1
|
Cheng Q, Guo Q, Zhang X, Zhu Y, Liu C, Wang H, Zhu C, Ni L, Li B, Yang H. An "EVs-in-ECM" mimicking system orchestrates transcription and translation of RUNX1 for in-situ cartilage regeneration. Mater Today Bio 2025; 31:101569. [PMID: 40040797 PMCID: PMC11876752 DOI: 10.1016/j.mtbio.2025.101569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 02/06/2025] [Accepted: 02/08/2025] [Indexed: 03/06/2025] Open
Abstract
The self-repair ability of articular cartilage is limited, which is one of the most difficult diseases to treat clinically. Kartogenin (KGN) induces chondrogenesis by regulating RUNX1 mRNA translation and the small molecule compound TD-198946 (TD) promotes chondrogenic differentiation of mesenchymal stem cells (MSCs) through increasing the transcription of RUNX1 mRNA. GelMA hydrogel and liposomes are respectively similar to the extracellular matrix (ECM) and extracellular vesicles (EVs). So, we developed an "EVs-in-ECM" mimicking system by incorporating GelMA and KGN/TD-loaded liposomes to investigate the repair effects of cartilage defect. First, western-blot, RNA fluorescence in situ hybridization (FISH), cellular immuno-fluorescence, co-immuno-precipitation (CO-IP), and qRT-PCR techniques showed that KGN regulated RUNX1 mRNA expression, and then promote chondrogenic differentiation of MSCs. Second, the role of RUNX1 was amplified by orchestrating RUNX1 transcription and translation through TD-198946 (TD) and KGN respectively, and the synergistic effects of TD and KGN on chondrogenesis of MSCs in vitro were discovered. Finally, an "EVs-in-ECM" mimicking system was designed for in situ cartilage repair. When GelMA loaded with KGN and TD liposomes, the hydrogel (KGN + TD@ GelMA) showed biological functions by the continuously controlled release of KGN and TD while maintaining its porous structure and mechanical strength, which enhanced the chondrogenesis of MSCs in one system. The repair performance of "EVs-in-ECM" in vivo was assessed using the articular osteochondral defect model of rat. The implantation of KGN + TD@ GelMA hydrogels effectively exerted favorable osteochondral repair effects showing structures similar to the native tissue, and prevented chondrocyte hypertrophy. The study indicate that the "EVs-in-ECM" mimicking system can act as a highly efficient and potent scaffold for osteochondral defect regeneration.
Collapse
Affiliation(s)
- Qi Cheng
- Department of Orthopaedic Surgery, Orthopaedic Institute, The First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215006, China
- Orthopaedic Department, Xuzhou Central Hospital, No. 199, The Jiefang South Road, Xuzhou, 221009, Jiangsu, China
| | - Qianping Guo
- Department of Orthopaedic Surgery, Orthopaedic Institute, The First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215006, China
| | - Xiaoyu Zhang
- Department of Orthopaedic Surgery, Orthopaedic Institute, The First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215006, China
| | - Yuanchen Zhu
- Department of Orthopaedic Surgery, Orthopaedic Institute, The First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215006, China
| | - Chengyuan Liu
- Department of Orthopaedic Surgery, Orthopaedic Institute, The First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215006, China
| | - Huan Wang
- Department of Orthopaedic Surgery, Orthopaedic Institute, The First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215006, China
| | - Caihong Zhu
- Department of Orthopaedic Surgery, Orthopaedic Institute, The First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215006, China
| | - Li Ni
- Department of Orthopaedic Surgery, Orthopaedic Institute, The First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215006, China
| | - Bin Li
- Department of Orthopaedic Surgery, Orthopaedic Institute, The First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215006, China
- Medical 3D Printing Center, Orthopedic Institute, Department of Orthopedic Surgery, The First Affiliated Hospital, School of Biology and Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215006, China
| | - Huilin Yang
- Department of Orthopaedic Surgery, Orthopaedic Institute, The First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215006, China
| |
Collapse
|
2
|
Chihab S, Khan NM, Eng T, Doan T, Kaiser JM, Drissi H. Kartogenin Enhances Chondrogenic Differentiation of iPSC Derived MSCs (iMSCs) and Improves Outcomes in an Osteochondral Defect Model in Male Rats. J Orthop Res 2025; 43:870-880. [PMID: 39800942 DOI: 10.1002/jor.26040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 12/12/2024] [Accepted: 12/16/2024] [Indexed: 02/14/2025]
Abstract
Osteochondral defects (OCD) pose a significant clinical challenge due to the limited self-repair capacity of cartilage, leading to pain, joint dysfunction, and progression to osteoarthritis. Cellular implantations of adult mesenchymal stem cells (MSCs) enhanced with treatment of factors, such as small molecule Kartogenin (KGN) to promote chondrogenic differentiation, are promising but these cells often encounter hypertrophy during differentiation, compromising long-term stability. Induced pluripotent stem cell-derived MSCs (iMSCs) offer greater proliferative and differentiation capacity than MSCs and may provide a superior source of cells for cartilage repair. We hypothesized that treatment of iMSCs with TGFβ3 and KGN would enhance chondrogenic differentiation and that implanting these pellets into a rat OCD model would promote de novo cartilage regeneration and reduce pain behavior. We pellet cultured iMSCs derived from articular chondrocytes and treated with various conditions of TGFβ3 and KGN. We then assessed the in vivo performance of the pellets using a trochlear osteochondral defect in male Lewis rats. Co-treatment of iMSC pellets with TGFβ3 and KGN showed more pronounced chondrogenic differentiation than sequential treatment and exhibited stronger expression of chondrogenic genes. Implantation of the TGFβ3/KGN-treated iMSC pellets into OCD resulted in modest repair, as observed via gross morphology, effectively prevented the onset of joint hyperalgesia, and helped to maintain normal gait out to 12 weeks post-implantation compared to untreated OCD rats. Our study highlights the potential of KGN to enhance iMSC pellet chondrogenesis, offering a scaffold-free, cell-based therapy that could simplify clinical translation and improve outcomes for patients with cartilage injuries.
Collapse
Affiliation(s)
- Samir Chihab
- Atlanta Veteran's Affairs Medical Center, Decatur, Georgia, USA
- Department of Orthopaedics, Emory University, Atlanta, Georgia, USA
| | - Nazir M Khan
- Atlanta Veteran's Affairs Medical Center, Decatur, Georgia, USA
- Department of Orthopaedics, Emory University, Atlanta, Georgia, USA
| | - Tracy Eng
- Atlanta Veteran's Affairs Medical Center, Decatur, Georgia, USA
- Department of Orthopaedics, Emory University, Atlanta, Georgia, USA
| | - Thanh Doan
- Atlanta Veteran's Affairs Medical Center, Decatur, Georgia, USA
- Department of Orthopaedics, Emory University, Atlanta, Georgia, USA
| | - Jarred M Kaiser
- Atlanta Veteran's Affairs Medical Center, Decatur, Georgia, USA
- Department of Orthopaedics, Emory University, Atlanta, Georgia, USA
| | - Hicham Drissi
- Atlanta Veteran's Affairs Medical Center, Decatur, Georgia, USA
- Department of Orthopaedics, Emory University, Atlanta, Georgia, USA
| |
Collapse
|
3
|
Zhu Y, Cheng Q, Liu C, Wang H, Zhu C, Qian J, Hu H, Li B, Guo Q, Shi J. Integrated GelMA and liposome composite hydrogel with effective coupling of angiogenesis and osteogenesis for promoting bone regeneration. Int J Biol Macromol 2025; 297:139835. [PMID: 39824404 DOI: 10.1016/j.ijbiomac.2025.139835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Revised: 12/12/2024] [Accepted: 01/11/2025] [Indexed: 01/20/2025]
Abstract
In clinical scenarios, bone defects stemming from trauma, infections, degenerative diseases, or hereditary conditions necessitate considerable bone grafts. Researchers ardently focus on creating diverse biomaterials to expedite and enhance these intricate restorative processes. These biomaterials play a pivotal role in aiding osteogenesis and angiogenesis factors for reconstructing stable, fully developed bone tissue. We observed the utilization of Desferoxamine (DFO) facilitated angiogenesis, thereby enabling Kartogenin (KGN) to activate the β-catenin/Runx-2 pathway. Our study introduces a composite hydrogel loaded with KGN and DFO via liposomes to enhance the coupling of angiogenesis and osteogenesis. Within this composite hydrogel system, KGN and DFO undergo effective release. This controlled release substantially promotes a conducive microenvironment for angiogenesis and osteogenesis. Our in vitro studies provide compelling evidence of the synergistic impact between KGN and DFO on osteogenic processes. Moreover, the composite hydrogel exhibits the capability to enhance the expression of proteins and genes associated with both angiogenesis and osteogenesis. In rat skull defect model, the composite hydrogel notably stimulates vascularization and osteogenic differentiation without infection or mortality. In summary, results underscore the potential of this composite hydrogel as an alternative to autografts for bone defect repair, offering a promising approach for future clinical and regenerative applications.
Collapse
Affiliation(s)
- Yuanchen Zhu
- Medical 3D Printing Center, Orthopedic Institute, Department of Orthopedic Surgery, The First Affiliated Hospital, School of Biology and Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu 215000, PR China; Department of Orthopedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, PR China
| | - Qi Cheng
- Medical 3D Printing Center, Orthopedic Institute, Department of Orthopedic Surgery, The First Affiliated Hospital, School of Biology and Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu 215000, PR China; Department of Orthopedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, PR China
| | - Chengyuan Liu
- Medical 3D Printing Center, Orthopedic Institute, Department of Orthopedic Surgery, The First Affiliated Hospital, School of Biology and Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu 215000, PR China
| | - Huan Wang
- Medical 3D Printing Center, Orthopedic Institute, Department of Orthopedic Surgery, The First Affiliated Hospital, School of Biology and Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu 215000, PR China
| | - Caihong Zhu
- Medical 3D Printing Center, Orthopedic Institute, Department of Orthopedic Surgery, The First Affiliated Hospital, School of Biology and Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu 215000, PR China
| | - Jin Qian
- Medical 3D Printing Center, Orthopedic Institute, Department of Orthopedic Surgery, The First Affiliated Hospital, School of Biology and Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu 215000, PR China; Department of Orthopedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, PR China
| | - Hanfeng Hu
- Medical 3D Printing Center, Orthopedic Institute, Department of Orthopedic Surgery, The First Affiliated Hospital, School of Biology and Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu 215000, PR China; Department of Orthopedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, PR China
| | - Bin Li
- Medical 3D Printing Center, Orthopedic Institute, Department of Orthopedic Surgery, The First Affiliated Hospital, School of Biology and Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu 215000, PR China.
| | - Qianping Guo
- Medical 3D Printing Center, Orthopedic Institute, Department of Orthopedic Surgery, The First Affiliated Hospital, School of Biology and Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu 215000, PR China.
| | - Jinhui Shi
- Department of Orthopedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, PR China.
| |
Collapse
|
4
|
Sarker M, Park S, Kumar V, Lee CH. Micro-thin hydrogel coating integrated in 3D printing for spatiotemporal delivery of bioactive small molecules. Biofabrication 2024; 17:015019. [PMID: 39437834 PMCID: PMC11552100 DOI: 10.1088/1758-5090/ad89fe] [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: 07/03/2024] [Revised: 09/17/2024] [Accepted: 10/22/2024] [Indexed: 10/25/2024]
Abstract
Three-dimensional (3D) printing incorporated with controlled delivery is an effective tool for complex tissue regeneration. Here, we explored a new strategy for spatiotemporal delivery of bioactive cues by establishing a precise-controlled micro-thin coating of hydrogel carriers on 3D-printed scaffolds. We optimized the printing parameters for three hydrogel carriers, fibrin cross-linked with genipin, methacrylate hyaluronic acid, and multidomain peptides, resulting in homogenous micro-coating on desired locations in 3D printed polycaprolactone microfibers at each layer. Using the optimized multi-head printing technique, we successfully established spatial-controlled micro-thin coating of hydrogel layers containing profibrogenic small molecules (SMs), Oxotremorine M and PPBP maleate, and a chondrogenic cue, Kartogenin. The delivered SMs showed sustained releases up to 28 d and guided regional differentiation of mesenchymal stem cells, thus leading to fibrous and cartilaginous tissue matrix formation at designated scaffold regionsin vitroandin vivo. Our micro-coating of hydrogel carriers may serve as an efficient approach to achieve spatiotemporal delivery of various bioactive cues through 3D printed scaffolds for engineering complex tissues.
