151
|
Ngo L, Knothe Tate ML. A spike in circulating cytokines TNF-α and TGF-β alters barrier function between vascular and musculoskeletal tissues. Sci Rep 2023; 13:9119. [PMID: 37277369 DOI: 10.1038/s41598-023-30322-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 02/21/2023] [Indexed: 06/07/2023] Open
Abstract
Molecular transport between the circulatory and musculoskeletal systems regulates articular joint physiology in health and disease. Osteoarthritis (OA) is a degenerative joint disease linked to systemic and local inflammation. Inflammatory events involve cytokines, which are secreted by cells of the immune system and modulate molecular transport across tissue interfaces (referred to as tight junction [TJ] barrier function). In a previous study from our group, OA knee joint tissues were shown to exhibit size separation of different sized molecules delivered as a single bolus to the heart (Ngo et al. in Sci. Rep. 8:10254, 2018). Here, in a follow up study of parallel design, we test the hypothesis that two common cytokines, with multifaceted roles in the etiology of osteoarthritis as well as immune state in general, modulate the barrier function properties of joint tissue interfaces. Specifically, we probe the effect of an acute cytokine increase (spike) on molecular transport within tissues and across tissue interfaces of the circulatory and musculoskeletal systems. A single bolus of fluorescent-tagged 70 kDa dextran, was delivered intracardially, either alone, or with either the pro-inflammatory cytokine TNF-α or the anti-inflammatory cytokine TGF-β, to skeletally mature (11 to 13-month-old) guinea pigs (Dunkin-Hartley, a spontaneous OA animal model). After five minutes' circulation, whole knee joints were serial sectioned and fluorescent block face cryo-imaged at near-single-cell resolution. The 70 kDa fluorescent-tagged tracer is analogous in size to albumin, the most prevalent blood transporter protein, and quantification of tracer fluorescence intensity gave a measure of tracer concentration. Within five minutes, a spike (acute doubling) in circulating cytokines TNF-α or TGF-β significantly disrupted barrier function between the circulatory and musculoskeletal systems, with barrier function essentially abrogated in the TNF-α group. In the entire volume of the joint (including all tissue compartments and the bounding musculature), tracer concentration was significantly decreased in the TGF-β- and TNF-α- compared to the control-group. These studies implicate inflammatory cytokines as gatekeepers for molecular passage within and between tissue compartments of our joints and may open new means to delay the onset and mitigate the progression of degenerative joint diseases such as OA, using pharmaceutical and/or physical measures.
Collapse
Affiliation(s)
- Lucy Ngo
- MechBio Team, Graduate School of Biomedical Engineering, University of New South Wales, Sydney, Australia
| | - Melissa L Knothe Tate
- Blue Mountains World Interdisciplinary Innovation Institute, New South Wales, Australia.
| |
Collapse
|
152
|
Poudel SB, Ruff RR, Yildirim G, Dixit M, Michot B, Gibbs JL, Ortiz SD, Kopchick JJ, Kirsch T, Yakar S. Excess Growth Hormone Triggers Inflammation-Associated Arthropathy, Subchondral Bone Loss, and Arthralgia. THE AMERICAN JOURNAL OF PATHOLOGY 2023; 193:829-842. [PMID: 36870529 PMCID: PMC10284029 DOI: 10.1016/j.ajpath.2023.02.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 01/29/2023] [Accepted: 02/10/2023] [Indexed: 03/06/2023]
Abstract
Growth hormone (GH) is a key mediator of skeletal growth. In humans, excess GH secretion due to pituitary adenoma, seen in patients with acromegaly, results in severe arthropathies. This study investigated the effects of long-term excess GH on the knee joint tissues. One year-old wild-type (WT) and bovine GH (bGH) transgenic mice were used as a model for excess GH. bGH mice showed increased sensitivity to mechanical and thermal stimuli, compared with WT mice. Micro-computed tomography analyses of the distal femur subchondral bone revealed significant reductions in trabecular thickness and significantly reduced bone mineral density of the tibial subchondral bone-plate associated with increased osteoclast activity in both male and female bGH compared with WT mice. bGH mice showed severe loss of matrix from the articular cartilage, osteophytosis, synovitis, and ectopic chondrogenesis. Articular cartilage loss in the bGH mice was associated with elevated markers of inflammation and chondrocyte hypertrophy. Finally, hyperplasia of synovial cells was associated with increased expression of Ki-67 and diminished p53 levels in the synovium of bGH mice. Unlike the low-grade inflammation seen in primary osteoarthritis, arthropathy caused by excess GH affects all joint tissues and triggers severe inflammatory response. Data from this study suggest that treatment of acromegalic arthropathy should involve inhibition of ectopic chondrogenesis and chondrocyte hypertrophy.
Collapse
Affiliation(s)
- Sher B Poudel
- Department of Molecular Pathobiology, David B. Kriser Dental Center, New York University College of Dentistry, New York, New York
| | - Ryan R Ruff
- Department of Epidemiology and Health Promotion, David B. Kriser Dental Center, New York University College of Dentistry, New York, New York
| | - Gozde Yildirim
- Department of Molecular Pathobiology, David B. Kriser Dental Center, New York University College of Dentistry, New York, New York
| | - Manisha Dixit
- Department of Molecular Pathobiology, David B. Kriser Dental Center, New York University College of Dentistry, New York, New York
| | - Benoit Michot
- Department of Restorative Dentistry and Biomaterials Sciences, Harvard School of Dental Medicine, Boston, Massachusetts
| | - Jennifer L Gibbs
- Department of Restorative Dentistry and Biomaterials Sciences, Harvard School of Dental Medicine, Boston, Massachusetts
| | - Silvana D Ortiz
- Department of Biomedical Sciences, Edison Biotechnology Institute, Ohio University, Athens, Ohio
| | - John J Kopchick
- Department of Biomedical Sciences, Edison Biotechnology Institute, Ohio University, Athens, Ohio
| | - Thorsten Kirsch
- Department of Orthopaedic Surgery, New York University Grossman School of Medicine, New York, New York; Department of Biomedical Engineering, New York University Tandon School of Engineering, New York, New York
| | - Shoshana Yakar
- Department of Molecular Pathobiology, David B. Kriser Dental Center, New York University College of Dentistry, New York, New York.
| |
Collapse
|
153
|
Zhang Z, Wang S, Liu X, Yang Y, Zhang Y, Li B, Guo F, Liang J, Hong X, Guo R, Zhang B. Secoisolariciresinol diglucoside Ameliorates Osteoarthritis via Nuclear factor-erythroid 2-related factor-2/ nuclear factor kappa B Pathway: In vitro and in vivo experiments. Biomed Pharmacother 2023; 164:114964. [PMID: 37269815 DOI: 10.1016/j.biopha.2023.114964] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 05/23/2023] [Accepted: 05/28/2023] [Indexed: 06/05/2023] Open
Abstract
Osteoarthritis (OA) is an age-related joint disease in which inflammation and extracellular matrix (ECM) degradation play a crucial role in the destruction of articular cartilage. Secoisolariciresinol diglucoside (SDG), the main lignan in wholegrain flaxseed, which has been reported to remarkably suppress inflammation and oxidative stress, may have potential therapeutic value in OA. In this study, the effect and mechanism of SDG against cartilage degeneration were verified in the destabilization of the medial meniscus (DMM) and collagen-induced (CIA) arthritis models and interleukin-1β (IL-1β)-stimulated osteoarthritis chondrocyte models. From our experiments, SDG treatment downregulated the expression of pro-inflammatory factors induced by IL-1β in vitro, including inducible nitric oxide synthase (INOS), cyclooxygenase-2 (COX2), tumor necrosis factor (TNF-α), and interleukin 6 (IL-6). Additionally, SDG promoted the expression of collagen II (COL2A1) and SRY-related high-mobility-group-box gene 9(SOX9), while suppressing the expression of a disintegrin and metalloproteinase with thrombospondin motifs 5(ADAMTS5) and matrix metalloproteinases 13(MMP13), which leads to catabolism. Consistently, in vivo, SDG has been identified to have chondroprotective effects in DMM-induced and collagen-induced arthritis models. Mechanistically, SDG exerted its anti-inflammation and anti-ECM degradation effects by activating the Nrf2/HO-1 pathway and inhibiting the nuclear factor kappa B (NF-κB) pathway. In conclusion, SDG ameliorates the progression of OA via the Nrf2/NF-κB pathway, which indicates that SDG may have therapeutic potential for OA.
Collapse
Affiliation(s)
- Zhiwei Zhang
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Artificial Joints Engineering and Technology Research Center of Jiangxi Province, Nanchang 330006, China
| | - Song Wang
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Artificial Joints Engineering and Technology Research Center of Jiangxi Province, Nanchang 330006, China
| | - Xuqiang Liu
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Artificial Joints Engineering and Technology Research Center of Jiangxi Province, Nanchang 330006, China
| | - Yuxin Yang
- Huankui academy, Nanchang University, Nanchang 330006, China
| | - Yiqin Zhang
- Huankui academy, Nanchang University, Nanchang 330006, China
| | - Bo Li
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Artificial Joints Engineering and Technology Research Center of Jiangxi Province, Nanchang 330006, China
| | - Fengfen Guo
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Artificial Joints Engineering and Technology Research Center of Jiangxi Province, Nanchang 330006, China
| | - Jianhui Liang
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Artificial Joints Engineering and Technology Research Center of Jiangxi Province, Nanchang 330006, China
| | - Xin Hong
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Artificial Joints Engineering and Technology Research Center of Jiangxi Province, Nanchang 330006, China
| | - Runsheng Guo
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Artificial Joints Engineering and Technology Research Center of Jiangxi Province, Nanchang 330006, China.
| | - Bin Zhang
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Artificial Joints Engineering and Technology Research Center of Jiangxi Province, Nanchang 330006, China.
| |
Collapse
|
154
|
Rai MF, Cai L, Zhang Q, Townsend RR, Brophy RH. Synovial Fluid Proteomics From Serial Aspirations of ACL-Injured Knees Identifies Candidate Biomarkers. Am J Sports Med 2023:3635465231169526. [PMID: 37191559 DOI: 10.1177/03635465231169526] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
BACKGROUND Anterior cruciate ligament (ACL) tears often result in knee effusion and an increased risk for developing knee osteoarthritis (OA) in the long run. The molecular profile of these effusions could be informative regarding initial steps in the development of posttraumatic OA after an ACL tear. HYPOTHESIS The proteomics of knee synovial fluid changes over time after ACL injury. STUDY DESIGN Descriptive laboratory study. METHODS Synovial fluid was collected from patients with an acute traumatic ACL tear presenting to the office for evaluation (18.31 ± 19.07 days from injury) (aspiration 1) and again at the time of surgery (35.41 ± 58.15 days after aspiration 1 (aspiration 2). High-resolution liquid chromatography mass spectrometry was used to assess the quantitative protein profile of synovial fluid, and differences in protein profile between the 2 aspirations were determined computationally. RESULTS A total of 58 synovial fluid samples collected from 29 patients (12 male, 17 female; 12 isolated ACL tear, 17 combined ACL and meniscal tear) with a mean age and body mass index of 27.01 ± 12.78 years and 26.30 ± 4.93, respectively, underwent unbiased proteomics analysis. The levels of 130 proteins in the synovial fluid changed over time (87 high, 43 low). Proteins of interest that were significantly higher in aspiration 2 included CRIP1, S100A11, PLS3, POSTN, and VIM, which represent catabolic/inflammatory activities in the joint. Proteins with a known role in chondroprotection and joint homeostasis such as CHI3L2 (YKL-39), TNFAIP6/TSG6, DEFA1, SPP1, and CILP were lower in aspiration 2. CONCLUSION Synovial fluid from knees with ACL tears exhibits an increased burden of inflammatory (catabolic) proteins relevant to OA with reduced levels of chondroprotective (anabolic) proteins. CLINICAL RELEVANCE This study identified a set of novel proteins that provide new biological insights into the aftermath of ACL tears. Elevated inflammation and decreased chondroprotection could represent initial disruption of homeostasis, potentially initiating the development of OA. Longitudinal follow-up and mechanistic studies are necessary to assess the functional role of these proteins in the joint. Ultimately, these investigations could lead to better approaches to predict and possibly improve patient outcomes.
Collapse
Affiliation(s)
- Muhammad Farooq Rai
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Lei Cai
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Qiang Zhang
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - R Reid Townsend
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Robert H Brophy
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
155
|
Li S, Cao P, Chen T, Ding C. Latest insights in disease-modifying osteoarthritis drugs development. Ther Adv Musculoskelet Dis 2023; 15:1759720X231169839. [PMID: 37197024 PMCID: PMC10184265 DOI: 10.1177/1759720x231169839] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 03/29/2023] [Indexed: 05/19/2023] Open
Abstract
Osteoarthritis (OA) is a prevalent and severely debilitating disease with an unmet medical need. In order to alleviate OA symptoms or prevent structural progression of OA, new drugs, particularly disease-modifying osteoarthritis drugs (DMOADs), are required. Several drugs have been reported to attenuate cartilage loss or reduce subchondral bone lesions in OA and thus potentially be DMOADs. Most biologics (including interleukin-1 (IL-1) and tumor necrosis factor (TNF) inhibitors), sprifermin, and bisphosphonates failed to yield satisfactory results when treating OA. OA clinical heterogeneity is one of the primary reasons for the failure of these clinical trials, which can require different therapeutic approaches based on different phenotypes. This review describes the latest insights into the development of DMOADs. We summarize in this review the efficacy and safety profiles of various DMOADs targeting cartilage, synovitis, and subchondral bone endotypes in phase 2 and 3 clinical trials. To conclude, we summarize the reasons for clinical trial failures in OA and suggest possible solutions.
Collapse
Affiliation(s)
- Shengfa Li
- Clinical Research Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Peihua Cao
- Clinical Research Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Tianyu Chen
- Clinical Research Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Department of Orthopedics, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Changhai Ding
- Clinical Research Center, Zhujiang Hospital, Southern Medical University, 261 Industry Road, Guangzhou 510515, China
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
- Clinical Research Center, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| |
Collapse
|
156
|
Wan J, Zhu Z, He Z, Wu H, Chen A, Zhu W, Cheng P. Stevioside protects primary articular chondrocytes against IL-1β-induced inflammation and catabolism by targeting integrin. Int Immunopharmacol 2023; 119:110261. [PMID: 37167638 DOI: 10.1016/j.intimp.2023.110261] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 04/26/2023] [Accepted: 04/28/2023] [Indexed: 05/13/2023]
Abstract
Osteoarthritis (OA) is a common, progressive, and chronic disorder of the joints that is characterized by the inflammation and degradation of articular cartilage and is known to significantly impair quality of daily life. Stevioside (SVS) is a natural diterpenoid glycoside that has anti-inflammatory benefits. Hence, in the current research, it was hypothesized that SVS might exert anti-inflammatory effects on articular chondrocytes and alleviate cartilage degradation in mice with OA. The expression of inflammatory cytokines, like inducible nitric oxide synthase (iNOS), NOD-, LRR- and pyrin domain-containing protein 3 (NLRP3), and cyclooxygenase-2 (COX-2) in chondrocytes after interleukin-1β (IL-1β) exposure, was inhibited by the pretreatment of SVS. As well, SVS inhibited the reduction of collagen II and sry-box transcription factor 9 (SOX9) in chondrocytes stimulated by IL-1β and suppressed the expression of MMP3 and MMP13. Further, after treatment with SVS, cell cytometry, autophagy flux, and related protein expression showed diminished cell apoptosis and reduced autophagy impairment. Moreover, SVS blocked the activation of phosphoinositide-3-kinase/Akt/nuclear factor-kappa beta (PI3K/Akt/NF-κB) and mitogen-activated protein kinase (MAPK) signaling pathways stimulated by IL-1β. This resulted in decreased cellular inflammation. In vivo experiments with intra-articular injections of SVS in mice with the DMM mouse model demonstrated a decrease in cartilage degradation and an improvement in subchondral bone remodeling. After the integrin αVβ3-related knockdown using siRNA, a reversed effect was observed on the anti-inflammatory, anabolic promoting, catabolic blocking, and NF-κB and MAPK signaling pathway inhibition of SVS on chondrocytes treated with IL-1β. The above findings highlighted that SVS blocked IL-1β, triggered an inflammatory response in mice chondrocytes, and prevented cartilage degradation in vivo through integrin αVβ3. This suggested that SVS might serve as a novel therapeutic option for OA.
