101
|
Bartels YL, van Lent PLEM, van der Kraan PM, Blom AB, Bonger KM, van den Bosch MHJ. Inhibition of TLR4 signalling to dampen joint inflammation in osteoarthritis. Rheumatology (Oxford) 2024; 63:608-618. [PMID: 37788083 PMCID: PMC10907820 DOI: 10.1093/rheumatology/kead493] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 08/23/2023] [Accepted: 08/29/2023] [Indexed: 10/05/2023] Open
Abstract
Local and systemic low-grade inflammation, mainly involving the innate immune system, plays an important role in the development of OA. A receptor playing a key role in initiation of this inflammation is the pattern-recognition receptor Toll-like receptor 4 (TLR4). In the joint, various ligands for TLR4, many of which are damage-associated molecular patterns (DAMPs), are present that can activate TLR4 signalling. This leads to the production of pro-inflammatory and catabolic mediators that cause joint damage. In this narrative review, we will first discuss the involvement of TLR4 ligands and signalling in OA. Furthermore, we will provide an overview of methods for inhibit, TLR4 signalling by RNA interference, neutralizing anti-TLR4 antibodies, small molecules and inhibitors targeting the TLR4 co-receptor MD2. Finally, we will focus on possible applications and challenges of these strategies in the dampening of inflammation in OA.
Collapse
Affiliation(s)
- Yvonne L Bartels
- Experimental Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Peter L E M van Lent
- Experimental Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Peter M van der Kraan
- Experimental Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Arjen B Blom
- Experimental Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Kimberly M Bonger
- Synthetic Organic Chemistry, Institute for Molecules and Materials, Radboud University, Nijmegen, The Netherlands
| | | |
Collapse
|
102
|
Clarke E, Varela L, Jenkins RE, Lozano-Andrés E, Cywińska A, Przewozny M, van Weeren PR, van de Lest CH, Peffers M, Wauben MH. Proteome and phospholipidome interrelationship of synovial fluid-derived extracellular vesicles in equine osteoarthritis: An exploratory 'multi-omics' study to identify composite biomarkers. Biochem Biophys Rep 2024; 37:101635. [PMID: 38298208 PMCID: PMC10828605 DOI: 10.1016/j.bbrep.2023.101635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 12/15/2023] [Accepted: 12/29/2023] [Indexed: 02/02/2024] Open
Abstract
Osteoarthritis causes progressive joint deterioration, severe morbidity, and reduced mobility in both humans and horses. Currently, osteoarthritis is diagnosed at late stages through clinical examination and radiographic imaging, hence it is challenging to address and provide timely therapeutic interventions to slow disease progression or ameliorate symptoms. Extracellular vesicles are cell-derived vesicles that play a key role in cell-to-cell communication and are potential sources for specific composite biomarker panel discovery. We here used a multi-omics strategy combining proteomics and phospholipidomics in an integral approach to identify composite biomarkers associated to purified extracellular vesicles from synovial fluid of healthy, mildly and severely osteoarthritic equine joints. Although the number of extracellular vesicles was unaffected by osteoarthritis, proteome profiling of extracellular vesicles by mass spectrometry identified 40 differentially expressed proteins (non-adjusted p < 0.05) in osteoarthritic joints associated with 7 significant canonical pathways in osteoarthritis. Moreover, pathway analysis unveiled changes in disease and molecular functions during osteoarthritis development. Phospholipidome profiling by mass spectrometry showed a relative increase in sphingomyelin and a decrease in phosphatidylcholine, phosphatidylinositol, and phosphatidylserine in extracellular vesicles derived from osteoarthritic joints compared to healthy joints. Unsupervised data integration revealed positive correlations between the proteome and the phospholipidome. Comprehensive analysis showed that some phospholipids and their related proteins increased as the severity of osteoarthritis progressed, while others decreased or remained stable. Altogether our data show interrelationships between synovial fluid extracellular vesicle-associated phospholipids and proteins responding to osteoarthritis pathology and which could be explored as potential composite diagnostic biomarkers of disease.
Collapse
Affiliation(s)
- Emily Clarke
- Department of Musculoskeletal Biology and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
| | - Laura Varela
- Division Equine Sciences, Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
- Division Cell Biology, Metabolism & Cancer, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - Rosalind E. Jenkins
- Centre for Drug Safety Science Bioanalytical Facility, Liverpool Shared Research Facilities, Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Estefanía Lozano-Andrés
- Division Cell Biology, Metabolism & Cancer, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
- Division of Infectious Diseases & Immunology, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - Anna Cywińska
- Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland
| | | | - P. René van Weeren
- Division Equine Sciences, Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - Chris H.A. van de Lest
- Division Equine Sciences, Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
- Division Cell Biology, Metabolism & Cancer, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - Mandy Peffers
- Department of Musculoskeletal Biology and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
| | - Marca H.M. Wauben
- Division Cell Biology, Metabolism & Cancer, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| |
Collapse
|
103
|
Mukherjee A, Das B. The role of inflammatory mediators and matrix metalloproteinases (MMPs) in the progression of osteoarthritis. BIOMATERIALS AND BIOSYSTEMS 2024; 13:100090. [PMID: 38440290 PMCID: PMC10910010 DOI: 10.1016/j.bbiosy.2024.100090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 12/04/2023] [Accepted: 02/20/2024] [Indexed: 03/06/2024] Open
Abstract
Osteoarthritis (OA) is a chronic musculoskeletal disorder characterized by an imbalance between (synthesis) and catabolism (degradation) in altered homeostasis of articular cartilage mediated primarily by the innate immune system. OA degenerates the joints resulting in synovial hyperplasia, degradation of articular cartilage with damage of the structural and functional integrity of the cartilage extracellular matrix, subchondral sclerosis, osteophyte formation, and is characterized by chronic pain, stiffness, and loss of function. Inflammation triggered by factors like biomechanical stress is involved in the development of osteoarthritis. In OA apart from catabolic effects, anti-inflammatory anabolic processes also occur continually. There is also an underlying chronic inflammation present, not only in cartilage tissue but also within the synovium, which perpetuates tissue destruction of the OA joint. The consideration of inflammation in OA considers synovitis and/or other cellular and molecular events in the synovium during the progression of OA. In this review, we have presented the progression of joint degradation that results in OA. The critical role of inflammation in the pathogenesis of OA is discussed in detail along with the dysregulation within the cytokine networks composed of inflammatory and anti-inflammatory cytokines that drive catabolic pathways, inhibit matrix synthesis, and promote cellular apoptosis. OA pathogenesis, fluctuation of synovitis, and its clinical impact on disease progression are presented here along with the role of synovial macrophages in promoting inflammatory and destructive responses in OA. The role of interplay between different cytokines, structure, and function of their receptors in the inter-cellular signaling pathway is further explored. The effect of cytokines in the increased synthesis and release of matrix-decomposing proteolytic enzymes, such as matrix metalloproteinase (MMPs) and a disintegrin-like and metalloproteinase with thrombospondin motif (ADAMTS), is elaborated emphasizing the potential impact of MMPs on the chondrocytes, synovial cells, articular and periarticular tissues, and other immune system cells migrating to the site of inflammation. We also shed light on the pathogenesis of OA via oxidative damage particularly due to nitric oxide (NO) via its angiogenic response to inflammation. We concluded by presenting the current knowledge about the tissue inhibitors of metalloproteinases (TIMPs). Synthetic MMP inhibitors include zinc binding group (ZBG), non-ZBG, and mechanism-based inhibitors, all of which have the potential to be therapeutically beneficial in the treatment of osteoarthritis. Improving our understanding of the signaling pathways and molecular mechanisms that regulate the MMP gene expression, may open up new avenues for the creation of therapies that can stop the joint damage associated with OA.
Collapse
Affiliation(s)
- Anwesha Mukherjee
- Department of Biomedical Engineering, Indian Institute of Technology Ropar, India
| | - Bodhisatwa Das
- Department of Biomedical Engineering, Indian Institute of Technology Ropar, India
| |
Collapse
|
104
|
Karjalainen K, Tanska P, Collins KH, Herzog W, Korhonen RK, Moo EK. Independent and combined effects of obesity and traumatic joint injury to the structure and composition of rat knee cartilage. Connect Tissue Res 2024; 65:117-132. [PMID: 38530304 DOI: 10.1080/03008207.2024.2310838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 01/22/2024] [Indexed: 03/27/2024]
Abstract
Osteoarthritis (OA) is a multifactorial joint disease characterized by articular cartilage degradation. Risk factors for OA include joint trauma, obesity, and inflammation, each of which can affect joint health independently, but their interaction and the associated consequences of such interaction were largely unexplored. Here, we studied compositional and structural alterations in knee joint cartilages of Sprague-Dawley rats exposed to two OA risk factors: joint injury and diet-induced obesity. Joint injury was imposed by surgical transection of anterior cruciate ligaments (ACLx), and obesity was induced by a high fat/high sucrose diet. Depth-dependent proteoglycan (PG) content and collagen structural network of cartilage were measured from histological sections collected previously in Collins et al.. (2015). We found that ACLx primarily affected the superficial cartilages. Compositionally, ACLx led to reduced PG content in lean animals, but increased PG content in obese rats. Structurally, ACLx caused disorganization of collagenous network in both lean and obese animals through increased collagen orientation in the superficial tissues and a change in the degree of fibrous alignment. However, the cartilage degradation attributed to joint injury and obesity was not necessarily additive when the two risk factors were present simultaneously, particularly for PG content and collagen orientation in the superficial tissues. Interestingly, sham surgeries caused a through-thickness disorganization of collagen network in lean and obese animals. We conclude that the interactions of multiple OA risk factors are complex and their combined effects cannot be understood by superposition principle. Further research is required to elucidate the interactive mechanism between OA subtypes.
Collapse
Affiliation(s)
- Kalle Karjalainen
- Department of Technical Physics, University of Eastern Finland, Kuopio, Finland
| | - Petri Tanska
- Department of Technical Physics, University of Eastern Finland, Kuopio, Finland
| | - Kelsey H Collins
- Laboratory of Musculoskeletal Crosstalk, Department of Orthopaedic Surgery, University of California San Francisco, San Francisco, USA
| | - Walter Herzog
- Human Performance Laboratory, Faculty of Kinesiology, University of Calgary, Calgary, Canada
| | - Rami K Korhonen
- Department of Technical Physics, University of Eastern Finland, Kuopio, Finland
| | - Eng Kuan Moo
- Department of Technical Physics, University of Eastern Finland, Kuopio, Finland
- Department of Mechanical and Aerospace Engineering, Carleton University, Ottawa, Canada
| |
Collapse
|
105
|
Pandey A, Singla M, Geller A, Goodman SB, Bhutani N. Targeting an inflammation-amplifying cell population can attenuate osteoarthritis-associated pain. Arthritis Res Ther 2024; 26:53. [PMID: 38368390 PMCID: PMC10874031 DOI: 10.1186/s13075-024-03284-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 01/31/2024] [Indexed: 02/19/2024] Open
Abstract
BACKGROUND Understanding of pain in osteoarthritis, its genesis, and perception is still in its early stages. Identification of precise ligand-receptor pairs that transduce pain and the cells and tissues in which they reside will elucidate new therapeutic approaches for pain management. Our recent studies had identified an inflammation-amplifying (Inf-A) cell population that is expanded in human OA cartilage and is distinctive in the expression of both IL1R1 and TNF-R2 receptors and active Jnk signaling cascade. METHODS In this study, we have tested the function of the cartilage-resident IL1R1+TNF-R2+ Inf-A cells in OA. We have identified that the IL1R1+TNF-R2+ Inf-A cells expand in aged mice as well as after anterior cruciate ligament tear upon tibia loading and OA initiation in mice. We targeted and modulated the Jnk signaling cascade in InfA through competitive inhibition of Jnk signaling in mice and human OA explants and tested the effects on joint structure and gait in mice. RESULTS Modulation of Jnk signaling led to attenuation of inflammatory cytokines CCL2 and CCL7 without showing any structural improvements in the joint architecture. Interestingly, Jnk inhibition and lowered CCL2 and 7 are sufficient to significantly improve the gait parameters in treated PTOA mice demonstrating reduced OA-associated pain. Consistent with the mice data, treatment with JNK inhibitor did not improve human OA cartilage explants. CONCLUSION These studies demonstrate that Inf-A, an articular-cartilage resident cell population, contributes to pain in OA via secretion of CCL2 and 7 and can be targeted via inhibition of Jnk signaling.
Collapse
Affiliation(s)
- Akshay Pandey
- Department of Orthopaedic Surgery, Stanford School of Medicine, 240, Pasteur Drive, Biomedical Innovations Bldg, Stanford, CA, 94034, USA
| | - Mamta Singla
- Department of Orthopaedic Surgery, Stanford School of Medicine, 240, Pasteur Drive, Biomedical Innovations Bldg, Stanford, CA, 94034, USA
| | - Ana Geller
- Department of Orthopaedic Surgery, Stanford School of Medicine, 240, Pasteur Drive, Biomedical Innovations Bldg, Stanford, CA, 94034, USA
| | - Stuart B Goodman
- Department of Orthopaedic Surgery, Stanford School of Medicine, 240, Pasteur Drive, Biomedical Innovations Bldg, Stanford, CA, 94034, USA
| | - Nidhi Bhutani
- Department of Orthopaedic Surgery, Stanford School of Medicine, 240, Pasteur Drive, Biomedical Innovations Bldg, Stanford, CA, 94034, USA.
| |
Collapse
|
106
|
Kim M, Kim J, Lee I. Mediating effect of lower extremity muscle on the relationship between obesity and osteoarthritis in middle-aged and elderly women in Korea: based on the 2009-2011 Korea National Health and Nutrition Examination Survey. Epidemiol Health 2024; 46:e2024027. [PMID: 38317528 PMCID: PMC11099567 DOI: 10.4178/epih.e2024027] [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: 10/20/2023] [Accepted: 01/08/2024] [Indexed: 02/07/2024] Open
Abstract
OBJECTIVES This study investigated whether the lower extremity muscle mass index (LMI) mediates the relationship between general obesity, central obesity, and knee osteoarthritis in middle-aged and elderly women in Korea. METHODS Data of 2,843 women aged ≥50 years were collected from the Korean National Health and Nutrition Examination Survey conducted between 2009 and 2011. General obesity and central obesity were evaluated based on body mass index (BMI) and waist circumference (WC), calculated through anthropometric measurements and body composition assessments. LMI was calculated by dividing the muscle mass in both legs-measured using the dual-energy X-ray absorptiometry-by body weight. Knee osteoarthritis was defined as a Kellgren-Lawrence scale (KL) grade of ≥2 as assessed through radiographic images. RESULTS Knee osteoarthritis prevalence, indicated by KL grades, was significantly higher in the general obesity and central obesity groups compared to the normal group, and conversely, lower with varying LMI levels. Using mediation analysis with bootstrapping and adjusting for covariates, we found that LMI mediated the relationship between BMI and KL (β, 0.005; 95% confidence interval [CI], 0.000 to 0.010) and WC and KL grade (β, 0.002; 95% CI, 0.001 to 0.003), explaining 4.8% and 6.7% of the total effects of BMI and WC on KL grade, respectively. CONCLUSIONS The study suggested that LMI partially mediates the link between general obesity and/or central obesity and knee osteoarthritis, proposing that a higher proportion of lower limb muscle mass relative to body weight can alleviate the increased risk of knee osteoarthritis caused by obesity.
Collapse
Affiliation(s)
- Minjun Kim
- Department of Physical Education, Yongin University, Yongin, Korea
| | - Joonwoong Kim
- Department of Convergence, Seowon University, Cheongju, Korea
| | - Inhwan Lee
- Department of Smart Healthcare, Changwon National University, Changwon, Korea
| |
Collapse
|
107
|
Wang M, Gao Z, Zhang Y, Zhao Q, Tan X, Wu S, Ding L, Liu Y, Qin S, Gu J, Xu L. Syringic acid promotes cartilage extracellular matrix generation and attenuates osteoarthritic cartilage degradation by activating TGF-β/Smad and inhibiting NF-κB signaling pathway. Phytother Res 2024; 38:1000-1012. [PMID: 38126609 DOI: 10.1002/ptr.8089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 11/14/2023] [Accepted: 11/26/2023] [Indexed: 12/23/2023]
Abstract
Osteoarthritis (OA) is a common chronic degenerative disease which is characterized by the disruption of articular cartilage. Syringic acid (SA) is a phenolic compound with anti-inflammatory, antioxidant, and other effects including promoting osteogenesis. However, the effect of SA on OA has not yet been reported. Therefore, the purpose of our study was to investigate the effect and mechanism of SA on OA in a mouse model of medial meniscal destabilization. The expressions of genes were evaluated by qPCR or western blot or immunofluorescence. RNA-seq analysis was performed to examine gene transcription alterations in chondrocytes treated with SA. The effect of SA on OA was evaluated using destabilization of the medial meniscus model of mice. We found that SA had no obvious toxic effect on chondrocytes, while promoting the expressions of chondrogenesis-related marker genes. The results of RNA-seq analysis showed that extracellular matrix-receptor interaction and transforming growth factor-β (TGF-β) signaling pathways were enriched among the up-regulated genes by SA. Mechanistically, we demonstrated that SA transcriptionally activated Smad3. In addition, we found that SA inhibited the overproduction of lipopolysaccharide-induced inflammation-related cytokines including tumor necrosis factor-α and interleukin-1β, as well as matrix metalloproteinase 3 and matrix metalloproteinase 13. The cell apoptosis and nuclear factor-kappa B (NF-κB) signaling were also inhibited by SA treatment. Most importantly, SA attenuated cartilage degradation in a mouse OA model. Taken together, our study demonstrated that SA could alleviate cartilage degradation in OA by activating the TGF-β/Smad and inhibiting NF-κB signaling pathway.
