, School of Pharmacy, Guangdong Medical University, Zhanjiang City, Guangdong Province, 524023, People’s Republic of China
| | - Xin Zhou
- Marine Medical Research Institute of Zhanjiang, Zhanjiang City, Guangdong Province, 524023, People’s Republic of China
| | - Liao Cui
- Guangdong Provincial Key Laboratory for Research and Development of Natural Drug, School of Pharmacy, Guangdong Medical University, Zhanjiang City, Guangdong Province, 524023, People’s Republic of China
| | - Yuyu Liu
- Guangdong Provincial Key Laboratory for Research and Development of Natural Drug, School of Pharmacy, Guangdong Medical University, Zhanjiang City, Guangdong Province, 524023, People’s Republic of China
| | - Yanzhi Liu
- Guangdong Provincial Key Laboratory for Research and Development of Natural Drug, School of Pharmacy, Guangdong Medical University, Zhanjiang City, Guangdong Province, 524023, People’s Republic of China
- Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang city, Guangdong province, 524045, People's Republic of China
- Shenzhen Osteomore Biotechnology Co., Ltd., Shenzhen city, Guangdong Province, 518118, People’s Republic of China
- Correspondence: Yanzhi Liu; Yuyu Liu, Tel +86-759-2388405; +86-759-2388588, Email ;
| |
Collapse
|
26
|
Wu G, Wu J, Li Z, Shi S, Wu D, Wang X, Xu H, Liu H, Huang Y, Wang R, Shen J, Dong Z, Wang S. Development of digital organ-on-a-chip to assess hepatotoxicity and extracellular vesicle-based anti-liver cancer immunotherapy. Biodes Manuf 2022. [DOI: 10.1007/s42242-022-00188-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
AbstractOrgan-on-a-chip systems have been increasingly recognized as attractive platforms to assess toxicity and to develop new therapeutic agents. However, current organ-on-a-chip platforms are limited by a “single pot” design, which inevitably requires holistic analysis and limits parallel processing. Here, we developed a digital organ-on-a-chip by combining a microwell array with cellular microspheres, which significantly increased the parallelism over traditional organ-on-a-chip for drug development. Up to 127 uniform liver cancer microspheres in this digital organ-on-a-chip format served as individual analytical units, allowing for analysis with high consistency and quick response. Our platform displayed evident anti-cancer efficacy at a concentration of 10 μM for sorafenib, and had greater alignment than the “single pot” organ-on-a-chip with a previous in vivo study. In addition, this digital organ-on-a-chip demonstrated the treatment efficacy of natural killer cell-derived extracellular vesicles for liver cancer at 50 μg/mL. The successful development of this digital organ-on-a-chip platform provides high-parallelism and a low-variability analytical tool for toxicity assessment and the exploration of new anticancer modalities, thereby accelerating the joint endeavor to combat cancer.
Graphic abstract
Collapse
|
27
|
Lei Y, Wang Y, Shen J, Cai Z, Zhao C, Chen H, Luo X, Hu N, Cui W, Huang W. Injectable hydrogel microspheres with self-renewable hydration layers alleviate osteoarthritis. SCIENCE ADVANCES 2022; 8:eabl6449. [PMID: 35108047 PMCID: PMC8809544 DOI: 10.1126/sciadv.abl6449] [Citation(s) in RCA: 138] [Impact Index Per Article: 46.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
Introducing hydration layers to hydrogel microspheres (HMs) by coating the surface with liposomes can effectively reduce friction. However, the lubrication can be inactivated when the surface coatings are damaged. To endow HMs with the ability to form self-renewable hydration layers and maintain cellular homeostasis, rapamycin-liposome-incorporating hyaluronic acid-based HMs (RAPA@Lipo@HMs) were created using microfluidic technology and photopolymerization processes. The RAPA@Lipo@HMs improve joint lubrication by using a smooth rolling mechanism and continuously exposing liposomes on the outer surface to form self-renewable hydration layers via frictional wear. In addition, the released autophagy activator (rapamycin)-loaded cationic liposomes can target negatively charged cartilage through electrostatic interactions and maintain cellular homeostasis by increasing autophagy. Furthermore, the in vivo data showed that the RAPA@Lipo@HMs can alleviate joint wear and delay the progression of osteoarthritis. The RAPA@Lipo@HMs can provide efficient lubrication and potentially alleviate friction-related diseases such as osteoarthritis.