Collapse
Affiliation(s)
- Junlai Wan
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Ziqing Zhu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Zhiyi He
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Hua Wu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Anmin Chen
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Wentao Zhu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China.
| | - Peng Cheng
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China.
| |
Collapse
|
157
|
Tran NT, Truong MD, Yun HW, Min BH. Potential of secretome of human fetal cartilage progenitor cells as disease modifying agent for osteoarthritis. Life Sci 2023; 324:121741. [PMID: 37149084 DOI: 10.1016/j.lfs.2023.121741] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 04/24/2023] [Accepted: 04/24/2023] [Indexed: 05/08/2023]
Abstract
AIMS Osteoarthritis (OA) is caused by an imbalance in the synthesis and degradation of cartilage tissue by chondrocytes. Therefore, a therapeutic agent for OA patients that can positively affect both synthesis and degradation is needed. However, current nonsurgical treatments for OA can barely achieve satisfactory long-term outcomes in cartilage repair. Human fetal cartilage progenitor cells-secretome (ShFCPC) has shown potent anti-inflammatory and tissue-repair effects; however, its underlying mechanisms and effects on OA have rarely been systematically elucidated. This study aims to analyze and evaluate the potency of ShFCPC in modifying OA process. MAIN METHODS Herein, secreted proteins enriched in ShFCPC have been characterized, and their biological functions both in vitro and in vivo in an OA model are compared with those of human bone marrow-derived mesenchymal stem cells-secretome (ShBMSC) and hyaluronan (HA). KEY FINDINGS Secretome analysis has shown that ShFCPC is significantly enriched with extracellular matrix molecules involved in many effects of cellular processes required for homeostasis during OA progression. Biological validation in vitro has shown that ShFCPC protects chondrocyte apoptosis by suppressing the expression of inflammatory mediators and matrix-degrading proteases and promotes the secretion of pro-chondrogenic cytokines in lipopolysaccharide-induced coculture of human chondrocytes and SW982 synovial cells compared with ShBMSC. Moreover, in a rat OA model, ShFCPC protects articular cartilage by reducing inflammatory cell infiltration and M1/M2 macrophage ratio in the synovium, which directly contributes to an increase in immunomodulatory atmosphere and enhances cartilage repair compared to ShBMSC and HA. SIGNIFICANCE Our findings support clinical translations of ShFCPC as a novel agent for modifying OA process.
Collapse
Affiliation(s)
- Ngoc-Trinh Tran
- Department of Molecular Science and Technology, Ajou University, Suwon, Republic of Korea; Cell Therapy Center, Ajou Medical Center, Suwon, Republic of Korea
| | - Minh-Dung Truong
- Cell Therapy Center, Ajou Medical Center, Suwon, Republic of Korea
| | - Hee-Woong Yun
- Cell Therapy Center, Ajou Medical Center, Suwon, Republic of Korea
| | - Byoung-Hyun Min
- Department of Orthopedic Surgery, School of Medicine, Ajou University, Suwon, Republic of Korea; Institute of Regenerative Medicine, Wake Forest University, NC, USA; Advanced Translational Engineering & Medical Science, Seoul, Republic of Korea.
| |
Collapse
|
158
|
Balaskas P, Goljanek-Whysall K, Clegg PD, Fang Y, Cremers A, Smagul A, Welting TJM, Peffers MJ. MicroRNA Signatures in Cartilage Ageing and Osteoarthritis. Biomedicines 2023; 11:1189. [PMID: 37189806 PMCID: PMC10136140 DOI: 10.3390/biomedicines11041189] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 04/08/2023] [Accepted: 04/12/2023] [Indexed: 05/17/2023] Open
Abstract
Osteoarthritis is the most common degenerative joint disorder. MicroRNAs are gene expression regulators that act post-transcriptionally to control tissue homeostasis. Microarray analysis was undertaken in osteoarthritic intact, lesioned and young intact cartilage. Principal component analysis showed that young intact cartilage samples were clustered together; osteoarthritic samples had a wider distribution; and osteoarthritic intact samples were separated into two subgroups, osteoarthritic-Intact-1 and osteoarthritic-Intact-2. We identified 318 differentially expressed microRNAs between young intact and osteoarthritic lesioned cartilage, 477 between young intact and osteoarthritic-Intact-1 cartilage and 332 between young intact and osteoarthritic-Intact-2 cartilage samples. For a selected list of differentially expressed microRNAs, results were verified in additional cartilage samples using qPCR. Of the validated DE microRNAs, four-miR-107, miR-143-3p, miR-361-5p and miR-379-5p-were selected for further experiments in human primary chondrocytes treated with IL-1β. Expression of these microRNAs decreased in human primary chondrocytes treated with IL-1β. For miR-107 and miR-143-3p, gain- and loss-of-function approaches were undertaken and associated target genes and molecular pathways were investigated using qPCR and mass spectrometry proteomics. Analyses showed that WNT4 and IHH, predicted targets of miR-107, had increased expression in osteoarthritic cartilage compared to young intact cartilage and in primary chondrocytes treated with miR-107 inhibitor, and decreased expression in primary chondrocytes treated with miR-107 mimic, suggesting a role of miR-107 in chondrocyte survival and proliferation. In addition, we identified an association between miR-143-3p and EIF2 signalling and cell survival. Our work supports the role of miR-107 and miR-143-3p in important chondrocyte mechanisms regulating proliferation, hypertrophy and protein translation.
Collapse
Affiliation(s)
- Panagiotis Balaskas
- Institute of Life Course and Medical Sciences, William Henry Duncan Building, 6 West Derby Street, Liverpool L7 8TX, UK
| | - Katarzyna Goljanek-Whysall
- Institute of Life Course and Medical Sciences, William Henry Duncan Building, 6 West Derby Street, Liverpool L7 8TX, UK
- Department of Physiology, College of Medicine, Nursing and Health Sciences, University of Galway, H91 TK33 Galway, Ireland
| | - Peter D. Clegg
- Institute of Life Course and Medical Sciences, William Henry Duncan Building, 6 West Derby Street, Liverpool L7 8TX, UK
| | - Yongxiang Fang
- Centre for Genomic Research, Institute of Integrative Biology, Biosciences Building, University of Liverpool, Crown Street, Liverpool L69 7ZB, UK
| | - Andy Cremers
- Department of Orthopaedic Surgery, Medical Centre, Maastricht University, 6202 AZ Maastricht, The Netherlands
| | - Aibek Smagul
- Institute of Life Course and Medical Sciences, William Henry Duncan Building, 6 West Derby Street, Liverpool L7 8TX, UK
| | - Tim J. M. Welting
- Department of Orthopaedic Surgery, Medical Centre, Maastricht University, 6202 AZ Maastricht, The Netherlands
| | - Mandy J. Peffers
- Institute of Life Course and Medical Sciences, William Henry Duncan Building, 6 West Derby Street, Liverpool L7 8TX, UK
| |
Collapse
|
159
|
Choi BE, Shin S, Evans S, Singh BB, Bandyopadhyay BC. Ablation of TRPC3 disrupts Ca 2+ signaling in salivary ductal cells and promotes sialolithiasis. Sci Rep 2023; 13:5772. [PMID: 37031239 PMCID: PMC10082769 DOI: 10.1038/s41598-023-32602-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 03/30/2023] [Indexed: 04/10/2023] Open
Abstract
Clinical studies and structural analyses of salivary stones strongly suggest a linkage between higher saliva calcium (Ca2+) and salivary stone formation, sialolithiasis; however, the process and the mechanism leading to Ca2+ overload during sialolithiasis is not well understood. Here, we show that TRPC3 null (-/-) mice presented with a reduction in Ca2+ entry and current in ductal cells with higher saliva [Ca2+] suggesting diminished transepithelial Ca2+ flux across the salivary ductal cells, leaving more Ca2+ in ductal fluid. Significantly, we found that TRPC3 was expressed in mice and human salivary ductal cells, while intraductal stones were detected in both mice (TRPC3-/-) and patient (sialolithiasis) salivary glands. To identify the mechanism, we found that TRPC3 was crucial in preventing the expression of calcification genes (BMP2/6, Runx2) in ductal cells which may be due to higher extracellular Ca2+ in SMG tissues. Similarly, inflammatory (IL6, NLRP3), fibrotic (FN1, TGFβ1) and apoptotic (Bax1/Bcl2) markers were also elevated, suggesting that the loss of TRPC3 induces genetic changes that leads to salivary gland cell death and induction of inflammatory response. Overall, ablation of TRPC3-/- leads to higher saliva [Ca2+], along with elevated detrimental gene expressions, altogether contributing to salivary gland stone formation.
Collapse
Affiliation(s)
- Bok-Eum Choi
- Calcium Signaling Laboratory, 151 Research Service, Veterans Affairs Medical Center, 50 Irving Street, NW, Washington, DC, 20422, USA
| | - Samuel Shin
- Calcium Signaling Laboratory, 151 Research Service, Veterans Affairs Medical Center, 50 Irving Street, NW, Washington, DC, 20422, USA
- Department of Biomedical Engineering, The Catholic University of America, 620 Michigan Avenue NE, Washington, DC, 20064, USA
| | - Sade Evans
- Calcium Signaling Laboratory, 151 Research Service, Veterans Affairs Medical Center, 50 Irving Street, NW, Washington, DC, 20422, USA
| | - Brij B Singh
- Department of Periodontics, The University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Dr, San Antonio, TX, 78229, USA
| | - Bidhan C Bandyopadhyay
- Calcium Signaling Laboratory, 151 Research Service, Veterans Affairs Medical Center, 50 Irving Street, NW, Washington, DC, 20422, USA.
- Department of Biomedical Engineering, The Catholic University of America, 620 Michigan Avenue NE, Washington, DC, 20064, USA.
| |
Collapse
|
160
|
Lu R, Wang YG, Qu Y, Wang SX, Peng C, You H, Zhu W, Chen A. Dihydrocaffeic acid improves IL-1β-induced inflammation and cartilage degradation via inhibiting NF-κB and MAPK signalling pathways. Bone Joint Res 2023; 12:259-273. [PMID: 37492935 PMCID: PMC10076109 DOI: 10.1302/2046-3758.124.bjr-2022-0384.r1] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/08/2023] Open
Abstract
Aims Osteoarthritis (OA) is a prevalent joint disorder with inflammatory response and cartilage deterioration as its main features. Dihydrocaffeic acid (DHCA), a bioactive component extracted from natural plant (gynura bicolor), has demonstrated anti-inflammatory properties in various diseases. We aimed to explore the chondroprotective effect of DHCA on OA and its potential mechanism. Methods In vitro, interleukin-1 beta (IL-1β) was used to establish the mice OA chondrocytes. Cell counting kit-8 evaluated chondrocyte viability. Western blotting analyzed the expression levels of collagen II, aggrecan, SOX9, inducible nitric oxide synthase (iNOS), IL-6, matrix metalloproteinases (MMPs: MMP1, MMP3, and MMP13), and signalling molecules associated with nuclear factor-kappa B (NF-κB) and mitogen-activated protein kinase (MAPK) pathways. Immunofluorescence analysis assessed the expression of aggrecan, collagen II, MMP13, and p-P65. In vivo, a destabilized medial meniscus (DMM) surgery was used to induce mice OA knee joints. After injection of DHCA or a vehicle into the injured joints, histological staining gauged the severity of cartilage damage. Results DHCA prevented iNOS and IL-6 from being upregulated by IL-1β. Moreover, the IL-1β-induced upregulation of MMPs could be inhibited by DHCA. Additionally, the administration of DHCA counteracted IL-1β-induced downregulation of aggrecan, collagen II, and SOX9. DHCA protected articular cartilage by blocking the NF-κB and MAPK pathways. Furthermore, DHCA mitigated the destruction of articular cartilage in vivo. Conclusion We present evidence that DHCA alleviates inflammation and cartilage degradation in OA chondrocytes via suppressing the NF-κB and MAPK pathways, indicating that DHCA may be a potential agent for OA treatment.
Collapse
Affiliation(s)
- Rui Lu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ying-Guang Wang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yunkun Qu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shan-Xi Wang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Cheng Peng
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongbo You
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wentao Zhu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Anmin Chen
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
161
|
Hsieh CY, Wang CC, Tayo LL, Deng SX, Tsai PW, Lee CJ. In vitro and in vivo anti-osteoarthritis effects of tradition Chinese prescription Ji-Ming-San. JOURNAL OF ETHNOPHARMACOLOGY 2023; 305:116084. [PMID: 36584922 DOI: 10.1016/j.jep.2022.116084] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 12/20/2022] [Accepted: 12/20/2022] [Indexed: 06/17/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Ji-Ming-Shan (JMS) is a traditional herbal prescription consisting of seven herbs including Areca cathechu Burm.f., Citrus reticulata Blanco, Chaenomeles speciosa (Sweet) Nakai, Euodia ruticarpa (A. Juss.) Benth., Perilla frutescens (L.) Britton, Zingiber officinale Roscoe, Platycodon grandiflorus (Jacq.). It was first recorded during the Song dynasty and has been used extensively for protection against rheumatism, treatment of swelling of tendons, relief from foot pain, gout and diuresis and other forms of inflammation. AIM OF THE STUDY The aim of this study is to evaluate the anti-inflammatory and anti-osteoarthritis activity of JMS extracts with the use of different cell lines (RAW 264.7 cells, SW1353 cells and primary cultured rat chondrocytes). MIA-induced rat animal models were used to assess the anti-osteoarthritis activity of the extract. MATERIALS AND METHODS This study investigated the anti-inflammatory activity of JMS-95E on LPS-induced RAW 264.7 macrophages and IL-1β-stimulated chondrocytes. For the in vivo study, male Wistar rats were used and they were randomly assigned in different groups: blank, control, positive control and three different JMS-95E treatment groups (200, 400, 800 mg/kg/d). Paw edema, hind-limb weight bearing, serum inflammatory cytokines including hematoxylin and eosin (HE) staining experiments were used to assess the efficacy of the extract in the rat model. RESULT JMS 95% ethanol extract (JMS-95E, marker substance: narirutin (5.10 mg/g) and hesperidin (11.33 mg/g) has been identified in the extract using high pressure liquid chromatography. For in vitro assays, JMS-95E did not exhibit cytotoxicity and was able to downregulate the protein expression of iNOS, COX-2 and MMP-13. The production of inflammatory mediators such as NO and PGE2 were also reduced with an increase in dose-dependent manner in various cell lines. Inhibitory activity on the key enzyme xanthine oxidase was also observed in this study. In rat animal models, JMS-95E reduced the inflammatory responses such as acute swelling, chondrocyte degradation and pain section of paw edema in rat model. Molecular marker studies of inflammation demonstrated that JMS-95E significantly decrease PGE2 expression in MIA model. CONCLUSION JMS-95E inhibited the inflammatory pathway leading to the production or expression levels of NO, iNOS, COX-2 and PGE2 in macrophage cells. In primary cultured rat chondrocytes iNOS and SW1353 MMP-13 expression were downregulated after JMS-95E treatment. For the in vivo study JMS-95E significantly reduced the paw volume of carrageenan-induced rat paw edema through each dose and significantly inhibited paw volume, counterweight the distribution of hind-paw weight bearing through the MIA model which means JMS-95E could promote recovery of the acute swelling and chondrocyte degradation of the ankle joints. The above results provided the multiple mechanism of JMS-95E in OA treatment of the scientific founding which supported the description of JMS in traditional use.