Collapse
Affiliation(s)
- Md Sarker
- Biomedical Engineering, University of Maryland Eastern Shore, 30665 Student Services Center, Princess Anne, MD 21853, United States of America
| | - Soomin Park
- Center for Dental and Craniofacial Research, College of Dental Medicine, Columbia University Medical Center, 630 W. 168th Street, VC12-210, New York, NY 10032, United States of America
| | - Vivek Kumar
- Department of Bio-Medical Engineering, New Jersey Institute of Technology, 138 Warren St., Room 316, Newark, NJ 07102, United States of America
| | - Chang H Lee
- Center for Dental and Craniofacial Research, College of Dental Medicine, Columbia University Medical Center, 630 W. 168th Street, VC12-210, New York, NY 10032, United States of America
| |
Collapse
|
5
|
Liu X, Liu P, Li H, Cen Y, Jiang G, Zhang W, Tian K, Wang X. Application of kartogenin for the treatment of cartilage defects: current practice and future directions. RSC Adv 2024; 14:33206-33222. [PMID: 39434994 PMCID: PMC11492430 DOI: 10.1039/d4ra06558a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 10/14/2024] [Indexed: 10/23/2024] Open
Abstract
Osteoarthritis and sports injuries often lead to cartilage defects. How to promote its repair and rebuild the smooth cartilage surface has been a hot spot of research in recent years. Kartogenin (KGN), a small molecule discovered in recent years, has been shown to promote the proliferation and chondrogenic differentiation of mesenchymal stem cells (MSCs). As more and more studies have been conducted on KGN, its mechanism of action has been gradually revealed. However, KGN is insoluble in water and therefore easily removed by body fluids. In order to address such issues, a number of systems for efficient intra-articular delivery of KGN have been developed. In addition, due to the complex pathology of cartilage repair, KGN is often used in combination with other drugs to target different stages. In addition, with the rapid development of tissue engineering, scholars have combined KGN with various scaffolds by physical or chemical methods. In this paper, we firstly introduce the general properties of KGN followed by a review of the latest advances in the intra-articular delivery modes of KGN. Finally, we discuss the prospects for the application of KGN in cartilage regeneration, which is aimed at providing a new idea and target for the treatment of cartilage defects.
Collapse
Affiliation(s)
- Xuemiao Liu
- First Affiliated Hospital of Dalian Medical University Dalian 116001 China
- Beijing National Laboratory for Molecular Sciences State Key Laboratory of Polymer Physics and Chemistry Institute of Chemistry Chinese Academy of Sciences Beijing 100190 China
| | - Pengfei Liu
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University Beijing 100191 China
| | - Han Li
- Xiongan Xuanwu Hospital Hebei 071700 China
| | - Ying Cen
- First Affiliated Hospital of Dalian Medical University Dalian 116001 China
| | - Guichun Jiang
- Liaoning Cancer Hospital & Institute, Clinical Skills Training Center Shenyang 110042 China
| | - Weiguo Zhang
- First Affiliated Hospital of Dalian Medical University Dalian 116001 China
| | - Kang Tian
- First Affiliated Hospital of Dalian Medical University Dalian 116001 China
| | - Xing Wang
- Beijing National Laboratory for Molecular Sciences State Key Laboratory of Polymer Physics and Chemistry Institute of Chemistry Chinese Academy of Sciences Beijing 100190 China
| |
Collapse
|
6
|
Jiang P, Hu K, Jin L, Luo Z. A brief review of current treatment options for osteoarthritis including disease-modifying osteoarthritis drugs (DMOADs) and novel therapeutics. Ann Med Surg (Lond) 2024; 86:4042-4048. [PMID: 38989236 PMCID: PMC11230824 DOI: 10.1097/ms9.0000000000002214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 05/13/2024] [Indexed: 07/12/2024] Open
Abstract
Osteoarthritis (OA) is a chronic disorder caused by degenerative changes in articular cartilage, which are mainly manifests as degeneration of cartilage, subchondral bone remodeling, as well as synovial inflammation. Over the next few decades, OA and its burden will continue to increase worldwide, posing a major public health challenge for the foreseeable future. Treatment for OA includes non-pharmacological, pharmacological, and surgical treatments. Existing conservative treatments and joint surgery can only alleviate the symptoms and cannot be cured, so new therapies for OA are urgently needed. Since advances in the understanding of OA pathophysiology, researchers have identified some potential therapeutic targets against degeneration of cartilage, subchondral bone remodeling and synovial inflammation, enabling development of the disease-modifying OA drugs (DMOADs). Additionally, a number of new technologies are also being investigated for treating OA, such as RNA interference (RNAi), CRISPR/Cas9 and PROTAC. The goal of this review is to describe the current development status of DMOADs and to discuss the potential of emerging therapeutic approaches for treating OA, thus providing a reference for OA treatments.
Collapse
Affiliation(s)
| | | | - Liang Jin
- The Second Affiliated Hospital Zhejiang University School of Medicine Changxing Campus, Taihu Middle Road, Changxing County, Zhejiang Province, China
| | - Zhicheng Luo
- The Second Affiliated Hospital Zhejiang University School of Medicine Changxing Campus, Taihu Middle Road, Changxing County, Zhejiang Province, China
| |
Collapse
|
7
|
Zhu J, Lu Y, Shan Y, Yuan L, Wu Q, Shen Z, Sun F, Shi H. Global Bibliometric and Visualized Analysis of Tracheal Tissue Engineering Research. TISSUE ENGINEERING. PART B, REVIEWS 2024; 30:198-216. [PMID: 37658839 DOI: 10.1089/ten.teb.2023.0129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
The development of tracheal tissue engineering (TTE) has seen a rapid growth in recent years. The purpose of this study was to investigate the global status, trends, and hotspots of TTE research based on bibliometrics and visualization analysis. Publications related to TTE were retrieved and included in the Web of Science Core Collection. VOSviewer and CiteSpace were used to generate knowledge maps. Six hundred fifty-five publications were identified, and the quantity of the annual publications worldwide was on the increase. International collaboration is a widespread reality. The United States led the world in the field of trachea tissue engineering, whereas University College London was the institution with the greatest contribution. In addition, Biomaterials had a great influence in this field, attracting the largest number of papers. Moreover, the topics of TTE research largely concentrated on the biomechanical scaffold preparation, the vascularization and epithelialization of scaffold, the tracheal cartilage regeneration, and the tissue-engineered tracheal transplantation. And the research on the application of decellularization and 3D printing for the construction of a tissue-engineered trachea was likely to receive more widespread attention in the future. Impact statement In recent years, tracheal tissue engineering (TTE) has experienced rapid growth. In this study, we investigated the worldwide status and trends of TTE research, and revealed the countries, institutions, journals, and authors that had made significant contributions to the field of TTE. Moreover, the possible research hotspots in the future were predicted. According to our research, researchers can gain a better understanding of the trends in this field, and stay informed of the most current research by tracking key journals, institutions, and authors.
Collapse
Affiliation(s)
- Jianwei Zhu
- Clinical Medical College, Yangzhou University, Yangzhou, China
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, China
| | - Yi Lu
- Clinical Medical College, Yangzhou University, Yangzhou, China
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, China
| | - Yibo Shan
- Clinical Medical College, Yangzhou University, Yangzhou, China
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, China
| | - Lei Yuan
- Clinical Medical College, Yangzhou University, Yangzhou, China
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, China
| | - Qiang Wu
- Clinical Medical College, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, China
- The Second Xiangya Hospital, Central South University, Changsha, China
| | - Zhiming Shen
- Clinical Medical College, Yangzhou University, Yangzhou, China
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, China
| | - Fei Sun
- Clinical Medical College, Yangzhou University, Yangzhou, China
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, China
| | - Hongcan Shi
- Clinical Medical College, Yangzhou University, Yangzhou, China
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, China
- The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
8
|
Lv H, Liu Y, Lu D, Wang Y. Kartogenin-loaded polyvinyl alcohol/nano-hydroxyapatite composite hydrogel promotes tendon-bone healing in rabbits after anterior cruciate ligament reconstruction. J Biomed Mater Res A 2024; 112:180-192. [PMID: 37694883 DOI: 10.1002/jbm.a.37605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 06/20/2023] [Accepted: 08/22/2023] [Indexed: 09/12/2023]
Abstract
Accumulating evidence supports the role of cartilage tissue engineering in cartilage defect repair, but the biological function has yet to be fully explained. In this work, kartogenin (KGN), an emerging chondroinductive nonprotein small molecule, was incorporated into a composite hydrogel of polyvinyl alcohol/nano-hydroxyapatite (PVA/n-HA) to fabricate an appropriate microenvironment for tendon-bone healing after anterior cruciate ligament (ACL) reconstruction. KGN/PVA/n-HA composite hydrogel scaffolds were prepared by in situ synthesis and physical adsorption, followed by characterization under a scanning electron microscope. The scaffolds were transplanted into healthy New Zealand White (NZW) rabbits. It was confirmed that KGN/PVA/n-HA scaffolds were successfully prepared and exhibited good supporting properties and excellent biocompatibility. Unilateral ACL reconstruction was constructed with tendon autograft in NZW rabbits, and the morphology and diameter of collagen fiber were analyzed. The scaffolds were shown to promote ACL growth and collagen fiber formation. Furthermore, microcomputerized tomography analysis and bone formation histology were performed to detect new bone formation. KGN/PVA/n-HA scaffolds effectively alleviated cartilage damage and prevented the occurrence of osteoarthritis. Meanwhile, ligament-bone healing and bone formation were observed in the presence of KGN/PVA/n-HA scaffolds. In conclusion, these results suggest that the KGN/PVA/n-HA scaffolds can facilitate tendon-bone healing after ACL reconstruction and might be considered novel hydrogel biomaterials in cartilage tissue engineering.
Collapse
Affiliation(s)
- Hao Lv
- Jinan Central Hospital, Jinan, People's Republic of China
| | - Yaobo Liu
- Jinan Central Hospital, Jinan, People's Republic of China
| | - Duyi Lu
- Jinan Central Hospital, Jinan, People's Republic of China
| | - Yuanrui Wang
- Jinan Central Hospital, Jinan, People's Republic of China
| |
Collapse
|
9
|
Gao J, Pei H, Lv F, Niu X, You Y, He L, Hu S, Shah KM, Liu M, Chen Y, Du B, Xiong H, Luo J. JD-312 - A novel small molecule that facilitates cartilage repair and alleviates osteoarthritis progression. J Orthop Translat 2024; 44:60-71. [PMID: 38269355 PMCID: PMC10805627 DOI: 10.1016/j.jot.2023.11.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 08/12/2023] [Accepted: 11/21/2023] [Indexed: 01/26/2024] Open
Abstract
Background The chondrogenic differentiation of mesenchymal stem cells (MSCs) to enhance cartilage repair and regeneration is a promising strategy to alleviate osteoarthritis (OA) progression. Method The potency of JD-312 in inducing chondrogenic differentiation of MSCs was assessed and verified. The efficacy of JD-312-treated MSCs was evaluated using a Sprague-Dawley rat DMM model. Additionally, the capacity of JD-312 to successfully recruit bone marrow-derived mesenchymal stem cells (BMSCs) for the treatment of OA in vitro was confirmed via intra-articular injection. The repair status of the articular cartilage was analyzed in vivo through histological examination. Result In this study, we identify JD-312 as a novel non-toxic small molecule that can promote chondrogenic differentiation in human umbilical cord-derived MSCs (hUCMSCs) and human bone marrow MSCS (hBMSCs) in vitro. We also show that transient differentiation of MSCs with JD-312 prior to in vivo administration remarkably improves the regeneration of cartilage and promotes Col2a1 and Acan expression in rat models of DMM, in comparison to kartogenin (KGN) pre-treatment or MSCs alone. Furthermore, direct intra-articular injection of JD-312 in murine model of OA showed reduced loss of articular cartilage and improved pain parameters. Lastly, we identified that the effects of JD-312 are at least in part mediated via upregulation of genes associated with the focal adhesion, PI3K-Akt signaling and the ECM-receptor interaction pathways, and specifically cartilage oligomeric matrix protein (COMP) may play a vital role. Conclusion Our study demonstrated that JD-312 showed encouraging repair effects for OA in vivo. The translational potential of this article Together, our findings demonstrate that JD-312 is a promising new therapeutic molecule for cartilage regeneration with clinical potential.
Collapse
Affiliation(s)
- Jingduo Gao
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, PR China
| | - Haixiang Pei
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, PR China
- Institute for Advanced Study, Shenzhen University and Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen, PR China
| | - Fang Lv
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, PR China
| | - Xin Niu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, PR China
| | - Yu You
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, PR China
| | - Liang He
- Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Tongji University School of Medicine, Shanghai, PR China
| | - Shijia Hu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, PR China
| | - Karan M. Shah
- Department of Oncology and Metabolism, The Medical School, The University of Sheffield, Sheffield, United Kingdom
| | - Mingyao Liu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, PR China
| | - Yihua Chen
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, PR China
| | - Bing Du
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, PR China
| | - Hai Xiong
- Institute for Advanced Study, Shenzhen University and Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen, PR China
| | - Jian Luo
- Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Tongji University School of Medicine, Shanghai, PR China
| |
Collapse
|
10
|
Chen P, Liao X. Kartogenin delivery systems for biomedical therapeutics and regenerative medicine. Drug Deliv 2023; 30:2254519. [PMID: 37665332 PMCID: PMC10478613 DOI: 10.1080/10717544.2023.2254519] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 06/14/2023] [Accepted: 08/21/2023] [Indexed: 09/05/2023] Open
Abstract
Kartogenin, a small and heterocyclic molecule, has emerged as a promising therapeutic agent for incorporation into biomaterials, owing to its unique physicochemical and biological properties. It holds potential for the regeneration of cartilage-related tissues in various common conditions and injuries. Achieving sustained release of kartogenin through appropriate formulation and efficient delivery systems is crucial for modulating cell behavior and tissue function. This review provides an overview of cutting-edge kartogenin-functionalized biomaterials, with a primarily focus on their design, structure, functions, and applications in regenerative medicine. Initially, we discuss the physicochemical properties and biological functions of kartogenin, summarizing the underlying molecular mechanisms. Subsequently, we delve into recent advancements in nanoscale and macroscopic materials for the carriage and delivery of kartogenin. Lastly, we address the opportunities and challenges presented by current biomaterial developments and explore the prospects for their application in tissue regeneration. We aim to enhance the generation of insightful ideas for the development of kartogenin delivery materials in the field of biomedical therapeutics and regenerative medicine by providing a comprehensive understanding of common preparation methods.