Collapse
Affiliation(s)
- Min Wang
- Key Laboratory of Orthopaedics and Traumatology, Lingnan Medical Research Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhao Gao
- Er Sha Sports Training Center of Guangdong Province, Guangzhou, China
| | - Yage Zhang
- Key Laboratory of Orthopaedics and Traumatology, Lingnan Medical Research Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qiangqiang Zhao
- Key Laboratory of Orthopaedics and Traumatology, Lingnan Medical Research Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xinfang Tan
- Key Laboratory of Orthopaedics and Traumatology, Lingnan Medical Research Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Siluo Wu
- Key Laboratory of Orthopaedics and Traumatology, Lingnan Medical Research Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lingli Ding
- Key Laboratory of Orthopaedics and Traumatology, Lingnan Medical Research Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yamei Liu
- School of Basic Medical Science, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shengnan Qin
- School of Biomedical Science, The University of Western Australia, Perth, Western Australia, Australia
| | - Jiangyong Gu
- School of Basic Medical Science, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Liangliang Xu
- Key Laboratory of Orthopaedics and Traumatology, Lingnan Medical Research Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
108
|
Balusani P, Shrivastava S, Pundkar A, Kale P. Navigating the Therapeutic Landscape: A Comprehensive Review of Platelet-Rich Plasma and Bone Marrow Aspirate Concentrate in Knee Osteoarthritis. Cureus 2024; 16:e54747. [PMID: 38524005 PMCID: PMC10960965 DOI: 10.7759/cureus.54747] [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: 12/07/2023] [Accepted: 02/23/2024] [Indexed: 03/26/2024] Open
Abstract
This comprehensive review provides an in-depth analysis of platelet-rich plasma (PRP) and bone marrow aspirate concentrate (BMAC) as potential treatments for knee osteoarthritis. It explores their mechanisms of action, clinical efficacy, safety considerations, and the importance of personalised treatment approaches. The review highlights promising findings regarding the ability of PRP and BMAC to alleviate symptoms, improve joint function, and potentially slow disease progression. It emphasises the need for further research into long-term outcomes, direct comparative studies, protocol standardisation, biomarker identification, and cost-effectiveness assessments to enhance clinical practice. While the review does not directly compare PRP and BMAC, it provides valuable insights into their respective roles in knee osteoarthritis management. The review aims to contribute to evidence-based advancements in regenerative therapies for knee osteoarthritis by addressing critical research priorities and refining treatment strategies.
Collapse
Affiliation(s)
- Prashanth Balusani
- Orthopaedics and Traumatology, Jawaharlal Nehru Medical College, Datta Meghe Institute of Medical Science, Wardha, IND
| | - Sandeep Shrivastava
- Orthopaedic Surgery, Jawaharlal Nehru Medical College, Datta Meghe Institute of Medical Science, Wardha, IND
| | - Aditya Pundkar
- Orthopaedics, Jawaharlal Nehru Medical College, Datta Meghe Institute of Medical Science, Wardha, IND
| | - Prathamesh Kale
- Orthopaedic Surgery, Jawaharlal Nehru Medical College, Datta Meghe Institute of Medical Science, Wardha, IND
| |
Collapse
|
109
|
Yang J, Li S, Li Z, Yao L, Liu M, Tong K, Xu Q, Yu B, Peng R, Gui T, Tang W, Xu Y, Chen J, He J, Zhao K, Wang X, Wang X, Zha Z, Zhang H. Targeting YAP1-regulated Glycolysis in Fibroblast-Like Synoviocytes Impairs Macrophage Infiltration to Ameliorate Diabetic Osteoarthritis Progression. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2304617. [PMID: 38044289 PMCID: PMC10837355 DOI: 10.1002/advs.202304617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 11/03/2023] [Indexed: 12/05/2023]
Abstract
The interplay between immune cells/macrophages and fibroblast-like synoviocytes (FLSs) plays a pivotal role in initiating synovitis; however, their involvement in metabolic disorders, including diabetic osteoarthritis (DOA), is largely unknown. In this study, single-cell RNA sequencing (scRNA-seq) is employed to investigate the synovial cell composition of DOA. A significant enrichment of activated macrophages within eight distinct synovial cell clusters is found in DOA synovium. Moreover, it is demonstrated that increased glycolysis in FLSs is a key driver for DOA patients' synovial macrophage infiltration and polarization. In addition, the yes-associated protein 1 (YAP1)/thioredoxin-interacting protein (TXNIP) signaling axis is demonstrated to play a crucial role in regulating glucose transporter 1 (GLUT1)-dependent glycolysis in FLSs, thereby controlling the expression of a series of adhesion molecules such as intercellular adhesion molecule-1 (ICAM-1) which may subsequently fine-tune the infiltration of M1-polarized synovial macrophages in DOA patients and db/db diabetic OA mice. For treatment, M1 macrophage membrane-camouflaged Verteporfin (Vt)-loaded PLGA nanoparticles (MVPs) are developed to ameliorate DOA progression by regulating the YAP1/TXNIP signaling axis, thus suppressing the synovial glycolysis and the infiltration of M1-polarized macrophages. The results provide several novel insights into the pathogenesis of DOA and offer a promising treatment approach for DOA.
Collapse
Affiliation(s)
- Jie Yang
- Department of Bone and Joint Surgerythe First Affiliated Hospital of Jinan UniversityKey Laboratory of Regenerative Medicine of Ministry of EducationJinan UniversityGuangzhouGuangdong510630China
| | - Shanshan Li
- State Key Laboratory of Pulp and Paper EngineeringSouth China University of TechnologyGuangzhou510640China
| | - Zhenyan Li
- Department of Bone and Joint Surgerythe First Affiliated Hospital of Jinan UniversityKey Laboratory of Regenerative Medicine of Ministry of EducationJinan UniversityGuangzhouGuangdong510630China
| | - Lutian Yao
- Department of OrthopedicsThe First Hospital of China Medical UniversityShenyang110001China
| | - Meijing Liu
- Key Laboratory of Big Data‐Based Precision MedicineSchool of Engineering MedicineBeihang UniversityBeijing100191China
- Clinical Research Platform for Interdisciplinary of Stomatologythe First Affiliated Hospital of Jinan University and Department of StomatologyJinan UniversityGuangzhou510632China
| | - Kui‐Leung Tong
- Department of Bone and Joint Surgerythe First Affiliated Hospital of Jinan UniversityKey Laboratory of Regenerative Medicine of Ministry of EducationJinan UniversityGuangzhouGuangdong510630China
| | - Qiutong Xu
- Department of Bone and Joint Surgerythe First Affiliated Hospital of Jinan UniversityKey Laboratory of Regenerative Medicine of Ministry of EducationJinan UniversityGuangzhouGuangdong510630China
| | - Bo Yu
- Department of Bone and Joint Surgerythe First Affiliated Hospital of Jinan UniversityKey Laboratory of Regenerative Medicine of Ministry of EducationJinan UniversityGuangzhouGuangdong510630China
| | - Rui Peng
- Department of Bone and Joint Surgerythe First Affiliated Hospital of Jinan UniversityKey Laboratory of Regenerative Medicine of Ministry of EducationJinan UniversityGuangzhouGuangdong510630China
| | - Tao Gui
- Department of Bone and Joint Surgerythe First Affiliated Hospital of Jinan UniversityKey Laboratory of Regenerative Medicine of Ministry of EducationJinan UniversityGuangzhouGuangdong510630China
| | - Wang Tang
- Department of Bone and Joint Surgerythe First Affiliated Hospital of Jinan UniversityKey Laboratory of Regenerative Medicine of Ministry of EducationJinan UniversityGuangzhouGuangdong510630China
| | - Yidi Xu
- Department of Bone and Joint Surgerythe First Affiliated Hospital of Jinan UniversityKey Laboratory of Regenerative Medicine of Ministry of EducationJinan UniversityGuangzhouGuangdong510630China
| | - Jiaxu Chen
- Guangzhou Key Laboratory of Formula‐Pattern Research CenterSchool of Traditional Chinese MedicineJinan UniversityGuangzhou510640China
| | - Jun He
- Institute of Laboratory Animal ScienceJinan UniversityGuangzhou510632China
| | - Kewei Zhao
- Guangzhou Key Laboratory of Chinese Medicine Research on Prevention and Treatment of Osteoporosisthe Third Affiliated Hospital of Guangzhou University of Chinese MedicineGuangzhou510375China
| | - Xiaogang Wang
- Key Laboratory of Big Data‐Based Precision MedicineSchool of Engineering MedicineBeihang UniversityBeijing100191China
- Clinical Research Platform for Interdisciplinary of Stomatologythe First Affiliated Hospital of Jinan University and Department of StomatologyJinan UniversityGuangzhou510632China
| | - Xiaoying Wang
- State Key Laboratory of Pulp and Paper EngineeringSouth China University of TechnologyGuangzhou510640China
| | - Zhengang Zha
- Department of Bone and Joint Surgerythe First Affiliated Hospital of Jinan UniversityKey Laboratory of Regenerative Medicine of Ministry of EducationJinan UniversityGuangzhouGuangdong510630China
| | - Huan‐Tian Zhang
- Department of Bone and Joint Surgerythe First Affiliated Hospital of Jinan UniversityKey Laboratory of Regenerative Medicine of Ministry of EducationJinan UniversityGuangzhouGuangdong510630China
- Guangzhou Key Laboratory of Chinese Medicine Research on Prevention and Treatment of Osteoporosisthe Third Affiliated Hospital of Guangzhou University of Chinese MedicineGuangzhou510375China
| |
Collapse
|
110
|
Xiao T, Cheng X, Zhi Y, Tian F, Wu A, Huang F, Tao L, Guo Z, Shen X. Ameliorative effect of Alangium chinense (Lour.) Harms on rheumatoid arthritis by reducing autophagy with targeting regulate JAK3-STAT3 and COX-2 pathways. JOURNAL OF ETHNOPHARMACOLOGY 2024; 319:117133. [PMID: 37690476 DOI: 10.1016/j.jep.2023.117133] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 08/30/2023] [Accepted: 09/04/2023] [Indexed: 09/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Alangium chinense has been used as traditional folk medicine for centuries to treat rheumatoid arthritis (RA) by Guizhou Miao nationality with remarkable clinical effect. But the mechanism of its anti-RA is not fully clarified. AIM OF THE STUDY To explore the effect and underlying mechanism of A. chinense against RA. MATERIAL AND METHODS RA rats were induced by CII/IFA, and oral administrated with or without ethyl acetate extracts of Alangium chinense (ACEE) and tripterygium glycosides (GTW). Then arthritis scores, inflammatory factors in serum and histological evaluation were evaluated to assess the degree of joints disease. Proteomics were conducted via LC-MS/MS to clarify the mechanism of ACEE preliminarily, and further examined by immunohistochemistry, immunofluorescence, western botting, and molecular docking. RESULTS ACEE decreased joints swelling, cell abscission and necrosis of joint tissues arthropathy of RA rats, and attenuated expression of TNF-α, IL-1β, IL-6, PGE2, TGF-β. Meanwhile, differentially expressed proteins in the ACEE treated groups were observed, which were involved in RA, spliceosome, cell adhesion molecules, phagosome and lysosome signaling pathways. Moreover, ACEE significantly ameliorated arthropathy, suppressed JAK-STAT pathway (JAK3, p-JAK3, STAT3, iNOS, RANKL), COX-2 pathway (COX-2, TNF-α, IL-6I, L-1β, 5-LOX), and autophagic signaling pathway (LC3-Ⅰ, LC3-Ⅱ, p62, mTOR). But it showed little effect on the expression of COX-1, JAK1, JAK2, TyK2. CONCLUSION It is the first evidence that A. chinense significantly ameliorates RA, and the underlying immune mechanism involves reducing autophagy with targeting regulate JAK3-STAT3 and COX-2 pathways.
Collapse
Affiliation(s)
- Ting Xiao
- The State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550025, China; The Department of Pharmacology of Materia Medica (The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province and The High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550031, China; The Key Laboratory of Optimal Utilization of Natural Medicine Resources (The Union Key Laboratory of Guiyang City-Guizhou Medical University), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550031, China.
| | - Xingyan Cheng
- The State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550025, China; The Department of Pharmacology of Materia Medica (The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province and The High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550031, China; The Key Laboratory of Optimal Utilization of Natural Medicine Resources (The Union Key Laboratory of Guiyang City-Guizhou Medical University), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550031, China.
| | - Yuan Zhi
- The State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550025, China; The Department of Pharmacology of Materia Medica (The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province and The High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550031, China; The Key Laboratory of Optimal Utilization of Natural Medicine Resources (The Union Key Laboratory of Guiyang City-Guizhou Medical University), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550031, China.
| | - Fangfang Tian
- The State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550025, China; The Department of Pharmacology of Materia Medica (The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province and The High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550031, China; The Key Laboratory of Optimal Utilization of Natural Medicine Resources (The Union Key Laboratory of Guiyang City-Guizhou Medical University), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550031, China.
| | - Ai Wu
- The State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550025, China; The Department of Pharmacology of Materia Medica (The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province and The High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550031, China; The Key Laboratory of Optimal Utilization of Natural Medicine Resources (The Union Key Laboratory of Guiyang City-Guizhou Medical University), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550031, China.
| | - Feilong Huang
- The State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550025, China; The Department of Pharmacology of Materia Medica (The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province and The High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550031, China; The Key Laboratory of Optimal Utilization of Natural Medicine Resources (The Union Key Laboratory of Guiyang City-Guizhou Medical University), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550031, China.
| | - Ling Tao
- The State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550025, China; The Department of Pharmacology of Materia Medica (The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province and The High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550031, China; The Key Laboratory of Optimal Utilization of Natural Medicine Resources (The Union Key Laboratory of Guiyang City-Guizhou Medical University), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550031, China.
| | - Zhenghong Guo
- School of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China.
| | - Xiangchun Shen
- The State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550025, China; The Department of Pharmacology of Materia Medica (The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province and The High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550031, China; The Key Laboratory of Optimal Utilization of Natural Medicine Resources (The Union Key Laboratory of Guiyang City-Guizhou Medical University), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550031, China.
| |
Collapse
|
111
|
Liu ZF, Zhang Y, Liu J, Wang YY, Chen M, Liu EY, Guo JM, Wang YH, Weng ZW, Liu CX, Yu CH, Wang XY. Effect of Traditional Chinese Non-Pharmacological Therapies on Knee Osteoarthritis: A Narrative Review of Clinical Application and Mechanism. Orthop Res Rev 2024; 16:21-33. [PMID: 38292459 PMCID: PMC10826518 DOI: 10.2147/orr.s442025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 01/16/2024] [Indexed: 02/01/2024] Open
Abstract
Knee osteoarthritis (KOA) stands as a degenerative ailment with a substantial and escalating prevalence. The practice of traditional Chinese non-pharmacological therapy has become a prevalent complementary and adjunctive approach. A mounting body of evidence suggests its efficacy in addressing KOA. Recent investigations have delved into its underlying mechanism, yielding some headway. Consequently, this comprehensive analysis seeks to encapsulate the clinical application and molecular mechanism of traditional Chinese non-pharmacological therapy in KOA treatment. The review reveals that various therapies, such as acupuncture, electroacupuncture, warm needle acupuncture, tuina, and acupotomy, primarily target localized knee components like cartilage, subchondral bone, and synovium. Moreover, their impact extends to the central nervous system and intestinal flora. More perfect experimental design and more comprehensive research remain a promising avenue in the future.
