1
|
Heidenberger J, Reihs EI, Strauss J, Frauenlob M, Gültekin S, Gerner I, Tögel S, Ertl P, Windhager R, Jenner F, Rothbauer M. The effect of cyclic fluid perfusion on the proinflammatory tissue environment in osteoarthritis using equine joint-on-a-chip models. LAB ON A CHIP 2025; 25:2256-2269. [PMID: 40162708 DOI: 10.1039/d4lc01078g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Osteoarthritis (OA) is a prevalent degenerative joint disorder characterized by cartilage degradation, chronic inflammation, and progressive joint dysfunction. Despite rising incidences driven by ageing and increasing obesity, potent treatments remain elusive, exacerbating the socioeconomic burden. OA pathogenesis involves an imbalance in extracellular matrix (ECM) turnover, mediated by inflammatory cytokines and matrix-degrading enzymes, leading to oxidative stress, chondrocyte apoptosis, and ECM degradation. Additionally, synovial inflammation (synovitis) plays a critical role in disease progression through molecular crosstalk with cartilage and other joint tissues. Existing in vitro and in vivo OA models face significant limitations in replicating human pathophysiology, particularly the complex interplay between joint tissues. Equine models, due to their anatomical and cellular similarities to humans, offer translational relevance but remain underutilized. This study aims to develop an advanced 3D coculture system using equine chondrocytes and synoviocytes to simulate tissue-level interactions and fluid mechanical forces involved in OA. By incorporating inflammatory stimuli and gravity-driven cyclic fluid actuation, this model enables the study of OA-related molecular interactions in both healthy and diseased conditions under dynamic fluid conditions. Findings from this research provide important insights into pathological tissue crosstalk. In turn, this can help to better understand underlying inflammatory pathways and the potential contribution of fluid flow as an influential factor on the tissue microenvironment.
Collapse
Affiliation(s)
- Johannes Heidenberger
- Department of Orthopedics and Trauma Surgery, Karl Chiari Lab for Orthopaedic Biology, Medizinische Universität Wien, Währinger Gürtel 18-20, 1090 Vienna, Austria.
| | - Eva I Reihs
- Department of Orthopedics and Trauma Surgery, Karl Chiari Lab for Orthopaedic Biology, Medizinische Universität Wien, Währinger Gürtel 18-20, 1090 Vienna, Austria.
- Faculty of Technical Chemistry, Technische Universität Wien, Getreidemarkt 9/163, 1060 Vienna, Austria.
| | - Jonathan Strauss
- Department of Orthopedics and Trauma Surgery, Karl Chiari Lab for Orthopaedic Biology, Medizinische Universität Wien, Währinger Gürtel 18-20, 1090 Vienna, Austria.
- Faculty of Technical Chemistry, Technische Universität Wien, Getreidemarkt 9/163, 1060 Vienna, Austria.
| | - Martin Frauenlob
- Faculty of Technical Chemistry, Technische Universität Wien, Getreidemarkt 9/163, 1060 Vienna, Austria.
| | - Sinan Gültekin
- Department for Small Animals and Horses, Centre for Equine Health and Research, Equine Surgery Unit, Veterinary Regenerative Medicine Laboratory, University of Veterinary Medicine Vienna, Veterinärplatz 1, 1210 Vienna, Austria
| | - Iris Gerner
- Department for Small Animals and Horses, Centre for Equine Health and Research, Equine Surgery Unit, Veterinary Regenerative Medicine Laboratory, University of Veterinary Medicine Vienna, Veterinärplatz 1, 1210 Vienna, Austria
| | - Stefan Tögel
- Department of Orthopedics and Trauma Surgery, Karl Chiari Lab for Orthopaedic Biology, Medizinische Universität Wien, Währinger Gürtel 18-20, 1090 Vienna, Austria.
- Ludwig Boltzmann Institute for Arthritis and Rehabilitation, Vienna, Austria
| | - Peter Ertl
- Faculty of Technical Chemistry, Technische Universität Wien, Getreidemarkt 9/163, 1060 Vienna, Austria.
| | - Reinhard Windhager
- Department of Orthopedics and Trauma Surgery, Karl Chiari Lab for Orthopaedic Biology, Medizinische Universität Wien, Währinger Gürtel 18-20, 1090 Vienna, Austria.
| | - Florien Jenner
- Department for Small Animals and Horses, Centre for Equine Health and Research, Equine Surgery Unit, Veterinary Regenerative Medicine Laboratory, University of Veterinary Medicine Vienna, Veterinärplatz 1, 1210 Vienna, Austria
| | - Mario Rothbauer
- Department of Orthopedics and Trauma Surgery, Karl Chiari Lab for Orthopaedic Biology, Medizinische Universität Wien, Währinger Gürtel 18-20, 1090 Vienna, Austria.
- Faculty of Technical Chemistry, Technische Universität Wien, Getreidemarkt 9/163, 1060 Vienna, Austria.
| |
Collapse
|
2
|
Asadikorayem M, Surman F, Weber P, Weber D, Zenobi-Wong M. Zwitterionic Granular Hydrogel for Cartilage Tissue Engineering. Adv Healthc Mater 2024; 13:e2301831. [PMID: 37501337 DOI: 10.1002/adhm.202301831] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 07/24/2023] [Indexed: 07/29/2023]
Abstract
Zwitterionic hydrogels have high potential for cartilage tissue engineering due to their ultra-hydrophilicity, nonimmunogenicity, and superior antifouling properties. However, their application in this field has been limited so far, due to the lack of injectable zwitterionic hydrogels that allow for encapsulation of cells in a biocompatible manner. Herein, a novel strategy is developed to engineer cartilage employing zwitterionic granular hydrogels that are injectable, self-healing, in situ crosslinkable and allow for direct encapsulation of cells with biocompatibility. The granular hydrogel is produced by mechanical fragmentation of bulk photocrosslinked hydrogels made of zwitterionic carboxybetaine acrylamide (CBAA), or a mixture of CBAA and zwitterionic sulfobetaine methacrylate (SBMA). The produced microgels are enzymatically crosslinkable using horseradish peroxidase, to quickly stabilize the construct, resulting in a microporous hydrogel. Encapsulated human primary chondrocytes are highly viable and able to proliferate, migrate, and produce cartilaginous extracellular matrix (ECM) in the zwitterionic granular hydrogel. It is also shown that by increasing hydrogel porosity and incorporation of SBMA, cell proliferation and ECM secretion are further improved. This strategy is a simple and scalable method, which has high potential for expanding the versatility and application of zwitterionic hydrogels for diverse tissue engineering applications.
Collapse
Affiliation(s)
- Maryam Asadikorayem
- Tissue Engineering + Biofabrication Laboratory, Department of Health Sciences and Technology, ETH Zürich, Otto-Stern-Weg 7, Zürich, 8093, Switzerland
| | - František Surman
- Tissue Engineering + Biofabrication Laboratory, Department of Health Sciences and Technology, ETH Zürich, Otto-Stern-Weg 7, Zürich, 8093, Switzerland
| | - Patrick Weber
- Tissue Engineering + Biofabrication Laboratory, Department of Health Sciences and Technology, ETH Zürich, Otto-Stern-Weg 7, Zürich, 8093, Switzerland
| | - Daniel Weber
- Division of Hand Surgery, University Children's Hospital, Zürich, 8032, Switzerland
| | - Marcy Zenobi-Wong
- Tissue Engineering + Biofabrication Laboratory, Department of Health Sciences and Technology, ETH Zürich, Otto-Stern-Weg 7, Zürich, 8093, Switzerland
| |
Collapse
|
3
|
Huang LW, Huang TC, Hu YC, Hsieh BS, Lin JS, Hsu HY, Lee CC, Chang KL. The Oral Administration of Lactobacillus delbrueckii subsp. lactis 557 (LDL557) Ameliorates the Progression of Monosodium Iodoacetate-Induced Osteoarthritis. Curr Issues Mol Biol 2024; 46:8969-8980. [PMID: 39194747 DOI: 10.3390/cimb46080530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 08/12/2024] [Accepted: 08/14/2024] [Indexed: 08/29/2024] Open
Abstract
Low-grade body inflammation is a major cause of osteoarthritis (OA), a common joint disease. Gut dysbiosis may lead to systemic inflammation which can be prevented by probiotic administration. The Lactobacillus delbrueckii subsp. lactis 557 (LDL557) has been demonstrated to have beneficial effects for anti-inflammation. This study investigated the effects of LDL557 on OA progress using monosodium iodoacetate (MIA)-induced OA of rats. Live or heat-killed (HK)-LDL557 of a low or high dose was administrated for two weeks before MIA-induced OA, and then continuously administrated for another six weeks. After taking supplements for eight weeks, OA progress was analyzed. Results showed that MIA induced knee joint swelling, chondrocyte damage, and cartilage degradation, and supplementation with a high dose of LDL557 reduced MIA-induced knee joint swelling, chondrocyte damage, and cartilage degradation. Additionally, MIA increased serum levels of the matrix-degrading enzyme MMP-13, while a high dose of HK-LDL557 decreased it for the controls. Simultaneously, bone turnover markers and inflammatory cytokines of serum were assayed, but no significant differences were found except for a TNF-α decrease from a low dose of live LDL557. These results demonstrated that supplementation with high doses of live LDL557 or HK-LDL557 can reduce the progression of MIA-induced OA in rats.
Collapse
Affiliation(s)
- Li-Wen Huang
- Department of Medical Laboratory Science and Biotechnology, College of Health Sciences, Kaohsiung Medical University, Kaohsiung 807378, Taiwan
| | - Tzu-Ching Huang
- Department of Biochemistry, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807378, Taiwan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807378, Taiwan
| | - Yu-Chen Hu
- Department of Biochemistry, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807378, Taiwan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807378, Taiwan
| | - Bau-Shan Hsieh
- Department of Biochemistry, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807378, Taiwan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807378, Taiwan
| | - Jin-Seng Lin
- Culture Collection & Research Institute, SYNBIO TECH INC., Kaohsiung 821011, Taiwan
| | - Han-Yin Hsu
- Culture Collection & Research Institute, SYNBIO TECH INC., Kaohsiung 821011, Taiwan
| | - Chia-Chia Lee
- Culture Collection & Research Institute, SYNBIO TECH INC., Kaohsiung 821011, Taiwan
| | - Kee-Lung Chang
- Department of Biochemistry, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807378, Taiwan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807378, Taiwan
| |
Collapse
|
4
|
Lu X, Dai S, Huang B, Li S, Wang P, Zhao Z, Li X, Li N, Wen J, Sun Y, Man Z, Liu B, Li W. Exosomes loaded a smart bilayer-hydrogel scaffold with ROS-scavenging and macrophage-reprogramming properties for repairing cartilage defect. Bioact Mater 2024; 38:137-153. [PMID: 38699244 PMCID: PMC11063794 DOI: 10.1016/j.bioactmat.2024.04.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 04/01/2024] [Accepted: 04/16/2024] [Indexed: 05/05/2024] Open
Abstract
Enhancing the regeneration of cartilage defects remains challenging owing to limited innate self-healing as well as acute inflammation arising from the overexpression of reactive oxygen species (ROS) in post-traumatic microenvironments. Recently, stem cell-derived exosomes (Exos) have been developed as potential cell-free therapy for cartilage regeneration. Although this approach promotes chondrogenesis, it neglects the emerging inflammatory microenvironment. In this study, a smart bilayer-hydrogel dual-loaded with sodium diclofenac (DC), an anti-inflammatory drug, and Exos from bone marrow-derived mesenchymal stem cells was developed to mitigate initial-stage inflammation and promote late-stage stem-cell recruitment and chondrogenic differentiation. First, the upper-hydrogel composed of phenylboronic-acid-crosslinked polyvinyl alcohol degrades in response to elevated levels of ROS to release DC, which mitigates oxidative stress, thus reprogramming macrophages to the pro-healing state. Subsequently, Exos are slowly released from the lower-hydrogel composed of hyaluronic acid into an optimal microenvironment for the stimulation of chondrogenesis. Both in vitro and in vivo assays confirmed that the dual-loaded bilayer-hydrogel reduced post-traumatic inflammation and enhanced cartilage regeneration by effectively scavenging ROS and reprogramming macrophages. The proposed platform provides multi-staged therapy, which allows for the optimal harnessing of Exos as a therapeutic for cartilage regeneration.
Collapse
Affiliation(s)
- Xiaoqing Lu
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, PR China
- College of Sports Medicine and Rehabilitation, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong, 271016, PR China
| | - Shimin Dai
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, PR China
| | - Benzhao Huang
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, PR China
| | - Shishuo Li
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, PR China
| | - Peng Wang
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, PR China
| | - Zhibo Zhao
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, PR China
| | - Xiao Li
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, PR China
| | - Ningbo Li
- School of Stomatology, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, PR China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, PR China
| | - Jie Wen
- School of Stomatology, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, PR China
| | - Yunhan Sun
- School of Stomatology, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, PR China
| | - Zhentao Man
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, PR China
- College of Sports Medicine and Rehabilitation, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong, 271016, PR China
- Endocrine and Metabolic Diseases Hospital of Shandong First Medical University, Shandong Institute of Endocrine and Metabolic Diseases, Jinan, Shandong, 250062, PR China
| | - Bing Liu
- School of Stomatology, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, PR China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, PR China
| | - Wei Li
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, PR China
- College of Sports Medicine and Rehabilitation, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong, 271016, PR China
| |
Collapse
|
5
|
Zhang G, Qin J, Xu W, Liu M, Wu R, Qin Y. Gene expression and immune infiltration analysis comparing lesioned and preserved subchondral bone in osteoarthritis. PeerJ 2024; 12:e17417. [PMID: 38827307 PMCID: PMC11141552 DOI: 10.7717/peerj.17417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 04/28/2024] [Indexed: 06/04/2024] Open
Abstract
Background Osteoarthritis (OA) is a degenerative disease requiring additional research. This study compared gene expression and immune infiltration between lesioned and preserved subchondral bone. The results were validated using multiple tissue datasets and experiments. Methods Differentially expressed genes (DEGs) between the lesioned and preserved tibial plateaus of OA patients were identified in the GSE51588 dataset. Moreover, functional annotation and protein-protein interaction (PPI) network analyses were performed on the lesioned and preserved sides to explore potential therapeutic targets in OA subchondral bones. In addition, multiple tissues were used to screen coexpressed genes, and the expression levels of identified candidate DEGs in OA were measured by quantitative real-time polymerase chain reaction. Finally, an immune infiltration analysis was conducted. Results A total of 1,010 DEGs were identified, 423 upregulated and 587 downregulated. The biological process (BP) terms enriched in the upregulated genes included "skeletal system development", "sister chromatid cohesion", and "ossification". Pathways were enriched in "Wnt signaling pathway" and "proteoglycans in cancer". The BP terms enriched in the downregulated genes included "inflammatory response", "xenobiotic metabolic process", and "positive regulation of inflammatory response". The enriched pathways included "neuroactive ligand-receptor interaction" and "AMP-activated protein kinase signaling". JUN, tumor necrosis factor α, and interleukin-1β were the hub genes in the PPI network. Collagen XI A1 and leucine-rich repeat-containing 15 were screened from multiple datasets and experimentally validated. Immune infiltration analyses showed fewer infiltrating adipocytes and endothelial cells in the lesioned versus preserved samples. Conclusion Our findings provide valuable information for future studies on the pathogenic mechanism of OA and potential therapeutic and diagnostic targets.