Collapse
Affiliation(s)
- Cheng-Yang Hsieh
- Ph.D. Program in Clinical Drug Development of Herbal Medicine, College of Pharmacy, Taipei Medical University, Taipei, 110, Taiwan.
| | - Ching-Chiung Wang
- Ph.D. Program in Clinical Drug Development of Herbal Medicine, College of Pharmacy, Taipei Medical University, Taipei, 110, Taiwan; Graduate Institute of Pharmacognosy, College of Pharmacy, Taipei Medical University, Taipei, 110, Taiwan; Traditional Herbal Medicine Research Center, Taipei Medical University Hospital, Taipei, 110, Taiwan; School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, 110, Taiwan.
| | - Lemmuel L Tayo
- School of Chemical, Biological, Materials Engineering and Sciences, Mapúa University, Intramuros, Manila, 1002, Metro Manila, Philippines.
| | - Shun-Xin Deng
- Ph.D. Program in Clinical Drug Development of Herbal Medicine, 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.
| | - Chia-Jung Lee
- Ph.D. Program in Clinical Drug Development of Herbal Medicine, College of Pharmacy, Taipei Medical University, Taipei, 110, Taiwan; Graduate Institute of Pharmacognosy, College of Pharmacy, Taipei Medical University, Taipei, 110, Taiwan; Traditional Herbal Medicine Research Center, Taipei Medical University Hospital, Taipei, 110, Taiwan.
| |
Collapse
|
162
|
Zhou K, Yang C, Shi K, Liu Y, Hu D, He X, Yang Y, Chu B, Peng J, Zhou Z, Qian Z. Activated macrophage membrane-coated nanoparticles relieve osteoarthritis-induced synovitis and joint damage. Biomaterials 2023; 295:122036. [PMID: 36804660 DOI: 10.1016/j.biomaterials.2023.122036] [Citation(s) in RCA: 49] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 01/27/2023] [Accepted: 02/03/2023] [Indexed: 02/13/2023]
Abstract
Osteoarthritis (OA) is a common joint condition that is a leading cause of disability worldwide. There are currently no disease-modifying treatments for osteoarthritis, which is associated with multiple kinds of inflammatory cytokines produced by M1 macrophages in the synovium of the joint. Despite recent therapeutic advancements with anti-cytokine biologics, the OA therapy response rate continues to be inadequate. To treat OA, the pro-inflammatory and anti-inflammatory responses of synoviocytes and macrophages must be controlled simultaneously. Therefore, the immune regulation capabilities of an ideal nano-drug should not only minimize pro-inflammatory responses but also effectively boost anti-inflammatory responses. In this paper, an M2H@RPK nanotherapeutic system was developed, KAFAK and shRNA-LEPR were condensed with polyethylenimine (PEI) to form a complex, which was then modified with hyaluronic acid (HA) to negatively charge to cover the M2 membrane. It was discovered that the repolarization of macrophages from the M1 to the M2 phenotype lowered pro-inflammatory responses while enhancing anti-inflammatory responses in macrophages and synoviocytes. In vitro and in vivo studies demonstrate that M2H@RPK dramatically decreases proinflammatory cytokines, controls synovial inflammation, and provides significant therapeutic efficacy by reducing joint damage. Overall, it has been demonstrated that M2H@RPK provides inflammation-targeted therapy by macrophage repolarization, and it represents a promising OA therapeutic strategy.
Collapse
Affiliation(s)
- Kai Zhou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, China; Department of Orthopedics, Orthopedic Research Institute, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Chengli Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, China; Department of Pharmacy, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Kun Shi
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, China
| | - Yue Liu
- Department of Anesthesiology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Danrong Hu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, China
| | - Xinlong He
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, China
| | - Yun Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, China
| | - Bingyang Chu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, China
| | - Jinrong Peng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, China
| | - Zongke Zhou
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Zhiyong Qian
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, China.
| |
Collapse
|
163
|
Shen Y, Teng L, Qu Y, Huang Y, Peng Y, Tang M, Fu Q. Hederagenin Suppresses Inflammation and Cartilage Degradation to Ameliorate the Progression of Osteoarthritis: An In vivo and In vitro Study. Inflammation 2023; 46:655-678. [PMID: 36348189 DOI: 10.1007/s10753-022-01763-5] [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: 07/09/2022] [Revised: 10/25/2022] [Accepted: 10/26/2022] [Indexed: 11/09/2022]
Abstract
Osteoarthritis (OA), a common degenerative joint disease, is characterized by the progressive degradation of articular cartilage and inflammation. Hederagenin (HE) is a pentacyclic triterpenoid saponin extracted from many herb plants. It has anti-inflammatory, anti-lipid peroxidative, anti-cancer, and neuroprotective activities. However, its effect on OA has not been investigated. Our study found that HE may be a potential anti-OA drug. In vitro, HE could suppress extracellular matrix (ECM) degradation via up-regulating aggrecan and Collagen II levels as well as downregulating MMPs and ADAMTS5 levels. It could also reduce proinflammatory and inflammatory cytokines or enzymes production, including TNF-α, IL-6, iNOS, COX-2, NO, and PGE2. Besides, HE markedly reduced IL-1β-induced C28/I2 cell apoptosis and ROS accumulation. Mechanistically, HE exerted chondroprotective and anti-inflammatory effects by partly inhibiting JAK2/STAT3/MAPK signalling pathway and the crosstalk of the two pathways. Also, HE exhibited anti-apoptotic and anti-oxidative effect via targeting Keap1-Nrf2/HO-1/ROS/Bax/Bcl-2 axis. In vivo, HE significantly reduced monosodium iodoacetate (MIA) induced cartilage destruction of rats with a lower OARSI score and inflammatory cytokine levels, further demonstrating its protective effects in OA progression. These results suggest that HE is a potential compound for the development of drugs to treat OA.
Collapse
Affiliation(s)
- Yue Shen
- Key Laboratory of Drug-Targeting and Drug Delivery System of Sichuan Province, Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu, 610106, China
| | - Li Teng
- School of Food and Biological Engineering, Chengdu University, Chengdu, 610106, China
| | - Yuhan Qu
- School of Food and Biological Engineering, Chengdu University, Chengdu, 610106, China
| | - Yuehui Huang
- Key Laboratory of Drug-Targeting and Drug Delivery System of Sichuan Province, Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu, 610106, China
| | - Yi Peng
- Key Laboratory of Drug-Targeting and Drug Delivery System of Sichuan Province, Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu, 610106, China
| | - Min Tang
- School of Food and Biological Engineering, Chengdu University, Chengdu, 610106, China
| | - Qiang Fu
- Key Laboratory of Drug-Targeting and Drug Delivery System of Sichuan Province, Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu, 610106, China.
| |
Collapse
|
164
|
Schubert EA, Johnstone MT, Benson MJ, Alffenaar JC, Wheate NJ. Medicinal cannabis for Australian patients with chronic refractory pain including arthritis. Br J Pain 2023; 17:206-217. [PMID: 37057257 PMCID: PMC10088421 DOI: 10.1177/20494637221147115] [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] [Indexed: 04/15/2023] Open
Abstract
Objectives To examine the tolerability and effectiveness of medicinal cannabis prescribed to patients for chronic, refractory pain, with a subset analysis on arthritis. Methods This was an interim analysis of the CA Clinics Observational Study investigating self-reported adverse events (AEs) and changes in health-related quality of life (HRQoL) outcomes over time after commencing medicinal cannabis. Patients were prescribed medicinal cannabis by a medical practitioner, containing various ratios of Δ9-tetrahydrocannabinol (THC) and/or cannabidiol (CBD). Results The overall chronic pain cohort, and specifically the balanced CBD:THC products, were associated with significantly reduced pain intensity scores (p = 0.003, p = 0.025), with 22% of patients reporting a clinically meaningful reduction in pain intensity. Patients in the arthritis subset (n = 199) reported significantly reduced pain intensity scores (p = 0.005) overall, and specifically for those taking CBD-only (p = 0.018) and balanced products (p = 0.005). Other HRQoL outcomes, including pain interference and pain impact scores were significantly improved depending on the CBD:THC ratio. Products that contained a balanced ratio of CBD:THC were associated with improvements in the most number of PROMIS-29 domains. Approximately half (n = 364; 51%) of the chronic pain cohort experienced at least one AE, the most common being dry mouth (24%), somnolence (19%) or fatigue (12%). These findings were similar in the arthritis subset. Discussion Medicinal cannabis was observed to improve pain intensity scores and HRQoL outcomes in patients with chronic, refractory pain, providing real-world insights into medicinal cannabis' therapeutic potential.
Collapse
Affiliation(s)
- Elise A Schubert
- School of Pharmacy, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | | | | | - Johannes C Alffenaar
- School of Pharmacy, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
- Westmead Hospital, Westmead, NSW, Australia
| | - Nial J Wheate
- School of Pharmacy, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
165
|
Warmink K, Vinod P, Korthagen NM, Weinans H, Rios JL. Macrophage-Driven Inflammation in Metabolic Osteoarthritis: Implications for Biomarker and Therapy Development. Int J Mol Sci 2023; 24:ijms24076112. [PMID: 37047082 PMCID: PMC10094694 DOI: 10.3390/ijms24076112] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 03/19/2023] [Accepted: 03/21/2023] [Indexed: 04/14/2023] Open
Abstract
Osteoarthritis (OA) is a common and debilitating joint disorder that leads to progressive joint breakdown and loss of articular cartilage. Accompanied by a state of low-grade inflammation, its etiology extends beyond that of a wear-and-tear disease, and the immune system might have a role in its initiation and progression. Obesity, which is directly associated with an increased incidence of OA, alters adipokine release, increases pro-inflammatory macrophage activity, and affects joint immune regulation. Studying inflammatory macrophage expression and strategies to inhibit inflammatory macrophage phenotype polarization might provide insights into disease pathogenesis and therapeutic applications. In pre-clinical studies, the detection of OA in its initial stages was shown to be possible using imaging techniques such as SPECT-CT, and advances are made to detect OA through blood-based biomarker analysis. In this review, obesity-induced osteoarthritis and its mechanisms in inducing joint degeneration are summarized, along with an analysis of the current developments in patient imaging and biomarker use for diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Kelly Warmink
- Department of Orthopedics, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Prateeksha Vinod
- Department of Orthopedics, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Nicoline M Korthagen
- Department of Orthopedics, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
- Department of Equine Sciences, Faculty of Veterinary Medicine, Utrecht University, 3584 CL Utrecht, The Netherlands
| | - Harrie Weinans
- Department of Orthopedics, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
- Department of Biomechanical Engineering, TU Delft, 2628 CD Delft, The Netherlands
| | - Jaqueline L Rios
- Department of Orthopedics, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| |
Collapse
|
166
|
Cao X, Wu S, Wang X, Huang J, Zhang W, Liang C. Receptor tyrosine kinase C-kit promotes a destructive phenotype of FLS in osteoarthritis via intracellular EMT signaling. Mol Med 2023; 29:38. [PMID: 36959556 PMCID: PMC10037859 DOI: 10.1186/s10020-023-00633-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 03/12/2023] [Indexed: 03/25/2023] Open
Abstract
BACKGROUND Chronic inflammation, mainly derived from fibroblast-like synoviocytes (FLSs), plays a central role in the pathomechanism of osteoarthritis (OA). Recently, epithelial-mesenchymal transition (EMT) signaling was found to be activated in OA-derived FLSs with a pro-inflammatory phenotype. However, the role of EMT signaling in regulating FLS function and OA-related inflammation remains unknown. METHODS The synovium of OA patients were evaluated for EMT and inflammation markers. The FLSs with activated EMT signaling were co-cultured with chondrocytes (chond). Gene expression of OA synovial samples were analyzed. The role of receptor tyrosine kinase C-kit was investigated in OA-FLSs and an OA rat model. The downstream pathways driven by C-kit were explored in OA-FLSs. RESULTS EMT marker N-cadherin (N-CDH) was upregulated in 40.0% of the OA samples. These N-CDH+ OA samples showed higher expression of pro-inflammatory factors. In co-culture, FLSs derived from N-CDH+ OA samples induced a typical degenerative phenotype of chonds and stimulated their production of matrix degrading enzymes. C-kit was significantly upregulated and spatially co-localized with N-CDH in N-CDH+ OA samples. In OA-FLSs, C-kit activated intracellular EMT signaling and induced destructive features of OA-FLSs. In OA rat model, C-kit largely promoted synovial inflammation and cartilage destruction, whereas knocking-down C-kit significantly restored the health of OA joints. Using GSK3β S9A mutant, we demonstrated that C-kit drives EMT signaling in OA-FLS by promoting phosphorylation of GSK3β and nuclear retention of the EMT transcription factor Snail. CONCLUSION C-kit drives EMT signaling in OA-FLSs and promotes a destructive FLS phenotype, leading to synovial inflammation and cartilage destruction.
Collapse
Affiliation(s)
- Xu Cao
- Department of Orthopaedics of the 3rd Xiangya Hospital, Central South University, 138 Tongzipo Road, Changsha, 410013, China
- Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Beijing, China
| | - Song Wu
- Department of Orthopaedics of the 3rd Xiangya Hospital, Central South University, 138 Tongzipo Road, Changsha, 410013, China
| | - Xinxing Wang
- Department of Orthopaedics of the 3rd Xiangya Hospital, Central South University, 138 Tongzipo Road, Changsha, 410013, China
| | - Junjie Huang
- Department of Orthopaedics of the 3rd Xiangya Hospital, Central South University, 138 Tongzipo Road, Changsha, 410013, China
| | - Wenxiu Zhang
- Department of Orthopaedics of the 3rd Xiangya Hospital, Central South University, 138 Tongzipo Road, Changsha, 410013, China
- Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Beijing, China
| | - Chi Liang
- Department of Orthopaedics of the 3rd Xiangya Hospital, Central South University, 138 Tongzipo Road, Changsha, 410013, China.
| |
Collapse
|
167
|
Kongdang P, Ongchai S, Chiranthanut N, Pruksakorn D. Anti-arthritis Effects of Zingiberaceae Extracts on Models of Inflammatory Joint Disease. PLANTA MEDICA 2023. [PMID: 36940710 DOI: 10.1055/a-2044-9129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Due to this becoming an aging society, the number of arthritis cases has been increasing. Unfortunately, some currently available medications can cause adverse effects. Using herbal remedies as a form of alternative medicine is becoming increasingly popular. Zingiber officinale (ZO), Curcuma longa (CL), and Kaempferia parviflora (KP) are herbal plants in the Zingiberaceae family that have potent anti-inflammatory effects. This study investigates the anti-inflammatory and chondroprotective effects of ZO, CL, and KP extracts on in vitro and ex vivo inflammatory models. The combinatorial anti-arthritis effect of each extract is also evaluated in an in vivo model. ZO extract preserves cartilaginous proteoglycans in proinflammatory cytokines-induced porcine cartilage explant in a fashion similar to that of CL and KP extracts and suppresses the expression of major inflammatory mediators in SW982 cells, particularly the COX2 gene. CL extract downregulates some inflammatory mediators and genes-associated cartilage degradation. Only KP extract shows a significant reduction in S-GAGs release in a cartilage explant model compared to the positive control, diacerein. In SW982 cells, it strongly suppresses many inflammatory mediators. The active constituents of each extract selectively downregulate inflammatory genes. The combined extracts show a reduction in inflammatory mediators to a similar degree as the combined active constituents. Reductions in paw swelling, synovial vascularity, inflammatory cell infiltration, and synovial hyperplasia are found in the combined extracts-treated arthritic rats. This study demonstrates that a combination of ZO, CL, and KP extracts has an anti-arthritis effect and could potentially be developed into an anti-arthritis cocktail for arthritis treatment.
Collapse
Affiliation(s)
- Patiwat Kongdang
- Center of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Siriwan Ongchai
- Thailand Excellence Center for Tissue Engineering and Stem Cells, Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Natthakarn Chiranthanut
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Dumnoensun Pruksakorn
- Center of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Musculoskeletal Science and Translational Research (MSTR) Center, Department of Orthopaedics, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| |
Collapse
|
168
|
Chen CC, Chang SS, Chang CH, Hu CC, Nakao Y, Yong SM, Mandy YLO, Lim CJ, Shim EKS, Shih HN. Randomized, double-blind, four-arm pilot study on the effects of chicken essence and type II collagen hydrolysate on joint, bone, and muscle functions. Nutr J 2023; 22:17. [PMID: 36918892 PMCID: PMC10015953 DOI: 10.1186/s12937-023-00837-w] [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: 01/27/2022] [Accepted: 01/05/2023] [Indexed: 03/16/2023] Open
Abstract
BACKGROUND Knee osteoarthritis (OA) is a leading cause of disability among older adults. Medical and surgical treatments are costly and associated with side effects. A natural nutraceutical, collagen hydrolysate, has received considerable attention due to its relieving effects on OA-associated symptoms. This study investigated the effects of hydrolyzed collagen type II (HC-II) and essence of chicken (BRAND'S Essence of Chicken) with added HC-II (EC-HC-II) on joint, muscle, and bone functions among older adults with OA. METHODS Patients (n = 160) with grade 1-3 knee OA according to the Kellgren-Lawrence classification system, joint pain for ≥ 3 months, and a Western Ontario and McMaster Universities Osteoarthritis Index (WOMAC) score of > 6 were randomly assigned with equal probability to consume EC-HC-II, HC-II, glucosamine HCl, or a placebo for 24 weeks in combination with resistance training. Outcome measurements were WOMAC score, visual analogue scale (VAS) pain score, grip strength, fat-free mass (FFM), and bone mass. RESULTS All groups exhibited similar levels of improvement in WOMAC index scores after 24 weeks. HC-II significantly reduced VAS pain score by 0.9 ± 1.89 (p = 0.034) after 14 days. A repeated-measures analysis of variance showed that HC-II reduced pain levels more than the placebo did (mean ± standard error: - 1.3 ± 0.45, p = 0.021) after 14 days; the EC-HC-II group also had significantly higher FFM than the glucosamine HCl (p = 0.02) and placebo (p = 0.017) groups and significantly higher grip strength than the glucosamine HCl group (p = 0.002) at 24 weeks. CONCLUSION HC-II reduces pain, and EC-HC-II may improve FFM and muscle strength. This suggests that EC-HC-II may be a novel holistic solution for mobility by improving joint, muscle, and bone health among older adults. Large-scale studies should be conducted to validate these findings. TRIAL REGISTRATION This trial was retrospectively registered at ClinicalTrials.gov (NCT04483024).