Collapse
Affiliation(s)
- Peixing Chen
- Chongqing Key Laboratory of Nano/Micro Composite Materials and Devices, School of Metallurgy and Materials Engineering, Chongqing University of Science and Technology, Chongqing, China
- Chongqing Engineering Laboratory of Nano/Micro Biomedical Detection Technology, School of Metallurgy and Materials Engineering, Chongqing University of Science and Technology, Chongqing, China
| | - Xiaoling Liao
- Chongqing Key Laboratory of Nano/Micro Composite Materials and Devices, School of Metallurgy and Materials Engineering, Chongqing University of Science and Technology, Chongqing, China
- Chongqing Engineering Laboratory of Nano/Micro Biomedical Detection Technology, School of Metallurgy and Materials Engineering, Chongqing University of Science and Technology, Chongqing, China
| |
Collapse
|
11
|
Kang J, Li Y, Qin Y, Huang Z, Wu Y, Sun L, Wang C, Wang W, Feng G, Qi Y. In Situ Deposition of Drug and Gene Nanoparticles on a Patterned Supramolecular Hydrogel to Construct a Directionally Osteochondral Plug. NANO-MICRO LETTERS 2023; 16:18. [PMID: 37975889 PMCID: PMC10656386 DOI: 10.1007/s40820-023-01228-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 09/20/2023] [Indexed: 11/19/2023]
Abstract
The integrated repair of bone and cartilage boasts advantages for osteochondral restoration such as a long-term repair effect and less deterioration compared to repairing cartilage alone. Constructing multifactorial, spatially oriented scaffolds to stimulate osteochondral regeneration, has immense significance. Herein, targeted drugs, namely kartogenin@polydopamine (KGN@PDA) nanoparticles for cartilage repair and miRNA@calcium phosphate (miRNA@CaP) NPs for bone regeneration, were in situ deposited on a patterned supramolecular-assembled 2-ureido-4 [lH]-pyrimidinone (UPy) modified gelation hydrogel film, facilitated by the dynamic and responsive coordination and complexation of metal ions and their ligands. This hydrogel film can be rolled into a cylindrical plug, mimicking the Haversian canal structure of natural bone. The resultant hydrogel demonstrates stable mechanical properties, a self-healing ability, a high capability for reactive oxygen species capture, and controlled release of KGN and miR-26a. In vitro, KGN@PDA and miRNA@CaP promote chondrogenic and osteogenic differentiation of mesenchymal stem cells via the JNK/RUNX1 and GSK-3β/β-catenin pathways, respectively. In vivo, the osteochondral plug exhibits optimal subchondral bone and cartilage regeneration, evidenced by a significant increase in glycosaminoglycan and collagen accumulation in specific zones, along with the successful integration of neocartilage with subchondral bone. This biomaterial delivery approach represents a significant toward improved osteochondral repair.
Collapse
Affiliation(s)
- Jiawei Kang
- Department of Orthopaedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, 310009, Zhejiang Province, People's Republic of China
| | - Yaping Li
- College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, 310027, People's Republic of China
- ZJU-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou, 311215, Zhejiang, People's Republic of China
| | - Yating Qin
- College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, 310027, People's Republic of China
- ZJU-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou, 311215, Zhejiang, People's Republic of China
| | - Zhongming Huang
- The Affiliated Nanhua Hospital, Orthopedic Research Centre, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China
| | - Yifan Wu
- Department of Orthopaedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, 310009, Zhejiang Province, People's Republic of China
| | - Long Sun
- Department of Radiology, Jining No. 1 People's Hospital, Jining Medical University, Jining, 272000, Shandong, People's Republic of China
| | - Cong Wang
- Department of Orthopaedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, 310009, Zhejiang Province, People's Republic of China
| | - Wei Wang
- College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, 310027, People's Republic of China.
- ZJU-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou, 311215, Zhejiang, People's Republic of China.
| | - Gang Feng
- Department of Orthopaedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, 310009, Zhejiang Province, People's Republic of China.
| | - Yiying Qi
- Department of Orthopaedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, 310009, Zhejiang Province, People's Republic of China.
| |
Collapse
|
12
|
Yu L, Cavelier S, Hannon B, Wei M. Recent development in multizonal scaffolds for osteochondral regeneration. Bioact Mater 2023; 25:122-159. [PMID: 36817819 PMCID: PMC9931622 DOI: 10.1016/j.bioactmat.2023.01.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 12/30/2022] [Accepted: 01/14/2023] [Indexed: 02/05/2023] Open
Abstract
Osteochondral (OC) repair is an extremely challenging topic due to the complex biphasic structure and poor intrinsic regenerative capability of natural osteochondral tissue. In contrast to the current surgical approaches which yield only short-term relief of symptoms, tissue engineering strategy has been shown more promising outcomes in treating OC defects since its emergence in the 1990s. In particular, the use of multizonal scaffolds (MZSs) that mimic the gradient transitions, from cartilage surface to the subchondral bone with either continuous or discontinuous compositions, structures, and properties of natural OC tissue, has been gaining momentum in recent years. Scrutinizing the latest developments in the field, this review offers a comprehensive summary of recent advances, current hurdles, and future perspectives of OC repair, particularly the use of MZSs including bilayered, trilayered, multilayered, and gradient scaffolds, by bringing together onerous demands of architecture designs, material selections, manufacturing techniques as well as the choices of growth factors and cells, each of which possesses its unique challenges and opportunities.
Collapse
Affiliation(s)
- Le Yu
- Department of Chemical and Biomolecular Engineering, Ohio University, Athens, OH, 45701, USA
| | - Sacha Cavelier
- Department of Chemical and Biomolecular Engineering, Ohio University, Athens, OH, 45701, USA
| | - Brett Hannon
- Biomedical Engineering Program, Ohio University, Athens, OH, 45701, USA
| | - Mei Wei
- Biomedical Engineering Program, Ohio University, Athens, OH, 45701, USA
- Department of Mechanical Engineering, Ohio University, Athens, OH, 45701, USA
| |
Collapse
|
13
|
Qiu J, Hua B, Ye X, Liu X. Intra-articular injection of kartogenin promotes fibrocartilage stem cell chondrogenesis and attenuates temporomandibular joint osteoarthritis progression. Front Pharmacol 2023; 14:1159139. [PMID: 37361231 PMCID: PMC10288139 DOI: 10.3389/fphar.2023.1159139] [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: 02/05/2023] [Accepted: 06/01/2023] [Indexed: 06/28/2023] Open
Abstract
Introduction: Kartogenin (KGN) is a small-molecule compound that has been reported to improve the chondrogenic differentiation of mesenchymal stem cells in vitro and to alleviate knee joint osteoarthritis in animal models. However, whether KGN has any effect on temporomandibular joint osteoarthritis (TMJOA) remains unclear. Methods: We first performed partial temporomandibular joint (TMJ) discectomy to induce TMJOA in rats. Histological analysis, tartrate-resistant acid phosphatase staining, and immunohistochemistry were used to assess the therapeutic effect of KGN on TMJOA in vivo. CCK8 and pellet cultures were used to determine whether KGN treatment could promote the proliferation and differentiation of FCSCs in vitro. Quantitative real-time polymerase chain reaction (qRT-PCR) was conducted to determine the expression of aggrecan, Col2a1, and Sox9 in FCSCs. Furthermore, we performed western blot to analysis the effect of KGN treatment on the expression of Sox9 and Runx2 in FCSCs. Results and discussion: Histological analysis, tartrate-resistant acid phosphatase staining, and immunohistochemistry showed that intra-articular injection of KGN attenuated cartilage degeneration and subchondral bone resorption in vivo. Further analyses of the underlying mechanisms revealed that KGN enhanced chondrocyte proliferation, increased the number of cells in both superficial and proliferative zones of TMJ condylar cartilage in vivo, enhanced the proliferation and chondrogenic differentiation of fibrocartilage stem cells (FCSCs), and upregulated the expression of chondrogenesis-related factors in vitro. Collectively, in our study, KGN was shown to promote FCSC chondrogenesis and restore TMJ cartilage, suggesting that KGN injections might be a potential treatment for TMJOA.
Collapse
|
14
|
Liu Z, Luo Z, Yu H, Zhao E, Guo J, Mou P, Chen A, Chen J, Zhou Z, Zeng W. Near-infrared light-controlled kartogenin delivery of multifunctional Prussian blue nanocomposites for cartilage defect repair. NANOSCALE 2023; 15:9076-9093. [PMID: 37129436 DOI: 10.1039/d3nr00205e] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Articular cartilage injury repair remains a challenge for clinicians and researchers. Mesenchymal stem cells (MSCs) have multiple differentiation potentials and can be induced to differentiate into the chondrogenic lineage for cartilage defect repair; however, the insufficient capacity of chondrogenic differentiation and excess reactive oxygen species (ROS)-mediated oxidative stress, which always lead to differentiation into hypertrophic chondrocytes, still need to be resolved. Accordingly, kartogenin (KGN), which can promote chondrogenic differentiation of MSCs, has shown promise in promoting infected cartilage repair. However, realizing controllable release to prolong its action time and avoid hypertrophic differentiation is critical. We herein developed a mesoporous Prussian blue nanoparticle (mPB)-based near-infrared (NIR) light-responsive controlled nanosystem. KGN was encapsulated in temperature-stimulated responsive phase change materials (PCMs), which were used as excellent gating materials (KGN-PCM@mPBs). In addition, the mPBs could efficiently scavenge ROS by their enzyme-like antioxidative activities. Our study demonstrates that the nanocomposites could efficiently promote chondrogenic differentiation and successfully inhibit the hypertrophic differentiation of MSCs. By intra-articular injection of KGN-PCM@mPBs and NIR-triggered precisely controlled release, satisfactory cartilage repair effects can be achieved in a rat chondral defect model. Thus, this constructed NIR-mediated KGN-PCM@mPB nanoplatform may represent an effective cartilage repair strategy with satisfactory biosafety in clinical applications.
Collapse
Affiliation(s)
- Zunhan Liu
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, 610041, China.
- Department of Sports Medicine Center, State Key Laboratory of Trauma, Burn and Combined Injury, the First Affiliated Hospital of the Army Military Medical University, Chongqing, 400038, China
| | - Zhenyu Luo
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, 610041, China.
| | - Haoda Yu
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, 610041, China.
| | - Enze Zhao
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, 610041, China.
| | - Junfeng Guo
- Department of Sports Medicine Center, State Key Laboratory of Trauma, Burn and Combined Injury, the First Affiliated Hospital of the Army Military Medical University, Chongqing, 400038, China
| | - Ping Mou
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, 610041, China.
| | - Anjing Chen
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, 610041, China.
| | - Jiali Chen
- West China School of Nursing, Sichuan University/Department of Orthopedics, West China Hospital, Sichuan University Chengdu, 610041, P.R. China
| | - Zongke Zhou
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, 610041, China.
| | - Weinan Zeng
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, 610041, China.
| |
Collapse
|
15
|
Sahu N, Grandi FC, Bhutani N. A single-cell mass cytometry platform to map the effects of preclinical drugs on cartilage homeostasis. JCI Insight 2022; 7:160702. [PMID: 36194485 PMCID: PMC9744259 DOI: 10.1172/jci.insight.160702] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 09/12/2022] [Indexed: 02/04/2023] Open
Abstract
No disease-modifying drug exists for osteoarthritis (OA). Despite success in animal models, candidate drugs continue to fail in clinical trials owing to the unmapped interpatient heterogeneity and disease complexity. We used a single-cell platform based on cytometry by time-of-flight (cyTOF) to precisely outline the effects of candidate drugs on human OA chondrocytes. OA chondrocytes harvested from patients undergoing total knee arthroplasty were treated with 2 drugs, an NF-κB pathway inhibitor, BMS-345541, and a chondroinductive small molecule, kartogenin, that showed preclinical success in animal models for OA. cyTOF conducted with 30 metal isotope-labeled antibodies parsed the effects of the drugs on inflammatory, senescent, and chondroprogenitor cell populations. The NF-κB pathway inhibition decreased the expression of p-NF-κB, HIF2A, and inducible NOS in multiple chondrocyte clusters and significantly depleted 4 p16ink4a-expressing senescent populations, including NOTCH1+STRO1+ chondroprogenitor cells. While kartogenin also affected select p16ink4a-expressing senescent clusters, there was a less discernible effect on chondroprogenitor cell populations. Overall, BMS-345541 elicited a uniform drug response in all patients, while only a few responded to kartogenin. These studies demonstrate that a single-cell cyTOF-based drug screening platform can provide insights into patient response assessment and patient stratification.
