1
|
Wang L, Chen X, Shi S, Yang X, Chen H, Xiao J. Advanced collagen-based scaffolds for cartilage and osteochondral regeneration: A review. Int J Biol Macromol 2025; 311:143992. [PMID: 40348245 DOI: 10.1016/j.ijbiomac.2025.143992] [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: 01/09/2025] [Revised: 04/21/2025] [Accepted: 05/05/2025] [Indexed: 05/14/2025]
Abstract
Osteoarthritis (OA), a prevalent degenerative joint disease, presents a formidable challenge to human health due to its complex pathophysiology. Despite various clinical treatments, a definitive cure for OA remains elusive, leaving patients with only symptomatic relief. Tissue engineering has emerged as a promising approach for OA treatment, offering the potential to restore damaged cartilage and osteochondral tissues. Collagen-based scaffolds, renowned for their superior biocompatibility and bioactivity, hold significant potential in promoting effective cartilage and osteochondral regeneration. Over the past decades, substantial progress has been made in the design and clinical translation of collagen-based scaffolds for cartilage and osteochondral tissue engineering. However, no comprehensive review has yet addressed the application of collagen scaffold materials for OA treatment. This review highlights the advanced fabrication of collagen-based scaffolds, including porous matrices, hydrogels, and microspheres, and their integration with cells, growth factors, and pharmaceuticals for OA therapy. Additionally, it examines the clinical translation of collagen-integrated constructs for managing OA. With continued innovation, collagen-enriched scaffolds are expected to play a pivotal role in improving outcomes for OA patients.
Collapse
Affiliation(s)
- Lili Wang
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, PR China; School of Life Science, Lanzhou University, Lanzhou 730000, PR China; Gansu Engineering Research Center of Medical Collagen, Lanzhou, 730000, PR China
| | - Xian Chen
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, PR China; Gansu Engineering Research Center of Medical Collagen, Lanzhou, 730000, PR China
| | - Shuangni Shi
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, PR China; Gansu Engineering Research Center of Medical Collagen, Lanzhou, 730000, PR China
| | - Xiaxia Yang
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, PR China; Gansu Engineering Research Center of Medical Collagen, Lanzhou, 730000, PR China
| | - Hongli Chen
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, PR China
| | - Jianxi Xiao
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, PR China; Gansu Engineering Research Center of Medical Collagen, Lanzhou, 730000, PR China.
| |
Collapse
|
2
|
Xiong X, Huang H, Wang N, Zhou K, Song X. Sirt1 overexpression inhibits chondrocyte ferroptosis via Ftl deacetylation to suppress the development of osteoarthritis. J Bone Miner Metab 2025; 43:203-215. [PMID: 39786573 DOI: 10.1007/s00774-024-01574-8] [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: 08/01/2024] [Accepted: 12/09/2024] [Indexed: 01/12/2025]
Abstract
INTRODUCTION Osteoarthritis (OA) is a chronic degenerative joint disorder characterized by an imbalance in chondrocyte metabolism. Ferroptosis has been implicated in the pathogenesis of OA. The role of Sirt1, a deacetylase, in mediating deacetylation during ferroptosis in OA chondrocytes remains underexplored. This study aimed to elucidate the mechanisms by which Sirt1 influences chondrocyte ferroptosis in the development of OA. MATERIALS AND METHODS In vitro and in vivo models of OA were established using IL-1β-induced mouse chondrocytes and a destabilization of the medial meniscus (DMM) mouse model, respectively. Ferroptosis was evaluated through measurements of cell viability, lactate dehydrogenase (LDH) release, intracellular levels of Fe2+, glutathione (GSH), malondialdehyde (MDA), lipid reactive oxygen species (ROS), propidium iodide staining, and Western blot analysis. The underlying mechanisms were further investigated using quantitative real-time polymerase chain reaction, Western blotting, immunoprecipitation (IP), co-immunoprecipitation (Co-IP), and glutathione-S-transferase pulldown assays. In vivo validation was performed via Safranin O staining. RESULTS IL-1β induced ferroptosis and increased histone acetylation, effects that were partially reversed by Sirt1 overexpression. Mechanistically, Sirt1 overexpression upregulated ferritin light polypeptide (Ftl) expression by deacetylating Ftl at the K181 residue. Ftl knockdown inhibited the ferroptosis-enhancing effect of Sirt1 overexpression in chondrocytes. In vivo studies showed that Sirt1 overexpression mitigated the progression of OA and reduced ferroptosis in the DMM-induced OA mouse model. CONCLUSION Our findings confirm that Sirt1 overexpression promotes Ftl expression through deacetylation at the K181 site, thereby suppressing chondrocyte ferroptosis and attenuating the progression of OA. These results suggest a potential therapeutic target for OA treatment.
Collapse
Affiliation(s)
- Xiaolong Xiong
- Department of Sports Medicine, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Hui Huang
- Department of Sports Medicine, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Ning Wang
- Department of Sports Medicine, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Kai Zhou
- Department of Sports Medicine, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Xinghui Song
- Universiti Kebangsaan Malaysia Health Science, UKM, 43600, Bandar Baru Bangi, Selangor, Malaysia.
| |
Collapse
|
3
|
Liang J, Huang X, Qin K, Wei H, Yang J, Liu B, Fan Z. Implanted Magnetoelectric Bionic Cartilage Hydrogel. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2415417. [PMID: 40134358 DOI: 10.1002/adma.202415417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 02/24/2025] [Indexed: 03/27/2025]
Abstract
Enhancing defective cartilage repair by creating a bionic cartilage hydrogel supplemented with in situ electromagnetic stimulation, replicating endogenous electromagnetic effects, remains challenging. To achieve this, a unique three-phase solvent system is designed to prepare a magnetoelectric bionic cartilage hydrogel incorporating piezoelectric poly(3-hydroxybutyric acid-3-hydroxyvaleric acid) (PHBV) and magnetostrictive triiron tetraoxide nanoparticles (Fe3O4 NPs) into sodium alginate (SA) hydrogel to form a dual-network, semi-crosslinked chain entanglement structure. The synthesized hydrogel features similar composition, structure, and mechanical properties to natural cartilage. In addition, after the implantation of cartilage, the motion-driven magnetoelectric-coupled cyclic transformation model is triggered by gentle joint forces, initiating a piezoelectric response that leads to magnetoelectric-coupled cyclic transformation. The freely excitable and cyclically enhanced electromagnetic stimulation it can provide, by simulating and amplifying endogenous electromagnetic effects, obtains induced defective cartilage repair efficacy superior to piezoelectric or magnetic stimulation alone.
Collapse
Affiliation(s)
- Jiachen Liang
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, Gansu Province, School of Stomatology, Lanzhou University, Lanzhou, 730000, P.R. China
| | - Xinyue Huang
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, Gansu Province, School of Stomatology, Lanzhou University, Lanzhou, 730000, P.R. China
| | - Kaiqi Qin
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, Gansu Province, School of Stomatology, Lanzhou University, Lanzhou, 730000, P.R. China
| | - Hui Wei
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, Gansu Province, School of Stomatology, Lanzhou University, Lanzhou, 730000, P.R. China
| | - Jiaxin Yang
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, Gansu Province, School of Stomatology, Lanzhou University, Lanzhou, 730000, P.R. China
| | - Bin Liu
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, Gansu Province, School of Stomatology, Lanzhou University, Lanzhou, 730000, P.R. China
| | - Zengjie Fan
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, Gansu Province, School of Stomatology, Lanzhou University, Lanzhou, 730000, P.R. China
| |
Collapse
|
4
|
Jin SC, Choi YY, Song M, Baek HK, Yi S, Kim E, Yang WM. Osteo-F, a Newly Developed Herbal Formula, Ameliorates Osteoarthritis Through the NF-κB/IκB/JNK Pathway Based on Network Pharmacology. Food Sci Nutr 2025; 13:e70239. [PMID: 40357339 PMCID: PMC12066244 DOI: 10.1002/fsn3.70239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 04/17/2025] [Accepted: 04/21/2025] [Indexed: 05/15/2025] Open
Abstract
Osteoarthritis (OA) is a painful joint condition primarily caused by cartilage degradation, leading to pain and reduced mobility. Given the side effects of current treatments, this study investigates Osteo-F, a novel herbal-based functional ingredient formulated with Schizandra chinensis, Lycium chinense, and Eucommia ulmoides, traditionally valued for their bioactive and health-promoting properties. Network pharmacology analysis identified significant interactions involving Osteo-F within the TNF signaling pathway, highlighting its role in modulating key inflammatory processes in OA. In vivo experiments using a monosodium iodoacetate-induced OA rat model demonstrated significant improvements in arthritis scores, bone mineral content, and bone mineral density, alongside preservation of cartilage integrity, as confirmed by histological analyses. In vitro studies further revealed that this formulation reduced the activation of JNK and NF-κB pathways, decreasing inflammatory cytokines and matrix metalloproteinases critical in cartilage breakdown. These findings underscore the potential of Osteo-F as a functional food candidate to reduce inflammation and support cartilage preservation in OA. Future clinical trials are required to validate these findings and explore its dietary integration in OA management.
Collapse
Affiliation(s)
- Seong Chul Jin
- Department of Convergence Korean Medical Science, College of Korean MedicineKyung Hee UniversitySeoulRepublic of Korea
| | - You Yeon Choi
- Department of Convergence Korean Medical Science, College of Korean MedicineKyung Hee UniversitySeoulRepublic of Korea
| | - Minwoo Song
- Department of Convergence Korean Medical Science, College of Korean MedicineKyung Hee UniversitySeoulRepublic of Korea
| | - Hee Kyung Baek
- KHU‐KIST Department of Converging Science and TechnologyCollege of Korean Medicine, Graduate School, Kyung Hee UniversitySeoulRepublic of Korea
| | - Seungyob Yi
- KHU‐KIST Department of Converging Science and TechnologyCollege of Korean Medicine, Graduate School, Kyung Hee UniversitySeoulRepublic of Korea
| | - Eun‐Jung Kim
- Department of Acupuncture and Moxibustion, Graduate School of Oriental MedicineDongguk UniversityGoyang‐siGyeonggi‐doKorea
| | - Woong Mo Yang
- Department of Convergence Korean Medical Science, College of Korean MedicineKyung Hee UniversitySeoulRepublic of Korea
- Korean Medicine Digital Convergence Center (KMDC)Kyung Hee UniversitySeoulRepublic of Korea
| |
Collapse
|
5
|
Tsutsumi-Arai C, Tran A, Arai Y, Ono W, Ono N. Mandibular Condylar Cartilage in Development and Diseases: A PTHrP-Centric View. Orthod Craniofac Res 2025. [PMID: 40251915 DOI: 10.1111/ocr.12936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Revised: 12/25/2024] [Accepted: 04/12/2025] [Indexed: 04/21/2025]
Abstract
The mandibular condylar cartilage (MCC) is a dual-function component of the temporomandibular joint (TMJ), acting as both articular cartilage for jaw movement and growth cartilage for vertical growth of the mandibular condyle. Parathyroid hormone-related protein (PTHrP) plays a critical role in orchestrating chondrogenesis in the long bone, and its importance is also highlighted in both MCC development and TMJ function. Here, we discuss the role of PTHrP in the development, growth and diseases of the MCC. PTHrP is a key morphogen in the MCC that regulates chondrogenesis by promoting chondrocyte proliferation and preventing premature hypertrophic differentiation. Exclusively expressed in the superficial layer, PTHrP diffuses across the MCC and targets chondrocytes in deeper layers, regulating transcription factors such as RUNX2 and SOX9. PTHrP regulates chondrocyte differentiation through two main pathways: the PTHrP-PTH1R signalling pathway, which suppresses hypertrophy and the PTHrP-Ihh negative feedback loop, which balances proliferation and hypertrophy. In the postnatal murine MCC, PTHrP levels are high early on and decrease after the onset of mastication around P21. Altered mechanical environments, such as those therapeutically induced as mandibular advancement, increase PTHrP expression, promoting chondrocyte proliferation and delaying hypertrophy. PTHrP also plays a dual role in adult TMJ diseases, particularly in osteoarthritis (OA); PTHrP expression transiently increases during the early stages of TMJ-OA to promote cell proliferation, but its eventual decrease contributes to the progression of the disease. This highlights the complex role of PTHrP in maintaining MCC homeostasis and its potential involvement in TMJ-OA pathology. The MCC combines the characteristics of growth and articular cartilage and functions distinctively in three phases: development before occlusion, growth after the occlusion is established, and maintenance after the growth is complete. While PTHrP plays a multifaceted role in all phases, further research is needed to fully understand how it regulates MCC development, growth and diseases.
Collapse
Affiliation(s)
- Chiaki Tsutsumi-Arai
- University of Texas Health Science Center at Houston School of Dentistry, Houston, Texas, USA
| | - Amy Tran
- University of Texas Health Science Center at Houston School of Dentistry, Houston, Texas, USA
| | - Yuki Arai
- University of Texas Health Science Center at Houston School of Dentistry, Houston, Texas, USA
| | - Wanida Ono
- University of Texas Health Science Center at Houston School of Dentistry, Houston, Texas, USA
| | - Noriaki Ono
- University of Texas Health Science Center at Houston School of Dentistry, Houston, Texas, USA
| |
Collapse
|
6
|
Fan X, Ong LJY, Sun AR, Prasadam I. From polarity to pathology: Decoding the role of cell orientation in osteoarthritis. J Orthop Translat 2024; 49:62-73. [PMID: 39430130 PMCID: PMC11488446 DOI: 10.1016/j.jot.2024.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 09/10/2024] [Accepted: 09/14/2024] [Indexed: 10/22/2024] Open
Abstract
UNLABELLED Cell polarity refers to the orientation of tissue and organelles within a cell and the direction of its function. It is one of the most critical characteristics of metazoans. The development, growth, and functional tissue distribution are closely related to holistic tissue or organ homeostasis. However, the connection between cell polarity and osteoarthritis (OA) is less well-known. In OA, multiple chondrocyte clusters and tissue disorganisation can be observed in the degraded cartilage tissue. The excessive upregulation of the planar cell polarity (PCP) signalling pathway leads to the loss of cell polarity and organisation in OA progression and aetiology. Recent research has become increasingly aware of the importance of cell polarity and its correlation with OA. Several cell polarity-related treatments have shed light on OA. A thorough understanding of cell polarity and OA would provide more insights for future investigations to treat this worldwide disease. THE TRANSLATIONAL POTENTIAL OF THIS ARTICLE Understanding cell polarity, associated signalling pathways, organelle changes, and cell movement in the development of OA could lead to advances in precision medicine and enhanced treatment strategies for OA patients.