Collapse
Affiliation(s)
- Gang Zhang
- The Second Affiliated Hospital of Harbin Medical University, Department of Orthopedics Surgery, Harbin Medical University, Harbin, China
- Department of Orthopedics, Harbin First Hospital, Harbin, China
- Future Medicine Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jinwei Qin
- Department of Emergency, Harbin First Hospital, Harbin, China
| | - Wenbo Xu
- The Second Affiliated Hospital of Harbin Medical University, Department of Orthopedics Surgery, Harbin Medical University, Harbin, China
| | - Meina Liu
- Department of Biostatistics, School of Public Health, Harbin Medical University, Harbin, China
| | - Rilige Wu
- Medical Big Data Research Center, Medical Innovation Research Division of PLA General Hospital, Beijing, China
| | - Yong Qin
- The Second Affiliated Hospital of Harbin Medical University, Department of Orthopedics Surgery, Harbin Medical University, Harbin, China
| |
Collapse
|
6
|
Hasson M, Fernandes LM, Solomon H, Pepper T, Huffman NL, Pucha SA, Bariteau JT, Kaiser JM, Patel JM. Considering the Cellular Landscape in Marrow Stimulation Techniques for Cartilage Repair. Cells Tissues Organs 2024; 213:523-537. [PMID: 38599194 PMCID: PMC11633897 DOI: 10.1159/000538530] [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: 11/30/2023] [Accepted: 03/21/2024] [Indexed: 04/12/2024] Open
Abstract
BACKGROUND Marrow stimulation is a common reparative approach to treat injuries to cartilage and other soft tissues (e.g., rotator cuff). It involves the recruitment of bone marrow elements and mesenchymal stem cells (MSCs) into the defect, theoretically initiating a regenerative process. However, the resulting repair tissue is often weak and susceptible to deterioration with time. The populations of cells at the marrow stimulation site (beyond MSCs), and their contribution to inflammation, vascularity, and fibrosis, may play a role in quality of the repair tissue. SUMMARY In this review, we accomplish three goals: (1) systematically review clinical trials on the augmentation of marrow stimulation and evaluate their assumptions on the biological elements recruited; (2) detail the cellular populations in bone marrow and their impact on healing; and (3) highlight emerging technologies and approaches that could better guide these specific cell populations towards enhanced cartilage or soft tissue formation. KEY MESSAGES We found that most clinical trials do not account for cell heterogeneity, nor do they specify the regenerative element recruited, and those that do typically utilize descriptions such as "clots," "elements," and "blood." Furthermore, our review of bone marrow cell populations demonstrates a dramatically heterogenous cell population, including hematopoietic cells, immune cells, fibroblasts, macrophages, and only a small population of MSCs. Finally, the field has developed numerous innovative techniques to enhance the chondrogenic potential (and reduce the anti-regenerative impacts) of these various cell types. We hope this review will guide approaches that account for cellular heterogeneity and improve marrow stimulation techniques to treat chondral defects.
Collapse
Affiliation(s)
- Maddie Hasson
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, GA, USA
- Atlanta VA Medical Center, Department of Veterans Affairs, Decatur, GA, USA
| | - Lorenzo M. Fernandes
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, GA, USA
- Atlanta VA Medical Center, Department of Veterans Affairs, Decatur, GA, USA
| | - Hanna Solomon
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, GA, USA
- Atlanta VA Medical Center, Department of Veterans Affairs, Decatur, GA, USA
| | - Tristan Pepper
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, GA, USA
| | - Nicholas L. Huffman
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, GA, USA
| | - Saitheja A. Pucha
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, GA, USA
- Atlanta VA Medical Center, Department of Veterans Affairs, Decatur, GA, USA
| | - Jason T. Bariteau
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, GA, USA
| | - Jarred M. Kaiser
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, GA, USA
- Atlanta VA Medical Center, Department of Veterans Affairs, Decatur, GA, USA
| | - Jay M. Patel
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, GA, USA
- Atlanta VA Medical Center, Department of Veterans Affairs, Decatur, GA, USA
| |
Collapse
|
7
|
Chen B, Lin C, Jin X, Zhang X, Yang K, Wang J, Zhang F, Zhang Y, Ji Y, Meng Z. Construction of a diagnostic model for osteoarthritis based on transcriptomic immune-related genes. Heliyon 2024; 10:e23636. [PMID: 38187306 PMCID: PMC10770511 DOI: 10.1016/j.heliyon.2023.e23636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 12/08/2023] [Accepted: 12/08/2023] [Indexed: 01/09/2024] Open
Abstract
BACKGROUND Osteoarthritis (OA) is a leading cause of disability globally, affecting over 500 million individuals worldwide. However, accurate and early diagnosis of OA is challenging to achieve. Immune-related genes play an essential role in OA development. Therefore, the objective of this study was to develop a diagnostic model for OA based on immune-related genes identified in synovial membrane. METHODS The gene expression profile of OA were downloaded based on four datasets. The significantly differentially expressed genes (DEGs) between OA and control groups were selected. The differential immune cells were analyzed, followed by immune-related DEGs screening. WGCNA was used to screen module genes and these genes were further selected through optimization algorithm. Then, nomogram model was constructed. Chemical drug small molecule related to OA was predicted. Finally, expression levels of several key genes were validated by qRT-PCR through construction of OA rat models. RESULTS The total 656 DEGs were obtained. Eight immune cells were significantly differential between two groups, and 317 immune-related DEGs were obtained. WGCNA identified three modules. The genes in modules were significantly involved in 15 pathways, involving in 65 genes. Then 12 DEGs were screened as the final optimal combination of DEGs, such as CEBPB, CXCL1, JUND, GABARAPL2 and PDGFC. The Nomogram model was also constructed. Furthermore, the chemical small molecules, such as acetaminophen, aspirin, and caffeine were predicted. The expression levels of CEBPB, CXCL1, GABARAPL2 and PDGFC were validated in OA rat models. CONCLUSION A diagnostic model based on twelve immune related genes was constructed. These model genes, such as CEBPB, CXCL1, GABARAPL2, and PDGFC, may serve as diagnostic biomarkers and immunotherapeutic targets.
Collapse
Affiliation(s)
- Bo Chen
- Rehabilitation Medicine Department, Northern Jiangsu People's Hospital, Yangzhou, Jiangsu, 225001, China
| | - Chun Lin
- Rehabilitation Medicine Department, Northern Jiangsu People's Hospital, Yangzhou, Jiangsu, 225001, China
| | - Xing Jin
- Rehabilitation Medicine Department, Northern Jiangsu People's Hospital, Yangzhou, Jiangsu, 225001, China
| | - Xibin Zhang
- Rehabilitation Medicine Department, Northern Jiangsu People's Hospital, Yangzhou, Jiangsu, 225001, China
| | - Kang Yang
- Rehabilitation Medicine Department, Northern Jiangsu People's Hospital, Yangzhou, Jiangsu, 225001, China
| | - Jianjian Wang
- Rehabilitation Medicine Department, Northern Jiangsu People's Hospital, Yangzhou, Jiangsu, 225001, China
| | - Feng Zhang
- Rehabilitation Medicine Department, Northern Jiangsu People's Hospital, Yangzhou, Jiangsu, 225001, China
| | - Yuxin Zhang
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai 200011, China
- Department of Rehabilitation Medicine, Huangpu Branch, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Yingying Ji
- The affiliated Wuxi Mental Health Center of Jiangnan University, Wuxi Central Rehabilitation Hospital, Wuxi, Jiangsu, 214151, China
| | - Zhaoxiang Meng
- Rehabilitation Medicine Department, Northern Jiangsu People's Hospital, Yangzhou, Jiangsu, 225001, China
| |
Collapse
|
8
|
Giannasi C, Della Morte E, Cadelano F, Valenza A, Casati S, Dei Cas M, Niada S, Brini AT. Boosting the therapeutic potential of cell secretome against osteoarthritis: Comparison of cytokine-based priming strategies. Biomed Pharmacother 2024; 170:115970. [PMID: 38042116 DOI: 10.1016/j.biopha.2023.115970] [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: 08/08/2023] [Revised: 11/22/2023] [Accepted: 11/27/2023] [Indexed: 12/04/2023] Open
Abstract
The secretome, or conditioned medium (CM), from Mesenchymal Stem/stromal Cells (MSCs) has recently emerged as a promising cell-free therapeutic against osteoarthritis (OA), capable of promoting cartilage regeneration and immunoregulation. Priming MSCs with 10 ng/ml tumor necrosis factor α (TNFα) and/or 10 ng/ml interleukin 1β (IL-1β) aims at mimicking the pathological milieu of OA joints in order to target their secretion towards a pathology-tailored phenotype. Here we compare the composition of the CM obtained after 24 or 72 h from untreated and cytokine-treated adipose-derived MSCs (ASCs). The 72-hour double-primed CM presents a higher total protein yield, a larger number of extracellular vesicles, and a greater concentration of bioactive lipids, in particular sphingolipids, fatty acids, and eicosanoids. Moreover, the levels of several factors involved in immunomodulation and regeneration, such as TGF-β1, PGE2, and CCL-2, are strongly upregulated. Additionally, the differential profiling of 80 bioactive molecules indicates that primed CM is enriched in immune cell chemotaxis and migration factors. Our results indicate that pre-conditioning ASCs with inflammatory cytokines can modulate the composition of their CM, promoting the release of factors with recognized anti-inflammatory, chondroprotective, and immunoregulatory properties.
Collapse
Affiliation(s)
- Chiara Giannasi
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milan, Italy; IRCCS Istituto Ortopedico Galeazzi, Milan, Italy.
| | | | - Francesca Cadelano
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milan, Italy; IRCCS Istituto Ortopedico Galeazzi, Milan, Italy
| | | | - Sara Casati
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milan, Italy
| | - Michele Dei Cas
- Department of Health Sciences, University of Milan, Milan, Italy
| | | | - Anna Teresa Brini
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milan, Italy; IRCCS Istituto Ortopedico Galeazzi, Milan, Italy
| |
Collapse
|
9
|
Du X, Xin R, Chen X, Wang G, Huang C, Zhou K, Zhang S. TAF15 regulates the BRD4/GREM1 axis and activates the gremlin-1-NF-κB pathway to promote OA progression. Regen Ther 2023; 24:227-236. [PMID: 37496731 PMCID: PMC10366938 DOI: 10.1016/j.reth.2023.06.016] [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: 04/06/2023] [Revised: 06/18/2023] [Accepted: 06/29/2023] [Indexed: 07/28/2023] Open
Abstract
Background Anterior cruciate ligament (ACL) injury is recognized as a risk factor for osteoarthritis (OA) progression. Herein, the function of TAF15 in ACL injury-induced OA was investigated. Methods OA cell model and OA mouse model were established by interleukin-1 beta (IL-1β) stimulation and ACL transection administration, respectively. The pathological changes were analyzed by histopathology. The mRNA and protein expressions were assessed using qRT-PCR, Western blot and IHC. Chondrocyte viability and apoptosis were examined by CCK8 assay and TUNEL staining, respectively. The interactions between TAF15, BRD4 and GREM1 were analyzed by RIP or ChIP assay. Results TAF15 expression was markedly elevated in OA, and its knockdown suppressed IL-1β-induced chondrocyte apoptosis and ECM degradation in vivo and cartilage pathological changes in vitro. TAF15 promoted BRD4 mRNA stability, and TAF15 silencing's repression on chondrocyte apoptosis and ECM degradation induced by IL-1β was abrogated following BRD4 overexpression. BRD4 promoted GREM1 expression by directly binding with GREM1. In addition, the GREM1/NF-κB pathway functioned as the downstream pathway of BRD4 in promoting OA progression. Conclusion TAF15 upregulation facilitated chondrocyte apoptosis and ECM degradation during OA development by acting on the BRD4/GREM1/NF-κB axis, which provided a theoretical basis for the development of novel therapies for OA.