Collapse
|
16
|
Elder S, Roberson JG, Warren J, Lawson R, Young D, Stokes S, Ross MK. Evaluation of Electrospun PCL-PLGA for Sustained Delivery of Kartogenin. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27123739. [PMID: 35744864 PMCID: PMC9229984 DOI: 10.3390/molecules27123739] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 06/06/2022] [Accepted: 06/08/2022] [Indexed: 01/01/2023]
Abstract
In this study, kartogenin was incorporated into an electrospun blend of polycaprolactone and poly(lactic-co-glycolic acid) (1:1) to determine the feasibility of this system for sustained drug delivery. Kartogenin is a small-molecule drug that could enhance the outcome of microfracture, a cartilage restoration procedure, by selectively stimulating chondrogenic differentiation of endogenous bone marrow mesenchymal stem cells. Experimental results showed that kartogenin did not affect the electrospinnability of the polymer blend, and it had negligible effects on fiber morphology and scaffold mechanical properties. The loading efficiency of kartogenin into electrospun membranes was nearly 100%, and no evidence of chemical reaction between kartogenin and the polymers was detected by Fourier transform infrared spectroscopy. Analysis of the released drug using high-performance liquid chromatography-photodiode array detection indicated an abundance of kartogenin and only a small amount of its major hydrolysis product. Kartogenin displayed a typical biphasic release profile, with approximately 30% being released within 24 h followed by a much slower, constant rate of release up to 28 days. Although additional development is needed to tune the release kinetics and address issues common to electrospun scaffolds (e.g., high fiber density), the results of this study demonstrated that a scaffold electrospun from biodegradable synthetic polymers is a suitable kartogenin delivery vehicle.
Collapse
Affiliation(s)
- Steven Elder
- Department of Agricultural & Biological Engineering, James Worth Bagley College of Engineering, Mississippi State University, Starkville, MS 39762, USA; (J.G.R.); (J.W.)
- Correspondence: ; Tel.: +1-662-325-9107
| | - John Graham Roberson
- Department of Agricultural & Biological Engineering, James Worth Bagley College of Engineering, Mississippi State University, Starkville, MS 39762, USA; (J.G.R.); (J.W.)
| | - James Warren
- Department of Agricultural & Biological Engineering, James Worth Bagley College of Engineering, Mississippi State University, Starkville, MS 39762, USA; (J.G.R.); (J.W.)
| | - Robert Lawson
- Department of Biochemistry, Molecular Biology, Entomology and Plant Pathology, College of Agriculture & Life Sciences, Mississippi State University, Starkville, MS 39762, USA;
| | - Daniel Young
- Department of Comparative Biomedical Sciences, College of Veterinary Medicine, Mississippi State University, Starkville, MS 39762, USA; (D.Y.); (M.K.R.)
| | - Sean Stokes
- Department of Chemistry, College of Arts and Sciences, Mississippi State University, Starkville, MS 39762, USA;
| | - Matthew K. Ross
- Department of Comparative Biomedical Sciences, College of Veterinary Medicine, Mississippi State University, Starkville, MS 39762, USA; (D.Y.); (M.K.R.)
| |
Collapse
|
17
|
Zhu M, Zhong W, Cao W, Zhang Q, Wu G. Chondroinductive/chondroconductive peptides and their-functionalized biomaterials for cartilage tissue engineering. Bioact Mater 2022; 9:221-238. [PMID: 34820567 PMCID: PMC8585793 DOI: 10.1016/j.bioactmat.2021.07.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 05/19/2021] [Accepted: 07/05/2021] [Indexed: 02/06/2023] Open
Abstract
The repair of articular cartilage defects is still challenging in the fields of orthopedics and maxillofacial surgery due to the avascular structure of articular cartilage and the limited regenerative capacity of mature chondrocytes. To provide viable treatment options, tremendous efforts have been made to develop various chondrogenically-functionalized biomaterials for cartilage tissue engineering. Peptides that are derived from and mimic the functions of chondroconductive cartilage extracellular matrix and chondroinductive growth factors, represent a unique group of bioactive agents for chondrogenic functionalization. Since they can be chemically synthesized, peptides bear better reproducibility, more stable efficacy, higher modifiability and yielding efficiency in comparison with naturally derived biomaterials and recombinant growth factors. In this review, we summarize the current knowledge in the designs of the chondroinductive/chondroconductive peptides, the underlying molecular mechanisms and their-functionalized biomaterials for cartilage tissue engineering. We also systematically compare their in-vitro and in-vivo efficacies in inducing chondrogenesis. Our vision is to stimulate the development of novel peptides and their-functionalized biomaterials for cartilage tissue engineering.
Collapse
Affiliation(s)
- Mingjing Zhu
- Department of Temporomandibular Joint, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, Guangdong, 510182, China
- Department of Oral and Maxillofacial Surgery/Pathology, Amsterdam UMC and Academic Center for Dentistry Amsterdam (ACTA), Vrije Universiteit Amsterdam (VU), Amsterdam Movement Science (AMS), Amsterdam, the Netherlands
| | - Wenchao Zhong
- Department of Temporomandibular Joint, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, Guangdong, 510182, China
| | - Wei Cao
- Department of Temporomandibular Joint, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, Guangdong, 510182, China
- Department of Oral and Maxillofacial Surgery/Pathology, Amsterdam UMC and Academic Center for Dentistry Amsterdam (ACTA), Vrije Universiteit Amsterdam (VU), Amsterdam Movement Science (AMS), Amsterdam, the Netherlands
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam, the Netherlands
| | - Qingbin Zhang
- Department of Temporomandibular Joint, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, Guangdong, 510182, China
| | - Gang Wu
- Department of Oral and Maxillofacial Surgery/Pathology, Amsterdam UMC and Academic Center for Dentistry Amsterdam (ACTA), Vrije Universiteit Amsterdam (VU), Amsterdam Movement Science (AMS), Amsterdam, the Netherlands
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam, the Netherlands
| |
Collapse
|
18
|
Dehghan-Baniani D, Mehrjou B, Wang D, Bagheri R, Solouk A, Chu PK, Wu H. A dual functional chondro-inductive chitosan thermogel with high shear modulus and sustained drug release for cartilage tissue engineering. Int J Biol Macromol 2022; 205:638-650. [PMID: 35217083 DOI: 10.1016/j.ijbiomac.2022.02.115] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 02/13/2022] [Accepted: 02/18/2022] [Indexed: 12/17/2022]
Abstract
We report a chitosan-based nanocomposite thermogel with superior shear modulus resembling that of cartilage and dual pro-chondrogenic and anti-inflammatory functions. Two therapeutic agents, kartogenin (KGN) and diclofenac sodium (DS), are employed to promote chondrogenesis of stem cells and suppress inflammation, respectively. To extend the release time in a controlled manner, KGN is encapsulated in the uniform-sized starch microspheres and DS is loaded into the halloysite nanotubes. Both drug carriers are doped into the maleimide-modified chitosan hydrogel to produce a shear modulus of 167 ± 5 kPa that is comparable to that of articular cartilage (50-250 kPa). Owing to the hydrogel injectability and relatively suitable gelation time (5 ± 0.5 min) at 37 °C, this system potentially constitutes a manageable platform for clinical practice. Moreover, sustained linear drug release for over a month boosts chondro-differentiation of stem cells to eliminate the necessity for multiple administrations. Considering virtues such as thermogel strength and ability to co-deliver anti-inflammatory and chondro-inductive biomolecules continuously, the materials and strategy have promising potential in functional cartilage tissue engineering.
Collapse
Affiliation(s)
- Dorsa Dehghan-Baniani
- Department of Chemical and Biological Engineering, Division of Biomedical Engineering, The Hong Kong University of Science and Technology, Hong Kong, China; Polymeric Materials Research Group, Department of Materials Science and Engineering, Sharif University of Technology, Tehran, P.O. Box 11155-9466, Iran
| | - Babak Mehrjou
- Department of Physics, Department of Materials Science and Engineering, Department of Biomedical Engineering, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, China
| | - Dong Wang
- Department of Chemistry, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Reza Bagheri
- Polymeric Materials Research Group, Department of Materials Science and Engineering, Sharif University of Technology, Tehran, P.O. Box 11155-9466, Iran
| | - Atefeh Solouk
- Department of Biomedical Engineering, Amirkabir University of Technology, Tehran, Iran
| | - Paul K Chu
- Department of Physics, Department of Materials Science and Engineering, Department of Biomedical Engineering, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, China
| | - Hongkai Wu
- Department of Chemical and Biological Engineering, Division of Biomedical Engineering, The Hong Kong University of Science and Technology, Hong Kong, China; Department of Chemistry, The Hong Kong University of Science and Technology, Hong Kong, China.
| |
Collapse
|
19
|
Woo Y, Patel M, Kim H, Park JK, Jung YJ, Cha SS, Jeong B. Pralatrexate Sustainably Released from Polypeptide Thermogel Is Effective for Chondrogenic Differentiation of Mesenchymal Stem Cells. ACS APPLIED MATERIALS & INTERFACES 2022; 14:3773-3783. [PMID: 35014790 DOI: 10.1021/acsami.1c20585] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Folic acid was reported to significantly improve chondrogenic differentiation of mesenchymal stem cells. In a similar mechanism of action, we investigated clinically approved antifolates by the U.S. Food and Drug Administration as chondrogenic-promoting compounds for tonsil-derived mesenchymal stem cells. A poly(ethylene glycol)-poly(l-alanine) thermogelling system was used as a three-dimensional cell culture matrix, where stem cells and antifolates could be incorporated simultaneously during a heat-induced in situ sol-to-gel transition. The antifolates could be supplied over several days by the sustained release of the drug from the thermogel. Initially, seven antifolates were prescreened based on cell viability and expression of a typical chondrogenic biomarker of type II collagen (COL II) at the mRNA level. Then, dapsone, pralatrexate, and trimethoprim were selected as candidate compounds in the second round screening, and detailed studies were carried out on the mRNA and protein expression of various chondrogenic biomarkers including COL II, SRY box transcription factor 9, and aggrecan. Three-dimensional cultures of stem cells in the thermogel in the absence of a chondrogenic promoter compound and in the presence of kartogenin (KGN) were performed as a negative control and positive control, respectively. The chondrogenic biomarkers were significantly increased in the selected antifolate-incorporating systems compared to the negative control system, without an increase in type I collagen (an osteogenic biomarker) expression. Pralatrexate was the best compound for inducing chondrogenic differentiation of the stem cells, even better than the positive control (KGN). Nuclear translocation of the core-binding factor β subunit (CBFβ) and enhanced nuclear runt-related transcription factor 1 (RUNX1) by antifolate treatment suggested that the chondrogenesis-enhancing mechanism is mediated by CBFβ and RUNX1. An in silico modeling study confirmed the mechanism by proving the high binding affinity of pralatrexate to a target protein of filamin A compared with other antifolate candidates. To conclude, pralatrexate was rediscovered as a lead compound, and the polypeptide thermogel incorporating pralatrexate and mesenchymal stem cells can be a very effective system in promoting chondrogenic differentiation of stem cells and might be used in injectable tissue engineering for cartilage repair.
Collapse
Affiliation(s)
- Yejin Woo
- Department of Chemistry and Nanoscience, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul 03760, Korea
| | - Madhumita Patel
- Department of Chemistry and Nanoscience, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul 03760, Korea
| | - Hyelin Kim
- Department of Chemistry and Nanoscience, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul 03760, Korea
| | - Jin Kyung Park
- Department of Chemistry and Nanoscience, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul 03760, Korea
| | - Yeon-Ju Jung
- Department of Chemistry and Nanoscience, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul 03760, Korea
| | - Sun-Shin Cha
- Department of Chemistry and Nanoscience, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul 03760, Korea
| | - Byeongmoon Jeong
- Department of Chemistry and Nanoscience, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul 03760, Korea
| |
Collapse
|
20
|
Liu H, Liu P. Kartogenin Promotes the BMSCs Chondrogenic Differentiation in Osteoarthritis by Down-Regulation of miR-145-5p Targeting Smad4 Pathway. Tissue Eng Regen Med 2021; 18:989-1000. [PMID: 34669172 DOI: 10.1007/s13770-021-00390-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Revised: 08/02/2021] [Accepted: 08/19/2021] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Transplantation of mesenchymal stem cells (MSCs) is a potential therapeutic strategy for cartilage degeneration of osteoarthritis (OA). But controlling chondrogenic differentiation of the implanted MSCs in the joints remains a challenge. The role of kartogenin (KGN) for chondrogenesis of MSCs has been widely reported, however, the mechanism of chondrogenesis has not been elucidated in OA. METHODS In this study, we investigated the miR-145-5p, TGF-β, Samd4, and p-stat3/stat3 expression in cartilage of OA patients and bone marrow mesenchymal stem cells (BMSCs) treated with KGN or miR-145-5p inhibitor. In addition, the cell proliferation and chondrogenic differentiation in vitro and in vivo of BMSCs treated with KGN was also detected. RESULTS In OA patients, the expression of miR-145-5p was up-regulated, and the expression of TGF-β, Samd4, and p-stat3/stat3 was inhibited. When the BMSCs treated with miR-145-5p inhibitor, the expression of TGF-β, Samd4, and p-stat3/stat3 was also significantly up-regulated. KGN-treated BMSCs had better proliferation and chondrogenic differentiation by up-regulating the expression of Sox 9, Col-2a1, aggrecan in vitro and in OA by down-regulation of miR-145-5p targeting Smad4 pathway. Moreover, intra-articular injection of KGN-treated BMSCs had a better pain relief effect in OA. CONCLUSION The double effect on cartilage protection and pain relief indicates a great potential of intra-articular injection of KGN-treated BMSCs for the treatment of OA.