Collapse
Affiliation(s)
- Zhi-Feng Liu
- Tuina and Pain Management Department, Beijing University of Chinese Medicine Affilliated Dongzhimen Hospital, Beijing, People’s Republic of China
| | - Yang Zhang
- Tuina and Pain Management Department, Beijing University of Chinese Medicine Affilliated Dongzhimen Hospital, Beijing, People’s Republic of China
| | - Jing Liu
- Tuina and Pain Management Department, Beijing University of Chinese Medicine Affilliated Dongzhimen Hospital, Beijing, People’s Republic of China
| | - Yu-Yan Wang
- Tuina and Pain Management Department, Beijing University of Chinese Medicine Affilliated Dongzhimen Hospital, Beijing, People’s Republic of China
| | - Mo Chen
- Tuina and Pain Management Department, Beijing University of Chinese Medicine Affilliated Dongzhimen Hospital, Beijing, People’s Republic of China
| | - Er-Yang Liu
- Tuina and Pain Management Department, Beijing University of Chinese Medicine Affilliated Dongzhimen Hospital, Beijing, People’s Republic of China
| | - Jun-Ming Guo
- Tuina and Pain Management Department, Beijing University of Chinese Medicine Affilliated Dongzhimen Hospital, Beijing, People’s Republic of China
| | - Yan-Hua Wang
- Tuina and Pain Management Department, Beijing University of Chinese Medicine Affilliated Dongzhimen Hospital, Beijing, People’s Republic of China
| | - Zhi-Wen Weng
- Tuina and Pain Management Department, Beijing University of Chinese Medicine Affilliated Dongzhimen Hospital, Beijing, People’s Republic of China
| | - Chang-Xin Liu
- Tuina and Pain Management Department, Beijing University of Chinese Medicine Affilliated Dongzhimen Hospital, Beijing, People’s Republic of China
| | - Chang-He Yu
- Tuina and Pain Management Department, Beijing University of Chinese Medicine Affilliated Dongzhimen Hospital, Beijing, People’s Republic of China
| | - Xi-You Wang
- Tuina and Pain Management Department, Beijing University of Chinese Medicine Affilliated Dongzhimen Hospital, Beijing, People’s Republic of China
| |
Collapse
|
112
|
Hannani MT, Thudium CS, Karsdal MA, Ladel C, Mobasheri A, Uebelhoer M, Larkin J, Bacardit J, Struglics A, Bay-Jensen AC. From biochemical markers to molecular endotypes of osteoarthritis: a review on validated biomarkers. Expert Rev Mol Diagn 2024; 24:23-38. [PMID: 38353446 DOI: 10.1080/14737159.2024.2315282] [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: 02/02/2024] [Indexed: 02/22/2024]
Abstract
INTRODUCTION Osteoarthritis (OA) affects over 500 million people worldwide. OA patients are symptomatically treated, and current therapies exhibit marginal efficacy and frequently carry safety-risks associated with chronic use. No disease-modifying therapies have been approved to date leaving surgical joint replacement as a last resort. To enable effective patient care and successful drug development there is an urgent need to uncover the pathobiological drivers of OA and how these translate into disease endotypes. Endotypes provide a more precise and mechanistic definition of disease subgroups than observable phenotypes, and a panel of tissue- and pathology-specific biochemical markers may uncover treatable endotypes of OA. AREAS COVERED We have searched PubMed for full-text articles written in English to provide an in-depth narrative review of a panel of validated biochemical markers utilized for endotyping of OA and their association to key OA pathologies. EXPERT OPINION As utilized in IMI-APPROACH and validated in OAI-FNIH, a panel of biochemical markers may uncover disease subgroups and facilitate the enrichment of treatable molecular endotypes for recruitment in therapeutic clinical trials. Understanding the link between biochemical markers and patient-reported outcomes and treatable endotypes that may respond to given therapies will pave the way for new drug development in OA.
Collapse
Affiliation(s)
- Monica T Hannani
- ImmunoScience, Nordic Bioscience A/S, Herlev, Denmark
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | | | | | - Ali Mobasheri
- Research Unit of Health Sciences and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania
- Department of Joint Surgery, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- World Health Organization Collaborating Centre for Public Health Aspects of Musculoskeletal Health and Aging, Université de Liège, Liège, Belgium
| | | | - Jonathan Larkin
- Research Unit of Health Sciences and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland
- SynOA Therapeutics, Philadelphia, PA, USA
| | - Jaume Bacardit
- School of Computing, Newcastle University, Newcastle upon Tyne, UK
| | - André Struglics
- Department of Clinical Sciences, Orthopaedics, Lund University, Lund, Sweden
| | | |
Collapse
|
113
|
Hu S, Liang Y, Chen J, Gao X, Zheng Y, Wang L, Jiang J, Zeng M, Luo M. Mechanisms of hydrogel-based microRNA delivery systems and its application strategies in targeting inflammatory diseases. J Tissue Eng 2024; 15:20417314241265897. [PMID: 39092451 PMCID: PMC11292707 DOI: 10.1177/20417314241265897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 06/18/2024] [Indexed: 08/04/2024] Open
Abstract
Hydrogels, composed of three-dimensional polymer networks, are excellent delivery carriers and have been extensively employed in the biomedical field. Inflammation acts as a protective mechanism to prevent harmful substances from entering living organisms, but chronic, long-lasting inflammation can cause oxidative stress, which damages tissue and organs and adversely affects patients' quality of life. The aberrant expression of microRNAs (miRNAs) has been found to play a significant part in the etiology and progression of inflammatory diseases, as suggested by growing evidence. Numerous hydrogels that can act as gene carriers for the intracellular delivery of miRNA have been described during ongoing research into innovative hydrogel materials. MiRNA hydrogel delivery systems, which are loaded with exogenous miRNA inhibitors or mimics, enable targeted miRNA intervention in inflammatory diseases and effectively prevent environmental stressors from degrading or inactivating miRNA. In this review, we summarize the classification of miRNA hydrogel delivery systems, the basic strategies and mechanisms for loading miRNAs into hydrogels, highlight the biomedical applications of miRNA hydrogel delivery systems in inflammatory diseases, and share our viewpoints on potential opportunities and challenges in the promising region of miRNA delivery systems. These findings may provide a new theoretical basis for the prevention and treatment of inflammation-related diseases and lay the foundation for clinical translation.
Collapse
Affiliation(s)
- Shaorun Hu
- Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Southwest Medical University, Luzhou, Sichuan, China
- Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
- Municipal Key Laboratory of Thrombosis and Vascular Biology, Luzhou, Sichuan, China
| | - Yu Liang
- Integrated Traditional Chinese and Western Medicine, Affiliated Hospital of Traditional Chinese Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - Jinxiang Chen
- Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Southwest Medical University, Luzhou, Sichuan, China
- Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
- Municipal Key Laboratory of Thrombosis and Vascular Biology, Luzhou, Sichuan, China
| | - Xiaojun Gao
- Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Southwest Medical University, Luzhou, Sichuan, China
- Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
- Municipal Key Laboratory of Thrombosis and Vascular Biology, Luzhou, Sichuan, China
| | - Youkun Zheng
- Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Southwest Medical University, Luzhou, Sichuan, China
- Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
- Municipal Key Laboratory of Thrombosis and Vascular Biology, Luzhou, Sichuan, China
| | - Liqun Wang
- Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Southwest Medical University, Luzhou, Sichuan, China
- Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
- Municipal Key Laboratory of Thrombosis and Vascular Biology, Luzhou, Sichuan, China
| | - Jun Jiang
- Department of General Surgery (Thyroid Surgery), The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
- Metabolic Vascular Diseases Key Laboratory of Sichuan Province, Luzhou, Sichuan, China
| | - Min Zeng
- Department of Pharmacy, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Mao Luo
- Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Southwest Medical University, Luzhou, Sichuan, China
- Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
- Municipal Key Laboratory of Thrombosis and Vascular Biology, Luzhou, Sichuan, China
| |
Collapse
|
114
|
Patnaik R, Riaz S, Sivani BM, Faisal S, Naidoo N, Rizzo M, Banerjee Y. Evaluating the potential of Vitamin D and curcumin to alleviate inflammation and mitigate the progression of osteoarthritis through their effects on human chondrocytes: A proof-of-concept investigation. PLoS One 2023; 18:e0290739. [PMID: 38157375 PMCID: PMC10756552 DOI: 10.1371/journal.pone.0290739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 08/14/2023] [Indexed: 01/03/2024] Open
Abstract
Osteoarthritis (OA) is a chronic degenerative joint disorder primarily affecting the elderly, characterized by a prominent inflammatory component. The long-term side effects associated with current therapeutic approaches necessitate the development of safer and more efficacious alternatives. Nutraceuticals, such as Vitamin D and curcumin, present promising therapeutic potentials due to their safety, efficacy, and cost-effectiveness. In this study, we utilized a proinflammatory human chondrocyte model of OA to assess the anti-inflammatory properties of Vitamin D and curcumin, with a particular focus on the Protease-Activated Receptor-2 (PAR-2) mediated inflammatory pathway. Employing a robust siRNA approach, we effectively modulated the expression of PAR-2 to understand its role in the inflammatory process. Our results reveal that both Vitamin D and curcumin attenuate the expression of PAR-2, leading to a reduction in the downstream proinflammatory cytokines, such as Tumor Necrosis Factor-alpha (TNF-α), Interleukin 6 (IL-6), and Interleukin 8 (IL-8), implicated in the OA pathogenesis. Concurrently, these compounds suppressed the expression of Receptor Activator of Nuclear Factor kappa-Β Ligand (RANKL) and its receptor RANK, which are associated with PAR-2 mediated TNF-α stimulation. Additionally, Vitamin D and curcumin downregulated the expression of Interferon gamma (IFN-γ), known to elevate RANKL levels, underscoring their potential therapeutic implications in OA. This study, for the first time, provides evidence of the mitigating effect of Vitamin D and curcumin on PAR-2 mediated inflammation, employing an siRNA approach in OA. Thus, our findings pave the way for future research and the development of novel, safer, and more effective therapeutic strategies for managing OA.
Collapse
Affiliation(s)
- Rajashree Patnaik
- College of Medicine and Health Sciences, Mohammed Bin Rashid University of Medicine, and Health Sciences (MBRU), Dubai, United Arab Emirates
| | - Sumbal Riaz
- College of Medicine and Health Sciences, Mohammed Bin Rashid University of Medicine, and Health Sciences (MBRU), Dubai, United Arab Emirates
| | - Bala Mohan Sivani
- College of Medicine and Health Sciences, Mohammed Bin Rashid University of Medicine, and Health Sciences (MBRU), Dubai, United Arab Emirates
| | - Shemima Faisal
- College of Medicine and Health Sciences, Mohammed Bin Rashid University of Medicine, and Health Sciences (MBRU), Dubai, United Arab Emirates
| | - Nerissa Naidoo
- College of Medicine and Health Sciences, Mohammed Bin Rashid University of Medicine, and Health Sciences (MBRU), Dubai, United Arab Emirates
| | - Manfredi Rizzo
- Department of Health Promotion, Mother and Child Care, Internal Medicine, and Medical Specialties (Promise), University of Palermo, Palermo, Italy
| | - Yajnavalka Banerjee
- College of Medicine and Health Sciences, Mohammed Bin Rashid University of Medicine, and Health Sciences (MBRU), Dubai, United Arab Emirates
- Centre for Medical Education, University of Dundee, Dundee, United Kingdom
| |
Collapse
|
115
|
Khan NM, Diaz-Hernandez ME, Martin WN, Patel B, Chihab S, Drissi H. pH-sensing G protein-coupled orphan receptor GPR68 is expressed in human cartilage and correlates with degradation of extracellular matrix during OA progression. PeerJ 2023; 11:e16553. [PMID: 38077417 PMCID: PMC10704986 DOI: 10.7717/peerj.16553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Accepted: 11/09/2023] [Indexed: 12/18/2023] Open
Abstract
Background Osteoarthritis (OA) is a debilitating joints disease affecting millions of people worldwide. As OA progresses, chondrocytes experience heightened catabolic activity, often accompanied by alterations in the extracellular environment's osmolarity and acidity. Nevertheless, the precise mechanism by which chondrocytes perceive and respond to acidic stress remains unknown. Recently, there has been growing interest in pH-sensing G protein-coupled receptors (GPCRs), such as GPR68, within musculoskeletal tissues. However, function of GPR68 in cartilage during OA progression remains unknown. This study aims to identify the role of GPR68 in regulation of catabolic gene expression utilizing an in vitro model that simulates catabolic processes in OA. Methods We examined the expression of GPCR by analyzing high throughput RNA-Seq data in human cartilage isolated from healthy donors and OA patients. De-identified and discarded OA cartilage was obtained from joint arthroplasty and chondrocytes were prepared by enzymatic digestion. Chondrocytes were treated with GPR68 agonist, Ogerin and then stimulated IL1β and RNA isolation was performed using Trizol method. Reverse transcription was done using the cDNA synthesis kit and the expression of GPR68 and OA related catabolic genes was quantified using SYBR® green assays. Results The transcriptome analysis revealed that pH sensing GPCR were expressed in human cartilage with a notable increase in the expression of GPR68 in OA cartilage which suggest a potential role for GPR68 in the pathogenesis of OA. Immunohistochemical (IHC) and qPCR analyses in human cartilage representing various stages of OA indicated a progressive increase in GPR68 expression in cartilage associated with higher OA grades, underscoring a correlation between GPR68 expression and the severity of OA. Furthermore, IHC analysis of Gpr68 in murine cartilage subjected to surgically induced OA demonstrated elevated levels of GPR68 in knee cartilage and meniscus. Using IL1β stimulated in vitro model of OA catabolism, our qPCR analysis unveiled a time-dependent increase in GPR68 expression in response to IL1β stimulation, which correlates with the expression of matrix degrading proteases suggesting the role of GPR68 in chondrocytes catabolism and matrix degeneration. Using pharmacological activator of GPR68, our results further showed that GPR68 activation repressed the expression of MMPs in human chondrocytes. Conclusions Our results demonstrated that GPR68 was robustly expressed in human cartilage and mice and its expression correlates with matrix degeneration and severity of OA progression in human and surgical model. GPR68 activation in human chondrocytes further repressed the expression of MMPs under OA pathological condition. These results identify GPR68 as a possible therapeutic target in the regulation of matrix degradation during OA.
Collapse
Affiliation(s)
- Nazir M. Khan
- Orthopaedics, Emory University, Atlanta, GA, United States
| | | | | | - Bhakti Patel
- Orthopaedics, Emory University, Atlanta, GA, United States
| | - Samir Chihab
- Orthopaedics, Emory University, Atlanta, GA, United States
| | - Hicham Drissi
- Orthopaedics, Emory University, Atlanta, GA, United States
| |
Collapse
|
116
|
Chun JM, Nam H, Lee JH, Seo YH, Kim HS, Moon BC, Park JH. Chondroprotective effects of Protaetia brevitarsis seulensis larvae as an edible insect on osteoarthritis in mice. Food Sci Nutr 2023; 11:7887-7899. [PMID: 38107146 PMCID: PMC10724628 DOI: 10.1002/fsn3.3706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 09/05/2023] [Accepted: 09/08/2023] [Indexed: 12/19/2023] Open
Abstract
Osteoarthritis (OA) is a common chronic joint inflammatory disease characterized by progressive destruction of the articular cartilage, bone remodeling, and excessive chronic pain. Most therapeutic approaches do not rescue the progression of OA effectively or provide relief of symptoms. Protaetia brevitarsis seulensis larva (PBSL), which is attracting attention, is an edible insect with very high nutritional value and herbal medicine for the treatment of blood stasis, hepatic disease, and various inflammatory diseases. However, the effect of PBSL on OA has not yet been investigated. This study aimed to demonstrate the effects of PBSL water extract on the progression of OA using monosodium iodoacetate (MIA)-induced mice and SW1353 chondrocytes or murine macrophages. We injected MIA into the intraarticular area of mice following pretreatment with either saline or PBSL (200 mg/kg) for 2 weeks, and then locomotor activity, microcomputed tomography and histopathological analysis, quantitative reverse transcriptase-polymerase chain reaction analysis, and western blot analysis were performed. To determine the molecular effects of PBSL, we used interleukin-1β (IL-1β)-induced SW1353 chondrosarcoma or lipopolysaccharide (LPS)-stimulated macrophages. Pretreatment with PBSL diminished the symptoms of OA. Physical activity, articular cartilage damage, and the generation of microfractures were rescued by pretreatment with PBSL in the mouse model. Pretreatment with PBSL suppressed the progress of OA through the regulation of articular cartilage degradation genes and inflammation in both in vivo and in vitro models. Our results demonstrated that PBSL has value as edible insect that can be used in the development of functional foods for OA.
Collapse
Affiliation(s)
- Jin Mi Chun
- Herbal Medicine Resources Research CenterKorea Institute of Oriental MedicineNaju‐siRepublic of Korea
| | - Hyeon‐Hwa Nam
- Herbal Medicine Resources Research CenterKorea Institute of Oriental MedicineNaju‐siRepublic of Korea
| | - Ji Hye Lee
- Herbal Medicine Resources Research CenterKorea Institute of Oriental MedicineNaju‐siRepublic of Korea
- School of Korean MedicinePusan National UniversityBusan‐siRepublic of Korea
| | - Young Hye Seo
- Herbal Medicine Resources Research CenterKorea Institute of Oriental MedicineNaju‐siRepublic of Korea
| | - Hyo Seon Kim
- Herbal Medicine Resources Research CenterKorea Institute of Oriental MedicineNaju‐siRepublic of Korea
| | - Byeong Cheol Moon
- Herbal Medicine Resources Research CenterKorea Institute of Oriental MedicineNaju‐siRepublic of Korea
| | - Jun Hong Park
- Herbal Medicine Resources Research CenterKorea Institute of Oriental MedicineNaju‐siRepublic of Korea
| |
Collapse
|
117
|
Rahman SO, Bariguian F, Mobasheri A. The Potential Role of Probiotics in the Management of Osteoarthritis Pain: Current Status and Future Prospects. Curr Rheumatol Rep 2023; 25:307-326. [PMID: 37656392 PMCID: PMC10754743 DOI: 10.1007/s11926-023-01108-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/07/2023] [Indexed: 09/02/2023]
Abstract
PURPOSE OF REVIEW This narrative review article comprehensively explains the pathophysiology of osteoarthritis (OA) pain perception, how the gut microbiota is correlated with it, possible molecular pathways involved in probiotics-mediated OA pain reduction, limitations in the current research approaches, and future perspectives. RECENT FINDINGS The initiation and progression of OA, including the development of chronic pain, is intricately associated with activation of the innate immune system and subsequent inflammatory responses. Trauma, lifestyle (e.g., obesity and metabolic disease), and chronic antibiotic treatment can disrupt commensal homeostasis of the human microbiome, thereby affecting intestinal integrity and promoting leakage of bacterial endotoxins and metabolites such as lipopolysaccharides (LPS) into circulation. Increased level of LPS is associated with knee osteophyte severity and joint pain. Both preclinical and clinical studies strongly suggest that probiotics may benefit patients with OA pain through positive gut microbiota modulation and attenuating low-grade inflammation via multiple pathways. Patent data also suggests increased interest in the development of new innovations that involve probiotic use for reducing OA and joint pain. Recent data suggest that probiotics are attracting more and more attention for OA pain management. The advancement of knowledge in this area may pave the way for developing different probiotic strains that can be used to support joint health, improve treatment outcomes in OA, and reduce the huge impact of the disease on healthcare systems worldwide.