Collapse
Affiliation(s)
- Xiufan Du
- Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Department of Sports Medicine, Haikou, 570311, Hainan, PR China
| | - Ruomei Xin
- Danzhou People's Hospital, Nursing Department, Danzhou, 571700, Hainan, PR China
| | - Xiaoyan Chen
- Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Department of Stomatology, Haikou, 570311, Hainan, PR China
| | - Guangji Wang
- Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Department of Sports Medicine, Haikou, 570311, Hainan, PR China
| | - Chunhang Huang
- Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Department of Sports Medicine, Haikou, 570311, Hainan, PR China
| | - Kai Zhou
- Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Department of Sports Medicine, Haikou, 570311, Hainan, PR China
| | - Shunli Zhang
- The Second Affiliated Hospital of Hainan Medical University, Haikou, 570216, Hainan, PR China
| |
Collapse
|
10
|
Takemura H, Okumo T, Tatsuo T, Izukashi K, Ikemoto H, Adachi N, Mochizuki M, Kanzaki K, Sunagawa M. The Preventive Effects of Platelet-Rich Plasma Against Knee Osteoarthritis Progression in Rats. Cureus 2023; 15:e48825. [PMID: 38106771 PMCID: PMC10722351 DOI: 10.7759/cureus.48825] [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] [Accepted: 11/14/2023] [Indexed: 12/19/2023] Open
Abstract
BACKGROUND In recent years, the intra-articular administration of platelet-rich plasma (PRP), a novel therapeutic strategy for knee osteoarthritis (KOA), has gained attention. However, the efficacy of PRP in inhibiting degenerative joint changes remains unclear. The current study aimed to evaluate the therapeutic effect of the intra-articular administration of PRP in rats with induced KOA. MATERIALS AND METHODS PRP was prepared from the whole blood of nine-week-old male Wistar rats via centrifugation at 25°C, 200 × g, for seven minutes. KOA was induced in the right knees of the rats via destabilization of the medial meniscus (DMM) surgery. The animals were divided into the control, sham, DMM, and DMM + PRP groups (n = 5 each). The rats in the DMM + PRP group received 50 μL of intra-articular PRP in the right knee joint four weeks after surgery. The rotarod test was conducted to assess locomotive function. Eight weeks after DMM surgery, the degree of medial meniscus extrusion was measured via computed tomography (CT) images on the right knee. Then, a histological analysis of the harvested knees was conducted. KOA progression was assessed using the Osteoarthritis Research Society International (OARSI) score. The number of multinucleated tartrate-resistant acid phosphatase (TRAP)-positive osteoclasts in the subchondral bone was counted via histological analysis. RESULTS The degree of medial meniscus extrusion did not significantly differ between the DMM and DMM + PRP groups. Similarly, there were no significant differences in the walking time based on the rotarod test between the DMM and DMM + PRP groups. However, the DMM group had a significantly higher OARSI score than the DMM + PRP group. The number of TRAP-positive osteoclasts in the subchondral bone of the DMM group increased over time, peaking four weeks after surgery. The DMM + PRP group had a higher number of TRAP-positive osteoclasts in the subchondral bone than the control group. However, there was no significant difference between the number of TRAP-positive osteoclasts between the DMM group and the control and sham groups. CONCLUSION The intra-articular administration of PRP may inhibit KOA progression in a rat model, especially in the articular cartilage degradation and osteophyte formation. The results can provide further evidence about the efficacy of PRP against KOA progression and can contribute to the current practice of healthcare professionals based on accurate knowledge.
Collapse
Affiliation(s)
- Haruka Takemura
- Department of Physiology, Showa University Graduate School of Medicine, Tokyo, JPN
- Department of Orthopedic Surgery, Showa University Fujigaoka Hospital, Yokohama, JPN
| | - Takayuki Okumo
- Department of Orthopedic Surgery, Showa University Fujigaoka Hospital, Yokohama, JPN
- Department of Physiology, Showa University Graduate School of Medicine, Tokyo, JPN
| | - Tokito Tatsuo
- Department of Pharmacology, Showa University Graduate School of Medicine, Tokyo, JPN
- Department of Orthopedic Surgery, Showa University Fujigaoka Hospital, Yokohama, JPN
| | - Kanako Izukashi
- Department of Physiology, Showa University Graduate School of Medicine, Tokyo, JPN
- Department of Orthopedics, Showa University Fujigaoka Hospital, Yokohama, JPN
| | - Hideshi Ikemoto
- Department of Physiology, Showa University Graduate School of Medicine, Tokyo, JPN
| | - Naoki Adachi
- Department of Physiology, Showa University Graduate School of Medicine, Tokyo, JPN
| | - Midori Mochizuki
- Department of Physiology, Showa University Graduate School of Medicine, Tokyo, JPN
| | - Koji Kanzaki
- Department of Orthopedic Surgery, Showa University Fujigaoka Hospital, Yokohama, JPN
| | - Masataka Sunagawa
- Department of Physiology, Showa University Graduate School of Medicine, Tokyo, JPN
| |
Collapse
|
11
|
Pinto-Cardoso R, Bessa-Andrês C, Correia-de-Sá P, Bernardo Noronha-Matos J. Could hypoxia rehabilitate the osteochondral diseased interface? Lessons from the interplay of hypoxia and purinergic signals elsewhere. Biochem Pharmacol 2023:115646. [PMID: 37321413 DOI: 10.1016/j.bcp.2023.115646] [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: 04/07/2023] [Revised: 06/03/2023] [Accepted: 06/07/2023] [Indexed: 06/17/2023]
Abstract
The osteochondral unit comprises the articular cartilage (90%), subchondral bone (5%) and calcified cartilage (5%). All cells present at the osteochondral unit that is ultimately responsible for matrix production and osteochondral homeostasis, such as chondrocytes, osteoblasts, osteoclasts and osteocytes, can release adenine and/or uracil nucleotides to the local microenvironment. Nucleotides are released by these cells either constitutively or upon plasma membrane damage, mechanical stress or hypoxia conditions. Once in the extracellular space, endogenously released nucleotides can activate membrane-bound purinoceptors. Activation of these receptors is fine-tuning regulated by nucleotides' breakdown by enzymes of the ecto-nucleotidase cascade. Depending on the pathophysiological conditions, both the avascular cartilage and the subchondral bone subsist to significant changes in oxygen tension, which has a tremendous impact on tissue homeostasis. Cell stress due to hypoxic conditions directly influences the expression and activity of several purinergic signalling players, namely nucleotide release channels (e.g. Cx43), NTPDase enzymes and purinoceptors. This review gathers experimental evidence concerning the interplay between hypoxia and the purinergic signalling cascade contributing to osteochondral unit homeostasis. Reporting deviations to this relationship resulting from pathological alterations of articular joints may ultimately unravel novel therapeutic targets for osteochondral rehabilitation. At this point, one can only hypothesize how hypoxia mimetic conditions can be beneficial to the ex vivo expansion and differentiation of osteo- and chondro-progenitors for auto-transplantation and tissue regenerative purposes.
Collapse
Affiliation(s)
- Rui Pinto-Cardoso
- Laboratório de Farmacologia e Neurobiologia; Center for Drug Discovery and Innovative Medicines (MedInUP), Departamento de Imuno-Fisiologia e Farmacologia, Instituto de Ciências Biomédicas Abel Salazar - Universidade do Porto (ICBAS-UP)
| | - Catarina Bessa-Andrês
- Laboratório de Farmacologia e Neurobiologia; Center for Drug Discovery and Innovative Medicines (MedInUP), Departamento de Imuno-Fisiologia e Farmacologia, Instituto de Ciências Biomédicas Abel Salazar - Universidade do Porto (ICBAS-UP)
| | - Paulo Correia-de-Sá
- Laboratório de Farmacologia e Neurobiologia; Center for Drug Discovery and Innovative Medicines (MedInUP), Departamento de Imuno-Fisiologia e Farmacologia, Instituto de Ciências Biomédicas Abel Salazar - Universidade do Porto (ICBAS-UP)
| | - José Bernardo Noronha-Matos
- Laboratório de Farmacologia e Neurobiologia; Center for Drug Discovery and Innovative Medicines (MedInUP), Departamento de Imuno-Fisiologia e Farmacologia, Instituto de Ciências Biomédicas Abel Salazar - Universidade do Porto (ICBAS-UP).
| |
Collapse
|
12
|
Stoddart MJ, Della Bella E, Armiento AR. Cartilage Tissue Engineering: An Introduction. Methods Mol Biol 2023; 2598:1-7. [PMID: 36355280 DOI: 10.1007/978-1-0716-2839-3_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Once damaged, cartilage has limited healing capability. This has led to a huge body of research that aims to repair or regenerate this important tissue. Despite the progress made, significant hurdles still need to be overcome. This chapter highlights some of the progress made, while elaborating on areas that need further research. The concept of translation and the route to clinical translation must be kept in mind if some of the promising preclinical research is to make it to routine clinical application.
Collapse
Affiliation(s)
| | | | - Angela R Armiento
- AO Research Institute Davos, Davos Platz, Switzerland
- UCB Pharma, Slough, UK
| |
Collapse
|
13
|
Four-octyl itaconate improves osteoarthritis by enhancing autophagy in chondrocytes via PI3K/AKT/mTOR signalling pathway inhibition. Commun Biol 2022; 5:641. [PMID: 35768581 PMCID: PMC9242998 DOI: 10.1038/s42003-022-03592-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 06/15/2022] [Indexed: 12/15/2022] Open
Abstract
Osteoarthritis (OA) is a highly prevalent and chronic disorder that is associated with a substantial social and economic burden. Itaconate, as an important regulator of cellular inflammation, is a metabolite synthesised by an enzyme encoded by immune-responsive gene 1. However, there are few studys regarding the effects of itaconate on OA. Here, we show the effect of the cell-permeable itaconate derivative 4-octyl itaconate (OI) on OA. OI attenuates the chondrocyte apoptosis induced by interleukin 1β (IL-1β) in vitro, indicating that OI protect chondrocytes against apoptosis. Moreover, OI ameliorates the chondrocyte autophagy inhibition induced by IL-1β via the inhibition of PI3K/AKT/mTOR signalling pathway. Finally, OI enhances autophagy and reduces cartilage degradation in a rat model of OA established by destabilization of medial meniscus (DMM). In summary, our findings reveal that OI is involved in regulating the progression of OA. The above results shed light on the treatment of OA. 4-octyl itaconate (OI) attenuates chondrocyte apoptosis and ameliorates cartilage degradation and defection of autophagy induced by IL-1 β via the PI3K/AKT/mTOR pathway. OI improved the autophagy and reduced the inflammation of rat models of osteoarthritis.
Collapse
|
14
|
Barisón MJ, Nogoceke R, Josino R, Horinouchi CDDS, Marcon BH, Correa A, Stimamiglio MA, Robert AW. Functionalized Hydrogels for Cartilage Repair: The Value of Secretome-Instructive Signaling. Int J Mol Sci 2022; 23:ijms23116010. [PMID: 35682690 PMCID: PMC9181449 DOI: 10.3390/ijms23116010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 05/20/2022] [Accepted: 05/23/2022] [Indexed: 02/07/2023] Open
Abstract
Cartilage repair has been a challenge in the medical field for many years. Although treatments that alleviate pain and injury are available, none can effectively regenerate the cartilage. Currently, regenerative medicine and tissue engineering are among the developed strategies to treat cartilage injury. The use of stem cells, associated or not with scaffolds, has shown potential in cartilage regeneration. However, it is currently known that the effect of stem cells occurs mainly through the secretion of paracrine factors that act on local cells. In this review, we will address the use of the secretome—a set of bioactive factors (soluble factors and extracellular vesicles) secreted by the cells—of mesenchymal stem cells as a treatment for cartilage regeneration. We will also discuss methodologies for priming the secretome to enhance the chondroregenerative potential. In addition, considering the difficulty of delivering therapies to the injured cartilage site, we will address works that use hydrogels functionalized with growth factors and secretome components. We aim to show that secretome-functionalized hydrogels can be an exciting approach to cell-free cartilage repair therapy.
Collapse
|
15
|
Li M, Yin H, Yan Z, Li H, Wu J, Wang Y, Wei F, Tian G, Ning C, Li H, Gao C, Fu L, Jiang S, Chen M, Sui X, Liu S, Chen Z, Guo Q. The immune microenvironment in cartilage injury and repair. Acta Biomater 2022; 140:23-42. [PMID: 34896634 DOI: 10.1016/j.actbio.2021.12.006] [Citation(s) in RCA: 172] [Impact Index Per Article: 57.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Revised: 12/01/2021] [Accepted: 12/05/2021] [Indexed: 02/07/2023]
Abstract
The ability of articular cartilage to repair itself is limited because it lacks blood vessels, nerves, and lymph tissue. Once damaged, it can lead to joint swelling and pain, accelerating the progression of osteoarthritis. To date, complete regeneration of hyaline cartilage exhibiting mechanical properties remains an elusive goal, despite the many available technologies. The inflammatory milieu created by cartilage damage is critical for chondrocyte death and hypertrophy, extracellular matrix breakdown, ectopic bone formation, and progression of cartilage injury to osteoarthritis. In the inflammatory microenvironment, mesenchymal stem cells (MSCs) undergo aberrant differentiation, and chondrocytes begin to convert or dedifferentiate into cells with a fibroblast phenotype, thereby resulting in fibrocartilage with poor mechanical qualities. All these factors suggest that inflammatory problems may be a major stumbling block to cartilage repair. To produce a milieu conducive to cartilage repair, multi-dimensional management of the joint inflammatory microenvironment in place and time is required. Therefore, this calls for elucidation of the immune microenvironment of cartilage repair after injury. This review provides a brief overview of: (1) the pathogenesis of cartilage injury; (2) immune cells in cartilage injury and repair; (3) effects of inflammatory cytokines on cartilage repair; (4) clinical strategies for treating cartilage defects; and (5) strategies for targeted immunoregulation in cartilage repair. STATEMENT OF SIGNIFICANCE: Immune response is increasingly considered the key factor affecting cartilage repair. It has both negative and positive regulatory effects on the process of regeneration and repair. Proinflammatory factors are secreted in large numbers, and necrotic cartilage is removed. During the repair period, immune cells can secrete anti-inflammatory factors and chondrogenic cytokines, which can inhibit inflammation and promote cartilage repair. However, inflammatory factors persist, which accelerate the degradation of the cartilage matrix. Furthermore, in an inflammatory microenvironment, MSCs undergo abnormal differentiation, and chondrocytes begin to transform or dedifferentiate into fibroblast-like cells, forming fibrocartilage with poor mechanical properties. Consequently, cartilage regeneration requires multi-dimensional regulation of the joint inflammatory microenvironment in space and time to make it conducive to cartilage regeneration.