Collapse
Affiliation(s)
- Huimin Liu
- Department of Paediatrics, Liyuan Hospital Affiliated To Tongji Medical College of Huazhong University of Science and Technology, 43006, Wuhan, People's Republic of China
| | - Ping Liu
- Department of Orthopaedics, Liyuan Hospital Affiliated To Tongji Medical College of Huazhong University of Science and Technology, 43006, Wuhan, People's Republic of China.
| |
Collapse
|
21
|
Zhang X, Jing H, Luo K, Shi B, Luo Q, Zhu Z, He X, Zheng J. Exosomes from 3T3-J2 promote expansion of tracheal basal cells to facilitate rapid epithelization of 3D-printed double-layer tissue engineered trachea. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 129:112371. [PMID: 34579890 DOI: 10.1016/j.msec.2021.112371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 08/06/2021] [Accepted: 08/09/2021] [Indexed: 11/29/2022]
Abstract
Functional epithelization plays a pivotal role in maintaining long-term lumen patency of tissue-engineered trachea (TET). Due to the slow migration of autologous epithelium, spontaneous epithelization process of transplanted TET is always tardive. Seeding tracheal basal cells (TBCs) on TET before transplantation might be favorable for accelerating epithelization, but rapid expansion of TBCs in vitro is still relatively intractable. In this study, we proposed a promising expansion strategy which enables the TBCs to proliferate rapidly in vitro. TBCs were isolated from the autologous tracheal mucosae of rabbit, and co-cultured with exosomes derived from 3T3-J2 cells. After co-culture with exosomal component, TBCs could vigorously proliferate in vitro and retained their multi-potency. It was in stark contrast to that the single-cultured TBCs could only be expand to passage 2 in about 30 days, moreover, the most majority of single-cultured cells entered late apoptotic stage. On the other hand, a bionic tubular double-layer scaffold with good mechanical property and bio-compatibility was designed and fabricated by 3D printing technology. Then TET with bi-lineage cell-type was constructed in vitro by implanting autologous chondrocytes on the outer-layer of scaffold, and TBCs on the inner-layer, respectively. And then TET was pre-vascularized in vivo, and pedicled transplanted to restore long-segmental defect in recipient rabbits. It was found that the chondrocytes and TBCs seeded on double-layer scaffolds developed well as expected. And almost complete coverage with ciliated epitheliums was observed on the lumen surface of TET 2-week after operation, in comparison with that the epithelization of TET without pre-seeding of TBCs accomplished nearly 2-month after operation. In conclusion, the promising expansion strategy of TBCs together with 3D-printed double-layer scaffolds facilitate the rapid epithelization process of transplanted TET, which might be of vital significance for clinical and translational medicine.
Collapse
Affiliation(s)
- Xiaoyang Zhang
- Department of Cardiothoracic Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 1678 Dongfang Road, Shanghai 200127, China
| | - Hui Jing
- Department of Thoracic Surgery, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kai Luo
- Department of Cardiothoracic Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 1678 Dongfang Road, Shanghai 200127, China
| | - Bozhong Shi
- Department of Cardiothoracic Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 1678 Dongfang Road, Shanghai 200127, China
| | - Qiancheng Luo
- Department of Cardiothoracic Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 1678 Dongfang Road, Shanghai 200127, China
| | - Zhongqun Zhu
- Department of Cardiothoracic Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 1678 Dongfang Road, Shanghai 200127, China
| | - Xiaomin He
- Department of Cardiothoracic Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 1678 Dongfang Road, Shanghai 200127, China.
| | - Jinghao Zheng
- Department of Cardiothoracic Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 1678 Dongfang Road, Shanghai 200127, China.
| |
Collapse
|
22
|
Szustak M, Gendaszewska-Darmach E. Nanocellulose-Based Scaffolds for Chondrogenic Differentiation and Expansion. Front Bioeng Biotechnol 2021; 9:736213. [PMID: 34485266 PMCID: PMC8415884 DOI: 10.3389/fbioe.2021.736213] [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: 07/04/2021] [Accepted: 08/03/2021] [Indexed: 11/13/2022] Open
Abstract
Nanocellulose deserves special attention among the large group of biocompatible biomaterials. It exhibits good mechanical properties, which qualifies it for potential use as a scaffold imitating cartilage. However, the reconstruction of cartilage is a big challenge due to this tissue's limited regenerative capacity resulting from its lack of vascularization, innervations, and sparsely distributed chondrocytes. This feature restricts the infiltration of progenitor cells into damaged sites. Unfortunately, differentiated chondrocytes are challenging to obtain, and mesenchymal stem cells have become an alternative approach to promote chondrogenesis. Importantly, nanocellulose scaffolds induce the differentiation of stem cells into chondrocyte phenotypes. In this review, we present the recent progress of nanocellulose-based scaffolds promoting the development of cartilage tissue, especially within the emphasis on chondrogenic differentiation and expansion.
Collapse
Affiliation(s)
| | - Edyta Gendaszewska-Darmach
- Faculty of Biotechnology and Food Sciences, Institute of Molecular and Industrial Biotechnology, Lodz University of Technology, Lodz, Poland
| |
Collapse
|
23
|
Zeng WN, Zhang Y, Wang D, Zeng YP, Yang H, Li J, Zhou CP, Liu JL, Yang QJ, Deng ZL, Zhou ZK. Intra-articular Injection of Kartogenin-Enhanced Bone Marrow-Derived Mesenchymal Stem Cells in the Treatment of Knee Osteoarthritis in a Rat Model. Am J Sports Med 2021; 49:2795-2809. [PMID: 34213976 DOI: 10.1177/03635465211023183] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND In this study, we investigated the in vitro and in vivo chondrogenic capacity of kartogenin (KGN)-enhanced bone marrow-derived mesenchymal stem cells (BMSCs) for cartilage regeneration. PURPOSE To determine (1) whether functionalized nanographene oxide (NGO) can effectively deliver KGN into BMSCs and (2) whether KGN would enhance BMSCs during chondrogenesis in vitro and in vivo in an animal model. STUDY DESIGN Controlled laboratory study. METHODS Functionalized NGO with line chain amine-terminated polyethylene glycol (PEG) and branched polyethylenimine (BPEI) were used to synthesize biocompatible NGO-PEG-BPEI (PPG) and for loading hydrophobic KGN molecules noncovalently via π-π stacking and hydrophobic interactions (PPG-KGN). Then, PPG-KGN was used for the intracellular delivery of hydrophobic KGN by simple mixing and co-incubation with BMSCs to acquire KGN-enhanced BMSCs. The chondrogenic efficacy of KGN-enhanced BMSCs was evaluated in vitro. In vivo, osteoarthritis (OA) was induced by anterior cruciate ligament transection in rats. A total of 5 groups were established: normal (OA treated with nothing), phosphate-buffered saline (PBS; intra-articular injection of PBS), PPG-KGN (intra-articular injection of PPG-KGN), BMSCs (intra-articular injection of BMSCs), and BMSCs + PPG-KGN (intra-articular injection of PPG-KGN-preconditioned BMSCs). At 6 and 9 weeks after the surgical induction of OA, the rats received intra-articular injections of PPG-KGN, BMSCs, or KGN-enhanced BMSCs. At 14 weeks after the surgical induction of OA, radiographic and behavioral evaluations as well as histological analysis of the knee joints were performed. RESULTS The in vitro study showed that PPG could be rapidly uptaken in the first 4 hours after incubation, reaching saturation at 12 hours and accumulating in the lysosome and cytoplasm of BMSCs. Thus, PPG-KGN could enhance the efficiency of the intracellular delivery of KGN, which showed a remarkably high chondrogenic differentiation capacity of BMSCs. When applied to an OA model of cartilage injuries in rats, PPG-KGN-preconditioned BMSCs contributed to protection from joint space narrowing, pathological mineralization, OA development, and OA-induced pain, as well as improved tissue regeneration, as evidenced by radiographic, weightbearing, and histological findings. CONCLUSION Our results demonstrate that KGN-enhanced BMSCs showed markedly improved capacities for chondrogenesis and articular cartilage repair. We believe that this work demonstrates that a multifunctional nanoparticle-based drug delivery system could be beneficial for stem cell therapy. Our results present an opportunity to reverse the symptoms and pathophysiology of OA. CLINICAL RELEVANCE The intracellular delivery of KGN to produce BMSCs with enhanced chondrogenic potential may offer a new approach for the treatment of OA.
Collapse
Affiliation(s)
- Wei-Nan Zeng
- Orthopedic Research Institution, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, China.,Department of Orthopedics, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing, China.,Department of Orthopedics, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yun Zhang
- Department of Traditional Chinese Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Duan Wang
- Orthopedic Research Institution, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, China
| | - Yi-Ping Zeng
- Department of Orthopedics, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing, China
| | - Hao Yang
- Center for Joint Surgery, Southwest Hospital, Army Medical University, Chongqing, China
| | - Juan Li
- Center for Joint Surgery, Southwest Hospital, Army Medical University, Chongqing, China
| | - Cheng-Pei Zhou
- Department of Orthopedics, Tangdu Hospital, Air Force Medical University, Xi'an, China
| | - Jun-Li Liu
- Department of Orthopedics, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing, China
| | - Qing-Jun Yang
- Department of Orthopedics, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing, China
| | - Zhong-Liang Deng
- Department of Orthopedics, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zong-Ke Zhou
- Orthopedic Research Institution, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
24
|
Hou M, Zhang Y, Zhou X, Liu T, Yang H, Chen X, He F, Zhu X. Kartogenin prevents cartilage degradation and alleviates osteoarthritis progression in mice via the miR-146a/NRF2 axis. Cell Death Dis 2021; 12:483. [PMID: 33986262 PMCID: PMC8119954 DOI: 10.1038/s41419-021-03765-x] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 03/29/2021] [Accepted: 03/30/2021] [Indexed: 02/06/2023]
Abstract
Osteoarthritis (OA) is a common articular degenerative disease characterized by loss of cartilage matrix and subchondral bone sclerosis. Kartogenin (KGN) has been reported to improve chondrogenic differentiation of mesenchymal stem cells. However, the therapeutic effect of KGN on OA-induced cartilage degeneration was still unclear. This study aimed to explore the protective effects and underlying mechanisms of KGN on articular cartilage degradation using mice with post-traumatic OA. To mimic the in vivo arthritic environment, in vitro cultured chondrocytes were exposed to interleukin-1β (IL-1β). We found that KGN barely affected the cell proliferation of chondrocytes; however, KGN significantly enhanced the synthesis of cartilage matrix components such as type II collagen and aggrecan in a dose-dependent manner. Meanwhile, KGN markedly suppressed the expression of matrix degradation enzymes such as MMP13 and ADAMTS5. In vivo experiments showed that intra-articular administration of KGN ameliorated cartilage degeneration and inhibited subchondral bone sclerosis in an experimental OA mouse model. Molecular biology experiments revealed that KGN modulated intracellular reactive oxygen species in IL-1β-stimulated chondrocytes by up-regulating nuclear factor erythroid 2-related factor 2 (NRF2), while barely affecting its mRNA expression. Microarray analysis further revealed that IL-1β significantly up-regulated miR-146a that played a critical role in regulating the protein levels of NRF2. KGN treatment showed a strong inhibitory effect on the expression of miR-146a in IL-1β-stimulated chondrocytes. Over-expression of miR-146a abolished the anti-arthritic effects of KGN not only by down-regulating the protein levels of NRF2 but also by up-regulating the expression of matrix degradation enzymes. Our findings demonstrate, for the first time, that KGN exerts anti-arthritic effects via activation of the miR-146a-NRF2 axis and KGN is a promising heterocyclic molecule to prevent OA-induced cartilage degeneration.