Collapse
Affiliation(s)
| | - Frédérique Bariguian
- Haleon (Formerly GSK Consumer Healthcare), Route de L'Etraz 2, Case Postale 1279, 1260, Nyon 1, Switzerland.
| | - Ali Mobasheri
- Research Unit of Health Sciences and Technology, Faculty of Medicine, University of Oulu, 90014, Oulu, FI, Finland.
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania.
- Department of Joint Surgery, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.
- World Health Organization Collaborating Centre for Public Health Aspects of Musculoskeletal Health and Aging, Liege, Belgium.
| |
Collapse
|
118
|
Estee MM, Cicuttini FM, Page MJ, Wluka AE, Wang Y. Efficacy of tumor necrosis factor inhibitors in hand osteoarthritis: A systematic review and meta-analysis of randomized controlled trials. OSTEOARTHRITIS AND CARTILAGE OPEN 2023; 5:100404. [PMID: 37649531 PMCID: PMC10462838 DOI: 10.1016/j.ocarto.2023.100404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 08/09/2023] [Indexed: 09/01/2023] Open
Abstract
Objectives This study aimed at systematically review the evidence for the efficacy of Tumor Necrosis Factor (TNF) inhibitors on symptoms and structural outcomes in hand osteoarthritis. Methods Three databases were searched for randomized controlled trials examining the efficacy of TNF inhibitors in hand osteoarthritis. Two authors extracted data and assessed the risk of bias. The mean difference (MD) was calculated, and a random-effects meta-analysis was performed. Results Four studies were identified involving 276 participants. Meta-analysis showed that TNF inhibitors had no effect on pain at 4-6 weeks (MD -0.93, 95%CI -7.41 to 5.55; 2 studies) and 24-26 weeks (MD -3.82, 95%CI -11.46 to 3.83; 2 studies) and no effect on grip strength at 12 months (MD -0.35, 95%CI -1.08 to 0.37; 2 studies). There was limited evidence for the effect of TNF inhibitors on structural outcomes at 12 months. Subgroup analysis from 2 studies showed beneficial effect of TNF inhibitors on reducing the progression of structural outcomes in hand OA patients with signs of inflammation but not in those without inflammation. The certainty of the evidence was low for the effect of TNF inhibitor on pain and moderate for the effect on grip strength. Conclusion This study found no effect of TNF inhibitors on clinical outcomes in hand osteoarthritis over the short term (<6 weeks) and within one year, with some evidence for beneficial effect on structural outcomes.
Collapse
Affiliation(s)
- Mahnuma Mahfuz Estee
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria 3004, Australia
| | - Flavia M. Cicuttini
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria 3004, Australia
| | - Matthew J. Page
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria 3004, Australia
| | - Anita E. Wluka
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria 3004, Australia
| | - Yuanyuan Wang
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria 3004, Australia
| |
Collapse
|
119
|
Saha S, Rebouh NY. Anti-Osteoarthritis Mechanism of the Nrf2 Signaling Pathway. Biomedicines 2023; 11:3176. [PMID: 38137397 PMCID: PMC10741080 DOI: 10.3390/biomedicines11123176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 11/23/2023] [Accepted: 11/27/2023] [Indexed: 12/24/2023] Open
Abstract
Osteoarthritis (OA) is a chronic degenerative disease and the primary pathogenic consequence of OA is inflammation, which can affect a variety of tissues including the synovial membrane, articular cartilage, and subchondral bone. The development of the intra-articular microenvironment can be significantly influenced by the shift of synovial macrophages between pro-inflammatory and anti-inflammatory phenotypes. By regulating macrophage inflammatory responses, the NF-κB signaling route is essential in the therapy of OA; whereas, the nuclear factor erythroid 2-related factor 2 (Nrf2) signaling pathway appears to manage the relationship between oxidative stress and inflammation. Additionally, it has been demonstrated that under oxidative stress and inflammation, there is a significant interaction between transcriptional pathways involving Nrf2 and NF-κB. Studying how Nrf2 signaling affects inflammation and cellular metabolism may help us understand how to treat OA by reprogramming macrophage behavior because Nrf2 signaling is thought to affect cellular metabolism. The candidates for treating OA by promoting an anti-inflammatory mechanism by activating Nrf2 are also reviewed in this paper.
Collapse
Affiliation(s)
- Sarmistha Saha
- Department of Biotechnology, Institute of Applied Sciences & Humanities, GLA University, Mathura 281406, Uttar Pradesh, India
| | - Nazih Y. Rebouh
- Department of Environmental Management, Institute of Environmental Engineering, RUDN University, 6 Miklukho-Maklaya St., 117198 Moscow, Russia
| |
Collapse
|
120
|
Bekaryssova D, Yessirkepov M, Mahmudov K. Structure, demography, and medico-social characteristics of articular syndrome in rheumatic diseases: a retrospective monocentric analysis of 2019-2021 data. Rheumatol Int 2023; 43:2057-2064. [PMID: 37624400 DOI: 10.1007/s00296-023-05435-x] [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: 07/20/2023] [Accepted: 08/14/2023] [Indexed: 08/26/2023]
Abstract
Rheumatic diseases encompass a wide range of conditions characterised by joint inflammation and pain, significantly impacting individuals' quality of life. Articular syndrome, manifested through joint-related symptoms such as pain, swelling, and reduced mobility, is a common feature of rheumatic diseases. This study aimed to analyze articular syndrome's structure, demography, and medico-social characteristics in rheumatic diseases. We retrieved case notes of 370 patients examined in 2019-2021 at the Rheumatology Department of the Regional Clinical Hospital, Shymkent, Kazakhstan. We processed data on gender, age, place of residence, social status, clinical diagnosis, comorbid conditions, complications, and delays. The material was counted by frequency analysis. Statistical and mathematical data processing was performed using the SPSS application software package version 26.0 (IBM). The identified rheumatic diseases among the patients included rheumatoid arthritis (183), systemic lupus erythematosus (47), osteoarthritis (42), ankylosing spondylitis (31), systemic scleroderma (30), reactive arthritis (18), gouty arthritis (14), psoriatic arthritis (3), and dermatomyositis (2). The distribution of patients with articular syndrome varied across the study years, with 102 patients in 2019, 216 patients in 2020, and 52 patients in 2021. The study revealed the age distribution of patients, with an average age of 46 at the time of examination and an average age of disease onset at 39. The study further investigated the distribution of rheumatic diseases categorized by gender, place of residence (urban or rural), and disease duration. Additionally, the study examined the prevalence of comorbid conditions and complications related to the underlying rheumatic disease. By examining the structure, demography, and medico-social characteristics of the articular syndrome in patients with rheumatic diseases, this retrospective analysis provides valuable insights into the epidemiological aspects of these conditions. The findings may contribute to a better understanding of the burden of rheumatic diseases on individuals and society. Such knowledge can aid in developing targeted interventions, improving healthcare delivery, and enhancing patients' overall well-being.
Collapse
Affiliation(s)
- Dana Bekaryssova
- Department of Biology and Biochemistry, South Kazakhstan Medical Academy, Shymkent, Kazakhstan.
| | - Marlen Yessirkepov
- Department of Biology and Biochemistry, South Kazakhstan Medical Academy, Shymkent, Kazakhstan
| | - Khaiyom Mahmudov
- Department of Propaedeutics of Internal Diseases, Avicenna Tajik State Medical University, Dushanbe, Tajikistan
| |
Collapse
|
121
|
Tagliero AJ, Foster MJ, Melugin HP, Su CA. Inflammatory and Immunologic Contributions in Femoroacetabular Impingement Syndrome. J Am Acad Orthop Surg 2023; 31:1097-1102. [PMID: 37311428 DOI: 10.5435/jaaos-d-22-01166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Accepted: 05/15/2023] [Indexed: 06/15/2023] Open
Abstract
Femoroacetabular impingement (FAI) is one of the most common causes of labral and early cartilage damage in the nondysplastic hip. FAI is increasingly recognized as a cause for hip and groin pain in the young, active patient, and the surgical treatment of FAI with hip arthroscopy has risen exponentially. Although our understanding of FAI and the progression to degenerative osteoarthritis of the hip has historically been considered a mechanical "wear-and-tear" disease of an imperfectly shaped, aspherical, femoral head within a deep or overcovering acetabulum leading to cartilage injury, our understanding of the intrinsic pathophysiologic mechanisms underlying the development of FAI and joint degeneration of the hip remains poor. For example, many patients with FAI morphology may never develop hip pain or osteoarthritis; there remains more to discover regarding the pathophysiology of arthritis in the setting of FAI. Recent work has begun to identify a strong inflammatory and immunologic component to the FAI disease process that affects the hip synovium, labrum, and cartilage and may be detectable from peripheral clinical samples (blood and urine). This review highlights our current understanding of the inflammatory and immunologic contributions to FAI and potential therapeutic strategies to supplement and augment the surgical management of FAI.
Collapse
Affiliation(s)
- Adam J Tagliero
- From the Department of Orthopaedic Surgery, University of Virginia (Dr. Tagliero, Dr. Su), Charlottesville, VA (Tagliero and Su), the University of Maryland Shore Regional Medical Center, Easton, MD (Foster), and the Ferrell-Duncan Clinic, Springfield, MO (Melugin)
| | | | | | | |
Collapse
|
122
|
Yang L, Ma J, Niu P, Zhang B, Wang Z. Synovial inducible costimulator is correlated with severity in knee osteoarthritis. Scand J Immunol 2023; 98:e13315. [PMID: 38441341 DOI: 10.1111/sji.13315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 07/09/2023] [Accepted: 07/13/2023] [Indexed: 03/07/2024]
Abstract
Osteoarthritis (OA) is a joint disease characterized by articular cartilage loss, which afflicts many people worldwide. Knowing the disease severity can improve the recovery rate of OA. Antibody array technology was utilized for protein expression profiling of synovial fluid from eight mild knee OA patients, eight severe knee OA patients and 16 healthy persons. Subsequently, 48 mild OA patients, 56 severe OA patients and 24 healthy controls were utilized for validation by ELISA. In the protein expression profiling, inducible costimulator (ICOS) levels were markedly higher in OA patients compared with those in the healthy population, and were significantly higher in severe OA than those in mild OA. Furthermore, ICOS levels were shown to be significantly correlated with WOMAC, MRI-MOAKS and MRI-UTE-T2* scores. The multivariate logistic regression analysis indicated that higher levels of ICOS could significantly increase the risk of severe OA. Synovial ICOS levels were positively correlated with the radiographic severity of OA. ICOS may represent a biomarker for predicting the OA severity and may be involved in the development and progression of knee OA.
Collapse
Affiliation(s)
- Lvlin Yang
- Department of Orthopedics, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, Ningxia Province, China
| | - Jun Ma
- Department of Orthopedics, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, Ningxia Province, China
| | - Pengying Niu
- Department of Medical Imaging Center, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, Ningxia Province, China
| | - Bowen Zhang
- Department of Orthopedics, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, Ningxia Province, China
| | - Zhiyan Wang
- Department of Emergency, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, Ningxia Province, China
| |
Collapse
|
123
|
Atwal A, Dale TP, Snow M, Forsyth NR, Davoodi P. Injectable hydrogels: An emerging therapeutic strategy for cartilage regeneration. Adv Colloid Interface Sci 2023; 321:103030. [PMID: 37907031 DOI: 10.1016/j.cis.2023.103030] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 10/17/2023] [Accepted: 10/19/2023] [Indexed: 11/02/2023]
Abstract
The impairment of articular cartilage due to traumatic incidents or osteoarthritis has posed significant challenges for healthcare practitioners, researchers, and individuals suffering from these conditions. Due to the absence of an approved treatment strategy for the complete restoration of cartilage defects to their native state, the tissue condition often deteriorates over time, leading to osteoarthritic (OA). However, recent advancements in the field of regenerative medicine have unveiled promising prospects through the utilization of injectable hydrogels. This versatile class of biomaterials, characterized by their ability to emulate the characteristics of native articular cartilage, offers the distinct advantage of minimally invasive administration directly to the site of damage. These hydrogels can also serve as ideal delivery vehicles for a diverse range of bioactive agents, including growth factors, anti-inflammatory drugs, steroids, and cells. The controlled release of such biologically active molecules from hydrogel scaffolds can accelerate cartilage healing, stimulate chondrogenesis, and modulate the inflammatory microenvironment to halt osteoarthritic progression. The present review aims to describe the methods used to design injectable hydrogels, expound upon their applications as delivery vehicles of biologically active molecules, and provide an update on recent advances in leveraging these delivery systems to foster articular cartilage regeneration.
Collapse
Affiliation(s)
- Arjan Atwal
- School of Pharmacy and Bioengineering, Hornbeam building, Keele University, Staffordshire ST5 5BG, United Kingdom; Guy Hilton Research Centre, School of Pharmacy and Bioengineering, Keele University, Staffordshire ST4 7QB, United Kingdom
| | - Tina P Dale
- School of Pharmacy and Bioengineering, Hornbeam building, Keele University, Staffordshire ST5 5BG, United Kingdom; Guy Hilton Research Centre, School of Pharmacy and Bioengineering, Keele University, Staffordshire ST4 7QB, United Kingdom
| | - Martyn Snow
- Department of Arthroscopy, Royal Orthopaedic Hospital NHS Foundation Trust, Birmingham B31 2AP, United Kingdom; The Robert Jones and Agnes Hunt Hospital, Oswestry, Shropshire SY10 7AG, United Kingdom
| | - Nicholas R Forsyth
- School of Pharmacy and Bioengineering, Hornbeam building, Keele University, Staffordshire ST5 5BG, United Kingdom; Guy Hilton Research Centre, School of Pharmacy and Bioengineering, Keele University, Staffordshire ST4 7QB, United Kingdom; Vice Principals' Office, University of Aberdeen, Kings College, Aberdeen AB24 3FX, United Kingdom
| | - Pooya Davoodi
- School of Pharmacy and Bioengineering, Hornbeam building, Keele University, Staffordshire ST5 5BG, United Kingdom; Guy Hilton Research Centre, School of Pharmacy and Bioengineering, Keele University, Staffordshire ST4 7QB, United Kingdom.
| |
Collapse
|
124
|
Huang SM, Hsieh CY, Ting JU, De Castro-Cruz KA, Wang CC, Lee CJ, Tsai PW. Anti-COVID-19, Anti-Inflammatory, and Anti-Osteoarthritis Activities of Sesamin from Sesamum indicum L. Bioengineering (Basel) 2023; 10:1263. [PMID: 38002386 PMCID: PMC10669907 DOI: 10.3390/bioengineering10111263] [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: 09/07/2023] [Revised: 10/16/2023] [Accepted: 10/19/2023] [Indexed: 11/26/2023] Open
Abstract
During the COVID-19 (coronavirus disease 2019) outbreak, many people were infected, and the symptoms may persist for several weeks or months for recovering patients. This is also known as "long COVID" and includes symptoms such as fatigue, joint pain, muscle pain, et cetera. The COVID-19 virus may trigger hyper-inflammation associated with cytokine levels in the body. COVID-19 can trigger inflammation in the joints, which can lead to osteoarthritis (OA), while long-term COVID-19 symptoms may lead to joint damage and other inflammation problems. According to several studies, sesame has potent anti-inflammatory properties due to its major constituent, sesamin. This study examined sesamin's anti-inflammatory, anti-osteoarthritis, and anti-COVID-19 effects. Moreover, in vivo and in vitro assays were used to determine sesamin's anti-inflammatory activity against the RAW264.7 and SW1353 cell lines. Sesamin had a dose-dependent effect (20 mg/kg) in a monoiodoacetic acid (MIA)-induced osteoarthritis rat model. Sesamin reduced paw swelling and joint discomfort. In addition, the findings indicated that sesamin suppressed the expression of iNOS (inducible nitric oxide synthase) and COX-2 (cyclooxygenase-2) in the RAW264.7 cell line within the concentration range of 6.25-50 μM. Furthermore, sesamin also had a suppressive effect on MMP (matrix metalloproteinase) expression in chondrocytes and the SW1353 cell line within the same concentration range of 6.25-50 μM. To examine the anti-viral activity, an in silico analysis was performed to evaluate sesamin's binding affinity with SARS-CoV-2 RdRp (severe acute respiratory syndrome coronavirus 2 RNA-dependent RNA polymerase) and human ACE2 (angiotensin-converting enzyme 2). Compared to the controls, sesamin exhibited strong binding affinities towards SARS-CoV-2 RdRp and human ACE2. Furthermore, sesamin had a higher binding affinity for the ACE2 target protein. This study suggests that sesamin shows potential anti-SARS-CoV-2 activity for drug development.
Collapse
Affiliation(s)
- Shu-Ming Huang
- Department of Nutrition, College of Medical and Health Care, Hungkuang University, Taichung 433, Taiwan;
- Department of Nutrition, Nantou Hospital of Ministry of Health and Welfare, Nantou 540, Taiwan
| | - Cheng-Yang Hsieh
- Ph.D. Program in Clinical Drug Development of Herbal Medicine, College of Pharmacy, Taipei Medical University, Taipei 110, Taiwan; (C.-Y.H.); (C.-C.W.)