Collapse
Affiliation(s)
- Xiwei Fan
- Department of Orthopaedic Surgery, The Second Xiangya Hospital of Central South University, Changsha, China
- School of Mechanical, Medical & Process Engineering, Queensland University of Technology, Brisbane, Australia
- Centre for Biomedical Technologies, Queensland University of Technology, Brisbane, Australia
| | - Louis Jun Ye Ong
- School of Mechanical, Medical & Process Engineering, Queensland University of Technology, Brisbane, Australia
- Centre for Biomedical Technologies, Queensland University of Technology, Brisbane, Australia
- Max Planck Queensland Centre (MPQC) for the Materials Science of Extracellular Matrices, Queensland University of Technology, Brisbane, Australia
| | - Antonia RuJia Sun
- School of Mechanical, Medical & Process Engineering, Queensland University of Technology, Brisbane, Australia
- Centre for Biomedical Technologies, Queensland University of Technology, Brisbane, Australia
| | - Indira Prasadam
- School of Mechanical, Medical & Process Engineering, Queensland University of Technology, Brisbane, Australia
- Centre for Biomedical Technologies, Queensland University of Technology, Brisbane, Australia
| |
Collapse
|
7
|
Singer J, Knezic N, Layne J, Gohring G, Christiansen J, Rothrauff B, Huard J. Enhancing Cartilage Repair: Surgical Approaches, Orthobiologics, and the Promise of Exosomes. Life (Basel) 2024; 14:1149. [PMID: 39337932 PMCID: PMC11432843 DOI: 10.3390/life14091149] [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: 07/28/2024] [Revised: 08/22/2024] [Accepted: 08/30/2024] [Indexed: 09/30/2024] Open
Abstract
Treating cartilage damage is challenging as its ability for self-regeneration is limited. Left untreated, it can progress to osteoarthritis (OA), a joint disorder characterized by the deterioration of articular cartilage and other joint tissues. Surgical options, such as microfracture and cell/tissue transplantation, have shown promise as techniques to harness the body's endogenous regenerative capabilities to promote cartilage repair. Nonetheless, these techniques have been scrutinized due to reported inconsistencies in long-term outcomes and the tendency for the defects to regenerate as fibrocartilage instead of the smooth hyaline cartilage native to joint surfaces. Orthobiologics are medical therapies that utilize biologically derived substances to augment musculoskeletal healing. These treatments are rising in popularity because of their potential to enhance surgical standards of care. More recent developments in orthobiologics have focused on the role of exosomes in articular cartilage repair. Exosomes are nano-sized extracellular vesicles containing cargo such as proteins, lipids, and nucleic acids, and are known to facilitate intercellular communication, though their regenerative potential still needs to be fully understood. This review aims to demonstrate the advancements in cartilage regeneration, highlight surgical and biological treatment options, and discuss the recent strides in understanding the precise mechanisms of action involved.
Collapse
Affiliation(s)
- Jacob Singer
- Linda and Mitch Hart Regenerative and Personalized Medicine, Steadman Philippon Research Institute, Vail, CO 81657, USA
| | - Noah Knezic
- Linda and Mitch Hart Regenerative and Personalized Medicine, Steadman Philippon Research Institute, Vail, CO 81657, USA
| | - Jonathan Layne
- Linda and Mitch Hart Regenerative and Personalized Medicine, Steadman Philippon Research Institute, Vail, CO 81657, USA
| | - Greta Gohring
- Linda and Mitch Hart Regenerative and Personalized Medicine, Steadman Philippon Research Institute, Vail, CO 81657, USA
| | - Jeff Christiansen
- Linda and Mitch Hart Regenerative and Personalized Medicine, Steadman Philippon Research Institute, Vail, CO 81657, USA
| | - Ben Rothrauff
- Linda and Mitch Hart Regenerative and Personalized Medicine, Steadman Philippon Research Institute, Vail, CO 81657, USA
| | - Johnny Huard
- Linda and Mitch Hart Regenerative and Personalized Medicine, Steadman Philippon Research Institute, Vail, CO 81657, USA
| |
Collapse
|
8
|
Krakowski P, Rejniak A, Sobczyk J, Karpiński R. Cartilage Integrity: A Review of Mechanical and Frictional Properties and Repair Approaches in Osteoarthritis. Healthcare (Basel) 2024; 12:1648. [PMID: 39201206 PMCID: PMC11353818 DOI: 10.3390/healthcare12161648] [Citation(s) in RCA: 58] [Impact Index Per Article: 58.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 08/09/2024] [Accepted: 08/14/2024] [Indexed: 09/02/2024] Open
Abstract
Osteoarthritis (OA) is one of the most common causes of disability around the globe, especially in aging populations. The main symptoms of OA are pain and loss of motion and function of the affected joint. Hyaline cartilage has limited ability for regeneration due to its avascularity, lack of nerve endings, and very slow metabolism. Total joint replacement (TJR) has to date been used as the treatment of end-stage disease. Various joint-sparing alternatives, including conservative and surgical treatment, have been proposed in the literature; however, no treatment to date has been fully successful in restoring hyaline cartilage. The mechanical and frictional properties of the cartilage are of paramount importance in terms of cartilage resistance to continuous loading. OA causes numerous changes in the macro- and microstructure of cartilage, affecting its mechanical properties. Increased friction and reduced load-bearing capability of the cartilage accelerate further degradation of tissue by exerting increased loads on the healthy surrounding tissues. Cartilage repair techniques aim to restore function and reduce pain in the affected joint. Numerous studies have investigated the biological aspects of OA progression and cartilage repair techniques. However, the mechanical properties of cartilage repair techniques are of vital importance and must be addressed too. This review, therefore, addresses the mechanical and frictional properties of articular cartilage and its changes during OA, and it summarizes the mechanical outcomes of cartilage repair techniques.
Collapse
Affiliation(s)
- Przemysław Krakowski
- Department of Trauma Surgery and Emergency Medicine, Medical University, 20-059 Lublin, Poland
- Orthopaedic and Sports Traumatology Department, Carolina Medical Center, Pory 78, 02-757 Warsaw, Poland; (A.R.); (J.S.)
| | - Adrian Rejniak
- Orthopaedic and Sports Traumatology Department, Carolina Medical Center, Pory 78, 02-757 Warsaw, Poland; (A.R.); (J.S.)
| | - Jakub Sobczyk
- Orthopaedic and Sports Traumatology Department, Carolina Medical Center, Pory 78, 02-757 Warsaw, Poland; (A.R.); (J.S.)
| | - Robert Karpiński
- Department of Machine Design and Mechatronics, Faculty of Mechanical Engineering, University of Technology, 20-618 Lublin, Poland
- Department of Psychiatry, Psychotherapy and Early Intervention, Medical University, 20-059 Lublin, Poland
| |
Collapse
|
9
|
Mamachan M, Sharun K, Banu SA, Muthu S, Pawde AM, Abualigah L, Maiti SK. Mesenchymal stem cells for cartilage regeneration: Insights into molecular mechanism and therapeutic strategies. Tissue Cell 2024; 88:102380. [PMID: 38615643 DOI: 10.1016/j.tice.2024.102380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 03/15/2024] [Accepted: 04/09/2024] [Indexed: 04/16/2024]
Abstract
The use of mesenchymal stem cells (MSCs) in cartilage regeneration has gained significant attention in regenerative medicine. This paper reviews the molecular mechanisms underlying MSC-based cartilage regeneration and explores various therapeutic strategies to enhance the efficacy of MSCs in this context. MSCs exhibit multipotent capabilities and can differentiate into various cell lineages under specific microenvironmental cues. Chondrogenic differentiation, a complex process involving signaling pathways, transcription factors, and growth factors, plays a pivotal role in the successful regeneration of cartilage tissue. The chondrogenic differentiation of MSCs is tightly regulated by growth factors and signaling pathways such as TGF-β, BMP, Wnt/β-catenin, RhoA/ROCK, NOTCH, and IHH (Indian hedgehog). Understanding the intricate balance between these pathways is crucial for directing lineage-specific differentiation and preventing undesirable chondrocyte hypertrophy. Additionally, paracrine effects of MSCs, mediated by the secretion of bioactive factors, contribute significantly to immunomodulation, recruitment of endogenous stem cells, and maintenance of chondrocyte phenotype. Pre-treatment strategies utilized to potentiate MSCs, such as hypoxic conditions, low-intensity ultrasound, kartogenin treatment, and gene editing, are also discussed for their potential to enhance MSC survival, differentiation, and paracrine effects. In conclusion, this paper provides a comprehensive overview of the molecular mechanisms involved in MSC-based cartilage regeneration and outlines promising therapeutic strategies. The insights presented contribute to the ongoing efforts in optimizing MSC-based therapies for effective cartilage repair.
Collapse
Affiliation(s)
- Merlin Mamachan
- Division of Surgery, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, India
| | - Khan Sharun
- Division of Surgery, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, India; Graduate Institute of Medicine, Yuan Ze University, Taoyuan, Taiwan.
| | - S Amitha Banu
- Division of Surgery, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, India
| | - Sathish Muthu
- Department of Biotechnology, Faculty of Engineering, Karpagam Academy of Higher Education, Coimbatore, Tamil Nadu, India; Orthopaedic Research Group, Coimbatore, Tamil Nadu, India; Department of Orthopaedics, Government Medical College, Kaur, Tamil Nadu, India
| | - Abhijit M Pawde
- Division of Surgery, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, India
| | - Laith Abualigah
- Artificial Intelligence and Sensing Technologies (AIST) Research Center, University of Tabuk, Tabuk 71491, Saudi Arabia; Hourani Center for Applied Scientific Research, Al-Ahliyya Amman University, Amman 19328, Jordan; Computer Science Department, Al al-Bayt University, Mafraq 25113, Jordan; MEU Research Unit, Middle East University, Amman 11831, Jordan; Department of Electrical and Computer Engineering, Lebanese American University, Byblos 13-5053, Lebanon; Applied Science Research Center, Applied Science Private University, Amman 11931, Jordan; School of Engineering and Technology, Sunway University Malaysia, Petaling Jaya 27500, Malaysia
| | - Swapan Kumar Maiti
- Division of Surgery, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, India
| |
Collapse
|
10
|
Ignatyeva N, Gavrilov N, Timashev PS, Medvedeva EV. Prg4-Expressing Chondroprogenitor Cells in the Superficial Zone of Articular Cartilage. Int J Mol Sci 2024; 25:5605. [PMID: 38891793 PMCID: PMC11171992 DOI: 10.3390/ijms25115605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 05/12/2024] [Accepted: 05/16/2024] [Indexed: 06/21/2024] Open
Abstract
Joint-resident chondrogenic precursor cells have become a significant therapeutic option due to the lack of regenerative capacity in articular cartilage. Progenitor cells are located in the superficial zone of the articular cartilage, producing lubricin/Prg4 to decrease friction of cartilage surfaces during joint movement. Prg4-positive progenitors are crucial in maintaining the joint's structure and functionality. The disappearance of progenitor cells leads to changes in articular hyaline cartilage over time, subchondral bone abnormalities, and the formation of ectopic ossification. Genetic labeling cell technology has been the main tool used to characterize Prg4-expressing progenitor cells of articular cartilage in vivo through drug injection at different time points. This technology allows for the determination of the origin of progenitor cells and the tracking of their progeny during joint development and cartilage damage. We endeavored to highlight the currently known information about the Prg4-producing cell population in the joint to underline the significance of the role of these cells in the development of articular cartilage and its homeostasis. This review focuses on superficial progenitors in the joint, how they contribute to postnatal articular cartilage formation, their capacity for regeneration, and the consequences of Prg4 deficiency in these cells. We have accumulated information about the Prg4+ cell population of articular cartilage obtained through various elegantly designed experiments using transgenic technologies to identify potential opportunities for further research.
Collapse
Affiliation(s)
- Nadezda Ignatyeva
- Institute for Regenerative Medicine, Sechenov First Moscow State Medical University (Sechenov University), 8-2 Trubetskaya St., Moscow 119048, Russia; (N.G.); (P.S.T.); (E.V.M.)
| | | | | | | |
Collapse
|
11
|
Xu J, Zhang Q, Jiang T, Liu L, Gu H, Tan Y, Wang H. Dose- and stage-dependent toxic effects of prenatal prednisone exposure on fetal articular cartilage development. Toxicol Lett 2024; 393:14-23. [PMID: 38211732 DOI: 10.1016/j.toxlet.2024.01.004] [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: 09/10/2023] [Revised: 12/16/2023] [Accepted: 01/06/2024] [Indexed: 01/13/2024]
Abstract
Prednisone is frequently used to treat rheumatoid diseases in pregnant women because of its high degree of safety. Whether prenatal prednisone exposure (PPE) negatively impacts fetal articular cartilage development is unclear. In this study, we simulated a clinical prednisone treatment regimen to examine the effects of different timings and doses of PPE on cartilage development in female and male fetal mice. Prednisone doses (0.25, 0.5, and 1 mg/kg/d) was administered to Kunming mice at different gestational stages (0-9 gestational days, GD0-9), mid-late gestation (GD10-18), or during the entire gestation (GD0-18) by oral gavage. The amount of matrix aggrecan (ACAN) and collagen type II a1(COL2a1), and expression of transforming growth factor β1 (TGFβ1) signaling pathway also demonstrated that the chondrocyte count and ACAN and COL2a1 expression reduced in fetal mice with early and mid-late PPE, with the reduction being more significant in the mice with early PPE than that in those with PPE at other stages. Prenatal exposure to different prednisone doses prevented the reduction of TGFβ signaling pathway-related genes [TGFβR1, SMAD family member 3 (Smad3), SRY-box9 (SOX9)] as well as ACAN and COL2a1 mRNA expression levels in fetal mouse cartilage, with the most significant decrease after 1 mg/kg·d PPE. In conclusion, PPE can inhibit/restrain fetal cartilage development, with the greatest effect at higher clinical dose (1 mg/kg·d) and early stage of pregnancy (GD0-9), and the mechanism may be related to TGFβ signaling pathway inhibition. The result of this study provide a theoretical and experimental foundation for the rational clinical use of prednisone.