Collapse
|
16
|
Saygili E, Saglam-Metiner P, Cakmak B, Alarcin E, Beceren G, Tulum P, Kim YW, Gunes K, Eren-Ozcan GG, Akakin D, Sun JY, Yesil-Celiktas O. Bilayered laponite/alginate-poly(acrylamide) composite hydrogel for osteochondral injuries enhances macrophage polarization: An in vivo study. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2022; 134:112721. [DOI: 10.1016/j.msec.2022.112721] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 02/05/2022] [Accepted: 02/14/2022] [Indexed: 12/14/2022]
|
17
|
Adipose Tissue-Derived Mesenchymal Stem Cells as a Potential Restorative Treatment for Cartilage Defects: A PRISMA Review and Meta-Analysis. Pharmaceuticals (Basel) 2021; 14:ph14121280. [PMID: 34959680 PMCID: PMC8705514 DOI: 10.3390/ph14121280] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 12/03/2021] [Accepted: 12/04/2021] [Indexed: 12/13/2022] Open
Abstract
Cartilage defects are a predisposing factor for osteoarthritis. Conventional therapies are mostly palliative and there is an interest in developing newer therapies that target the disease’s progression. Mesenchymal stem cells (MSCs) have been suggested as a promising therapy to restore hyaline cartilage to cartilage defects, though the optimal cell source has remained under investigation. A PRISMA systematic review was conducted utilising five databases (MEDLINE, EMBASE, Cochrane Library, Scopus, Web of Science) which identified nineteen human studies that used adipose tissue-derived MSC (AMSC)-based therapies, including culture-expanded AMSCs and stromal vascular fraction, to treat cartilage defects. Clinical, imaging and histological outcomes, as well as other relevant details pertaining to cartilage regeneration, were extracted from each study. Pooled analysis revealed a significant improvement in WOMAC scores (mean difference: −25.52; 95%CI (−30.93, −20.10); p < 0.001), VAS scores (mean difference: −3.30; 95%CI (−3.72, −2.89); p < 0.001), KOOS scores and end point MOCART score (mean: 68.12; 95%CI (62.18, 74.05)), thus showing improvement. The studies in this review demonstrate the safety and efficacy of AMSC-based therapies for cartilage defects. Establishing standardised methods for MSC extraction and delivery, and performing studies with long follow-up should enable future high-quality research to provide the evidence needed to bring AMSC-based therapies into the market.
Collapse
|
18
|
Ribitsch I, Bileck A, Egerbacher M, Gabner S, Mayer RL, Janker L, Gerner C, Jenner F. Fetal Immunomodulatory Environment Following Cartilage Injury-The Key to CARTILAGE Regeneration? Int J Mol Sci 2021; 22:ijms222312969. [PMID: 34884768 PMCID: PMC8657887 DOI: 10.3390/ijms222312969] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 11/27/2021] [Accepted: 11/28/2021] [Indexed: 01/15/2023] Open
Abstract
Fetal cartilage fully regenerates following injury, while in adult mammals cartilage injury leads to osteoarthritis (OA). Thus, in this study, we compared the in vivo injury response of fetal and adult ovine articular cartilage histologically and proteomically to identify key factors of fetal regeneration. In addition, we compared the secretome of fetal ovine mesenchymal stem cells (MSCs) in vitro with injured fetal cartilage to identify potential MSC-derived therapeutic factors. Cartilage injury caused massive cellular changes in the synovial membrane, with macrophages dominating the fetal, and neutrophils the adult, synovial cellular infiltrate. Correspondingly, proteomics revealed differential regulation of pro- and anti-inflammatory mediators and growth-factors between adult and fetal joints. Neutrophil-related proteins and acute phase proteins were the two major upregulated protein groups in adult compared to fetal cartilage following injury. In contrast, several immunomodulating proteins and growth factors were expressed significantly higher in the fetus than the adult. Comparison of the in vitro MSCs proteome with the in vivo fetal regenerative signature revealed shared upregulation of 17 proteins, suggesting their therapeutic potential. Biomimicry of the fetal paracrine signature to reprogram macrophages and modulate inflammation could be an important future research direction for developing novel therapeutics.
Collapse
Affiliation(s)
- Iris Ribitsch
- VETERM, Equine Surgery Unit, Department of Companion Animals and Horses, University of Veterinary Medicine Vienna, 1210 Vienna, Austria;
| | - Andrea Bileck
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, 1090 Vienna, Austria; (A.B.); (R.L.M.); (L.J.)
| | - Monika Egerbacher
- Administrative Unit Veterinary Medicine, UMIT—Private University for Health Sciences, Medical Informatics and Technology GmbH, 6060 Hall in Tirol, Austria;
| | - Simone Gabner
- Histology & Embryology, Department of Pathobiology, University of Veterinary Medicine, 1210 Vienna, Austria;
| | - Rupert L. Mayer
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, 1090 Vienna, Austria; (A.B.); (R.L.M.); (L.J.)
| | - Lukas Janker
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, 1090 Vienna, Austria; (A.B.); (R.L.M.); (L.J.)
| | - Christopher Gerner
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, 1090 Vienna, Austria; (A.B.); (R.L.M.); (L.J.)
- Correspondence: (C.G.); (F.J.)
| | - Florien Jenner
- VETERM, Equine Surgery Unit, Department of Companion Animals and Horses, University of Veterinary Medicine Vienna, 1210 Vienna, Austria;
- Correspondence: (C.G.); (F.J.)
| |
Collapse
|
19
|
Jiang X, Zhong R, Dai W, Huang H, Yu Q, Zhang JA, Cai Y. Exploring Diagnostic Biomarkers and Comorbid Pathogenesis for Osteoarthritis and Metabolic Syndrome via Bioinformatics Approach. Int J Gen Med 2021; 14:6201-6213. [PMID: 34616175 PMCID: PMC8487858 DOI: 10.2147/ijgm.s325561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 09/14/2021] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Metabolic syndrome (MS) has grown in recognition to contribute to the pathogenesis of osteoarthritis (OA), which is the most prevalent arthritis characterized by joint dysfunction. However, the specific mechanism between OA and MS remains unclear. METHODS The gene expression profiles and clinical information data of OA and MS were retrieved from the Gene Expression Omnibus (GEO) database. The genes in the key module of MS were identified by weighted gene co-expression network analysis (WGCNA), which intersected with the differentially expressed genes (DEGs) between control and MS samples to obtain hub genes for MS. The potential functions and pathways of hub genes were detected through the Gene Ontology (GO) and Kyoto Encyclopedia of Gene and Genome (KEGG) analyses. The genes involved in the different KEGG pathways between the control and OA samples overlapped with the DEGs between the two groups via the Venn analysis to gain the hub genes for OA affected by MS (MOHGs). Additionally, the least absolute shrinkage and selection operator (LASSO) was performed on the MOHGs to establish a diagnostic model for each disease. RESULTS A total of 61 hub genes for MS were identified that significantly enriched in platelet activation, complement and coagulation cascades, and hematopoietic cell lineage. Besides, 4 candidate genes (ELOVL7, F2RL3, GP9, and ITGA2B) were screened among the 6 MOHGs to construct a diagnostic model, showing good performance for distinguishing controls from patients with MS and OA. GSEA suggested that these diagnostic genes were closely associated with immune response, adipocytokine signaling, fatty acid metabolism, cell cycle, and platelet activation. CONCLUSION Taken together, we identified 4 potential gene biomarkers for diagnosing MS and OA patients, providing a theoretical basis and reference for the diagnostics and treatment targets of MS and OA.
Collapse
Affiliation(s)
- Xiang Jiang
- Department of Orthopaedics and Rehabilitation, Shanghai Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Tongji University School of Medicine, Shanghai, 201619, People’s Republic of China
| | - Rongzhou Zhong
- Department of Orthopaedics and Rehabilitation, Shanghai Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Tongji University School of Medicine, Shanghai, 201619, People’s Republic of China
| | - Weifan Dai
- Department of Digital Hub, Decathlon International, Shanghai, 200131, People’s Republic of China
| | - Hui Huang
- Department of Orthopaedics and Rehabilitation, Shanghai Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Tongji University School of Medicine, Shanghai, 201619, People’s Republic of China
| | - Qinyuan Yu
- Department of Orthopaedics and Rehabilitation, Shanghai Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Tongji University School of Medicine, Shanghai, 201619, People’s Republic of China
| | - Jiji Alexander Zhang
- Department of Medicine, Heidelberg University Hospital, University of Heidelberg, Heidelberg, 69120, Germany
| | - Yanrong Cai
- Department of Medicine, Heidelberg University Hospital, University of Heidelberg, Heidelberg, 69120, Germany
| |
Collapse
|
20
|
Neefjes M, Housmans BAC, van Beuningen HM, Vitters EL, van den Akker GGH, Welting TJM, van Caam APM, van der Kraan PM. Prediction of the Effect of the Osteoarthritic Joint Microenvironment on Cartilage Repair. Tissue Eng Part A 2021; 28:27-37. [PMID: 34039008 DOI: 10.1089/ten.tea.2021.0051] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Osteoarthritis (OA) is characterized by progressive articular cartilage loss. Human mesenchymal stromal cells (MSCs) can be used for cartilage repair therapies based on their potential to differentiate into chondrocytes. However, the joint microenvironment is a major determinant of the success of MSC-based cartilage formation. Currently, there is no tool that is able to predict the effect of a patient's OA joint microenvironment on MSC-based cartilage formation. Our goal was to develop a molecular tool that can predict this effect before the start of cartilage repair therapies. Six different promoter reporters (hIL6, hIL8, hADAMTS5, hWISP1, hMMP13, and hADAM28) were generated and evaluated in an immortalized human articular chondrocyte for their responsiveness to an osteoarthritic microenvironment by stimulation with OA synovium-conditioned medium (OAs-cm) obtained from 32 different knee OA patients. To study the effect of this OA microenvironment on MSC-based cartilage formation, MSCs were cultured in a three-dimensional pellet culture model, while stimulated with OAs-cm. Cartilage formation was assessed histologically and by quantifying sulfated glycosaminoglycan (sGAG) production. We confirmed that OAs-cm of different patients had significantly different effects on sGAG production. In addition, significant correlations were obtained between the effect of the OAs-cm on cartilage formation and promoter reporter outcome. Furthermore, we validated the predictive value of measuring two promoter reporters with an independent cohort of OAs-cm and the effect of 87.5% of the OAs-cm on MSC-based cartilage formation could be predicted. Together, we developed a novel tool to predict the effect of the OA joint microenvironment on MSC-based cartilage formation. This is an important first step toward personalized cartilage repair strategies for OA patients.
Collapse
Affiliation(s)
- Margot Neefjes
- Experimental Rheumatology, Department of Rheumatology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Bas A C Housmans
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Henk M van Beuningen
- Experimental Rheumatology, Department of Rheumatology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Elly L Vitters
- Experimental Rheumatology, Department of Rheumatology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Guus G H van den Akker
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Tim J M Welting
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Arjan P M van Caam
- Experimental Rheumatology, Department of Rheumatology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Peter M van der Kraan
- Experimental Rheumatology, Department of Rheumatology, Radboud University Medical Center, Nijmegen, the Netherlands
| |
Collapse
|
21
|
Jiang G, Li S, Yu K, He B, Hong J, Xu T, Meng J, Ye C, Chen Y, Shi Z, Feng G, Chen W, Yan S, He Y, Yan R. A 3D-printed PRP-GelMA hydrogel promotes osteochondral regeneration through M2 macrophage polarization in a rabbit model. Acta Biomater 2021; 128:150-162. [PMID: 33894346 DOI: 10.1016/j.actbio.2021.04.010] [Citation(s) in RCA: 135] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 03/25/2021] [Accepted: 04/01/2021] [Indexed: 12/17/2022]
Abstract
Osteochondral regeneration is an orchestrated process of inflammatory immunity, host cell response, and implant degradation in tissue engineering. Here, the effects of a platelet-rich plasma (PRP)-gelatin methacryloyl (GelMA) hydrogel scaffold fabricated using the digital micro-mirror device (DMD) technique for osteochondral repair were investigated in a rabbit model. GelMA hydrogels with different PRP concentrations were fabricated, and their roles in bone marrow mesenchymal stem cells (BMSCs) and macrophage polarization in vitro were investigated. The incorporation of 20% PRP into the hydrogel showed optimal effects on the proliferation, migration, and osteogenic and chondrogenic differentiation of BMSCs. The 20% PRP-GelMA (v/v) hydrogel also promoted M2 polarization with high expression of Arg1 and CD206. Compared to the 20% PRP group, the 50% PRP group showed similar biological roles in BMSCs but less extent of osteogenesis. In the vivo study, the 20% PRP-GelMA composite was used for osteochondral reconstruction and showed more cartilage and subchondral bone regeneration than that observed using the pure GelMA hydrogel. The PRP-GelMA group exhibited more M2 macrophage infiltration and less M1 macrophage presentation at three time points as compared to the nontreatment group. The expression of Arg1 in the PRP-GelMA group increased significantly at 6 weeks but decreased to a lower level at 12 weeks, while CD163 showed sustained high expression until 18 weeks. Our findings demonstrated that the 3D-printed PRP-GelMA composite could promote osteochondral repair through immune regulation by M2 polarization and could be a potential candidate for osteochondral tissue engineering. STATEMENT OF SIGNIFICANCE: PRP-GelMA hydrogels promoted the migration and osteogenic and chondrogenic differentiation of BMSCs. PRP-GelMA hydrogels participated in immune regulation and M1-to-M2 transition of macrophages. PRP-GelMA hydrogels coordinated and promoted several overlapping osteochondral repair events, including dynamic immune regulation, chemotaxis of MSCs, and osteochondral differentiation. PRP-GelMA hydrogels showed superior cartilage and subchondral bone repair properties.
