1
|
Li J, Zhang Y, Gan X, Li J, Xia G, He L, Xia C, Zhang W, Akhtar Ali K, Zhu M, Huang H. Blocking the LRH-1/LCN2 axis by ML-180, an LRH-1 inverse agonist, ameliorates osteoarthritis via inhibiting the MAPK pathway. Biochem Pharmacol 2025; 237:116922. [PMID: 40194607 DOI: 10.1016/j.bcp.2025.116922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 02/28/2025] [Accepted: 04/02/2025] [Indexed: 04/09/2025]
Abstract
Osteoarthritis (OA) is a chronic and degenerative disease marked by inflammation and extracellular matrix (ECM) degeneration, contributing to synovial inflammation and cartilage destruction. Accumulating evidence has demonstrated that Liver receptor homolog-1 (LRH-1), an orphan nuclear receptor, mediates inflammatory response. However, there is a lack of evidence regarding the regulatory role of LRH-1 in OA pathogenesis. In this study, we confirmed that chondrocytes expressed LRH-1, and observed its upregulation in both IL-1β-treated chondrocytes and cartilage of destabilization of the medial meniscus (DMM)-operated mice. Overexpression of LRH-1 promoted inflammation and dysregulation of ECM metabolism in IL-1β-induced chondrocytes, reversed by inhibition of LRH-1 with ML-180 or gene silencing to protect chondrocytes. Moreover, ML-180 treatment in vivo improved the deteriorated OA phenotypes in mouse models, alleviating OA development. Mechanistically, RNA sequencing revealed that Lipocalin-2 (LCN2), a member of the lipocalin family associated with inflammation, is located downstream of LRH-1 and is positively regulated by it. Furthermore, the LRH-1/LCN2 axis mainly relied on activating the mitogen-activated protein kinase (MAPK) signaling pathway to promote inflammation and dysregulation of ECM metabolism, ultimately damaging chondrocytes. Our findings demonstrate that LRH-1 positively modulates LCN2,activating the MAPK pathway, indicating that targeting the LRH-1/LCN2/MAPK axis may represent a potential therapeutic strategy for OA.
Collapse
Affiliation(s)
- Jianwen Li
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Yayun Zhang
- Department of Traumatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Xin Gan
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Junhong Li
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Ganqing Xia
- Department of Orthopedics, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, China
| | - Lingxiao He
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Chengyan Xia
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Weikai Zhang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Khan Akhtar Ali
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Meipeng Zhu
- Department of Pediatric Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
| | - Hui Huang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
| |
Collapse
|
2
|
Ponce A, Jimenez L, Roldan ML, Shoshani L. Ion Currents Mediated by TRPA1 Channels in Freshly Dissociated Rat Articular Chondrocytes: Biophysical Properties and Regulation by Inflammatory Processes. Pharmaceuticals (Basel) 2025; 18:332. [PMID: 40143111 PMCID: PMC11944639 DOI: 10.3390/ph18030332] [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: 01/28/2025] [Revised: 02/19/2025] [Accepted: 02/23/2025] [Indexed: 03/28/2025] Open
Abstract
Background: Articular chondrocytes are specialized cells in synovial joint cartilage, responsible for maintaining and regenerating the extracellular matrix. Inflammation disrupts the balance between matrix synthesis and degradation, leading to cartilage breakdown. This process, commonly observed in conditions such as osteoarthritis, results in chondrocyte dysfunction and accelerates joint degeneration. Since TRPA1 channels are implicated in inflammatory processes, this study investigates the expression of TRPA1 channels in freshly dissociated rat articular chondrocytes and their modulation by anti-inflammatory agents. Methods: We used the whole-cell patch-clamp method to assess TRPA1 channel expression and modulation. Results: Freshly dissociated chondrocytes exhibit ion currents attributable to TRPA1 channel expression, with higher magnitudes observed in medium-sized cells. These currents decrease over time in primary culture. Treatment with pro-inflammatory agents (IL-1α, IL-1β, and LPS) increases TRPA1's current magnitude. IL-1β treatment directly induces transient TRPA1 currents. Several signaling components activated during inflammation contribute to the IL-1β-induced enhancement of TRPA1 current density, including IL-1 R1, the adaptor protein MyD88, and the downstream kinases IRAK1 and IRAK4. Conclusions: Our findings demonstrate that healthy rat chondrocytes express functional TRPA1 channels and that inflammatory processes modulate their expression.
Collapse
Affiliation(s)
- Arturo Ponce
- Department of Physiology, Biophysics and Neurosciences, CINVESTAV-IPN, Mexico City 07360, Mexico; (L.J.); (M.L.R.); (L.S.)
| | | | | | | |
Collapse
|
3
|
Brahmachary PP, Erdogan AE, Myers EP, June RK. Metabolomic Profiling and Characterization of a Novel 3D Culture System for Studying Chondrocyte Mechanotransduction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.06.10.598340. [PMID: 38915493 PMCID: PMC11195103 DOI: 10.1101/2024.06.10.598340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Background/Objective Articular chondrocytes synthesize and maintain the avascular and aneural articular cartilage. In vivo these cells are surrounded by a 3D pericellular matrix (PCM) containing predominantly collagen VI. The PCM protects chondrocytes and facilitates mechanotransduction. PCM stiffness is critical in transmitting biomechanical signals to chondrocytes. Various culture systems with different hydrogels are used to encapsulate chondrocytes for 3D culture, but many lack either the PCM or the in vivo stiffness of the cartilage matrix. This study aimed at establishing a culture system to investigate a) if chondrocytes cultured in alginate will develop a PCM and b) study mechanotransduction via metabolic changes induced in 3D agarose-embedded chondrocytes upon mechanical stimulation. Methods We cultured primary human and bovine chondrocytes in monolayers or as alginate encapsulated cells in media containing sodium L-ascorbate. PCM expression was analyzed by immunofluorescence and western blots. We further characterized the response of chondrocytes embedded in physiologically stiff agarose to dynamic compression through metabolomic profiling. Results We found that primary human and bovine chondrocytes, when cultured in alginate beads with addition of sodium L-ascorbate for 7 days, had a pronounced PCM, retained their phenotype, and synthesized both collagens VI and II. This novel culture system enables alginate-encapsulated chondrocytes to develop a robust PCM thereby creating a model system to study mechanotransduction in the presence of an endogenous PCM. We also observed distinct compression-induced changes in metabolomic profiles between the monolayer-agarose and alginate-released agarose-embedded chondrocytes indicating physiological changes in cell metabolism. Conclusion/Significance These data show that 3D preculture of chondrocytes in alginate before encapsulation in physiologically stiff agarose leads to pronounced development of pericellular matrix that is sustained in the presence of ascorbate. This model can be useful in studying the mechanism by which chondrocytes respond to cyclical compression and other types of loading simulating in vivo physiological conditions.
Collapse
Affiliation(s)
- Priyanka P Brahmachary
- Department of Mechanical & Industrial Engineering, Montana State University, Bozeman, MT 59717
| | - Ayten E Erdogan
- Department of Mechanical & Industrial Engineering, Montana State University, Bozeman, MT 59717
| | - Erik P Myers
- Department of Mechanical & Industrial Engineering, Montana State University, Bozeman, MT 59717
| | - Ronald K June
- Department of Mechanical & Industrial Engineering, Montana State University, Bozeman, MT 59717
- Department of Microbiology & Cell Biology, Montana State University, Bozeman, MT 59717
| |
Collapse
|
4
|
Semitela A, Marques PAAP, Completo A. Strategies to engineer articular cartilage with biomimetic zonal features: a review. Biomater Sci 2024; 12:5961-6005. [PMID: 39463257 DOI: 10.1039/d4bm00579a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Articular cartilage (AC) is a highly specialized tissue with restricted ability for self-regeneration, given its avascular and acellular nature. Although a considerable number of surgical treatments is available for the repair, reconstruction, and regeneration of AC defects, most of them do not prioritize the development of engineered cartilage with zonal stratification derived from biomimetic biochemical, biomechanical and topographic cues. In the absence of these zonal elements, engineered cartilage will exhibit increased susceptibility to failure and will neither be able to withstand the mechanical loading to which AC is subjected nor will it integrate well with the surrounding tissue. In this regard, new breakthroughs in the development of hierarchical stratified engineered cartilage are highly sought after. Initially, this review provides a comprehensive analysis of the composition and zonal organization of AC, aiming to enhance our understanding of the significance of the structure of AC for its function. Next, we direct our attention towards the existing in vitro and in vivo studies that introduce zonal elements in engineered cartilage to elicit appropriate AC regeneration by employing tissue engineering strategies. Finally, the advantages, challenges, and future perspectives of these approaches are presented.
Collapse
Affiliation(s)
- Angela Semitela
- Centre of Mechanical Technology and Automation (TEMA), Department of Mechanical Engineering, University of Aveiro, 3810-193 Aveiro, Portugal.
| | - Paula A A P Marques
- Centre of Mechanical Technology and Automation (TEMA), Department of Mechanical Engineering, University of Aveiro, 3810-193 Aveiro, Portugal.
| | - António Completo
- Centre of Mechanical Technology and Automation (TEMA), Department of Mechanical Engineering, University of Aveiro, 3810-193 Aveiro, Portugal.
| |
Collapse
|
5
|
Carton F, Rizzi M, Canciani E, Sieve G, Di Francesco D, Casarella S, Di Nunno L, Boccafoschi F. Use of Hydrogels in Regenerative Medicine: Focus on Mechanical Properties. Int J Mol Sci 2024; 25:11426. [PMID: 39518979 PMCID: PMC11545898 DOI: 10.3390/ijms252111426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 10/16/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024] Open
Abstract
Bioengineered materials represent an innovative option to support the regenerative processes of damaged tissues, with the final objective of creating a functional environment closely mimicking the native tissue. Among the different available biomaterials, hydrogels represent the solution of choice for tissue regeneration, thanks to the easy synthesis process and the highly tunable physical and mechanical properties. Moreover, hydrogels are biocompatible and biodegradable, able to integrate in biological environments and to support cellular interactions in order to restore damaged tissues' functionality. This review offers an overview of the current knowledge concerning hydrogel synthesis and characterization and of the recent achievements in their experimental use in supporting skin, bone, cartilage, and muscle regeneration. The currently available in vitro and in vivo results are of great interest, highlighting the need for carefully designed and controlled preclinical studies and clinical trials to support the transition of these innovative biomaterials from the bench to the bedside.
Collapse
Affiliation(s)
- Flavia Carton
- Department of Health Sciences, Università del Piemonte Orientale, 28100 Novara, Italy (E.C.); (S.C.)
| | - Manuela Rizzi
- Department of Health Sciences, Università del Piemonte Orientale, 28100 Novara, Italy (E.C.); (S.C.)
| | - Elena Canciani
- Department of Health Sciences, Università del Piemonte Orientale, 28100 Novara, Italy (E.C.); (S.C.)
| | - Gianluca Sieve
- Department of Health Sciences, Università del Piemonte Orientale, 28100 Novara, Italy (E.C.); (S.C.)
| | - Dalila Di Francesco
- Department of Health Sciences, Università del Piemonte Orientale, 28100 Novara, Italy (E.C.); (S.C.)
- Laboratory for Biomaterials and Bioengineering, CRC-I, Department of Min-Met-Materials Engineering, University Hospital Research Center, Regenerative Medicine, Laval University, Quebec City, QC G1V 0A6, Canada
| | - Simona Casarella
- Department of Health Sciences, Università del Piemonte Orientale, 28100 Novara, Italy (E.C.); (S.C.)
| | - Luca Di Nunno
- Department of Health Sciences, Università del Piemonte Orientale, 28100 Novara, Italy (E.C.); (S.C.)
- Laboratory for Biomaterials and Bioengineering, CRC-I, Department of Min-Met-Materials Engineering, University Hospital Research Center, Regenerative Medicine, Laval University, Quebec City, QC G1V 0A6, Canada
| | - Francesca Boccafoschi
- Department of Health Sciences, Università del Piemonte Orientale, 28100 Novara, Italy (E.C.); (S.C.)
| |
Collapse
|
6
|
Clement RGE, Wong SJ, Hall A, Howie SEM, Simpson AHRW. The long-term time course of septic arthritis. Bone Jt Open 2024; 5:785-792. [PMID: 39293801 PMCID: PMC11410400 DOI: 10.1302/2633-1462.59.bjo-2024-0048.r1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/20/2024] Open
Abstract
Aims The aims of this study were to: 1) report on a cohort of skeletally mature patients with native hip and knee septic arthritis over a 14-year period; 2) to determine the rate of joint failure in patients who had experienced an episode of hip or knee septic arthritis; and 3) to assess the outcome following septic arthritis relative to the infecting organism, whether those patients infected by Staphylococcus aureus would be more likely to have adverse outcomes than those infected by other organisms. Methods All microbiological samples from joint aspirations between March 2000 and December 2014 at our institution were reviewed in order to identify cases of culture-proven septic arthritis. Cases in children (aged < 16 years) and prosthetic joints were excluded. Data were abstracted on age at diagnosis, sex, joint affected (hip or knee), type of organisms isolated, cause of septic arthritis, comorbidities within the Charlson Comorbidity Index (CCI), details of treatment, and outcome. Results A total of 142 patients were confirmed to have had an episode of septic arthritis in a native hip (n = 17) or knee joint (n = 125). S. aureus accounted for 57.7% of all hip and knee joint infections. There were 13 inpatient deaths attributed to septic arthritis. The median age of the patients who died was 77.5 (46.9 to 92.2) and their median age-adjusted CCI was 8 (6 to 12). A failure of the joint occurred in 26 knees (21%) and nine hips (53%). Of the knee joints infected by S. aureus (n = 71), 23 knees (32%) went into failure of joint, whereas of those infected by other organisms (n = 54), only three knees (6%) failed. Conclusion Based on our study findings, hip and knee septic arthritis long-term outcomes were substantially worse than their immediate outcome suggested. Failure of knee joint is 6.1 times more likely to occur in those infected with S. aureus.
Collapse
Affiliation(s)
| | - Seng J. Wong
- University of Edinburgh, Edinburgh, UK
- Singapore General Hospital, Singapore, Singapore
| | | | | | | |
Collapse
|
7
|
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: 0] [Impact Index Per Article: 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
|
8
|
Zhang Z, Shen C, Zhang P, Xu S, Kong L, Liang X, Li C, Qiu X, Huang J, Cui X. Fundamental, mechanism and development of hydration lubrication: From bio-inspiration to artificial manufacturing. Adv Colloid Interface Sci 2024; 327:103145. [PMID: 38615561 DOI: 10.1016/j.cis.2024.103145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 03/26/2024] [Accepted: 03/30/2024] [Indexed: 04/16/2024]
Abstract
Friction and lubrication are ubiquitous in all kinds of movements and play a vital role in the smooth operation of production machinery. Water is indispensable both in the lubrication systems of natural organisms and in hydration lubrication systems. There exists a high degree of similarity between these systems, which has driven the development of hydration lubrication from biomimetic to artificial manufacturing. In particular, significant advancements have been made in the understanding of the mechanisms of hydration lubrication over the past 30 years. This enhanced understanding has further stimulated the exploration of biomimetic inspiration from natural hydration lubrication systems, to develop novel artificial hydration lubrication systems that are cost-effective, easily transportable, and possess excellent capability. This review summarizes the recent experimental and theoretical advances in the understanding of hydration-lubrication processes. The entire paper is divided into three parts. Firstly, surface interactions relevant to hydration lubrication are discussed, encompassing topics such as hydrogen bonding, hydration layer, electric double layer force, hydration force, and Stribeck curve. The second part begins with an introduction to articular cartilage in biomaterial lubrication, discussing its compositional structure and lubrication mechanisms. Subsequently, three major categories of bio-inspired artificial manufacturing lubricating material systems are presented, including hydrogels, polymer brushes (e.g., neutral, positive, negative and zwitterionic brushes), hydration lubricant additives (e.g., nano-particles, polymers, ionic liquids), and their related lubrication mechanism is also described. Finally, the challenges and perspectives for hydration lubrication research and materials development are presented.