Collapse
Affiliation(s)
- Junmiao Xu
- Division of Joint surgery and sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Qi Zhang
- Division of Joint surgery and sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Tao Jiang
- Division of Joint surgery and sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Liang Liu
- Division of Joint surgery and sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Hanwen Gu
- Division of Joint surgery and sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Yang Tan
- Division of Joint surgery and sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, China.
| | - Hui Wang
- Department of Pharmacology, Basic Medical School of Wuhan University, Wuhan 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, China.
| |
Collapse
|
12
|
Ladner YD, Stoddart MJ. Tribological loading of cartilage and chondrogenic cells. BIOMATERIALS AND BIOSYSTEMS 2024; 13:100088. [PMID: 38389863 PMCID: PMC10881303 DOI: 10.1016/j.bbiosy.2024.100088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 12/06/2023] [Accepted: 02/05/2024] [Indexed: 02/24/2024] Open
Abstract
Novel cartilage regeneration therapies often look promising in-vitro but fail when implanted in vivo. One of the possible reasons for this discrepancy is the simplified, static in-vitro chondrogenesis models typically used. Complex mechanical stimulation plays a key role in physiological cartilage and chondrogenic cell metabolism, including the development of cartilage structure, yet it is routinely lacking during in-vitro studies. Multiaxial load bioreactors are becoming more widespread and offer advantages over more simple loading devices. Within this article, we highlight some of the important findings from in-vitro assays and key aspects relating to tribological loading of cartilage and chondrogenic cells.
Collapse
Affiliation(s)
- Yann D Ladner
- AO Research Institute Davos, Clavadelerstrasse 8, 7270 Davos Platz, Switzerland
| | - Martin J Stoddart
- AO Research Institute Davos, Clavadelerstrasse 8, 7270 Davos Platz, Switzerland
| |
Collapse
|
13
|
Francis DV, Rajeswari AJ, Stephen JB, Parasuraman G, Lisha J J, Livingston A, Rani S, Daniel AJ, Sathishkumar S, Vinod E. An ultrastructural report of human articular cartilage resident cells in correlation with their phenotypic characteristics. J Histotechnol 2024; 47:23-38. [PMID: 37966827 DOI: 10.1080/01478885.2023.2278118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 10/26/2023] [Indexed: 11/16/2023]
Abstract
The recent discovery of progenitors based on their differential fibronectin-adhesion (FAA-CPs) and migratory-based (MCPs) assay has evoked interest due to their superiority in terms of their efficient chondrogenesis and reduced hypertrophic propensity. This study aims to isolate and enrich three articular cartilage subsets, chondrocytes, FAA-CPs, and MCPs, and compare their undifferentiated and chondrogenic differentiated status, using in-vitro phenotypical characterization in correlation with ultrastructural analysis using Transmission Electron Microscopy (TEM). Following informed consent, cartilage shavings were procured from a non-diseased human ankle joint and cultured to obtain the three subsets. Chondrocytes exhibited higher CD106 and lower CD49b and CD146 levels. Following chondrogenic differentiation, corroborative results were seen, with the MCP group showing the highest GAG/DNA ratio levels and uptake of extracellular matrix stain as compared to the FAA-CP group. TEM analysis of the chondrocytes revealed the presence of more autolytic cells with disintegrated cytoplasm and plasma membrane. The differentiated FAA-CPs and MCPs displayed higher collagen and rough endoplasmic reticulum. The results presented in this study provide novel information on the ultrastructural characteristics of cartilage resident cells, with the chondrocyte group displaying features of terminal differentiation. Both progenitor subtypes showed superiority in varied contexts, with greater collagen fibrils and greater GAG content in MCPs. The display of preferential and differentiation traits sheds insight on the necessity to enrich progenitors and coculturing them with the general pool of constituent cells to combine their advantages and reduce their drawbacks to achieve a regenerative tissue displaying genuine hyaline-like repair while limiting their terminal differentiation.
Collapse
Affiliation(s)
| | | | | | - Ganesh Parasuraman
- Centre for Stem Cell Research, (A unit of InStem, Bengaluru), Christian Medical College, Vellore, India
| | - Jeya Lisha J
- Department of Physiology, Christian Medical College, Vellore, India
| | - Abel Livingston
- Department of Orthopaedics, Christian Medical College, Vellore, India
| | - Sandya Rani
- Centre for Stem Cell Research, (A unit of InStem, Bengaluru), Christian Medical College, Vellore, India
| | - Alfred Job Daniel
- Department of Orthopaedics, Christian Medical College, Vellore, India
| | | | - Elizabeth Vinod
- Centre for Stem Cell Research, (A unit of InStem, Bengaluru), Christian Medical College, Vellore, India
- Department of Physiology, Christian Medical College, Vellore, India
| |
Collapse
|
14
|
Taghizadeh S, Tayebi L, Akbarzadeh M, Lohrasbi P, Savardashtaki A. Magnetic hydrogel applications in articular cartilage tissue engineering. J Biomed Mater Res A 2024; 112:260-275. [PMID: 37750666 DOI: 10.1002/jbm.a.37620] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 09/02/2023] [Accepted: 09/11/2023] [Indexed: 09/27/2023]
Abstract
Articular cartilage defects afflict millions of individuals worldwide, presenting a significant challenge due to the tissue's limited self-repair capability and anisotropic nature. Hydrogel-based biomaterials have emerged as promising candidates for scaffold production in artificial cartilage construction, owing to their water-rich composition, biocompatibility, and tunable properties. Nevertheless, conventional hydrogels typically lack the anisotropic structure inherent to natural cartilage, impeding their clinical and preclinical applications. Recent advancements in tissue engineering (TE) have introduced magnetically responsive hydrogels, a type of intelligent hydrogel that can be remotely controlled using an external magnetic field. These innovative materials offer a means to create the desired anisotropic architecture required for successful cartilage TE. In this review, we first explore conventional techniques employed for cartilage repair and subsequently delve into recent breakthroughs in the application and utilization of magnetic hydrogels across various aspects of articular cartilage TE.
Collapse
Affiliation(s)
- Saeed Taghizadeh
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
- Pharmaceutical Science Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Lobat Tayebi
- Marquette University School of Dentistry, Milwaukee, Wisconsin, USA
| | - Majid Akbarzadeh
- Department of Internal Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Parvin Lohrasbi
- Department of Reproductive Biology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amir Savardashtaki
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
- Infertility Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
15
|
Zhang F, Liu L, Wang H, Chen L. Effects of prenatal acetaminophen exposure at different stages, doses and courses on articular cartilage of offspring mice. Food Chem Toxicol 2023; 180:114003. [PMID: 37633638 DOI: 10.1016/j.fct.2023.114003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 08/21/2023] [Accepted: 08/22/2023] [Indexed: 08/28/2023]
Abstract
Previous studies showed that chondrodysplasia has intrauterine origin. Although prenatal acetaminophen exposure (PAcE) can cause nervous and reproductive abnormalities in offspring, its effect on cartilage is uninvestigated. Herein, mice were treated with different doses and courses of acetaminophen at various gestational stages (100 or 400 mg/kg∙d on gestational days 10-12 (GD10-12), 400 mg/kg∙d on GD12 or GD15-17) based on clinical administration and conversion between humans and mice. Fetal knee joints were harvested on GD18 to analyze cartilage morphology, chondrocyte proliferation and apoptosis, and matrix content, synthesis and degradation. Results showed that 400 mg/kg∙d acetaminophen exposure during GD10-12 decreased chondrocyte numbers, safranin O staining, proliferation and matrix synthesis, without elevating matrix degradation and apoptosis. Low-dose, single-course, or late-pregnancy exposure had no effect on above indexes. Moreover, Tgfβ pathway was inhibited, showing a positive correlation with the expression of Col2a1, Acan, Ki67, and Pcna. Overall, clinical doses of PAcE can inhibit chondrocyte proliferation and matrix synthesis, causing fetal mice chondrodysplasia, especially after multi-course exposure of 400 mg/kg∙d acetaminophen during GD10-12, the mechanism of which might involve Tgfβ pathway inhibition. This study provides an experimental basis for assessing fetal developmental toxicity and standardizing the clinical use of acetaminophen during pregnancy.
Collapse
Affiliation(s)
- Fan Zhang
- Department of Orthopedic Surgery, Division of Joint Surgery and Sports Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Liang Liu
- Department of Orthopedic Surgery, Division of Joint Surgery and Sports Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Hui Wang
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China.
| | - Liaobin Chen
- Department of Orthopedic Surgery, Division of Joint Surgery and Sports Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China.
| |
Collapse
|
16
|
Liu G, Guo Q, Liu C, Bai J, Wang H, Li J, Liu D, Yu Q, Shi J, Liu C, Zhu C, Li B, Zhang H. Cytomodulin-10 modified GelMA hydrogel with kartogenin for in-situ osteochondral regeneration. Acta Biomater 2023; 169:317-333. [PMID: 37586447 DOI: 10.1016/j.actbio.2023.08.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 07/27/2023] [Accepted: 08/09/2023] [Indexed: 08/18/2023]
Abstract
The incidence of osteochondral defect is increasing year by year, but there is still no widely accepted method for repairing the defect. Hydrogels loaded with bioactive molecules have provided promising alternatives for in-situ osteochondral regeneration. Kartogenin (KGN) is an effective and steady small molecule with the function of cartilage regeneration and protection which can be further boosted by TGF-β. However, the high cost, instability, and immunogenicity of TGF-β would limit its combined effect with KGN in clinical application. In this study, a composite hydrogel CM-KGN@GelMA, which contained TGF-β1 analog short peptide cytomodulin-10 (CM-10) and KGN, was fabricated. The results indicated that CM-10 modified on GelMA hydrogels exerted an equivalent role in enhancing chondrogenesis as TGF-β1, and this effect was also boosted when combined with KGN. Moreover, it was revealed that CM-10 and KGN had a synergistic effect on promoting the chondrogenesis of BMSCs by up-regulating the expression of RUNX1 and SOX9 at both mRNA and protein levels in vitro. Finally, the composite hydrogel exhibited a satisfactory osteochondral defect repair effect in vivo, showing similar structures close to the native tissue. Taken together, this study has revealed that CM-10 may serve as an alternative for TGF-β1 and can collaborate with KGN to accelerate chondrogenesis, which suggests that the fabricated CM-KGN@GelMA composite hydrogel can be acted as a potential scaffold for osteochondral defect regeneration. STATEMENT OF SIGNIFICANCE: Kartogenin and TGF-β have shown great value in promoting osteochondral defect regeneration, and their combined application can enhance the effect and show great potential for clinical application. Herein, a functional CM-KGN@GelMA hydrogel was fabricated, which was composed of TGF-β1 mimicking peptide CM-10 and KGN. CM-10 in hydrogel retained an activity like TGF-β1 to facilitate BMSC chondrogenesis and exhibited boosting chondrogenesis by up-regulating RUNX1 and SOX9 when being co-applied with KGN. In vivo, the hydrogel promoted cartilage regeneration and subchondral bone reconstruction, showing similar structures as the native tissue, which might be vital in recovering the bio-function of cartilage. Thus, this study developed an effective scaffold and provided a promising way for osteochondral defect repair.
Collapse
Affiliation(s)
- Guoping Liu
- Department of Orthopedic Surgery, Medical 3D Printing Center, Orthopedic Institute, the First Affiliated Hospital, School of Biology and Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu 215000, China; Department of Spine Surgery, the Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421000, China
| | - Qianping Guo
- Department of Orthopedic Surgery, Medical 3D Printing Center, Orthopedic Institute, the First Affiliated Hospital, School of Biology and Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu 215000, China
| | - Changjiang Liu
- Department of Orthopedic Surgery, Medical 3D Printing Center, Orthopedic Institute, the First Affiliated Hospital, School of Biology and Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu 215000, China
| | - Jianzhong Bai
- Department of Orthopedic Surgery, Medical 3D Printing Center, Orthopedic Institute, the First Affiliated Hospital, School of Biology and Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu 215000, China
| | - Huan Wang
- Department of Orthopedic Surgery, Medical 3D Printing Center, Orthopedic Institute, the First Affiliated Hospital, School of Biology and Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu 215000, China
| | - Jiaying Li
- Department of Orthopedic Surgery, Medical 3D Printing Center, Orthopedic Institute, the First Affiliated Hospital, School of Biology and Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu 215000, China
| | - Dachuan Liu
- Department of Orthopedic Surgery, Medical 3D Printing Center, Orthopedic Institute, the First Affiliated Hospital, School of Biology and Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu 215000, China
| | - Qifan Yu
- Department of Orthopedic Surgery, Medical 3D Printing Center, Orthopedic Institute, the First Affiliated Hospital, School of Biology and Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu 215000, China
| | - Jinhui Shi
- Department of Orthopedic Surgery, Medical 3D Printing Center, Orthopedic Institute, the First Affiliated Hospital, School of Biology and Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu 215000, China
| | - Chengyuan Liu
- Department of Orthopedic Surgery, Medical 3D Printing Center, Orthopedic Institute, the First Affiliated Hospital, School of Biology and Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu 215000, China
| | - Caihong Zhu
- Department of Orthopedic Surgery, Medical 3D Printing Center, Orthopedic Institute, the First Affiliated Hospital, School of Biology and Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu 215000, China.
| | - Bin Li
- Department of Orthopedic Surgery, Medical 3D Printing Center, Orthopedic Institute, the First Affiliated Hospital, School of Biology and Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu 215000, China; Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu 215000, China; Department of Spinal Surgery, the Third Affiliated Hospital, Soochow University, Changzhou, Jiangsu 213003, China.
| | - Hongtao Zhang
- Department of Orthopedic Surgery, Medical 3D Printing Center, Orthopedic Institute, the First Affiliated Hospital, School of Biology and Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu 215000, China.
| |
Collapse
|
17
|
Kwok B, Chandrasekaran P, Wang C, He L, Mauck RL, Dyment NA, Koyama E, Han L. Rapid specialization and stiffening of the primitive matrix in developing articular cartilage and meniscus. Acta Biomater 2023; 168:235-251. [PMID: 37414114 PMCID: PMC10529006 DOI: 10.1016/j.actbio.2023.06.047] [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: 02/20/2023] [Revised: 06/02/2023] [Accepted: 06/28/2023] [Indexed: 07/08/2023]
Abstract
Understanding early patterning events in the extracellular matrix (ECM) formation can provide a blueprint for regenerative strategies to better recapitulate the function of native tissues. Currently, there is little knowledge on the initial, incipient ECM of articular cartilage and meniscus, two load-bearing counterparts of the knee joint. This study elucidated distinctive traits of their developing ECMs by studying the composition and biomechanics of these two tissues in mice from mid-gestation (embryonic day 15.5) to neo-natal (post-natal day 7) stages. We show that articular cartilage initiates with the formation of a pericellular matrix (PCM)-like primitive matrix, followed by the separation into distinct PCM and territorial/interterritorial (T/IT)-ECM domains, and then, further expansion of the T/IT-ECM through maturity. In this process, the primitive matrix undergoes a rapid, exponential stiffening, with a daily modulus increase rate of 35.7% [31.9 39.6]% (mean [95% CI]). Meanwhile, the matrix becomes more heterogeneous in the spatial distribution of properties, with concurrent exponential increases in the standard deviation of micromodulus and the slope correlating local micromodulus with the distance from cell surface. In comparison to articular cartilage, the primitive matrix of meniscus also exhibits exponential stiffening and an increase in heterogeneity, albeit with a much slower daily stiffening rate of 19.8% [14.9 24.9]% and a delayed separation of PCM and T/IT-ECM. These contrasts underscore distinct development paths of hyaline versus fibrocartilage. Collectively, these findings provide new insights into how knee joint tissues form to better guide cell- and biomaterial-based repair of articular cartilage, meniscus and potentially other load-bearing cartilaginous tissues. STATEMENT OF SIGNIFICANCE: Successful regeneration of articular cartilage and meniscus is challenged by incomplete knowledge of early events that drive the initial formation of the tissues' extracellular matrix in vivo. This study shows that articular cartilage initiates with a pericellular matrix (PCM)-like primitive matrix during embryonic development. This primitive matrix then separates into distinct PCM and territorial/interterritorial domains, undergoes an exponential daily stiffening of ≈36% and an increase in micromechanical heterogeneity. At this early stage, the meniscus primitive matrix shows differential molecular traits and exhibits a slower daily stiffening of ≈20%, underscoring distinct matrix development between these two tissues. Our findings thus establish a new blueprint to guide the design of regenerative strategies to recapitulate the key developmental steps in vivo.