Collapse
|
22
|
Bartolotti I, Roseti L, Petretta M, Grigolo B, Desando G. A Roadmap of In Vitro Models in Osteoarthritis: A Focus on Their Biological Relevance in Regenerative Medicine. J Clin Med 2021; 10:1920. [PMID: 33925222 PMCID: PMC8124812 DOI: 10.3390/jcm10091920] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 04/20/2021] [Accepted: 04/26/2021] [Indexed: 01/15/2023] Open
Abstract
Osteoarthritis (OA) is a multifaceted musculoskeletal disorder, with a high prevalence worldwide. Articular cartilage and synovial membrane are among the main biological targets in the OA microenvironment. Gaining more knowledge on the accuracy of preclinical in vitro OA models could open innovative avenues in regenerative medicine to bridge major gaps, especially in translation from animals to humans. Our methodological approach entailed searches on Scopus, the Web of Science Core Collection, and EMBASE databases to select the most relevant preclinical in vitro models for studying OA. Predicting the biological response of regenerative strategies requires developing relevant preclinical models able to mimic the OA milieu influencing tissue responses and organ complexity. In this light, standard 2D culture models lack critical properties beyond cell biology, while animal models suffer from several limitations due to species differences. In the literature, most of the in vitro models only recapitulate a tissue compartment, by providing fragmented results. Biotechnological advances may enable scientists to generate new in vitro models that combine easy manipulation and organ complexity. Here, we review the state-of-the-art of preclinical in vitro models in OA and outline how the different preclinical systems (inflammatory/biomechanical/microfluidic models) may be valid tools in regenerative medicine, describing their pros and cons. We then discuss the prospects of specific and combinatorial models to predict biological responses following regenerative approaches focusing on mesenchymal stromal cells (MSCs)-based therapies to reduce animal testing.
Collapse
Affiliation(s)
- Isabella Bartolotti
- Laboratorio RAMSES, IRCCS Istituto Ortopedico Rizzoli, via di Barbiano 1/10, 40136 Bologna, Italy; (I.B.); (M.P.); (B.G.)
| | - Livia Roseti
- Laboratorio RAMSES, IRCCS Istituto Ortopedico Rizzoli, via di Barbiano 1/10, 40136 Bologna, Italy; (I.B.); (M.P.); (B.G.)
| | - Mauro Petretta
- Laboratorio RAMSES, IRCCS Istituto Ortopedico Rizzoli, via di Barbiano 1/10, 40136 Bologna, Italy; (I.B.); (M.P.); (B.G.)
- RegenHu Company, Z.I Du Vivier 22, 1690 Villaz-St-Pierre, Switzerland
| | - Brunella Grigolo
- Laboratorio RAMSES, IRCCS Istituto Ortopedico Rizzoli, via di Barbiano 1/10, 40136 Bologna, Italy; (I.B.); (M.P.); (B.G.)
| | - Giovanna Desando
- Laboratorio RAMSES, IRCCS Istituto Ortopedico Rizzoli, via di Barbiano 1/10, 40136 Bologna, Italy; (I.B.); (M.P.); (B.G.)
| |
Collapse
|
23
|
Bansal H, Leon J, Pont JL, Wilson DA, Bansal A, Agarwal D, Preoteasa I. Platelet-rich plasma (PRP) in osteoarthritis (OA) knee: Correct dose critical for long term clinical efficacy. Sci Rep 2021; 11:3971. [PMID: 33597586 PMCID: PMC7889864 DOI: 10.1038/s41598-021-83025-2] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 01/22/2021] [Indexed: 12/11/2022] Open
Abstract
Despite encouraging results reported with regards to Platelet-rich plasma (PRP) application in osteoarthritis (OA) knee, still critical issues like conclusive structural evidence of its efficacy, standard dose and good manual method of preparation to obtain high yield remains unanswered. Present study is an attempt to optimise the dose and concentration of therapeutic PRP and its correlation with structural, physiologic efficacy with a new manual method of PRP preparation. A total of one hundred and fifty patients were randomized to receive either PRP (10 billion platelets) or hyaluronic acid (HA; 4 ml; 75 patients in each group) and followed up till 1 year. An addition of filtration step with 1 µm filter in manual PRP processing improved platelet recovery upto 90%. Significant improvements in WOMAC (51.94 ± 7.35 vs. 57.33 ± 8.92; P < 0.001), IKDC scores (62.8 ± 6.24 vs 52.7 ± 6.39; P < 0.001), 6-min pain free walking distance (+ 120 vs. + 4; P < 0.001) persisted in PRP compared to HA group at 1 year. Significant decline IL-6 and TNF-α levels observed in PRP group (P < 0.05) compared to HA at 1 month. Study demonstrated that an absolute count of 10 billion platelets is crucial in a PRP formulation to have long sustained chondroprotective effect upto one year in moderate knee OA.
Collapse
Affiliation(s)
- Himanshu Bansal
- Mother Cell Spinal Injury and Stem Cell Research, Anupam Hospital, Rudrapur, Uttarakhand, India.
| | - Jerry Leon
- PMR Advance Health Institute Mayaguez, Puerto Rico, USA
| | | | | | - Anupama Bansal
- Mother Cell Spinal Injury and Stem Cell Research, Anupam Hospital, Rudrapur, Uttarakhand, India
| | | | | |
Collapse
|
24
|
Regenerative Medicine for Equine Musculoskeletal Diseases. Animals (Basel) 2021; 11:ani11010234. [PMID: 33477808 PMCID: PMC7832834 DOI: 10.3390/ani11010234] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/08/2021] [Accepted: 01/15/2021] [Indexed: 01/15/2023] Open
Abstract
Simple Summary Lameness due to musculoskeletal disease is the most common diagnosis in equine veterinary practice. Many of these orthopaedic disorders are chronic problems, for which no clinically satisfactory treatment exists. Thus, high hopes are pinned on regenerative medicine, which aims to replace or regenerate cells, tissues, or organs to restore or establish normal function. Some regenerative medicine therapies have already made their way into equine clinical practice mainly to treat tendon injures, tendinopathies, cartilage injuries and degenerative joint disorders with promising but diverse results. This review summarises the current knowledge of commonly used regenerative medicine treatments and critically discusses their use. Abstract Musculoskeletal injuries and chronic degenerative diseases commonly affect both athletic and sedentary horses and can entail the end of their athletic careers. The ensuing repair processes frequently do not yield fully functional regeneration of the injured tissues but biomechanically inferior scar or replacement tissue, causing high reinjury rates, degenerative disease progression and chronic morbidity. Regenerative medicine is an emerging, rapidly evolving branch of translational medicine that aims to replace or regenerate cells, tissues, or organs to restore or establish normal function. It includes tissue engineering but also cell-based and cell-free stimulation of endogenous self-repair mechanisms. Some regenerative medicine therapies have made their way into equine clinical practice mainly to treat tendon injures, tendinopathies, cartilage injuries and degenerative joint disorders with promising results. However, the qualitative and quantitative spatiotemporal requirements for specific bioactive factors to trigger tissue regeneration in the injury response are still unknown, and consequently, therapeutic approaches and treatment results are diverse. To exploit the full potential of this burgeoning field of medicine, further research will be required and is ongoing. This review summarises the current knowledge of commonly used regenerative medicine treatments in equine patients and critically discusses their use.
Collapse
|
25
|
Ghosh B, Kirtania MD. Clinical applications of biopolymer-based hydrogels. PLANT AND ALGAL HYDROGELS FOR DRUG DELIVERY AND REGENERATIVE MEDICINE 2021:535-568. [DOI: 10.1016/b978-0-12-821649-1.00015-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
26
|
Down-regulated ciRS-7/up-regulated miR-7 axis aggravated cartilage degradation and autophagy defection by PI3K/AKT/mTOR activation mediated by IL-17A in osteoarthritis. Aging (Albany NY) 2020; 12:20163-20183. [PMID: 33099538 PMCID: PMC7655186 DOI: 10.18632/aging.103731] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Accepted: 06/25/2020] [Indexed: 12/13/2022]
Abstract
Osteoarthritis (OA) is one of the most painful and widespread chronic degenerative joint diseases and is characterized by destructed articular cartilage and inflamed joints. Previously, our findings indicated that circular RNA ciRS-7 (ciRS-7)/microRNA 7 (miR-7) axis is abnormally expressed in OA, and regulates proliferation, inflammatory responses, and apoptosis of interleukin-1β (IL-1β)-stimulated chondrocytes. However, its underlying role in OA remains unknown. In this study, we first validated cartilage degradation and defection of autophagy in samples of OA patients. IL-1β initially stimulated autophagy of chondrocytes, and ultimately significantly suppressed autophagy. Upregulated ciRS-7/down-regulated miR-7 aggravated IL-1β-induced cartilage degradation, and restrained autophagy in vitro. Gene sequencing and bioinformatics analysis performed on a control group, IL-1β group, and IL-1β+miR-7-mimics group demonstrated that seven of the most significant mRNA candidates were enriched in the interleukin-17 (IL-17) signaling pathway. Increased IL-17A levels were also observed by qRT-PCR and ELISA. In addition, it was revealed that the ciRS-7/miR-7 axis ameliorated cartilage degradation and defection of autophagy by PI3K/AKT/mTOR activation in IL-1β-induced chondrocytes. Furthermore, an OA model was established in rats with medial meniscus destabilization. miR-7-siRNA-expressing lentiviruses alleviated surgical resection-induced cartilage destruction of OA mice, whereas miR-7 mimics worsened the effects. Thus, these findings revealed that the mechanism of the ciRS-7/miR-7 axis involved regulating OA progression and provided valuable directions for OA treatment.
Collapse
|
27
|
The Immune Cell Landscape in Different Anatomical Structures of Knee in Osteoarthritis: A Gene Expression-Based Study. BIOMED RESEARCH INTERNATIONAL 2020; 2020:9647072. [PMID: 32258161 PMCID: PMC7106908 DOI: 10.1155/2020/9647072] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 11/10/2019] [Accepted: 01/04/2020] [Indexed: 01/10/2023]
Abstract
Background Immunological mechanisms play a vital role in the pathogenesis of knee osteoarthritis (KOA). Moreover, the immune phenotype is a relevant prognostic factor in various immune-related diseases. In this study, we used CIBERSORT for deconvolution of global gene expression data to define the immune cell landscape of different structures of knee in osteoarthritis. Methods and Findings. By applying CIBERSORT, we assessed the relative proportions of immune cells in 76 samples of knee cartilage, 146 samples of knee synovial tissue, 40 samples of meniscus, and 50 samples of knee subchondral bone. Enumeration and activation status of 22 immune cell subtypes were provided by the obtained immune cell profiles. In synovial tissues, the differences in proportions of plasma cells, M1 macrophages, M2 macrophages, activated dendritic cells, resting mast cells, and eosinophils between normal tissues and osteoarthritic tissues were statistically significant (P < 0.05). The area under the curve was relatively large in resting mast cells, dendritic cells, and M2 macrophages in receiver operating characteristic analyses. In subchondral bones, the differences in proportions of resting master cells and neutrophils between normal tissues and osteoarthritic tissues were statistically significant (P < 0.05). In subchondral bones, the proportions of immune cells, from the principle component analyses, displayed distinct group-bias clustering. Resting mast cells and T cell CD8 were the major component of first component. Moreover, we revealed the potential interaction between immune cells. There was almost no infiltration of immune cells in the meniscus and cartilage of the knee joint. Conclusions The immune cell composition in KOA differed substantially from that of healthy joint tissue, while it also differed in different anatomical structures of the knee. Meanwhile, activated mast cells were mainly associated with high immune cell infiltration in OA. Furthermore, we speculate M2 macrophages in synovium and mast cells in subchondral bone may play an important role in the pathogenesis of OA.
Collapse
|
28
|
Chen C, Cui S, Li W, Jin H, Fan J, Sun Y, Cui Z. Ingenuity pathway analysis of human facet joint tissues: Insight into facet joint osteoarthritis. Exp Ther Med 2020; 19:2997-3008. [PMID: 32256786 PMCID: PMC7086291 DOI: 10.3892/etm.2020.8555] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2019] [Accepted: 01/30/2020] [Indexed: 12/14/2022] Open
Abstract
Facet joint osteoarthritis (FJOA) is a common degenerative joint disorder with high prevalence in the elderly. FJOA causes lower back pain and lower extremity pain, and thus severely impacts the quality of life of affected patients. Emerging studies have focused on the histomorphological and histomorphometric changes in FJOA. However, the dynamic genetic changes in FJOA have remained to be clearly determined. In the present study, previously obtained RNA deep sequencing data were subjected to an ingenuity pathway analysis (IPA) and canonical signaling pathways of differentially expressed genes (DEGs) in FJOA were studied. The top 25 enriched canonical signaling pathways were identified and canonical signaling pathways with high absolute values of z-scores, specifically leukocyte extravasation signaling, Tec kinase signaling and osteoarthritis pathway, were investigated in detail. DEGs were further categorized by disease, biological function and toxicity (tox) function. The genetic networks between DEGs as well as hub genes in these functional networks were also investigated. It was demonstrated that C-X-C motif chemokine ligand 8, elastase, neutrophil expressed, growth factor independent 1 transcriptional repressor, Spi-1 proto-oncogene, CCAAT enhancer binding protein epsilon, GATA binding protein 1, TAL bHLH transcription factor 1, erythroid differentiation factor, minichromosome maintenance complex component 4, BTG anti-proliferation factor 2, BRCA1 DNA repair-associated, cyclin D1, chromatin assembly factor 1 subunit A, triggering receptor expressed on myeloid cells 1 and tumor protein p63 were hub genes in the top 5 IPA networks (with a score >30). The present study provides insight into the pathological processes of FJOA from a genetic perspective and may thus benefit the clinical treatment of FJOA.