Collapse
Affiliation(s)
- Zekai Zhang
- Center for Advanced Jet Engineering Technologies (CaJET), Key Laboratory of High Efficiency and Clean Mechanical Manufacture of Ministry of Education, School of Mechanical Engineering, Shandong University, Jinan, Shandong 25006, China
| | - Chaojie Shen
- Center for Advanced Jet Engineering Technologies (CaJET), Key Laboratory of High Efficiency and Clean Mechanical Manufacture of Ministry of Education, School of Mechanical Engineering, Shandong University, Jinan, Shandong 25006, China
| | - Peipei Zhang
- Advanced Interdisciplinary Technology Research Center, National Innovation Institute of Defense Technology, Beijing 100071, China
| | - Shulei Xu
- Center for Advanced Jet Engineering Technologies (CaJET), Key Laboratory of High Efficiency and Clean Mechanical Manufacture of Ministry of Education, School of Mechanical Engineering, Shandong University, Jinan, Shandong 25006, China
| | - Lingchao Kong
- Advanced Interdisciplinary Technology Research Center, National Innovation Institute of Defense Technology, Beijing 100071, China
| | - Xiubing Liang
- Advanced Interdisciplinary Technology Research Center, National Innovation Institute of Defense Technology, Beijing 100071, China
| | - Chengcheng Li
- Advanced Interdisciplinary Technology Research Center, National Innovation Institute of Defense Technology, Beijing 100071, China
| | - Xiaoyong Qiu
- Center for Advanced Jet Engineering Technologies (CaJET), Key Laboratory of High Efficiency and Clean Mechanical Manufacture of Ministry of Education, School of Mechanical Engineering, Shandong University, Jinan, Shandong 25006, China
| | - Jun Huang
- Center for Advanced Jet Engineering Technologies (CaJET), Key Laboratory of High Efficiency and Clean Mechanical Manufacture of Ministry of Education, School of Mechanical Engineering, Shandong University, Jinan, Shandong 25006, China.
| | - Xin Cui
- Advanced Interdisciplinary Technology Research Center, National Innovation Institute of Defense Technology, Beijing 100071, China.
| |
Collapse
|
9
|
Chen M, Jiang Z, Zou X, You X, Cai Z, Huang J. Advancements in tissue engineering for articular cartilage regeneration. Heliyon 2024; 10:e25400. [PMID: 38352769 PMCID: PMC10862692 DOI: 10.1016/j.heliyon.2024.e25400] [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: 11/03/2023] [Revised: 01/25/2024] [Accepted: 01/25/2024] [Indexed: 02/16/2024] Open
Abstract
Articular cartilage injury is a prevalent clinical condition resulting from trauma, tumors, infection, osteoarthritis, and other factors. The intrinsic lack of blood vessels, nerves, and lymphatic vessels within cartilage tissue severely limits its self-regenerative capacity after injury. Current treatment options, such as conservative drug therapy and joint replacement, have inherent limitations. Achieving perfect regeneration and repair of articular cartilage remains an ongoing challenge in the field of regenerative medicine. Tissue engineering has emerged as a key focus in articular cartilage injury research, aiming to utilize cultured and expanded tissue cells combined with suitable scaffold materials to create viable, functional tissues. This review article encompasses the latest advancements in seed cells, scaffolds, and cytokines. Additionally, the role of stimulatory factors including cytokines and growth factors, genetic engineering techniques, biophysical stimulation, and bioreactor systems, as well as the role of scaffolding materials including natural scaffolds, synthetic scaffolds, and nanostructured scaffolds in the regeneration of cartilage tissues are discussed. Finally, we also outline the signaling pathways involved in cartilage regeneration. Our review provides valuable insights for scholars to address the complex problem of cartilage regeneration and repair.
Collapse
Affiliation(s)
- Maohua Chen
- Department of Plastic Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Zhiyuan Jiang
- Department of Plastic Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Xiuyuan Zou
- Department of Plastic Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Xiaobo You
- Department of Plastic Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Zhen Cai
- Department of Plastic Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Jinming Huang
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
10
|
Jarraya M, Guermazi A, Roemer FW. Osteoarthritis year in review 2023: Imaging. Osteoarthritis Cartilage 2024; 32:18-27. [PMID: 37879600 DOI: 10.1016/j.joca.2023.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 09/24/2023] [Accepted: 10/17/2023] [Indexed: 10/27/2023]
Abstract
PURPOSE This narrative review summarizes the original research in the field of in vivo osteoarthritis (OA) imaging between 1 January 2022 and 1 April 2023. METHODS A PubMed search was conducted using the following several terms pertaining to OA imaging, including but not limited to "Osteoarthritis / OA", "Magnetic resonance imaging / MRI", "X-ray" "Computed tomography / CT", "artificial intelligence /AI", "deep learning", "machine learning". This review is organized by topics including the anatomical structure of interest and modality, AI, challenges of OA imaging in the context of clinical trials, and imaging biomarkers in clinical trials and interventional studies. Ex vivo and animal studies were excluded from this review. RESULTS Two hundred and forty-nine publications were relevant to in vivo human OA imaging. Among the articles included, the knee joint (61%) and MRI (42%) were the predominant anatomical area and imaging modalities studied. Marked heterogeneity of structural tissue damage in OA knees was reported, a finding of potential relevance to clinical trial inclusion. The use of AI continues to rise rapidly to be applied in various aspect of OA imaging research but a lack of generalizability beyond highly standardized datasets limit interpretation and wide-spread application. No pharmacologic clinical trials using imaging data as outcome measures have been published in the period of interest. CONCLUSIONS Recent advances in OA imaging continue to heavily weigh on the use of AI. MRI remains the most important modality with a growing role in outcome prediction and classification.
Collapse
Affiliation(s)
- Mohamed Jarraya
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| | - Ali Guermazi
- Department of Radiology, VA Boston Healthcare System, Boston University School of Medicine, Boston, MA, USA.
| | - Frank W Roemer
- Department of Radiology, VA Boston Healthcare System, Boston University School of Medicine, Boston, MA, USA; Department of Radiology, Universitätsklinikum Erlangen & Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.
| |
Collapse
|
11
|
Fredrikson JP, Brahmachary PP, June RK, Cox LM, Chang CB. Pericellular Matrix Formation and Atomic Force Microscopy of Single Primary Human Chondrocytes Cultured in Alginate Microgels. Adv Biol (Weinh) 2024; 8:e2300268. [PMID: 37688354 PMCID: PMC10843004 DOI: 10.1002/adbi.202300268] [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: 06/30/2023] [Revised: 08/21/2023] [Indexed: 09/10/2023]
Abstract
One of the main components of articular cartilage is the chondrocyte's pericellular matrix (PCM), which is critical for regulating mechanotransduction, biochemical cues, and healthy cartilage development. Here, individual primary human chondrocytes (PHC) are encapsulated and cultured in 50 µm diameter alginate microgels using drop-based microfluidics. This unique culturing method enables PCM formation and manipulation of individual cells. Over ten days, matrix formation is observed using autofluorescence imaging, and the elastic moduli of isolated cells are measured using AFM. Matrix production and elastic modulus increase are observed for the chondrons cultured in microgels. Furthermore, the elastic modulus of cells grown in microgels increases ≈ten-fold over ten days, nearly reaching the elastic modulus of in vivo PCM. The AFM data is further analyzed using a Gaussian mixture model and shows that the population of PHCs grown in microgels exhibit two distinct populations with elastic moduli averaging 9.0 and 38.0 kPa. Overall, this work shows that microgels provide an excellent culture platform for the growth and isolation of PHCs, enabling PCM formation that is mechanically similar to native PCM. The microgel culture platform presented here has the potential to revolutionize cartilage regeneration procedures through the inclusion of in vitro developed PCM.
Collapse
Affiliation(s)
- Jacob P Fredrikson
- Department of Chemical & Biological Engineering, Montana State University, P.O. Box 173920, Bozeman, MT, 59717, USA
- Center for Biofilm Engineering, Montana State University, P.O. Box 173980, Bozeman, MT, 59717, USA
| | - Priyanka P Brahmachary
- Department of Mechanical & Industrial Engineering, Montana State University, P.O. Box 173800, Bozeman, MT, 59717, USA
| | - Ronald K June
- Department of Mechanical & Industrial Engineering, Montana State University, P.O. Box 173800, Bozeman, MT, 59717, USA
- Department of Microbiology & Cell Biology, Montana State University, P.O. Box 173520, Bozeman, MT, 59717, USA
| | - Lewis M Cox
- Department of Mechanical & Industrial Engineering, Montana State University, P.O. Box 173800, Bozeman, MT, 59717, USA
| | - Connie B Chang
- Department of Chemical & Biological Engineering, Montana State University, P.O. Box 173920, Bozeman, MT, 59717, USA
- Center for Biofilm Engineering, Montana State University, P.O. Box 173980, Bozeman, MT, 59717, USA
- Department of Physiology & Biomedical Engineering, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA
| |
Collapse
|
12
|
Semitela Â, Pinto SC, Capitão A, Marques PAAP, Completo A. Fabrication of Customizable and Reproducible 3D Chondrocyte-Laden Nanofibrous Architectures: Effect of Specific Fiber Alignments and Porosities on Chondrocyte Response under Cyclic Compression. ACS APPLIED BIO MATERIALS 2023; 6:5541-5554. [PMID: 37947854 DOI: 10.1021/acsabm.3c00737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2023]
Abstract
Electrospinning has been widely employed to fabricate complex extracellular matrix-like microenvironments for tissue engineering due to its ability to replicate structurally biomimetic micro- and nanotopographic cues. Nevertheless, these nanofibrous structures are typically either confined to bidimensional systems or confined to three-dimensional ones that are unable to provide controlled multiscale patterns. Thus, an electrospinning modality was used in this work to fabricate chondrocyte-laden nanofibrous scaffolds with highly customizable three-dimensional (3D) architectures in an automated manner, with the ultimate goal of recreating a suitable 3D scaffold for articular cartilage tissue engineering. Three distinct architectures were designed and fabricated by combining multiple nanofibrous and chondrocyte-laden hydrogel layers and tested in vitro in a compression bioreactor system. Results demonstrated that it was possible to precisely control the placement and alignment of electrospun polycaprolactone and gelatin nanofibers, generating three unique architectures with distinctive macroscale porosity, water absorption capacity, and mechanical properties. The architecture organized in a lattice-like fashion was highly porous with substantial pore interconnectivity, resulting in a high-water absorption capacity but a poor compression modulus and relatively weaker energy dissipation capacity. The donut-like 3D geometry was the densest, with lower swelling, but the highest compression modulus and improved energy dissipation ability. The third architecture combined a lattice and donut-like fibrous arrangement, exhibiting intermediary behavior in terms of porosity, water absorption, compression modulus, and energy dissipation capacity. The properties of the donut-like 3D architecture demonstrated great potential for articular cartilage tissue engineering, as it mimicked key topographic, chemical, and mechanical characteristics of chondrocytes' surrounding environment. In fact, the combination of these architectural features with a dynamically compressive mechanical stimulus triggered the best in vitro results in terms of viability and biosynthetic production.
Collapse
Affiliation(s)
- Ângela Semitela
- Centre of Mechanical Technology and Automation (TEMA), Department of Mechanical Engineering, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Susana C Pinto
- Centre of Mechanical Technology and Automation (TEMA), Department of Mechanical Engineering, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Ana Capitão
- Centre for Neuroscience and Cell Biology (CNC), University of Coimbra, 3004-504 Coimbra, Portugal
| | - Paula A A P Marques
- Centre of Mechanical Technology and Automation (TEMA), Department of Mechanical Engineering, University of Aveiro, 3810-193 Aveiro, Portugal
| | - António Completo
- Centre of Mechanical Technology and Automation (TEMA), Department of Mechanical Engineering, University of Aveiro, 3810-193 Aveiro, Portugal
| |
Collapse
|
13
|
Fan H, Xu P, Chen X, Li Y, Zhang Z, Hsu J, Le M, Ye E, Gao B, Demos H, Yao H, Ye T. Mask R-CNN provides efficient and accurate measurement of chondrocyte viability in the label-free assessment of articular cartilage. OSTEOARTHRITIS AND CARTILAGE OPEN 2023; 5:100415. [PMID: 38025155 PMCID: PMC10679817 DOI: 10.1016/j.ocarto.2023.100415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 11/03/2023] [Indexed: 12/01/2023] Open
Abstract
Objective Chondrocyte viability (CV) can be measured with the label-free method using second harmonic generation (SHG) and two-photon excitation autofluorescence (TPAF) imaging. To automate the image processing for the label-free CV measurement, we previously demonstrated a two-step deep-learning method: Step 1 used a U-Net to segment the lacuna area on SHG images; Step 2 used dual CNN networks to count live cells and the total number of cells in extracted cell clusters from TPAF images. This study aims to develop one-step deep learning methods to improve the efficiency of CV measurement. Method TPAF/SHG images were acquired simultaneously on cartilage samples from rats and pigs using two-photon microscopes and were merged to form RGB color images with red, green, and blue channels assigned to emission bands of oxidized flavoproteins, reduced forms of nicotinamide adenine dinucleotide, and SHG signals, respectively. Based on the Mask R-CNN, we designed a deep learning network and its denoising version using Wiener deconvolution for CV measurement. Results Using training and test datasets from rat and porcine cartilage, we have demonstrated that Mask R-CNN-based networks can segment and classify individual cells with a single-step processing flow. The absolute error (difference between the measured and the ground-truth CV) of the CV measurement using the Mask R-CNN with or without Wiener deconvolution denoising reaches 0.01 or 0.08, respectively; the error of the previous CV networks is 0.18, significantly larger than that of the Mask R-CNN methods. Conclusions Mask R-CNN-based deep-learning networks improve efficiency and accuracy of the label-free CV measurement.