- Laboratory of Oncology, Pharmacy Practice and Sciences, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8577, Japan
| | - Jasmine U. Ting
- Department of Chemistry, College of Science, De La Salle University, Metro Manila 1004, Philippines;
| | - Kathlia A. De Castro-Cruz
- School of Chemical, Biological, and Materials Engineering and Sciences, Mapúa University, Metro Manila 1002, Philippines;
| | - Ching-Chiung Wang
- Ph.D. Program in Clinical Drug Development of Herbal Medicine, College of Pharmacy, Taipei Medical University, Taipei 110, Taiwan; (C.-Y.H.); (C.-C.W.)
- Traditional Herbal Medicine Research Center, Taipei Medical University Hospital, Taipei 110, Taiwan
- Graduate Institute of Pharmacognosy, College of Pharmacy, Taipei Medical University, Taipei 110, Taiwan
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei 110, Taiwan
- Orthopedics Research Center, Taipei Medical University Hospital, Taipei 110, Taiwan
| | - Chia-Jung Lee
- Ph.D. Program in Clinical Drug Development of Herbal Medicine, College of Pharmacy, Taipei Medical University, Taipei 110, Taiwan; (C.-Y.H.); (C.-C.W.)
- Traditional Herbal Medicine Research Center, Taipei Medical University Hospital, Taipei 110, Taiwan
- Graduate Institute of Pharmacognosy, College of Pharmacy, Taipei Medical University, Taipei 110, Taiwan
| | - Po-Wei Tsai
- Department of Medical Science Industries, College of Health Sciences, Chang Jung Christian University, Tainan 711, Taiwan
| |
Collapse
|
125
|
Gupta A. StemOne TM/Stempeucel ®: CDSCO Approved, Adult Human Bone Marrow-Derived, Cultured, Pooled, Allogenic Mesenchymal Stem Cells for Knee Osteoarthritis. Biomedicines 2023; 11:2894. [PMID: 38001895 PMCID: PMC10669254 DOI: 10.3390/biomedicines11112894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 10/25/2023] [Indexed: 11/26/2023] Open
Abstract
The knee, the most likely joint to present osteoarthritis (OA), is accountable for approximately 80% of the global burden of the OA [...].
Collapse
Affiliation(s)
- Ashim Gupta
- Regenerative Orthopaedics, Noida 201301, India;
- Future Biologics, Lawrenceville, GA 30043, USA
| |
Collapse
|
126
|
Kojima Y, Watanabe T. Low-intensity pulsed ultrasound irradiation attenuates collagen degradation of articular cartilage in early osteoarthritis-like model mice. J Exp Orthop 2023; 10:106. [PMID: 37870591 PMCID: PMC10593698 DOI: 10.1186/s40634-023-00672-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 10/11/2023] [Indexed: 10/24/2023] Open
Abstract
PURPOSE Osteoarthritis (OA) is a combination of degeneration and destruction of articular cartilage due to mechanical stress, secondary synovitis, and bone remodelling. In recent years, early knee OA, a preliminary stage of structural failure in OA, has attracted attention as a potential target for therapy to prevent the onset of OA. Intra-articular administration of monoiodoacetic acid (MIA) induces OA-like symptoms, and low doses of MIA induce early OA like symptoms. In this experiment, a low-dose of MIA was induced to early OA model mice, which were then irradiated with low-intensity pulsed ultrasound (LIPUS) to examine whether LIPUS improves symptoms of early OA. METHODS After 4 weeks of LIPUS irradiation, articular cartilage was observed at 1 and 4 weeks. The Osteoarthritis Research Society International (OARSI) scores were calculated using Safranin-O staining results. Cartilage degeneration was detected using Denatured Collagen Detection Reagent (DCDR). RESULTS We observed a significant decrease in OARSI scores in the LIPUS irradiated group at week 4. The non-LIPUS group showed widespread areas of double positivity for Type II collagen and DCDR, whereas the LIPUS group showed only a small number of DCDR-positive areas. In addition, macrophage numbers counted in the articular capsule at week 1 showed a significant decrease in the LIPUS irradiated group. Lubricin detection showed that lubricin positive cell number was significantly increased by LIPUS irradiation at week 4. CONCLUSIONS These results suggest that LIPUS attenuates cartilage degeneration in early OA by relieving inflammation and enhancing the inhibitory effect of lubricin on cartilage degeneration.
Collapse
Affiliation(s)
- Yoshitsugu Kojima
- Clinical Pharmacology Research Laboratory, Yokohama University of Pharmacy, 601 Matanocho Totsukaku, Yokohama, Kanagawa, 245-0066, Japan.
- Planning and Product Development Division, Nippon Sigmax Co., Ltd., 7th Floor, 1-24-1 Nishi-shinjuku, Shinjuku-Ku, Tokyo, 160-0023, Japan.
| | - Takayuki Watanabe
- Clinical Pharmacology Research Laboratory, Yokohama University of Pharmacy, 601 Matanocho Totsukaku, Yokohama, Kanagawa, 245-0066, Japan
- Planning and Product Development Division, Nippon Sigmax Co., Ltd., 7th Floor, 1-24-1 Nishi-shinjuku, Shinjuku-Ku, Tokyo, 160-0023, Japan
| |
Collapse
|
127
|
Salamanna F, Caravelli S, Marchese L, Carniato M, Vocale E, Gardini G, Puccetti G, Mosca M, Giavaresi G. Proprioception and Mechanoreceptors in Osteoarthritis: A Systematic Literature Review. J Clin Med 2023; 12:6623. [PMID: 37892761 PMCID: PMC10607296 DOI: 10.3390/jcm12206623] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 09/26/2023] [Accepted: 10/17/2023] [Indexed: 10/29/2023] Open
Abstract
PURPOSE Osteoarthritis (OA) is one of the most common chronic diseases in the world. It is frequently accompanied by high levels of persistent pain, as well as substantial impairments in function and functional capacity. This review aims to systematically analyze the changes in proprioception and related mechanoreceptors in OA patients. METHODS Studies from September 2013 to September 2023 were identified by conducting searches on the PubMed, Web of Science, and Scopus electronic databases following the PRISMA statement. One reviewer independently assessed and screened the literature, extracted the data, and graded the studies. The body of evidence underwent an evaluation and grading process using the ROBINS-I tool, which was specifically designed to assess the risk of bias in non-randomized studies of interventions. Results were summarized using descriptive methods. RESULTS A search through 37 studies yielded 14 clinical studies that were ultimately included. The primary focus of the studies was on the knee joint, particularly the posterior cruciate ligament (PCL). The studies found that PCL in OA patients had impaired proprioceptive accuracy, possibly due to changes in mechanoreceptors (Ruffini, Pacini, and Golgi Mazzoni corpuscles). This suggests that dysfunctional articular mechanoreceptors, especially in severe cases of OA, may contribute to reduced proprioception. Dynamic stabilometry also identified significant proprioceptive deficits in patients with knee articular cartilage lesions, underscoring the impact of such lesions on knee proprioception. CONCLUSIONS Literature data have shown that proprioceptive accuracy may play an important role in OA, particularly in the knee PCL and cartilage. However, the role of proprioception and related mechanoreceptors needs to be further clarified. Future studies focusing on the relationship between proprioception, OA disease, and symptoms, considering age and gender differences, and exploring OA joints other than the knee should be conducted to improve clinical and surgical outcomes in cases where proprioception and mechanoreceptors are impaired in OA patients.
Collapse
Affiliation(s)
- Francesca Salamanna
- Surgical Sciences and Technologies, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy; (L.M.); (M.C.); (G.G.)
| | - Silvio Caravelli
- IRCCS Istituto Ortopedico Rizzoli, Via Pupilli 1, 40136 Bologna, Italy; (S.C.); (M.M.)
| | - Laura Marchese
- Surgical Sciences and Technologies, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy; (L.M.); (M.C.); (G.G.)
| | - Melania Carniato
- Surgical Sciences and Technologies, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy; (L.M.); (M.C.); (G.G.)
| | - Emanuele Vocale
- 2nd Orthopaedic and Traumatologic Clinic, IRCCS Istituto Ortopedico Rizzoli, Via Pupilli 1, 40136 Bologna, Italy; (E.V.); (G.G.); (G.P.)
| | - Giammarco Gardini
- 2nd Orthopaedic and Traumatologic Clinic, IRCCS Istituto Ortopedico Rizzoli, Via Pupilli 1, 40136 Bologna, Italy; (E.V.); (G.G.); (G.P.)
| | - Giulia Puccetti
- 2nd Orthopaedic and Traumatologic Clinic, IRCCS Istituto Ortopedico Rizzoli, Via Pupilli 1, 40136 Bologna, Italy; (E.V.); (G.G.); (G.P.)
| | - Massimiliano Mosca
- IRCCS Istituto Ortopedico Rizzoli, Via Pupilli 1, 40136 Bologna, Italy; (S.C.); (M.M.)
| | - Gianluca Giavaresi
- Surgical Sciences and Technologies, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy; (L.M.); (M.C.); (G.G.)
| |
Collapse
|
128
|
Tsukahara T, Imamura S, Morohoshi T. A Review of Cyclic Phosphatidic Acid and Other Potential Therapeutic Targets for Treating Osteoarthritis. Biomedicines 2023; 11:2790. [PMID: 37893163 PMCID: PMC10603845 DOI: 10.3390/biomedicines11102790] [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: 09/19/2023] [Revised: 10/09/2023] [Accepted: 10/12/2023] [Indexed: 10/29/2023] Open
Abstract
Osteoarthritis (OA), a chronic degenerative joint disease, is the most common form of arthritis. OA occurs when the protective cartilage that cushions the ends of bones gradually breaks down. This leads to the rubbing of bones against each other, resulting in pain and stiffness. Cyclic phosphatidic acid (cPA) shows promise as a treatment for OA. In this article, we review the most recent findings regarding the biological functions of cPA signaling in mammalian systems, specifically in relation to OA. cPA is a naturally occurring phospholipid mediator with unique cyclic phosphate rings at the sn-2 and sn-3 positions in the glycerol backbone. cPA promotes various responses, including cell proliferation, migration, and survival. cPA possesses physiological activities that are distinct from those elicited by lysophosphatidic acid; however, its biochemical origin has rarely been studied. Although there is currently no cure for OA, advances in medical research may lead to new therapies or strategies in the future, and cPA has potential therapeutic applications.
Collapse
Affiliation(s)
- Tamotsu Tsukahara
- Department of Pharmacology and Therapeutic Innovation, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8521, Japan
| | | | | |
Collapse
|
129
|
Thompson CL, Hopkins T, Bevan C, Screen HRC, Wright KT, Knight MM. Human vascularised synovium-on-a-chip: a mechanically stimulated, microfluidic model to investigate synovial inflammation and monocyte recruitment. Biomed Mater 2023; 18:065013. [PMID: 37703884 DOI: 10.1088/1748-605x/acf976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 09/13/2023] [Indexed: 09/15/2023]
Abstract
Healthy synovium is critical for joint homeostasis. Synovial inflammation (synovitis) is implicated in the onset, progression and symptomatic presentation of arthritic joint diseases such as rheumatoid arthritis and osteoarthritis. Thus, the synovium is a promising target for the development of novel, disease-modifying therapeutics. However, target exploration is hampered by a lack of good pre-clinical models that accurately replicate human physiology and that are developed in a way that allows for widespread uptake. The current study presents a multi-channel, microfluidic, organ-on-a-chip (OOAC) model, comprising a 3D configuration of the human synovium and its associated vasculature, with biomechanical and inflammatory stimulation, built upon a commercially available OOAC platform. Healthy human fibroblast-like synoviocytes (hFLS) were co-cultured with human umbilical vein endothelial cells (HUVECs) with appropriate matrix proteins, separated by a flexible, porous membrane. The model was developed within the Emulate organ-chip platform enabling the application of physiological biomechanical stimulation in the form of fluid shear and cyclic tensile strain. The hFLS exhibited characteristic morphology, cytoskeletal architecture and matrix protein deposition. Synovial inflammation was initiated through the addition of interleukin-1β(IL-1β) into the synovium channel resulting in the increased secretion of inflammatory and catabolic mediators, interleukin-6 (IL-6), prostaglandin E2 (PGE2), matrix metalloproteinase 1 (MMP-1), as well as the synovial fluid constituent protein, hyaluronan. Enhanced expression of the inflammatory marker, intercellular adhesion molecule-1 (ICAM-1), was observed in HUVECs in the vascular channel, accompanied by increased attachment of circulating monocytes. This vascularised human synovium-on-a-chip model recapitulates a number of the functional characteristics of both healthy and inflamed human synovium. Thus, this model offers the first human synovium organ-chip suitable for widespread adoption to understand synovial joint disease mechanisms, permit the identification of novel therapeutic targets and support pre-clinical testing of therapies.
Collapse
Affiliation(s)
- Clare L Thompson
- Centre for Predictive In Vitro Models, Queen Mary University of London, London, United Kingdom
- Centre for Bioengineering, School of Engineering and Materials Science, Queen Mary University of London, London, United Kingdom
| | - Timothy Hopkins
- Centre for Predictive In Vitro Models, Queen Mary University of London, London, United Kingdom
- Centre for Bioengineering, School of Engineering and Materials Science, Queen Mary University of London, London, United Kingdom
- School of Pharmacy and Bioengineering, Keele University, Staffordshire, United Kingdom
- Robert Jones and Agnes Hunt Orthopaedic Hospital, Shropshire, United Kingdom
| | - Catrin Bevan
- Centre for Predictive In Vitro Models, Queen Mary University of London, London, United Kingdom
- Centre for Bioengineering, School of Engineering and Materials Science, Queen Mary University of London, London, United Kingdom
| | - Hazel R C Screen
- Centre for Predictive In Vitro Models, Queen Mary University of London, London, United Kingdom
- Centre for Bioengineering, School of Engineering and Materials Science, Queen Mary University of London, London, United Kingdom
| | - Karina T Wright
- School of Pharmacy and Bioengineering, Keele University, Staffordshire, United Kingdom
- Robert Jones and Agnes Hunt Orthopaedic Hospital, Shropshire, United Kingdom
| | - Martin M Knight
- Centre for Predictive In Vitro Models, Queen Mary University of London, London, United Kingdom
- Centre for Bioengineering, School of Engineering and Materials Science, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
130
|
Moore-Lotridge SN, Hajdu KS, Hou BQ, Gibson BHY, Schoenecker JG. Maintaining the balance: the critical role of plasmin activity in orthopedic surgery injury response. J Thromb Haemost 2023; 21:2653-2665. [PMID: 37558131 PMCID: PMC10926148 DOI: 10.1016/j.jtha.2023.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 08/01/2023] [Accepted: 08/01/2023] [Indexed: 08/11/2023]
Abstract
The musculoskeletal system plays vital roles in the body, facilitating movement, protecting vital structures, and regulating hematopoiesis and mineral metabolism. Injuries to this system are common and can cause chronic pain, loss of range of motion, and disability. The acute phase response (APR) is a complex process necessary for surviving and repairing injured musculoskeletal tissue. To conceptualize the APR, it is useful to divide it into 2 distinct phases, survival and repair. During the survival-APR, a "damage matrix" primarily composed of fibrin, via thrombin activity, is produced to contain the zone of injury. Once containment is achieved, the APR transitions to the repair phase, where reparative inflammatory cells use plasmin to systematically remove the damage matrix and replace it with new permanent matrices produced by differentiated mesenchymal stem cells. The timing of thrombin and plasmin activation during their respective APR phases is crucial for appropriate regulation of the damage matrix. This review focuses on evidence indicating that inappropriate exuberant activation of plasmin during the survival-APR can result in an overactive APR, leading to an "immunocoagulopathy" that may cause "immunothrombosis" and death. Conversely, preclinical data suggest that too little plasmin activity during the repair-APR may contribute to failed tissue repair, such as a fracture nonunion, and chronic inflammatory degenerative diseases like osteoporosis. Future clinical studies are required to affirm these findings. Therefore, the temporal-spatial functions of plasmin in response to musculoskeletal injury and its pharmacologic manipulation are intriguing new targets for improving orthopedic care.
Collapse
Affiliation(s)
- Stephanie N Moore-Lotridge
- Department of Orthopaedics, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Center for Bone Biology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Katherine S Hajdu
- School of Medicine, Vanderbilt University, Nashville, Tennessee, USA
| | - Brian Q Hou
- School of Medicine, Vanderbilt University, Nashville, Tennessee, USA
| | - Breanne H Y Gibson
- Department of Orthopaedics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Jonathan G Schoenecker
- Department of Orthopaedics, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Center for Bone Biology, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, USA.
| |
Collapse
|
131
|
Saengsiwaritt W, Ngamtipakon P, Udomsinprasert W. Vitamin D and autophagy in knee osteoarthritis: A review. Int Immunopharmacol 2023; 123:110712. [PMID: 37523972 DOI: 10.1016/j.intimp.2023.110712] [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: 05/05/2023] [Revised: 07/21/2023] [Accepted: 07/24/2023] [Indexed: 08/02/2023]
Abstract
Knee osteoarthritis (KOA), the highly prevalent degenerative disease affecting the joint, perpetually devastates the health of the elderly. Of various mechanisms known to participate in KOA etiology, apoptosis of chondrocytes is widely regarded as the primary cause of cartilage degradation. It has been suggested that the induction of autophagy in chondrocytes could potentially prolong the progression of KOA by modulating intracellular metabolic processes, which may be helpful for ameliorating chondrocyte apoptosis and eventual cartilage degeneration. Autophagy, a physiological process characterized by intracellular self-degradation, has been reportedly implicated in various pathologic conditions including KOA. Interestingly, vitamin D has been shown to regulate autophagy in human chondrocytes through multiple pathways, specifically AMPK/mTOR signaling pathway. This observation underscores the potential of vitamin D as a novel approach for restoring the functionality and survivability of chondrocytes in KOA. Supporting vitamin D's clinical significance, previous studies have demonstrated its substantial involvement in the symptoms and irregular joint morphology observed in KOA patients, strengthening potential therapeutic efficacy of vitamin D in treatment of KOA. Herein, the purpose of this review was to determine the mechanisms underlying the multi-processes of vitamin D implicated in autophagy in several cells including chondrocytes, which would bring unique insights into KOA pathogenesis.