Collapse
Affiliation(s)
- Chun-Chieh Chen
- Department of Orthopedic Surgery, Chang Gung Memorial Hospital, Linkou, No. 5, Fuxing Street, Guishan District, Taoyuan City 333, Taiwan.,College of Medicine, Chang Gung University, No. 259, Wenhua 1st Road, Guishan District, Taoyuan City, 333, Taiwan.,Bone and Joint Research Center, Chang Gung Memorial Hospital, Linkou, No. 5, Fuxing Street, Guishan District, Taoyuan City, 333, Taiwan
| | - Shih-Sheng Chang
- Department of Orthopedic Surgery, Chang Gung Memorial Hospital, Linkou, No. 5, Fuxing Street, Guishan District, Taoyuan City 333, Taiwan.,College of Medicine, Chang Gung University, No. 259, Wenhua 1st Road, Guishan District, Taoyuan City, 333, Taiwan.,Bone and Joint Research Center, Chang Gung Memorial Hospital, Linkou, No. 5, Fuxing Street, Guishan District, Taoyuan City, 333, Taiwan
| | - Chih-Hsiang Chang
- Department of Orthopedic Surgery, Chang Gung Memorial Hospital, Linkou, No. 5, Fuxing Street, Guishan District, Taoyuan City 333, Taiwan.,College of Medicine, Chang Gung University, No. 259, Wenhua 1st Road, Guishan District, Taoyuan City, 333, Taiwan.,Bone and Joint Research Center, Chang Gung Memorial Hospital, Linkou, No. 5, Fuxing Street, Guishan District, Taoyuan City, 333, Taiwan.,Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, No. 259, Wenhua 1St Road, Guishan District, Taoyuan City, 333, Taiwan
| | - Chih-Chien Hu
- Department of Orthopedic Surgery, Chang Gung Memorial Hospital, Linkou, No. 5, Fuxing Street, Guishan District, Taoyuan City 333, Taiwan.,College of Medicine, Chang Gung University, No. 259, Wenhua 1st Road, Guishan District, Taoyuan City, 333, Taiwan.,Bone and Joint Research Center, Chang Gung Memorial Hospital, Linkou, No. 5, Fuxing Street, Guishan District, Taoyuan City, 333, Taiwan
| | - Yoshihiro Nakao
- Research and Development, Suntory Beverage and Food Asia, 3 Biopolis Drive, #06-14/19, Synapse, Singapore, 138623, Singapore
| | - Shan May Yong
- Research and Development, Suntory Beverage and Food Asia, 3 Biopolis Drive, #06-14/19, Synapse, Singapore, 138623, Singapore
| | - Yen Ling Ow Mandy
- Research and Development, Suntory Beverage and Food Asia, 3 Biopolis Drive, #06-14/19, Synapse, Singapore, 138623, Singapore
| | - Chia Juan Lim
- Research and Development, Suntory Beverage and Food Asia, 3 Biopolis Drive, #06-14/19, Synapse, Singapore, 138623, Singapore
| | - Eric Kian-Shiun Shim
- Research and Development, Suntory Beverage and Food Asia, 3 Biopolis Drive, #06-14/19, Synapse, Singapore, 138623, Singapore
| | - Hsin-Nung Shih
- Department of Orthopedic Surgery, Chang Gung Memorial Hospital, Linkou, No. 5, Fuxing Street, Guishan District, Taoyuan City 333, Taiwan. .,College of Medicine, Chang Gung University, No. 259, Wenhua 1st Road, Guishan District, Taoyuan City, 333, Taiwan. .,Bone and Joint Research Center, Chang Gung Memorial Hospital, Linkou, No. 5, Fuxing Street, Guishan District, Taoyuan City, 333, Taiwan.
| |
Collapse
|
169
|
Liu S, Pan Y, Li T, Zou M, Liu W, Li Q, Wan H, Peng J, Hao L. The Role of Regulated Programmed Cell Death in Osteoarthritis: From Pathogenesis to Therapy. Int J Mol Sci 2023; 24:5364. [PMID: 36982438 PMCID: PMC10049357 DOI: 10.3390/ijms24065364] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 03/08/2023] [Accepted: 03/09/2023] [Indexed: 03/14/2023] Open
Abstract
Osteoarthritis (OA) is a worldwide chronic disease that can cause severe inflammation to damage the surrounding tissue and cartilage. There are many different factors that can lead to osteoarthritis, but abnormally progressed programmed cell death is one of the most important risk factors that can induce osteoarthritis. Prior studies have demonstrated that programmed cell death, including apoptosis, pyroptosis, necroptosis, ferroptosis, autophagy, and cuproptosis, has a great connection with osteoarthritis. In this paper, we review the role of different types of programmed cell death in the generation and development of OA and how the different signal pathways modulate the different cell death to regulate the development of OA. Additionally, this review provides new insights into the radical treatment of osteoarthritis rather than conservative treatment, such as anti-inflammation drugs or surgical operation.
Collapse
Affiliation(s)
- Suqing Liu
- Department of Orthopedics, Second Affifiliated Hospital of Nanchang University, Nanchang 330006, China
- Queen Marry College, Nanchang University, Nanchang 330006, China
| | - Yurong Pan
- Department of Orthopedics, Second Affifiliated Hospital of Nanchang University, Nanchang 330006, China
- Queen Marry College, Nanchang University, Nanchang 330006, China
| | - Ting Li
- Department of Orthopedics, Second Affifiliated Hospital of Nanchang University, Nanchang 330006, China
- The Second Clinical Medical College, Nanchang University, Nanchang 330006, China
| | - Mi Zou
- Department of Orthopedics, Second Affifiliated Hospital of Nanchang University, Nanchang 330006, China
- The Second Clinical Medical College, Nanchang University, Nanchang 330006, China
| | - Wenji Liu
- Department of Orthopedics, Second Affifiliated Hospital of Nanchang University, Nanchang 330006, China
- The Second Clinical Medical College, Nanchang University, Nanchang 330006, China
| | - Qingqing Li
- Department of Orthopedics, Second Affifiliated Hospital of Nanchang University, Nanchang 330006, China
- The Second Clinical Medical College, Nanchang University, Nanchang 330006, China
| | - Huan Wan
- Department of Orthopedics, Second Affifiliated Hospital of Nanchang University, Nanchang 330006, China
- The Second Clinical Medical College, Nanchang University, Nanchang 330006, China
| | - Jie Peng
- The Second Clinical Medical College, Nanchang University, Nanchang 330006, China
| | - Liang Hao
- Department of Orthopedics, Second Affifiliated Hospital of Nanchang University, Nanchang 330006, China
| |
Collapse
|
170
|
Botoseneanu A, Markwardt S, Quiñones AR. Multimorbidity and Functional Disability among Older Adults: The Role of Inflammation and Glycemic Status - An Observational Longitudinal Study. Gerontology 2023; 69:826-838. [PMID: 36858034 PMCID: PMC10442862 DOI: 10.1159/000528648] [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: 01/07/2022] [Accepted: 12/07/2022] [Indexed: 03/03/2023] Open
Abstract
INTRODUCTION Specific multimorbidity combinations, in particular those including arthritis, stroke, and cognitive impairment, have been associated with high burden of activities of daily living (ADL)-instrumental activities of daily living (IADL) disability in older adults. The biologic underpinnings of these associations are still unclear. METHODS Observational longitudinal study using data from the Health and Retirement Study (N = 8,618, mean age = 74 years, 58% female, 25% non-white) and negative binomial regression models stratified by sex to evaluate the role of inflammatory and glycemic biomarkers (high-sensitivity C-reactive protein (hs-CRP) and HbA1c) in the association between specific multimorbidity combinations (grouped around one of eight index diseases: arthritis, cancer, cognitive impairment, diabetes, heart disease, hypertension, lung disease, and stroke; assessed between 2006 and 2014) and prospective ADL-IADL disability (2 years later, 2008-2016). Results were adjusted for sociodemographic characteristics, body mass index, number of coexisting diseases, and baseline ADL-IADL score. RESULTS Multimorbidity combinations indexed by arthritis (IRR = 1.1, 95% CI = 1.01-1.20), diabetes (IRR = 1.19, 95% CI = 1.09-1.30), and cognitive impairment (IRR = 1.11, 95% CI = 1.01-1.23) among men and diabetes-indexed multimorbidity combinations (IRR = 1.07, 95% CI = 1.01-1.14) among women were associated with higher ADL-IADL scores at increasing levels of HbA1c. Across higher levels of hs-CRP, multimorbidity combinations indexed by arthritis (IRR = 1.06, 95% CI = 1.02-1.11), hypertension (IRR = 1.06, 95% CI = 1.02-1.11), heart disease (IRR = 1.06, 95% CI = 1.01-1.12), and lung disease (IRR = 1.14, 95% CI = 1.07-1.23) were associated with higher ADL-IADL scores among women, while there were no significant associations among men. CONCLUSION The findings suggest potential for anti-inflammatory management among older women and optimal glycemic control among older men with these particular multimorbidity combinations as focus for therapeutic/preventive options for maintaining functional health.
Collapse
Affiliation(s)
- Anda Botoseneanu
- Department of Health & Human Services, University of Michigan, Dearborn, MI, USA
- Institute of Gerontology, University of Michigan, Ann Arbor, MI, USA
| | - Sheila Markwardt
- School of Public Health, Oregon Health & Science University, Portland, OR, USA
| | - Ana R. Quiñones
- School of Public Health, Oregon Health & Science University, Portland, OR, USA
- Department of Family Medicine, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
171
|
Fine N, Lively S, Séguin CA, Perruccio AV, Kapoor M, Rampersaud R. Intervertebral disc degeneration and osteoarthritis: a common molecular disease spectrum. Nat Rev Rheumatol 2023; 19:136-152. [PMID: 36702892 DOI: 10.1038/s41584-022-00888-z] [Citation(s) in RCA: 54] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/05/2022] [Indexed: 01/27/2023]
Abstract
Intervertebral disc degeneration (IDD) and osteoarthritis (OA) affecting the facet joint of the spine are biomechanically interdependent, typically occur in tandem, and have considerable epidemiological and pathophysiological overlap. Historically, the distinctions between these degenerative diseases have been emphasized. Therefore, research in the two fields often occurs independently without adequate consideration of the co-dependence of the two sites, which reside within the same functional spinal unit. Emerging evidence from animal models of spine degeneration highlight the interdependence of IDD and facet joint OA, warranting a review of the parallels between these two degenerative phenomena for the benefit of both clinicians and research scientists. This Review discusses the pathophysiological aspects of IDD and OA, with an emphasis on tissue, cellular and molecular pathways of degeneration. Although the intervertebral disc and synovial facet joint are biologically distinct structures that are amenable to reductive scientific consideration, substantial overlap exists between the molecular pathways and processes of degeneration (including cartilage destruction, extracellular matrix degeneration and osteophyte formation) that occur at these sites. Thus, researchers, clinicians, advocates and policy-makers should consider viewing the burden and management of spinal degeneration holistically as part of the OA disease continuum.
Collapse
Affiliation(s)
- Noah Fine
- Osteoarthritis Research Program, Division of Orthopaedics, Schroeder Arthritis Institute, University Health Network, Toronto, Ontario, Canada.,Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Starlee Lively
- Osteoarthritis Research Program, Division of Orthopaedics, Schroeder Arthritis Institute, University Health Network, Toronto, Ontario, Canada.,Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Cheryle Ann Séguin
- Department of Physiology & Pharmacology, Schulich School of Medicine & Dentistry, Bone and Joint Institute, University of Western Ontario London, London, Ontario, Canada
| | - Anthony V Perruccio
- Osteoarthritis Research Program, Division of Orthopaedics, Schroeder Arthritis Institute, University Health Network, Toronto, Ontario, Canada.,Krembil Research Institute, University Health Network, Toronto, Ontario, Canada.,Institute of Health Policy, Management and Evaluation, Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada.,Department of Surgery, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Mohit Kapoor
- Osteoarthritis Research Program, Division of Orthopaedics, Schroeder Arthritis Institute, University Health Network, Toronto, Ontario, Canada.,Krembil Research Institute, University Health Network, Toronto, Ontario, Canada.,Department of Surgery, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Raja Rampersaud
- Osteoarthritis Research Program, Division of Orthopaedics, Schroeder Arthritis Institute, University Health Network, Toronto, Ontario, Canada. .,Krembil Research Institute, University Health Network, Toronto, Ontario, Canada. .,Department of Surgery, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
172
|
Zapata A, Fernández-Parra R. Management of Osteoarthritis and Joint Support Using Feed Supplements: A Scoping Review of Undenatured Type II Collagen and Boswellia serrata. Animals (Basel) 2023; 13:870. [PMID: 36899726 PMCID: PMC10000124 DOI: 10.3390/ani13050870] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 02/17/2023] [Accepted: 02/19/2023] [Indexed: 03/06/2023] Open
Abstract
In the multimodal management of osteoarthritis (OA) in recent decades, the use of feed supplements to maintain joint cartilage has been advocated. The aim of this scoping review is to present the results found in the veterinary literature on the use of undenatured type II collagen and Boswellia serrata in dogs, specifically its use in dogs with clinical signs of OA, healthy dogs after intense exercise or dogs with diseases that predispose the individual to OA. For this purpose, a literature review was carried out using the electronic databases PubMed, Web of Science and Google Scholar, from which a total of 26 records were included in this review: fourteen evaluating undenatured type II collagen, ten evaluating Boswellia serrata and two evaluating the combination of undenatured type II collagen and Boswellia serrata. The review of the records showed that undenatured type II collagen decreases the clinical signs associated with OA, improving the general clinical state with a reduction in the degree of lameness and increase in physical activity or mobility. Evaluating the response to supplementation with Boswellia serrata alone is complicated due to the limited publication of studies and variations in the purity and compositions of the products used, but in general terms, its combination with other feed supplements produces benefits by relieving pain and reducing the clinical signs of OA in dogs. The combination of both in the same product provides results similar to those obtained in undenatured type II collagen studies. In conclusion, undenatured type II collagen and Boswellia serrata are considered a valid option for the multimodal approach to the management of OA and for improving activity during intense exercise, but more studies are needed to conclude whether or not it prevents OA in dogs.
Collapse
Affiliation(s)
- Ana Zapata
- Hospital Veterinario de Referencia UCV, Departamento de Medicina y Cirugía Animal, Facultad de Veterinaria y Ciencias Experimentales, Universidad Católica de Valencia San Vicente Mártir, 46018 Valencia, Spain
- Escuela de Doctorado, Programa de Doctorado en Ciencias de la Vida y del Medio Natural, Universidad Católica de Valencia San Vicente Mártir, 46002 Valencia, Spain
| | - Rocio Fernández-Parra
- Hospital Veterinario de Referencia UCV, Departamento de Medicina y Cirugía Animal, Facultad de Veterinaria y Ciencias Experimentales, Universidad Católica de Valencia San Vicente Mártir, 46018 Valencia, Spain
- Departamento Medicina y Cirugía Animal, Facultad de Veterinaria y Ciencias Experimentales, Universidad Católica de Valencia San Vicente Mártir, 46002 Valencia, Spain
| |
Collapse
|
173
|
Intracellular Delivery of Itaconate by Metal–Organic Framework-Anchored Hydrogel Microspheres for Osteoarthritis Therapy. Pharmaceutics 2023; 15:pharmaceutics15030724. [PMID: 36986584 PMCID: PMC10051475 DOI: 10.3390/pharmaceutics15030724] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 02/01/2023] [Accepted: 02/20/2023] [Indexed: 02/24/2023] Open
Abstract
Treatment of osteoarthritis (OA) remains a significant clinical challenge. Itaconate (IA), an emerging regulator of intracellular inflammation and oxidative stress, may potentially be harnessed to treat OA. However, the short joint residence time, inefficient drug delivery, and cell-impermeable property of IA can seriously hamper the clinical translation. Herein, IA-encapsulated zeolitic imidazolate framework-8 (IA-ZIF-8) nanoparticles were self-assembled by zinc ions, 2-methylimidazole, and IA to render them pH-responsive. Subsequently, IA-ZIF-8 nanoparticles were firmly immobilized in hydrogel microspheres via one-step microfluidic technology. It was demonstrated in vitro experiments that IA-ZIF-8-loaded hydrogel microspheres (IA-ZIF-8@HMs) exhibited good anti-inflammatory and anti-oxidative stress effects by releasing pH-responsive nanoparticles into chondrocytes. Importantly, compared with IA-ZIF-8, IA-ZIF-8@HMs showed better performance in the treatment of OA due to their superior performance in sustained release. Thus, such hydrogel microspheres not only hold enormous potential for OA therapy, but also provide a novel avenue for cell-impermeable drugs by constructing appropriate drug delivery systems.