Collapse
Affiliation(s)
- Hongming Fan
- Department of Bioengineering, Clemson University, SC, USA
| | - Pei Xu
- School of Computing, Clemson University, SC, USA
| | - Xun Chen
- Department of Bioengineering, Clemson University, SC, USA
| | - Yang Li
- School of Medicine, Yale University, New Haven, CT, USA
| | - Zhao Zhang
- Department of Bioengineering, Clemson University, SC, USA
| | - Jennifer Hsu
- Department of Bioengineering, Clemson University, SC, USA
- School of Computing, Clemson University, SC, USA
| | - Michael Le
- Department of Bioengineering, Clemson University, SC, USA
| | - Emily Ye
- College of Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Bruce Gao
- Department of Bioengineering, Clemson University, SC, USA
| | - Harry Demos
- Department of Orthopaedics & Physical Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Hai Yao
- Department of Bioengineering, Clemson University, SC, USA
- Department of Orthopaedics & Physical Medicine, Medical University of South Carolina, Charleston, SC, USA
- Department of Oral Health Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Tong Ye
- Department of Bioengineering, Clemson University, SC, USA
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA
| |
Collapse
|
14
|
Macica CM, Tommasini SM. Biomechanical Impact of Phosphate Wasting on Articular Cartilage Using the Murine Hyp Model of X-linked hypophosphatemia. JBMR Plus 2023; 7:e10796. [PMID: 37808393 PMCID: PMC10556269 DOI: 10.1002/jbm4.10796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 06/05/2023] [Accepted: 06/19/2023] [Indexed: 10/10/2023] Open
Abstract
Degenerative osteoarthritis (OA) is recognized as an early-onset comorbidity of X-linked hypophosphatemia (XLH), contributing to pain and stiffness and limiting range of motion and activities of daily living. Here, we extend prior findings describing biochemical and cellular changes of articular cartilage (AC) in the phosphate-wasting environment of XLH to determine the impact of these changes on the biomechanical properties of AC in compression and potential role in the etiology of OA. We hypothesize that despite increased proteoglycan biosynthesis, disruption of the mineralized zone of AC impacts the mechanical properties of cartilage that function to accommodate loads and that therapeutic restoration of this zone will improve the mechanical properties of AC. Data were compared between three groups: wild type (WT), Hyp, and Hyp mice treated with calcitriol and oral phosphate. EPIC microCT confirmed AC mineral deficits and responsiveness to therapy. MicroCT of the Hyp subchondral bone plate revealed that treatment improved trabecular bone volume (BV/TV) but remained significantly lower than WT mice in other trabecular microstructures (p < 0.05). Microindentation AC studies revealed that, compared with WT mice, the mean stiffness of tibial AC was significantly lower in untreated Hyp mice (2.65 ± 0.95 versus 0.87 ± 0.33 N/mm, p < 0.001) and improved with therapy (2.15 + 0.38 N/mm) to within WT values. Stress relaxation of AC under compressive loading displayed similar biphasic relaxation time constants (Taufast and Tauslow) between controls and Hyp mice, although Tauslow trended toward slowed relaxation times. In addition, Taufast and Tauslow times correlated with peak load in WT mice (r = 0.80; r = 0.78, respectively), whereas correlation coefficient values for Hyp mice (r = 0.46; r = 0.21) improved with treatment (r = 0.71; r = 0.56). These data provide rationale for therapies that both preserve AC stiffness and recovery from compression. The Hyp mouse also provides unique insight into determinants of structural stiffness and the viscoelastic properties of AC in the progression of OA. © 2023 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Carolyn M Macica
- Connecticut Children's Research InstituteHartfordCTUSA
- Department of PharmacologyYale University School of MedicineNew HavenCTUSA
| | - Steven M Tommasini
- Department of Orthopaedics and RehabilitationYale University School of MedicineNew HavenCTUSA
| |
Collapse
|
15
|
Zhang Y, Tawiah GK, Wu X, Zhang Y, Wang X, Wei X, Qiao X, Zhang Q. Primary cilium-mediated mechanotransduction in cartilage chondrocytes. Exp Biol Med (Maywood) 2023; 248:1279-1287. [PMID: 37897221 PMCID: PMC10625344 DOI: 10.1177/15353702231199079] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/29/2023] Open
Abstract
Osteoarthritis (OA) is one of the most prevalent joint disorders associated with the degradation of articular cartilage and an abnormal mechanical microenvironment. Mechanical stimuli, including compression, shear stress, stretching strain, osmotic challenge, and the physical properties of the matrix microenvironment, play pivotal roles in the tissue homeostasis of articular cartilage. The primary cilium, as a mechanosensory and chemosensory organelle, is important for detecting and transmitting both mechanical and biochemical signals in chondrocytes within the matrix microenvironment. Growing evidence indicates that primary cilia are critical for chondrocytes signaling transduction and the matrix homeostasis of articular cartilage. Furthermore, the ability of primary cilium to regulate cellular signaling is dynamic and dependent on the cellular matrix microenvironment. In the current review, we aim to elucidate the key mechanisms by which primary cilia mediate chondrocytes sensing and responding to the matrix mechanical microenvironment. This might have potential therapeutic applications in injuries and OA-associated degeneration of articular cartilage.
Collapse
Affiliation(s)
- Yang Zhang
- Department of Histology and Embryology, Shanxi Medical University, Jinzhong 030604, Shanxi, China
- College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan 030024, Shanxi, China
| | - Godfred K Tawiah
- College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan 030024, Shanxi, China
| | - Xiaoan Wu
- Department of Physiology and Biophysics, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Yanjun Zhang
- College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan 030024, Shanxi, China
| | - Xiaohu Wang
- Department of Orthopaedics, The Second Hospital of Shanxi Medical University, Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Shanxi Medical University, Taiyuan 030001, Shanxi, China
| | - Xiaochun Wei
- Department of Orthopaedics, The Second Hospital of Shanxi Medical University, Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Shanxi Medical University, Taiyuan 030001, Shanxi, China
| | - Xiaohong Qiao
- Department of Histology and Embryology, Shanxi Medical University, Jinzhong 030604, Shanxi, China
- Department of Orthopaedics, Lvliang Hospital Affiliated to Shanxi Medical University, Lvliang 033099, Shanxi, China
| | - Quanyou Zhang
- College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan 030024, Shanxi, China
- Department of Orthopaedics, The Second Hospital of Shanxi Medical University, Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Shanxi Medical University, Taiyuan 030001, Shanxi, China
| |
Collapse
|
16
|
Jaramillo-Rangel G, Chávez-Briones MDL, Ancer-Arellano A, Miranda-Maldonado I, Ortega-Martínez M. Back to the Basics: Usefulness of Naturally Aged Mouse Models and Immunohistochemical and Quantitative Morphologic Methods in Studying Mechanisms of Lung Aging and Associated Diseases. Biomedicines 2023; 11:2075. [PMID: 37509714 PMCID: PMC10377355 DOI: 10.3390/biomedicines11072075] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 06/17/2023] [Accepted: 07/19/2023] [Indexed: 07/30/2023] Open
Abstract
Aging-related molecular and cellular alterations in the lung contribute to an increased susceptibility of the elderly to devastating diseases. Although the study of the aging process in the lung may benefit from the use of genetically modified mouse models and omics techniques, these approaches are still not available to most researchers and produce complex results. In this article, we review works that used naturally aged mouse models, together with immunohistochemistry (IHC) and quantitative morphologic (QM) methods in the study of the mechanisms of the aging process in the lung and its most commonly associated disorders: cancer, chronic obstructive pulmonary disease (COPD), and infectious diseases. The advantage of using naturally aged mice is that they present characteristics similar to those observed in human aging. The advantage of using IHC and QM methods lies in their simplicity, economic accessibility, and easy interpretation, in addition to the fact that they provide extremely important information. The study of the aging process in the lung and its associated diseases could allow the design of appropriate therapeutic strategies, which is extremely important considering that life expectancy and the number of elderly people continue to increase considerably worldwide.
Collapse
Affiliation(s)
- Gilberto Jaramillo-Rangel
- Department of Pathology, School of Medicine, Autonomous University of Nuevo León, Monterrey 64460, Mexico
| | | | - Adriana Ancer-Arellano
- Department of Pathology, School of Medicine, Autonomous University of Nuevo León, Monterrey 64460, Mexico
| | - Ivett Miranda-Maldonado
- Department of Pathology, School of Medicine, Autonomous University of Nuevo León, Monterrey 64460, Mexico
| | - Marta Ortega-Martínez
- Department of Pathology, School of Medicine, Autonomous University of Nuevo León, Monterrey 64460, Mexico
| |
Collapse
|
17
|
Yu L, Cavelier S, Hannon B, Wei M. Recent development in multizonal scaffolds for osteochondral regeneration. Bioact Mater 2023; 25:122-159. [PMID: 36817819 PMCID: PMC9931622 DOI: 10.1016/j.bioactmat.2023.01.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 12/30/2022] [Accepted: 01/14/2023] [Indexed: 02/05/2023] Open
Abstract
Osteochondral (OC) repair is an extremely challenging topic due to the complex biphasic structure and poor intrinsic regenerative capability of natural osteochondral tissue. In contrast to the current surgical approaches which yield only short-term relief of symptoms, tissue engineering strategy has been shown more promising outcomes in treating OC defects since its emergence in the 1990s. In particular, the use of multizonal scaffolds (MZSs) that mimic the gradient transitions, from cartilage surface to the subchondral bone with either continuous or discontinuous compositions, structures, and properties of natural OC tissue, has been gaining momentum in recent years. Scrutinizing the latest developments in the field, this review offers a comprehensive summary of recent advances, current hurdles, and future perspectives of OC repair, particularly the use of MZSs including bilayered, trilayered, multilayered, and gradient scaffolds, by bringing together onerous demands of architecture designs, material selections, manufacturing techniques as well as the choices of growth factors and cells, each of which possesses its unique challenges and opportunities.
Collapse
Affiliation(s)
- Le Yu
- Department of Chemical and Biomolecular Engineering, Ohio University, Athens, OH, 45701, USA
| | - Sacha Cavelier
- Department of Chemical and Biomolecular Engineering, Ohio University, Athens, OH, 45701, USA
| | - Brett Hannon
- Biomedical Engineering Program, Ohio University, Athens, OH, 45701, USA
| | - Mei Wei
- Biomedical Engineering Program, Ohio University, Athens, OH, 45701, USA
- Department of Mechanical Engineering, Ohio University, Athens, OH, 45701, USA
| |
Collapse
|
18
|
Cho GH, Bae HC, Cho WY, Jeong EM, Park HJ, Yang HR, Wang SY, Kim YJ, Shin DM, Chung HM, Kim IG, Han HS. High-glutathione mesenchymal stem cells isolated using the FreSHtracer probe enhance cartilage regeneration in a rabbit chondral defect model. Biomater Res 2023; 27:54. [PMID: 37259149 PMCID: PMC10233867 DOI: 10.1186/s40824-023-00398-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 05/20/2023] [Indexed: 06/02/2023] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) are a promising cell source for cartilage regeneration. However, the function of MSC can vary according to cell culture conditions, donor age, and heterogeneity of the MSC population, resulting in unregulated MSC quality control. To overcome these limitations, we previously developed a fluorescent real-time thiol tracer (FreSHtracer) that monitors cellular levels of glutathione (GSH), which are known to be closely associated with stem cell function. In this study, we investigated whether using FreSHtracer could selectively separate high-functioning MSCs based on GSH levels and evaluated the chondrogenic potential of MSCs with high GSH levels to repair cartilage defects in vivo. METHODS Flow cytometry was conducted on FreSHtracer-loaded MSCs to select cells according to their GSH levels. To determine the function of FreSHtracer-isolated MSCs, mRNA expression, migration, and CFU assays were conducted. The MSCs underwent chondrogenic differentiation, followed by analysis of chondrogenic-related gene expression. For in vivo assessment, MSCs with different cellular GSH levels or cell culture densities were injected in a rabbit chondral defect model, followed by histological analysis of cartilage-regenerated defect sites. RESULTS FreSHtracer successfully isolated MSCs according to GSH levels. MSCs with high cellular GSH levels showed enhanced MSC function, including stem cell marker mRNA expression, migration, CFU, and oxidant resistance. Regardless of the stem cell tissue source, FreSHtracer selectively isolated MSCs with high GSH levels and high functionality. The in vitro chondrogenic potential was the highest in pellets generated by MSCs with high GSH levels, with increased ECM formation and chondrogenic marker expression. Furthermore, the MSCs' function was dependent on cell culture conditions, with relatively higher cell culture densities resulting in higher GSH levels. In vivo, improved cartilage repair was achieved by articular injection of MSCs with high levels of cellular GSH and MSCs cultured under high-density conditions, as confirmed by Collagen type 2 IHC, Safranin-O staining and O'Driscoll scores showing that more hyaline cartilage was formed on the defects. CONCLUSION FreSHtracer selectively isolates highly functional MSCs that have enhanced in vitro chondrogenesis and in vivo hyaline cartilage regeneration, which can ultimately overcome the current limitations of MSC therapy.
Collapse
Affiliation(s)
- Gun Hee Cho
- Department of Orthopedic Surgery, College of Medicine, Seoul National University, 101 Daehak-Ro, Jongno-Gu, Seoul, 03080, Republic of Korea
- Department of Orthopedic Surgery, Seoul National University Hospital, Yongondong Chongnogu, Seoul, 110-744, Republic of Korea
| | - Hyun Cheol Bae
- Department of Orthopedic Surgery, Seoul National University Hospital, Yongondong Chongnogu, Seoul, 110-744, Republic of Korea
| | - Won Young Cho
- Department of Orthopedic Surgery, Seoul National University Hospital, Yongondong Chongnogu, Seoul, 110-744, Republic of Korea
| | - Eui Man Jeong
- Department of Pharmacy, College of Pharmacy, Jeju National University, Jeju Special Self-Governing Province, Jeju-do, Republic of Korea
| | - Hee Jung Park
- Department of Orthopedic Surgery, Seoul National University Hospital, Yongondong Chongnogu, Seoul, 110-744, Republic of Korea
| | - Ha Ru Yang
- Department of Orthopedic Surgery, Seoul National University Hospital, Yongondong Chongnogu, Seoul, 110-744, Republic of Korea
| | - Sun Young Wang
- Department of Orthopedic Surgery, Seoul National University Hospital, Yongondong Chongnogu, Seoul, 110-744, Republic of Korea
| | - You Jung Kim
- Department of Orthopedic Surgery, Seoul National University Hospital, Yongondong Chongnogu, Seoul, 110-744, Republic of Korea
| | - Dong Myung Shin
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, 88 Olymic-Ro 43-Gil, Songpa-Gu, Seoul, 05505, Republic of Korea
| | - Hyung Min Chung
- Department of Stem Cell Biology, School of Medicine, Konkuk University, Seoul, 05029, Republic of Korea
| | - In Gyu Kim
- Laboratory for Cellular Response to Oxidative Stress, Cell2in, Inc, Seoul, 03127, Republic of Korea
| | - Hyuk-Soo Han
- Department of Orthopedic Surgery, College of Medicine, Seoul National University, 101 Daehak-Ro, Jongno-Gu, Seoul, 03080, Republic of Korea.
- Department of Orthopedic Surgery, Seoul National University Hospital, Yongondong Chongnogu, Seoul, 110-744, Republic of Korea.
| |
Collapse
|
19
|
Nazarov DA, Denisenko GM, Budylin GS, Kozlova EA, Lipina MM, Lazarev VA, Shirshin EA, Tarabrin MK. Diffuse reflectance spectroscopy of the cartilage tissue in the fourth optical window. BIOMEDICAL OPTICS EXPRESS 2023; 14:1509-1521. [PMID: 37078039 PMCID: PMC10110295 DOI: 10.1364/boe.483135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 02/08/2023] [Accepted: 02/27/2023] [Indexed: 05/03/2023]
Abstract
Studies of the optical properties of biological tissues in the infrared range have demonstrated significant potential for diagnostic tasks. One of the insufficiently explored ranges for diagnostic problems at the moment is the fourth transparency window, or short wavelength infrared region II (SWIR II). A Cr2+:ZnSe laser with tuning capability in the range from 2.1 to 2.4 µm was developed to explore the possibilities in this region. The capability of diffuse reflectance spectroscopy to analyze water and collagen content in biosamples was investigated using the optical gelatin phantoms and the cartilage tissue samples during their drying process. It was demonstrated that decomposition components of the optical density spectra correlated with the partial content of the collagen and water in the samples. The present study indicates the possibility of using this spectral range for the development of diagnostic methods, in particular, for observation of the changes in the content of cartilage tissue components in degenerative diseases such as osteoarthritis.