Collapse
Affiliation(s)
- Yiting Lei
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Orthopedic Laboratory of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing 400016, P. R. China
| | - Yuping Wang
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Orthopedic Laboratory of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing 400016, P. R. China
| | - Jieliang Shen
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Orthopedic Laboratory of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing 400016, P. R. China
| | - Zhengwei Cai
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, P. R. China
| | - Chen Zhao
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Orthopedic Laboratory of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing 400016, P. R. China
| | - Hong Chen
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Orthopedic Laboratory of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing 400016, P. R. China
| | - Xiaoji Luo
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Orthopedic Laboratory of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing 400016, P. R. China
| | - Ning Hu
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Orthopedic Laboratory of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing 400016, P. R. China
- Corresponding author. (N.H.); (W.C.); (W.H.)
| | - Wenguo Cui
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, P. R. China
- Corresponding author. (N.H.); (W.C.); (W.H.)
| | - Wei Huang
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Orthopedic Laboratory of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing 400016, P. R. China
- Corresponding author. (N.H.); (W.C.); (W.H.)
| |
Collapse
|
28
|
Intra-Articular Drug Delivery for Osteoarthritis Treatment. Pharmaceutics 2021; 13:pharmaceutics13122166. [PMID: 34959445 PMCID: PMC8703898 DOI: 10.3390/pharmaceutics13122166] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 12/13/2021] [Accepted: 12/13/2021] [Indexed: 02/07/2023] Open
Abstract
Osteoarthritis (OA) is the most prevalent degenerative joint disease affecting millions of people worldwide. Currently, clinical nonsurgical treatments of OA are only limited to pain relief, anti-inflammation, and viscosupplementation. Developing disease-modifying OA drugs (DMOADs) is highly demanded for the efficient treatment of OA. As OA is a local disease, intra-articular (IA) injection directly delivers drugs to synovial joints, resulting in high-concentration drugs in the joint and reduced side effects, accompanied with traditional oral or topical administrations. However, the injected drugs are rapidly cleaved. By properly designing the drug delivery systems, prolonged retention time and targeting could be obtained. In this review, we summarize the drugs investigated for OA treatment and recent advances in the IA drug delivery systems, including micro- and nano-particles, liposomes, and hydrogels, hoping to provide some information for designing the IA injected formulations.
Collapse
|
29
|
Yu Y, Wang Z, Yang Q, Ding Q, Wang R, Li Z, Fang Y, Liao J, Qi W, Chen K, Li M, Zhu YZ. A novel dendritic mesoporous silica based sustained hydrogen sulfide donor for the alleviation of adjuvant-induced inflammation in rats. Drug Deliv 2021; 28:1031-1042. [PMID: 34060389 PMCID: PMC8172227 DOI: 10.1080/10717544.2021.1921075] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 04/15/2021] [Accepted: 04/20/2021] [Indexed: 12/29/2022] Open
Abstract
PURPOSE S-propargyl-cysteine (SPRC), an excellent endogenous hydrogen sulfide (H2S) donor, could elevate H2S levels via the cystathionine γ-lyase (CSE)/H2S pathway both in vitro and in vivo. However, the immediate release of H2S in vivo and daily administration of SPRC potentially limited its clinical use. METHODS To solve the fore-mentioned problem, in this study, the dendritic mesoporous silica nanoparticles (DMSN) was firstly prepared, and a sustained H2S delivery system consisted of SPRC and DMSN (SPRC@DMSN) was then constructed. Their release profiles, both in vitro and in vivo, were investigated, and their therapeutical effect toward adjuvant-induced arthritis (AIA) rats was also studied. RESULTS The spherical morphology of DMSN could be observed under scanning Electron Microscope (SEM), and the transmission electron microscope (TEM) images showed a central-radiational pore channel structure of DMSN. DMSN showed excellent SPRC loading capacity and attaining a sustained releasing ability than SPRC both in vitro and in vivo, and the prolonged SPRC releasing could further promote the release of H2S in a sustained manner through CSE/H2S pathway both in vitro and in vivo. Importantly, the SPRC@DMSN showed promising anti-inflammation effect against AIA in rats was also observed. CONCLUSIONS A sustained H2S releasing donor consisting of SPRC and DMSN was constructed in this study, and this sustained H2S releasing donor might be of good use for the treatment of AIA.