Collapse
Affiliation(s)
- Mingzhuang Hou
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, China.,Orthopaedic Institute, Medical College, Soochow University, Suzhou, China
| | - Yijian Zhang
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, China.,Orthopaedic Institute, Medical College, Soochow University, Suzhou, China
| | - Xinfeng Zhou
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, China.,Orthopaedic Institute, Medical College, Soochow University, Suzhou, China
| | - Tao Liu
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, China
| | - Huilin Yang
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, China.,Orthopaedic Institute, Medical College, Soochow University, Suzhou, China
| | - Xi Chen
- Department of Pathology, The Third Affiliated Hospital of Soochow University, Changzhou, China.
| | - Fan He
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, China. .,Orthopaedic Institute, Medical College, Soochow University, Suzhou, China.
| | - Xuesong Zhu
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, China. .,Orthopaedic Institute, Medical College, Soochow University, Suzhou, China.
| |
Collapse
|
25
|
Ma X, Long C, Wang F, Lou B, Yuan M, Duan F, Yang Y, Li J, Qian X, Zeng J, Lin S, Shen H, Lin X. METTL3 attenuates proliferative vitreoretinopathy and epithelial-mesenchymal transition of retinal pigment epithelial cells via wnt/β-catenin pathway. J Cell Mol Med 2021; 25:4220-4234. [PMID: 33759344 PMCID: PMC8093987 DOI: 10.1111/jcmm.16476] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 02/28/2021] [Accepted: 03/03/2021] [Indexed: 12/11/2022] Open
Abstract
Proliferative vitreoretinopathy (PVR) is a refractory vitreoretinal fibrosis disease, and epithelial-mesenchymal transition (EMT) of retinal pigment epithelial (RPE) cells is the key pathological mechanism of PVR. However, few studies focused on the role of METTL3, the dominating methyltransferase for m6A RNA modification in PVR pathogenesis. Immunofluorescence staining and qRT-PCR were used to determine the expression of METTL3 in human tissues. Lentiviral transfection was used to stably overexpress and knockdown METTL3 in ARPE-19 cells. MTT assay was employed to study the effects of METTL3 on cell proliferation. The impact of METTL3 on the EMT of ARPE-19 cells was assessed by migratory assay, morphological observation and expression of EMT markers. Intravitreal injection of cells overexpressing METTL3 was used to assess the impact of METTL3 on the establishment of the PVR model. We found that METTL3 expression was less in human PVR membranes than in the normal RPE layers. In ARPE-19 cells, total m6A abundance and the METTL3 expression were down-regulated after EMT. Additionally, METTL3 overexpression inhibited cell proliferation through inducing cell cycle arrest at G0/G1 phase. Furthermore, METTL3 overexpression weakened the capacity of TGFβ1 to trigger EMT by regulating wnt/β -catenin pathway. Oppositely, knockdown of METTL3 facilitated proliferation and EMT of ARPE-19 cells. In vivo, intravitreal injection of METTL3-overexpressing cells delayed the development of PVR compared with injection of control cells. In summary, this study suggested that METTL3 is involved in the PVR process, and METTL3 overexpression inhibits the EMT of ARPE-19 cells in vitro and suppresses the PVR process in vivo.
Collapse
Affiliation(s)
- Xinqi Ma
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Chongde Long
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Fangyu Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Bingsheng Lou
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Miner Yuan
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Fang Duan
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Yao Yang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Jiaqing Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Xiaobing Qian
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Jieting Zeng
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Shuibin Lin
- Center for Translational Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Huangxuan Shen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China.,Biobank of Eye, State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Xiaofeng Lin
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
26
|
Recent Developed Strategies for Enhancing Chondrogenic Differentiation of MSC: Impact on MSC-Based Therapy for Cartilage Regeneration. Stem Cells Int 2021; 2021:8830834. [PMID: 33824665 PMCID: PMC8007380 DOI: 10.1155/2021/8830834] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 02/20/2021] [Accepted: 03/04/2021] [Indexed: 12/19/2022] Open
Abstract
Articular cartilage is susceptible to damage, but its self-repair is hindered by its avascular nature. Traditional treatment methods are not able to achieve satisfactory repair effects, and the development of tissue engineering techniques has shed new light on cartilage regeneration. Mesenchymal stem cells (MSCs) are one of the most commonly used seed cells in cartilage tissue engineering. However, MSCs tend to lose their multipotency, and the composition and structure of cartilage-like tissues formed by MSCs are far from those of native cartilage. Thus, there is an urgent need to develop strategies that promote MSC chondrogenic differentiation to give rise to durable and phenotypically correct regenerated cartilage. This review provides an overview of recent advances in enhancement strategies for MSC chondrogenic differentiation, including optimization of bioactive factors, culture conditions, cell type selection, coculture, gene editing, scaffolds, and physical stimulation. This review will aid the further understanding of the MSC chondrogenic differentiation process and enable improvement of MSC-based cartilage tissue engineering.
Collapse
|
27
|
Gelatin reduced Graphene Oxide Nanosheets as Kartogenin Nanocarrier Induces Rat ADSCs Chondrogenic Differentiation Combining with Autophagy Modification. MATERIALS 2021; 14:ma14051053. [PMID: 33668133 PMCID: PMC7956601 DOI: 10.3390/ma14051053] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 02/18/2021] [Accepted: 02/19/2021] [Indexed: 12/19/2022]
Abstract
Biocompatible reduced graphene oxide (rGO) could deliver drugs for synergistically stimulating stem cells directed differentiation with influences on specific cellular activities. Here, we prepared a biodegradable gelatin reduced graphene oxide (rGO@Ge) to evaluate its functions in promoting rat adipose derived mesenchymal stem cells (ADSCs) chondrogenic differentiation through delivering kartogenin (KGN) into the stem cell efficiently. The optimum KGN concentration (approximately 1 μM) that promoted the proliferation and chondrogenic differentiation of ADSCs was clarified by a series of experiments, including immunofluorescent (IF) staining (Sox-9, Col II), alcian blue (Ab) staining, toluidine blue (Tb) staining and real-time quantitative PCR analysis of the chondrogenic markers. Meanwhile, the biocompatibility of rGO@Ge was evaluated to clearly define the nonhazardous concentration range, and the drug loading and releasing properties of rGO@Ge were tested with KGN for its further application in inducing ADSCs chondrogenic differentiation. Furthermore, the mechanism of rGO@Ge entering ADSCs was investigated by the different inhibitors that are involved in the endocytosis of the nanocarrier, and the degradation of the rGO@Ge in ADSCs was observed by transmission electron microscopy (TEM). The synergistic promoting effect of rGO@Ge nanocarrier on ADSCs chondrogenesis with KGN was also studied by the IF, Ab, Tb stainings and PCR analysis of the chondrogenic markers. Finally, the intracellular Reactive Oxygen Species (ROS) and autophagy induced by KGN/rGO@Ge complex composites were tested in details for clarification on the correlation between the autophagy and chondrogenesis in ADSCs induced by rGO@Ge. All the results show that rGO@Ge as a biocompatible nanocarrier can deliver KGN into ADSCs for exerting a pro-chondrogenic effect and assist the drug to promote ADSCs chondrogenesis synergistically through modification of the autophagy in vitro, which promised its further application in repairing cartilage defect in vivo.
Collapse
|
28
|
Chitosan/polycaprolactone multilayer hydrogel: A sustained Kartogenin delivery model for cartilage regeneration. Int J Biol Macromol 2021; 177:589-600. [PMID: 33610607 DOI: 10.1016/j.ijbiomac.2021.02.122] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 02/13/2021] [Accepted: 02/16/2021] [Indexed: 12/11/2022]
Abstract
Cartilage regeneration using biomaterial-guided delivery systems presents improved therapeutic efficacy of the biomolecules while minimizing side effects. Here, our hypothesis was to design a multilayer scaffold of chitosan (CS) hydrogel and polycaprolactone (PCL) mat to enhance the mechanical properties, integrity and stability of CS, especially for subsequent in vivo transplantation. After conjugation of the Kartogenin (KGN) into this structure, its gradual release can promote chondrogenesis of mesenchymal stem cells (MSCs). Initially, a thin electrospun PCL layer was sandwiched between two CS hydrogels. Subsequently, KGN was superficially immobilized onto the CS matrix. The successful conjugation was confirmed by scanning electron microscopy (SEM) and infrared spectroscopy. These novel KGN-conjugated scaffolds possessed lower swelling and higher compressive modulus and showed gradual release of KGN in longer retention times. Immunofluorescent and histological staining represented more cells located in lacunae as well as more Coll2 and Sox9 positive cells on KGN-conjugated scaffolds. Gene expression analysis also revealed that SOX9, COLL2 and ACAN expression levels were higher in the presence of KGN, while COLLX expression was down-regulated, indicating a hypertrophy phenomenon with synergistic effect of TGF-β. This multilayer structure not only facilitates the effective treatment, but also provides a proper mechanical structure for cartilage engineering.
Collapse
|
29
|
Research Progress on Stem Cell Therapies for Articular Cartilage Regeneration. Stem Cells Int 2021; 2021:8882505. [PMID: 33628274 PMCID: PMC7895563 DOI: 10.1155/2021/8882505] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 01/11/2021] [Accepted: 01/28/2021] [Indexed: 02/07/2023] Open
Abstract
Injury of articular cartilage can cause osteoarthritis and seriously affect the physical and mental health of patients. Unfortunately, current surgical treatment techniques that are commonly used in the clinic cannot regenerate articular cartilage. Regenerative medicine involving stem cells has entered a new stage and is considered the most promising way to regenerate articular cartilage. In terms of theories on the mechanism, it was thought that stem cell-mediated articular cartilage regeneration was achieved through the directional differentiation of stem cells into chondrocytes. However, recent evidence has shown that the stem cell secretome plays an important role in biological processes such as the immune response, inflammation regulation, and drug delivery. At the same time, the stem cell secretome can effectively mediate the process of tissue regeneration. This new theory has attributed the therapeutic effect of stem cells to their paracrine effects. The application of stem cells is not limited to exogenous stem cell transplantation. Endogenous stem cell homing and in situ regeneration strategies have received extensive attention. The application of stem cell derivatives, such as conditioned media, extracellular vesicles, and extracellular matrix, is an extension of stem cell paracrine theory. On the other hand, stem cell pretreatment strategies have also shown promising therapeutic effects. This article will systematically review the latest developments in these areas, summarize challenges in articular cartilage regeneration strategies involving stem cells, and describe prospects for future development.
Collapse
|
30
|
Kobayashi M, Chijimatsu R, Hart DA, Hamamoto S, Jacob G, Yano F, Saito T, Shimomura K, Ando W, Chung UI, Tanaka S, Yoshikawa H, Nakamura N. Evidence that TD-198946 enhances the chondrogenic potential of human synovium-derived stem cells through the NOTCH3 signaling pathway. J Tissue Eng Regen Med 2020; 15:103-115. [PMID: 33169924 DOI: 10.1002/term.3149] [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: 10/16/2019] [Revised: 11/03/2020] [Accepted: 11/04/2020] [Indexed: 11/10/2022]
Abstract
Human synovium-derived stem cells (hSSCs) are an attractive source of cells for cartilage repair. At present, the quality of tissue and techniques used for cartilage regeneration have scope for improvement. A small compound, TD-198946, was reported to enhance chondrogenic induction from hSSCs; however, other applications of TD-198946, such as priming the cell potential of hSSCs, remain unknown. Our study aimed to examine the effect of TD-198946 pretreatment on hSSCs. HSSCs were cultured with or without TD-198946 for 7 days during expansion culture and then converted into a three-dimensional pellet culture supplemented with bone morphogenetic protein-2 (BMP2) and/or transforming growth factor beta-3 (TGFβ3). Chondrogenesis in cultures was assessed based on the GAG content, histology, and expression levels of chondrogenic marker genes. Cell pellets derived from TD-198946-pretreated hSSCs showed enhanced chondrogenic potential when chondrogenesis was induced by both BMP2 and TGFβ3. Moreover, cartilaginous tissue was efficiently generated from TD-198946-pretreated hSSCs using a combination of BMP2 and TGFβ3. Microarray analysis revealed that NOTCH pathway-related genes and their target genes were significantly upregulated in TD-198946-treated hSSCs, although TD-198946 alone did not upregulate chondrogenesis related markers. The administration of the NOTCH signal inhibitor diminished the effect of TD-198946. Thus, TD-198946 enhances the chondrogenic potential of hSSCs via the NOTCH3 signaling pathway. This study is the first to demonstrate the gradual activation of NOTCH3 signaling during chondrogenesis in hSSCs. The priming of NOTCH3 using TD-198946 provides a novel insight regarding the regulation of the differentiation of hSSCs into chondrocytes.