Collapse
Affiliation(s)
- Bryan Kwok
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| | - Prashant Chandrasekaran
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| | - Chao Wang
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| | - Lan He
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| | - Robert L Mauck
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States; Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz Veterans Administration Medical Center, Philadelphia, PA 19104, United States
| | - Nathaniel A Dyment
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Eiki Koyama
- Translational Research Program in Pediatric Orthopaedics, Division of Orthopaedic Surgery, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, United States
| | - Lin Han
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States.
| |
Collapse
|
18
|
Gezer HH, Ostor A. What is new in pharmacological treatment for osteoarthritis? Best Pract Res Clin Rheumatol 2023; 37:101841. [PMID: 37302928 DOI: 10.1016/j.berh.2023.101841] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 05/10/2023] [Indexed: 06/13/2023]
Abstract
Osteoarthritis (OA) is a degenerative joint disease in which structural changes of hyaline articular cartilage, subchondral bone, ligaments, capsule, synovium, muscles, and periarticular changes are involved. The knee is the most commonly affected joint, followed by the hand, hip, spine, and feet. Different pathological mechanisms are at play in each of these various involvement sites. Although systemic inflammation is more prominent in hand OA, knee and hip OA have been associated with excessive joint load and injury. As OA has varied phenotypes and the primarily affected tissues differ, treatment options must be tailored accordingly. In recent years, ongoing efforts have been made to develop disease-modifying options that halt or slow disease progression. Many are still in clinical trials, and as insights into the pathogenesis of OA evolve, novel therapeutic strategies will be developed. In this chapter, we provide an overview of the novel and emerging strategies in the management of OA.
Collapse
Affiliation(s)
- Halise Hande Gezer
- Marmara University School of Medicine, PMR Department Rheumatology Division, Istanbul, Turkiye
| | - Andrew Ostor
- Cabrini Medical Centre, Monash University, Melbourne & ANU, Canberra, Australia.
| |
Collapse
|
19
|
Zhang H, Wu S, Chen W, Hu Y, Geng Z, Su J. Bone/cartilage targeted hydrogel: Strategies and applications. Bioact Mater 2023; 23:156-169. [PMID: 36406248 PMCID: PMC9661677 DOI: 10.1016/j.bioactmat.2022.10.028] [Citation(s) in RCA: 70] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 10/26/2022] [Accepted: 10/27/2022] [Indexed: 11/13/2022] Open
Abstract
The skeletal system is responsible for weight-bearing, organ protection, and movement. Bone diseases caused by trauma, infection, and aging can seriously affect a patient's quality of life. Bone targeted biomaterials are suitable for the treatment of bone diseases. Biomaterials with bone-targeted properties can improve drug utilization and reduce side effects. A large number of bone-targeted micro-nano materials have been developed. However, only a few studies addressed bone-targeted hydrogel. The large size of hydrogel makes it difficult to achieve systematic targeting. However, local targeted hydrogel still has significant prospects. Molecules in bone/cartilage extracellular matrix and bone cells provide binding sites for bone-targeted hydrogel. Drug delivery systems featuring microgels with targeting properties is a key construction strategy for bone-targeted hydrogel. Besides, injectable hydrogel drug depot carrying bone-targeted drugs is another strategy. In this review, we summarize the bone-targeted hydrogel through application environment, construction strategies and disease applications. We hope this article will provide a reference for the development of bone-targeted hydrogels. We also hope this article could increase awareness of bone-targeted materials. Introducing the microenvironment and target molecules in different parts of long bones. Summarizing the construction strategy of micro/nanoparticle hydrogel with bone targeting properties. Summarizing the construction strategy of hydrogel based depot carrying bone-targeted drugs. Reporting the application and effect of bone targeting hydrogel in common bone diseases.
Collapse
|
20
|
Patel J, Chen S, Katzmeyer T, Pei YA, Pei M. Sex-dependent variation in cartilage adaptation: from degeneration to regeneration. Biol Sex Differ 2023; 14:17. [PMID: 37024929 PMCID: PMC10077643 DOI: 10.1186/s13293-023-00500-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 03/20/2023] [Indexed: 04/08/2023] Open
Abstract
Despite acknowledgement in the scientific community of sex-based differences in cartilage biology, the implications for study design remain unclear, with many studies continuing to arbitrarily assign demographics. Clinically, it has been well-established that males and females differ in cartilage degeneration, and accumulating evidence points to the importance of sex differences in the field of cartilage repair. However, a comprehensive review of the mechanisms behind this trend and the influence of sex on cartilage regeneration has not yet been presented. This paper aims to summarize current findings regarding sex-dependent variation in knee anatomy, sex hormones' effect on cartilage, and cartilaginous degeneration and regeneration, with a focus on stem cell therapies. Findings suggest that the stem cells themselves, as well as their surrounding microenvironment, contribute to sex-based differences. Accordingly, this paper underscores the contribution of both stem cell donor and recipient sex to sex-related differences in treatment efficacy. Cartilage regeneration is a field that needs more research to optimize strategies for better clinical results; taking sex into account could be a big factor in developing more effective and personalized treatments. The compilation of this information emphasizes the importance of investing further research in sex differences in cartilage biology.
Collapse
Affiliation(s)
- Jhanvee Patel
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, 64 Medical Center Drive, PO Box 9196, Morgantown, WV, 26506-9196, USA
| | - Song Chen
- Department of Orthopaedics, The General Hospital of Western Theater Command, Chengdu, 610083, Sichuan, China
| | - Torey Katzmeyer
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, 64 Medical Center Drive, PO Box 9196, Morgantown, WV, 26506-9196, USA
| | - Yixuan Amy Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, 64 Medical Center Drive, PO Box 9196, Morgantown, WV, 26506-9196, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Ming Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, 64 Medical Center Drive, PO Box 9196, Morgantown, WV, 26506-9196, USA.
- WVU Cancer Institute, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, WV, 26506, USA.
| |
Collapse
|
21
|
Pattnaik A, Sanket AS, Pradhan S, Sahoo R, Das S, Pany S, Douglas TEL, Dandela R, Liu Q, Rajadas J, Pati S, De Smedt SC, Braeckmans K, Samal SK. Designing of gradient scaffolds and their applications in tissue regeneration. Biomaterials 2023; 296:122078. [PMID: 36921442 DOI: 10.1016/j.biomaterials.2023.122078] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 02/19/2023] [Accepted: 03/02/2023] [Indexed: 03/07/2023]
Abstract
Gradient scaffolds are isotropic/anisotropic three-dimensional structures with gradual transitions in geometry, density, porosity, stiffness, etc., that mimic the biological extracellular matrix. The gradient structures in biological tissues play a major role in various functional and metabolic activities in the body. The designing of gradients in the scaffold can overcome the current challenges in the clinic compared to conventional scaffolds by exhibiting excellent penetration capacity for nutrients & cells, increased cellular adhesion, cell viability & differentiation, improved mechanical stability, and biocompatibility. In this review, the recent advancements in designing gradient scaffolds with desired biomimetic properties, and their implication in tissue regeneration applications have been briefly explained. Furthermore, the gradients in native tissues such as bone, cartilage, neuron, cardiovascular, skin and their specific utility in tissue regeneration have been discussed in detail. The insights from such advances using gradient-based scaffolds can widen the horizon for using gradient biomaterials in tissue regeneration applications.
Collapse
Affiliation(s)
- Ananya Pattnaik
- Laboratory of Biomaterials and Regenerative Medicine for Advanced Therapies, ICMR-Regional Medical Research Centre, Bhubaneswar, 751023, Odisha, India
| | - A Swaroop Sanket
- Laboratory of Biomaterials and Regenerative Medicine for Advanced Therapies, ICMR-Regional Medical Research Centre, Bhubaneswar, 751023, Odisha, India
| | - Sanghamitra Pradhan
- Department of Chemistry, Institute of Technical Education and Research, Siksha 'O' Anusandhan University, Bhubaneswar, 751030, Odisha, India
| | - Rajashree Sahoo
- Laboratory of Biomaterials and Regenerative Medicine for Advanced Therapies, ICMR-Regional Medical Research Centre, Bhubaneswar, 751023, Odisha, India
| | - Sudiptee Das
- Laboratory of Biomaterials and Regenerative Medicine for Advanced Therapies, ICMR-Regional Medical Research Centre, Bhubaneswar, 751023, Odisha, India
| | - Swarnaprbha Pany
- Laboratory of Biomaterials and Regenerative Medicine for Advanced Therapies, ICMR-Regional Medical Research Centre, Bhubaneswar, 751023, Odisha, India
| | - Timothy E L Douglas
- Engineering Department, Lancaster University, Lancaster, United Kingdom; Materials Science Institute, Lancaster University, Lancaster, United Kingdom
| | - Rambabu Dandela
- Department of Industrial and Engineering Chemistry, Institute of Chemical Technology, Indian Oil Odisha Campus, Bhubaneswar, Odisha, India
| | - Qiang Liu
- Advanced Drug Delivery and Regenerative Biomaterials Laboratory, Cardiovascular Institute, Stanford University School of Medicine, Department of Medicine, Stanford University, California, 94304, USA
| | - Jaykumar Rajadas
- Advanced Drug Delivery and Regenerative Biomaterials Laboratory, Cardiovascular Institute, Stanford University School of Medicine, Department of Medicine, Stanford University, California, 94304, USA; Department of Bioengineering and Therapeutic Sciences, University of California San Francusco (UCSF) School of Parmacy, California, USA
| | - Sanghamitra Pati
- Laboratory of Biomaterials and Regenerative Medicine for Advanced Therapies, ICMR-Regional Medical Research Centre, Bhubaneswar, 751023, Odisha, India
| | - Stefaan C De Smedt
- Laboratory of General Biochemistry and Physical Pharmacy, University of Ghent, Ghent, 9000, Belgium.
| | - Kevin Braeckmans
- Laboratory of General Biochemistry and Physical Pharmacy, University of Ghent, Ghent, 9000, Belgium
| | - Sangram Keshari Samal
- Laboratory of Biomaterials and Regenerative Medicine for Advanced Therapies, ICMR-Regional Medical Research Centre, Bhubaneswar, 751023, Odisha, India.
| |
Collapse
|
22
|
Nischal N, Iyengar KP, Herlekar D, Botchu R. Imaging of Cartilage and Chondral Defects: An Overview. Life (Basel) 2023; 13:life13020363. [PMID: 36836719 PMCID: PMC9960762 DOI: 10.3390/life13020363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 01/15/2023] [Accepted: 01/20/2023] [Indexed: 02/03/2023] Open
Abstract
A healthy articular cartilage is paramount to joint function. Cartilage defects, whether acute or chronic, are a significant source of morbidity. This review summarizes various imaging modalities used for cartilage assessment. While radiographs are insensitive, they are still widely used to indirectly assess cartilage. Ultrasound has shown promise in the detection of cartilage defects, but its efficacy is limited in many joints due to inadequate visualization. CT arthrography has the potential to assess internal derangements of joints along with cartilage, especially in patients with contraindications to MRI. MRI remains the favored imaging modality to assess cartilage. The conventional imaging techniques are able to assess cartilage abnormalities when cartilage is already damaged. The newer imaging techniques are thus targeted at detecting biochemical and structural changes in cartilage before an actual visible irreversible loss. These include, but are not limited to, T2 and T2* mapping, dGEMRI, T1ρ imaging, gagCEST imaging, sodium MRI and integrated PET with MRI. A brief discussion of the advances in the surgical management of cartilage defects and post-operative imaging assessment is also included.
Collapse
Affiliation(s)
- Neha Nischal
- Department of Musculoskeletal Radiology, Royal Orthopedic Hospital, Birmingham B31 2AP, UK
- Department of Radiology, Holy Family Hospital, New Delhi 110025, India
| | | | - Deepak Herlekar
- Department of Orthopaedics, University Hospitals of Morecambe Bay NHS Foundation Trust, Kendal LA9 7RG, UK
| | - Rajesh Botchu
- Department of Musculoskeletal Radiology, Royal Orthopedic Hospital, Birmingham B31 2AP, UK
- Correspondence:
| |
Collapse
|
23
|
Sorvina A, Antoniou M, Esmaeili Z, Kochetkova M. Unusual Suspects: Bone and Cartilage ECM Proteins as Carcinoma Facilitators. Cancers (Basel) 2023; 15:cancers15030791. [PMID: 36765749 PMCID: PMC9913341 DOI: 10.3390/cancers15030791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 01/25/2023] [Accepted: 01/25/2023] [Indexed: 01/31/2023] Open
Abstract
The extracellular matrix (ECM) is the complex three-dimensional network of fibrous proteins and proteoglycans that constitutes an essential part of every tissue to provide support for normal tissue homeostasis. Tissue specificity of the ECM in its topology and structure supports unique biochemical and mechanical properties of each organ. Cancers, like normal tissues, require the ECM to maintain multiple processes governing tumor development, progression and spread. A large body of experimental and clinical evidence has now accumulated to demonstrate essential roles of numerous ECM components in all cancer types. Latest findings also suggest that multiple tumor types express, and use to their advantage, atypical ECM components that are not found in the cancer tissue of origin. However, the understanding of cancer-specific expression patterns of these ECM proteins and their exact roles in selected tumor types is still sketchy. In this review, we summarize the latest data on the aberrant expression of bone and cartilage ECM proteins in epithelial cancers and their specific functions in the pathogenesis of carcinomas and discuss future directions in exploring the utility of this selective group of ECM components as future drug targets.