Collapse
|
174
|
Xu W, Wang X, Liu D, Lin X, Wang B, Xi C, Kong P, Yan J. Identification and validation of hub genes and potential drugs involved in osteoarthritis through bioinformatics analysis. Front Genet 2023; 14:1117713. [PMID: 36845391 PMCID: PMC9947480 DOI: 10.3389/fgene.2023.1117713] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 02/01/2023] [Indexed: 02/11/2023] Open
Abstract
Purpose: Osteoarthritis (OA) is a common degenerative disease, which still lacks specific therapeutic drugs. Synovitis is one of the most important pathological process in OA. Therefore, we aim to identify and analyze the hub genes and their related networks of OA synovium with bioinformatics tools to provide theoretical basis for potential drugs. Materials and methods: Two datasets were obtained from GEO. DEGs and hub genes of OA synovial tissue were screened through Gene Ontology (GO) annotation, Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment as well as protein-protein interaction (PPI) network analysis. Subsequently, the correlation between expression of hub genes and ferroptosis or pyroptosis was analyzed. CeRNA regulatory network was constructed after predicting the upstream miRNAs and lncRNAs. The validation of hub genes was undertook through RT-qPCR and ELISA. Finally, potential drugs targeting pathways and hub genes were identified, followed by the validation of the effect of two potential drugs on OA. Results: A total of 161 commom DEGs were obtained, of which 8 genes were finally identified as hub genes through GO and KEGG enrichment analysis as well as PPI network analysis. Eight genes related to ferroptosis and pyroptosis respectively were significantly correlated to the expression of hub genes. 24 miRNAs and 69 lncRNAs were identified to construct the ceRNA regulatory network. The validation of EGR1, JUN, MYC, FOSL1, and FOSL2 met the trend of bioinformatics analysis. Etanercept and Iguratimod reduced the secretion of MMP-13 and ADAMTS5 of fibroblast-like synoviocyte. Conclusion: EGR1, JUN, MYC, FOSL1, and FOSL2 were identified as hub genes in the development of OA after series of bioinformatics analysis and validation. Etanercept and Iguratimod seemed to have opportunities to be novel drugs for OA.
Collapse
Affiliation(s)
- Wenbo Xu
- The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xuyao Wang
- Department of Pharmacy, Harbin Second Hospital, Harbin, China
| | - Donghui Liu
- Department of Oncology, Heilongjiang Provincial Hospital, Harbin, China
| | - Xin Lin
- The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Bo Wang
- The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Chunyang Xi
- The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Pengyu Kong
- The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jinglong Yan
- The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
175
|
Lambova SN. Pleiotropic Effects of Metformin in Osteoarthritis. Life (Basel) 2023; 13:life13020437. [PMID: 36836794 PMCID: PMC9960992 DOI: 10.3390/life13020437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 01/15/2023] [Accepted: 01/25/2023] [Indexed: 02/09/2023] Open
Abstract
The involvement of the knee joint is the most common localization of the pathological process in osteoarthritis (OA), which is associated with obesity in over 50% of the patients and is mediated by mechanical, inflammatory, and metabolic mechanisms. Obesity and the associated conditions (hyperglycemia, dyslipidemia, and hypertension) have been found to be risk factors for the development of knee OA, which has led to the emerging concept of the existence of a distinct phenotype, i.e., metabolic knee OA. Combined assessment of markers derived from dysfunctional adipose tissue, markers of bone and cartilage metabolism, as well as high-sensitivity inflammatory markers and imaging, might reveal prognostic signs for metabolic knee OA. Interestingly, it has been suggested that drugs used for the treatment of other components of the metabolic syndrome may also affect the clinical course and retard the progression of metabolic-associated knee OA. In this regard, significant amounts of new data are accumulating about the role of metformin-a drug, commonly used in clinical practice with suggested multiple pleiotropic effects. The aim of the current review is to analyze the current views about the potential pleiotropic effects of metformin in OA. Upon the analysis of the different effects of metformin, major mechanisms that might be involved in OA are the influence of inflammation, oxidative stress, autophagy, adipokine levels, and microbiome modulation. There is an increasing amount of evidence from in vitro studies, animal models, and clinical trials that metformin can slow OA progression by modulating inflammatory and metabolic factors that are summarized in the current up-to-date review. Considering the contemporary concept about the existence of metabolic type knee OA, in which the accompanying obesity and systemic low-grade inflammation are suggested to influence disease course, metformin could be considered as a useful and safe component of the personalized therapeutic approach in knee OA patients with accompanying type II diabetes or obesity.
Collapse
Affiliation(s)
- Sevdalina Nikolova Lambova
- Department of Propaedeutics of Internal Diseases “Prof Dr Anton Mitov”, Faculty of Medicine, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria;
- Department in Rheumatology, MHAT “Sveti Mina”, 4002 Plovdiv, Bulgaria
| |
Collapse
|
176
|
Rapp AE, Zaucke F. Cartilage extracellular matrix-derived matrikines in osteoarthritis. Am J Physiol Cell Physiol 2023; 324:C377-C394. [PMID: 36571440 DOI: 10.1152/ajpcell.00464.2022] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Osteoarthritis (OA) is among the most frequent diseases of the musculoskeletal system. Degradation of cartilage extracellular matrix (ECM) is a hallmark of OA. During the degradation process, intact/full-length proteins and proteolytic fragments are released which then might induce different downstream responses via diverse receptors, therefore leading to different biological consequences. Collagen type II and the proteoglycan aggrecan are the most abundant components of the cartilage ECM. However, over the last decades, a large number of minor components have been identified and for some of those, a role in the manifold processes associated with OA has already been demonstrated. To date, there is still no therapy able to halt or cure OA. A better understanding of the matrikine landscape occurring with or even preceding obvious degenerative changes in joint tissues is needed and might help to identify molecules that could serve as biomarkers, druggable targets, or even be blueprints for disease modifying drug OA drugs. For this narrative review, we screened PubMed for relevant literature in the English language and summarized the current knowledge regarding the function of selected ECM molecules and the derived matrikines in the context of cartilage and OA.
Collapse
Affiliation(s)
- Anna E Rapp
- Dr. Rolf M. Schwiete Research Unit for Osteoarthritis, Department of Orthopedics (Friedrichsheim), University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - Frank Zaucke
- Dr. Rolf M. Schwiete Research Unit for Osteoarthritis, Department of Orthopedics (Friedrichsheim), University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| |
Collapse
|
177
|
Bradley PX, Thomas KN, Kratzer AL, Robinson AC, Wittstein JR, DeFrate LE, McNulty AL. The Interplay of Biomechanical and Biological Changes Following Meniscus Injury. Curr Rheumatol Rep 2023; 25:35-46. [PMID: 36479669 PMCID: PMC10267895 DOI: 10.1007/s11926-022-01093-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/31/2022] [Indexed: 12/12/2022]
Abstract
PURPOSE OF REVIEW Meniscus injury often leads to joint degeneration and post-traumatic osteoarthritis (PTOA) development. Therefore, the purpose of this review is to outline the current understanding of biomechanical and biological repercussions following meniscus injury and how these changes impact meniscus repair and PTOA development. Moreover, we identify key gaps in knowledge that must be further investigated to improve meniscus healing and prevent PTOA. RECENT FINDINGS Following meniscus injury, both biomechanical and biological alterations frequently occur in multiple tissues in the joint. Biomechanically, meniscus tears compromise the ability of the meniscus to transfer load in the joint, making the cartilage more vulnerable to increased strain. Biologically, the post-injury environment is often characterized by an increase in pro-inflammatory cytokines, catabolic enzymes, and immune cells. These multi-faceted changes have a significant interplay and result in an environment that opposes tissue repair and contributes to PTOA development. Additionally, degenerative changes associated with OA may cause a feedback cycle, negatively impacting the healing capacity of the meniscus. Strides have been made towards understanding post-injury biological and biomechanical changes in the joint, their interplay, and how they affect healing and PTOA development. However, in order to improve clinical treatments to promote meniscus healing and prevent PTOA development, there is an urgent need to understand the physiologic changes in the joint following injury. In particular, work is needed on the in vivo characterization of the temporal biomechanical and biological changes that occur in patients following meniscus injury and how these changes contribute to PTOA development.
Collapse
Affiliation(s)
- Patrick X Bradley
- Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC, USA
| | - Karl N Thomas
- Department of Orthopaedic Surgery, Duke University School of Medicine, DUMC Box 3093, Durham, NC, 27710, USA
| | - Avery L Kratzer
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Allison C Robinson
- Department of Orthopaedic Surgery, Duke University School of Medicine, DUMC Box 3093, Durham, NC, 27710, USA
| | - Jocelyn R Wittstein
- Department of Orthopaedic Surgery, Duke University School of Medicine, DUMC Box 3093, Durham, NC, 27710, USA
| | - Louis E DeFrate
- Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC, USA
- Department of Orthopaedic Surgery, Duke University School of Medicine, DUMC Box 3093, Durham, NC, 27710, USA
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Amy L McNulty
- Department of Orthopaedic Surgery, Duke University School of Medicine, DUMC Box 3093, Durham, NC, 27710, USA.
- Department of Biomedical Engineering, Duke University, Durham, NC, USA.
- Department of Pathology, Duke University School of Medicine, Durham, NC, USA.
| |
Collapse
|
178
|
Cai T, Ye H, Jiang H, Lin C, Lou C, Wang W, Yan Z, Xue X, Pan X, Lin J. Stevioside targets the NF-κB and MAPK pathways for inhibiting inflammation and apoptosis of chondrocytes and ameliorates osteoarthritis in vivo. Int Immunopharmacol 2023; 115:109683. [PMID: 36630751 DOI: 10.1016/j.intimp.2023.109683] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 12/21/2022] [Accepted: 12/31/2022] [Indexed: 01/11/2023]
Abstract
Osteoarthritis (OA) is a joint disease that is characterized by articular cartilage degeneration and destruction. Stevioside (SVS) is a diterpenoid glycoside extracted from Stevia rebaudiana Bertoni with some specific effects against inflammatory and apoptotic, whereas it is still unclear what function SVS has in osteoarthritis. This study focuses on the anti-inflammatory and anti-apoptosis functions of SVS on chondrocytes induced by interleukin (IL)-1beta, and the role of SVS in an osteoarthritis model for mice. We can detect the production of inflammatory factors such as nitric oxide (NO) and prostaglandin E2 (PGE2) using real-time quantitative polymerase chain reaction (RT-qPCR), the Griess reaction, and enzyme linked immunosorbent assay (ELISA). On the basis of Western blot, we have observed the protein expressions of cartilage matrix metabolism, inflammatory factors, and apoptosis of chondrocytes. Simultaneously, the pharmacological effects of SVS in mice were evaluated by hematoxylin and eosin (HE), toluidine blue, Safranin O, and immunohistochemical staining. The results show that SVS slows extracellular matrix degradation and chondrocyte apoptosis. In addition, SVS mediates its cellular effect by inhibiting the activation of mitogen-activated protein kinase (MAPK) and nuclear factor kappa B (NF-κB) signaling pathways. Meanwhile, molecular docking studies revealed that SVS has excellent binding capabilities to p65, extracellular signal-regulated kinase (ERK), p38, and c-Jun N-terminal kinase (JNK). The study suggests that SVS can be developed as a potential osteoarthritis treatment.
Collapse
Affiliation(s)
- Tingwen Cai
- Department of Orthopedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Key Laboratory of Orthopedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Hantao Ye
- Department of Orthopedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Key Laboratory of Orthopedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Hongyi Jiang
- Department of Orthopedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Key Laboratory of Orthopedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Chihao Lin
- Department of Orthopedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Key Laboratory of Orthopedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Chao Lou
- Department of Orthopedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Key Laboratory of Orthopedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Weidan Wang
- Department of Orthopedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Key Laboratory of Orthopedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Zijian Yan
- Department of Orthopedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Key Laboratory of Orthopedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Xinghe Xue
- Department of Orthopedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Xiaoyun Pan
- Department of Orthopedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China.
| | - Jian Lin
- Department of Orthopedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China.
| |
Collapse
|
179
|
Effects of 2-carba-cyclic phosphatidic acid derivatives on IL-1β-stimulated human chondrocytes. Prostaglandins Other Lipid Mediat 2023; 164:106699. [PMID: 36513319 DOI: 10.1016/j.prostaglandins.2022.106699] [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: 08/30/2022] [Revised: 11/21/2022] [Accepted: 12/05/2022] [Indexed: 12/14/2022]
Abstract
Osteoarthritis (OA) is a common joint disease characterized by the breakdown of subchondral bone and cartilage damage, most often affecting middle-aged and elderly people. Although the etiology of OA is still unknown, some reports suggest that inflammatory factors such as interleukin (IL)- 1β mediate the progression of OA. To investigate the effect of IL-1β and the possibility of treatment for OA, we applied 2-carba-cyclic phosphatidic acid (2ccPA) and its derivatives on human chondrocytes. 2ccPA is a synthesized phospholipid derived from a bioactive phospholipid mediator: cyclic phosphatidic acid (cPA). It has been previously reported that 2ccPA exhibits anti-inflammatory and chondroprotective effects in an OA animal model. 2ccPA and its ring-opened body (ROB) derivative significantly suppressed IL-1β-induced upregulation of IL-6, matrix metalloproteinase-13, and cyclooxygenase-2, as well as the degradation of type II collagen and aggrecan. However, the other two derivatives, namely the deacylated and ring-opened deacylated bodies, showed little effect on an IL-1β-exposed human chondrosarcoma cell-line. These data suggest that the intactness of 2ccPA and ROB is essential for anti-inflammatory effects on OA. Collectively, this study provides evidence that 2ccPA and ROB would be novel therapeutic agents for OA.
Collapse
|
180
|
Ma M, Cui G, Liu Y, Tang Y, Lu X, Yue C, Zhang X. Mesenchymal stem cell-derived extracellular vesicles, osteoimmunology and orthopedic diseases. PeerJ 2023; 11:e14677. [PMID: 36710868 PMCID: PMC9881470 DOI: 10.7717/peerj.14677] [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: 08/29/2022] [Accepted: 12/12/2022] [Indexed: 01/26/2023] Open
Abstract
Mesenchymal stem cells (MSCs) play an important role in tissue healing and regenerative medicine due to their self-renewal and multi-directional differentiation properties. MSCs exert their therapeutic effects mainly via the paracrine pathway, which involves the secretion of extracellular vesicles (EVs). EVs have a high drug loading capacity and can transport various molecules, such as proteins, nucleic acids, and lipids, that can modify the course of diverse diseases. Due to their ability to maintain the therapeutic effects of their parent cells, MSC-derived EVs have emerged as a promising, safe cell-free treatment approach for tissue regeneration. With advances in inflammation research and emergence of the field of osteoimmunology, evidence has accumulated pointing to the role of inflammatory and osteoimmunological processes in the occurrence and progression of orthopedic diseases. Several studies have shown that MSC-derived EVs participate in bone regeneration and the pathophysiology of orthopedic diseases by regulating the inflammatory environment, enhancing angiogenesis, and promoting the differentiation and proliferation of osteoblasts and osteoclasts. In this review, we summarize recent advances in the application and functions of MSC-derived EVs as potential therapies against orthopedic diseases, including osteoarthritis, intervertebral disc degeneration, osteoporosis and osteonecrosis.