Collapse
Affiliation(s)
- Masato Kobayashi
- Orthopaedic Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Ryota Chijimatsu
- Bone and Cartilage Regenerative Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - David A Hart
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, Canada
| | - Shuichi Hamamoto
- Orthopaedic Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - George Jacob
- Orthopaedic Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Fumiko Yano
- Bone and Cartilage Regenerative Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Taku Saito
- Sensory and Motor System Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Kazunori Shimomura
- Orthopaedic Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Wataru Ando
- Orthopaedic Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Ung-Il Chung
- Center for Disease Biology and Integrative Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Sakae Tanaka
- Sensory and Motor System Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Hideki Yoshikawa
- Orthopaedic Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Norimasa Nakamura
- Institute for Medical Science in Sports, Osaka Health Science University, Osaka, Japan
| |
Collapse
|
31
|
Li T, Liu B, Chen K, Lou Y, Jiang Y, Zhang D. Small molecule compounds promote the proliferation of chondrocytes and chondrogenic differentiation of stem cells in cartilage tissue engineering. Biomed Pharmacother 2020; 131:110652. [PMID: 32942151 DOI: 10.1016/j.biopha.2020.110652] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 08/15/2020] [Accepted: 08/17/2020] [Indexed: 02/09/2023] Open
Abstract
The application of tissue engineering to generate cartilage is limited because of low proliferative ability and unstable phenotype of chondrocytes. The sources of cartilage seed cells are mainly chondrocytes and stem cells. A variety of methods have been used to obtain large numbers of chondrocytes, including increasing chondrocyte proliferation and stem cell chondrogenic differentiation via cytokines, genes, and proteins. Natural or synthetic small molecule compounds can provide a simple and effective method to promote chondrocyte proliferation, maintain a stable chondrocyte phenotype, and promote stem cell chondrogenic differentiation. Therefore, the study of small molecule compounds is of great importance for cartilage tissue engineering. Herein, we review a series of small molecule compounds and their mechanisms that can promote chondrocyte proliferation, maintain chondrocyte phenotype, or induce stem cell chondrogenesis. The studies in this field represent significant contributions to the research in cartilage tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Tian Li
- Department of Plastic and Reconstructive Surgery, The First Bethune Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| | - Bingzhang Liu
- Department of Plastic and Reconstructive Surgery, The First Bethune Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| | - Kang Chen
- Department of Plastic and Reconstructive Surgery, The First Bethune Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| | - Yingyue Lou
- Department of Plastic and Reconstructive Surgery, The First Bethune Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| | - Yuhan Jiang
- Department of Plastic and Reconstructive Surgery, The First Bethune Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| | - Duo Zhang
- Department of Plastic and Reconstructive Surgery, The First Bethune Hospital of Jilin University, Changchun, Jilin, People's Republic of China.
| |
Collapse
|
32
|
Transforming growth factor-beta stimulates human bone marrow-derived mesenchymal stem/stromal cell chondrogenesis more so than kartogenin. Sci Rep 2020; 10:8340. [PMID: 32433527 PMCID: PMC7239921 DOI: 10.1038/s41598-020-65283-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 03/20/2020] [Indexed: 12/21/2022] Open
Abstract
A previous study identified kartogenin (KGN) as a potent modulator of bone marrow mesenchymal stem/stromal cell (BMSC) chondrogenesis. This initial report did not contrast KGN directly against transforming growth factor-beta 1 (TGF-β1), the most common growth factor used in chondrogenic induction medium. Herein, we directly compared the in vitro chondrogenic potency of TGF-β1 and KGN using a high resolution micropellet model system. Micropellets were cultured for 7–14 days in medium supplemented with TGF-β1, KGN, or both TGF-β1 + KGN. Following 14 days of induction, micropellets exposed to TGF-β1 alone or TGF-β1 + KGN in combination were larger and produced more glycosominoglycan (GAG) than KGN-only cultures. When TGF-β1 + KGN was used, GAG quantities were similar or slightly greater than the TGF-β1-only cultures, depending on the BMSC donor. BMSC micropellet cultures supplemented with KGN alone contracted in size over the culture period and produced minimal GAG. Indicators of hypertrophy were not mitigated in TGF-β1 + KGN cultures, suggesting that KGN does not obstruct BMSC hypertrophy. KGN appears to have weak chondrogenic potency in human BMSC cultures relative to TGF-β1, does not obstruct hypertrophy, and may not be a viable alternative to growth factors in cartilage tissue engineering.
Collapse
|
33
|
Budgude P, Kale V, Vaidya A. Mesenchymal stromal cell‐derived extracellular vesicles as cell‐free biologics for the ex vivo expansion of hematopoietic stem cells. Cell Biol Int 2020; 44:1078-1102. [DOI: 10.1002/cbin.11313] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 01/31/2020] [Indexed: 12/11/2022]
Affiliation(s)
- Pallavi Budgude
- Symbiosis Centre for Stem Cell ResearchSymbiosis International (Deemed University) Pune 412115 India
| | - Vaijayanti Kale
- Symbiosis Centre for Stem Cell ResearchSymbiosis International (Deemed University) Pune 412115 India
| | - Anuradha Vaidya
- Symbiosis Centre for Stem Cell ResearchSymbiosis International (Deemed University) Pune 412115 India
- Symbiosis School of Biological SciencesSymbiosis International (Deemed University) Pune 412115 India
| |
Collapse
|
34
|
Chen C, Huang K, Zhu J, Bi Y, Wang L, Jiang J, Zhu T, Yan X, Zhao J. A novel elastic and controlled-release poly(ether-ester-urethane)urea scaffold for cartilage regeneration. J Mater Chem B 2020; 8:4106-4121. [PMID: 32253395 DOI: 10.1039/c9tb02754h] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
In the tissue engineering of cartilage, scaffolds with appropriate elasticity and controlled-release properties are essential. Herein, we synthesized a poly(ether-ester-urethane)urea scaffold with a pendant amino group (PEEUUN) through a de-protection process from PEEUU-Boc polymers and grafted kartogenin (KGN) onto the PEEUUN scaffolds (PEEUUN-KGN). Characterization, performance tests, scaffold biocompatibility analysis, and chondrogenesis evaluation both in vitro and in vivo were conducted. The results revealed that the PEEUUN-KGN scaffolds were degradable and three-dimensional (3D) with interconnected pores, and possessed good elasticity, as well as excellent cytocompatibility. Meanwhile, KGN on the PEEUUN-KGN scaffolds underwent stable sustained release for a long time and promoted human umbilical cord mesenchymal stem cells (HUCMSCs) to differentiate into chondrocytes in vitro, thus enhancing cartilage regeneration in vivo. In conclusion, the present PEEUUN-KGN scaffolds would have application potential for cartilage tissue engineering.
Collapse
Affiliation(s)
- Chang'an Chen
- Department of Sports Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, People's Republic of China.
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Jiang C, Sun ZM, Zhu DC, Guo Q, Xu JJ, Lin JH, Chen ZX, Wu YS. Inhibition of Rac1 activity by NSC23766 prevents cartilage endplate degeneration via Wnt/β-catenin pathway. J Cell Mol Med 2020; 24:3582-3592. [PMID: 32040269 PMCID: PMC7131937 DOI: 10.1111/jcmm.15049] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Revised: 11/22/2019] [Accepted: 01/22/2020] [Indexed: 12/19/2022] Open
Abstract
Cartilage endplate (CEP) degeneration has been considered as one of important factors related to intervertebral disc degeneration (IVDD). Previous researches have showed that Rac1 played a pivotal role in chondrocyte differentiation. However, the effect of Rac1 during the process of CEP degeneration remains unclear. Herein, we explored the effect of Rac1 on CEP degeneration and elucidated the underlying molecular mechanism. We found expression of Rac1‐GTP increased in human‐degenerated CEP tissue and IL‐1β‐stimulated rat endplate chondrocytes (EPCs). Our study revealed that Rac1 inhibitor NSC23766 treatment promoted the expression of collagen II, aggrecan and Sox‐9, and decreased the expression of ADTAMTS5 and MMP13 in IL‐1β‐stimulated rat EPCs. Moreover, we also found that NSC23766 could suppress the activation of Wnt/β‐catenin pathway, suggesting that the beneficial effects of Rac1 inhibition in EPCs are mediated through the Wnt/β‐catenin signalling. Besides, puncture‐induced rats models showed that NSC23766 played a protective role on CEP and disc degeneration. Collectively, these findings demonstrated that Rac1 inhibition delayed the EPCs degeneration and its potential mechanism may be associated with Wnt/β‐catenin pathway regulation, which may help us better understand the association between Rac1 and CEP degeneration and provide a promising strategy for delaying the progression of IVDD.
Collapse
Affiliation(s)
- Chao Jiang
- Department of Orthopaedic surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, China.,The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Ze-Ming Sun
- Department of Orthopaedic surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, China.,The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Ding-Chao Zhu
- Department of Orthopaedic surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, China.,The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Qiang Guo
- Department of Orthopaedic surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, China.,The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Jia-Jing Xu
- Department of Orthopaedic surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, China.,The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Jia-Hao Lin
- Department of Orthopaedic surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, China.,The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Ze-Xin Chen
- Department of Orthopaedic surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, China
| | - Yao-Sen Wu
- Department of Orthopaedic surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, China
| |
Collapse
|
36
|
Jing H, Zhang X, Luo K, Luo Q, Yin M, Wang W, Zhu Z, Zheng J, He X. miR-381-abundant small extracellular vesicles derived from kartogenin-preconditioned mesenchymal stem cells promote chondrogenesis of MSCs by targeting TAOK1. Biomaterials 2020; 231:119682. [DOI: 10.1016/j.biomaterials.2019.119682] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 12/08/2019] [Accepted: 12/11/2019] [Indexed: 02/07/2023]
|
37
|
Zhao Y, Zhao X, Zhang R, Huang Y, Li Y, Shan M, Zhong X, Xing Y, Wang M, Zhang Y, Zhao Y. Cartilage Extracellular Matrix Scaffold With Kartogenin-Encapsulated PLGA Microspheres for Cartilage Regeneration. Front Bioeng Biotechnol 2020; 8:600103. [PMID: 33363129 PMCID: PMC7756004 DOI: 10.3389/fbioe.2020.600103] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 10/30/2020] [Indexed: 12/13/2022] Open
Abstract
Repair of articular cartilage defects is a challenging aspect of clinical treatment. Kartogenin (KGN), a small molecular compound, can induce the differentiation of bone marrow-derived mesenchymal stem cells (BMSCs) into chondrocytes. Here, we constructed a scaffold based on chondrocyte extracellular matrix (CECM) and poly(lactic-co-glycolic acid) (PLGA) microspheres (MP), which can slowly release KGN, thus enhancing its efficiency. Cell adhesion, live/dead staining, and CCK-8 results indicated that the PLGA(KGN)/CECM scaffold exhibited good biocompatibility. Histological staining and quantitative analysis demonstrated the ability of the PLGA(KGN)/CECM composite scaffold to promote the differentiation of BMSCs. Macroscopic observations, histological tests, and specific marker analysis showed that the regenerated tissues possessed characteristics similar to those of normal hyaline cartilage in a rabbit model. Use of the PLGA(KGN)/CECM scaffold may mimic the regenerative microenvironment, thereby promoting chondrogenic differentiation of BMSCs in vitro and in vivo. Therefore, this innovative composite scaffold may represent a promising approach for acellular cartilage tissue engineering.
Collapse
Affiliation(s)
- Yanhong Zhao
- Stomatological Hospital of Tianjin Medical University, Tianjin, China
- Tianjin Medical University, Tianjin, China
- *Correspondence: Yanhong Zhao,
| | - Xige Zhao
- Stomatological Hospital of Tianjin Medical University, Tianjin, China
- Tianjin Medical University, Tianjin, China
| | - Rui Zhang
- Stomatological Hospital of Tianjin Medical University, Tianjin, China
- Tianjin Medical University, Tianjin, China
| | - Ying Huang
- Stomatological Hospital of Tianjin Medical University, Tianjin, China
- Tianjin Medical University, Tianjin, China
| | - Yunjie Li
- Stomatological Hospital of Tianjin Medical University, Tianjin, China
- Tianjin Medical University, Tianjin, China
| | - Minhui Shan
- Stomatological Hospital of Tianjin Medical University, Tianjin, China
- Tianjin Medical University, Tianjin, China
| | - Xintong Zhong
- Stomatological Hospital of Tianjin Medical University, Tianjin, China
- Tianjin Medical University, Tianjin, China
| | - Yi Xing
- Stomatological Hospital of Tianjin Medical University, Tianjin, China
- Tianjin Medical University, Tianjin, China
| | - Min Wang
- Stomatological Hospital of Tianjin Medical University, Tianjin, China
- Tianjin Medical University, Tianjin, China
| | | | - Yanmei Zhao
- Institute of Disaster Medicine, Tianjin University, Tianjin, China
- Yanmei Zhao,
| |
Collapse
|
38
|
Porte B, Marguerit G, Thomasseau S, Paquet C, Hugon J. Dose-dependent neuroprotective effect of the JNK inhibitor Brimapitide in 5xFAD transgenic mice. Brain Res 2019; 1727:146587. [PMID: 31811838 DOI: 10.1016/j.brainres.2019.146587] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 11/28/2019] [Accepted: 11/30/2019] [Indexed: 01/13/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease mainly affecting old people. According to the "amyloid cascade hypothesis", the accumulation of Aβ oligomers could lead to kinase activation and Tau phosphorylation. Activated kinases include c-Jun N-terminal kinase (JNK) and previous studies highlighted the beneficial effects of the JNK-specific inhibitor Brimapitide (10 mg/kg) in 5xFAD transgenic mice. Our aim was to evaluate the effects of decreasing doses of Brimapitide on cognition and neurodegeneration in early treated 5xFAD mice. Three month-old 5xFAD were intravenously treated for 6 months with either Brimapitide (3 mg/kg or 0.3 mg/kg) or Nacl. Cognition and amyloid burden, neuronal and synaptic impairments were evaluated. Low doses of Brimapitide (0.3 mg/kg) reduced neuronal degeneration and improved cognition in treated mice compared to non-treated mice. Amyloid burden and synaptic degeneration only decreased with the 3 mg/kg dose. This JNK inhibitor can afford neuroprotection but with a differential effect on amyloid deposition in 5xFAD mice. Brimapitide might partially prevent ongoing neurodegeneration in 5xFAD mice.