Collapse
Affiliation(s)
| | - Georgy M. Denisenko
- Bauman Moscow State Technical University, Moscow, 105005, Russia
- Institute for Regenerative Medicine, Sechenov First Moscow State Medical University (Sechenov University), Moscow 119991, Russia
- Laboratory of Clinical Biophotonics, Biomedical Science and Technology Park, Sechenov First Moscow State Medical University, 119048 Moscow, Russia
| | - Gleb S. Budylin
- Laboratory of Clinical Biophotonics, Biomedical Science and Technology Park, Sechenov First Moscow State Medical University, 119048 Moscow, Russia
| | | | - Marina M. Lipina
- Department of Trauma, Orthopedics and Disaster Surgery, Sechenov First Moscow State Medical University, Moscow 119991, Russia
| | - Vladimir A. Lazarev
- Bauman Moscow State Technical University, Moscow, 105005, Russia
- World-Class Research Center Digital Biodesign and Personalized Healthcare, Sechenov First Moscow State Medical University, 119048 Moscow, Russia
| | - Evgeny A. Shirshin
- Laboratory of Clinical Biophotonics, Biomedical Science and Technology Park, Sechenov First Moscow State Medical University, 119048 Moscow, Russia
- Faculty of Physics, M.V. Lomonosov Moscow State University, Moscow 119991, Russia
| | | |
Collapse
|
20
|
Gu Z, Wang J, Fu Y, Pan H, He H, Gan Q, Liu C. Smart Biomaterials for Articular Cartilage Repair and Regeneration. ADVANCED FUNCTIONAL MATERIALS 2023; 33. [DOI: 10.1002/adfm.202212561] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Indexed: 01/06/2025]
Abstract
AbstractArticular cartilage defects bring about disability and worldwide socioeconomic loss, therefore, articular cartilage repair and regeneration is recognized as a global issue. However, due to its avascular and nearly acellular characteristic, cartilage tissue regeneration ability is limited to some extent. Despite the availability of various treatment methods, including palliative drugs and surgical regenerative therapy, articular cartilage repair and regeneration still face major challenges due to the lack of appropriate methods and materials. Smart biomaterials can regulate cell behavior and provide excellent tissue repair and regeneration microenvironment, thus inducing articular cartilage repair and regeneration. This process is adjusted by controlling drug/bioactive factors release via responding to exogenous/endogenous stimuli, tailoring materials’ structure and function similar to native cartilage or providing physiochemical and physical signaling factors. Herein, smart biomaterials, recently applied in articular cartilage repair and regeneration, are elaborated from two aspects: smart drug release system and smart scaffolds. Furthermore, articular cartilage and its defects and advanced manufacturing techniques of smart biomaterials are discussed in brief. Finally, perspectives for smart biomaterials used in articular cartilage repair and regeneration are presented and the clinical translation of smart biomaterials is emphasized.
Collapse
Affiliation(s)
- Zhanghao Gu
- Key Laboratory for Ultrafine Materials of Ministry of Education East China University of Science and Technology Shanghai 200237 P. R. China
- School of Materials Science and Engineering East China University of Science and Technology Shanghai 200237 P. R. China
| | - Jiayi Wang
- Key Laboratory for Ultrafine Materials of Ministry of Education East China University of Science and Technology Shanghai 200237 P. R. China
- School of Materials Science and Engineering East China University of Science and Technology Shanghai 200237 P. R. China
| | - Yu Fu
- School of Aerospace Engineering and Applied Mechanics Tongji University Zhangwu Road 100 Shanghai 200092 P. R. China
| | - Hao Pan
- Key Laboratory for Ultrafine Materials of Ministry of Education East China University of Science and Technology Shanghai 200237 P. R. China
- School of Materials Science and Engineering East China University of Science and Technology Shanghai 200237 P. R. China
| | - Hongyan He
- Key Laboratory for Ultrafine Materials of Ministry of Education East China University of Science and Technology Shanghai 200237 P. R. China
- School of Materials Science and Engineering East China University of Science and Technology Shanghai 200237 P. R. China
- Engineering Research Center for Biomedical Materials of the Ministry of Education East China University of Science and Technology Shanghai 200237 P. R. China
| | - Qi Gan
- Key Laboratory for Ultrafine Materials of Ministry of Education East China University of Science and Technology Shanghai 200237 P. R. China
- School of Materials Science and Engineering East China University of Science and Technology Shanghai 200237 P. R. China
- Engineering Research Center for Biomedical Materials of the Ministry of Education East China University of Science and Technology Shanghai 200237 P. R. China
| | - Changsheng Liu
- Key Laboratory for Ultrafine Materials of Ministry of Education East China University of Science and Technology Shanghai 200237 P. R. China
- School of Materials Science and Engineering East China University of Science and Technology Shanghai 200237 P. R. China
- Engineering Research Center for Biomedical Materials of the Ministry of Education East China University of Science and Technology Shanghai 200237 P. R. China
- Frontiers Science Center for Materiobiology and Dynamic Chemistry East China University of Science and Technology Shanghai 200237 P. R. China
| |
Collapse
|
21
|
Fan H, Xu P, Chen X, Li Y, Hsu J, Le M, Zhang Z, Ye E, Gao B, Ye T. Measuring chondrocyte viability of articular cartilage based on label-free two-photon microscopy and deep learning image analysis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.13.527931. [PMID: 36824870 PMCID: PMC9949096 DOI: 10.1101/2023.02.13.527931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
Objective Chondrocyte viability (CV) is an important indicator of articular cartilage health. Two-photon excitation autofluorescence (TPAF) and second harmonic generation (SHG) microscopy provide a label-free method for imaging chondrocytes. In this study, we propose an automated assessment of CV using deep learning cell segmentation and counting based on acquired TPAF/SHG images. Design Label-free TPAF/SHG images of cartilage samples from rats and porcine were acquired using both commercial and home-built two-photon microscopes, respectively. TPAF/SHG images were merged to form RGB color images with red, green, and blue channels assigned to TPAF (two channels) and SHG signals, respectively. To make the training datasets for the deep learning networks, individual chondrocyte areas on the RGB color images were manually circled and live or dead chondrocytes were validated by using Calcein-AM and Ethidium homodimer-1 dye labeling. We first built a chondrocyte viability network (MCV-Net) using the Mask R-CNN architecture, which could provide individual segmented cellular areas with live or dead status. Wiener deconvolution preprocessing was added before the input of MCV-Net to improve the accuracy of the CV analysis, forming the Wiener deconvolution CV network (wMCV-Net). Results Training (300 images) and test (120 images) datasets were built for rats and porcine cartilage respectively. Wiener deconvolution could improve the Peak Signal-to-Noise Ratio (PSNR) for 30-40%. We demonstrated that both MCV-Net and wMCV-Net significantly improved the accuracy of the CV measurement. Conclusion A custom desktop TPAF/SHG microscope was used in collaboration with deep learning algorithm wMCV-Net based label-free method to assess the CV and get 95% accuracy with both rats and porcine samples.
Collapse
|
22
|
Sarkar A, Liu NQ, Magallanes J, Tassey J, Lee S, Shkhyan R, Lee Y, Lu J, Ouyang Y, Tang H, Bian F, Tao L, Segil N, Ernst J, Lyons K, Horvath S, Evseenko D. STAT3 promotes a youthful epigenetic state in articular chondrocytes. Aging Cell 2023; 22:e13773. [PMID: 36638270 PMCID: PMC9924946 DOI: 10.1111/acel.13773] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 12/21/2022] [Indexed: 01/14/2023] Open
Abstract
Epigenetic mechanisms guiding articular cartilage regeneration and age-related disease such as osteoarthritis (OA) are poorly understood. STAT3 is a critical age-patterned transcription factor highly active in fetal and OA chondrocytes, but the context-specific role of STAT3 in regulating the epigenome of cartilage cells remain elusive. In this study, DNA methylation profiling was performed across human chondrocyte ontogeny to build an epigenetic clock and establish an association between CpG methylation and human chondrocyte age. Exposure of adult chondrocytes to a small molecule STAT3 agonist decreased DNA methylation, while genetic ablation of STAT3 in fetal chondrocytes induced global hypermethylation. CUT&RUN assay and subsequent transcriptional validation revealed DNA methyltransferase 3 beta (DNMT3B) as one of the putative STAT3 targets in chondrocyte development and OA. Functional assessment of human OA chondrocytes showed the acquisition of progenitor-like immature phenotype by a significant subset of cells. Finally, conditional deletion of Stat3 in cartilage cells increased DNMT3B expression in articular chondrocytes in the knee joint in vivo and resulted in a more prominent OA progression in a post-traumatic OA (PTOA) mouse model induced by destabilization of the medial meniscus (DMM). Taken together these data reveal a novel role for STAT3 in regulating DNA methylation in cartilage development and disease. Our findings also suggest that elevated levels of active STAT3 in OA chondrocytes may indicate an intrinsic attempt of the tissue to regenerate by promoting a progenitor-like phenotype. However, it is likely that chronic activation of this pathway, induced by IL-6 cytokines, is detrimental and leads to tissue degeneration.
Collapse
Affiliation(s)
- Arijita Sarkar
- Department of Orthopaedic Surgery, Keck School of Medicine of USCUniversity of Southern California (USC)Los AngelesCaliforniaUSA
| | - Nancy Q. Liu
- Department of Orthopaedic Surgery, Keck School of Medicine of USCUniversity of Southern California (USC)Los AngelesCaliforniaUSA
| | - Jenny Magallanes
- Department of Orthopaedic Surgery, Keck School of Medicine of USCUniversity of Southern California (USC)Los AngelesCaliforniaUSA
| | - Jade Tassey
- Department of Orthopaedic Surgery, Keck School of Medicine of USCUniversity of Southern California (USC)Los AngelesCaliforniaUSA
| | - Siyoung Lee
- Department of Orthopaedic Surgery, Keck School of Medicine of USCUniversity of Southern California (USC)Los AngelesCaliforniaUSA
| | - Ruzanna Shkhyan
- Department of Orthopaedic Surgery, Keck School of Medicine of USCUniversity of Southern California (USC)Los AngelesCaliforniaUSA
| | - Youngjoo Lee
- Department of Orthopaedic Surgery, Keck School of Medicine of USCUniversity of Southern California (USC)Los AngelesCaliforniaUSA
| | - Jinxiu Lu
- Department of Orthopaedic Surgery, Keck School of Medicine of USCUniversity of Southern California (USC)Los AngelesCaliforniaUSA
| | - Yuxin Ouyang
- Department of Orthopaedic Surgery, Keck School of Medicine of USCUniversity of Southern California (USC)Los AngelesCaliforniaUSA
| | - Hanhan Tang
- Department of Orthopaedic Surgery, Keck School of Medicine of USCUniversity of Southern California (USC)Los AngelesCaliforniaUSA
| | - Fangzhou Bian
- Department of Orthopaedic Surgery, Keck School of Medicine of USCUniversity of Southern California (USC)Los AngelesCaliforniaUSA
| | - Litao Tao
- Department of Biomedical SciencesCreighton UniversityNebraskaOmahaUSA
| | - Neil Segil
- Department of Stem Cell and Regenerative MedicineUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
- Eli and Edythe Broad CenterUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Jason Ernst
- Department of Biological ChemistryUniversity of CaliforniaLos AngelesCaliforniaUSA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research at UCLALos AngelesCaliforniaUSA
- Computer Science DepartmentUniversity of CaliforniaLos AngelesCaliforniaUSA
- Jonsson Comprehensive Cancer Center, University of CaliforniaLos AngelesCaliforniaUSA
- Molecular Biology Institute, University of CaliforniaLos AngelesCaliforniaUSA
- Department of Computational MedicineUniversity of CaliforniaLos AngelesCaliforniaUSA
| | - Karen Lyons
- Department of Orthopaedic SurgeryUniversity of CaliforniaLos AngelesCaliforniaUSA
| | - Steve Horvath
- Department of Biostatistics, Fielding School of Public HealthUniversity of CaliforniaLos AngelesCaliforniaUSA
- Department of Human Genetics, David Geffen School of MedicineUniversity of CaliforniaLos AngelesCaliforniaUSA
| | - Denis Evseenko
- Department of Orthopaedic Surgery, Keck School of Medicine of USCUniversity of Southern California (USC)Los AngelesCaliforniaUSA
- Department of Stem Cell and Regenerative MedicineUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
- Eli and Edythe Broad CenterUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| |
Collapse
|
23
|
The Biomimetics of Mg 2+-Concentration-Resolved Microenvironment for Bone and Cartilage Repairing Materials Design. Biomimetics (Basel) 2022; 7:biomimetics7040227. [PMID: 36546928 PMCID: PMC9775637 DOI: 10.3390/biomimetics7040227] [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: 10/15/2022] [Revised: 11/28/2022] [Accepted: 12/02/2022] [Indexed: 12/12/2022] Open
Abstract
With the increase in population aging, the tendency of osteochondral injury will be accelerated, and repairing materials are increasingly needed for the optimization of the regenerative processes in bone and cartilage recovery. The local environment of the injury sites and the deficiency of Mg2+ retards the repairing period via inhibiting the progenitor osteogenesis and chondrogenesis cells’ recruitment, proliferation, and differentiation, which results in the sluggish progress in the osteochondral repairing materials design. In this article, we elucidate the Mg2+-concentration specified effect on the cell proliferation, osteochondral gene expression, and differentiation of modeling chondrocytes (extracted from New Zealand white rabbit) and osteoblasts (MC3T3-E1). The concentration of Mg2+ in the culture medium affects the proliferation, chondrogenesis, and osteogenesis: (i) Appropriate concentrations of Mg2+ promote the proliferation of chondrocytes (1.25−10.0 mM) and MC3T3-E1 cells (2.5−30.0 mM); (ii) the optimal concentration of Mg2+ that promotes the gene expression of noncalcified cartilage is 15 mM, calcified cartilage 10 mM, and subchondral bone 5 mM, respectively; (iii) overdosed Mg2+ leads to the inhibition of cell activity for either chondrocytes (>20 mM) or osteoblasts (>30 mM). The biomimetic elucidation for orchestrating the allocation of gradient concentration of Mg2+ in accordance of the physiological condition is crucial for designing the accurate microenvironment in osteochondral injury defects for optimization of bone and cartilage repairing materials in the future.
Collapse
|
24
|
O'Connell CD, Duchi S, Onofrillo C, Caballero‐Aguilar LM, Trengove A, Doyle SE, Zywicki WJ, Pirogova E, Di Bella C. Within or Without You? A Perspective Comparing In Situ and Ex Situ Tissue Engineering Strategies for Articular Cartilage Repair. Adv Healthc Mater 2022; 11:e2201305. [PMID: 36541723 PMCID: PMC11468013 DOI: 10.1002/adhm.202201305] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 10/21/2022] [Indexed: 11/23/2022]
Abstract
Human articular cartilage has a poor ability to self-repair, meaning small injuries often lead to osteoarthritis, a painful and debilitating condition which is a major contributor to the global burden of disease. Existing clinical strategies generally do not regenerate hyaline type cartilage, motivating research toward tissue engineering solutions. Prospective cartilage tissue engineering therapies can be placed into two broad categories: i) Ex situ strategies, where cartilage tissue constructs are engineered in the lab prior to implantation and ii) in situ strategies, where cells and/or a bioscaffold are delivered to the defect site to stimulate chondral repair directly. While commonalities exist between these two approaches, the core point of distinction-whether chondrogenesis primarily occurs "within" or "without" (outside) the body-can dictate many aspects of the treatment. This difference influences decisions around cell selection, the biomaterials formulation and the surgical implantation procedure, the processes of tissue integration and maturation, as well as, the prospects for regulatory clearance and clinical translation. Here, ex situ and in situ cartilage engineering strategies are compared: Highlighting their respective challenges, opportunities, and prospects on their translational pathways toward long term human cartilage repair.