Collapse
Affiliation(s)
- Yue Yu
- State Key Laboratory of Quality Research in Chinese Medicine & School of Pharmacy, Macau University of Science and Technology, Taipa, China
| | - Zhou Wang
- State Key Laboratory of Quality Research in Chinese Medicine & School of Pharmacy, Macau University of Science and Technology, Taipa, China
| | - Qinyan Yang
- State Key Laboratory of Quality Research in Chinese Medicine & School of Pharmacy, Macau University of Science and Technology, Taipa, China
| | - Qian Ding
- State Key Laboratory of Quality Research in Chinese Medicine & School of Pharmacy, Macau University of Science and Technology, Taipa, China
| | - Ran Wang
- State Key Laboratory of Quality Research in Chinese Medicine & School of Pharmacy, Macau University of Science and Technology, Taipa, China
| | - Zhaoyi Li
- State Key Laboratory of Quality Research in Chinese Medicine & School of Pharmacy, Macau University of Science and Technology, Taipa, China
| | - Yudong Fang
- State Key Laboratory of Quality Research in Chinese Medicine & School of Pharmacy, Macau University of Science and Technology, Taipa, China
| | - Junyi Liao
- State Key Laboratory of Quality Research in Chinese Medicine & School of Pharmacy, Macau University of Science and Technology, Taipa, China
| | - Wei Qi
- State Key Laboratory of Quality Research in Chinese Medicine & School of Pharmacy, Macau University of Science and Technology, Taipa, China
| | - Keyuan Chen
- State Key Laboratory of Quality Research in Chinese Medicine & School of Pharmacy, Macau University of Science and Technology, Taipa, China
| | - Meng Li
- State Key Laboratory of Quality Research in Chinese Medicine & School of Pharmacy, Macau University of Science and Technology, Taipa, China
| | - Yi Zhun Zhu
- State Key Laboratory of Quality Research in Chinese Medicine & School of Pharmacy, Macau University of Science and Technology, Taipa, China
- Shanghai Key Laboratory of Bioactive Small Molecules & School of Pharmacy, Fudan University, Shanghai, China
| |
Collapse
|
30
|
Pape E, Parent M, Pinzano A, Sapin-Minet A, Henrionnet C, Gillet P, Scala-Bertola J, Gambier N. Rapamycin-loaded Poly(lactic-co-glycolic) acid nanoparticles: Preparation, characterization, and in vitro toxicity study for potential intra-articular injection. Int J Pharm 2021; 609:121198. [PMID: 34662644 DOI: 10.1016/j.ijpharm.2021.121198] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 09/29/2021] [Accepted: 10/10/2021] [Indexed: 12/11/2022]
Abstract
Osteoarthritis (OA) is the most common degenerative joint disease. Rapamycin is a potential candidate for OA treatment by increasing the autophagy process implicated in its physiopathology. To optimize Rapamycin profit and avoid systemic side effects, intra-articular (i.a.) administration appeared helpful. However, Rapamycin's highly hydrophobic nature and low bioavailability made it challenging to develop purpose-made drug delivery systems to overcome these limitations. We developed Rapamycin-loaded nanoparticles (NPs) using poly (lactic-co-glycolic acid) by emulsion/evaporation method. We evaluated these NPs' cytocompatibility towards cartilage (chondrocytes) and synovial membrane cells (synoviocytes) for a potential i.a. administration. The in vitro characterization of Rapamycin-loaded NPs had shown a suitable profile for an i.a. administration. In vitro biocompatibility of NPs was highlighted to 10 µM of Rapamycin for both synoviocytes and chondrocytes, but significant toxicity was observed with higher concentrations. Besides, synoviocytes are more sensitive to Rapamycin-loaded NPs than chondrocytes. Finally, we observed in vitro that an adapted formulated Rapamycin-loaded NPs could be safe at suitable i.a. injection concentrations. The toxic effect of Rapamycin encapsulated in these NPs on both articular cells was dose-dependent. After Rapamycin-loaded NPs i.a. administration, local retention, in situ safety, and systemic release should be evaluated with experimental in vivo models.