Collapse
Affiliation(s)
| | - Phatchana Ngamtipakon
- Department of Biochemistry, Faculty of Pharmacy, Mahidol University, Bangkok 10400, Thailand
| | - Wanvisa Udomsinprasert
- Department of Biochemistry, Faculty of Pharmacy, Mahidol University, Bangkok 10400, Thailand.
| |
Collapse
|
132
|
Park H, Lee JS, Lee N, Kwon K, Kim JB, Kim SB, Kim HG, Kim DW. Red stem of spinach promotes antioxidant and anti-inflammatory chondroprotection in a rat model of osteoarthritis. J Funct Foods 2023; 109:105789. [DOI: 10.1016/j.jff.2023.105789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025] Open
|
133
|
Varela L, van de Lest CHA, Boere J, Libregts SFWM, Lozano-Andrés E, van Weeren PR, Wauben MHM. Acute joint inflammation induces a sharp increase in the number of synovial fluid EVs and modifies their phospholipid profile. Biochim Biophys Acta Mol Cell Biol Lipids 2023; 1868:159367. [PMID: 37473834 DOI: 10.1016/j.bbalip.2023.159367] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 07/12/2023] [Accepted: 07/17/2023] [Indexed: 07/22/2023]
Abstract
Inflammation is the hallmark of most joint disorders. However, the precise regulation of induction, perpetuation, and resolution of joint inflammation is not entirely understood. Since extracellular vesicles (EVs) are critical for intercellular communication, we aim to unveil their role in these processes. Here, we investigated the EVs' dynamics and phospholipidome profile from synovial fluid (SF) of healthy equine joints and from horses with lipopolysaccharide (LPS)-induced synovitis. LPS injection triggered a sharp increase of SF-EVs at 5-8 h post-injection, which started to decline at 24 h post-injection. Importantly, we identified significant changes in the lipid profile of SF-EVs after synovitis induction. Compared to healthy joint-derived SF-EVs (0 h), SF-EVs collected at 5, 24, and 48 h post-LPS injection were strongly increased in hexosylceramides. At the same time, phosphatidylserine, phosphatidylcholine, and sphingomyelin were decreased in SF-EVs at 5 h and 24 h post-LPS injection. Based on the lipid changes during acute inflammation, we composed specific lipid profiles associated with healthy and inflammatory state-derived SF-EVs. The sharp increase in SF-EVs during acute synovitis and the correlation of specific lipids with either healthy or inflamed states-derived SF-EVs are findings of potential interest for unveiling the role of SF-EVs in joint inflammation, as well as for the identification of EV-biomarkers of joint inflammation.
Collapse
Affiliation(s)
- Laura Varela
- Division Equine Sciences, Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands; Division Cell Biology, Metabolism & Cancer, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - Chris H A van de Lest
- Division Equine Sciences, Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands; Division Cell Biology, Metabolism & Cancer, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - Janneke Boere
- Division Equine Sciences, Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands; Department of Orthopaedics, University Medical Center Utrecht, Utrecht, Netherlands
| | - Sten F W M Libregts
- Division Cell Biology, Metabolism & Cancer, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - Estefanía Lozano-Andrés
- Division Cell Biology, Metabolism & Cancer, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands; Division of Infectious Diseases & Immunology, Department Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - P René van Weeren
- Division Equine Sciences, Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - Marca H M Wauben
- Division Cell Biology, Metabolism & Cancer, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands.
| |
Collapse
|
134
|
Zou Z, Li H, Yu K, Ma K, Wang Q, Tang J, Liu G, Lim K, Hooper G, Woodfield T, Cui X, Zhang W, Tian K. The potential role of synovial cells in the progression and treatment of osteoarthritis. EXPLORATION (BEIJING, CHINA) 2023; 3:20220132. [PMID: 37933282 PMCID: PMC10582617 DOI: 10.1002/exp.20220132] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 06/15/2023] [Indexed: 11/08/2023]
Abstract
Osteoarthritis (OA), the commonest arthritis, is characterized by the progressive destruction of cartilage, leading to disability. The Current early clinical treatment strategy for OA often centers on anti-inflammatory or analgesia medication, weight loss, improved muscular function and articular cartilage repair. Although these treatments can relieve symptoms, OA tends to be progressive, and most patients require arthroplasty at the terminal stages of OA. Recent studies have shown a close correlation between joint pain, inflammation, cartilage destruction and synovial cells. Consequently, understanding the potential mechanisms associated with the action of synovial cells in OA could be beneficial for the clinical management of OA. Therefore, this review comprehensively describes the biological functions of synovial cells, the synovium, together with the pathological changes of synovial cells in OA, and the interaction between the cartilage and synovium, which is lacking in the present literature. Additionally, therapeutic approaches based on synovial cells for OA treatment are further discussed from a clinical perspective, highlighting a new direction in the treatment of OA.
Collapse
Affiliation(s)
- Zaijun Zou
- Department of Sports MedicineThe First Affiliated Hospital of Dalian Medical UniversityDalianLiaoningChina
| | - Han Li
- Department of Sports MedicineThe First Affiliated Hospital of Dalian Medical UniversityDalianLiaoningChina
| | - Kai Yu
- Department of Bone and JointCentral Hospital of Zhuang He CityDalianLiaoningChina
| | - Ke Ma
- Department of Clinical MedicineChina Medical UniversityShenyangLiaoningChina
| | - Qiguang Wang
- National Engineering Research Center for BiomaterialsSichuan UniversityChengduSichuanChina
| | - Junnan Tang
- Department of CardiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Guozhen Liu
- School of MedicineThe Chinese University of Hong Kong (Shenzhen)ShenzhenGuangdongChina
| | - Khoon Lim
- Christchurch Regenerative Medicine and Tissue Engineering Group (CReaTE)Department of Orthopaedic Surgery and Musculoskeletal MedicineUniversity of OtagoChristchurchNew Zealand
| | - Gary Hooper
- Christchurch Regenerative Medicine and Tissue Engineering Group (CReaTE)Department of Orthopaedic Surgery and Musculoskeletal MedicineUniversity of OtagoChristchurchNew Zealand
| | - Tim Woodfield
- Christchurch Regenerative Medicine and Tissue Engineering Group (CReaTE)Department of Orthopaedic Surgery and Musculoskeletal MedicineUniversity of OtagoChristchurchNew Zealand
| | - Xiaolin Cui
- Department of Sports MedicineThe First Affiliated Hospital of Dalian Medical UniversityDalianLiaoningChina
- School of MedicineThe Chinese University of Hong Kong (Shenzhen)ShenzhenGuangdongChina
- Christchurch Regenerative Medicine and Tissue Engineering Group (CReaTE)Department of Orthopaedic Surgery and Musculoskeletal MedicineUniversity of OtagoChristchurchNew Zealand
| | - Weiguo Zhang
- Department of Sports MedicineThe First Affiliated Hospital of Dalian Medical UniversityDalianLiaoningChina
- Key Laboratory of Molecular Mechanisms for Repair and Remodeling of Orthopaedic DiseasesLiaoning ProvinceDalianLiaoningChina
| | - Kang Tian
- Department of Sports MedicineThe First Affiliated Hospital of Dalian Medical UniversityDalianLiaoningChina
- Key Laboratory of Molecular Mechanisms for Repair and Remodeling of Orthopaedic DiseasesLiaoning ProvinceDalianLiaoningChina
| |
Collapse
|
135
|
De Roover A, Escribano-Núñez A, Monteagudo S, Lories R. Fundamentals of osteoarthritis: Inflammatory mediators in osteoarthritis. Osteoarthritis Cartilage 2023; 31:1303-1311. [PMID: 37353140 DOI: 10.1016/j.joca.2023.06.005] [Citation(s) in RCA: 61] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 06/15/2023] [Accepted: 06/16/2023] [Indexed: 06/25/2023]
Abstract
OBJECTIVES As more has become known of the pathophysiology of osteoarthritis (OA), evidence that inflammation plays a critical role in its development and progression has accumulated. Here, we aim to review current knowledge of the complex inflammatory network in the OA joint. DESIGN This narrative review is presented in three main sections: local inflammation, systemic inflammation, and therapeutic implications. We focused on inflammatory mediators and their link to OA structural changes in the joint. RESULTS OA is characterized by chronic and low-grade inflammation mediated mostly by the innate immune system, which results in cartilage degradation, bone remodeling and synovial changes. Synovitis is regarded as an OA characteristic and associated with increased severity of symptoms and joint dysfunction. However, the articular cartilage and the subchondral bone also produce several pro-inflammatory mediators thus establishing a complex interplay between the different tissues of the joint. In addition, systemic low-grade inflammation induced by aging, obesity and metabolic syndrome can contribute to OA development and progression. The main inflammatory mediators associated with OA include cytokines, chemokines, growth factors, adipokines, and neuropeptides. CONCLUSIONS Future research is needed to deeper understand the molecular pathways mediating the inflammation in OA to provide new therapeutics that target these pathways, or to repurpose existing drugs.
Collapse
Affiliation(s)
- Astrid De Roover
- Laboratory of Tissue Homeostasis and Disease, Skeletal Biology and Engineering Research Center, Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium
| | - Ana Escribano-Núñez
- Laboratory of Tissue Homeostasis and Disease, Skeletal Biology and Engineering Research Center, Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium
| | - Silvia Monteagudo
- Laboratory of Tissue Homeostasis and Disease, Skeletal Biology and Engineering Research Center, Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium
| | - Rik Lories
- Laboratory of Tissue Homeostasis and Disease, Skeletal Biology and Engineering Research Center, Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium; Division of Rheumatology, University Hospitals Leuven, 3000 Leuven, Belgium.
| |
Collapse
|
136
|
Ku SK, Kim JK, Chun YS, Song CH. Anti-Osteoarthritic Effects of Antarctic Krill Oil in Primary Chondrocytes and a Surgical Rat Model of Knee Osteoarthritis. Mar Drugs 2023; 21:513. [PMID: 37888448 PMCID: PMC10608626 DOI: 10.3390/md21100513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 09/07/2023] [Accepted: 09/27/2023] [Indexed: 10/28/2023] Open
Abstract
Osteoarthritis (OA) is characterized by progressive cartilage destruction and synovitis; however, there are no approved disease-modifying OA drugs. Krill oil (KO) has been reported to possess anti-inflammatory properties and alleviate joint pain in knee OA, indicating its potential to target the inflammatory mechanism of OA. Therefore, the anti-OA effects of KO were investigated in primary chondrocytes and a surgical rat model of knee OA. The oral administration of KO at 200 and 100 mg/kg for 8 weeks improved joint swelling and mobility in the animal model and led to increased bone mineral density and compressive strength in the cartilage. The oral KO doses upregulated chondrogenic genes (type 2 collagen, aggrecan, and Sox9), with inhibition of inflammation markers (5-lipoxygenase and prostaglandin E2) and extracellular matrix (ECM)-degrading enzymes (MMP-2 and MMP-9) in the cartilage and synovium. Consistently, KO treatments increased the viability of chondrocytes exposed to interleukin 1α, accompanied by the upregulation of the chondrogenic genes and the inhibition of the ECM-degrading enzymes. Furthermore, KO demonstrated inhibitory effects on lipopolysaccharide-induced chondrocyte inflammation. Histopathological and immunohistochemical analyses revealed that KO improved joint destruction and synovial inflammation, probably due to the anti-inflammatory, anti-apoptotic, and chondrogenic effects. These findings suggest the therapeutic potential of KO for knee OA.
Collapse
Affiliation(s)
- Sae-Kwang Ku
- Department of Anatomy and Histology, College of Korean Medicine, Daegu Haany University, Gyeongsan 38610, Republic of Korea;
| | - Jong-Kyu Kim
- AriBnC Co., Ltd., Yongin 16914, Republic of Korea; (J.-K.K.); (Y.-S.C.)
| | - Yoon-Seok Chun
- AriBnC Co., Ltd., Yongin 16914, Republic of Korea; (J.-K.K.); (Y.-S.C.)
| | - Chang-Hyun Song
- Department of Anatomy and Histology, College of Korean Medicine, Daegu Haany University, Gyeongsan 38610, Republic of Korea;
| |
Collapse
|
137
|
Gherghel R, Macovei LA, Burlui MA, Cardoneanu A, Rezus II, Mihai IR, Rezus E. Osteoarthritis—The Role of Mesenchymal Stem Cells in Cartilage Regeneration. APPLIED SCIENCES 2023; 13:10617. [DOI: 10.3390/app131910617] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Abstract
Osteoarthritis (OA) is a condition that can cause substantial pain, loss of joint function, and a decline in quality of life in patients. Numerous risk factors, including aging, genetics, and injury, have a role in the onset of OA, characterized by structural changes within the joints. Most therapeutic approaches focus on the symptoms and try to change or improve the structure of the joint tissues. Even so, no treatments have been able to stop or slow the progression of OA or give effective and long-lasting relief of symptoms. In the absence of disease-modifying drugs, regenerative medicine is being investigated as a possible treatment that can change the course of OA by changing the structure of damaged articular cartilage. In regenerative therapy for OA, mesenchymal stem cells (MSCs) have been the mainstay of translational investigations and clinical applications. In recent years, MSCs have been discovered to be an appropriate cell source for treating OA due to their ability to expand rapidly in culture, their nontumorigenic nature, and their ease of collection. MSCs’ anti-inflammatory and immunomodulatory capabilities may provide a more favorable local environment for the regeneration of injured articular cartilage, which was thought to be one of the reasons why they were seen as more suited for OA. In addition to bone marrow, MSCs have also been isolated from adipose tissue, synovium, umbilical cord, cord blood, dental pulp, placenta, periosteum, and skeletal muscle. Adipose tissue and bone marrow are two of the most essential tissues for therapeutic MSCs. Positive preclinical and clinical trial results have shown that, despite current limitations and risks, MSC-based therapy is becoming a promising approach to regenerative medicine in treating OA.
Collapse
Affiliation(s)
- Robert Gherghel
- Department of Orthopedics and Trauma Surgery, Piatra Neamt Emergency Hospital, 700115 Piatra Neamt, Romania
| | - Luana Andreea Macovei
- Department of Rheumatology and Rehabilitation, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy Iasi, 700115 Iasi, Romania
| | - Maria-Alexandra Burlui
- Department of Rheumatology and Rehabilitation, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy Iasi, 700115 Iasi, Romania
| | - Anca Cardoneanu
- Department of Rheumatology and Rehabilitation, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy Iasi, 700115 Iasi, Romania
| | - Ioana-Irina Rezus
- Department of Dermatology, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy Iasi, 700115 Iasi, Romania
| | - Ioana Ruxandra Mihai
- Department of Rheumatology and Rehabilitation, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy Iasi, 700115 Iasi, Romania
| | - Elena Rezus
- Department of Rheumatology and Rehabilitation, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy Iasi, 700115 Iasi, Romania
| |
Collapse
|
138
|
Peng BY, Singh AK, Tsai CY, Chan CH, Deng YH, Wu CM, Chou YR, Tsao W, Wu CY, Deng WP. Platelet-derived biomaterial with hyaluronic acid alleviates temporal-mandibular joint osteoarthritis: clinical trial from dish to human. J Biomed Sci 2023; 30:77. [PMID: 37691117 PMCID: PMC10494357 DOI: 10.1186/s12929-023-00962-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Accepted: 08/07/2023] [Indexed: 09/12/2023] Open
Abstract
BACKGROUND Bioactive materials have now raised considerable attention for the treatment of osteoarthritis (OA), such as knee OA, rheumatoid OA, and temporomandibular joint (TMJ) OA. TMJ-OA is a common disease associated with an imbalance of cartilage regeneration, tissue inflammation, and disability in mouth movement. Recently, biological materials or molecules have been developed for TMJ-OA therapy; however, ideal treatment is still lacking. In this study, we used the combination of a human platelet rich plasma with hyaluronic acid (hPRP/HA) for TMJ-OA therapy to perform a clinical trial in dish to humans. METHOD Herein, hPRP was prepared, and the hPRP/HA combined concentration was optimized by MTT assay. For the clinical trial in dish, pro-inflammatory-induced in-vitro and in-vivo mimic 3D TMJ-OA models were created, and proliferation, gene expression, alcian blue staining, and IHC were used to evaluate chondrocyte regeneration. For the animal studies, complete Freund's adjuvant (CFA) was used to induce the TMJ-OA rat model, and condyle and disc regeneration were investigated through MRI. For the clinical trial in humans, 12 patients with TMJ-OA who had disc displacement and pain were enrolled. The disc displacement and pain at baseline and six months were measured by MRI, and clinical assessment, respectively. RESULTS Combined hPRP/HA treatment ameliorated the proinflammatory-induced TMJ-OA model and promoted chondrocyte proliferation by activating SOX9, collagen type I/II, and aggrecan. TMJ-OA pathology-related inflammatory factors were efficiently downregulated with hPRP/HA treatment. Moreover, condylar cartilage was regenerated by hPRP/HA treatment in a proinflammatory-induced 3D neocartilage TMJ-OA-like model. During the animal studies, hPRP/HA treatment strongly repaired the condyle and disc in a CFA-induced TMJ-OA rat model. Furthermore, we performed a clinical trial in humans, and the MRI data demonstrated that after 6 months of treatment, hPRP/HA regenerated the condylar cartilage, reduced disc displacement, alleviated pain, and increased the maximum mouth opening (MMO). Overall, clinical trials in dish to human results revealed that hPRP/HA promoted cartilage regeneration, inhibited inflammation, reduced pain, and increased joint function in TMJ-OA. CONCLUSION Conclusively, this study highlighted the therapeutic potential of the hPRP and HA combination for TMJ-OA therapy, with detailed evidence from bench to bedside. Trial registration Taipei Medical University Hospital (TMU-JIRB No. N201711041). Registered 24 November 2017. https://tmujcrc.tmu.edu.tw/inquiry_general.php .