Collapse
Affiliation(s)
- Maoxiao Ma
- Department of Orthopedics, Luoyang Orthopedic Hospital of Henan Province, Orthopedic Hospital of Henan Province, Luoyang, Henan, China
| | - Guofeng Cui
- Department of Orthopedics, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, Henan, China
| | - Youwen Liu
- Department of Orthopedics, Luoyang Orthopedic Hospital of Henan Province, Orthopedic Hospital of Henan Province, Luoyang, Henan, China
| | - Yanfeng Tang
- Department of Orthopedics, Luoyang Orthopedic Hospital of Henan Province, Orthopedic Hospital of Henan Province, Luoyang, Henan, China
| | - Xiaoshuai Lu
- Department of Orthopedics, Luoyang Orthopedic Hospital of Henan Province, Orthopedic Hospital of Henan Province, Luoyang, Henan, China
| | - Chen Yue
- Department of Orthopedics, Luoyang Orthopedic Hospital of Henan Province, Orthopedic Hospital of Henan Province, Luoyang, Henan, China
| | - Xue Zhang
- Department of Orthopedics, Luoyang Orthopedic Hospital of Henan Province, Orthopedic Hospital of Henan Province, Luoyang, Henan, China
| |
Collapse
|
181
|
Loucks A, Maerz T, Hankenson K, Moeser A, Colbath A. The multifaceted role of mast cells in joint inflammation and arthritis. Osteoarthritis Cartilage 2023; 31:567-575. [PMID: 36682447 DOI: 10.1016/j.joca.2023.01.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 01/03/2023] [Accepted: 01/10/2023] [Indexed: 01/22/2023]
Abstract
OBJECTIVE To review current knowledge surrounding the role of mast cells in joint inflammation and arthritis. METHOD Narrative review. RESULTS Mast cells (MCs) are commonly observed in the synovium of the joint, particularly surrounding blood vessels and nerve endings. Some studies have reported increased MC number and degranulation in patients with osteoarthritis (OA). In two studies, MCs were the only immune cell type found in higher concentrations in synovium of OA patients compared to rheumatoid arthritis patients. Activation of MCs in OA includes signaling pathways such as immunoglobulin E/Fc epsilon Receptor 1 (IgE/FcεR1), immunoglobulin G/Fc gamma receptor (IgG/FcγR), complement, and toll-like cell surface receptor-mediated signaling, resulting in context-dependent release of either pro-inflammatory and/or anti-inflammatory mediators within the joint. Activation of MCs results in the release of pro-inflammatory mediators that ultimately contribute to inflammation of the synovium, bone remodeling, and cartilage damage. However, some studies have proposed that MCs can also exhibit anti-inflammatory effects by secreting mediators that inactivate pro-inflammatory cytokines such as interleukin 6 (IL-6). CONCLUSIONS MCs may play a role in mediating synovial inflammation and OA progression. However, the mechanisms governing MC activation, the downstream pro- and/or anti-inflammatory effects, and their impact on osteoarthritis pathogenesis remains to be elucidated and requires extensive further study. Furthermore, it is important to establish the pathways of MC activation in OA to determine whether MCs exhibit varying phenotypes as a function of disease stage. Ultimately, such research is needed before understanding whether MCs could be targeted in OA treatments.
Collapse
Affiliation(s)
- A Loucks
- Department of Clinical Sciences, Cornell University, Ithaca, NY, USA.
| | - T Maerz
- Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, USA.
| | - K Hankenson
- Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, USA.
| | - A Moeser
- Department of Large Animal Clinical Sciences, Michigan State University, East Lansing, MI, USA.
| | - A Colbath
- Department of Clinical Sciences, Cornell University, Ithaca, NY, USA.
| |
Collapse
|
182
|
Jin H, Luo J, Jiang Y, Lin J, Jiang J, Ren R, Fang W, Wu Y, Wang X. Osteophyte formation causes neurological symptoms after anterior cervical discectomy and fusion (ACDF): A case report. Front Surg 2023; 9:1029743. [PMID: 36713656 PMCID: PMC9880155 DOI: 10.3389/fsurg.2022.1029743] [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: 08/27/2022] [Accepted: 12/27/2022] [Indexed: 01/15/2023] Open
Abstract
Spinal surgeons have been drawn to the incidence of osteophytes following intervertebral disc degeneration in clinical practice. However, the production of osteophytes, particularly in the spinal canal, after anterior cervical discectomy and fusion (ACDF) is uncommon. We described a 42-year-old male patient who underwent C4-6 ACDF due to cervical stenosis two years prior in another public hospital in the province. His primary symptoms were significantly relieved, but he developed new pain and weakness in his right leg six months after surgery. The imaging results revealed a large posterior osteophyte at C5/6, compressing the spinal cord anteriorly. Accordingly, we performed cervical open-door laminoplasty to decompress the spinal cord. The patient's clinical symptoms had significantly improved at the one-year follow-up. This case seeks to inform surgeons that cautious, routine follow-ups are necessary for the event that a severe intracanal osteophyte develops at the operated level following ACDF. The comprehensive osteophyte removal and strong fixation at the operative level during ACDF warrant more consideration as these procedures may lower the incidence of new osteophytes. Additionally, surgical procedures may be required.
Collapse
Affiliation(s)
- Haiming Jin
- Division of Spine Surgery, Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China,Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jiangtao Luo
- School of the Second Clinical Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Yuhan Jiang
- School of the Second Clinical Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Jinghao Lin
- School of the Second Clinical Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Junchen Jiang
- School of the Second Clinical Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Rufeng Ren
- School of the Second Clinical Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Weiyuan Fang
- School of the Second Clinical Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Yaosen Wu
- Division of Spine Surgery, Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China,Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiangyang Wang
- Division of Spine Surgery, Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China,Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China,Correspondence: Xiangyang Wang
| |
Collapse
|
183
|
Gupta DP, Lee YS, Choe Y, Kim KT, Song GJ, Hwang SC. Knee osteoarthritis accelerates amyloid beta deposition and neurodegeneration in a mouse model of Alzheimer's disease. Mol Brain 2023; 16:1. [PMID: 36593507 PMCID: PMC9809050 DOI: 10.1186/s13041-022-00986-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 12/10/2022] [Indexed: 01/03/2023] Open
Abstract
Knee osteoarthritis (OA) is characterized by knee cartilage degeneration and secondary bone hyperplasia, resulting in pain, stiffness, and gait disturbance. The relationship between knee OA and neurodegenerative diseases is still unclear. This study used an Alzheimer's disease (AD) mouse model to observe whether osteoarthritis accelerates dementia progression by analyzing brain histology and neuroinflammation. Knee OA was induced by destabilizing the medial meniscus (DMM) in control (WT) and AD (5xFAD) mice before pathological symptoms. Mouse knee joints were scanned with a micro-CT scanner. A sham operation was used as control. Motor and cognitive abilities were tested after OA induction. Neurodegeneration, β-amyloid plaque formation, and neuroinflammation were analyzed by immunostaining, Western blotting, and RT-PCR in brain tissues. Compared with sham controls, OA in AD mice increased inflammatory cytokine levels in brain tissues. Furthermore, OA significantly increased β-amyloid deposition and neuronal loss in AD mice compared to sham controls. In conclusion, knee OA accelerated amyloid plaque deposition and neurodegeneration in AD-OA mice, suggesting that OA is a risk factor for AD.
Collapse
Affiliation(s)
- Deepak Prasad Gupta
- grid.411199.50000 0004 0470 5702Department of Medical Science, College of Medicine, Catholic Kwandong University, Gangneung, Gangwon-Do Republic of Korea ,grid.411199.50000 0004 0470 5702Translational Brain Research Center, International St. Mary’s Hospital, Catholic Kwandong University, Incheon, Republic of Korea
| | - Young-Sun Lee
- grid.411199.50000 0004 0470 5702Department of Medical Science, College of Medicine, Catholic Kwandong University, Gangneung, Gangwon-Do Republic of Korea ,grid.411199.50000 0004 0470 5702Translational Brain Research Center, International St. Mary’s Hospital, Catholic Kwandong University, Incheon, Republic of Korea
| | - Youngshik Choe
- grid.452628.f0000 0004 5905 0571Korea Brain Research Institute, Daegu, Republic of Korea
| | - Kun-Tae Kim
- grid.411899.c0000 0004 0624 2502Department of Orthopaedic Surgery, Gyeongsang National University College of Medicine, Gyeongsang National University Hospital, Jinju-Si, Gyeongsangnam-Do Republic of Korea
| | - Gyun Jee Song
- grid.411199.50000 0004 0470 5702Department of Medical Science, College of Medicine, Catholic Kwandong University, Gangneung, Gangwon-Do Republic of Korea ,grid.411199.50000 0004 0470 5702Translational Brain Research Center, International St. Mary’s Hospital, Catholic Kwandong University, Incheon, Republic of Korea
| | - Sun-Chul Hwang
- grid.411899.c0000 0004 0624 2502Department of Orthopaedic Surgery, Gyeongsang National University College of Medicine, Gyeongsang National University Hospital, Jinju-Si, Gyeongsangnam-Do Republic of Korea
| |
Collapse
|
184
|
Manukyan G, Gallo J, Mikulkova Z, Trajerova M, Savara J, Slobodova Z, Fidler E, Shrestha B, Kriegova E. Phenotypic and functional characterisation of synovial fluid-derived neutrophils in knee osteoarthritis and knee infection. Osteoarthritis Cartilage 2023; 31:72-82. [PMID: 36216277 DOI: 10.1016/j.joca.2022.09.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 08/25/2022] [Accepted: 09/24/2022] [Indexed: 11/16/2022]
Abstract
OBJECTIVE An increase in the number of neutrophils (NEUs) has long been associated with infections in the knee joints; however, their impact on knee osteoarthritis (KOA) pathophysiology remains largely unexplored. DESIGN This study compared the phenotypic and functional characteristics of synovial fluid (SF)-derived NEUs in KOA and knee infection (INF). RESULTS KOA NEUs were characterised by a lower expression of CD11b, CD54, and CD64 and higher expression of CD62L, TLR2, and TLR4 compared with INF NEUs. Except for CCL2, lower levels of inflammatory mediators and proteases were detected in KOA SF than in INF SF. Functionally, KOA NEUs displayed increased reactive oxygen species production and phagocytic activity compared with INF NEUs. Moreover, KOA and INF NEUs differed in cell sizes, histological characteristics of the surrounding synovial tissues, and their effects on the endothelial cells assessed by human umbilical vein endothelial cells. When KOA patients were subdivided based on the SF NEU abundance, patients with high NEUs (10%-60%) were characterised by i) elevated SF protein levels of TNF-α, IL-1RA, MMP-9, sTREM-1, VILIP-1 and ii) lower CD54, CD64, TLR2 and TLR4 expression compared to patients with low NEUs (<10%). Analysis of paired SF samples suggests that low or high NEU percentages, respectively, persist throughout the course of disease. CONCLUSIONS Our findings suggest that NEU may play a significant role in KOA pathophysiology. Further studies should explore the mechanisms that contribute to the increased number of NEUs in SF and the clinical consequences of neutrophilic phenotype in KOA.
Collapse
Affiliation(s)
- G Manukyan
- Department of Immunology, Faculty of Medicine and Dentistry, Palacký University Olomouc and University Hospital Olomouc, Olomouc, Czech Republic; Laboratory of Molecular and Cellular Immunology, Institute of Molecular Biology NAS RA, Yerevan, Armenia.
| | - J Gallo
- Department of Orthopedics, Palacký University Olomouc and University Hospital Olomouc, Olomouc, Czech Republic.
| | - Z Mikulkova
- Department of Immunology, Faculty of Medicine and Dentistry, Palacký University Olomouc and University Hospital Olomouc, Olomouc, Czech Republic.
| | - M Trajerova
- Department of Immunology, Faculty of Medicine and Dentistry, Palacký University Olomouc and University Hospital Olomouc, Olomouc, Czech Republic.
| | - J Savara
- Department of Immunology, Faculty of Medicine and Dentistry, Palacký University Olomouc and University Hospital Olomouc, Olomouc, Czech Republic; Department of Computer Science, Faculty of Electrical Engineering and Computer Science, VSB-Technical University of Ostrava, Ostrava, Czech Republic.
| | - Z Slobodova
- Department of Clinical and Molecular Pathology, Palacký University Olomouc and University Hospital Olomouc, Olomouc, Czech Republic.
| | - E Fidler
- Department of Orthopedics, Palacký University Olomouc and University Hospital Olomouc, Olomouc, Czech Republic.
| | - B Shrestha
- Department of Immunology, Faculty of Medicine and Dentistry, Palacký University Olomouc and University Hospital Olomouc, Olomouc, Czech Republic.
| | - E Kriegova
- Department of Immunology, Faculty of Medicine and Dentistry, Palacký University Olomouc and University Hospital Olomouc, Olomouc, Czech Republic.
| |
Collapse
|
185
|
Ramezanpour S, Kanthawang T, Lynch J, McCulloch CE, Nevitt MC, Link TM, Joseph GB. Impact of Sustained Synovitis on Knee Joint Structural Degeneration: 4-Year MRI Data from the Osteoarthritis Initiative. J Magn Reson Imaging 2023; 57:153-164. [PMID: 35561016 PMCID: PMC9653513 DOI: 10.1002/jmri.28223] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 04/20/2022] [Accepted: 04/21/2022] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND Synovial inflammation is a risk factor for osteoarthritis (OA). But to date, there is limited information on how inflammation impacts progression of knee OA. PURPOSE To investigate how sustained synovitis, assessed with semi-quantitative magnetic resonance imaging (MRI) scores, impacts progression of knee degenerative changes over 4 years. STUDY TYPE Retrospective cohort study. SUBJECTS In 249 participants (N = 132 women [53%]), from the Osteoarthritis Initiative (OAI) two definitions for synovitis were used resulting in two groups of participants with sustained synovitis at baseline, 2-year, and 4-year follow-up (N = 80 and N = 132), and two groups without synovitis at all three time points (N = 81 and N = 47). FIELD STRENGTH/SEQUENCE 3 T intermediate-weighted (IW) turbo spin-echo (TSE) sequence and three-dimensional (3D) dual-echo steady-state (DESS) sequence. ASSESSMENT Synovitis was scored semi-quantitatively using the Anterior Cruciate Ligament Osteoarthritis Score (ACLOAS), MRI Osteoarthritis Knee Score (MOAKS), and synovial proliferation score (SPS). Two MRI-based definitions of synovitis were used: (i) score ≥2 based on cumulative score of MOAKS and ACLOAS, and (ii) score ≥3 based on the cumulative score of ACLOAS, MOAKS, and SPS. Changes in structural abnormalities from baseline to year 4 measured using the whole-organ MRI score (WORMS) were defined as outcomes. STATISTICAL TESTS Linear regression models were used to compare the differences in longitudinal changes in WORMS scores between participants with and without sustained synovitis for each definition of sustained synovitis. A P-value of <0.05 was considered statistically significant. RESULTS Significantly higher rates of progression were found in participants with synovitis for patellar (Beta coeff. = 0.29) and medial tibial cartilage abnormalities (Beta coeff. = 0.29) for definition (i). For definition (ii), patellar (Beta coeff. = 0.36) and medial femoral cartilage (Beta coeff. = 0.30) abnormalities demonstrated significant differences. DATA CONCLUSION Greater progression of structural degenerative disease was observed in individuals with sustained synovitis compared to those without sustained synovitis, suggesting that sustained synovitis is associated with progressive OA. LEVEL OF EVIDENCE 3 TECHNICAL EFFICACY: Stage 2.
Collapse
Affiliation(s)
- Sara Ramezanpour
- Department of Radiology and Biomedical Imaging, University of California, San Francisco
| | - Thanat Kanthawang
- Department of Radiology and Biomedical Imaging, University of California, San Francisco
- Department of Radiology, Faculty of Medicine, Chiang Mai University, Thailand
| | - John Lynch
- Department of Epidemiology and Biostatistics, University of California, San Francisco
| | - Charles E. McCulloch
- Department of Epidemiology and Biostatistics, University of California, San Francisco
| | - Michael C. Nevitt
- Department of Epidemiology and Biostatistics, University of California, San Francisco
| | - Thomas M. Link
- Department of Radiology and Biomedical Imaging, University of California, San Francisco
| | - Gabby B. Joseph
- Department of Radiology and Biomedical Imaging, University of California, San Francisco
| |
Collapse
|
186
|
Hu L, Luo D, Zhang H, He L. Nardosinone suppresses monoiodoacetate-induced osteoarthritis in rats: The key role of the miR-218-5p/NUMB axis. Chem Biol Drug Des 2023; 101:120-130. [PMID: 35962465 DOI: 10.1111/cbdd.14127] [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: 05/09/2022] [Revised: 07/15/2022] [Accepted: 07/30/2022] [Indexed: 12/15/2022]
Abstract
Nardosinone is a bioactive compound with a sesquiterpenoid structure isolated from Nardostachys jatamansi. The compound has shown treatment effects against skeletal disorders. In the current study, the effects of nardosinone on osteoarthritis (OA) were first assessed and the mechanism underlying the effects was explored by detecting changes in the miR-218-5p/NUMB axis. The miR, as a potential target mediating the effects of nardosinone on OA, was first determined with microarray and RT-qPCR detections. Then, OA symptoms were induced in rats using monoiodoacetate (MIA) and treated with nardosinone. The anti-OA effects of nardosinone were assessed via the detection of the histological structure and inflammation. The role of miR-218-5p was delineated by modulating its levels in OA-affected rats. Based on the results of microarray and RT-qPCR detections, miR-218-5p was selected as the therapeutic target for nardosinone. The induction of OA resulted in tissue destruction and the production of cytokines in rat joint tissues, which was associated with the up-regulation of miR-218-5p and the downregulation of NUMB. For OA-affected rats treated with nardosinone, the joint structure was improved and the inflammatory response was suppressed, along with the restored expression levels of miR-218-5p and NUMB. The re-induced level of miR-218-5p compromised the anti-OA effects of nardosinone, indicating that the inhibition of the miR played an indispensable role in the anti-OA function of nardosinone. Collectively, the findings of our study demonstrated that nardosinone exerts treatment effects against OA by modulating the miR-218-5p/NUMB axis. Future studies will provide more detailed information on the interaction between nardosinone and miR in the attenuation of OA.