Collapse
|
24
|
HS, an Ancient Molecular Recognition and Information Storage Glycosaminoglycan, Equips HS-Proteoglycans with Diverse Matrix and Cell-Interactive Properties Operative in Tissue Development and Tissue Function in Health and Disease. Int J Mol Sci 2023; 24:ijms24021148. [PMID: 36674659 PMCID: PMC9867265 DOI: 10.3390/ijms24021148] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/23/2022] [Accepted: 12/27/2022] [Indexed: 01/11/2023] Open
Abstract
Heparan sulfate is a ubiquitous, variably sulfated interactive glycosaminoglycan that consists of repeating disaccharides of glucuronic acid and glucosamine that are subject to a number of modifications (acetylation, de-acetylation, epimerization, sulfation). Variable heparan sulfate chain lengths and sequences within the heparan sulfate chains provide structural diversity generating interactive oligosaccharide binding motifs with a diverse range of extracellular ligands and cellular receptors providing instructional cues over cellular behaviour and tissue homeostasis through the regulation of essential physiological processes in development, health, and disease. heparan sulfate and heparan sulfate-PGs are integral components of the specialized glycocalyx surrounding cells. Heparan sulfate is the most heterogeneous glycosaminoglycan, in terms of its sequence and biosynthetic modifications making it a difficult molecule to fully characterize, multiple ligands also make an elucidation of heparan sulfate functional properties complicated. Spatio-temporal presentation of heparan sulfate sulfate groups is an important functional determinant in tissue development and in cellular control of wound healing and extracellular remodelling in pathological tissues. The regulatory properties of heparan sulfate are mediated via interactions with chemokines, chemokine receptors, growth factors and morphogens in cell proliferation, differentiation, development, tissue remodelling, wound healing, immune regulation, inflammation, and tumour development. A greater understanding of these HS interactive processes will improve therapeutic procedures and prognoses. Advances in glycosaminoglycan synthesis and sequencing, computational analytical carbohydrate algorithms and advanced software for the evaluation of molecular docking of heparan sulfate with its molecular partners are now available. These advanced analytic techniques and artificial intelligence offer predictive capability in the elucidation of heparan sulfate conformational effects on heparan sulfate-ligand interactions significantly aiding heparan sulfate therapeutics development.
Collapse
|
25
|
Ahmadian E, Eftekhari A, Janas D, Vahedi P. Nanofiber scaffolds based on extracellular matrix for articular cartilage engineering: A perspective. Nanotheranostics 2023; 7:61-69. [PMID: 36593799 PMCID: PMC9760364 DOI: 10.7150/ntno.78611] [Citation(s) in RCA: 65] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 10/27/2022] [Indexed: 11/24/2022] Open
Abstract
Articular cartilage has a low self-repair capacity due to the lack of vessels and nerves. In recent times, nanofiber scaffolds have been widely used for this purpose. The optimum nanofiber scaffold should stimulate new tissue's growth and mimic the articular cartilage nature. Furthermore, the characteristics of the scaffold should match those of the cellular matrix components of the native tissue to best merge with the target tissue. Therefore, selective modification of prefabricated scaffolds based on the structure of the repaired tissues is commonly conducted to promote restoring the tissue. A thorough analysis is required to find out the architectural features of scaffolds that are essential to make the treatment successful. The current review aims to target this challenge. The article highlights different optimization approaches of nanofibrous scaffolds for improved cartilage tissue engineering. In this context, the influence of the architecture of nanoscaffolds on performance is discussed in detail. Finally, based on the gathered information, a future outlook is provided to catalyze development in this promising field.
Collapse
Affiliation(s)
- Elham Ahmadian
- Kidney Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Aziz Eftekhari
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran,Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran,✉ Corresponding authors: Aziz Eftekhari (), Dawid Janas (), Parviz Vahedi ()
| | - Dawid Janas
- Department of Organic Chemistry, Bioorganic Chemistry and Biotechnology, Silesian University of Technology, B. Krzywoustego 4, 44-100, Gliwice, Poland,✉ Corresponding authors: Aziz Eftekhari (), Dawid Janas (), Parviz Vahedi ()
| | - Parviz Vahedi
- Department of Anatomical Sciences, Maragheh University of Medical Sciences, Maragheh 78151-55158, Iran,✉ Corresponding authors: Aziz Eftekhari (), Dawid Janas (), Parviz Vahedi ()
| |
Collapse
|
26
|
DIA mass spectrometry characterizes urinary proteomics in neonatal and adult donkeys. Sci Rep 2022; 12:22590. [PMID: 36585464 PMCID: PMC9803668 DOI: 10.1038/s41598-022-27245-0] [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: 01/27/2022] [Accepted: 12/28/2022] [Indexed: 12/31/2022] Open
Abstract
Health monitoring is critical for newborn animals due to their vulnerability to diseases. Urine can be not only a useful and non-invasive tool (free-catch samples) to reflect the physiological status of animals but also to help monitor the progression of diseases. Proteomics involves the study of the whole complement of proteins and peptides, including structure, quantities, functions, variations and interactions. In this study, urinary proteomics of neonatal donkeys were characterized and compared to the profiles of adult donkeys to provide a reference database for healthy neonatal donkeys. The urine samples were collected from male neonatal donkeys on their sixth to tenth days of life (group N) and male adult donkeys aging 4-6 years old (group A). Library-free data-independent acquisition (direct DIA) mass spectrometry-based proteomics were applied to analyze the urinary protein profiles. Total 2179 urinary proteins were identified, and 411 proteins were differentially expressed (P < 0.05) between the two groups. 104 proteins were exclusively expressed in group N including alpha fetoprotein (AFP), peptidase-mitochondrial processing data unit (PMPCB), and upper zone of growth plate and cartilage matrix associated (UCMA), which might be used to monitor the health status of neonatal donkeys. In functional analysis, some differentially expressed proteins were identified related to immune system pathways, which might provide more insight in the immature immunity of neonatal donkeys. To the best of our knowledge, this is the first time to report donkey urinary proteome and our results might provide reference for urinary biomarker discovery used to monitor and evaluate health status of neonatal donkeys.
Collapse
|
27
|
Zhang CH, Gao Y, Hung HH, Zhuo Z, Grodzinsky AJ, Lassar AB. Creb5 coordinates synovial joint formation with the genesis of articular cartilage. Nat Commun 2022; 13:7295. [PMID: 36435829 PMCID: PMC9701237 DOI: 10.1038/s41467-022-35010-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 11/15/2022] [Indexed: 11/28/2022] Open
Abstract
While prior work has established that articular cartilage arises from Prg4-expressing perichondrial cells, it is not clear how this process is specifically restricted to the perichondrium of synovial joints. We document that the transcription factor Creb5 is necessary to initiate the expression of signaling molecules that both direct the formation of synovial joints and guide perichondrial tissue to form articular cartilage instead of bone. Creb5 promotes the generation of articular chondrocytes from perichondrial precursors in part by inducing expression of signaling molecules that block a Wnt5a autoregulatory loop in the perichondrium. Postnatal deletion of Creb5 in the articular cartilage leads to loss of both flat superficial zone articular chondrocytes coupled with a loss of both Prg4 and Wif1 expression in the articular cartilage; and a non-cell autonomous up-regulation of Ctgf. Our findings indicate that Creb5 promotes joint formation and the subsequent development of articular chondrocytes by driving the expression of signaling molecules that both specify the joint interzone and simultaneously inhibit a Wnt5a positive-feedback loop in the perichondrium.
Collapse
Affiliation(s)
- Cheng-Hai Zhang
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute at Harvard Medical School, 240 Longwood Ave., Boston, MA, 02115, USA.
| | - Yao Gao
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute at Harvard Medical School, 240 Longwood Ave., Boston, MA, 02115, USA
| | - Han-Hwa Hung
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Zhu Zhuo
- Bioinformatics Core, Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
| | - Alan J Grodzinsky
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Andrew B Lassar
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute at Harvard Medical School, 240 Longwood Ave., Boston, MA, 02115, USA.
| |
Collapse
|
28
|
Liu H, Craig SEL, Molchanov V, Floramo JS, Zhao Y, Yang T. SUMOylation in Skeletal Development, Homeostasis, and Disease. Cells 2022; 11:cells11172710. [PMID: 36078118 PMCID: PMC9454984 DOI: 10.3390/cells11172710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 08/19/2022] [Accepted: 08/27/2022] [Indexed: 11/18/2022] Open
Abstract
The modification of proteins by small ubiquitin-related modifier (SUMO) molecules, SUMOylation, is a key post-translational modification involved in a variety of biological processes, such as chromosome organization, DNA replication and repair, transcription, nuclear transport, and cell signaling transduction. In recent years, emerging evidence has shown that SUMOylation regulates the development and homeostasis of the skeletal system, with its dysregulation causing skeletal diseases, suggesting that SUMOylation pathways may serve as a promising therapeutic target. In this review, we summarize the current understanding of the molecular mechanisms by which SUMOylation pathways regulate skeletal cells in physiological and disease contexts.
Collapse
Affiliation(s)
| | | | | | | | | | - Tao Yang
- Laboratory of Skeletal Biology, Department of Cell Biology, Van Andel Institute, 333 Bostwick Ave NE, Grand Rapids, MI 49503, USA
- Correspondence: ; Tel.: +1-616-234-5820
| |
Collapse
|
29
|
Double – Network Hydrogel Based on Exopolysaccharides as a Biomimetic Extracellular Matrix to Augment Articular Cartilage Regeneration. Acta Biomater 2022; 152:124-143. [DOI: 10.1016/j.actbio.2022.08.062] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 08/25/2022] [Accepted: 08/25/2022] [Indexed: 11/01/2022]
|
30
|
Li K, Ji X, Seeley R, Lee WC, Shi Y, Song F, Liao X, Song C, Huang X, Rux D, Cao J, Luo X, Anderson SM, Huang W, Long F. Impaired glucose metabolism underlies articular cartilage degeneration in osteoarthritis. FASEB J 2022; 36:e22377. [PMID: 35608871 DOI: 10.1096/fj.202200485r] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Revised: 05/05/2022] [Accepted: 05/13/2022] [Indexed: 01/09/2023]
Abstract
Osteoarthritis (OA) is the leading joint disease characterized by cartilage destruction and loss of mobility. Accumulating evidence indicates that the incidence and severity of OA increases with diabetes, implicating systemic glucose metabolism in joint health. However, a definitive link between cellular metabolism in articular cartilage and OA pathogenesis is not yet established. Here, we report that in mice surgically induced to develop knee OA through destabilization of medial meniscus (DMM), expression of the main glucose transporter Glut1 is notably reduced in joint cartilage. Inducible deletion of Glut1 specifically in the Prg4-expressing articular cartilage accelerates cartilage loss in DMM-induced OA. Conversely, forced expression of Glut1 protects against cartilage destruction following DMM. Moreover, in mice with type I diabetes, both Glut1 expression and the rate of glycolysis are diminished in the articular cartilage, and the diabetic mice exhibit more severe cartilage destruction than their nondiabetic counterparts following DMM. The results provide proof of concept that boosting glucose metabolism in articular chondrocytes may ameliorate cartilage degeneration in OA.
Collapse
Affiliation(s)
- Ke Li
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Translational Research Program of Pediatric Orthopedics, Department of Surgery, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Xing Ji
- Translational Research Program of Pediatric Orthopedics, Department of Surgery, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Rebecca Seeley
- Translational Research Program of Pediatric Orthopedics, Department of Surgery, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Wen-Chih Lee
- Translational Research Program of Pediatric Orthopedics, Department of Surgery, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Yu Shi
- Translational Research Program of Pediatric Orthopedics, Department of Surgery, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Fangfang Song
- Translational Research Program of Pediatric Orthopedics, Department of Surgery, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Xueyang Liao
- Translational Research Program of Pediatric Orthopedics, Department of Surgery, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Chao Song
- Translational Research Program of Pediatric Orthopedics, Department of Surgery, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Xiaobin Huang
- Translational Research Program of Pediatric Orthopedics, Department of Surgery, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Danielle Rux
- Translational Research Program of Pediatric Orthopedics, Department of Surgery, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Ju Cao
- Department of Laboratory Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaoji Luo
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Steven M Anderson
- Department of Pathology, University of Colorado School of Medicine, Denver, Colorado, USA
| | - Wei Huang
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Fanxin Long
- Translational Research Program of Pediatric Orthopedics, Department of Surgery, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| |
Collapse
|
31
|
Shah FH, Kim SJ. Therapeutic role of medicinal plant extracts and bioactive compounds in osteoarthritis. ADVANCES IN TRADITIONAL MEDICINE 2022. [DOI: 10.1007/s13596-022-00635-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
32
|
Castro NJ, Babakhanova G, Hu J, Athanasiou K. Nondestructive testing of native and tissue-engineered medical products: adding numbers to pictures. Trends Biotechnol 2022; 40:194-209. [PMID: 34315621 PMCID: PMC8772387 DOI: 10.1016/j.tibtech.2021.06.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 06/25/2021] [Accepted: 06/28/2021] [Indexed: 02/03/2023]
Abstract
Traditional destructive tests are used for quality assurance and control within manufacturing workflows. Their applicability to biomanufacturing is limited due to inherent constraints of the biomanufacturing process. To address this, photo- and acoustic-based nondestructive testing has risen in prominence to interrogate not only structure and function, but also to integrate quantitative measurements of biochemical composition to cross-correlate structural, compositional, and functional variances. We survey relevant literature related to single-mode and multimodal nondestructive testing of soft tissues, which adds numbers (quantitative measurements) to pictures (qualitative data). Native and tissue-engineered articular cartilage is highlighted because active biomanufacturing processes are being developed. Included are recent efforts and prominent trends focused on technologies for clinical and in-process biomanufacturing applications.
Collapse
Affiliation(s)
- Nathan J. Castro
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA 92617, USA
| | - Greta Babakhanova
- Biosystems and Biomaterials Division, National Institute of Standards and Technology, Gaithersburg, MD, 20899, USA
| | - Jerry Hu
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA 92617, USA
| | - K.A. Athanasiou
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA 92617, USA,Correspondence:
| |
Collapse
|
33
|
Ge D, O'Brien MJ, Savoie FH, Gimble JM, Wu X, Gilbert MH, Clark-Patterson GL, Schuster JD, Miller KS, Wang A, Myers L, You Z. Human adipose-derived stromal/stem cells expressing doublecortin improve cartilage repair in rabbits and monkeys. NPJ Regen Med 2021; 6:82. [PMID: 34848747 PMCID: PMC8633050 DOI: 10.1038/s41536-021-00192-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 10/29/2021] [Indexed: 11/25/2022] Open
Abstract
Localized cartilage lesions in early osteoarthritis and acute joint injuries are usually treated surgically to restore function and relieve pain. However, a persistent clinical challenge remains in how to repair the cartilage lesions. We expressed doublecortin (DCX) in human adipose-derived stromal/stem cells (hASCs) and engineered hASCs into cartilage tissues using an in vitro 96-well pellet culture system. The cartilage tissue constructs with and without DCX expression were implanted in the knee cartilage defects of rabbits (n = 42) and monkeys (n = 12). Cohorts of animals were euthanized at 6, 12, and 24 months after surgery to evaluate the cartilage repair outcomes. We found that DCX expression in hASCs increased expression of growth differentiation factor 5 (GDF5) and matrilin 2 in the engineered cartilage tissues. The cartilage tissues with DCX expression significantly enhanced cartilage repair as assessed macroscopically and histologically at 6, 12, and 24 months after implantation in the rabbits and 24 months after implantation in the monkeys, compared to the cartilage tissues without DCX expression. These findings suggest that hASCs expressing DCX may be engineered into cartilage tissues that can be used to treat localized cartilage lesions.