Collapse
Affiliation(s)
- Bou-Yue Peng
- Department of Dentistry, Taipei Medical University Hospital, Taipei, 110301, Taiwan
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, 110301, Taiwan
| | - Abhinay Kumar Singh
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, 110301, Taiwan
- Stem Cell Research Center, College of Oral Medicine, Taipei Medical University, Taipei, 110301, Taiwan
| | - Ching-Yu Tsai
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, 110301, Taiwan
- Stem Cell Research Center, College of Oral Medicine, Taipei Medical University, Taipei, 110301, Taiwan
| | - Chun-Hao Chan
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, 110301, Taiwan
- Stem Cell Research Center, College of Oral Medicine, Taipei Medical University, Taipei, 110301, Taiwan
| | - Yue-Hua Deng
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, 110301, Taiwan
- Stem Cell Research Center, College of Oral Medicine, Taipei Medical University, Taipei, 110301, Taiwan
| | - Chi-Ming Wu
- Stem Cell Research Center, College of Oral Medicine, Taipei Medical University, Taipei, 110301, Taiwan
| | - Yen-Ru Chou
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, 110301, Taipei, Taiwan
| | - Wen Tsao
- Stem Cell Research Center, College of Oral Medicine, Taipei Medical University, Taipei, 110301, Taiwan
- School of Oral Hygiene, College of Oral Medicine, Taipei Medical University, Taipei, 110301, Taiwan
| | - Chia-Yu Wu
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, 110301, Taiwan.
- Division of Oral and Maxillofacial Surgery, Department of Dentistry, Taipei Medical University Hospital, Taipei, 110301, Taiwan.
| | - Win-Ping Deng
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, 110301, Taiwan.
- Stem Cell Research Center, College of Oral Medicine, Taipei Medical University, Taipei, 110301, Taiwan.
- Graduate Institute of Biomedical and Pharmaceutical Science, Fu Jen Catholic University, Taipei, 242062, Taiwan.
| |
Collapse
|
139
|
Darraj H, Hakami KM, Zogel B, Maghrabi R, Khired Z. Septic Arthritis of the Knee in Children. Cureus 2023; 15:e45659. [PMID: 37868524 PMCID: PMC10590147 DOI: 10.7759/cureus.45659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/21/2023] [Indexed: 10/24/2023] Open
Abstract
Septic arthritis of the knee is the most common form of septic arthritis in children and can lead to irreversible damage to the joint. Staphylococcus aureus is the primary pathogen associated with septic arthritis, although other causative pathogens may be isolate in children with specific risk factors. The diagnosis of knee septic arthritis is based on comprehensive evaluation, including the patient's medical history, physical examination, blood tests, and arthrocentesis. Empirical treatment typically involves anti-staphylococcal penicillin or a first-generation cephalosporin, although modifications may be made based on local resistance patterns and clinical culture data. Surgical debridement, either through open surgery or arthroscopy, involving extensive debridement of the joint, is effective in eliminating the infection. In most cases, additional surgical intervention is not necessary.
Collapse
Affiliation(s)
| | | | - Basem Zogel
- Medicine and Surgery, Jazan University, Jazan, SAU
| | | | | |
Collapse
|
140
|
Sampath SJP, Venkatesan V, Ghosh S, Kotikalapudi N. Obesity, Metabolic Syndrome, and Osteoarthritis-An Updated Review. Curr Obes Rep 2023; 12:308-331. [PMID: 37578613 DOI: 10.1007/s13679-023-00520-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/07/2023] [Indexed: 08/15/2023]
Abstract
PURPOSE OF REVIEW Metabolic syndrome (MetS), also called the 'deadly quartet' comprising obesity, diabetes, dyslipidemia, and hypertension, has been ascertained to have a causal role in the pathogenesis of osteoarthritis (OA). This review is aimed at discussing the current knowledge on the contribution of metabolic syndrome and its various components to OA pathogenesis and progression. RECENT FINDINGS Lately, an increased association identified between the various components of metabolic syndrome (obesity, diabetes, dyslipidemia, and hypertension) with OA has led to the identification of the 'metabolic phenotype' of OA. These metabolic perturbations alongside low-grade systemic inflammation have been identified to inflict detrimental effects upon multiple tissues of the joint including cartilage, bone, and synovium leading to complete joint failure in OA. Recent epidemiological and clinical findings affirm that adipokines significantly contribute to inflammation, tissue degradation, and OA pathogenesis mediated through multiple signaling pathways. OA is no longer perceived as just a 'wear and tear' disease and the involvement of the metabolic components in OA pathogenesis adds up to the complexity of the disease. Given the global surge in obesity and its allied metabolic perturbations, this review aims to throw light on the current knowledge on the pathophysiology of MetS-associated OA and the need to address MetS in the context of metabolic OA management. Better regulation of the constituent factors of MetS could be profitable in preventing MetS-associated OA. The identification of key roles for several metabolic regulators in OA pathogenesis has also opened up newer avenues in the recognition and development of novel therapeutic agents.
Collapse
Affiliation(s)
- Samuel Joshua Pragasam Sampath
- Department of Biotechnology, Faculty of Science & Humanities, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu District, Tamil Nadu, 603203, India.
- Molecular Biology Division, Indian Council of Medical Research - National Institute of Nutrition, Hyderabad, Telangana, 500007, India.
| | | | - Sudip Ghosh
- Molecular Biology Division, Indian Council of Medical Research - National Institute of Nutrition, Hyderabad, Telangana, 500007, India
| | - Nagasuryaprasad Kotikalapudi
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School Teaching Hospital, Boston, MA, 02115, USA
| |
Collapse
|
141
|
Tarabeih N, Kalinkovich A, Shalata A, Higla O, Livshits G. Pro-Inflammatory Biomarkers Combined with Body Composition Display a Strong Association with Knee Osteoarthritis in a Community-Based Study. Biomolecules 2023; 13:1315. [PMID: 37759715 PMCID: PMC10527309 DOI: 10.3390/biom13091315] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 08/22/2023] [Accepted: 08/25/2023] [Indexed: 09/29/2023] Open
Abstract
Knee osteoarthritis (KOA) is one of the most common progressive, age-dependent chronic degenerative joint diseases. KOA often develops as a result of a gradual articular cartilage loss caused by its wear and tear. Numerous studies suggest that the degradation of the knee joint involves inflammatory components. This process is also associated with body composition, particularly being overweight and muscle mass loss. The present study aimed to search for novel circulating KOA inflammatory biomarkers, taking into account body composition characteristics. To this aim, we recruited 98 patients diagnosed and radiologically confirmed with KOA and 519 healthy controls from the Arab community in Israel. A panel of soluble molecules, related to inflammatory, metabolic, and musculoskeletal disorders, was measured by ELISA in plasma samples, while several body composition parameters were assessed with bioimpedance analysis. Statistical analysis, including multivariable logistic regression, revealed a number of the factors significantly associated with KOA, independently of age and sex. The most significant independent associations [OR (95% CI)] were fat body mass/body weight index-1.56 (1.20-2.02), systemic immune-inflammation index-4.03 (2.23-7.27), circulating vaspin levels-1.39 (1.15-1.68), follistatin/FSTL1 ratio-1.32 (1.02-1.70), and activin A/FSTL1 ratio-1.33 (1.01-1.75). Further clinical studies are warranted to confirm the relevance of these KOA-associated biological factors. Hereafter, they could serve as reliable biomarkers for KOA in the general human population.
Collapse
Affiliation(s)
- Nader Tarabeih
- Department of Morphological Studies, Adelson School of Medicine, Ariel University, Ariel 40700, Israel;
| | - Alexander Kalinkovich
- Department of Anatomy and Anthropology, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 6905126, Israel;
| | - Adel Shalata
- The Simon Winter Institute for Human Genetics, Bnai Zion Medical Center, The Ruth and Bruce Rappaport Faculty of Medicine, Technion, Haifa 32000, Israel;
| | - Orabi Higla
- Orthopedics Clinic, Clalit, Migdal HaMeah, Tel-Aviv 6203854, Israel;
| | - Gregory Livshits
- Department of Anatomy and Anthropology, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 6905126, Israel;
| |
Collapse
|
142
|
Cachon T, Frykman O, Innes JF, Lascelles BDX, Okumura M, Sousa P, Staffieri F, Steagall PV, Van Ryssen B. COAST Development Group's international consensus guidelines for the treatment of canine osteoarthritis. Front Vet Sci 2023; 10:1137888. [PMID: 37601753 PMCID: PMC10436090 DOI: 10.3389/fvets.2023.1137888] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 04/10/2023] [Indexed: 08/22/2023] Open
Abstract
This report describes consensus guidelines and recommendations for the treatment of canine osteoarthritis (OA) according to the "Canine OsteoArthritis Staging Tool excluding radiography" (COASTeR) stage of OA, by the COAST Development Group. The recommendations are based on evidence-based medicine and clinical experience and are proposed with international relevance in mind. The aim is to provide veterinarians with a practical reference to consolidated information and to support the development of patient-specific OA management protocols and informed treatment choices based on the stage of OA.
Collapse
Affiliation(s)
- Thibaut Cachon
- Service de chirurgie, Campus Vétérinaire de Lyon VetAgro-Sup, Marcy l'Etoile, France
- Unité de recherche ICE, UPSP 2007-03-135, VetAgro-Sup Campus Vétérinaire, Université de Lyon, Marcy l'Etoile, France
| | | | - John F. Innes
- Movement Veterinary Referrals, Runcorn, United Kingdom
| | - B. Duncan X. Lascelles
- Translational Research in Pain (TRiP) Program, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
- Comparative Pain Research and Education Centre, Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
- Thurston Arthritis Centre, UNC School of Medicine, Chapel Hill, NC, United States
- Center for Translational Pain Research, Department of Anesthesiology, Duke University, Durham, NC, United States
| | - Masahiro Okumura
- Department of Veterinary Clinical Sciences, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Pedro Sousa
- Hospital Veterinari Montjuic - Vetpartners España, Barcelona, Spain
| | - Francesco Staffieri
- Section of Veterinary Clinicas and Animal Production, Department of Emergency and Organ Transplantations (D.E.O.T.), ‘Aldo Moro' University of Bari, Bari, Italy
| | - Paulo V. Steagall
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Université de Montréal, Saint Hyacinthe, QC, Canada
- Department of Veterinary Clinical Sciences, Centre for Animal Health and Welfare, Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Bernadette Van Ryssen
- Department of Medical Imaging and Small Animal Orthopedics, Faculty of Veterinary Medicine, Ghent University, Ghent, Belgium
| |
Collapse
|
143
|
Li W, Yu L, Li W, Ge G, Ma Y, Xiao L, Qiao Y, Huang W, Huang W, Wei M, Wang Z, Bai J, Geng D. Prevention and treatment of inflammatory arthritis with traditional Chinese medicine: Underlying mechanisms based on cell and molecular targets. Ageing Res Rev 2023; 89:101981. [PMID: 37302756 DOI: 10.1016/j.arr.2023.101981] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 05/25/2023] [Accepted: 06/08/2023] [Indexed: 06/13/2023]
Abstract
Inflammatory arthritis, primarily including rheumatoid arthritis, osteoarthritis and ankylosing spondylitis, is a group of chronic inflammatory diseases, whose general feature is joint dysfunction with chronic pain and eventually causes disability in older people. To date, both Western medicine and traditional Chinese medicine (TCM) have developed a variety of therapeutic methods for inflammatory arthritis and achieved excellent results. But there is still a long way to totally cure these diseases. TCM has been used to treat various joint diseases for thousands of years in Asia. In this review, we summarize clinical efficacies of TCM in inflammatory arthritis treatment after reviewing the results demonstrated in meta-analyses, systematic reviews, and clinical trials. We pioneered taking inflammatory arthritis-related cell targets of TCM as the entry point and further elaborated the molecular targets inside the cells of TCM, especially the signaling pathways. In addition, we also briefly discussed the relationship between gut microbiota and TCM and described the role of drug delivery systems for using TCM more accurately and safely. We provide updated and comprehensive insights into the clinical application of TCM for inflammatory arthritis treatment. We hope this review can guide and inspire researchers to further explore mechanisms of the anti-arthritis activity of TCM and make a great leap forward in comprehending the science of TCM.
Collapse
Affiliation(s)
- Wenhao Li
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, China; Orthopedic Institute, Medical College, Soochow University, Suzhou 215006, Jiangsu, China
| | - Lei Yu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, China; Orthopedic Institute, Medical College, Soochow University, Suzhou 215006, Jiangsu, China
| | - Wenming Li
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, China; Orthopedic Institute, Medical College, Soochow University, Suzhou 215006, Jiangsu, China
| | - Gaoran Ge
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, China; Orthopedic Institute, Medical College, Soochow University, Suzhou 215006, Jiangsu, China
| | - Yong Ma
- Department of Integrated Chinese and Western Medicine, School of Chinese Medicine & School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu, China
| | - Long Xiao
- Translational Medical Innovation Center, Department of Orthopedics, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang 215600, Jiangsu, China
| | - Yusen Qiao
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, China; Orthopedic Institute, Medical College, Soochow University, Suzhou 215006, Jiangsu, China
| | - Wei Huang
- Department of Orthopedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230031, Anhui, China
| | - Wenli Huang
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei 230031, Anhui, China
| | - Minggang Wei
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, China
| | - Zhirong Wang
- Translational Medical Innovation Center, Department of Orthopedics, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang 215600, Jiangsu, China.
| | - Jiaxiang Bai
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, China; Department of Orthopedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230031, Anhui, China.
| | - Dechun Geng
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, China; Orthopedic Institute, Medical College, Soochow University, Suzhou 215006, Jiangsu, China.
| |
Collapse
|
144
|
Takeda H, Nakajima Y, Yamaguchi T, Watanabe I, Miyoshi S, Nagashio K, Sekine H, Motohashi H, Yano H, Tanaka J. The anti-inflammatory and anti-oxidative effect of a classical hypnotic bromovalerylurea mediated by the activation of NRF2. J Biochem 2023; 174:131-142. [PMID: 37039781 PMCID: PMC10372716 DOI: 10.1093/jb/mvad030] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 03/14/2023] [Accepted: 04/05/2023] [Indexed: 04/12/2023] Open
Abstract
The Kelch-like ECH-associated protein 1-nuclear factor erythroid 2-related factor 2 (KEAP1-NRF2) system plays a central role in redox homeostasis and inflammation control. Oxidative stress or electrophilic compounds promote NRF2 stabilization and transcriptional activity by negatively regulating its inhibitor, KEAP1. We have previously reported that bromovalerylurea (BU), originally developed as a hypnotic, exerts anti-inflammatory effects in various inflammatory disease models. However, the molecular mechanism underlying its effect remains uncertain. Herein, we found that by real-time multicolor luciferase assay using stable luciferase red3 (SLR3) and green-emitting emerald luciferase (ELuc), BU potentiates NRF2-dependent transcription in the human hepatoblastoma cell line HepG2 cells, which lasted for more than 60 h. Further analysis revealed that BU promotes NRF2 accumulation and the transcription of its downstream cytoprotective genes in the HepG2 and the murine microglial cell line BV2. Keap1 knockdown did not further enhance NRF2 activity, suggesting that BU upregulates NRF2 by targeting KEAP1. Knockdown of Nfe2l2 in BV2 cells diminished the suppressive effects of BU on the production of pro-inflammatory mediators, like nitric oxide (NO) and its synthase NOS2, indicating the involvement of NRF2 in the anti-inflammatory effects of BU. These data collectively suggest that BU could be repurposed as a novel NRF2 activator to control inflammation and oxidative stress.