Collapse
Affiliation(s)
- Elise Pape
- Université de Lorraine, CNRS, IMoPA, F-54000 Nancy, France; Laboratoire de Pharmacologie, Toxicologie et Pharmacovigilance, Bâtiment de Biologie Médicale et de Biopathologie, CHRU de Nancy-Brabois, 5 Rue du Morvan, F54511 Vandœuvre-Lès-Nancy, France.
| | | | - Astrid Pinzano
- Université de Lorraine, CNRS, IMoPA, F-54000 Nancy, France.
| | | | | | - Pierre Gillet
- Université de Lorraine, CNRS, IMoPA, F-54000 Nancy, France; Laboratoire de Pharmacologie, Toxicologie et Pharmacovigilance, Bâtiment de Biologie Médicale et de Biopathologie, CHRU de Nancy-Brabois, 5 Rue du Morvan, F54511 Vandœuvre-Lès-Nancy, France.
| | - Julien Scala-Bertola
- Université de Lorraine, CNRS, IMoPA, F-54000 Nancy, France; Laboratoire de Pharmacologie, Toxicologie et Pharmacovigilance, Bâtiment de Biologie Médicale et de Biopathologie, CHRU de Nancy-Brabois, 5 Rue du Morvan, F54511 Vandœuvre-Lès-Nancy, France.
| | - Nicolas Gambier
- Université de Lorraine, CNRS, IMoPA, F-54000 Nancy, France; Laboratoire de Pharmacologie, Toxicologie et Pharmacovigilance, Bâtiment de Biologie Médicale et de Biopathologie, CHRU de Nancy-Brabois, 5 Rue du Morvan, F54511 Vandœuvre-Lès-Nancy, France.
| |
Collapse
|
31
|
Ma X, Zhang Z, Kang X, Deng C, Sun Y, Li Y, Huang D, Liu X. Defining matrix Gla protein expression in the Dunkin-Hartley guinea pig model of spontaneous osteoarthritis. BMC Musculoskelet Disord 2021; 22:870. [PMID: 34641845 PMCID: PMC8513366 DOI: 10.1186/s12891-021-04735-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 09/24/2021] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Matrix Gla (γ-carboxyglutamate) protein (MGP) is considered a strong inhibitor of ectopic calcification, and it has been associated with OA severity, although not conclusively. We utilized male Dunkin-Hartley (DH) guinea pigs to investigate the expression of MGP throughout aging and disease pathogenesis in a spontaneous model. METHOD Twenty-five male DH guinea pigs were obtained and nurtured to several timepoints, and then randomly and equally divided by age into five subgroups (1-, 3-, 6-, 9-, and 12-months, with the 1-month group as the reference group). DH guinea pigs in each group were euthanized at the designated month-age and the left or right medial tibial plateaus cartilages were randomly excised. OA severity was described by modified Mankin Score (MMS) at microscopy (Safranin O/Fast Green stain). Proteomic evaluation using isobaric tags for relative and absolute quantification (iTRAQ) was performed to validate the age-related changes in the MGP profiles, and immunohistochemistry (IHC) methods were applied for semi-quantitative determination of MGP expression in articular cartilage. RESULTS The histopathologic findings validated the increasing severity of cartilage degeneration with age in the DH guinea pigs. The MMS showed significant, stepwise (every adjacent comparison P < 0.05) disease progression with month-age. The iTRAQ indicated that MGP levels increased significantly with advancing age (P < 0.05), as supported by the IHC result (P < 0.05). CONCLUSION Increased expression of MGP in male DH guinea pigs was present throughout aging and disease progression and may be link to increased OA severity. Further studies are needed to investigate and confirm the association between MGP levels and OA severity.
Collapse
Affiliation(s)
- Xun Ma
- Department of Rehabilitation, Shengjing Hospital of China Medical University, No.16, Puhe Street, Shenyang North New Area, Shenyang, 110134, Liaoning Province, China
| | - Zhan Zhang
- Department of Orthopaedics, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Xinyuan Kang
- Department of Orthopaedics, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Chunbo Deng
- Department of Orthopaedics, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, China
- Department of Orthopedics, Central Hospital of Shenyang Medical College, Shenyang, Liaoning Province, China
| | - Yingwei Sun
- Department of Radiology, Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning Province, China
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Yanjun Li
- Department of Mathematics, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Desheng Huang
- Department of Mathematics, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Xueyong Liu
- Department of Rehabilitation, Shengjing Hospital of China Medical University, No.16, Puhe Street, Shenyang North New Area, Shenyang, 110134, Liaoning Province, China.