Collapse
Affiliation(s)
- Linyong Hu
- Department of Orthopedics, Ganzhou Municipal Hospital, Ganzhou, Jiangxi, China
| | - Dejun Luo
- Department of Orthopedics, The People's Hospital of Jianyang City, Jianyang, Sichuan, China
| | - Hong Zhang
- Department of Orthopedics, Ganzhou Municipal Hospital, Ganzhou, Jiangxi, China
| | - Ling He
- Department of Orthopaedic, The People's Hospital of Dazu, Chongqing, China
| |
Collapse
|
187
|
Golmohammadi M, Kheirouri S, Ebrahimzadeh Attari V, Moludi J, Sulistyowati R, Nachvak SM, Mostafaei R, Mansordehghan M. Is there any association between dietary inflammatory index and quality of life? A systematic review. Front Nutr 2022; 9:1067468. [PMID: 36618692 PMCID: PMC9815464 DOI: 10.3389/fnut.2022.1067468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 11/23/2022] [Indexed: 12/24/2022] Open
Abstract
Background The inflammatory potential of unhealthy diets can lead to the development of chronic diseases and also exacerbating their complications. Therefore, the present systematic review aimed to evaluate the association of dietary inflammatory index (DII) and quality of life (QOL) in human subjects. Methods A systematic search was conducted in PubMed, Web of Science, and Scopus databases, using the combination of all search terms related to DII and QOL until May 2022. All eligible human studies published in English were included. Results Three hundred twenty-seven studies were obtained from the first systematic search of the databases although, only eight studies were eligible for the evaluation. Seven studies reported that there was a significant reverse association between DII scores and overall QOL and/or its subscales in different populations including patients with asthma, osteoarthritis, hemodialysis patients, multiple sclerosis, obese women, and also in healthy subjects. While, one study on postmenopausal women found no evidence of this association. Conclusion This systematic review demonstrated that an anti-inflammatory diet might be associated with better QOL. However, future well-designed clinical trials can provide better conclusions especially regarding the quantifying of this relationship.
Collapse
Affiliation(s)
- Mona Golmohammadi
- Department of Nutritional Sciences, School of Nutritional Sciences and Food Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Sorayya Kheirouri
- Department of Community Medicine, School of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Jalal Moludi
- Department of Nutritional Sciences, School of Nutritional Sciences and Food Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | | | - Seyed Mostafa Nachvak
- Department of Nutritional Sciences, School of Nutritional Sciences and Food Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Roghayeh Mostafaei
- Department of Nutritional Sciences, School of Nutritional Sciences and Food Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Maryam Mansordehghan
- Department of Nutritional Sciences, School of Nutritional Sciences and Food Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
188
|
Synovial Fluid in Knee Osteoarthritis Extends Proinflammatory Niche for Macrophage Polarization. Cells 2022; 11:cells11244115. [PMID: 36552878 PMCID: PMC9776803 DOI: 10.3390/cells11244115] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 12/11/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
Macrophage polarization is a steering factor of osteoarthritis (OA) progression. Synovial fluid (SF) obtained from OA patients with different Kellgren-Lawrence grades (KL grades) holds several proinflammatory factors and was hypothesized to induce macrophage differentiation and polarization by providing the needed microenvironment. U937 cells and peripheral-blood-mononuclear-cell-derived monocytes (PBMC-derived CD14+ cells) were induced with SFs of progressive KL grades for 48 h, and the status of the differentiated cells was evaluated by cell surface markers representing M1 and M2 macrophage phenotypes. Functional viability assessment of the differentiated cells was performed by cytokine estimation. The fraction of macrophages and their phenotypes were estimated by immunophenotyping of SF-isolated cells of different KL grades. A grade-wise proteome analysis of SFs was performed in search of the factors which are influential in macrophage differentiation and polarization. In the assay on U937 cells, induction with SF of KL grade III and IV showed a significant increase in M1 type (CD86+). The percentage of M2 phenotype (CD163+) was significantly higher after the induction with SF of KL grade II. A Significantly higher M1/M2 ratio was estimated in the cells induced with KL grade III and IV. The cell differentiation pattern in the assay on PBMC-derived CD14+ cells showed a grade-wise decline in both M1 (CD11C+, CD86+) and M2 phenotype (CD163+). Cytokine estimation specific to M1 (TNF-α, IL-6, IL-1β, IFN-γ) and M2 (IL-4 and IL-10) macrophages corelated with the differentiation pattern in the U937 cell assay, while it did not reveal any significant changes in the PBMC-derived CD14+ cells assay. SF cells' immunophenotyping showed the highest percentage of CD14+ macrophages in KL grade II; CD86+ and CD163+ cells were minimal in all KL grades' SFs. The proteome analysis revealed significantly expressed MIF, CAPG/MCP, osteopontin, and RAS-related RAB proteins in KL grade III and IV samples, which are linked with macrophages' movement, polarization, and migration-behavior. In conclusion, this study demonstrated that SF in OA joints acts as a niche and facilitates M1 phenotype polarization by providing a proinflammatory microenvironment.
Collapse
|
189
|
Gushcha S, Nasibullin B, Nikolaieva G, Plakida A. External use of radon and sulfide mineral waters in the treat-ment of experimental arthrosis. BALNEO AND PRM RESEARCH JOURNAL 2022. [DOI: 10.12680/balneo.2022.528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Abstract: The article presents the results of studies of the effect of radon and sulfide min-eral waters (MW) on Wistar rats with experimental arthrosis. materials and methods. Rats were randomized into 4 groups. Group 1 consisted of intact rats (control group). In the remaining three groups, a model of knee arthrosis was reproduced using dexamethasone injections. Group 2 consisted of rats with untreated pathology. Group 3 consisted of rats using radon MW procedures, and group 4 consisted of rats using sulfide MW. Results. Morphological studies have determined that using radon and sulfide MW has a curative effect on the structural and functional organization of the joint and cartilage - the manifes-tations of inflammation in the knee joints are significantly reduced, dystrophic manifesta-tions disappear, and reparative processes in cartilage improve. There are signs of an im-provement in the state of metabolic processes in the body of rats: the balance in the lipid peroxidation and antioxidant systems is restored, and the indicators of the processes of energy supply of transmembrane transport (according to the activity of magnesium-dependent Na+/K+-ATP-ase and Ca2+-ATP-ase) and protein exchange.Conclusion. Based on the data obtained, it was concluded that both applied MWs have a unidirectional, but somewhat different in strength, curative effect on the course of experimental arthrosis, which is due to different mechanisms of the biological action of radon and hydrogen sul-fide.
Keywords: experimental arthrosis; morphological and metabolic indicators; radon; hydrogen sulfide; mineral water.
Collapse
Affiliation(s)
- Sergey Gushcha
- State Institution «Ukrainian Research Institute of Medical Rehabilitation and Resort Therapy of the Ministry of Health of Ukraine», Odesa, Ukraine
| | - Boris Nasibullin
- State Institution «Ukrainian Research Institute of Medical Rehabilitation and Resort Therapy of the Ministry of Health of Ukraine», Odesa, Ukraine
| | - Ganna Nikolaieva
- State Establishment «The Institute of Stomatology and Maxillo-Facial Surgery National Academy of Medical Science of Ukraine», Odesa, Ukraine
| | - Alexander Plakida
- State Institution «Ukrainian Research Institute of Medical Rehabilitation and Resort Therapy of the Ministry of Health of Ukraine», Odesa, Ukraine
| |
Collapse
|
190
|
Dietary inflammation index and osteoarthritis in the elderly: is there a mediating role of physical activity? Br J Nutr 2022; 128:2258-2266. [PMID: 35067232 PMCID: PMC9661367 DOI: 10.1017/s0007114522000265] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
We examined whether physical activity (PA) explains the association between dietary inflammatory potential and osteoarthritis (OA) in the elderly. A total of 1249 elderly people (≥65 years) were eligible for this study from the National Health and Nutrition Examination Survey (NHANES) from 2011 to 2016. The semi-quantitative Food Frequency Questionnaire (FFQ) and Global PA Questionnaire (GPAQ) were used to evaluate the diet and PA of the elderly, respectively. The multivariable logistic regression model estimated the odds ratio (OR) and 95% confidence interval (CI) between Energy-adjusted Dietary Inflammatory Index (E-DII) and OA. The interaction of E-DII and PA on depressive events was tested, and the mediation analysis of PA was performed. The average E-DII in this study was +0.68 (SE 0.08), and the score ranges from -5.32 (most anti-inflammatory) to +4.26 (most pro-inflammatory). In comparison with the first quartile, the elderly from the second quartile (OR: 1.16 [95% CI: 1.06, 1.68]) to the fourth quartile (OR: 1.64 [95% CI: 1.13, 2.37]) had a higher risk of OA before adjustment for PA. An interaction was observed between E-DII and PA in terms of the risk of OA (PInteraction < 0.001). The whole related part was mediated by PA (20.08%). Our findings indicated that the higher pro-inflammatory potential of diet was associated with a higher risk of OA, and low PA was an important part of the mediating factor in the relationship between systemic low-grade dietary inflammation and the risk of OA.
Collapse
|
191
|
Gambari L, Cellamare A, Grassi F, Grigolo B, Panciera A, Ruffilli A, Faldini C, Desando G. Overview of Anti-Inflammatory and Anti-Nociceptive Effects of Polyphenols to Halt Osteoarthritis: From Preclinical Studies to New Clinical Insights. Int J Mol Sci 2022; 23:ijms232415861. [PMID: 36555503 PMCID: PMC9779856 DOI: 10.3390/ijms232415861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/10/2022] [Accepted: 12/11/2022] [Indexed: 12/15/2022] Open
Abstract
Knee osteoarthritis (OA) is one of the most multifactorial joint disorders in adults. It is characterized by degenerative and inflammatory processes that are responsible for joint destruction, pain and stiffness. Despite therapeutic advances, the search for alternative strategies to target inflammation and pain is still very challenging. In this regard, there is a growing body of evidence for the role of several bioactive dietary molecules (BDMs) in targeting inflammation and pain, with promising clinical results. BDMs may be valuable non-pharmaceutical solutions to treat and prevent the evolution of early OA to more severe phenotypes, overcoming the side effects of anti-inflammatory drugs. Among BDMs, polyphenols (PPs) are widely studied due to their abundance in several plants, together with their benefits in halting inflammation and pain. Despite their biological relevance, there are still many questionable aspects (biosafety, bioavailability, etc.) that hinder their clinical application. This review highlights the mechanisms of action and biological targets modulated by PPs, summarizes the data on their anti-inflammatory and anti-nociceptive effects in different preclinical in vitro and in vivo models of OA and underlines the gaps in the knowledge. Furthermore, this work reports the preliminary promising results of clinical studies on OA patients treated with PPs and discusses new perspectives to accelerate the translation of PPs treatment into the clinics.
Collapse
Affiliation(s)
- Laura Gambari
- Laboratorio RAMSES, IRCCS Istituto Ortopedico Rizzoli, via di Barbiano 1/10, 40136 Bologna, Italy
| | - Antonella Cellamare
- Laboratorio RAMSES, IRCCS Istituto Ortopedico Rizzoli, via di Barbiano 1/10, 40136 Bologna, Italy
| | - Francesco Grassi
- Laboratorio RAMSES, IRCCS Istituto Ortopedico Rizzoli, via di Barbiano 1/10, 40136 Bologna, Italy
| | - Brunella Grigolo
- Laboratorio RAMSES, IRCCS Istituto Ortopedico Rizzoli, via di Barbiano 1/10, 40136 Bologna, Italy
| | - Alessandro Panciera
- 1st Orthopedic and Traumatology Clinic, IRCCS Istituto Ortopedico Rizzoli, via G.C. Pupilli 1, 40136 Bologna, Italy
| | - Alberto Ruffilli
- 1st Orthopedic and Traumatology Clinic, IRCCS Istituto Ortopedico Rizzoli, via G.C. Pupilli 1, 40136 Bologna, Italy
| | - Cesare Faldini
- 1st Orthopedic and Traumatology Clinic, IRCCS Istituto Ortopedico Rizzoli, via G.C. Pupilli 1, 40136 Bologna, Italy
| | - Giovanna Desando
- Laboratorio RAMSES, IRCCS Istituto Ortopedico Rizzoli, via di Barbiano 1/10, 40136 Bologna, Italy
- Correspondence: ; Tel.: +39-0516366803
| |
Collapse
|
192
|
Rai MF, Cai L, Tycksen ED, Keener J, Chamberlain A. RNA-Seq reveals distinct transcriptomic differences in rotator cuff tendon based on tear etiology and patient sex. J Orthop Res 2022; 40:2728-2742. [PMID: 35174534 DOI: 10.1002/jor.25296] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 01/16/2022] [Accepted: 02/03/2022] [Indexed: 02/04/2023]
Abstract
Rotator cuff tears are a common pathology in the shoulder and generally have two underlying etiologies: traumatic and degenerative. Little is known about the molecular underpinning of these etiologies. Here we queried transcript level differences in tear etiology stratified by sex in 31 patients with rotator cuff tears. Tendon tissues were isolated from females (N = 16) and males (N = 15) with traumatic (N = 16) or degenerative (N = 15) tears during arthroscopy. Differentially expressed transcripts were identified by RNA-seq and biological processes were probed computationally. Expression of some transcripts was validated by real-time quantitative polymerase chain reaction (qPCR). We identified 339 and 336 transcripts differentially expressed by tear etiology in females and males, respectively, at a fold-change greater than |2|. In females, GSTM1, MT1G, S1008A, ACSM3, DSC, FAM110C, and VNN2 were elevated in traumatic tears representing metabolic/catabolic processes, and immune response whereas CHAD, CLEC3A, IBSP, TNMD, APLNR, and CPA3 were elevated in degenerative tears representing tissue morphogenesis and developmental processes, angiogenesis, and extracellular matrix organization. In males, ELOA3B, CXCL8, ADM, TNS4, and SPOCK1 were elevated in traumatic tears representing localization of endoplasmic reticulum, chromosome organization, leukocyte/neutrophil degranulation, and protein transport whereas MYL2, TNNC1, MB, CPA3, APLNR, and CA3 were highly upregulated in degenerative tears representing muscle cell differentiation and development and angiogenesis. Numerous novel lncRNAs were identified to be differentially expressed by tear etiology in both sexes. Real-time qPCR confirmed RNA-seq data. This study improves our understanding of tendon biology based on underlying etiology (trauma or degeneration) in a sex-specific manner. These findings may help drive clinical decision-making in females and males with traumatic and degenerative shoulder injuries.
Collapse
Affiliation(s)
- Muhammad Farooq Rai
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri, USA.,Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Lei Cai
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Eric D Tycksen
- Genome Technology Access Center, McDonell Genome Institute, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Jay Keener
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Aaron Chamberlain
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
193
|
Sawvell E, Wright N, Ode G, Mercuri J. Perinatal Tissue-Derived Allografts and Stromal Cells for the Treatment of Knee Osteoarthritis: A Review of Preclinical and Clinical Evidence. Cartilage 2022; 13:184-199. [PMID: 36398763 PMCID: PMC9924983 DOI: 10.1177/19476035221137725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE The use of perinatal-derived tissues and mesenchymal stromal cells (MSCs) as alternative treatment options to corticosteroid and hyaluronic acid injections has been gaining popularity. However, their ability to attenuate osteoarthritic (OA) symptoms while also slowing the progression of the disease remains controversial. Thus, the objective of this article is to summarize the results from both preclinical and clinical studies evaluating the efficacy of perinatal-derived tissue allografts and MSCs for the treatment of OA. DESIGN A comprehensive literature search was conducted on databases including Pubmed, ScienceDirect, and Google Scholar beginning in March 2020 for both preclinical and clinical studies evaluating perinatal-derived tissues and MSCs in OA. Eighteen studies met the inclusion criteria and were used for this review. RESULTS Both animal models and early human clinical trials demonstrated that perinatal tissues could reduce joint inflammation and pain as well as improve range of motion and function in OA. Perinatal tissue-derived MSCs in animal studies have shown the potential to support chondrocyte proliferation while also decreasing inflammatory gene and protein expression. Limited clinical results suggest perinatal tissue-derived MSC sources may also be a viable alternative or adjunct to hyaluronic acid in reducing pain and symptoms in an arthritic joint. CONCLUSIONS Perinatal tissue-derived allografts and MSCs have promise as potential therapeutics for mitigating OA progression. However, further research is warranted to fully define the therapeutic mechanism(s) of action and safety of these biological therapies.