Collapse
Affiliation(s)
- Baptiste Porte
- Université de Paris, INSERM UMR-S 1144, F-75006 Paris, France; Centre de Neurologie Cognitive, AP-HP, Hôpital Fernand-Widal, F-75475 Paris, France.
| | | | | | - Claire Paquet
- Université de Paris, INSERM UMR-S 1144, F-75006 Paris, France; Centre de Neurologie Cognitive, AP-HP, Hôpital Fernand-Widal, F-75475 Paris, France.
| | - Jacques Hugon
- Université de Paris, INSERM UMR-S 1144, F-75006 Paris, France; Centre de Neurologie Cognitive, AP-HP, Hôpital Fernand-Widal, F-75475 Paris, France.
| |
Collapse
|
39
|
Li Y, Qiao Z, Yu F, Hu H, Huang Y, Xiang Q, Zhang Q, Yang Y, Zhao Y. Transforming Growth Factor-β3/Chitosan Sponge (TGF-β3/CS) Facilitates Osteogenic Differentiation of Human Periodontal Ligament Stem Cells. Int J Mol Sci 2019; 20:E4982. [PMID: 31600954 PMCID: PMC6834328 DOI: 10.3390/ijms20204982] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 10/04/2019] [Accepted: 10/07/2019] [Indexed: 12/12/2022] Open
Abstract
Periodontal disease is the main reason for tooth loss in adults. Tissue engineering and regenerative medicine are advanced technologies used to manage soft and hard tissue defects caused by periodontal disease. We developed a transforming growth factor-β3/chitosan sponge (TGF-β3/CS) to repair periodontal soft and hard tissue defects. We investigated the proliferation and osteogenic differentiation behaviors of primary human periodontal ligament stem cells (hPDLSCs) to determine the bioactivity and potential application of TGF-β3 in periodontal disease. We employed calcein-AM/propidium iodide (PI) double labeling or cell membranes (CM)-Dil labeling coupled with fluorescence microscopy to trace the survival and function of cells after implantation in vitro and in vivo. The mineralization of osteogenically differentiated hPDLSCs was confirmed by measuring alkaline phosphatase (ALP) activity and calcium content. The levels of COL I, ALP, TGF-βRI, TGF-βRII, and Pp38/t-p38 were assessed by western blotting to explore the mechanism of bone repair prompted by TGF-β3. When hPDLSCs were implanted with various concentrations of TGF-β3/CS (62.5-500 ng/mL), ALP activity was the highest in the TGF-β3 (250 ng/mL) group after 7 d (p < 0.05 vs. control). The calcium content in each group was increased significantly after 21 and 28 d (p < 0.001 vs. control). The optimal result was achieved by the TGF-β3 (500 ng/mL) group. These results showed that TGF-β3/CS promotes osteogenic differentiation of hPDLSCs, which may involve the p38 mitogen-activated protein kinase (MAPK) signaling pathway. TGF-β3/CS has the potential for application in the repair of incomplete alveolar bone defects.
Collapse
Affiliation(s)
- Yangfan Li
- Institute of Biomedicine and Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou 510632, China (Y.H.)
| | - Zhifen Qiao
- Institute of Biomedicine and Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou 510632, China (Y.H.)
| | - Fenglin Yu
- Institute of Biomedicine and Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou 510632, China (Y.H.)
| | - Huiting Hu
- Department of Stomatology, Jinan University Medical College, Guangzhou 510632, China;
| | - Yadong Huang
- Institute of Biomedicine and Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou 510632, China (Y.H.)
| | - Qi Xiang
- Institute of Biomedicine and Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou 510632, China (Y.H.)
| | - Qihao Zhang
- Institute of Biomedicine and Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou 510632, China (Y.H.)
| | - Yan Yang
- Institute of Biomedicine and Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou 510632, China (Y.H.)
| | - Yueping Zhao
- Department of Stomatology, Jinan University Medical College, Guangzhou 510632, China;
| |
Collapse
|
40
|
Hua J, Shen N, Wang J, Tao Y, Li F, Chen Q, Zhou X. Small Molecule-Based Strategy Promotes Nucleus Pulposus Specific Differentiation of Adipose-Derived Mesenchymal Stem Cells. Mol Cells 2019; 42:661-671. [PMID: 31564076 PMCID: PMC6776160 DOI: 10.14348/molcells.2019.0098] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Revised: 08/06/2019] [Accepted: 08/21/2019] [Indexed: 12/28/2022] Open
Abstract
Adipose tissue-derived mesenchymal stem cells (ADSCs) are promising for regenerating degenerated intervertebral discs (IVDs), but the low efficiency of nucleus pulposus (NP)-specific differentiation limits their clinical applications. The Sonic hedgehog (Shh) signaling pathway is important in NP-specific differentiation of ADSCs, and Smoothened Agonist (SAG) is a highly specific and effective agonist of Shh signaling. In this study, we proposed a new differentiation strategy with the use of the small molecule SAG. The NP-specific differentiation and extracellular matrix (ECM) synthesis of ADSCs were measured in vitro , and the regenerative effects of SAG pretreated ADSCs in degenerated IVDs were verified in vivo . The results showed that the combination of SAG and transforming growth factor-β3 (TGF-β3) is able to increase the ECM synthesis of ADSCs. In addition, the gene and protein expression levels of NP-specific markers were increased by treatment with SAG and TGF-β3. Furthermore, SAG pretreated ADSCs can also improve the disc height, water content, ECM content, and structure of degenerated IVDs in vivo . Our new differentiation scheme has high efficiency in inducing NP-specific differentiation of ADSCs and is promising for stem cell-based treatment of degenerated IVDs.
Collapse
Affiliation(s)
- Jianming Hua
- Department of Radiology, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009,
China
| | - Ning Shen
- Department of Rheumatology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016,
China
| | - Jingkai Wang
- Department of Orthopedics Surgery, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009,
China
- Orthopedics Research Institute of Zhejiang University, Hangzhou 310009,
China
| | - Yiqing Tao
- Department of Orthopedics Surgery, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009,
China
- Orthopedics Research Institute of Zhejiang University, Hangzhou 310009,
China
| | - Fangcai Li
- Department of Orthopedics Surgery, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009,
China
- Orthopedics Research Institute of Zhejiang University, Hangzhou 310009,
China
| | - Qixin Chen
- Department of Orthopedics Surgery, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009,
China
- Orthopedics Research Institute of Zhejiang University, Hangzhou 310009,
China
| | - Xiaopeng Zhou
- Department of Orthopedics Surgery, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009,
China
- Orthopedics Research Institute of Zhejiang University, Hangzhou 310009,
China
| |
Collapse
|
41
|
Zhang S, Hu P, Liu T, Li Z, Huang Y, Liao J, Hamid MR, Wen L, Wang T, Mo C, Alini M, Grad S, Wang T, Chen D, Zhou G. Kartogenin hydrolysis product 4-aminobiphenyl distributes to cartilage and mediates cartilage regeneration. Theranostics 2019; 9:7108-7121. [PMID: 31695756 PMCID: PMC6831301 DOI: 10.7150/thno.38182] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 08/10/2019] [Indexed: 12/14/2022] Open
Abstract
Rationale The small molecule Kartogenin (KGN) promotes cartilage regeneration in osteoarthritis (OA) by activating stem cells differentiation, but its pharmacological mode-of-action remains unclear. KGN can be cleaved into 4-aminobiphenyl (4-ABP) and phthalic acid (PA) following enzymolysis of an amide bond. Therefore, this study investigated whether 4-ABP or PA exerted the same action as KGN. Methods KGN, 4-ABP and PA were analyzed in cartilage of mice after oral, intravenous or intra-articular administration of KGN by liquid chromatography-mass spectrometry method. Their effect on proliferation and chondrogenic differentiation of mesenchymal stem cells (MSC) was evaluated in vitro. Furthermore, their effect on cartilage preservation was tested in mice OA model induced by destabilization of medial meniscus. OA severity was quantified using OARSI histological scoring. Transcriptional analysis was used to find the possible targets of the chemicals, which were further validated. Results We demonstrated that while oral or intra-articular KGN delivery effectively ameliorated OA phenotypes in mice, only 4-ABP was detectable in cartilage. 4-ABP could induce chondrogenic differentiation and proliferation of MSC in vitro and promote cartilage repair in OA mouse models mainly by increasing the number of CD44+/CD105+ stem-cell and prevention of matrix loss. These effect of 4-ABP was stronger than that of KGN. Transcriptional profiling of 4-ABP-stimulated MSC suggested that RPS6KA2 and the PI3K-Akt pathway were 4-ABP targets; 4-ABP could activate the PI3K-Akt pathway to promote MSC proliferation and repair OA injury, which was blocked in RPS6KA2-knockdown MSC or RPS6KA2-deficient mice.Conclusion 4-ABP bio-distribution in cartilage promotes proliferation and chondrogenic differentiation of MSC, and repairs osteoarthritic lesions via PI3K-Akt pathway activation.
Collapse
Affiliation(s)
- Shuai Zhang
- Department of Medical Cell Biology and Genetics, Guangdong Key Laboratory of Genomic Stability and Disease Prevention, Shenzhen Key Laboratory of Anti-aging and Regenerative Medicine, and Shenzhen Engineering Laboratory of Regenerative Technologies for Orthopaedic Diseases, Health Sciences Center, Shenzhen University, Shenzhen, 518060, China
| | - Peilin Hu
- Department of Medical Cell Biology and Genetics, Guangdong Key Laboratory of Genomic Stability and Disease Prevention, Shenzhen Key Laboratory of Anti-aging and Regenerative Medicine, and Shenzhen Engineering Laboratory of Regenerative Technologies for Orthopaedic Diseases, Health Sciences Center, Shenzhen University, Shenzhen, 518060, China
| | - Tao Liu
- Department of Medical Cell Biology and Genetics, Guangdong Key Laboratory of Genomic Stability and Disease Prevention, Shenzhen Key Laboratory of Anti-aging and Regenerative Medicine, and Shenzhen Engineering Laboratory of Regenerative Technologies for Orthopaedic Diseases, Health Sciences Center, Shenzhen University, Shenzhen, 518060, China
- Department of Biotherapy and Oncology, Shenzhen Luohu People's Hospital, Shenzhen, 518001, China
| | - Zhen Li
- Department of Medical Cell Biology and Genetics, Guangdong Key Laboratory of Genomic Stability and Disease Prevention, Shenzhen Key Laboratory of Anti-aging and Regenerative Medicine, and Shenzhen Engineering Laboratory of Regenerative Technologies for Orthopaedic Diseases, Health Sciences Center, Shenzhen University, Shenzhen, 518060, China
- AO Research Institute Davos, Davos, Switzerland
| | - Yongcan Huang
- Shenzhen Engineering Laboratory of Orthopaedic Regenerative Technologies, Departments of Orthopaedics, Peking University Shenzhen Hospital, Shenzhen, Guangdong, 510086, China
| | - Jinqi Liao
- Department of Medical Cell Biology and Genetics, Guangdong Key Laboratory of Genomic Stability and Disease Prevention, Shenzhen Key Laboratory of Anti-aging and Regenerative Medicine, and Shenzhen Engineering Laboratory of Regenerative Technologies for Orthopaedic Diseases, Health Sciences Center, Shenzhen University, Shenzhen, 518060, China
| | - Md Rana Hamid
- Department of Medical Cell Biology and Genetics, Guangdong Key Laboratory of Genomic Stability and Disease Prevention, Shenzhen Key Laboratory of Anti-aging and Regenerative Medicine, and Shenzhen Engineering Laboratory of Regenerative Technologies for Orthopaedic Diseases, Health Sciences Center, Shenzhen University, Shenzhen, 518060, China
| | - Liru Wen
- Department of Medical Cell Biology and Genetics, Guangdong Key Laboratory of Genomic Stability and Disease Prevention, Shenzhen Key Laboratory of Anti-aging and Regenerative Medicine, and Shenzhen Engineering Laboratory of Regenerative Technologies for Orthopaedic Diseases, Health Sciences Center, Shenzhen University, Shenzhen, 518060, China
| | - Ting Wang
- Shenzhen Apls Cell Technologies LTD., Yinxing Scientific Building, Lonhua District, Shenzhen, 510086, China
| | - Cuiping Mo
- Shenzhen Apls Cell Technologies LTD., Yinxing Scientific Building, Lonhua District, Shenzhen, 510086, China
| | - Mauro Alini
- AO Research Institute Davos, Davos, Switzerland
| | | | - Tianfu Wang
- Guangdong Key Laboratory for Biomedical Measurements & Ultrasound Imaging, School of Biomedical Engineering, Health Sciences Center, Shenzhen University, Shenzhen, 518060, China
| | - Di Chen
- Department of Orthopedic Surgery, Rush University Medical Center, Chicago, IL 60612, USA
| | - Guangqian Zhou
- Department of Medical Cell Biology and Genetics, Guangdong Key Laboratory of Genomic Stability and Disease Prevention, Shenzhen Key Laboratory of Anti-aging and Regenerative Medicine, and Shenzhen Engineering Laboratory of Regenerative Technologies for Orthopaedic Diseases, Health Sciences Center, Shenzhen University, Shenzhen, 518060, China
| |
Collapse
|