Collapse
Affiliation(s)
- Cathal D. O'Connell
- Discipline of Electrical and Biomedical EngineeringRMIT UniversityMelbourneVictoria3000Australia
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
| | - Serena Duchi
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
- Department of SurgerySt Vincent's HospitalUniversity of MelbourneFitzroyVictoria3065Australia
| | - Carmine Onofrillo
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
- Department of SurgerySt Vincent's HospitalUniversity of MelbourneFitzroyVictoria3065Australia
| | - Lilith M. Caballero‐Aguilar
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
- School of ScienceComputing and Engineering TechnologiesSwinburne University of TechnologyMelbourneVictoria3122Australia
| | - Anna Trengove
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
- Department of Biomedical EngineeringUniversity of MelbourneMelbourneVictoria3010Australia
| | - Stephanie E. Doyle
- Discipline of Electrical and Biomedical EngineeringRMIT UniversityMelbourneVictoria3000Australia
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
| | - Wiktor J. Zywicki
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
- Department of Biomedical EngineeringUniversity of MelbourneMelbourneVictoria3010Australia
| | - Elena Pirogova
- Discipline of Electrical and Biomedical EngineeringRMIT UniversityMelbourneVictoria3000Australia
| | - Claudia Di Bella
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
- Department of SurgerySt Vincent's HospitalUniversity of MelbourneFitzroyVictoria3065Australia
- Department of MedicineSt Vincent's Hospital MelbourneFitzroyVictoria3065Australia
| |
Collapse
|
25
|
Kutaish H, Bengtsson L, Tscholl PM, Marteyn A, Braunersreuther V, Guérin A, Béna F, Gimelli S, Longet D, Ilmjärv S, Dietrich PY, Gerstel E, Jaquet V, Hannouche D, Menetrey J, Assal M, Krause KH, Cosset E, Tieng V. Hyaline Cartilage Microtissues Engineered from Adult Dedifferentiated Chondrocytes: Safety and Role of WNT Signaling. Stem Cells Transl Med 2022; 11:1219-1231. [PMID: 36318262 PMCID: PMC9801297 DOI: 10.1093/stcltm/szac074] [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: 03/23/2022] [Accepted: 09/18/2022] [Indexed: 11/05/2022] Open
Abstract
The repair of damaged articular cartilage is an unmet medical need. Chondrocyte-based cell therapy has been used to repair cartilage for over 20 years despite current limitations. Chondrocyte dedifferentiation upon expansion in monolayer is well known and is the main obstacle to their use as cell source for cartilage repair. Consequently, current approaches often lead to fibrocartilage, which is biomechanically different from hyaline cartilage and not effective as a long-lasting treatment. Here, we describe an innovative 3-step method to engineer hyaline-like cartilage microtissues, named Cartibeads, from high passage dedifferentiated chondrocytes. We show that WNT5A/5B/7B genes were highly expressed in dedifferentiated chondrocytes and that a decrease of the WNT signaling pathway was instrumental for full re-differentiation of chondrocytes, enabling production of hyaline matrix instead of fibrocartilage matrix. Cartibeads showed hyaline-like characteristics based on GAG quantity and type II collagen expression independently of donor age and cartilage quality. In vivo, Cartibeads were not tumorigenic when transplanted into SCID mice. This simple 3-step method allowed a standardized production of hyaline-like cartilage microtissues from a small cartilage sample, making Cartibeads a promising candidate for the treatment of cartilage lesions.
Collapse
Affiliation(s)
| | | | - Philippe Matthias Tscholl
- University Medical Center, University of Geneva, Geneva, Switzerland,Department of Orthopaedics Surgery, Geneva University Hospital, Geneva, Switzerland
| | - Antoine Marteyn
- Department of Pathology and Immunology, Medical School, University of Geneva, Geneva, Switzerland,University Medical Center, University of Geneva, Geneva, Switzerland
| | - Vincent Braunersreuther
- Service of Clinical Pathology, Diagnostic Department, Geneva University Hospitals, Geneva, Switzerland
| | - Alexandre Guérin
- Department of Pathology and Immunology, Medical School, University of Geneva, Geneva, Switzerland,University Medical Center, University of Geneva, Geneva, Switzerland
| | - Frédérique Béna
- Service of Genetic Medicine, Diagnostic Department, Geneva University Hospitals, Geneva, Switzerland
| | - Stefania Gimelli
- Service of Genetic Medicine, Diagnostic Department, Geneva University Hospitals, Geneva, Switzerland
| | - David Longet
- Department of Pathology and Immunology, Medical School, University of Geneva, Geneva, Switzerland,University Medical Center, University of Geneva, Geneva, Switzerland
| | - Sten Ilmjärv
- Department of Pathology and Immunology, Medical School, University of Geneva, Geneva, Switzerland,University Medical Center, University of Geneva, Geneva, Switzerland
| | - Pierre-Yves Dietrich
- Laboratory of Tumor Immunology, Oncology Department, Center for Translational Research in Onco-Hematology, Geneva University Hospitals, University of Geneva, Geneva, Switzerland
| | - Eric Gerstel
- University Medical Center, University of Geneva, Geneva, Switzerland,Clinique la Colline, Hirslanden, Geneva, Switzerland
| | - Vincent Jaquet
- Department of Pathology and Immunology, Medical School, University of Geneva, Geneva, Switzerland,University Medical Center, University of Geneva, Geneva, Switzerland,READS Unit, Medical School, University of Geneva, Geneva, Switzerland
| | - Didier Hannouche
- University Medical Center, University of Geneva, Geneva, Switzerland,Department of Orthopaedics Surgery, Geneva University Hospital, Geneva, Switzerland
| | - Jacques Menetrey
- University Medical Center, University of Geneva, Geneva, Switzerland,Centre for Sports Medicine and Exercise, Clinique la Colline, Hirslanden, Geneva, Switzerland
| | - Mathieu Assal
- University Medical Center, University of Geneva, Geneva, Switzerland,Foot and Ankle Surgery Centre, Centre Assal, Clinique La Colline, Hirslanden Geneva, Switzerland
| | - Karl-Heinz Krause
- Department of Pathology and Immunology, Medical School, University of Geneva, Geneva, Switzerland,University Medical Center, University of Geneva, Geneva, Switzerland
| | | | - Vannary Tieng
- Corresponding author: Vannary Tieng, Vanarix SA, Avenue Mon-Repos 14, 1005 Lausanne, Switzerland.
| |
Collapse
|
26
|
Fredrikson JP, Brahmachary PP, Erdoğan AE, Archambault ZK, Wilking JN, June RK, Chang CB. Metabolomic Profiling and Mechanotransduction of Single Chondrocytes Encapsulated in Alginate Microgels. Cells 2022; 11:900. [PMID: 35269522 PMCID: PMC8909502 DOI: 10.3390/cells11050900] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 02/25/2022] [Accepted: 03/02/2022] [Indexed: 02/01/2023] Open
Abstract
Articular cartilage is comprised of two main components, the extracellular matrix (ECM) and the pericellular matrix (PCM). The PCM helps to protect chondrocytes in the cartilage from mechanical loads, but in patients with osteoarthritis, the PCM is weakened, resulting in increased chondrocyte stress. As chondrocytes are responsible for matrix synthesis and maintenance, it is important to understand how mechanical loads affect the cellular responses of chondrocytes. Many studies have examined chondrocyte responses to in vitro mechanical loading by embedding chondrocytes in 3-D hydrogels. However, these experiments are mostly performed in the absence of PCM, which may obscure important responses to mechanotransduction. Here, drop-based microfluidics is used to culture single chondrocytes in alginate microgels for cell-directed PCM synthesis that closely mimics the in vivo microenvironment. Chondrocytes formed PCM over 10 days in these single-cell 3-D microenvironments. Mechanotransduction studies were performed, in which single-cell microgels mimicking the cartilage PCM were embedded in high-stiffness agarose. After physiological dynamic compression in a custom-built bioreactor, microgels exhibited distinct metabolomic profiles from both uncompressed and monolayer controls. These results demonstrate the potential of single cell encapsulation in alginate microgels to advance cartilage tissue engineering and basic chondrocyte mechanobiology.
Collapse
Affiliation(s)
- Jacob P. Fredrikson
- Department of Chemical & Biological Engineering, Montana State University, P.O. Box 173920, Bozeman, MT 59717, USA; (J.P.F.); (A.E.E.); (J.N.W.)
| | - Priyanka P. Brahmachary
- Department of Mechanical & Industrial Engineering, Montana State University, P.O. Box 173800, Bozeman, MT 59717, USA; (P.P.B.); (Z.K.A.)
| | - Ayten E. Erdoğan
- Department of Chemical & Biological Engineering, Montana State University, P.O. Box 173920, Bozeman, MT 59717, USA; (J.P.F.); (A.E.E.); (J.N.W.)
| | - Zachary K. Archambault
- Department of Mechanical & Industrial Engineering, Montana State University, P.O. Box 173800, Bozeman, MT 59717, USA; (P.P.B.); (Z.K.A.)
| | - James N. Wilking
- Department of Chemical & Biological Engineering, Montana State University, P.O. Box 173920, Bozeman, MT 59717, USA; (J.P.F.); (A.E.E.); (J.N.W.)
- Center for Biofilm Engineering, Montana State University, P.O. Box 173980, Bozeman, MT 59717, USA
| | - Ronald K. June
- Department of Mechanical & Industrial Engineering, Montana State University, P.O. Box 173800, Bozeman, MT 59717, USA; (P.P.B.); (Z.K.A.)
- Department of Microbiology & Cell Biology, Montana State University, P.O. Box 173520, Bozeman, MT 59717, USA
| | - Connie B. Chang
- Department of Chemical & Biological Engineering, Montana State University, P.O. Box 173920, Bozeman, MT 59717, USA; (J.P.F.); (A.E.E.); (J.N.W.)
- Center for Biofilm Engineering, Montana State University, P.O. Box 173980, Bozeman, MT 59717, USA
| |
Collapse
|
27
|
Kang D, Lee J, Jung J, Carlson BA, Chang MJ, Chang CB, Kang SB, Lee BC, Gladyshev VN, Hatfield DL, Lee BJ, Kim JH. Selenophosphate synthetase 1 deficiency exacerbates osteoarthritis by dysregulating redox homeostasis. Nat Commun 2022; 13:779. [PMID: 35140209 PMCID: PMC8828855 DOI: 10.1038/s41467-022-28385-7] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 01/24/2022] [Indexed: 12/13/2022] Open
Abstract
Aging and mechanical overload are prominent risk factors for osteoarthritis (OA), which lead to an imbalance in redox homeostasis. The resulting state of oxidative stress drives the pathological transition of chondrocytes during OA development. However, the specific molecular pathways involved in disrupting chondrocyte redox homeostasis remain unclear. Here, we show that selenophosphate synthetase 1 (SEPHS1) expression is downregulated in human and mouse OA cartilage. SEPHS1 downregulation impairs the cellular capacity to synthesize a class of selenoproteins with oxidoreductase functions in chondrocytes, thereby elevating the level of reactive oxygen species (ROS) and facilitating chondrocyte senescence. Cartilage-specific Sephs1 knockout in adult mice causes aging-associated OA, and augments post-traumatic OA, which is rescued by supplementation of N-acetylcysteine (NAC). Selenium-deficient feeding and Sephs1 knockout have synergistic effects in exacerbating OA pathogenesis in mice. Therefore, we propose that SEPHS1 is an essential regulator of selenium metabolism and redox homeostasis, and its dysregulation governs the progression of OA. Osteoarthritis is caused by the gradual accumulation of oxidative stress in cartilage. Here, the authors show that dysregulation of the selenium metabolic pathway underlies a shift in redox homeostasis in chondrocytes, leading to chronic osteoarthritic changes in joints.
Collapse
Affiliation(s)
- Donghyun Kang
- Center for RNA Research, Institute for Basic Science, Seoul, 08826, South Korea.,Department of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul, 08826, South Korea
| | - Jeeyeon Lee
- Center for RNA Research, Institute for Basic Science, Seoul, 08826, South Korea.,Department of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul, 08826, South Korea
| | - Jisu Jung
- Department of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul, 08826, South Korea
| | - Bradley A Carlson
- Mouse Cancer Genetics Program, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Moon Jong Chang
- Department of Orthopaedic Surgery, Seoul National University College of Medicine, Boramae Hospital, Seoul, 07061, South Korea
| | - Chong Bum Chang
- Department of Orthopaedic Surgery, Seoul National University Bundang Hospital, Seongnam, 13620, South Korea
| | - Seung-Baik Kang
- Department of Orthopaedic Surgery, Seoul National University College of Medicine, Boramae Hospital, Seoul, 07061, South Korea
| | - Byung Cheon Lee
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, South Korea
| | - Vadim N Gladyshev
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Dolph L Hatfield
- Mouse Cancer Genetics Program, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Byeong Jae Lee
- Department of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul, 08826, South Korea. .,Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul, 08826, South Korea.
| | - Jin-Hong Kim
- Center for RNA Research, Institute for Basic Science, Seoul, 08826, South Korea. .,Department of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul, 08826, South Korea. .,Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul, 08826, South Korea.
| |
Collapse
|
28
|
Application of Alginate Hydrogels for Next-Generation Articular Cartilage Regeneration. Int J Mol Sci 2022; 23:ijms23031147. [PMID: 35163071 PMCID: PMC8835677 DOI: 10.3390/ijms23031147] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/14/2022] [Accepted: 01/18/2022] [Indexed: 12/28/2022] Open
Abstract
The articular cartilage has insufficient intrinsic healing abilities, and articular cartilage injuries often progress to osteoarthritis. Alginate-based scaffolds are attractive biomaterials for cartilage repair and regeneration, allowing for the delivery of cells and therapeutic drugs and gene sequences. In light of the heterogeneity of findings reporting the benefits of using alginate for cartilage regeneration, a better understanding of alginate-based systems is needed in order to improve the approaches aiming to enhance cartilage regeneration with this compound. This review provides an in-depth evaluation of the literature, focusing on the manipulation of alginate as a tool to support the processes involved in cartilage healing in order to demonstrate how such a material, used as a direct compound or combined with cell and gene therapy and with scaffold-guided gene transfer procedures, may assist cartilage regeneration in an optimal manner for future applications in patients.
Collapse
|
29
|
Molecular Responses to High Hydrostatic Pressure in Eukaryotes: Genetic Insights from Studies on Saccharomyces cerevisiae. BIOLOGY 2021; 10:biology10121305. [PMID: 34943220 PMCID: PMC8698847 DOI: 10.3390/biology10121305] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 11/27/2021] [Accepted: 12/06/2021] [Indexed: 01/19/2023]
Abstract
Simple Summary High hydrostatic pressure generally has an adverse effect on the biological systems of organisms inhabiting lands or shallow sea regions. Deep-sea piezophiles that prefer high hydrostatic pressure for growth have garnered considerable scientific attention. However, the underlying molecular mechanisms of their adaptation to high pressure remains unclear owing to the challenges of culturing and manipulating the genome of piezophiles. Humans also experience high hydrostatic pressure during exercise. A long-term stay in space can cause muscle weakness in astronauts. Thus, the human body indubitably senses mechanical stresses such as hydrostatic pressure and gravity. Nonetheless, the mechanisms underlying biological responses to high pressures are not clearly understood. This review summarizes the occurrence and significance of high-pressure effects in eukaryotic cells and how the cell responds to increasing pressure by particularly focusing on the physiology of S. cerevisiae at the molecular level. Abstract High hydrostatic pressure is common mechanical stress in nature and is also experienced by the human body. Organisms in the Challenger Deep of the Mariana Trench are habitually exposed to pressures up to 110 MPa. Human joints are intermittently exposed to hydrostatic pressures of 3–10 MPa. Pressures less than 50 MPa do not deform or kill the cells. However, high pressure can have various effects on the cell’s biological processes. Although Saccharomyces cerevisiae is not a deep-sea piezophile, it can be used to elucidate the molecular mechanism underlying the cell’s responses to high pressures by applying basic knowledge of the effects of pressure on industrial processes involving microorganisms. We have explored the genes associated with the growth of S. cerevisiae under high pressure by employing functional genomic strategies and transcriptomics analysis and indicated a strong association between high-pressure signaling and the cell’s response to nutrient availability. This review summarizes the occurrence and significance of high-pressure effects on complex metabolic and genetic networks in eukaryotic cells and how the cell responds to increasing pressure by particularly focusing on the physiology of S. cerevisiae at the molecular level. Mechanosensation in humans has also been discussed.