Collapse
Affiliation(s)
- Dongxia Ge
- Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, LA, USA
- Department of Orthopaedic Surgery, Tulane University School of Medicine, New Orleans, LA, USA
| | - Michael J O'Brien
- Department of Orthopaedic Surgery, Tulane University School of Medicine, New Orleans, LA, USA
| | - Felix H Savoie
- Department of Orthopaedic Surgery, Tulane University School of Medicine, New Orleans, LA, USA
| | - Jeffrey M Gimble
- Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, LA, USA
- LaCell LLC and Obatala Sciences Inc., New Orleans, LA, USA
- Tulane Center for Stem Cell Research and Regenerative Medicine, Tulane University School of Medicine, New Orleans, LA, USA
- John W. Deming Department of Medicine, Tulane University School of Medicine, New Orleans, LA, USA
- Department of Surgery, Tulane University School of Medicine, New Orleans, LA, USA
| | - Xiying Wu
- LaCell LLC and Obatala Sciences Inc., New Orleans, LA, USA
| | - Margaret H Gilbert
- Tulane National Primate Research Center, Tulane University, New Orleans, LA, USA
| | | | - Jason D Schuster
- Department of Biomedical Engineering, Tulane University, New Orleans, LA, USA
| | - Kristin S Miller
- Department of Biomedical Engineering, Tulane University, New Orleans, LA, USA
| | - Alun Wang
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Leann Myers
- Department of Biostatistics and Data Science, Tulane University School of Public Health and Tropic Medicine, New Orleans, LA, USA
| | - Zongbing You
- Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, LA, USA.
- Department of Orthopaedic Surgery, Tulane University School of Medicine, New Orleans, LA, USA.
- Tulane Center for Stem Cell Research and Regenerative Medicine, Tulane University School of Medicine, New Orleans, LA, USA.
- Tulane Cancer Center and Louisiana Cancer Research Consortium, Tulane University School of Medicine, New Orleans, LA, USA.
- Tulane Center for Aging, Tulane University School of Medicine, New Orleans, LA, USA.
- Southeast Louisiana Veterans Health Care System, New Orleans, LA, USA.
| |
Collapse
|
34
|
Doyle SE, Snow F, Duchi S, O’Connell CD, Onofrillo C, Di Bella C, Pirogova E. 3D Printed Multiphasic Scaffolds for Osteochondral Repair: Challenges and Opportunities. Int J Mol Sci 2021; 22:12420. [PMID: 34830302 PMCID: PMC8622524 DOI: 10.3390/ijms222212420] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 11/11/2021] [Accepted: 11/12/2021] [Indexed: 12/19/2022] Open
Abstract
Osteochondral (OC) defects are debilitating joint injuries characterized by the loss of full thickness articular cartilage along with the underlying calcified cartilage through to the subchondral bone. While current surgical treatments can provide some relief from pain, none can fully repair all the components of the OC unit and restore its native function. Engineering OC tissue is challenging due to the presence of the three distinct tissue regions. Recent advances in additive manufacturing provide unprecedented control over the internal microstructure of bioscaffolds, the patterning of growth factors and the encapsulation of potentially regenerative cells. These developments are ushering in a new paradigm of 'multiphasic' scaffold designs in which the optimal micro-environment for each tissue region is individually crafted. Although the adoption of these techniques provides new opportunities in OC research, it also introduces challenges, such as creating tissue interfaces, integrating multiple fabrication techniques and co-culturing different cells within the same construct. This review captures the considerations and capabilities in developing 3D printed OC scaffolds, including materials, fabrication techniques, mechanical function, biological components and design.
Collapse
Affiliation(s)
- Stephanie E. Doyle
- Electrical and Biomedical Engineering, School of Engineering, RMIT University, Melbourne, VIC 3000, Australia; (F.S.)
- ACMD, St Vincent’s Hospital Melbourne, Fitzroy, VIC 3065, Australia; (S.D.); (C.O.); (C.D.B.)
| | - Finn Snow
- Electrical and Biomedical Engineering, School of Engineering, RMIT University, Melbourne, VIC 3000, Australia; (F.S.)
| | - Serena Duchi
- ACMD, St Vincent’s Hospital Melbourne, Fitzroy, VIC 3065, Australia; (S.D.); (C.O.); (C.D.B.)
- Department of Surgery, The University of Melbourne, St Vincent’s Hospital Melbourne, Fitzroy, VIC 3065, Australia
- ARC Centre of Excellence for Electromaterials Science, Intelligent Polymer Research Institute, University of Wollongong, Wollongong, NSW 2522, Australia
| | - Cathal D. O’Connell
- Electrical and Biomedical Engineering, School of Engineering, RMIT University, Melbourne, VIC 3000, Australia; (F.S.)
- ACMD, St Vincent’s Hospital Melbourne, Fitzroy, VIC 3065, Australia; (S.D.); (C.O.); (C.D.B.)
| | - Carmine Onofrillo
- ACMD, St Vincent’s Hospital Melbourne, Fitzroy, VIC 3065, Australia; (S.D.); (C.O.); (C.D.B.)
- Department of Surgery, The University of Melbourne, St Vincent’s Hospital Melbourne, Fitzroy, VIC 3065, Australia
- ARC Centre of Excellence for Electromaterials Science, Intelligent Polymer Research Institute, University of Wollongong, Wollongong, NSW 2522, Australia
| | - Claudia Di Bella
- ACMD, St Vincent’s Hospital Melbourne, Fitzroy, VIC 3065, Australia; (S.D.); (C.O.); (C.D.B.)
- Department of Surgery, The University of Melbourne, St Vincent’s Hospital Melbourne, Fitzroy, VIC 3065, Australia
- Department of Orthopaedics, St Vincent’s Hospital Melbourne, Fitzroy, VIC 3065, Australia
| | - Elena Pirogova
- Electrical and Biomedical Engineering, School of Engineering, RMIT University, Melbourne, VIC 3000, Australia; (F.S.)
| |
Collapse
|
35
|
Li XJ, Zhu F, Li B, Zhang D, Liang CW. Recombinant human regenerating gene 4 attenuates the severity of osteoarthritis by promoting the proliferation of articular chondrocyte in an animal model. Curr Mol Pharmacol 2021; 15:693-699. [PMID: 34488597 DOI: 10.2174/1874467214666210901163144] [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: 03/05/2021] [Revised: 06/24/2021] [Accepted: 07/09/2021] [Indexed: 11/22/2022]
Abstract
INTRODUCTION Osteoarthritis (OA) is a dominant cause of morbidity and disability. As a chronic disease, its etiological risk factors and most therapies at present, are empirical and symptomatic. Regenerating gene 4 (Reg4) is involved in cell growth, survival, regeneration, adhesion, and resistance to apoptosis, which are partially thought to be the pathogenic mechanisms of OA. However, the proper role of Reg4 in OA is still unknown. METHODS In this study, a consecutive administration of rhReg4 was applied to normal Sprague-Dawley rats or rats after OA induction. Histological changes and chondrocyte proliferation in the articular cartilage were measured. RESULTS We found that RhReg4 promotes chondrocyte proliferation in normal rats, and RhReg4 attenuated the severity of OA in rats by promoting chondrocytes' proliferation in OA rats. CONCLUSION In conclusion, recombinant human regenerating gene 4 (rhReg4) attenuates the severity of osteoarthritis in OA animal models and may be used as a new method for the treatment of osteoarthritis.
Collapse
Affiliation(s)
- Xue-Jia Li
- Department of Orthopaedics, Yueyang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai. China
| | - Fei Zhu
- Department of Orthopaedics, Guanghua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai. China
| | - Bo Li
- Department of Orthopaedics, Yueyang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai. China
| | - Dong Zhang
- Department of Orthopaedics, Yueyang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai. China
| | - Cheng-Wei Liang
- Department of Orthopaedics, Huadong Hospital Affiliated to Fudan University, Shanghai. China
| |
Collapse
|
36
|
Joint Degeneration in a Mouse Model of Pseudoachondroplasia: ER Stress, Inflammation, and Block of Autophagy. Int J Mol Sci 2021; 22:ijms22179239. [PMID: 34502142 PMCID: PMC8431545 DOI: 10.3390/ijms22179239] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/05/2021] [Accepted: 08/18/2021] [Indexed: 01/05/2023] Open
Abstract
Pseudoachondroplasia (PSACH), a short limb skeletal dysplasia associated with premature joint degeneration, is caused by misfolding mutations in cartilage oligomeric matrix protein (COMP). Here, we define mutant-COMP-induced stress mechanisms that occur in articular chondrocytes of MT-COMP mice, a murine model of PSACH. The accumulation of mutant-COMP in the ER occurred early in MT-COMP articular chondrocytes and stimulated inflammation (TNFα) at 4 weeks, and articular chondrocyte death increased at 8 weeks while ER stress through CHOP was elevated by 12 weeks. Importantly, blockage of autophagy (pS6), the major mechanism that clears the ER, sustained cellular stress in MT-COMP articular chondrocytes. Degeneration of MT-COMP articular cartilage was similar to that observed in PSACH and was associated with increased MMPs, a family of degradative enzymes. Moreover, chronic cellular stresses stimulated senescence. Senescence-associated secretory phenotype (SASP) may play a role in generating and propagating a pro-degradative environment in the MT-COMP murine joint. The loss of CHOP or resveratrol treatment from birth preserved joint health in MT-COMP mice. Taken together, these results indicate that ER stress/CHOP signaling and autophagy blockage are central to mutant-COMP joint degeneration, and MT-COMP mice joint health can be preserved by decreasing articular chondrocyte stress. Future joint sparing therapeutics for PSACH may include resveratrol.
Collapse
|
37
|
Deletion of Glut1 in early postnatal cartilage reprograms chondrocytes toward enhanced glutamine oxidation. Bone Res 2021; 9:38. [PMID: 34426569 PMCID: PMC8382841 DOI: 10.1038/s41413-021-00153-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 02/04/2021] [Accepted: 02/28/2021] [Indexed: 12/18/2022] Open
Abstract
Glucose metabolism is fundamental for the functions of all tissues, including cartilage. Despite the emerging evidence related to glucose metabolism in the regulation of prenatal cartilage development, little is known about the role of glucose metabolism and its biochemical basis in postnatal cartilage growth and homeostasis. We show here that genetic deletion of the glucose transporter Glut1 in postnatal cartilage impairs cell proliferation and matrix production in growth plate (GPs) but paradoxically increases cartilage remnants in the metaphysis, resulting in shortening of long bones. On the other hand, articular cartilage (AC) with Glut1 deficiency presents diminished cellularity and loss of proteoglycans, which ultimately progress to cartilage fibrosis. Moreover, predisposition to Glut1 deficiency severely exacerbates injury-induced osteoarthritis. Regardless of the disparities in glucose metabolism between GP and AC chondrocytes under normal conditions, both types of chondrocytes demonstrate metabolic plasticity to enhance glutamine utilization and oxidation in the absence of glucose availability. However, uncontrolled glutamine flux causes collagen overmodification, thus affecting extracellular matrix remodeling in both cartilage compartments. These results uncover the pivotal and distinct roles of Glut1-mediated glucose metabolism in two of the postnatal cartilage compartments and link some cartilage abnormalities to altered glucose/glutamine metabolism.
Collapse
|
38
|
Zhuang C, Wang Z, Chen W, Tian B, Li J, Lin H. Osteoporosis and Endplate Damage Correlation Using a Combined Approach of Hounsfield Unit Values and Total Endplate Scores: A Retrospective Cross-Sectional Study. Clin Interv Aging 2021; 16:1275-1283. [PMID: 34262267 PMCID: PMC8275111 DOI: 10.2147/cia.s315213] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Accepted: 06/20/2021] [Indexed: 12/13/2022] Open
Abstract
Purpose Osteoporosis and endplate damage, two primary orthopedic disorders that have adverse effects on the quality of life of older adults, may have some previously unknown relationship. The purpose of this study was to determine the potential association between osteoporosis and endplate damage with two specific imaging scoring systems and analyze the underlying mechanisms. Patients and Methods A cross-sectional study including 156 patients with degenerative disc disease (DDD) who visited our department in 2018 was performed. Data including age, sex, body mass index, Hounsfield unit (HU) values utilizing computed tomography (CT), and total endplate scores (TEPSs) using magnetic resonance imaging (MRI) of all patients were retrospectively collected and analyzed. The average HU value and TEPS of L1–L4 were used to represent the degrees of bone mineral density (BMD) and endplate damage, respectively. Patients with an HU value < 110 were defined as having osteoporosis and placed in the low-BMD group; otherwise, they were placed in the normal-BMD group. Multivariate logistic regression models were used to determine the independent factors of endplate damage. Results The TEPSs in the low-BMD group were significantly higher (6.4 ± 1.6 vs 5.0 ± 0.9, p < 0.001) overall and in every segment of L1–L4 (p < 0.01). A significant negative correlation was found between TEPS and HU values (p < 0.001). The HU value (odds ratio [OR] 0.221; 95% confidence interval [CI], 0.148–0.295, p < 0.001), age (OR 0.047; 95% CI, 0.029–0.224, p < 0.001), and BMD (OR 3.796; 95% CI, 2.11–7.382, p < 0.05) were independent factors influencing endplate damage. Conclusion A significantly positive correlation was observed between osteoporosis and endplate damage, indicating the requirement for a more comprehensive therapeutic regimen for treating patients with DDD complicated with osteoporosis.