Collapse
Affiliation(s)
- Chu Chen
- Department of Spine Surgery, The Second Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Shengyu Cui
- Department of Spine Surgery, The Second Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Weidong Li
- Department of Spine Surgery, The Second Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Huricha Jin
- Department of Spine Surgery, The Second Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Jianbo Fan
- Department of Spine Surgery, The Second Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Yuyu Sun
- Department of Spine Surgery, The Second Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Zhiming Cui
- Department of Spine Surgery, The Second Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| |
Collapse
|
29
|
Inflammation-Modulating Hydrogels for Osteoarthritis Cartilage Tissue Engineering. Cells 2020; 9:cells9020419. [PMID: 32059502 PMCID: PMC7072320 DOI: 10.3390/cells9020419] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 02/03/2020] [Accepted: 02/11/2020] [Indexed: 12/30/2022] Open
Abstract
Osteoarthritis (OA) is the most common form of the joint disease associated with age, obesity, and traumatic injury. It is a disabling degenerative disease that affects synovial joints and leads to cartilage deterioration. Despite the prevalence of this disease, the understanding of OA pathophysiology is still incomplete. However, the onset and progression of OA are heavily associated with the inflammation of the joint. Therefore, studies on OA treatment have sought to intra-articularly deliver anti-inflammatory drugs, proteins, genes, or cells to locally control inflammation in OA joints. These therapeutics have been delivered alone or increasingly, in delivery vehicles for sustained release. The use of hydrogels in OA treatment can extend beyond the delivery of anti-inflammatory components to have inherent immunomodulatory function via regulating immune cell polarization and activity. Currently, such immunomodulatory biomaterials are being developed for other applications, which can be translated into OA therapy. Moreover, anabolic and proliferative levels of OA chondrocytes are low, except initially, when chondrocytes temporarily increase anabolism and proliferation in response to structural changes in their extracellular environment. Therefore, treatments need to restore matrix protein synthesis and proliferation to healthy levels to reverse OA-induced damage. In conjugation with injectable and/or adhesive hydrogels that promote cartilage tissue regeneration, immunomodulatory tissue engineering solutions will have robust potential in OA treatment. This review describes the disease, its current and future immunomodulatory therapies as well as cartilage-regenerative injectable and adhesive hydrogels.
Collapse
|
30
|
Ji X, Yuan X, Ma L, Bi B, Zhu H, Lei Z, Liu W, Pu H, Jiang J, Jiang X, Zhang Y, Xiao J. Mesenchymal stem cell-loaded thermosensitive hydroxypropyl chitin hydrogel combined with a three-dimensional-printed poly(ε-caprolactone) /nano-hydroxyapatite scaffold to repair bone defects via osteogenesis, angiogenesis and immunomodulation. Theranostics 2020; 10:725-740. [PMID: 31903147 PMCID: PMC6929983 DOI: 10.7150/thno.39167] [Citation(s) in RCA: 121] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 10/08/2019] [Indexed: 12/01/2022] Open
Abstract
Chitin-derived hydrogels are commonly used in bone regeneration because of their high cell compatibility; however, their poor mechanical properties and little knowledge of the interaction between the materials and host cells have limited their practical application. Methods: To evaluate osteoinductivity and enhance the mechanical properties of a newly synthesized thermosensitive hydroxypropyl chitin hydrogel (HPCH), a mesenchymal stem cell (MSC)-encapsulated HPCH was infused into a three-dimensional-printed poly (ε-caprolactone) (PCL)/ nano-hydroxyapatite (nHA) scaffold to form a hybrid scaffold. The mechanical properties and cell compatibility of the scaffold were tested. The interaction between macrophages and scaffold for angiogenesis and osteogenesis were explored in vitro and in vivo. Results: The hybrid scaffold showed improved mechanical properties and high cell viability. When MSCs were encapsulated in HPCH, osteo-differentiation was promoted properly via endochondral ossification. The co-culture experiments showed that the hybrid scaffold facilitated growth factor secretion from macrophages, thus promoting vascularization and osteoinduction. The Transwell culture proved that MSCs modulated the inflammatory response of HPCH. Additionally, subcutaneous implantation of MSC-encapsulated HPCH confirmed M2 activation. In situ evaluation of calvarial defects confirmed that the repair was optimal in the MSC-loaded HPCH + PCL/nHA group. Conclusions: PCL/nHA + HPCH hybrid scaffolds effectively promoted vascularization and osteoinduction via osteogenesis promotion and immunomodulation, which suggests promising applications for bone regeneration.
Collapse
Affiliation(s)
- Xiongfa Ji
- Department of Orthopedics, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, PR China
- Department of Orthopaedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xi Yuan
- Department of Orthopaedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Limin Ma
- Department of Orthopedics, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, PR China
| | - Bo Bi
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan, 430072, China
| | - Hao Zhu
- Department of Orthopaedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Zehua Lei
- Department of Orthopaedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Wenbin Liu
- Department of Orthopaedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - HongXu Pu
- Department of Orthopaedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jiawei Jiang
- Department of Orthopaedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xulin Jiang
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan, 430072, China
| | - Yu Zhang
- Department of Orthopedics, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, PR China
| | - Jun Xiao
- Department of Orthopaedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| |
Collapse
|
31
|
A comprehensive characterisation of large-scale expanded human bone marrow and umbilical cord mesenchymal stem cells. Stem Cell Res Ther 2019; 10:99. [PMID: 30885254 PMCID: PMC6421680 DOI: 10.1186/s13287-019-1202-4] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 02/28/2019] [Accepted: 03/01/2019] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND The manufacture of mesenchymal stem/stromal cells (MSCs) for clinical use needs to be cost effective, safe and scaled up. Current methods of expansion on tissue culture plastic are labour-intensive and involve several 'open' procedures. We have used the closed Quantum® hollow fibre bioreactor to expand four cultures each of MSCs derived from bone marrow (BM) and, for the first time, umbilical cords (UCs) and assessed extensive characterisation profiles for each, compared to parallel cultures grown on tissue culture plastic. METHODS Bone marrow aspirate was directly loaded into the Quantum®, and cells were harvested and characterised at passage (P) 0. Bone marrow cells were re-seeded into the Quantum®, harvested and further characterised at P1. UC-MSCs were isolated enzymatically and cultured once on tissue culture plastic, before loading cells into the Quantum®, harvesting and characterising at P1. Quantum®-derived cultures were phenotyped in terms of immunoprofile, tri-lineage differentiation, response to inflammatory stimulus and telomere length, as were parallel cultures expanded on tissue culture plastic. RESULTS Bone marrow cell harvests from the Quantum® were 23.1 ± 16.2 × 106 in 14 ± 2 days (P0) and 131 ± 84 × 106 BM-MSCs in 13 ± 1 days (P1), whereas UC-MSC harvests from the Quantum® were 168 ± 52 × 106 UC-MSCs after 7 ± 2 days (P1). Quantum®- and tissue culture plastic-expanded cultures at P1 adhered to criteria for MSCs in terms of cell surface markers, multipotency and plastic adherence, whereas the integrins, CD29, CD49c and CD51/61, were found to be elevated on Quantum®-expanded BM-MSCs. Rapid culture expansion in the Quantum® did not cause shortened telomeres when compared to cultures on tissue culture plastic. Immunomodulatory gene expression was variable between donors but showed that all MSCs upregulated indoleamine 2, 3-dioxygenase (IDO). CONCLUSIONS The results presented here demonstrate that the Quantum® can be used to expand large numbers of MSCs from bone marrow and umbilical cord tissues for next-generation large-scale manufacturing, without impacting on many of the properties that are characteristic of MSCs or potentially therapeutic. Using the Quantum®, we can obtain multiple MSC doses from a single manufacturing run to treat many patients. Together, our findings support the development of cheaper cell-based treatments.
Collapse
|
32
|
Lolli A, Colella F, De Bari C, van Osch GJVM. Targeting anti-chondrogenic factors for the stimulation of chondrogenesis: A new paradigm in cartilage repair. J Orthop Res 2019; 37:12-22. [PMID: 30175861 DOI: 10.1002/jor.24136] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 08/09/2018] [Indexed: 02/04/2023]
Abstract
Trauma and age-related cartilage disorders represent a major global cause of morbidity, resulting in chronic pain and disability in patients. A lack of effective therapies, together with a rapidly aging population, creates an impressive clinical and economic burden on healthcare systems. In this scenario, experimental therapies based on transplantation or in situ stimulation of skeletal Mesenchymal Stem/progenitor Cells (MSCs) have raised great interest for cartilage repair. Nevertheless, the challenge of guiding MSC differentiation and preventing cartilage hypertrophy and calcification still needs to be overcome. While research has mostly focused on the stimulation of cartilage anabolism using growth factors, several issues remain unresolved prompting the field to search for novel solutions. Recently, inhibition of anti-chondrogenic regulators has emerged as an intriguing opportunity. Anti-chondrogenic regulators include extracellular proteins as well as intracellular transcription factors and microRNAs that act as potent inhibitors of pro-chondrogenic signals. Suppression of these inhibitors can enhance MSC chondrogenesis and production of cartilage matrix. We here review the current knowledge concerning different types of anti-chondrogenic regulators. We aim to highlight novel therapeutic targets for cartilage repair and discuss suitable tools for suppressing their anti-chondrogenic functions. Further effort is needed to unveil the therapeutic perspectives of this approach and pave the way for effective treatment of cartilage injuries in patients. © 2018 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res.
Collapse
Affiliation(s)
- Andrea Lolli
- Department of Orthopaedics, Erasmus MC, University Medical Center, Wytemaweg 80, 3015CN Rotterdam, the Netherlands
| | - Fabio Colella
- Arthritis and Regenerative Medicine Laboratory, Aberdeen Centre for Arthritis and Musculoskeletal Health, Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Cosimo De Bari
- Arthritis and Regenerative Medicine Laboratory, Aberdeen Centre for Arthritis and Musculoskeletal Health, Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Gerjo J V M van Osch
- Department of Orthopaedics, Erasmus MC, University Medical Center, Wytemaweg 80, 3015CN Rotterdam, the Netherlands.,Department of Otorhinolaryngology, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
33
|
Kargozar S, Baino F, Hoseini SJ, Hamzehlou S, Darroudi M, Verdi J, Hasanzadeh L, Kim HW, Mozafari M. Biomedical applications of nanoceria: new roles for an old player. Nanomedicine (Lond) 2018; 13:3051-3069. [PMID: 30507347 DOI: 10.2217/nnm-2018-0189] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2018] [Accepted: 09/26/2018] [Indexed: 02/08/2023] Open
Abstract
The use of different biomaterials with the ability to accelerate the repair and regeneration processes is of great importance in tissue engineering strategies. On this point, cerium oxide nanoparticles (CNPs or nanoceria) have recently attracted much attention due to their excellent biological properties including anti-oxidant, anti-inflammation and antibacterial activities as well as high angiogenic potential. The results of incorporation of these nano-sized particles into various constructs and scaffolds designed for tissue engineering applications have proven the success of this strategy in terms of improving healing process of different tissues. In this review, we first summarize the physicochemical and biological properties of nanoceria in brief and then present its usability in tissue engineering strategies based on the currently available published reports.
Collapse
Affiliation(s)
- Saeid Kargozar
- Department of Modern Sciences & Technologies, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Francesco Baino
- Institute of Materials Physics & Engineering, Department of Applied Science & Technology (DISAT), Politecnico di Torino, Torino, Italy
| | - Seyed Javad Hoseini
- Department of Medical Biotechnology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sepideh Hamzehlou
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Medical Genetics Network (MeGeNe), Universal Scientific Education & Research Network (USERN), Tehran, Iran
| | - Majid Darroudi
- Nuclear Medicine Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Javad Verdi
- Tissue Engineering & Applied Cell Sciences Department, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Leila Hasanzadeh
- Department of Medical Biotechnology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hae-Won Kim
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, Republic of Korea
- Department of Nanobiomedical Science & BK21 PLUS NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, Republic of Korea
- Department of Biomaterials Science, School of Dentistry, Dankook University, Cheonan, Republic of Korea
| | - Masoud Mozafari
- Bioengineering Research Group, Nanotechnology & Advanced Materials Department, Materials & Energy Research Center (MERC), Tehran, Iran
- Cellular & Molecular Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran
- Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran
| |
Collapse
|
34
|
Abstract
PURPOSE OF REVIEW To assess the utilization and efficacy of platelet-rich plasma (PRP), for the treatment of articular cartilage injury, most commonly characterized by progressive pain and loss of joint function in the setting of osteoarthritis (OA). RECENT FINDINGS PRP modulates the inflammatory and catabolic environment through a locally applied concentrate of platelets, leukocytes, and growth factors. Clinically, PRP has been shown to be possibly a viable treatment adjuvant for a variety of inflammatory and degenerative conditions. Recent efforts have focused on optimizing delivery methods that enable platelets to slowly degranulate their biological constituents, which may promote healing and improve OA symptoms for a longer duration. There are various factors that affect the progression of OA within joints, including inhibition of inflammatory cytokines and altering the level of enzymatic expression. PRP therapy aims to mediate inflammatory and catabolic factors in a degenerative environment through the secretion of anti-inflammatory factors and chemotaxic effects. There are a growing number of studies that have demonstrated the clinical benefit of PRP for non-operative management of OA. Additional randomized controlled trials with long-term follow-up are needed in order to validate PRP's therapeutic efficacy in this setting. Additionally, continued basic research along with well-designed pre-clinical studies and reporting standards are necessary in order to clarify the effectiveness of PRP for cartilage repair and regeneration for future clinical applications.