| |
Collapse
|
32
|
Chen CH, Kang L, Chang LH, Cheng TL, Lin SY, Wu SC, Lin YS, Chuang SC, Lee TC, Chang JK, Ho ML. Intra-articular low-dose parathyroid hormone (1-34) improves mobility and articular cartilage quality in a preclinical age-related knee osteoarthritis model. Bone Joint Res 2021; 10:514-525. [PMID: 34387115 PMCID: PMC8414442 DOI: 10.1302/2046-3758.108.bjr-2020-0165.r2] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Aims Osteoarthritis (OA) is prevalent among the elderly and incurable. Intra-articular parathyroid hormone (PTH) ameliorated OA in papain-induced and anterior cruciate ligament transection-induced OA models; therefore, we hypothesized that PTH improved OA in a preclinical age-related OA model. Methods Guinea pigs aged between six and seven months of age were randomized into control or treatment groups. Three- or four-month-old guinea pigs served as the young control group. The knees were administered 40 μl intra-articular injections of 10 nM PTH or vehicle once a week for three months. Their endurance as determined from time on the treadmill was evaluated before kill. Their tibial plateaus were analyzed using microcalculated tomography (μCT) and histological studies. Results PTH increased the endurance on the treadmill test, preserved glycosaminoglycans, and reduced Osteoarthritis Research Society International score and chondrocyte apoptosis rate. No difference was observed in the subchondral plate bone density or metaphyseal trabecular bone volume and bone morphogenetic 2 protein staining. Conclusion Subchondral bone is crucial in the initiation and progression of OA. Although previous studies have shown that subcutaneous PTH alleviates knee OA by improving subchondral and metaphyseal bone mass, we demonstrated that intra-articular PTH injections improved spontaneous OA by directly affecting the cartilage rather than the subchondral or metaphyseal bone in a preclinical age-related OA model. Cite this article: Bone Joint Res 2021;10(8):514–525.
Collapse
Affiliation(s)
- Chung-Hwan Chen
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan.,Institute of Medical Science and Technology, National Sun Yat-Sen University, Kaohsiung, Taiwan.,Department of Healthcare Administration and Medical Informatics, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Orthopedics, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Division of Adult Reconstruction Surgery, Department of Orthopedics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Orthopedics, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan.,Regeneration Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Lin Kang
- Department of Obstetrics and Gynecology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Ling-Hua Chang
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Tsung-Lin Cheng
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan.,Regeneration Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Physiology, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Sung-Yen Lin
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Orthopedics, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Division of Adult Reconstruction Surgery, Department of Orthopedics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Orthopedics, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Shun-Cheng Wu
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan.,Regeneration Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yi-Shan Lin
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Shu-Chun Chuang
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan.,Regeneration Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Tien-Ching Lee
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Orthopedics, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Division of Adult Reconstruction Surgery, Department of Orthopedics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Orthopedics, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan.,Regeneration Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Je-Ken Chang
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Orthopedics, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Division of Adult Reconstruction Surgery, Department of Orthopedics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Orthopedics, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan.,Regeneration Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Mei-Ling Ho
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan.,Regeneration Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Physiology, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung, Taiwan.,Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
33
|
Yi X, Liu J, Cheng MS, Zhou Q. Low-intensity pulsed ultrasound inhibits IL-6 in subchondral bone of temporomandibular joint osteoarthritis by suppressing the TGF-β1/Smad3 pathway. Arch Oral Biol 2021; 125:105110. [PMID: 33774341 DOI: 10.1016/j.archoralbio.2021.105110] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 03/12/2021] [Accepted: 03/17/2021] [Indexed: 01/24/2023]
Abstract
OBJECTIVE This study aimed to provide further information on the exact mechanisms involved in the anti-inflammatory effect of low-intensity pulsed ultrasound (LIPUS) on rabbit temporomandibular joint osteoarthritis (TMJOA) on interleukin-6 (IL-6) production in subchondral bone, IL-6 production in IL-1β stimulated via inhibition of the TGF-β1/Smad3 pathway in mouse embryo osteoblast precursor (MC3T3-E1) cells. DESIGN Bilateral joints were injected with type II collagenase to establish TMJOA models in two male and four female rabbits. The left joint was continuously stimulated by LIPUS, while the right joint was treated with the power off in this model. One male and two female rabbits were used as normal healthy controls without treatment. The histological features of subchondral bone were examined by Safranin-O/Fast staining. Immunohistochemistry was conducted to evaluate IL-6 expression. Then, cells were stimulated by LIPUS with IL-1β. IL-6 expression and activity of the TGF-β1/Smad3 pathway were evaluated by Enzyme-linked immunosorbent assay (ELISA), Immunofluorescence and Western blotting, respectively. Specific inhibition of the TGF-β1/Smad3 pathway was conducted by transfecting with small interfering RNA (siRNA) of type II receptor (siTβRII). RESULTS LIPUS significantly ameliorated the production of IL-6 in vitro and in vivo. Its inhibitory effect on the production of IL-6 induced by IL-1β in MC3T3-E1 cells was partly reversed by siTβRII knockdown. CONCLUSIONS LIPUS inhibited IL-6 production by suppressing the TGF-β1/Smad3 pathway of subchondral bone in TMJOA. These data revealed the part of the pathways involved in the anti-inflammatory effect of LIPUS and provided a possible treatment strategy for TMJOA patients and other inflammatory diseases.