Collapse
|
30
|
Yan J, Liu C, Tu C, Zhang R, Tang X, Li H, Wang H, Ma Y, Zhang Y, Wu H, Sheng G. Hydrogel-hydroxyapatite-monomeric collagen type-I scaffold with low-frequency electromagnetic field treatment enhances osteochondral repair in rabbits. Stem Cell Res Ther 2021; 12:572. [PMID: 34774092 PMCID: PMC8590294 DOI: 10.1186/s13287-021-02638-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 10/25/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Cartilage damage is a common medical issue in clinical practice. Complete cartilage repair remains a significant challenge owing to the inferior quality of regenerative tissue. Safe and non-invasive magnetic therapy combined with tissue engineering to repair cartilage may be a promising breakthrough. METHODS In this study, a composite scaffold made of Hydroxyapatite-Collagen type-I (HAC) and PLGA-PEG-PLGA thermogel was produced to match the cartilage and subchondral layers in osteochondral defects, respectively. Bone marrow mesenchymal stem cells (BMSC) encapsulated in the thermogel were stimulated by an electromagnetic field (EMF). Effect of EMF on the proliferation and chondrogenic differentiation potential was evaluated in vitro. 4 mm femoral condyle defect was constructed in rabbits. The scaffolds loaded with BMSCs were implanted into the defects with or without EMF treatment. Effects of the combination treatment of the EMF and composite scaffold on rabbit osteochondral defect was detected in vivo. RESULTS In vitro experiments showed that EMF could promote proliferation and chondrogenic differentiation of BMSCs partly by activating the PI3K/AKT/mTOR and Wnt1/LRP6/β-catenin signaling pathway. In vivo results further confirmed that the scaffold with EMF enhances the repair of osteochondral defects in rabbits, and, in particular, cartilage repair. CONCLUSION Hydrogel-Hydroxyapatite-Monomeric Collagen type-I scaffold with low-frequency EMF treatment has the potential to enhance osteochondral repair.
Collapse
Affiliation(s)
- Jiyuan Yan
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue 1095, Wuhan, 430030, Hubei, People's Republic of China
| | - Chaoxu Liu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue 1095, Wuhan, 430030, Hubei, People's Republic of China
| | - Chang Tu
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China
| | - Ruizhuo Zhang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue 1095, Wuhan, 430030, Hubei, People's Republic of China
| | - Xiangyu Tang
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Hao Li
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue 1095, Wuhan, 430030, Hubei, People's Republic of China
| | - Huaixi Wang
- Department of Spine and Spinal Cord Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Henan, Zhengzhou, People's Republic of China
| | - Yongzhuang Ma
- Department of Orthopedics, Shanxi Bethune Hospital, Taiyuan, Shanxi, People's Republic of China
| | - Yingchi Zhang
- Department of Traumatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue 1095, Wuhan, 430030, Hubei, People's Republic of China.
| | - Hua Wu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue 1095, Wuhan, 430030, Hubei, People's Republic of China.
| | - Gaohong Sheng
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue 1095, Wuhan, 430030, Hubei, People's Republic of China.
| |
Collapse
|
31
|
Ravichandran D, Pillai J, Krishnamurthy K. Genetics of intervertebral disc disease: A review. Clin Anat 2021; 35:116-120. [PMID: 34689354 DOI: 10.1002/ca.23803] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 09/23/2021] [Accepted: 10/14/2021] [Indexed: 01/08/2023]
Abstract
Intervertebral disc disease (IVDD) is a common musculoskeletal disease affecting about 5% of all individuals. It is characterized by lumbar disc herniation, which causes nerve root irritation, either mechanically or via inflammatory mediators, and results in radiating pain, known as sciatica. Numerous studies have been conducted to identify the causes and risk factors for IVDD. Lifting heavy loads, torsional stress, and motor vehicle driving are among the best-identified environmental risk factors. However, it has become evident recently from family and twin studies that genetic factors may also be important in IVDD. This hypothesis was strengthened by the identification of two collagen IX alleles associated with sciatica and lumbar disc herniation. In addition, disc degeneration has been shown to be related to an aggrecan gene polymorphism, a Vitamin D receptor and matrix metalloproteinase-3 gene alleles. This review highlights the genetic role and occupational aspects of IVDD.
Collapse
|
32
|
Khan B, Kafian-Attari I, Nippolainen E, Shaikh R, Semenov D, Hauta-Kasari M, Töyräs J, Afara IO. Articular cartilage optical properties in the near-infrared (NIR) spectral range vary with depth and tissue integrity. BIOMEDICAL OPTICS EXPRESS 2021; 12:6066-6080. [PMID: 34745722 PMCID: PMC8548021 DOI: 10.1364/boe.430053] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 07/27/2021] [Accepted: 08/09/2021] [Indexed: 05/02/2023]
Abstract
Optical properties of biological tissues in the NIR spectral range have demonstrated significant potential for in vivo diagnostic applications and are critical parameters for modelling light interaction in biological tissues. This study aims to investigate the optical properties of articular cartilage as a function of tissue depth and integrity. The results suggest consistent wavelength-dependent variation in optical properties between cartilage depth-wise zones, as well as between healthy and degenerated tissue. Also, statistically significant differences (p<0.05) in both optical properties were observed between the different cartilage depth-wise zones and as a result of tissue degeneration. When taken into account, the outcome of this study could enable accurate modelling of light interaction in cartilage matrix and could provide useful diagnostic information on cartilage integrity.
Collapse
Affiliation(s)
- Bilour Khan
- University of Eastern Finland, Department of Applied Physics, Yliopistonranta 1, Kuopio, Finland, 70120, Finland
| | - Iman Kafian-Attari
- University of Eastern Finland, Department of Applied Physics, Yliopistonranta 1, Kuopio, Finland, 70120, Finland
| | - Ervin Nippolainen
- University of Eastern Finland, Department of Applied Physics, Yliopistonranta 1, Kuopio, Finland, 70120, Finland
| | - Rubina Shaikh
- University of Eastern Finland, Department of Applied Physics, Yliopistonranta 1, Kuopio, Finland, 70120, Finland
| | - Dmitry Semenov
- University of Eastern Finland, School of Computing, Lämsikatu 15, Joensuu, Finland, 80110, Finland
| | - Markku Hauta-Kasari
- University of Eastern Finland, School of Computing, Lämsikatu 15, Joensuu, Finland, 80110, Finland
| | - Juha Töyräs
- University of Eastern Finland, Department of Applied Physics, Yliopistonranta 1, Kuopio, Finland, 70120, Finland
- The University of Queensland, School of Information Technology, and Electrical Engineering, St. Lucia, Australia, QLD 4072, Australia
| | - Isaac O. Afara
- University of Eastern Finland, Department of Applied Physics, Yliopistonranta 1, Kuopio, Finland, 70120, Finland
- The University of Queensland, School of Information Technology, and Electrical Engineering, St. Lucia, Australia, QLD 4072, Australia
| |
Collapse
|
33
|
Deloney M, Garoosi P, Dartora VFC, Christiansen BA, Panitch A. Hyaluronic Acid-Binding, Anionic, Nanoparticles Inhibit ECM Degradation and Restore Compressive Stiffness in Aggrecan-Depleted Articular Cartilage Explants. Pharmaceutics 2021; 13:1503. [PMID: 34575579 PMCID: PMC8469381 DOI: 10.3390/pharmaceutics13091503] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 09/10/2021] [Accepted: 09/11/2021] [Indexed: 12/03/2022] Open
Abstract
Joint trauma results in the production of inflammatory cytokines that stimulate the secretion of catabolic enzymes, which degrade articular cartilage. Molecular fragments of the degraded articular cartilage further stimulate inflammatory cytokine production, with this process eventually resulting in post-traumatic osteoarthritis (PTOA). The loss of matrix component aggrecan occurs early in the progression of PTOA and results in the loss of compressive stiffness in articular cartilage. Aggrecan is highly sulfated, associates with hyaluronic acid (HA), and supports the compressive stiffness in cartilage. Presented here, we conjugated the HA-binding peptide GAHWQFNALTVRGSG (GAH) to anionic nanoparticles (hNPs). Nanoparticles conjugated with roughly 19 GAH peptides, termed 19 GAH-hNP, bound to HA in solution and increased the dynamic viscosity by 94.1% compared to an HA solution treated with unconjugated hNPs. Moreover, treating aggrecan-depleted (AD) cartilage explants with 0.10 mg of 19 GAH-hNP restored the cartilage compressive stiffness to healthy levels six days after a single nanoparticle treatment. Treatment of AD cartilage with 0.10 mg of 19 GAH-hNP inhibited the degradation of articular cartilage. Treated AD cartilage had 409% more collagen type II and 598% more GAG content than untreated-AD explants. The 19 GAH-hNP therapeutic slowed ECM degradation in AD cartilage explants, restored the compressive stiffness of damaged cartilage, and showed promise as a localized treatment for PTOA.
Collapse
Affiliation(s)
- Marcus Deloney
- Biomedical Engineering Department, 451 E. Health Sciences Dr. Room 2303, University of California Davis, Davis, CA 95616, USA; (M.D.); (P.G.); (V.F.C.D.)
| | - Parssa Garoosi
- Biomedical Engineering Department, 451 E. Health Sciences Dr. Room 2303, University of California Davis, Davis, CA 95616, USA; (M.D.); (P.G.); (V.F.C.D.)
| | - Vanessa F. C. Dartora
- Biomedical Engineering Department, 451 E. Health Sciences Dr. Room 2303, University of California Davis, Davis, CA 95616, USA; (M.D.); (P.G.); (V.F.C.D.)
- Department of Surgery, School of Medicine, University of California Davis, Sacramento, CA 95817, USA
| | - Blaine A. Christiansen
- Lawrence J. Ellison Musculoskeletal Research Center, Department of Orthopedic Surgery, University of California Davis Health, 4635 2nd Avenue, Suite 2000, Sacramento, CA 95817, USA;
| | - Alyssa Panitch
- Biomedical Engineering Department, 451 E. Health Sciences Dr. Room 2303, University of California Davis, Davis, CA 95616, USA; (M.D.); (P.G.); (V.F.C.D.)
- Department of Surgery, School of Medicine, University of California Davis, Sacramento, CA 95817, USA
| |
Collapse
|
34
|
The Influence of Running on Lower Limb Cartilage: A Systematic Review and Meta-analysis. Sports Med 2021; 52:55-74. [PMID: 34478109 DOI: 10.1007/s40279-021-01533-7] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/01/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND Running is a popular activity practiced worldwide. It is important to understand how running affects joint health to provide recommendations to sports medicine practitioners and runners. OBJECTIVE Our aim was to summarize the influence of running on lower limb cartilage morphology and composition using quantitative magnetic resonance imaging (MRI). METHODS Prospective repeated-measures studies evaluating cartilage using MRI before and after running were included. Data sources included Pubmed, Embase, CINAHL, SportDiscus, Web of Science, and Cochrane Central Registry of Controlled Trials. Qualitative analyses considered the number and methodological quality ratings of studies based on the QualSyst tool, and recommendations were based on the strength of evidence (strong, moderate, limited, or very limited). Quantitative analysis involved meta-analyses, for which effect sizes were calculated as Hedge's g standardized mean differences. RESULTS We included 43 articles, assessing seven outcomes (lesions, volume, thickness, glycosaminoglycan content, and T1ρ, T2, and T2* relaxation times). Nineteen articles were rated as high quality, 24 were rated as moderate quality, and none were rated as low quality. Qualitative analyses suggest that running may cause an immediate reduction in knee cartilage volume, thickness, as well as T1ρ and T2 relaxation times immediately; however, these changes did not persist. Meta-analyses revealed a small and moderate decrease immediately following a single running bout in T2 relaxation time in the medial femur and tibia, respectively. Qualitative analyses indicated that the influence of repeated exposure to running on cartilage morphology and composition was limited. Despite conflicting evidence regarding pre-existing knee cartilage lesions, moderate evidence suggests that running does not lead to the formation of new lesions. Repeated running exposure did not cause changes to foot and ankle cartilage thickness or composition. CONCLUSIONS Changes to lower limb cartilage following running are transient. Immediate changes to cartilage morphology and composition, which likely reflect natural fluid dynamics, do not persist and were generally not significant when pooled statistically. Results suggest that cartilage recovers well from a single running bout and adapts to repeated exposure. Given that moderate evidence indicates that running does not lead to new lesions, future trials should focus on clinical populations, such as those with osteoarthritis. TRIAL REGISTRATION Not applicable.
Collapse
|
35
|
Gherghel R, Iordan DA, Mocanu MD, Onu A, Onu I. Osteoarthritis is not a disease, but rather an accumulation of predisposing factors. A systematic review. BALNEO AND PRM RESEARCH JOURNAL 2021. [DOI: 10.12680/balneo.2021.441] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Introduction: Although they do not endanger the life of the individual, the major symptoms of osteoarthritis (OA), such as pain, inflammation and dysfunction, it will slowly decrease quality of life and performance, leading finally to disabilities. Due to the fact that this disease has no cure, strategies are still being sought to slow its evolution. The lack of understanding of the predisposing and triggering factors of OA, has led to different approaches to this pathology so discussed, but with modest results. This systematic review aims to debate the main phenomena underlying joint destruction in OA, and etiopathogenic theories.
Materials and Methods: In this study were included 58 bibliographic sources, of which title 39 refers to OA, 6 with inflammation, 28 with cartilage, 3 with chondrocytes, and 5 with synovitis. In this study were discussed the etiopathogenic theories of OA which include: age, alteration of the cartilaginous matrix, alteration of chondrocyte metabolism, microtrauma and major trauma, inflammation of the joints - synovitis and obesity.
Results: Increasing the level of understanding of predisposing factors, the occurrence of acute inflammatory phenomenon and the perpetuation of mechanisms that latently maintain chronic inflammation that over time develops a destructive effect on articular cartilage, would limit the negative effects of OA, delay the evolution and optimally combat that maintain the vicious circle: inflammation → production of enzymes → chondrolysis → inflammation.
Conclusions: These studies contribute significantly to the understanding of destructive phenomena in OA. More studies are needed on the risk factors of OA and its production mechanisms, to find increasingly effective therapies that limiting its progression.
Keywords: Osteoarthritis, etiopathogenic theories, chronic inflammation, age, obesity, cartilaginous matrixm, chondrocyte metabolism
Collapse
Affiliation(s)
- Robert Gherghel
- Department of Biomedical Sciences, Faculty of Medical Bioengineering, “Grigore T. Popa” University of Medicine and Pharmacy Iasi, România 2. Departments of orthopedics and physiotherapy Micromedica Medical Clinic, Piatra Neamț
| | - Daniel-Andrei Iordan
- “Dunărea de Jos” University, Faculty of Physical Education and Sports, Galați, România
| | - Mircea-Dan Mocanu
- “Dunărea de Jos” University, Faculty of Physical Education and Sports, Galați, România
| | - Ana Onu
- Departments of orthopedics and physiotherapy Micromedica Medical Clinic, Piatra Neamț
| | - Ilie Onu
- Department of Biomedical Sciences, Faculty of Medical Bioengineering, “Grigore T. Popa” University of Medicine and Pharmacy Iasi, România 2. Departments of orthopedics and physiotherapy Micromedica Medical Clinic, Piatra Neamț
| |
Collapse
|
36
|
Ge Y, Li Y, Wang Z, Li L, Teng H, Jiang Q. Effects of Mechanical Compression on Chondrogenesis of Human Synovium-Derived Mesenchymal Stem Cells in Agarose Hydrogel. Front Bioeng Biotechnol 2021; 9:697281. [PMID: 34350163 PMCID: PMC8327094 DOI: 10.3389/fbioe.2021.697281] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 06/22/2021] [Indexed: 01/22/2023] Open
Abstract
Mechanical compression is a double-edged sword for cartilage remodeling, and the effect of mechanical compression on chondrogenic differentiation still remains elusive to date. Herein, we investigate the effect of mechanical dynamic compression on the chondrogenic differentiation of human synovium-derived mesenchymal stem cells (SMSCs). To this aim, SMSCs encapsulated in agarose hydrogels were cultured in chondrogenic-induced medium with or without dynamic compression. Dynamic compression was applied at either early time-point (day 1) or late time-point (day 21) during chondrogenic induction period. We found that dynamic compression initiated at early time-point downregulated the expression level of chondrocyte-specific markers as well as hypertrophy-specific markers compared with unloaded control. On the contrary, dynamic compression applied at late time-point not only enhanced the levels of cartilage matrix gene expression, but also suppressed the hypertrophic development of SMSCs compared with unloaded controls. Taken together, our findings suggest that dynamic mechanical compression loading not only promotes chondrogenic differentiation of SMSCs, but also plays a vital role in the maintenance of cartilage phenotype, and our findings also provide an experimental guide for stem cell-based cartilage repair and regeneration.