Collapse
|
35
|
Repair of Damaged Articular Cartilage: Current Approaches and Future Directions. Int J Mol Sci 2018; 19:ijms19082366. [PMID: 30103493 PMCID: PMC6122081 DOI: 10.3390/ijms19082366] [Citation(s) in RCA: 174] [Impact Index Per Article: 24.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 08/07/2018] [Accepted: 08/07/2018] [Indexed: 12/28/2022] Open
Abstract
Articular hyaline cartilage is extensively hydrated, but it is neither innervated nor vascularized, and its low cell density allows only extremely limited self-renewal. Most clinical and research efforts currently focus on the restoration of cartilage damaged in connection with osteoarthritis or trauma. Here, we discuss current clinical approaches for repairing cartilage, as well as research approaches which are currently developing, and those under translation into clinical practice. We also describe potential future directions in this area, including tissue engineering based on scaffolding and/or stem cells as well as a combination of gene and cell therapy. Particular focus is placed on cell-based approaches and the potential of recently characterized chondro-progenitors; progress with induced pluripotent stem cells is also discussed. In this context, we also consider the ability of different types of stem cell to restore hyaline cartilage and the importance of mimicking the environment in vivo during cell expansion and differentiation into mature chondrocytes.
Collapse
|
36
|
O'Leary SA, White JL, Hu JC, Athanasiou KA. Biochemical and biomechanical characterisation of equine cervical facet joint cartilage. Equine Vet J 2018; 50:800-808. [DOI: 10.1111/evj.12845] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2017] [Accepted: 03/30/2018] [Indexed: 11/28/2022]
Affiliation(s)
- S. A. O'Leary
- Department of Biomedical Engineering; University of California; Davis California USA
| | - J. L. White
- Department of Biomedical Engineering; University of California; Davis California USA
| | - J. C. Hu
- Department of Biomedical Engineering; University of California; Davis California USA
| | - K. A. Athanasiou
- Department of Biomedical Engineering; University of California; Davis California USA
- Department of Biomedical Engineering; University of California; Irvine California USA
| |
Collapse
|
37
|
Chen X, Gao C. Influences of surface coating of PLGA nanoparticles on immune activation of macrophages. J Mater Chem B 2018; 6:2065-2077. [DOI: 10.1039/c7tb03080k] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Uptake of BSA-coated PLGA NPs induces a stronger inflammatory response which is represented by the up-expression of TNF-α.
Collapse
Affiliation(s)
- Xinyi Chen
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization
- Department of Polymer Science and Engineering
- Zhejiang University
- Hangzhou 310027
- China
| | - Changyou Gao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization
- Department of Polymer Science and Engineering
- Zhejiang University
- Hangzhou 310027
- China
| |
Collapse
|
38
|
Chen X, Gao C. Influences of size and surface coating of gold nanoparticles on inflammatory activation of macrophages. Colloids Surf B Biointerfaces 2017; 160:372-380. [DOI: 10.1016/j.colsurfb.2017.09.046] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 09/19/2017] [Accepted: 09/20/2017] [Indexed: 12/31/2022]
|
39
|
Dai Y, Shen T, Ma L, Wang D, Gao C. Regeneration of osteochondral defects in vivo by a cell-free cylindrical poly(lactide-co-glycolide) scaffold with a radially oriented microstructure. J Tissue Eng Regen Med 2017; 12:e1647-e1661. [PMID: 29047223 DOI: 10.1002/term.2592] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 09/12/2017] [Accepted: 10/09/2017] [Indexed: 12/15/2022]
Abstract
A scaffold with an oriented porous architecture to facilitate cell infiltration and bioactive interflow between neo-host tissues is of great importance for in situ inductive osteochondral regeneration. In this study, a poly(lactide-co-glycolide) (PLGA) scaffold with oriented pores in its radial direction was fabricated via unidirectional cooling of the PLGA solution in the radial direction, following with lyophilization. Micro-computed tomography evaluation and scanning electron microscopy observation confirmed the radially oriented microtubular pores in the scaffold. The scaffold had porosity larger than 90% and a compressive modulus of 4 MPa in a dry state. Culture of bone marrow stem cells in vitro revealed faster migration and regular distribution of cells in the poly(lactide-co-glycolide) scaffold with oriented pores compared with the random PLGA scaffold. The cell-free oriented macroporous PLGA scaffold was implanted into rabbit articular osteochondral defect in vivo for 12 weeks to evaluate its inductive tissue regeneration function. Histological analysis confirmed obvious tide mark formation and abundant chondrocytes distributed regularly with obvious lacunae in the cartilage layer. Safranin O-fast green staining showed an obvious boundary between the two layers with distinct staining results, indicating the simultaneous regeneration of the cartilage and subchondral bone layers, which is not the case for the random poly(lactide-co-glycolide) scaffold after the same implantation in vivo. The oriented macroporous PLGA scaffold is a promising material for the in situ inductive osteochondral regeneration without the necessity of preseeding cells.
Collapse
Affiliation(s)
- Yuankun Dai
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China
| | - Tao Shen
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China
| | - Lie Ma
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China
| | - Dongan Wang
- Division of Bioengineering, School of Chemical & Biomedical Engineering, Nanyang Technological University, Singapore, Singapore
| | - Changyou Gao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China.,Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
40
|
In Vivo Articular Cartilage Regeneration Using Human Dental Pulp Stem Cells Cultured in an Alginate Scaffold: A Preliminary Study. Stem Cells Int 2017; 2017:8309256. [PMID: 28951745 PMCID: PMC5603743 DOI: 10.1155/2017/8309256] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 07/04/2017] [Indexed: 01/09/2023] Open
Abstract
Osteoarthritis is an inflammatory disease in which all joint-related elements, articular cartilage in particular, are affected. The poor regeneration capacity of this tissue together with the lack of pharmacological treatment has led to the development of regenerative medicine methodologies including microfracture and autologous chondrocyte implantation (ACI). The effectiveness of ACI has been shown in vitro and in vivo, but the use of other cell types, including bone marrow and adipose-derived mesenchymal stem cells, is necessary because of the poor proliferation rate of isolated articular chondrocytes. In this investigation, we assessed the chondrogenic ability of human dental pulp stem cells (hDPSCs) to regenerate cartilage in vitro and in vivo. hDPSCs and primary isolated rabbit chondrocytes were cultured in chondrogenic culture medium and found to express collagen II and aggrecan. Both cell types were cultured in 3% alginate hydrogels and implanted in a rabbit model of cartilage damage. Three months after surgery, significant cartilage regeneration was observed, particularly in the animals implanted with hDPSCs. Although the results presented here are preliminary, they suggest that hDPSCs may be useful for regeneration of articular cartilage.
Collapse
|
41
|
Karateev AE, Karateev DE, Davydov OS. PAIN AND INFLAMMATION. PART 1. PATHOGENETIC ASPECTS. RHEUMATOLOGY SCIENCE AND PRACTICE 2017. [DOI: 10.14412/1995-4484-2016-693-704] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The relief of suffering, which is associated with a rapid and complete elimination of painful sensations, is the most important challenge facing physicians of many specialties. It is obvious that it can be solved only when you understand clearly the processes governing the development and chronization of pain. Inflammation, a universal adaptive mechanism that always accompanies damage to living tissues, plays a key role. Part 1 of this review considers the main stages of development of an inflammatory response, beginning with primary damage accompanied by the release of molecules acting as an alarm and ending with the deployment of a complete picture of the inflammatory response with the involvement of many cell elements and the overexpression of cytokines and proinflammatory mediators. The biological basis of the peripheral and central nociceptive sensitization phenomenon that is rigidly associated with inflammation is presented. Particular emphasis is placed on the possible natural completion of the inflammatory response, on the adaptive mechanisms regulating this process and on the reasons that prevent this and determines inflammation chronization.
Collapse
Affiliation(s)
| | | | - O. S. Davydov
- Z.P. Solovyev Research and Practical Center of Psychoneurology, Moscow Healthcare Department
| |
Collapse
|
42
|
Dong S, Xia T, Wang L, Zhao Q, Tian J. Investigation of candidate genes for osteoarthritis based on gene expression profiles. ACTA ORTHOPAEDICA ET TRAUMATOLOGICA TURCICA 2016; 50:686-690. [PMID: 27866912 PMCID: PMC6197383 DOI: 10.1016/j.aott.2016.04.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 02/22/2016] [Accepted: 04/30/2016] [Indexed: 02/07/2023]
Abstract
OBJECTIVE To explore the mechanism of osteoarthritis (OA) and provide valid biological information for further investigation. METHODS Gene expression profile of GSE46750 was downloaded from Gene Expression Omnibus database. The Linear Models for Microarray Data (limma) package (Bioconductor project, http://www.bioconductor.org/packages/release/bioc/html/limma.html) was used to identify differentially expressed genes (DEGs) in inflamed OA samples. Gene Ontology function enrichment analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways enrichment analysis of DEGs were performed based on Database for Annotation, Visualization and Integrated Discovery data, and protein-protein interaction (PPI) network was constructed based on the Search Tool for the Retrieval of Interacting Genes/Proteins database. Regulatory network was screened based on Encyclopedia of DNA Elements. Molecular Complex Detection was used for sub-network screening. Two sub-networks with highest node degree were integrated with transcriptional regulatory network and KEGG functional enrichment analysis was processed for 2 modules. RESULTS In total, 401 up- and 196 down-regulated DEGs were obtained. Up-regulated DEGs were involved in inflammatory response, while down-regulated DEGs were involved in cell cycle. PPI network with 2392 protein interactions was constructed. Moreover, 10 genes including Interleukin 6 (IL6) and Aurora B kinase (AURKB) were found to be outstanding in PPI network. There are 214 up- and 8 down-regulated transcription factor (TF)-target pairs in the TF regulatory network. Module 1 had TFs including SPI1, PRDM1, and FOS, while module 2 contained FOSL1. The nodes in module 1 were enriched in chemokine signaling pathway, while the nodes in module 2 were mainly enriched in cell cycle. CONCLUSION The screened DEGs including IL6, AGT, and AURKB might be potential biomarkers for gene therapy for OA by being regulated by TFs such as FOS and SPI1, and participating in the cell cycle and cytokine-cytokine receptor interaction pathway.
Collapse
Affiliation(s)
| | - Tian Xia
- Shanghai First People's Hospital, Shanghai, China
| | - Lei Wang
- Shanghai First People's Hospital, Shanghai, China
| | - Qinghua Zhao
- Shanghai First People's Hospital, Shanghai, China
| | - Jiwei Tian
- Shanghai First People's Hospital, Shanghai, China.
| |
Collapse
|
43
|
Modulation of Synovial Fluid-Derived Mesenchymal Stem Cells by Intra-Articular and Intraosseous Platelet Rich Plasma Administration. Stem Cells Int 2016; 2016:1247950. [PMID: 27818688 PMCID: PMC5080490 DOI: 10.1155/2016/1247950] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Revised: 09/12/2016] [Accepted: 09/20/2016] [Indexed: 11/28/2022] Open
Abstract
The aim of this study was to evaluate the effect of intra-articular (IA) or a combination of intra-articular and intraosseous (IO) infiltration of Platelet Rich Plasma (PRP) on the cellular content of synovial fluid (SF) of osteoarthritic patients. Thirty-one patients received a single infiltration of PRP either in the IA space (n = 14) or in the IA space together with two IO infiltrations, one in the medial femoral condyle and one in the tibial plateau (n = 17). SF was collected before and after one week of the infiltration. The presence in the SF of mesenchymal stem cells (MSCs), monocytes, and lymphocytes was determined and quantified by flow cytometry. The number and identity of the MSCs were further confirmed by colony-forming and differentiation assays. PRP infiltration into the subchondral bone (SB) and the IA space induced a reduction in the population of MSCs in the SF. This reduction in MSCs was further confirmed by colony-forming (CFU-F) assay. On the contrary, IA infiltration alone did not cause variations in any of the cellular populations by flow cytometry or CFU-F assay. The SF of osteoarthritic patients contains a population of MSCs that can be modulated by PRP infiltration of the SB compartment.
Collapse
|
44
|
Farrell E, Fahy N, Ryan AE, Flatharta CO, O'Flynn L, Ritter T, Murphy JM. vIL-10-overexpressing human MSCs modulate naïve and activated T lymphocytes following induction of collagenase-induced osteoarthritis. Stem Cell Res Ther 2016; 7:74. [PMID: 27194025 PMCID: PMC4870800 DOI: 10.1186/s13287-016-0331-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Revised: 03/14/2016] [Accepted: 04/26/2016] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Recent efforts in osteoarthritis (OA) research have highlighted synovial inflammation and involvement of immune cells in disease onset and progression. We sought to establish the in-vivo immune response in collagenase-induced OA and investigate the ability of human mesenchymal stem cells (hMSCs) overexpressing viral interleukin 10 (vIL-10) to modulate immune populations and delay/prevent disease progression. METHODS Eight-week-old male C57BL/6 mice were injected with 1 U type VII collagenase over two consecutive days. At day 7, 20,000 hMSCs overexpressing vIL-10 were injected into the affected knee. Control groups comprised of vehicle, 20,000 untransduced or adNull-transduced MSCs or virus alone. Six weeks later knees were harvested for histological analysis and popliteal and inguinal lymph nodes for flow cytometric analysis. RESULTS At this time there was no significant difference in knee OA scores between any of the groups. A trend toward more damage in animals treated with hMSCs was observed. Interestingly there was a significant reduction in the amount of activated CD4 and CD8 T cells in the vIL-10-expressing hMSC group. CONCLUSIONS vIL-10-overexpressing hMSCs can induce long-term reduction in activated T cells in draining lymph nodes of mice with collagenase-induced OA. This could lead to reduced OA severity or disease progression over the long term.