Collapse
Affiliation(s)
- Xin Yi
- Department of Oral Anatomy and Physiology, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Disease, Shenyang, 110002, China.
| | - Jie Liu
- Department of Science Experiment Center of China Medical University, Shenyang, 110122, China.
| | - Mo-Sha Cheng
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Disease, Shenyang, 110002, China.
| | - Qing Zhou
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Disease, Shenyang, 110002, China.
| |
Collapse
|
34
|
Huang HT, Cheng TL, Yang CD, Chang CF, Ho CJ, Chuang SC, Li JY, Huang SH, Lin YS, Shen HY, Yu TH, Kang L, Lin SY, Chen CH. Intra-Articular Injection of (-)-Epigallocatechin 3-Gallate (EGCG) Ameliorates Cartilage Degeneration in Guinea Pigs with Spontaneous Osteoarthritis. Antioxidants (Basel) 2021; 10:178. [PMID: 33530594 PMCID: PMC7910837 DOI: 10.3390/antiox10020178] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 01/17/2021] [Accepted: 01/20/2021] [Indexed: 12/18/2022] Open
Abstract
Osteoarthritis (OA) is the most prevalent joint disease that causes an enormous burden of disease worldwide. (-)-Epigallocatechin 3-gallate (EGCG) has been reported to reduce post-traumatic OA progression through its anti-inflammatory property. Aging is the most crucial risk factor of OA, and the majority of OA incidences are related to age and not trauma. In this study, we assess whether EGCG can ameliorate cartilage degradation in primary OA. In an in-vitro study, real-time PCR was performed to assess the expression of genes associated with human articular chondrocyte homeostasis. A spontaneously occurring OA model in guinea pigs was used to investigate the effect of EGCG in vivo. OA severity was evaluated using Safranin O staining and Osteoarthritis Research Society International (OARSI) scores, as well as by immunohistochemical (IHC) analysis to determine the protein level of type II collagen (Col II), matrix metalloproteinase 13 (MMP-13), and p16 ink4a in articular cartilage. In the in-vitro study, EGCG increased the gene expression of aggrecan and Col II and decreased the expression of interleukin-1, cyclooxygenase 2, MMP-13, alkaline phosphatase, Col X, and p16 Ink4a; EGCG treatment also attenuated the degraded cartilage with a lower OARSI score. Meanwhile, IHC results showed that EGCG exerted an anti-OA effect by reducing ECM degradation, cartilage inflammation, and cell senescence with a less-immunostained Col II, MMP-13, and p16 Ink4a. In conclusion, these findings suggest that EGCG may be a potential disease-modifying OA drug for the treatment of primary OA.
Collapse
Affiliation(s)
- Hsuan-Ti Huang
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan; (H.-T.H.); (T.-L.C.); (C.-J.H.); (S.-C.C.); (J.-Y.L.); (S.-H.H.); (Y.-S.L.); (H.-Y.S.); (T.-H.Y.)
- Department of Orthopedics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Regeneration Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Departments of Orthopedics, College of Medicine, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Department of Orthopedics, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung 80145, Taiwan
- Musculoskeletal Regeneration Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
| | - Tsung-Lin Cheng
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan; (H.-T.H.); (T.-L.C.); (C.-J.H.); (S.-C.C.); (J.-Y.L.); (S.-H.H.); (Y.-S.L.); (H.-Y.S.); (T.-H.Y.)