Collapse
Affiliation(s)
- Chenyang Zhuang
- Department of Orthopedics, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Zixiang Wang
- Department of Orthopedics, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Weisin Chen
- Department of Orthopedics, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Bo Tian
- Department of Orthopedics, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Juan Li
- Department of Orthopedics, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China.,Department of Orthopedics, Shanghai Geriatrics Center, Fudan University, Shanghai, People's Republic of China
| | - Hong Lin
- Department of Orthopedics, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China.,Department of Orthopedics, Shanghai Geriatrics Center, Fudan University, Shanghai, People's Republic of China
| |
Collapse
|
39
|
Bhatti FUR, Karydis A, Lee BS, Deguchi T, Kim DG, Cho H. Understanding Early-Stage Posttraumatic Osteoarthritis for Future Prospects of Diagnosis: from Knee to Temporomandibular Joint. Curr Osteoporos Rep 2021; 19:166-174. [PMID: 33523424 DOI: 10.1007/s11914-021-00661-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/19/2021] [Indexed: 12/23/2022]
Abstract
PURPOSE OF REVIEW Many mechanical load-bearing joints of the body are prone to posttraumatic osteoarthritis (PTOA), including the knee joint and temporomandibular joint (TMJ). Early detection of PTOA can be beneficial in prevention or alleviating further progression of the disease. RECENT FINDINGS Various mouse models, similar to those used in development of novel diagnosis strategies for early stages of OA, have been proposed to study early PTOA. While many studies have focused on OA and PTOA in the knee joint, early diagnostic methods for OA and PTOA of the TMJ are still not well established. Previously, we showed that fluorescent near-infrared imaging can diagnose inflammation and cartilage damage in mouse models of knee PTOA. Here we propose that the same approach can be used for early diagnosis of TMJ-PTOA. In this review, we present a brief overview of PTOA, application of relevant mouse models, current imaging methods available to examine TMJ-PTOA, and the prospects of near-infrared optical imaging to diagnose early-stage TMJ-OA.
Collapse
Affiliation(s)
- Fazal-Ur-Rehman Bhatti
- Department of Orthopaedic Surgery and Biomedical Engineering, University of Tennessee Health Science Center, Research 151, VAMC, 1030 Jefferson Ave, Memphis, TN, 38104 , USA
| | - Anastasios Karydis
- Department of Orthopaedic Surgery and Biomedical Engineering, University of Tennessee Health Science Center, Research 151, VAMC, 1030 Jefferson Ave, Memphis, TN, 38104 , USA
| | - Beth S Lee
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University , Graves Hall, 333 West 10th Avenue, Columbus, OH, 43210, USA
| | - Toru Deguchi
- Division of Orthodontics, College of Dentistry, The Ohio State University, 4088 Postle Hall, 305 W. 12th Ave., Columbus, OH, 43210, USA
| | - Do-Gyoon Kim
- Division of Orthodontics, College of Dentistry, The Ohio State University, 4088 Postle Hall, 305 W. 12th Ave., Columbus, OH, 43210, USA.
| | - Hongsik Cho
- Department of Orthopaedic Surgery and Biomedical Engineering, University of Tennessee Health Science Center, Research 151, VAMC, 1030 Jefferson Ave, Memphis, TN, 38104 , USA.
- Campbell Clinic, Memphis, TN, USA.
- Veterans Affairs Medical Center, Memphis, TN, USA.
| |
Collapse
|
40
|
Sun K, Guo J, Yao X, Guo Z, Guo F. Growth differentiation factor 5 in cartilage and osteoarthritis: A possible therapeutic candidate. Cell Prolif 2021; 54:e12998. [PMID: 33522652 PMCID: PMC7941218 DOI: 10.1111/cpr.12998] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 01/01/2021] [Accepted: 01/05/2021] [Indexed: 12/11/2022] Open
Abstract
Growth differentiation factor 5 (GDF-5) is essential for cartilage development and homeostasis. The expression and function of GDF-5 are highly associated with the pathogenesis of osteoarthritis (OA). OA, characterized by progressive degeneration of joint, particularly in cartilage, causes severe social burden. However, there is no effective approach to reverse the progression of this disease. Over the past decades, extensive studies have demonstrated the protective effects of GDF-5 against cartilage degeneration and defects. Here, we summarize the current literature describing the role of GDF-5 in development of cartilage and joints, and the association between the GDF-5 gene polymorphisms and OA susceptibility. We also shed light on the protective effects of GDF-5 against OA in terms of direct GDF-5 supplementation and modulation of the GDF-5-related signalling. Finally, we discuss the current limitations in the application of GDF-5 for the clinical treatment of OA. This review provides a comprehensive insight into the role of GDF-5 in cartilage and emphasizes GDF-5 as a potential therapeutic candidate in OA.
Collapse
Affiliation(s)
- Kai Sun
- Department of OrthopedicsTongji Medical CollegeTongji HospitalHuazhong University of Science and TechnologyWuhanChina
| | - Jiachao Guo
- Department of OrthopedicsTongji Medical CollegeTongji HospitalHuazhong University of Science and TechnologyWuhanChina
| | - Xudong Yao
- Department of OrthopedicsTongji Medical CollegeTongji HospitalHuazhong University of Science and TechnologyWuhanChina
| | - Zhou Guo
- Department of OrthopedicsTongji Medical CollegeTongji HospitalHuazhong University of Science and TechnologyWuhanChina
| | - Fengjing Guo
- Department of OrthopedicsTongji Medical CollegeTongji HospitalHuazhong University of Science and TechnologyWuhanChina
| |
Collapse
|
41
|
Gille J, Reiss E, Freitag M, Schagemann J, Steinwachs M, Piontek T, Reiss E. Autologous Matrix-Induced Chondrogenesis for Treatment of Focal Cartilage Defects in the Knee: A Follow-up Study. Orthop J Sports Med 2021; 9:2325967120981872. [PMID: 33738308 PMCID: PMC7934047 DOI: 10.1177/2325967120981872] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 08/17/2020] [Indexed: 12/19/2022] Open
Abstract
Background: Autologous matrix-induced chondrogenesis (AMIC) is a well-established treatment for full-thickness cartilage defects. Purpose: To evaluate the long-term clinical outcomes of AMIC for the treatment of chondral lesions of the knee. Study Design: Case series; Level of evidence, 4. Methods: A multisite prospective registry recorded demographic data and outcomes for patients who underwent repair of chondral defects. In total, 131 patients were included in the study. Lysholm, Knee injury and Osteoarthritis Outcome Score (KOOS), and visual analog scale (VAS) score for pain were used for outcome analysis. Across all patients, the mean ± SD age of patients was 36.6 ± 11.7 years. The mean body weight was 80.0 ± 16.8 kg, mean height was 176.3 ± 7.9 cm, and mean defect size was 3.3 ± 1.8 cm2. Defects were classified as Outerbridge grade III or IV. A repeated-measures analysis of variance was used to compare outcomes across all time points. Results: The median follow-up time for the patients in this cohort was 4.56 ± 2.92 years. Significant improvement (P < .001) in all scores was observed at 1 to 2 years after AMIC, and improved values were noted up to 7 years postoperatively. Among all patients, the mean preoperative Lysholm score was 46.9 ± 19.6. At the 1-year follow-up, a significantly higher mean Lysholm score was noted, with maintenance of the favorable outcomes at 7-year follow-up. The KOOS also showed a significant improvement of postoperative values compared with preoperative data. The mean VAS had significantly decreased during the 7-year follow-up. Age, sex, and defect size did not have a significant effect on the outcomes. Conclusion: AMIC is an effective method of treating chondral defects of the knee and leads to reliably favorable results up to 7 years postoperatively.
Collapse
Affiliation(s)
- Justus Gille
- Department of Orthopaedic and Trauma Surgery, University Hospital Schleswig-Holstein, Campus Luebeck, Luebeck, Germany
| | | | - Moritz Freitag
- Department of Orthopaedic and Trauma Surgery, University Hospital Schleswig-Holstein, Campus Luebeck, Luebeck, Germany
| | - Jan Schagemann
- Department of Orthopaedic and Trauma Surgery, University Hospital Schleswig-Holstein, Campus Luebeck, Luebeck, Germany
| | | | - Tomasz Piontek
- Sport Medicine Lab, Department of Spine Disorders and Pediatric Orthopaedics, University of Medical Sciences Poznań, Poznán, Poland
| | | |
Collapse
|
42
|
Querido W, Kandel S, Pleshko N. Applications of Vibrational Spectroscopy for Analysis of Connective Tissues. Molecules 2021; 26:922. [PMID: 33572384 PMCID: PMC7916244 DOI: 10.3390/molecules26040922] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/30/2021] [Accepted: 02/04/2021] [Indexed: 02/07/2023] Open
Abstract
Advances in vibrational spectroscopy have propelled new insights into the molecular composition and structure of biological tissues. In this review, we discuss common modalities and techniques of vibrational spectroscopy, and present key examples to illustrate how they have been applied to enrich the assessment of connective tissues. In particular, we focus on applications of Fourier transform infrared (FTIR), near infrared (NIR) and Raman spectroscopy to assess cartilage and bone properties. We present strengths and limitations of each approach and discuss how the combination of spectrometers with microscopes (hyperspectral imaging) and fiber optic probes have greatly advanced their biomedical applications. We show how these modalities may be used to evaluate virtually any type of sample (ex vivo, in situ or in vivo) and how "spectral fingerprints" can be interpreted to quantify outcomes related to tissue composition and quality. We highlight the unparalleled advantage of vibrational spectroscopy as a label-free and often nondestructive approach to assess properties of the extracellular matrix (ECM) associated with normal, developing, aging, pathological and treated tissues. We believe this review will assist readers not only in better understanding applications of FTIR, NIR and Raman spectroscopy, but also in implementing these approaches for their own research projects.
Collapse
Affiliation(s)
| | | | - Nancy Pleshko
- Department of Bioengineering, Temple University, Philadelphia, PA 19122, USA; (W.Q.); (S.K.)
| |
Collapse
|
43
|
Zhu J, Fu Q, Shao J, Peng J, Qian Q, Zhou Y, Chen Y. Over-expression of MEG3 promotes differentiation of bone marrow mesenchymal stem cells into chondrocytes by regulating miR-129-5p/RUNX1 axis. Cell Cycle 2021; 20:96-111. [PMID: 33410373 DOI: 10.1080/15384101.2020.1863043] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
This study explored the role of MEG3 in the cartilage differentiation of bone marrow mesenchymal stem cells (BMSCs). We investigated the effects of over-expression and knockdown of MEG3 on cell viability, cell differentiation, and the expressions of MEG3, miR-129-5p, COL2, chondrocyte differentiation-related genes (sry-type high-mobility-group box 9 (SOX9), SOX5, Aggrecan, silent information regulator 1 (SIRT1), and Cartilage oligomeric matrix protein (COMP)). The targeting relationship between MEG3 and miR-129-5p and the target gene of miR-129-5p was confirmed through Starbase, TargetScan and luciferase experiments. Finally, a series of rescue experiments were conducted to study the regulatory effects of MEG3 and miR-129-5p. BMSCs were identified as CD29+ and CD44+ positive, and their differentiation was time-dependent. As BMSCs differentiated, MEG3 expression was up-regulated, but miR-129-5p was down-regulated. Over-expressed MEG3 promoted the viability and differentiation of BMSCs, up-regulated the expressions of COL2 and chondrocyte differentiation-related genes, and inhibited miR-129-5p. Runt-related transcription factor 1 (RUNX1) was negatively regulated as a target gene of miR-129-5p. Results of rescue experiments showed that the inhibitory effect of miR-129-5p mimic on BMSCs could be partially reversed by MEG3. Over-expression of MEG3 regulated the miR-129-5p/RUNX1 axis to promote the differentiation of BMSCs into chondrocytes. This study provides a reliable basis for the application of lncRNA in articular cartilage injury.
Collapse
Affiliation(s)
- Jun Zhu
- Department of Joint Surgery and Orthopedic Medicine, Shanghai Changzheng Hospital, Second Military Medical University , Shanghai, China
| | - Qiwei Fu
- Department of Joint Surgery and Orthopedic Medicine, Shanghai Changzheng Hospital, Second Military Medical University , Shanghai, China
| | - Jiahua Shao
- Department of Joint Surgery and Orthopedic Medicine, Shanghai Changzheng Hospital, Second Military Medical University , Shanghai, China
| | - Jinhui Peng
- Department of Joint Surgery and Orthopedic Medicine, Shanghai Changzheng Hospital, Second Military Medical University , Shanghai, China
| | - Qirong Qian
- Department of Joint Surgery and Orthopedic Medicine, Shanghai Changzheng Hospital, Second Military Medical University , Shanghai, China
| | - Yiqin Zhou
- Department of Joint Surgery and Orthopedic Medicine, Shanghai Changzheng Hospital, Second Military Medical University , Shanghai, China
| | - Yi Chen
- Department of Joint Surgery and Orthopedic Medicine, Shanghai Changzheng Hospital, Second Military Medical University , Shanghai, China
| |
Collapse
|
44
|
Abstract
As our understanding of hip function and disease improves, it is evident that the acetabular fossa has received little attention, despite it comprising over half of the acetabulum’s surface area and showing the first signs of degeneration. The fossa’s function is expected to be more than augmenting static stability with the ligamentum teres and being a templating landmark in arthroplasty. Indeed, the fossa, which is almost mature at 16 weeks of intrauterine development, plays a key role in hip development, enabling its nutrition through vascularization and synovial fluid, as well as the influx of chondrogenic stem/progenitor cells that build articular cartilage. The pulvinar, a fibrofatty tissue in the fossa, has the same developmental origin as the synovium and articular cartilage and is a biologically active area. Its unique anatomy allows for homogeneous distribution of the axial loads into the joint. It is composed of intra-articular adipose tissue (IAAT), which has adipocytes, fibroblasts, leucocytes, and abundant mast cells, which participate in the inflammatory cascade after an insult to the joint. Hence, the fossa and pulvinar should be considered in decision-making and surgical outcomes in hip preservation surgery, not only for their size, shape, and extent, but also for their biological capacity as a source of cytokines, immune cells, and chondrogenic stem cells. Cite this article: Bone Joint Res 2020;9(12):857–869.