Collapse
Affiliation(s)
- Yuxiang Ge
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China.,Laboratory for Bone and Joint Disease, Model Animal Research Center, Nanjing University, Nanjing, China
| | - Yixuan Li
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China.,Laboratory for Bone and Joint Disease, Model Animal Research Center, Nanjing University, Nanjing, China
| | - Zixu Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China.,Laboratory for Bone and Joint Disease, Model Animal Research Center, Nanjing University, Nanjing, China
| | - Lan Li
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China.,Jiangsu Engineering Research Center for 3D Bioprinting, Nanjing, China
| | - Huajian Teng
- Laboratory for Bone and Joint Disease, Model Animal Research Center, Nanjing University, Nanjing, China
| | - Qing Jiang
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China.,Laboratory for Bone and Joint Disease, Model Animal Research Center, Nanjing University, Nanjing, China
| |
Collapse
|
37
|
Miller RP, Berlouis ME, Hall AG, Simpson AHR, Smith IDM, Hall AC. Effects of Antibiotics on α-Toxin Levels during Staphylococcus aureus Culture: Implications for the Protection of Chondrocytes in a Model of Septic Arthritis. Cartilage 2021; 12:362-376. [PMID: 30762428 PMCID: PMC8236659 DOI: 10.1177/1947603519828433] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
OBJECTIVE Septic arthritis results from joint infection by Staphylococcus aureus, which produces potent α-toxin causing cell death, potentially leading to permanent cartilage damage. Treatment is by joint irrigation and antibiotics, although it is unclear if, following treatment with antibiotics which cause bacterial lysis, there is release of additional stored α-toxin. DESIGN A rabbit erythrocyte hemolysis assay was optimised to assess biologically-active α-toxin from cultured S. aureus α-toxin strain DU5946. Hemoglobin release was measured spectrophotometrically following addition of a bacteriostatic antibiotic (linezolid) or a bacteriolytic antibiotic (penicillin). A bovine cartilage model of septic arthritis was used to test the protective effects of antibiotics against S. aureus infection. RESULTS During S. aureus culture, α-toxin levels increased rapidly but the rate of rise was quickly (within 20 minutes) suppressed by linezolid (25 μg/mL). Penicillin also reduced the increase in α-toxin levels; however, the time course was relatively slow compared to linezolid even at high concentrations (50,000 U/mL). The efficacy of penicillin (250,000 U/mL) at reducing the rise in α-toxin was approximately 8% less than that of linezolid (P < 0.05) suggesting the presence of additional toxin. This could be due to a delayed action of penicillin, and/or release of a small pool of stored α-toxin from dying bacteria. In a bovine cartilage model, however, there was no difference between the protection of in situ chondrocytes against S. aureus by penicillin or linezolid (P > 0.05). CONCLUSION The results suggested that equally effective protection of chondrocytes against S. aureus septic arthritis may be obtained by the bacteriostatic or bacteriolytic antibiotics tested.
Collapse
Affiliation(s)
- Robbie P. Miller
- Centre for Integrative Physiology, Deanery of Biomedical Sciences, University of Edinburgh, Edinburgh, Scotland, UK
| | - Marie E. Berlouis
- Centre for Integrative Physiology, Deanery of Biomedical Sciences, University of Edinburgh, Edinburgh, Scotland, UK
| | - Alan G. Hall
- Centre for Integrative Physiology, Deanery of Biomedical Sciences, University of Edinburgh, Edinburgh, Scotland, UK
| | - A. Hamish R.W. Simpson
- Musculoskeletal Research Unit, Department of Orthopaedic Surgery, University of Edinburgh, Edinburgh, Scotland, UK
| | - Innes D. M. Smith
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, Scotland, UK
| | - Andrew C. Hall
- Centre for Integrative Physiology, Deanery of Biomedical Sciences, University of Edinburgh, Edinburgh, Scotland, UK,Andrew C. Hall, Centre for Integrative Physiology, Deanery of Biomedical Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, EH8 9AG, Scotland, UK
| |
Collapse
|
38
|
Wu X, Liu Y, Du J, Li X, Lin J, Ni L, Zhu P, Zhou H, Kong F, Yang H, Geng D, Mao H. Melatonin Attenuates Intervertebral Disk Degeneration via Maintaining Cartilaginous Endplate Integrity in Rats. Front Physiol 2021; 12:672572. [PMID: 34220535 PMCID: PMC8248798 DOI: 10.3389/fphys.2021.672572] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 05/10/2021] [Indexed: 01/08/2023] Open
Abstract
Objective The aim of this study is to verify whether melatonin (Mel) could mitigate intervertebral disk degeneration (IVDD) in rats and to investigate the potential mechanism of it. Method A rat acupuncture model of IVDD was established with intraperitoneal injection of Mel. The effect of Mel on IVDD was analyzed via radiologic and histological evaluations. The specific Mel receptors were investigated in both the nucleus pulposus (NP) and cartilaginous endplates (EPs). In vitro, EP cartilaginous cells (EPCs) were treated by different concentrations of Mel under lipopolysaccharide (LPS) and Luzindole conditions. In addition, LPS-induced inflammatory response and matrix degradation following nuclear factor kappa-B (NF-κB) pathway activation were investigated to confirm the potential mechanism of Mel on EPCs. Results The percent disk height index (%DHI) and MRI signal decreased after initial puncture in the degeneration group compared with the control group, while Mel treatment protected disk height from decline and prevented the loss of water during the degeneration process. In the meantime, the histological staining of the Mel groups showed more integrity and well-ordered construction of the NP and EPs in both low and high concentration than that of the degeneration group. In addition, more deep-brown staining of type II collagen (Coll-II) was shown in the Mel groups compared with the degeneration group. Furthermore, in rat samples, immunohistochemical staining showed more positive cells of Mel receptors 1a and 1b in the EPs, instead of in the NP. Moreover, evident osteochondral lacuna formation was observed in rat EPs in the degeneration group; after Mel treatment, the osteochondral destruction alleviated accompanying fewer receptor activator for nuclear factor-κB ligand (RANKL) and tartrate-resistant acid phosphatase (TRAP)-stained positive cells expressed in the EPs. In vitro, Mel could promote the proliferation of EPCs, which protected EPCs from degeneration under LPS treatment. What is more, Mel downregulated the inflammatory response and matrix degradation of EPCs activated by NF-κB pathway through binding to its specific receptors. Conclusion These results indicate that Mel protects the integrity of the EPs and attenuates IVDD by binding to the Mel receptors in the EPs. It may alleviate the inflammatory response and matrix degradation of EPCs activated by NF-κB pathway.
Collapse
Affiliation(s)
- Xiexing Wu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yijie Liu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jiacheng Du
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xiaoping Li
- Department of Clinical Education, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jiayi Lin
- Department of Orthopedics Center, Ningbo No. 2 Hospital, Ningbo, China
| | - Li Ni
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Pengfei Zhu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Hong Zhou
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Fanchen Kong
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Huilin Yang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Dechun Geng
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Haiqing Mao
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
39
|
Wu SC, Chang CH, Chang LH, Wu CW, Chen JW, Chen CH, Lin YS, Chang JK, Ho ML. Simvastatin Enhances the Chondrogenesis But Not the Osteogenesis of Adipose-Derived Stem Cells in a Hyaluronan Microenvironment. Biomedicines 2021; 9:biomedicines9050559. [PMID: 34067739 PMCID: PMC8156330 DOI: 10.3390/biomedicines9050559] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 05/11/2021] [Accepted: 05/14/2021] [Indexed: 11/16/2022] Open
Abstract
Directing adipose-derived stem cells (ADSCs) toward chondrogenesis is critical for ADSC-based articular cartilage regeneration. Simvastatin (SIM) was reported to promote both chondrogenic and osteogenic differentiation of ADSCs by upregulating bone morphogenetic protein-2 (BMP-2). We previously found that ADSC chondrogenesis is initiated and promoted in a hyaluronan (HA) microenvironment (HAM). Here, we further hypothesized that SIM augments HAM-induced chondrogenesis but not osteogenesis of ADSCs. ADSCs were treated with SIM in a HAM (SIM plus HAM) by HA-coated wells or HA-enriched fibrin (HA/Fibrin) hydrogel, and chondrogenic differentiation of ADSCs was evaluated. SIM plus HAM increased chondrogenesis more than HAM or SIM alone, including cell aggregation, chondrogenic gene expression (collagen type II and aggrecan) and cartilaginous tissue formation (collagen type II and sulfated glycosaminoglycan). In contrast, SIM-induced osteogenesis in ADSCs was reduced in SIM plus HAM, including mRNA expression of osteogenic genes, osteocalcin and alkaline phosphatase (ALP), ALP activity and mineralization. SIM plus HAM also showed the most effective increases in the mRNA expression of BMP-2 and transcription factors of SOX-9 and RUNX-2 in ADSCs, while these effects were reversed by CD44 blockade. HAM suppressed the levels of JNK, p-JNK, P38 and p-P38 in ADSCs, and SIM plus HAM also decreased SIM-induced phosphorylated JNK and p38 levels. In addition, SIM enhanced articular cartilage regeneration, as demonstrated by implantation of an ADSCs/HA/Fibrin construct in an ex vivo porcine articular chondral defect model. The results from this study indicate that SIM may be an enhancer of HAM-initiated MSC-based chondrogenesis and avoid osteogenesis.
Collapse
Affiliation(s)
- Shun-Cheng Wu
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan; (S.-C.W.); (L.-H.C.); (C.-W.W.); (J.-W.C.); (C.-H.C.); (Y.-S.L.)
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan;
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Post-Baccalaureate Program in Nursing, Asia University, Taichung 41354, Taiwan
| | - Chih-Hsiang Chang
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan;
| | - Ling-Hua Chang
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan; (S.-C.W.); (L.-H.C.); (C.-W.W.); (J.-W.C.); (C.-H.C.); (Y.-S.L.)
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan;
| | - Che-Wei Wu
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan; (S.-C.W.); (L.-H.C.); (C.-W.W.); (J.-W.C.); (C.-H.C.); (Y.-S.L.)
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan;
| | - Jhen-Wei Chen
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan; (S.-C.W.); (L.-H.C.); (C.-W.W.); (J.-W.C.); (C.-H.C.); (Y.-S.L.)
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan;
| | - Chung-Hwan Chen
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan; (S.-C.W.); (L.-H.C.); (C.-W.W.); (J.-W.C.); (C.-H.C.); (Y.-S.L.)
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan;
- Department of Orthopaedics, College of Medicine, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Division of Adult Reconstruction Surgery, Department of Orthopedics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Department of Orthopedics, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
| | - Yi-Shan Lin
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan; (S.-C.W.); (L.-H.C.); (C.-W.W.); (J.-W.C.); (C.-H.C.); (Y.-S.L.)
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan;
| | - Je-Ken Chang
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan; (S.-C.W.); (L.-H.C.); (C.-W.W.); (J.-W.C.); (C.-H.C.); (Y.-S.L.)
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan;
- Department of Orthopaedics, College of Medicine, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Correspondence: (J.-K.C.); (M.-L.H.)
| | - Mei-Ling Ho
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan; (S.-C.W.); (L.-H.C.); (C.-W.W.); (J.-W.C.); (C.-H.C.); (Y.-S.L.)
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan;
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Department of Physiology, College of Medicine, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 80424, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Correspondence: (J.-K.C.); (M.-L.H.)
| |
Collapse
|
40
|
Fan X, Wu X, Crawford R, Xiao Y, Prasadam I. Macro, Micro, and Molecular. Changes of the Osteochondral Interface in Osteoarthritis Development. Front Cell Dev Biol 2021; 9:659654. [PMID: 34041240 PMCID: PMC8142862 DOI: 10.3389/fcell.2021.659654] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 04/12/2021] [Indexed: 01/05/2023] Open
Abstract
Osteoarthritis (OA) is a long-term condition that causes joint pain and reduced movement. Notably, the same pathways governing cell growth, death, and differentiation during the growth and development of the body are also common drivers of OA. The osteochondral interface is a vital structure located between hyaline cartilage and subchondral bone. It plays a critical role in maintaining the physical and biological function, conveying joint mechanical stress, maintaining chondral microenvironment, as well as crosstalk and substance exchange through the osteochondral unit. In this review, we summarized the progress in research concerning the area of osteochondral junction, including its pathophysiological changes, molecular interactions, and signaling pathways that are related to the ultrastructure change. Multiple potential treatment options were also discussed in this review. A thorough understanding of these biological changes and molecular mechanisms in the pathologic process will advance our understanding of OA progression, and inform the development of effective therapeutics targeting OA.
Collapse
Affiliation(s)
- Xiwei Fan
- Faculty of Science and Engineering, School of Mechanical, Medical and Process Engineering, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD, Australia
| | - Xiaoxin Wu
- Faculty of Science and Engineering, School of Mechanical, Medical and Process Engineering, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD, Australia
| | - Ross Crawford
- Faculty of Science and Engineering, School of Mechanical, Medical and Process Engineering, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD, Australia
- Orthopaedic Department, The Prince Charles Hospital, Brisbane, QLD, Australia
| | - Yin Xiao
- Faculty of Science and Engineering, School of Mechanical, Medical and Process Engineering, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD, Australia
- Australia-China Centre for Tissue Engineering and Regenerative Medicine, Queensland University of Technology, Brisbane, QLD, Australia
| | - Indira Prasadam
- Faculty of Science and Engineering, School of Mechanical, Medical and Process Engineering, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD, Australia
| |
Collapse
|
41
|
Smith K, Mercuri J. Microgravity and Radiation Effects on Astronaut Intervertebral Disc Health. Aerosp Med Hum Perform 2021; 92:342-352. [PMID: 33875067 DOI: 10.3357/amhp.5713.2021] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
INTRODUCTION: The effects of spaceflight on the intervertebral disc (IVD) have not been thoroughly studied, despite the knowledge that spaceflight increases the risk of herniation of IVDs in astronauts upon return to Earth. However, as long duration missions become more common, fully characterizing the mechanisms behind space-induced IVD degeneration becomes increasingly imperative for mission success. This review therefore surveys current literature to outline the results of human, animal, and cell-level studies investigating the effect of microgravity and radiation exposure on IVD health. Overall, recurring study findings include increases in IVD height in microgravity conditions, upregulation of catabolic proteases leading to a weakening extracellular matrix (ECM), and both nucleus pulposus (NP) swelling and loss of annulus fibrosus (AF) fiber alignment which are hypothesized to contribute to the increased risk of herniation when reloading is experienced. However, the limitations of current studies are also discussed. For example, human studies do not allow for invasive measures of the underpinning biochemical mechanisms, correlating animal model results to the human condition may be difficult, and cellular studies lack incorporation of ECM and other complexities that mimic the native IVD microarchitecture and environment. Moving forward, the use of three-dimensional organoid culture models that incorporate IVD-specific human cells, ECM, and signals as well as the development of cell- and ECM-level computational models may further improve our understanding of the impacts that spaceflight has on astronaut IVD health.Smith K, Mercuri J. Microgravity and radiation effects on astronaut intervertebral disc health. Aerosp Med Hum Perform. 2021; 92(5):342352.