- Regeneration Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Musculoskeletal Regeneration Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Department of Physiology, College of Medicine, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
| | - Chung-Da Yang
- Graduate Institute of Animal Vaccine Technology, College of Veterinary Medicine, National Pingtung University of Science and Technology, Pingtung 91201, Taiwan;
| | - Chi-Fen Chang
- Department of Anatomy, School of Medicine, China Medical University, Taichung 40402, Taiwan;
| | - Cheng-Jung Ho
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan; (H.-T.H.); (T.-L.C.); (C.-J.H.); (S.-C.C.); (J.-Y.L.); (S.-H.H.); (Y.-S.L.); (H.-Y.S.); (T.-H.Y.)
- Department of Orthopedics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Regeneration Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Departments of Orthopedics, College of Medicine, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Department of Orthopedics, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung 80145, Taiwan
- Musculoskeletal Regeneration Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
| | - Shu-Chun Chuang
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan; (H.-T.H.); (T.-L.C.); (C.-J.H.); (S.-C.C.); (J.-Y.L.); (S.-H.H.); (Y.-S.L.); (H.-Y.S.); (T.-H.Y.)
- Department of Orthopedics, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung 80145, Taiwan
- Musculoskeletal Regeneration Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
| | - Jhong-You Li
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan; (H.-T.H.); (T.-L.C.); (C.-J.H.); (S.-C.C.); (J.-Y.L.); (S.-H.H.); (Y.-S.L.); (H.-Y.S.); (T.-H.Y.)
- Department of Orthopedics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Regeneration Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Departments of Orthopedics, College of Medicine, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Musculoskeletal Regeneration Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Department of Orthopedics, Kaohsiung Municipal Hsiao-Kang Hospital, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
| | - Shih-Hao Huang
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan; (H.-T.H.); (T.-L.C.); (C.-J.H.); (S.-C.C.); (J.-Y.L.); (S.-H.H.); (Y.-S.L.); (H.-Y.S.); (T.-H.Y.)
- Department of Orthopedics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Regeneration Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Departments of Orthopedics, College of Medicine, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Department of Orthopedics, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung 80145, Taiwan
| | - Yi-Shan Lin
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan; (H.-T.H.); (T.-L.C.); (C.-J.H.); (S.-C.C.); (J.-Y.L.); (S.-H.H.); (Y.-S.L.); (H.-Y.S.); (T.-H.Y.)
- Regeneration Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Musculoskeletal Regeneration Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
| | - Hsin-Yi Shen
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan; (H.-T.H.); (T.-L.C.); (C.-J.H.); (S.-C.C.); (J.-Y.L.); (S.-H.H.); (Y.-S.L.); (H.-Y.S.); (T.-H.Y.)
| | - Tsung-Han Yu
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan; (H.-T.H.); (T.-L.C.); (C.-J.H.); (S.-C.C.); (J.-Y.L.); (S.-H.H.); (Y.-S.L.); (H.-Y.S.); (T.-H.Y.)
| | - Lin Kang
- Department of Obstetrics and Gynecology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70457, Taiwan
| | - Sung-Yen Lin
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan; (H.-T.H.); (T.-L.C.); (C.-J.H.); (S.-C.C.); (J.-Y.L.); (S.-H.H.); (Y.-S.L.); (H.-Y.S.); (T.-H.Y.)
- Department of Orthopedics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Regeneration Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Departments of Orthopedics, College of Medicine, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Department of Orthopedics, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung 80145, Taiwan
- Musculoskeletal Regeneration Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
| | - Chung-Hwan Chen
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan; (H.-T.H.); (T.-L.C.); (C.-J.H.); (S.-C.C.); (J.-Y.L.); (S.-H.H.); (Y.-S.L.); (H.-Y.S.); (T.-H.Y.)