Collapse
Affiliation(s)
- Pablo A Slullitel
- 'Sir John Charnley' Hip Surgery Unit, Institute of Orthopaedics 'Carlos E. Ottolenghi', Italian Hospital of Buenos Aires, Buenos Aires, Argentina
| | - Daniel Coutu
- Regenerative Medicine Program, The Ottawa Hospital Research Institute, Ottawa, Canada
| | - Martin A Buttaro
- 'Sir John Charnley' Hip Surgery Unit, Institute of Orthopaedics 'Carlos E. Ottolenghi', Italian Hospital of Buenos Aires, Buenos Aires, Argentina
| | - Paul Edgar Beaule
- Division of Orthopaedic Surgery, The Ottawa Hospital, Ottawa, Canada
| | | |
Collapse
|
45
|
Shang L, Ma B, Wang F, Li J, Shen S, Li X, Liu H, Ge S. Nanotextured silk fibroin/hydroxyapatite biomimetic bilayer tough structure regulated osteogenic/chondrogenic differentiation of mesenchymal stem cells for osteochondral repair. Cell Prolif 2020; 53:e12917. [PMID: 33001510 PMCID: PMC7653257 DOI: 10.1111/cpr.12917] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 09/09/2020] [Accepted: 09/11/2020] [Indexed: 12/29/2022] Open
Abstract
OBJECTIVES Articular cartilage plays a vital role in bearing and buffering. Injured cartilage and subchondral bone repair is a crucial challenge in cartilage tissue engineering due to the peculiar structure of osteochondral unit and the requirement of osteogenic/chondrogenic bi-directional differentiation. Based on the bionics principle, a nanotextured silk fibroin (SF)-chondroitin sulphate (CS)/hydroxyapatite (HAp) nanowire tough bilayer structure was prepared for osteochondral repair. METHODS The SF-CS/HAp membrane was constructed by alcohol-induced β-sheet formation serving as the physical crosslink. Its osteochondral repairing capacity was evaluated by culturing bone marrow mesenchymal stem cells (BMSCs) in vitro and constructing a rat osteochondral defect model in vivo. RESULTS The bilayer SF-CS/HAp membrane with satisfactory mechanical properties similar to natural cartilage imitated the natural osteochondral unit structural layers and exerted the function of bearing and buffering timely after in vivo implantation. SF-CS layer upregulated the expression of chondrogenesis-related genes of BMSCs by surface nanotopography and sustained release CS. Meanwhile, nanotextured HAp layer assembled with nanowire endowed the membrane with an osteogenic differentiation tendency for BMSCs. In vivo results proved that the biomimetic bilayer structure dramatically promoted new cartilage formation and subchondral bone remodelling for osteochondral defect model after implantation. CONCLUSIONS The SF-CS/HAp biomimetic bilayer membrane provides a promising strategy for precise osteochondral repair.
Collapse
Affiliation(s)
- Lingling Shang
- Department of PeriodontologySchool and Hospital of StomatologyCheeloo College of MedicineShandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue RegenerationJinanChina
| | - Baojin Ma
- State Key Laboratory of Crystal MaterialsShandong UniversityJinanChina
| | - Fulei Wang
- State Key Laboratory of Crystal MaterialsShandong UniversityJinanChina
| | - Jianhua Li
- Department of PeriodontologySchool and Hospital of StomatologyCheeloo College of MedicineShandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue RegenerationJinanChina
| | - Song Shen
- Department of PeriodontologySchool and Hospital of StomatologyCheeloo College of MedicineShandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue RegenerationJinanChina
| | - Xiaoyuan Li
- Department of PeriodontologySchool and Hospital of StomatologyCheeloo College of MedicineShandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue RegenerationJinanChina
| | - Hong Liu
- State Key Laboratory of Crystal MaterialsShandong UniversityJinanChina
| | - Shaohua Ge
- Department of PeriodontologySchool and Hospital of StomatologyCheeloo College of MedicineShandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue RegenerationJinanChina
| |
Collapse
|
46
|
Schreiner AJ, Stoker AM, Bozynski CC, Kuroki K, Stannard JP, Cook JL. Clinical Application of the Basic Science of Articular Cartilage Pathology and Treatment. J Knee Surg 2020; 33:1056-1068. [PMID: 32583400 DOI: 10.1055/s-0040-1712944] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The joint is an organ with each tissue playing critical roles in health and disease. Intact articular cartilage is an exquisite tissue that withstands incredible biologic and biomechanical demands in allowing movement and function, which is why hyaline cartilage must be maintained within a very narrow range of biochemical composition and morphologic architecture to meet demands while maintaining health and integrity. Unfortunately, insult, injury, and/or aging can initiate a cascade of events that result in erosion, degradation, and loss of articular cartilage such that joint pain and dysfunction ensue. Importantly, articular cartilage pathology affects the health of the entire joint and therefore should not be considered or addressed in isolation. Treating articular cartilage lesions is challenging because left alone, the tissue is incapable of regeneration or highly functional and durable repair. Nonoperative treatments can alleviate symptoms associated with cartilage pathology but are not curative or lasting. Current surgical treatments range from stimulation of intrinsic repair to whole-surface and whole-joint restoration. Unfortunately, there is a relative paucity of prospective, randomized controlled, or well-designed cohort-based clinical trials with respect to cartilage repair and restoration surgeries, such that there is a gap in knowledge that must be addressed to determine optimal treatment strategies for this ubiquitous problem in orthopedic health care. This review article discusses the basic science rationale and principles that influence pathology, symptoms, treatment algorithms, and outcomes associated with articular cartilage defects in the knee.
Collapse
Affiliation(s)
- Anna J Schreiner
- Thompson Laboratory for Regenerative Orthopaedics, University of Missouri, Columbia, Missouri.,Department of Orthopaedic Surgery, University of Missouri, Columbia, Missouri.,BG Center for Trauma and Reconstructive Surgery, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Aaron M Stoker
- Thompson Laboratory for Regenerative Orthopaedics, University of Missouri, Columbia, Missouri.,Department of Orthopaedic Surgery, University of Missouri, Columbia, Missouri
| | - Chantelle C Bozynski
- Thompson Laboratory for Regenerative Orthopaedics, University of Missouri, Columbia, Missouri.,Department of Orthopaedic Surgery, University of Missouri, Columbia, Missouri
| | - Keiichi Kuroki
- Thompson Laboratory for Regenerative Orthopaedics, University of Missouri, Columbia, Missouri
| | - James P Stannard
- Thompson Laboratory for Regenerative Orthopaedics, University of Missouri, Columbia, Missouri.,Department of Orthopaedic Surgery, University of Missouri, Columbia, Missouri
| | - James L Cook
- Thompson Laboratory for Regenerative Orthopaedics, University of Missouri, Columbia, Missouri.,Department of Orthopaedic Surgery, University of Missouri, Columbia, Missouri
| |
Collapse
|
47
|
Barsch F, Niedermair T, Mamilos A, Schmitt VH, Grevenstein D, Babel M, Burgoyne T, Shoemark A, Brochhausen C. Physiological and Pathophysiological Aspects of Primary Cilia-A Literature Review with View on Functional and Structural Relationships in Cartilage. Int J Mol Sci 2020; 21:ijms21144959. [PMID: 32674266 PMCID: PMC7404129 DOI: 10.3390/ijms21144959] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 07/09/2020] [Accepted: 07/10/2020] [Indexed: 02/06/2023] Open
Abstract
Cilia are cellular organelles that project from the cell. They occur in nearly all non-hematopoietic tissues and have different functions in different tissues. In mesenchymal tissues primary cilia play a crucial role in the adequate morphogenesis during embryological development. In mature articular cartilage, primary cilia fulfil chemo- and mechanosensitive functions to adapt the cellular mechanisms on extracellular changes and thus, maintain tissue homeostasis and morphometry. Ciliary abnormalities in osteoarthritic cartilage could represent pathophysiological relationships between ciliary dysfunction and tissue deformation. Nevertheless, the molecular and pathophysiological relationships of ‘Primary Cilia’ (PC) in the context of osteoarthritis is not yet fully understood. The present review focuses on the current knowledge about PC and provide a short but not exhaustive overview of their role in cartilage.
Collapse
Affiliation(s)
- Friedrich Barsch
- Institute of Pathology, University Regensburg, 93053 Regensburg, Germany and Institute of Exercise and Occupational Medicine, Department of Medicine, University of Freiburg, 79106 Freiburg, Germany;
| | - Tanja Niedermair
- Institute of Pathology, University Regensburg, 93053 Regensburg, Germany; (T.N.); (A.M.); (M.B.)
| | - Andreas Mamilos
- Institute of Pathology, University Regensburg, 93053 Regensburg, Germany; (T.N.); (A.M.); (M.B.)
| | - Volker H. Schmitt
- Cardiology I, Centre for Cardiology, University Medical Centre, Johannes Gutenberg University of Mainz, 55122 Mainz, Germany;
| | - David Grevenstein
- Department for Orthopedic and Trauma Surgery, University of Cologne, 50923 Köln, Germany;
| | - Maximilian Babel
- Institute of Pathology, University Regensburg, 93053 Regensburg, Germany; (T.N.); (A.M.); (M.B.)
| | - Thomas Burgoyne
- Royal Brompton Hospital and Harefield NHS Trust, SW3 6NP London and UCL Institute of Ophthalmology, University College London, London EC1V 9EL, UK;
| | - Amelia Shoemark
- Royal Brompton Hospital and Harefield NHS Trust, University of Dundee, Dundee DD1 4HN, UK;
| | - Christoph Brochhausen
- Institute of Pathology, University Regensburg, 93053 Regensburg, Germany; (T.N.); (A.M.); (M.B.)
- Correspondence: ; Tel.: +49-941-944-6636
| |
Collapse
|
48
|
McCreery KP, Calve S, Neu CP. Ontogeny informs regeneration: explant models to investigate the role of the extracellular matrix in cartilage tissue assembly and development. Connect Tissue Res 2020; 61:278-291. [PMID: 32186210 PMCID: PMC7190409 DOI: 10.1080/03008207.2019.1698556] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Accepted: 11/22/2019] [Indexed: 02/03/2023]
Abstract
Osteoarthritis (OA) is typically managed in late stages by replacement of the articular cartilage surface with a prosthesis as an effective, though undesirable outcome. As an alternative, hydrogel implants or growth factor treatments are currently of great interest in the tissue engineering community, and scaffold materials are often designed to emulate the mechanical and chemical composition of mature extracellular matrix (ECM) tissue. However, scaffolds frequently fail to capture the structure and organization of cartilage. Additionally, many current scaffold designs do not mimic processes by which structurally sound cartilage is formed during musculoskeletal development. The objective of this review is to highlight methods that investigate cartilage ontogenesis with native and model systems in the context of regenerative medicine. Specific emphasis is placed on the use of cartilage explant cultures that provide a physiologically relevant microenvironment to study tissue assembly and development. Ex vivo cartilage has proven to be a cost-effective and accessible model system that allows researchers to control the culture conditions and stimuli and perform proteomics and imaging studies that are not easily possible using in vivo experiments, while preserving native cell-matrix interactions. We anticipate our review will promote a developmental biology approach using explanted tissues to guide cartilage tissue engineering and inform new treatment methods for OA and joint damage.
Collapse
Affiliation(s)
| | - Sarah Calve
- Department of Mechanical Engineering, University of Colorado, Boulder, USA
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, USA
| | - Corey P. Neu
- Department of Mechanical Engineering, University of Colorado, Boulder, USA
| |
Collapse
|
49
|
Li J, Xiao H, Luo H, Tan Y, Ni Q, He C, Magdalou J, Chen L, Wang H. GR/HDAC2/TGFβR1 pathway contributes to prenatal caffeine induced-osteoarthritis susceptibility in male adult offspring rats. Food Chem Toxicol 2020; 140:111279. [PMID: 32199975 DOI: 10.1016/j.fct.2020.111279] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 03/05/2020] [Accepted: 03/15/2020] [Indexed: 12/25/2022]
Abstract
Prenatal caffeine exposure (PCE) induces developmental toxicity of multi-organ and susceptibility to multi-disease in offspring. However, the effects of PCE on osteoarthritis susceptibility in adult offspring and its intrauterine programming mechanism remain to be further investigated. Here, we found that PCE induced susceptibility to osteoarthritis in male adult offspring rats, which was related to the inhibited function of cartilage matrix synthesis from fetuses to adults. Meanwhile, PCE consistently downregulated the H3K9ac and expression levels of transforming growth factor β receptor 1 (TGFβR1), and then blocked TGFβ signaling pathway, which contributed to the suppressed cartilage matrix synthesis. Moreover, the high level of corticosterone caused by PCE reduced the H3K9ac level on TGFβR1 promoter region through acting on glucocorticoids receptor (GR) and recruiting histone deacetylase 2 (HDAC2) into the nucleus of fetal chondrocytes. Taken together, PCE induced osteoarthritis susceptibility in male adult offspring rats, which was attributed to the low-functional programming of TGFβR1 induced by corticosterone via GR/HDAC2 signaling.
Collapse
Affiliation(s)
- Jing Li
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China
| | - Hao Xiao
- Department of Orthopedic Surgery, Wuhan University Zhongnan Hospital, Wuhan, 430071, China
| | - Hanwen Luo
- Department of Orthopedic Surgery, Wuhan University Zhongnan Hospital, Wuhan, 430071, China
| | - Yang Tan
- Department of Orthopedic Surgery, Wuhan University Zhongnan Hospital, Wuhan, 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China
| | - Qubo Ni
- Department of Orthopedic Surgery, Wuhan University Zhongnan Hospital, Wuhan, 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China
| | - Chunjiang He
- Department of Medical Genetics, Basic Medical School of Wuhan University, Wuhan, 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China
| | | | - Liaobin Chen
- Department of Orthopedic Surgery, Wuhan University Zhongnan Hospital, Wuhan, 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China.
| | - Hui Wang
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China.
| |
Collapse
|
50
|
Johnstone B, Stoddart MJ, Im GI. Multi-Disciplinary Approaches for Cell-Based Cartilage Regeneration. J Orthop Res 2020; 38:463-472. [PMID: 31478253 DOI: 10.1002/jor.24458] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Accepted: 08/23/2019] [Indexed: 02/04/2023]
Abstract
Articular cartilage does not regenerate in adults. A lot of time and resources have been dedicated to cartilage regeneration research. The current understanding suggests that multi-disciplinary approach including biologic, genetic, and mechanical stimulations may be needed for cell-based cartilage regeneration. This review summarizes contents of a workshop sponsored by International Combined Orthopaedic Societies during the 2019 annual meeting of the Orthopaedic Research Society held in Austin, Texas. Three approaches for cell-based cartilage regeneration were introduced, including cellular basis of chondrogenesis, gene-enhanced cartilage regeneration, and physical modulation to divert stem cells to chondrogenic cell fate. While the complicated nature of cartilage regeneration has not allowed us to achieve successful regeneration of hyaline articular cartilage so far, the utilization of multi-disciplinary approaches in various fields of biomedical engineering will provide means to achieve this goal faster. © 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 38:463-472, 2020.
Collapse
Affiliation(s)
- Brian Johnstone
- Department of Orthopaedics and Rehabilitation, Oregon Health & Science University, Portland, Oregon
| | | | - Gun-Il Im
- Integrative Research Institute for Regenerative Biomedical Engineering, Dongguk University, Goyang, Republic of Korea
| |
Collapse
|