Collapse
Affiliation(s)
- Emily Sawvell
- Laboratory of Orthopaedic Tissue
Regeneration & Orthobiologics, Department of Bioengineering, Clemson University,
Clemson, SC, USA,Frank H. Stelling and C. Dayton Riddle
Orthopaedic Education and Research Laboratory, Clemson University Biomedical
Engineering Innovation Campus, Greenville, SC, USA
| | - Noah Wright
- Laboratory of Orthopaedic Tissue
Regeneration & Orthobiologics, Department of Bioengineering, Clemson University,
Clemson, SC, USA,Frank H. Stelling and C. Dayton Riddle
Orthopaedic Education and Research Laboratory, Clemson University Biomedical
Engineering Innovation Campus, Greenville, SC, USA
| | - Gabriella Ode
- Department of Orthopaedic Surgery,
Prisma Health–Upstate, Greenville, SC, USA
| | - Jeremy Mercuri
- Laboratory of Orthopaedic Tissue
Regeneration & Orthobiologics, Department of Bioengineering, Clemson University,
Clemson, SC, USA,Frank H. Stelling and C. Dayton Riddle
Orthopaedic Education and Research Laboratory, Clemson University Biomedical
Engineering Innovation Campus, Greenville, SC, USA,Jeremy Mercuri, Laboratory of Orthopaedic
Tissue Regeneration & Orthobiologics, Department of Bioengineering, Clemson
University, 401-5 Rhodes Engineering Research Center, Clemson, SC 29634, USA.
| |
Collapse
|
194
|
Guowei G, Yuzhong Z, Xuan Z, Zhi D, Juanhui D, Jing W, Peikui Y, Xiangzhi L, Zhen W. Zhuanggu Guanjie herbal formula mitigates osteoarthritis via the NF-κB transduction mechanism. Front Pharmacol 2022; 13:896397. [PMID: 36532734 PMCID: PMC9751418 DOI: 10.3389/fphar.2022.896397] [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: 03/15/2022] [Accepted: 11/15/2022] [Indexed: 12/05/2022] Open
Abstract
The Zhuanggu Guanjie herbal formula has been a famous Chinese prescription for treating bone diseases since time immemorial. The anti-osteoarthritis (OA) properties of this botanical prescription are well documented in the Chinese Pharmacopoeia. However, the detailed mechanisms behind the phenomenon have not been elucidated. Hence, we aimed to investigate the anti-OA efficacy of the Zhuanggu Guanjie herbal formula and its underlying mechanism. The anti-OA properties of Zhuanggu Guanjie capsule (ZGC) were determined by the cytokine contents and inflammatory-related proteins, which were measured by RT-PCR, flow cytometry, Western blot, and laser confocal assay in ATDC5 cells. The levels of interleukin-6, tumor necrosis factor-α, inducible nitric oxide synthase, cyclooxygenase-2, and prostaglandin synthesis E2 have been markedly reduced after being treated with ZGC for 48 h in a dose-dependent manner. Furthermore, ZGC prevented the translocation of NF-κB from the cytosol to the nucleus. On the other hand, we used the mono-iodoacetate (MIA)-induced OA model to confirm the in vivo efficacies of this herbal formula. Oral administration of ZGC attenuated MIA-induced OA damage through changes in histopathological and knee joint volumes. The serum matrix metalloproteinase-13 contents in the ZGC treatment group declined as compared to those in the MIA model group. Through our in vitro and in vivo studies, we confirmed the anti-OA efficacy of ZGC and uncovered its detailed mechanism, and this treatment shed light on OA pathophysiology.
Collapse
Affiliation(s)
- Gong Guowei
- Department of Bioengineering, Zunyi Medical University, Zhuhai Campus, Zhuhai, China,Guangdong Provincial Key Laboratory of Functional Substances in Medicinal Edible Resources and Healthcare Products, Hanshan Normal University, Chaozhou, China,*Correspondence: Gong Guowei, ; Zheng Yuzhong,
| | - Zheng Yuzhong
- Guangdong Provincial Key Laboratory of Functional Substances in Medicinal Edible Resources and Healthcare Products, Hanshan Normal University, Chaozhou, China,*Correspondence: Gong Guowei, ; Zheng Yuzhong,
| | - Zhou Xuan
- Department of Bioengineering, Zunyi Medical University, Zhuhai Campus, Zhuhai, China
| | - Dai Zhi
- China Resources Sanjiu Medical & Pharmaceutical Co., Ltd., Shenzhen, China
| | - Duan Juanhui
- China Resources Sanjiu Medical & Pharmaceutical Co., Ltd., Shenzhen, China
| | - Wang Jing
- China Resources Sanjiu Medical & Pharmaceutical Co., Ltd., Shenzhen, China
| | - Yang Peikui
- Department of Medical Laboratory, Chaozhou People’s Hospital, Chaozhou, China
| | - Liu Xiangzhi
- Department of Medical Laboratory, Chaozhou People’s Hospital, Chaozhou, China
| | - Wen Zhen
- College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen, China
| |
Collapse
|
195
|
Guo Y, Li R, Dang X. S100A10 regulates tumor necrosis factor alpha-induced apoptosis in chondrocytes via the reactive oxygen species/nuclear factor-kappa B pathway. Biotechnol Appl Biochem 2022; 69:2284-2295. [PMID: 34787893 DOI: 10.1002/bab.2285] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 11/11/2021] [Indexed: 01/06/2023]
Abstract
Aberrant chondrocyte apoptosis and inflammation are the most critical causes of osteoarthritis (OA) development. This study was designed to demonstrate the relationship between S100A10 and OA. In this study, S100A10 was overexpressed or silenced in rat chondrocytes. Cell viability, apoptosis, reactive oxidative species (ROS), and calcium ion detection were assessed using Cell Counting Kit-8 assay and flow cytometry. The levels of key oxidation-related enzymes and tumor necrosis factor-alpha (TNF-α) were quantified using enzyme-linked immunosorbent assay, quantitative polymerase chain reaction, and Western blotting. S100A10 was highly expressed in patients with OA and positively correlated with TNF-α level. Knockdown of S100A10 effectively counteracted TNF-α-induced ROS level, apoptosis, and calcium level and associated with decreased inflammation-related metalloproteinase 1 (MMP1), MMP13, and nuclear necrosis factor-kappa B (NF-κB)-p65 and increased survivin and cytoplasmic NF-κB-p65. Overexpression of S100A10 had an effect similar to TNF-α, which was significantly counteracted by pyrrolidine dithiocarbamate, an NF-κB inhibitor, or verapamil, a calcium-channel blocker. S100A10 contributed to chondrocyte apoptosis through the ROS/NF-κB pathway. This study has established the relationship between S100A10 and the NF-κB pathway, thus providing novel perspectives for exploring S100A10 functions.
Collapse
Affiliation(s)
- Yanjie Guo
- Department of Orthopaedics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shanxi Province, China
| | - Ruofei Li
- Department of Orthopaedics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shanxi Province, China
| | - Xiaoqian Dang
- Department of Orthopaedics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shanxi Province, China
| |
Collapse
|
196
|
Gui T, Wei Y, Luo L, Li J, Zhong L, Yao L, Beier F, Nelson CL, Tsourkas A, Liu XS, Enomoto-Iwamoto M, Yu F, Cheng Z, Qin L. Activating EGFR Signaling Attenuates Osteoarthritis Development Following Loading Injury in Mice. J Bone Miner Res 2022; 37:2498-2511. [PMID: 36178273 PMCID: PMC10183199 DOI: 10.1002/jbmr.4717] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 09/08/2022] [Accepted: 09/22/2022] [Indexed: 01/21/2023]
Abstract
Posttraumatic osteoarthritis (PTOA) results in joint pain, loss of joint function, and impaired quality of daily life in patients with limited treatment options. We previously demonstrated that epidermal growth factor receptor (EGFR) signaling is essential for maintaining chondroprogenitors during articular cartilage development and homeostasis. Here, we used a nonsurgical, loading-induced PTOA mouse model to investigate the protective action of EGFR signaling. A single bout of cyclic tibial loading at a peak force of 6 N injured cartilage at the posterior aspect of lateral femoral condyle. Similar loading at a peak force of 9 N ruptured the anterior cruciate ligament, causing additional cartilage damage at the medial compartment and ectopic cartilage formation in meniscus and synovium. Constitutively overexpression of an EGFR ligand, heparin binding EGF-like growth factor (HBEGF), in chondrocytes significantly reduced cartilage injury length, synovitis, and pain after 6 N loading and mitigated medial side cartilage damage and ectopic cartilage formation after 9 N loading. Mechanistically, overactivation of EGFR signaling protected chondrocytes from loading-induced apoptosis and loss of proliferative ability and lubricant synthesis. Overexpressing HBEGF in adult cartilage starting right before 6 N loading had similar beneficial effects. In contrast, inactivating EGFR in adult cartilage led to accelerated PTOA progression with elevated cartilage Mankin score and synovitis score and increased ectopic cartilage formation. As a therapeutic approach, we constructed a nanoparticle conjugated with the EGFR ligand TGFα. Intra-articular injections of this nanoconstruct once every 3 weeks for 12 weeks partially mitigated PTOA symptoms in cartilage and synovium after 6 N loading. Our findings demonstrate the anabolic actions of EGFR signaling in maintaining articular cartilage during PTOA development and shed light on developing a novel nanomedicine for PTOA. © 2022 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Tao Gui
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Center for Joint Surgery and Sports Medicine, the First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Yulong Wei
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lijun Luo
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
- School of Agricultural Engineering, Jiangsu University, Zhenjiang, China
| | - Jun Li
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Leilei Zhong
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Lutian Yao
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Frank Beier
- Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, Canada
| | - Charles L. Nelson
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Andrew Tsourkas
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - X. Sherry Liu
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Motomi Enomoto-Iwamoto
- Department of Orthopaedics, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Feifan Yu
- AlphaThera, LLC, Philadelphia, PA, USA
| | - Zhiliang Cheng
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Ling Qin
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
197
|
Effects of knee prosthetic surgery on overactive bladder symptoms and incontinence in women. Int Urogynecol J 2022:10.1007/s00192-022-05409-w. [DOI: 10.1007/s00192-022-05409-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 10/31/2022] [Indexed: 11/26/2022]
|
198
|
Collins DP, Elsouri KN, Demory Beckler M. Osteoarthritis: Can We Do Better? Cureus 2022; 14:e31505. [DOI: 10.7759/cureus.31505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 10/20/2022] [Indexed: 11/16/2022] Open
|
199
|
Peshkova M, Kosheleva N, Shpichka A, Radenska-Lopovok S, Telyshev D, Lychagin A, Li F, Timashev P, Liang XJ. Targeting Inflammation and Regeneration: Scaffolds, Extracellular Vesicles, and Nanotechnologies as Cell-Free Dual-Target Therapeutic Strategies. Int J Mol Sci 2022; 23:13796. [PMID: 36430272 PMCID: PMC9694395 DOI: 10.3390/ijms232213796] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/25/2022] [Accepted: 11/02/2022] [Indexed: 11/11/2022] Open
Abstract
Osteoarthritis (OA) affects over 250 million people worldwide and despite various existing treatment strategies still has no cure. It is a multifactorial disease characterized by cartilage loss and low-grade synovial inflammation. Focusing on these two targets together could be the key to developing currently missing disease-modifying OA drugs (DMOADs). This review aims to discuss the latest cell-free techniques applied in cartilage tissue regeneration, since they can provide a more controllable approach to inflammation management than the cell-based ones. Scaffolds, extracellular vesicles, and nanocarriers can be used to suppress inflammation, but they can also act as immunomodulatory agents. This is consistent with the latest tissue engineering paradigm, postulating a moderate, controllable inflammatory reaction to be beneficial for tissue remodeling and successful regeneration.
Collapse
Affiliation(s)
- Maria Peshkova
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov University, 119991 Moscow, Russia
- Institute for Regenerative Medicine, Sechenov University, 119991 Moscow, Russia
- Laboratory of Clinical Smart Nanotechnologies, Institute for Regenerative Medicine, Sechenov University, 119991 Moscow, Russia
| | - Nastasia Kosheleva
- Institute for Regenerative Medicine, Sechenov University, 119991 Moscow, Russia
- Laboratory of Clinical Smart Nanotechnologies, Institute for Regenerative Medicine, Sechenov University, 119991 Moscow, Russia
- FSBSI Institute of General Pathology and Pathophysiology, 125315 Moscow, Russia
| | - Anastasia Shpichka
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov University, 119991 Moscow, Russia
- Laboratory of Clinical Smart Nanotechnologies, Institute for Regenerative Medicine, Sechenov University, 119991 Moscow, Russia
- Chemistry Department, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Stefka Radenska-Lopovok
- Institute for Clinical Morphology and Digital Pathology, Sechenov University, 119991 Moscow, Russia
| | - Dmitry Telyshev
- Institute of Biomedical Systems, National Research University of Electronic Technology, 124498 Moscow, Russia
- Institute of Bionic Technologies and Engineering, Sechenov University, 119991 Moscow, Russia
| | - Alexey Lychagin
- Laboratory of Clinical Smart Nanotechnologies, Institute for Regenerative Medicine, Sechenov University, 119991 Moscow, Russia
- Department of Traumatology, Orthopedics and Disaster Surgery, Sechenov University, 119991 Moscow, Russia
| | - Fangzhou Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China
| | - Peter Timashev
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov University, 119991 Moscow, Russia
- Laboratory of Clinical Smart Nanotechnologies, Institute for Regenerative Medicine, Sechenov University, 119991 Moscow, Russia
- Chemistry Department, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Xing-Jie Liang
- Laboratory of Clinical Smart Nanotechnologies, Institute for Regenerative Medicine, Sechenov University, 119991 Moscow, Russia
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
200
|
Qi L, Wang J, Chen X, Ding Y, Ling B, Wang W, Xu J, Xue Z. Single-cell transcriptomics reveals variable trajectories of CSPCs in the progression of osteoarthritis. Heliyon 2022; 8:e11148. [PMID: 36339749 PMCID: PMC9634280 DOI: 10.1016/j.heliyon.2022.e11148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 05/05/2022] [Accepted: 10/13/2022] [Indexed: 01/15/2023] Open
Abstract
Osteoarthritis (OA) is characterised by cartilage destruction; however, there are no specific drugs available for its treatment. Cartilage-derived stem/progenitor cells (CSPCs) are multipotent cells that play an essential role in cartilage renewal and may provide critical insights into the medical needs for OA treatment. However, alterations in cell function and fate of CSPCs during OA progression have seldom been analysed, especially at the single-cell level. Additionally, it has been reported that CSPCs can migrate to the cartilage injury area, although the mechanism of migration remains elusive. Thus, understanding the changing patterns of CSPCs in the pathological process of OA is important in the effort to develop stem cell therapy for OA. Here, we downloaded single-cell transcriptomic data of patients with OA from the Gene Expression Omnibus (GEO) database and performed unbiased clustering of the cells based on gene expression patterns using the Seurat package. Using common stem cell markers and chondrogenic transcription factors, we traced CSPCs throughout all stages of OA. We further explored the dynamics of CSPCs in OA progression and validated the single-cell RNA sequencing data in vitro using qPCR, immunofluorescence, and western blotting. Specifically, we primarily explored the heterogeneity of CSPCs at the single-cell level and found that it was closely associated with OA progression. Our results indicate significantly reduced chondrogenic differentiation capacity in CSPCs during the late stage of OA, while their proliferation capacity tended to increase. We also found that genes implicated in fibrosis, cell motility, and extracellular matrix remodelling were upregulated in CSPCs during the progression of OA. Our study revealed the dynamics of stem cells in OA progression and may inform the development of stem cell therapy for OA.
Collapse
Affiliation(s)
- Lingbin Qi
- Tongji Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Jian Wang
- Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Xian Chen
- Tongji Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Yanhui Ding
- Tongji Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Bin Ling
- The Second People’s Hospital of Yunnan Province, Kunming, China
| | - Wenjun Wang
- Stem Cell and Regenerative Medicine Engineering Research Center of Hunan Province, Hunan Yuanpin Cell Technology Co. Ltd, 102 Dongwu Road, Changsha City 410100, Hunan Province, China
- Corresponding author.
| | - Jun Xu
- Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
- Corresponding author.
| | - Zhigang Xue
- Tongji Hospital, School of Medicine, Tongji University, Shanghai 200092, China
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopaedic Department of Tongji Hospital, School of Medicine, Tongji University, 200065 Shanghai, China
- Corresponding author.
| |
Collapse
|