- Department of Orthopedics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Regeneration Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Departments of Orthopedics, College of Medicine, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Department of Orthopedics, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung 80145, Taiwan
- Musculoskeletal Regeneration Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Department of Healthcare Administration and Medical Informatics, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Institute of Medical Science and Technology, National Sun Yat-Sen University, Kaohsiung 80424, Taiwan
| |
Collapse
|
35
|
Salgado C, Jordan O, Allémann E. Osteoarthritis In Vitro Models: Applications and Implications in Development of Intra-Articular Drug Delivery Systems. Pharmaceutics 2021; 13:60. [PMID: 33466397 PMCID: PMC7824837 DOI: 10.3390/pharmaceutics13010060] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 12/23/2020] [Accepted: 12/28/2020] [Indexed: 02/08/2023] Open
Abstract
Osteoarthritis (OA) is a complex multi-target disease with an unmet medical need for the development of therapies that slow and potentially revert disease progression. Intra-articular (IA) delivery has seen a surge in osteoarthritis research in recent years. As local administration of molecules, this represents a way to circumvent systemic drug delivery struggles. When developing intra-articular formulations, the main goals are a sustained and controlled release of therapeutic drug doses, taking into account carrier choice, drug molecule, and articular joint tissue target. Therefore, the selection of models is critical when developing local administration formulation in terms of accurate outcome assessment, target and off-target effects and relevant translation to in vivo. The current review highlights the applications of OA in vitro models in the development of IA formulation by means of exploring their advantages and disadvantages. In vitro models are essential in studies of OA molecular pathways, understanding drug and target interactions, assessing cytotoxicity of carriers and drug molecules, and predicting in vivo behaviors. However, further understanding of molecular and tissue-specific intricacies of cellular models for 2D and 3D needs improvement to accurately portray in vivo conditions.
Collapse
Affiliation(s)
- Carlota Salgado
- School of Pharmaceutical Sciences, University of Geneva, 1211 Geneva, Switzerland; (C.S.); (O.J.)
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1211 Geneva, Switzerland
| | - Olivier Jordan
- School of Pharmaceutical Sciences, University of Geneva, 1211 Geneva, Switzerland; (C.S.); (O.J.)
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1211 Geneva, Switzerland
| | - Eric Allémann
- School of Pharmaceutical Sciences, University of Geneva, 1211 Geneva, Switzerland; (C.S.); (O.J.)
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1211 Geneva, Switzerland
| |
Collapse
|
36
|
Intra-Articular Injection of (-)-Epigallocatechin 3-Gallate to Attenuate Articular Cartilage Degeneration by Enhancing Autophagy in a Post-Traumatic Osteoarthritis Rat Model. Antioxidants (Basel) 2020; 10:antiox10010008. [PMID: 33374730 PMCID: PMC7824012 DOI: 10.3390/antiox10010008] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Revised: 12/05/2020] [Accepted: 12/14/2020] [Indexed: 12/13/2022] Open
Abstract
(-)-Epigallocatechin 3-gallate (EGCG) is the main active green tea catechin and has a wide variety of benefits for health. Post-traumatic osteoarthritis (PTOA) occurs as a consequence of joint injuries that commonly happen in the young population. In this study, we investigated the effects of EGCG on PTOA prevention by using the anterior cruciate ligament transection (ACLT)–OA model and further investigated the roles of autophagy in OA treatment. Our results showed that intra-articular injection of EGCG significantly improved the functional performances and decreased cartilage degradation. EGCG treatment attenuated the inflammation on synovial tissue and cartilage through less immunostained cyclooxygenase-2 and matrix metalloproteinase-13. We further noted EGCG may modulate the chondrocyte apoptosis by activation of the cytoprotective autophagy through reducing the expression of the mTOR and enhancing the expression of microtubule-associated protein light chain 3, beclin-1, and p62. In conclusion, intra-articular injection of EGCG after ACL injury inhibited the joint inflammation and cartilage degradation, thereby increasing joint function. EGCG treatment also reduced the chondrocyte apoptosis, possibly by activating autophagy. These findings suggested that EGCG may be a potential disease-modifying drug for preventing OA progression.
Collapse
|
37
|
Current Nanoparticle-Based Technologies for Osteoarthritis Therapy. NANOMATERIALS 2020; 10:nano10122368. [PMID: 33260493 PMCID: PMC7760945 DOI: 10.3390/nano10122368] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 11/17/2020] [Accepted: 11/24/2020] [Indexed: 12/21/2022]
Abstract
Osteoarthritis (OA) is a common chronic joint disease that is characterized by joint pain and stiffness, and limitation of motion and the major cause of disability, which reduces life quality of patients and brings a large economic burden to the family and society. Current clinical treatment is mostly limited to symptomatic treatment aimed at pain alleviation and functional improvement, rather than suppressing the progression of OA. Nanotechnology is a promising strategy for the treatment of OA. In this review, we summarize the current experimental progress that focuses on technologies such as liposomes, micelles, dendrimers, polymeric nanoparticles (PNPs), exosomes, and inorganic nanoparticles (NPs) for their potential treatment of OA.
Collapse
|