Collapse
|
42
|
Lin W, Klein J. Recent Progress in Cartilage Lubrication. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2005513. [PMID: 33759245 DOI: 10.1002/adma.202005513] [Citation(s) in RCA: 200] [Impact Index Per Article: 50.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 11/23/2020] [Indexed: 05/18/2023]
Abstract
Healthy articular cartilage, covering the ends of bones in major joints such as hips and knees, presents the most efficiently-lubricated surface known in nature, with friction coefficients as low as 0.001 up to physiologically high pressures. Such low friction is indeed essential for its well-being. It minimizes wear-and-tear and hence the cartilage degradation associated with osteoarthritis, the most common joint disease, and, by reducing shear stress on the mechanotransductive, cartilage-embedded chondrocytes (the only cell type in the cartilage), it regulates their function to maintain homeostasis. Understanding the origins of such low friction of the articular cartilage, therefore, is of major importance in order to alleviate disease symptoms, and slow or even reverse its breakdown. This progress report considers the relation between frictional behavior and the cellular mechanical environment in the cartilage, then reviews the mechanism of lubrication in the joints, in particular focusing on boundary lubrication. Following recent advances based on hydration lubrication, a proposed synergy between different molecular components of the synovial joints, acting together in enabling the low friction, has been proposed. Additionally, recent development of natural and bio-inspired lubricants is reviewed.
Collapse
Affiliation(s)
- Weifeng Lin
- Department of Materials and Interfaces, Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Jacob Klein
- Department of Materials and Interfaces, Weizmann Institute of Science, Rehovot, 76100, Israel
| |
Collapse
|
43
|
The Biomechanics of Cartilage-An Overview. Life (Basel) 2021; 11:life11040302. [PMID: 33915881 PMCID: PMC8065530 DOI: 10.3390/life11040302] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 03/21/2021] [Accepted: 03/26/2021] [Indexed: 12/15/2022] Open
Abstract
Articular cartilage (AC) sheathes joint surfaces and minimizes friction in diarthrosis. The resident cell population, chondrocytes, are surrounded by an extracellular matrix and a multitude of proteins, which bestow their unique characteristics. AC is characterized by a zonal composition (superficial (tangential) zone, middle (transitional) zone, deep zone, calcified zone) with different mechanical properties. An overview is given about different testing (load tests) methods as well as different modeling approaches. The widely accepted biomechanical test methods, e.g., the indentation analysis, are summarized and discussed. A description of the biphasic theory is also shown. This is required to understand how interstitial water contributes toward the viscoelastic behavior of AC. Furthermore, a short introduction to a more complex model is given.
Collapse
|
44
|
Amann E, Amirall A, Franco AR, Poh PSP, Sola Dueñas FJ, Fuentes Estévez G, Leonor IB, Reis RL, van Griensven M, Balmayor ER. A Graded, Porous Composite of Natural Biopolymers and Octacalcium Phosphate Guides Osteochondral Differentiation of Stem Cells. Adv Healthc Mater 2021; 10:e2001692. [PMID: 33448144 PMCID: PMC11468142 DOI: 10.1002/adhm.202001692] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 11/19/2020] [Indexed: 01/08/2023]
Abstract
Lesions involving the osteochondral unit are difficult to treat. Biomimetic scaffolds are previously shown as promising alternative. Such devices often lack multiple functional layers that mimic bone, cartilage, and the interface. In this study, multilayered scaffolds are developed based on the use of natural extracellular matrix (ECM)-like biopolymers. Particular attention is paid to obtain a complex matrix that mimics the native osteochondral transition. Porous, sponge-like chitosan-collagen-octacalcium phosphate (OCP) scaffolds are obtained. Collagen content increases while the amount of OCP particles decreases toward the cartilage layer. The scaffolds are bioactive as a mineral layer is deposited containing hydroxyapatite at the bony side. The scaffolds stimulate proliferation of human adipose-derived mesenchymal stem cells, but the degree of proliferation depends on the cell seeding density. The scaffolds give rise to a zone-specific gene expression. RUNX2, COL1A1, BGLAP, and SPP1 are upregulated in the bony layer of the scaffold. SOX9 is upregulated concomitant with COL2A1 expression in the cartilage zone. Mineralization in presence of the cells is prominent in the bone area with Ca and P steadily increasing over time. These results are encouraging for the fabrication of biomimetic scaffolds using ECM-like materials and featuring gradients that mimic native tissues and their interface.
Collapse
Affiliation(s)
- Elisabeth Amann
- Experimental Trauma SurgeryKlinikum rechts der IsarTechnical University of MunichMunich81675Germany
| | | | - Albina R. Franco
- 3B's Research GroupI3Bs‐Research Institute on BiomaterialsBiodegradables and BiomimeticsUniversity of MinhoHeadquarters of the European Institute of Excellence on Tissue Engineering and Regenerative MedicineAveparkBarcoGuimarães4805‐017Portugal
- ICVS/3B's—PT Government Associate LaboratoryBragaGuimarãesPortugal
| | - Patrina S. P. Poh
- Experimental Trauma SurgeryKlinikum rechts der IsarTechnical University of MunichMunich81675Germany
- Julius Wolff InstituteCharité—Universitätsmedizin Berlin13353BerlinGermany
| | | | | | - Isabel B. Leonor
- 3B's Research GroupI3Bs‐Research Institute on BiomaterialsBiodegradables and BiomimeticsUniversity of MinhoHeadquarters of the European Institute of Excellence on Tissue Engineering and Regenerative MedicineAveparkBarcoGuimarães4805‐017Portugal
- ICVS/3B's—PT Government Associate LaboratoryBragaGuimarãesPortugal
| | - Rui L. Reis
- 3B's Research GroupI3Bs‐Research Institute on BiomaterialsBiodegradables and BiomimeticsUniversity of MinhoHeadquarters of the European Institute of Excellence on Tissue Engineering and Regenerative MedicineAveparkBarcoGuimarães4805‐017Portugal
- ICVS/3B's—PT Government Associate LaboratoryBragaGuimarãesPortugal
- The Discoveries Centre for Regenerative and Precision MedicineHeadquarters at University of MinhoAveparkBarcoGuimarães4805‐017Portugal
| | - Martijn van Griensven
- Experimental Trauma SurgeryKlinikum rechts der IsarTechnical University of MunichMunich81675Germany
- Department of Cell Biology‐Inspired Tissue EngineeringMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityMaastricht6229 ERThe Netherlands
| | - Elizabeth R. Balmayor
- Experimental Trauma SurgeryKlinikum rechts der IsarTechnical University of MunichMunich81675Germany
- Department of Instructive Biomaterials EngineeringMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityMaastricht6229 ERThe Netherlands
| |
Collapse
|
45
|
Deviated nose: Physiological and pathological changes of the nasal cavity. Arch Plast Surg 2020; 47:505-515. [PMID: 33238336 PMCID: PMC7700847 DOI: 10.5999/aps.2020.01781] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 10/27/2020] [Indexed: 11/08/2022] Open
Abstract
Deviated nose is highly challenging in rhinoplasty since the surgeon should consider both aesthetic and functional aspects of the nose. Deviated nose correction is surgically complex, and a thorough understanding of the mechanical and physiological changes of intranasal structures, including the septum and turbinates, is necessary for functional improvement.
Collapse
|
46
|
Sultan S, Alalmie A, Noorwali A, Alyamani A, Shaabad M, Alfakeeh S, Bahmaid A, Ahmed F, Pushparaj P, Kalamegam G. Resveratrol promotes chondrogenesis of human Wharton’s jelly stem cells in a hyperglycemic state by modulating the expression of inflammation-related cytokines. ALL LIFE 2020. [DOI: 10.1080/26895293.2020.1835739] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Affiliation(s)
- Samar Sultan
- Medical Laboratory Technology Department, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ali Alalmie
- Medical Laboratory Technology Department, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Abdulwahab Noorwali
- Stem Cell Unit, King Fahd Medical Research Centre, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Clinical Biochemistry, College of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Aisha Alyamani
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Manal Shaabad
- Center of Excellence in Genomic Medicine Research (CEGMR), King Abdulaziz University, Jeddah, Saudi Arabia
| | - Saadiah Alfakeeh
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Afnan Bahmaid
- Center of Excellence in Genomic Medicine Research (CEGMR), King Abdulaziz University, Jeddah, Saudi Arabia
| | - Farid Ahmed
- Center of Excellence in Genomic Medicine Research (CEGMR), King Abdulaziz University, Jeddah, Saudi Arabia
| | - Peter Pushparaj
- Center of Excellence in Genomic Medicine Research (CEGMR), King Abdulaziz University, Jeddah, Saudi Arabia
| | - Gauthaman Kalamegam
- Center of Excellence in Genomic Medicine Research (CEGMR), King Abdulaziz University, Jeddah, Saudi Arabia
- Faculty of Medicine, AIMST University, Bedong, Malaysia
| |
Collapse
|
47
|
Chansoria P, Schuchard K, Shirwaiker RA. Process hybridization schemes for multiscale engineered tissue biofabrication. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2020; 13:e1673. [PMID: 33084240 DOI: 10.1002/wnan.1673] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 09/08/2020] [Accepted: 09/10/2020] [Indexed: 12/18/2022]
Abstract
Recapitulation of multiscale structure-function properties of cells, cell-secreted extracellular matrix, and 3D architecture of natural tissues is central to engineering biomimetic tissue substitutes. Toward achieving biomimicry, a variety of biofabrication processes have been developed, which can be broadly classified into five categories-fiber and fabric formation, additive manufacturing, surface modification, remote fields, and other notable processes-each with specific advantages and limitations. The majority of biofabrication literature has focused on using a single process at a time, which often limits the range of tissues that could be created with relevant features that span nano to macro scales. With multiscale biomimicry as the goal, development of hybrid biofabrication strategies that synergistically unite two or more processes to complement each other's strengths and limitations has been steadily increasing. This work discusses recent literature in this domain and attempts to equip the reader with the understanding of selecting appropriate processes that can harmonize toward creating engineered tissues with appropriate multiscale structure-function properties. Opportunities related to various hybridization schemes and a future outlook on scale-up biofabrication have also been discussed. This article is categorized under: Nanotechnology Approaches to Biology > Nanoscale Systems in Biology Implantable Materials and Surgical Technologies > Nanotechnology in Tissue Repair and Replacement.
Collapse
Affiliation(s)
- Parth Chansoria
- Edward P. Fitts Department of Industrial and Systems Engineering, North Carolina State University, Raleigh, North Carolina, USA.,Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina, USA
| | - Karl Schuchard
- Edward P. Fitts Department of Industrial and Systems Engineering, North Carolina State University, Raleigh, North Carolina, USA.,Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina, USA
| | - Rohan A Shirwaiker
- Edward P. Fitts Department of Industrial and Systems Engineering, North Carolina State University, Raleigh, North Carolina, USA.,Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina, USA.,Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina at Chapel Hill, Raleigh, North Carolina, USA
| |
Collapse
|
48
|
Oliveira Silva M, Gregory JL, Ansari N, Stok KS. Molecular Signaling Interactions and Transport at the Osteochondral Interface: A Review. Front Cell Dev Biol 2020; 8:750. [PMID: 32974333 PMCID: PMC7466715 DOI: 10.3389/fcell.2020.00750] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Accepted: 07/17/2020] [Indexed: 12/11/2022] Open
Abstract
Articular joints are comprised of different tissues, including cartilage and bone, with distinctive structural and mechanical properties. Joint homeostasis depends on mechanical and biological integrity of these components and signaling exchanges between them. Chondrocytes and osteocytes actively sense, integrate, and convert mechanical forces into biochemical signals in cartilage and bone, respectively. The osteochondral interface between the bone and cartilage allows these tissues to communicate with each other and exchange signaling and nutritional molecules, and by that ensure an integrated response to mechanical stimuli. It is currently not well known how molecules are transported between these tissues. Measuring molecular transport in vivo is highly desirable for tracking cartilage degeneration and osteoarthritis progression. Since transport of contrast agents, which are used for joint imaging, also depend on diffusion through the cartilage extracellular matrix, contrast agent enhanced imaging may provide a high resolution, non-invasive method for investigating molecular transport in the osteochondral unit. Only a few techniques have been developed to track molecular transport at the osteochondral interface, and there appear opportunities for development in this field. This review will describe current knowledge of the molecular interactions and transport in the osteochondral interface and discuss the potential of using contrast agents for investigating molecular transport and structural changes of the joint.
Collapse
Affiliation(s)
| | | | | | - Kathryn S. Stok
- Department of Biomedical Engineering, University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
49
|
Guo JB, Liang T, Che YJ, Yang HL, Luo ZP. Structure and mechanical properties of high-weight-bearing and low-weight-bearing areas of hip cartilage at the micro- and nano-levels. BMC Musculoskelet Disord 2020; 21:425. [PMID: 32616028 PMCID: PMC7333404 DOI: 10.1186/s12891-020-03468-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Accepted: 06/29/2020] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Articular cartilage has a high-weight-bearing area and a low-weight-bearing area, the macroscopic elastic moduli of the two regions are different. Chondrocytes are affected by the applied force at the microscopic level. Currently, the modulus of the two areas at the micro and nano levels is unknown, and studies on the relationship between macro-, micro- and nano-scale elastic moduli are limited. Such information may be important for further understanding of cartilage mechanics. Moreover, the surface morphology, proteoglycan content, and micro and nano structure of the two areas, which influences the mechanical properties of cartilage should be discussed. METHODS Safranin-O/Fast Green staining was used to evaluate the surface morphology and semi-quantify proteoglycan content of porcine femoral head cartilage between the two weight-bearing areas. The unconfined compression test was used to determine the macro elastic modulus. Atomic force microscope was used to measure the micro and nano compressive elastic modulus as well as the nano structure. Scanning electron microscope was employed to evaluate the micro structure. RESULTS No significant differences in the fibrillation index were observed between two areas (P = 0.5512). The Safranin-O index of the high-weight-bearing area was significantly higher than that of the low-weight-bearing area (P = 0.0387). The compressive elastic modulus of the high-weight-bearing area at the macro and micro level was significantly higher than that of the low-weight-bearing area (P = 0.0411 for macro-scale, and P = 0.0001 for micro-scale), while no statistically significant differences were observed in the elastic modulus of collagen fibrils at the nano level (P = 0.8544). The density of the collagen fibers was significantly lower in the high-weight-bearing area (P = 0.0177). No significant differences were observed in the structure and diameter of the collagen fibers between the two areas (P = 0.7361). CONCLUSIONS A higher proteoglycan content correlated with a higher compressive elastic modulus of the high-weight-bearing area at the micro level than that of the low-weight-bearing area, which was consistent with the trend observed from the macroscopic compressive elastic modulus. The weight-bearing level was not associated with the elastic modulus of individual collagen fibers and the diameter at the nano level. The micro structure of cartilage may influence the macro- and micro-scale elastic modulus.
Collapse
Affiliation(s)
- Jiang-Bo Guo
- Department of Orthopaedics, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, People's Republic of China.,Department of Orthopaedics, Orthopaedic Institute, the First Affiliated Hospital, Soochow University, Suzhou, Jiangsu, 215006, People's Republic of China
| | - Ting Liang
- Department of Orthopaedics, Orthopaedic Institute, the First Affiliated Hospital, Soochow University, Suzhou, Jiangsu, 215006, People's Republic of China
| | - Yan-Jun Che
- Department of Orthopaedics, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, People's Republic of China.,Department of Orthopaedics, Orthopaedic Institute, the First Affiliated Hospital, Soochow University, Suzhou, Jiangsu, 215006, People's Republic of China
| | - Hui-Lin Yang
- Department of Orthopaedics, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, People's Republic of China.,Department of Orthopaedics, Orthopaedic Institute, the First Affiliated Hospital, Soochow University, Suzhou, Jiangsu, 215006, People's Republic of China
| | - Zong-Ping Luo
- Department of Orthopaedics, Orthopaedic Institute, the First Affiliated Hospital, Soochow University, Suzhou, Jiangsu, 215006, People's Republic of China.
| |
Collapse
|
50
|
García-Fernández L, Olmeda-Lozano M, Benito-Garzón L, Pérez-Caballer A, San Román J, Vázquez-Lasa B. Injectable hydrogel-based drug delivery system for cartilage regeneration. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 110:110702. [DOI: 10.1016/j.msec.2020.110702] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 12/18/2019] [Accepted: 01/27/2020] [Indexed: 01/12/2023]
|