Collapse
Affiliation(s)
- Eric Farrell
- Department of Oral and Maxillofacial Surgery, Special Dental Care and Orthodontics, Erasmus MC, University Medical Centre, Room Ee1614, Erasmus MC, Wytemaweg 80, Rotterdam, 3015CN, The Netherlands. .,Regenerative Medicine Institute, National University of Ireland Galway, Galway, Ireland.
| | - Niamh Fahy
- Regenerative Medicine Institute, National University of Ireland Galway, Galway, Ireland.,Musculoskeletal Regeneration, AO Research Institute Davos (ARI), Davos, Switzerland
| | - Aideen E Ryan
- Regenerative Medicine Institute, National University of Ireland Galway, Galway, Ireland.,College of Medicine, Nursing and Health Sciences, National University of Ireland Galway, Galway, Ireland.,Discipline of Pharmacology and Therapeutics, National University of Ireland Galway, Galway, Ireland
| | - Cathal O Flatharta
- Regenerative Medicine Institute, National University of Ireland Galway, Galway, Ireland
| | - Lisa O'Flynn
- Regenerative Medicine Institute, National University of Ireland Galway, Galway, Ireland.,College of Medicine, Nursing and Health Sciences, National University of Ireland Galway, Galway, Ireland.,Orbsen Therapeutics Ltd, Galway, Ireland
| | - Thomas Ritter
- Regenerative Medicine Institute, National University of Ireland Galway, Galway, Ireland.,College of Medicine, Nursing and Health Sciences, National University of Ireland Galway, Galway, Ireland
| | - J Mary Murphy
- Regenerative Medicine Institute, National University of Ireland Galway, Galway, Ireland
| |
Collapse
|
45
|
Sánchez M, Anitua E, Delgado D, Sanchez P, Prado R, Goiriena JJ, Prosper F, Orive G, Padilla S. A new strategy to tackle severe knee osteoarthritis: Combination of intra-articular and intraosseous injections of Platelet Rich Plasma. Expert Opin Biol Ther 2016; 16:627-43. [PMID: 26930117 DOI: 10.1517/14712598.2016.1157162] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
INTRODUCTION Knee osteoarthritis (KOA) is a mechanically induced, cytokine and enzyme-mediated disorder involving all the joint tissue of the knee. Rebuilding a physiological-homeostatic network at the tissue level following knee organ failure, such as in severe KOA, is a daunting task with therapeutic targets encompassing the articular cartilage, synovium and subchondral bone. Intraarticular infiltration of plasma rich in growth factors (PRP) has emerged as a promising symptomatic approach, although it is insufficient to reach the subchondral bone. AREAS COVERED This review addresses current molecular and cellular data in joint homeostasis and osteoarthritis pathophysiology. In particular, it focuses on changes that subchondral bone undergoes in knee osteoarthritis and evaluates recent observations on the crosstalk among articular cartilage, subchondral bone and synovial membrane. In addition, we review some mechanistic aspects that have been proposed and provide the rationale for using PRP intraosseously in KOA. EXPERT OPINION The knee joint is a paradigm of autonomy and connectedness of its anatomical structures and tissues from which it is made. We propose an innovative approach to the treatment of severe knee osteoarthritis consisting of a combination of intraarticular and intraosseous infiltrations of PRP, which might offer a new therapeutic tool in KOA therapy.
Collapse
Affiliation(s)
- Mikel Sánchez
- a Arthroscopic Surgery Unit , Hospital Vithas San José , Vitoria-Gasteiz , Spain
| | - Eduardo Anitua
- b Department of Regenerative Medicine, Laboratory of Regenerative Medicine, BTI Biotechnology Institute , Vitoria , Spain
| | - Diego Delgado
- c Arthroscopic Surgery Unit Research , Hospital Vithas San José , Vitoria-Gasteiz , Spain
| | - Peio Sanchez
- c Arthroscopic Surgery Unit Research , Hospital Vithas San José , Vitoria-Gasteiz , Spain
| | - Roberto Prado
- b Department of Regenerative Medicine, Laboratory of Regenerative Medicine, BTI Biotechnology Institute , Vitoria , Spain
| | | | - Felipe Prosper
- e Cell Therapy Program, Foundation for Applied Medical Research , University of Navarra , Pamplona , Spain.,f Hematology and Cell Therapy Department , Clínica Universidad de Navarra, University of Navarra , Pamplona , Spain
| | - Gorka Orive
- b Department of Regenerative Medicine, Laboratory of Regenerative Medicine, BTI Biotechnology Institute , Vitoria , Spain.,g Laboratory of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy , University of the Basque Country , Vitoria , Spain.,h Networking Biomedical Research Center on Bioengineering, Biomaterials and Nanomedicine , CIBER-BBN, SLFPB-EHU , Vitoria-Gasteiz , Spain
| | - Sabino Padilla
- b Department of Regenerative Medicine, Laboratory of Regenerative Medicine, BTI Biotechnology Institute , Vitoria , Spain
| |
Collapse
|
46
|
Lohan P, Treacy O, Lynch K, Barry F, Murphy M, Griffin MD, Ritter T, Ryan AE. Culture expanded primary chondrocytes have potent immunomodulatory properties and do not induce an allogeneic immune response. Osteoarthritis Cartilage 2016; 24:521-33. [PMID: 26493330 DOI: 10.1016/j.joca.2015.10.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Revised: 10/06/2015] [Accepted: 10/10/2015] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Allogeneic cell therapies, such as mesenchymal stromal cells (MSC), which have potent regenerative and anti-inflammatory potential are being investigated as a therapy for osteoarthritis (OA) and cartilage injury. Here we describe another potential source of regenerative and anti-inflammatory allogeneic cells, culture expanded primary chondrocytes (CEPC). In direct comparison to allogeneic MSC, we extensively assess the immunological interactions of CEPC in an allogeneic setting. METHODS Chondrocytes were isolated from rat articular cartilage and cultured in normoxic or hypoxic conditions. In vitro co-culture assays with allogeneic lymphocytes and macrophages were used to assess the immunomodulatory capacities of the chondrocytes, followed by immune response analysis by flow cytometry, ELISA and qPCR. RESULTS CEPC showed reduced induction of proliferation, activation and cytotoxic granzyme B expression in allogeneic T cells. Importantly, exposure to pro-inflammatory cytokines did not increase CEPC immunogenicity despite increases in MHC-I. Furthermore, CEPC had a potent ability to suppress allogeneic T cell proliferation, which was dependent on nitric oxide production. This suppression was contact independent in hypoxia cultured CEPC. Finally, chondrocytes were shown to have the capacity to modulate pro-inflammatory macrophage activity by reducing MHC-II expression and TNF-α secretion. CONCLUSION These data indicate the potential use of allogeneic chondrocytes in OA and cartilage defects. The lack of evident immunogenicity, despite exposure to a pro-inflammatory environment, coupled with the immunomodulatory ability indicates that these cells have the potential to evade the host immune system and suppress inflammation, thus potentially facilitating the resolution of OA induced inflammation and cartilage regeneration.
Collapse
Affiliation(s)
- P Lohan
- Regenerative Medicine Institute (REMEDI), School of Medicine, College of Medicine, Nursing and Health Sciences, National University of Ireland, Galway, Ireland
| | - O Treacy
- Regenerative Medicine Institute (REMEDI), School of Medicine, College of Medicine, Nursing and Health Sciences, National University of Ireland, Galway, Ireland
| | - K Lynch
- Regenerative Medicine Institute (REMEDI), School of Medicine, College of Medicine, Nursing and Health Sciences, National University of Ireland, Galway, Ireland
| | - F Barry
- Regenerative Medicine Institute (REMEDI), School of Medicine, College of Medicine, Nursing and Health Sciences, National University of Ireland, Galway, Ireland
| | - M Murphy
- Regenerative Medicine Institute (REMEDI), School of Medicine, College of Medicine, Nursing and Health Sciences, National University of Ireland, Galway, Ireland
| | - M D Griffin
- Regenerative Medicine Institute (REMEDI), School of Medicine, College of Medicine, Nursing and Health Sciences, National University of Ireland, Galway, Ireland
| | - T Ritter
- Regenerative Medicine Institute (REMEDI), School of Medicine, College of Medicine, Nursing and Health Sciences, National University of Ireland, Galway, Ireland
| | - A E Ryan
- Regenerative Medicine Institute (REMEDI), School of Medicine, College of Medicine, Nursing and Health Sciences, National University of Ireland, Galway, Ireland; Discipline of Pharmacology and Therapeutics, School of Medicine, College of Medicine, Nursing and Health Sciences, National University of Ireland, Galway, Ireland.
| |
Collapse
|
47
|
Scotti C, Gobbi A, Karnatzikos G, Martin I, Shimomura K, Lane JG, Peretti GM, Nakamura N. Cartilage Repair in the Inflamed Joint: Considerations for Biological Augmentation Toward Tissue Regeneration. TISSUE ENGINEERING PART B-REVIEWS 2015; 22:149-59. [PMID: 26467024 DOI: 10.1089/ten.teb.2015.0297] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Cartilage repair/regeneration procedures (e.g., microfracture, autologous chondrocyte implantation [ACI]) typically result in a satisfactory outcome in selected patients. However, the vast majority of patients with chronic symptoms and, in general, a more diseased joint, do not benefit from these surgical techniques. The aims of this work were to (1) review factors negatively influencing the joint environment; (2) review current adjuvant therapies that can be used to improve results of cartilage repair/regeneration procedures in patients with more diseased joints, (3) outline future lines of research and promising experimental approaches. Chronicity of symptoms and advancing patient age appear to be the most relevant factors negatively affecting clinical outcome of cartilage repair/regeneration. Preliminary experience with hyaluronic acid, platelet-rich plasma, and mesenchymal stem cell has been positive but there is no strong evidence supporting the use of these products and this requires further assessment with high-quality, prospective clinical trials. The use of a Tissue Therapy strategy, based on more mature engineered tissues, holds promise to tackle limitations of standard ACI procedures. Current research has highlighted the need for more targeted therapies, and (1) induction of tolerance with granulocyte colony-stimulating factor (G-CSF) or by preventing IL-6 downregulation; (2) combined IL-4 and IL-10 local release; and (3) selective activation of the prostaglandin E2 (PGE2) signaling appear to be the most promising innovative strategies. For older patients and for those with chronic symptoms, adjuvant therapies are needed in combination with microfracture and ACI.
Collapse
Affiliation(s)
| | - Alberto Gobbi
- 2 Orthopedic Arthroscopic Surgery International (O.A.S.I.) Bioresearch Foundation , Gobbi Onlus, Milan, Italy
| | - Georgios Karnatzikos
- 2 Orthopedic Arthroscopic Surgery International (O.A.S.I.) Bioresearch Foundation , Gobbi Onlus, Milan, Italy
| | - Ivan Martin
- 3 Departments of Surgery and of Biomedicine, University Hospital Basel, University of Basel , Basel, Switzerland
| | - Kazunori Shimomura
- 4 Department of Orthopedics, Osaka University Graduate School of Medicine , Osaka, Japan
| | - John G Lane
- 5 COAST Surgery Center, University of California , San Diego, California
| | - Giuseppe Michele Peretti
- 1 IRCCS Istituto Ortopedico Galeazzi , Milan, Italy .,6 Department of Biomedical Sciences for Health, University of Milan , Milan, Italy
| | - Norimasa Nakamura
- 7 Institute for Medical Science in Sports, Osaka Health Science University , Osaka, Japan .,8 Center for Advanced Medical Engineering and Informatics, Osaka University , Osaka, Japan
| |
Collapse
|
48
|
Luo L, Wei Q, Liu L, Lin X, Lin C, Zheng LI, Zhao J. Protocatechuic acid benefits proliferation and phenotypic maintenance of rabbit articular chondrocytes: An in vitro study. Exp Ther Med 2015; 9:1865-1870. [PMID: 26136906 DOI: 10.3892/etm.2015.2326] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 02/06/2015] [Indexed: 12/22/2022] Open
Abstract
Numerous antioxidants exhibit antiarthritic effects due to their inhibitory effect on inflammatory factors. Certain antioxidants, such as protocatechuic acid (PCA) and its analogs, have been reported to be effective in the treatment of arthritis. However, the effect of PCA on chondro-protection may be alleviated due to the induction of apoptosis, as has been demonstrated in stomatocytes. To clearly determine the effect of PCA on the biological and cellular metabolism of rabbit articular chondrocytes in vitro, examinations of cytotoxicity, proliferation and morphology were performed, in addition to analyses of glycosaminoglycan (GAG) synthesis and the expression of cartilage-specific genes. The results revealed that PCA effectively promoted chondrocyte growth, the synthesis of the extracellular matrix and the mRNA expression of aggrecan, collagen II and Sox9, while downregulating the expression of the collagen I gene, a marker of chondrocyte dedifferentiation. In addition, hypertrophy, which may result in chondrocyte ossification, was not detected in the groups. Among the doses (range, 0.05-0.3 mmol/l) of PCA that promoted the proliferation of chondrocytes, a concentration of 0.125 mmol/l produced the optimum performance. The results indicated that PCA, particularly at a dose of 0.125 mmol/l, accelerated the proliferation of rabbit articular chondrocytes in vitro and maintained their phenotype. This study may provide a basis for further research concerning the treatment of cartilage defects.
Collapse
Affiliation(s)
- Like Luo
- Department of Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China ; Guangxi Key Laboratory of Regenerative Medicine, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Qingjun Wei
- Department of Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China ; Guangxi Key Laboratory of Regenerative Medicine, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Lei Liu
- Department of Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China ; Guangxi Key Laboratory of Regenerative Medicine, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Xiao Lin
- School of Chemistry and Chemical Engineering, Guangxi University, Nanning, Guangxi 530004, P.R. China ; Guangxi Key Laboratory of Traditional Chinese Medicine Quality Standards, Guangxi Institute of Traditional Medical and Pharmaceutical Sciences, Nanning, Guangxi 530022, P.R. China
| | - Cuiwu Lin
- School of Chemistry and Chemical Engineering, Guangxi University, Nanning, Guangxi 530004, P.R. China
| | - L I Zheng
- Guangxi Key Laboratory of Regenerative Medicine, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China ; The Medical and Scientific Research Center, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Jinmin Zhao
- Department of Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China ; Guangxi Key Laboratory of Regenerative Medicine, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| |
Collapse
|