Collapse
Key Words
- Abbreviations: ARE, antioxidant responsive element; BU, bromovalerylurea; CCL2, C-C motif chemokine 2; DMF, dimethyl fumarate; GCLC, glutamate–cysteine ligase catalytic subunit; GCLM, glutamate–cysteine ligase modifier subunit; GSS, glutathione synthetase; GSH, glutathione; Hmox-1, heme oxygenase-1; IL-1β, interluekin-1β; IL-6, interluekin-6; JAK, Janus kinase; KEAP1, Kelch-like ECH-associated protein; NO, nitric oxide; NOS2, NO synthase 2; NRE, NF-κB responsive element; NQO-1, NAD(P)H quinone dehydrogenase; NRF2, nuclear factor erythroid 2-related factor 2/nuclear factor erythroid-derived 2-like 2; TXNRD, thioredoxin–disulfide reductase; ROS, reactive oxygen species
- KEAP1–NRF2
- anti-inflammation
- anti-oxidant oxygen
- bromovalerylurea
- drug action toxins/drugs/xenobiotics
Collapse
Affiliation(s)
- Haruna Takeda
- Department of Molecular and Cellular Physiology, Graduate School of Medicine, Ehime University, 454, Shitsukawa, Toon, Ehime, 791-0295, Japan
- Department of Gene Expression Regulation, Institute of Development, Aging and Cancer, Tohoku University, 4-1, Seiryo-cho, Aoba-ku, Sendai, Miyagi, 980-8575, Japan
| | - Yoshihiro Nakajima
- Health and Medical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), 2 217-14, Hayashi-cho, Takamatsu, Kagawa, 761-0301, Japan
| | - Teruaki Yamaguchi
- Department of Molecular and Cellular Physiology, Graduate School of Medicine, Ehime University, 454, Shitsukawa, Toon, Ehime, 791-0295, Japan
| | - Itaru Watanabe
- Department of Molecular and Cellular Physiology, Graduate School of Medicine, Ehime University, 454, Shitsukawa, Toon, Ehime, 791-0295, Japan
| | - Shoko Miyoshi
- Department of Molecular and Cellular Physiology, Graduate School of Medicine, Ehime University, 454, Shitsukawa, Toon, Ehime, 791-0295, Japan
| | - Kodai Nagashio
- Department of Molecular and Cellular Physiology, Graduate School of Medicine, Ehime University, 454, Shitsukawa, Toon, Ehime, 791-0295, Japan
| | - Hiroki Sekine
- Department of Gene Expression Regulation, Institute of Development, Aging and Cancer, Tohoku University, 4-1, Seiryo-cho, Aoba-ku, Sendai, Miyagi, 980-8575, Japan
| | - Hozumi Motohashi
- Department of Gene Expression Regulation, Institute of Development, Aging and Cancer, Tohoku University, 4-1, Seiryo-cho, Aoba-ku, Sendai, Miyagi, 980-8575, Japan
| | - Hajime Yano
- Department of Molecular and Cellular Physiology, Graduate School of Medicine, Ehime University, 454, Shitsukawa, Toon, Ehime, 791-0295, Japan
| | - Junya Tanaka
- Department of Molecular and Cellular Physiology, Graduate School of Medicine, Ehime University, 454, Shitsukawa, Toon, Ehime, 791-0295, Japan
| |
Collapse
|
145
|
Chang NP, DaPrano EM, Evans WR, Nissenbaum M, McCourt M, Alzate D, Lindman M, Chou TW, Atkins C, Kusnecov AW, Huda R, Daniels BP. Neuronal DAMPs exacerbate neurodegeneration via astrocytic RIPK3 signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.21.550097. [PMID: 37546744 PMCID: PMC10401942 DOI: 10.1101/2023.07.21.550097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
Astrocyte activation is a common feature of neurodegenerative diseases. However, the ways in which dying neurons influence the activity of astrocytes is poorly understood. RIPK3 signaling has recently been described as a key regulator of neuroinflammation, but whether this kinase mediates astrocytic responsiveness to neuronal death has not yet been studied. Here, we used the MPTP model of Parkinson's disease to show that activation of astrocytic RIPK3 drives dopaminergic cell death and axon damage. Transcriptomic profiling revealed that astrocytic RIPK3 promoted gene expression associated with neuroinflammation and movement disorders, and this coincided with significant engagement of DAMP signaling. Using human cell culture systems, we show that factors released from dying neurons signal through RAGE to induce RIPK3-dependent astrocyte activation. These findings highlight a mechanism of neuron-glia crosstalk in which neuronal death perpetuates further neurodegeneration by engaging inflammatory astrocyte activation via RIPK3.
Collapse
Affiliation(s)
- Nydia P. Chang
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
| | - Evan M. DaPrano
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
| | - Wesley R. Evans
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
- W. M. Keck Center for Collaborative Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
| | | | - Micheal McCourt
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
| | - Diego Alzate
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
| | - Marissa Lindman
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
| | - Tsui-Wen Chou
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
| | - Colm Atkins
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
| | | | - Rafiq Huda
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
- W. M. Keck Center for Collaborative Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
| | - Brian P. Daniels
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
| |
Collapse
|
146
|
Lana JF, Purita J, Everts PA, De Mendonça Neto PAT, de Moraes Ferreira Jorge D, Mosaner T, Huber SC, Azzini GOM, da Fonseca LF, Jeyaraman M, Dallo I, Santos GS. Platelet-Rich Plasma Power-Mix Gel (ppm)-An Orthobiologic Optimization Protocol Rich in Growth Factors and Fibrin. Gels 2023; 9:553. [PMID: 37504432 PMCID: PMC10379106 DOI: 10.3390/gels9070553] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 07/03/2023] [Accepted: 07/05/2023] [Indexed: 07/29/2023] Open
Abstract
Platelet- and fibrin-rich orthobiologic products, such as autologous platelet concentrates, have been extensively studied and appreciated for their beneficial effects on multiple conditions. Platelet-rich plasma (PRP) and its derivatives, including platelet-rich fibrin (PRF), have demonstrated encouraging outcomes in clinical and laboratory settings, particularly in the treatment of musculoskeletal disorders such as osteoarthritis (OA). Although PRP and PRF have distinct characteristics, they share similar properties. The relative abundance of platelets, peripheral blood cells, and molecular components in these orthobiologic products stimulates numerous biological pathways. These include inflammatory modulation, augmented neovascularization, and the delivery of pro-anabolic stimuli that regulate cell recruitment, proliferation, and differentiation. Furthermore, the fibrinolytic system, which is sometimes overlooked, plays a crucial role in musculoskeletal regenerative medicine by regulating proteolytic activity and promoting the recruitment of inflammatory cells and mesenchymal stem cells (MSCs) in areas of tissue regeneration, such as bone, cartilage, and muscle. PRP acts as a potent signaling agent; however, it diffuses easily, while the fibrin from PRF offers a durable scaffolding effect that promotes cell activity. The combination of fibrin with hyaluronic acid (HA), another well-studied orthobiologic product, has been shown to improve its scaffolding properties, leading to more robust fibrin polymerization. This supports cell survival, attachment, migration, and proliferation. Therefore, the administration of the "power mix" containing HA and autologous PRP + PRF may prove to be a safe and cost-effective approach in regenerative medicine.
Collapse
Affiliation(s)
- José Fábio Lana
- OrthoRegen Group, Max-Planck University, Indaiatuba 13343-060, Brazil
| | | | | | | | | | - Tomas Mosaner
- Brazilian Institute of Regenerative Medicine (BIRM), Indaiatuba 13334-170, Brazil
| | - Stephany Cares Huber
- Brazilian Institute of Regenerative Medicine (BIRM), Indaiatuba 13334-170, Brazil
| | | | | | - Madhan Jeyaraman
- Department of Orthopaedics, Faculty of Medicine, Sri Lalithambigai Medical College and Hospital, Tamil Nadu 600095, India
| | - Ignacio Dallo
- SportMe Medical Center, Department of Orthopaedic Surgery and Sports Medicine, Unit of Biological Therapies and MSK Interventionism, 41013 Seville, Spain
| | - Gabriel Silva Santos
- Brazilian Institute of Regenerative Medicine (BIRM), Indaiatuba 13334-170, Brazil
| |
Collapse
|
147
|
Xie Z, Wang L, Chen J, Zheng Z, Srinual S, Guo A, Sun R, Hu M. Reduction of systemic exposure and side effects by intra-articular injection of anti-inflammatory agents for osteoarthritis: what is the safer strategy? J Drug Target 2023; 31:596-611. [PMID: 37249274 DOI: 10.1080/1061186x.2023.2220083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 03/03/2023] [Accepted: 04/05/2023] [Indexed: 05/31/2023]
Abstract
Osteoarthritis (OA) is a chronic degenerative joint disease associated with pain, inflammation, and cartilage degradation. However, no current treatment can effectively halt the progression of the disease. Therefore, the use of NSAIDs and intra-articular corticosteroids is usually recommended as the primary treatment for OA-associated pain and inflammation. However, there is accumulating evidence that the long-term use of oral NSAIDs and intra-articular corticosteroids can lead to a myriad of negative side effects. Although numerous efforts have been made to develop intra-articular formulations for NSAIDs, the systemic exposure of intra-articular injection of NSAIDs and its potential side effects have not been explicitly investigated. To ascertain the evident and potential side effects of intra-articular injection of anti-inflammatory agents, we have summarised in this review the systemic exposure, local side effects, and systemic side effects of intra-articular injections of anti-inflammatory agents, including NSAIDs and corticosteroids. For developing a safer treatment to fulfil the unmet long-term use needs of patients, a new therapy, which combines the locally active drug and a sustained-release formulation, has been proposed in this review.
Collapse
Affiliation(s)
- Zuoxu Xie
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, USA
- Drug Metabolism and Pharmacokinetics, Biogen, Cambridge, MA, USA
| | - Lu Wang
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, USA
| | - Jie Chen
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, USA
| | - Zicong Zheng
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, USA
| | - Songpol Srinual
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, USA
| | - Annie Guo
- Drug Metabolism and Pharmacokinetics, Biogen, Cambridge, MA, USA
| | - Rongjin Sun
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, USA
| | - Ming Hu
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, USA
| |
Collapse
|
148
|
Geczy QE, Thirumaran AJ, Carroll PR, McLachlan AJ, Hunter DJ. What is the most effective and safest Non-steroidal anti-inflammatory drug for treating osteoarthritis in patients with comorbidities? Expert Opin Drug Metab Toxicol 2023; 19:681-695. [PMID: 37817419 DOI: 10.1080/17425255.2023.2267424] [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: 04/20/2023] [Accepted: 10/03/2023] [Indexed: 10/12/2023]
Abstract
INTRODUCTION Understanding what the most effective and safe non-steroidal anti-inflammatory drug (NSAID) is for managing osteoarthritis (OA) is complicated. OA is prevalent worldwide and people living with OA commonly have multiple comorbidities. The efficacy and safety of NSAIDs in a patient are influenced by their intrinsic and extrinsic factors. Current guidelines recommend the lowest dose for the shortest duration, monitoring patients for risk factors and comorbidities but generally do not specify, which NSAID is most suitable for a patient with specific comorbidities. AREAS COVERED This paper looks at the mechanism of action of all NSAIDs and reviews the current literature concerning their safety in patients with and without comorbidities. Relevant publications were identified by searching PubMed and Cochrane Library using key terms. The search was conducted from inception to 18 July 2023 and included results published before 18 July 2023. The search results and their references were then manually reviewed. EXPERT OPINION In the paper, we determine whether the current practice of 'lowest dose for shortest duration' is in fact the best approach for prescribing NSAIDs and identify which NSAIDs are most suitable given a patient's risk factors and comorbidities. Our aim is to help guide health professionals in recommending the most suitable NSAID for each patient.
Collapse
Affiliation(s)
- Quentin E Geczy
- Sydney Medical Program, The University of Sydney, Sydney, NSW, Australia
| | | | - Peter R Carroll
- School of Medicine Sydney, University of Notre Dame, Darlinghurst, NSW, Australia
- Sydney Pharmacy School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Andrew J McLachlan
- Sydney Pharmacy School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - David J Hunter
- Sydney Musculoskeletal Health, Arabanoo Precinct, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
- Rheumatology Department, Royal North Shore Hospital, St Leonards, NSW, Australia
| |
Collapse
|
149
|
Alexander A, Herz J, Calvier L. Reelin through the years: From brain development to inflammation. Cell Rep 2023; 42:112669. [PMID: 37339050 PMCID: PMC10592530 DOI: 10.1016/j.celrep.2023.112669] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 05/09/2023] [Accepted: 06/02/2023] [Indexed: 06/22/2023] Open
Abstract
Reelin was originally identified as a regulator of neuronal migration and synaptic function, but its non-neuronal functions have received far less attention. Reelin participates in organ development and physiological functions in various tissues, but it is also dysregulated in some diseases. In the cardiovascular system, Reelin is abundant in the blood, where it contributes to platelet adhesion and coagulation, as well as vascular adhesion and permeability of leukocytes. It is a pro-inflammatory and pro-thrombotic factor with important implications for autoinflammatory and autoimmune diseases such as multiple sclerosis, Alzheimer's disease, arthritis, atherosclerosis, or cancer. Mechanistically, Reelin is a large secreted glycoprotein that binds to several membrane receptors, including ApoER2, VLDLR, integrins, and ephrins. Reelin signaling depends on the cell type but mostly involves phosphorylation of NF-κB, PI3K, AKT, or JAK/STAT. This review focuses on non-neuronal functions and the therapeutic potential of Reelin, while highlighting secretion, signaling, and functional similarities between cell types.
Collapse
Affiliation(s)
- Anna Alexander
- Department of Molecular Genetics, University of Texas (UT) Southwestern Medical Center, Dallas, TX, USA; Center for Translational Neurodegeneration Research, UT Southwestern Medical Center, Dallas, TX, USA
| | - Joachim Herz
- Department of Molecular Genetics, University of Texas (UT) Southwestern Medical Center, Dallas, TX, USA; Center for Translational Neurodegeneration Research, UT Southwestern Medical Center, Dallas, TX, USA; Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX, USA; Department of Neurology and Neurotherapeutics, UT Southwestern Medical Center, Dallas, TX, USA
| | - Laurent Calvier
- Department of Molecular Genetics, University of Texas (UT) Southwestern Medical Center, Dallas, TX, USA; Center for Translational Neurodegeneration Research, UT Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
150
|
Holik H, Krečak I, Lucijanić M, Samardžić I, Pilipac D, Vučinić Ljubičić I, Coha B, Kitter Pipić A, Miškić B, Zupančić-Šalek S. Hip and Knee Osteoarthritis in Patients with Chronic Myeloproliferative Neoplasms: A Cross-Sectional Study. Life (Basel) 2023; 13:1388. [PMID: 37374170 PMCID: PMC10300951 DOI: 10.3390/life13061388] [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: 04/21/2023] [Revised: 06/01/2023] [Accepted: 06/12/2023] [Indexed: 06/29/2023] Open
Abstract
BACKGROUND Osteoarthritis (OA) is a progressive degenerative disease with an inflammatory background. Chronic myeloproliferative neoplasms (MPN) are clonal hematopoietic disorders characterized by chronic inflammation and a tendency for connective tissue remodeling. AIM This study aimed to investigate the prevalence and associated risk factors of symptomatic OA (sOA) in MPN patients. PATIENTS AND METHODS A total of 100 consecutive MPN (39 essential-thrombocythemia, 34 polycythemia-vera, 27 myelofibrosis) patients treated in two community hematologic centers were cross-sectionally evaluated. Patients were required to have both symptoms attributable to hip and/or knee OA and radiographic confirmation to be considered as having sOA. RESULTS The prevalence of hip and/or knee sOA was significantly higher among MPN patients than the previously reported prevalence in the general population of similar age (61% vs. 22%, p < 0.001). Hip sOA was present in 50%, knee sOA in 51% and sOA of both localizations in 41% of patients. A high proportion of MPN patients had radiographic signs of hip OA (94%) and knee OA (98%) in the presence of attributable symptoms. Among the other factors, sOA was univariately associated with the presence of JAK2 mutation, myelofibrosis phenotype, older age, higher body weight, and higher MPN-SAF score (p < 0.050 for all analyses). In the multivariate analysis, older age (odds ratio = 1.19, 95% confidence interval-CI 1.06-1.33) and higher body weight (OR = 1.15, 95% CI 1.06-1.25) were recognized as independent risk factors for sOA. On the other hand, cytoreductive treatment was a protective factor for sOA (OR = 0.07, 95% CI 0.006-0.86). CONCLUSIONS The prevalence of sOA in MPN patients was higher than that in the general population and seems to correlate with older age, increased myeloproliferation and a higher inflammatory state. Whether cytoreductive treatment may postpone OA development in MPN patients warrants additional confirmation.
Collapse
Affiliation(s)
- Hrvoje Holik
- Department of Internal Medicine, Dr. Josip Benčević General Hospital, 35000 Slavonski Brod, Croatia
- Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
| | - Ivan Krečak
- Department of Internal Medicine, General Hospital of Šibenik-Knin County, 22000 Šibenik, Croatia
- Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
| | - Marko Lucijanić
- University Hospital Dubrava, 10000 Zagreb, Croatia
- Faculty of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Ivan Samardžić
- Department of Orthopaedic Surgery, Dr. Josip Benčević General Hospital, 35000 Slavonski Brod, Croatia
| | - Danijel Pilipac
- Department of Orthopaedic Surgery, General Hospital of Šibenik-Knin County, 22000 Šibenik, Croatia
| | - Ivana Vučinić Ljubičić
- Department of Internal Medicine, Dr. Josip Benčević General Hospital, 35000 Slavonski Brod, Croatia
| | - Božena Coha
- Department of Internal Medicine, Dr. Josip Benčević General Hospital, 35000 Slavonski Brod, Croatia
| | - Alma Kitter Pipić
- Department of Laboratory Diagnostics, General Hospital ‘Dr Josip Benčević’, 35000 Slavonski Brod, Croatia
| | - Blaženka Miškić
- Department of Internal Medicine, Dr. Josip Benčević General Hospital, 35000 Slavonski Brod, Croatia
- Faculty of Dental Medicine and Health Osijek, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
| | - Silva Zupančić-Šalek
- Department of Hematology and Coagulation, University Hospital Holy Spirit, 10000 Zagreb, Croatia
| |
Collapse
|