1
|
Shah YY, Partain BD, Aldrich JL, Strinden M, Dobson J, Rinaldi-Ramos C, Allen KD. Proteomic characterization of particle-protein coronas shows differences between osteoarthritic and contralateral knees in a rat model. Connect Tissue Res 2025; 66:59-72. [PMID: 39988892 DOI: 10.1080/03008207.2025.2459242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 12/11/2024] [Accepted: 01/22/2025] [Indexed: 02/25/2025]
Abstract
OBJECTIVE When synthetic particles are injected into a biofluid, proteins nonspecifically adsorb onto the particle surface and form a protein corona. Protein coronas are known to alter how particles function in blood; however, little is known about protein corona formation in synovial fluid or how these coronas change with osteoarthritis (OA). In this study, protein coronas were characterized on particles incubated within OA-affected or healthy rat knees. DESIGN First, to evaluate particle collection techniques, magnetic polystyrene particles were placed in bovine synovial fluid and separated using either magnetics or centrifugation. In a second experiment, 12 male and 12 female Lewis rats received a simulated medial meniscal injury. At 2, 5, or 8 weeks post-surgery, operated and contralateral limbs were injected with clean magnetic particles (n = 8 per timepoint). After a 4-h incubation, animals were euthanized and particles were magnetically recovered. In both experiments, protein coronas were characterized using an Orbitrap fusion mass spectrometer. RESULTS In the first experiment, the particle separation method affected the identified proteins, likely due to centrifugation forces causing some large proteins to spin-down with the particles. In the OA model, 300-500 proteins were identified in the particle-protein coronas with 35, 59, and 13 proteins differing between the OA-affected and contralateral limbs at 2, 5, and 8 weeks, respectively. In particular, plectin, a serine (or cysteine) proteinase inhibitor, and cathepsin B were more prominent in the particle-protein coronas of OA-affected knees. CONCLUSIONS Synthetic particles nonspecifically adsorb proteins in synovial fluid, and these binding events differ with OA severity.
Collapse
Affiliation(s)
- Yash Y Shah
- Department of Materials Science and Engineering, University of Florida, Gainesville, FL, USA
| | - Brittany D Partain
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Jessica L Aldrich
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Michael Strinden
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Jon Dobson
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Carlos Rinaldi-Ramos
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
- Department of Chemical Engineering, University of Florida, Gainesville, FL, USA
| | - Kyle D Allen
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| |
Collapse
|
2
|
Elashry MI, Speer J, De Marco I, Klymiuk MC, Wenisch S, Arnhold S. Extracellular Vesicles: A Novel Diagnostic Tool and Potential Therapeutic Approach for Equine Osteoarthritis. Curr Issues Mol Biol 2024; 46:13078-13104. [PMID: 39590374 PMCID: PMC11593097 DOI: 10.3390/cimb46110780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 11/12/2024] [Accepted: 11/15/2024] [Indexed: 11/28/2024] Open
Abstract
Osteoarthritis (OA) is a chronic progressive degenerative joint disease that affects a significant portion of the equine population and humans worldwide. Current treatment options for equine OA are limited and incompletely curative. Horses provide an excellent large-animal model for studying human OA. Recent advances in the field of regenerative medicine have led to the exploration of extracellular vesicles (EVs)-cargoes of microRNA, proteins, lipids, and nucleic acids-to evaluate their diagnostic value in terms of disease progression and severity, as well as a potential cell-free therapeutic approach for equine OA. EVs transmit molecular signals that influence various biological processes, including the inflammatory response, apoptosis, proliferation, and cell communication. In the present review, we summarize recent advances in the isolation and identification of EVs, the use of their biologically active components as biomarkers, and the distribution of the gap junction protein connexin 43. Moreover, we highlight the role of mesenchymal stem cell-derived EVs as a potential therapeutic tool for equine musculoskeletal disorders. This review aims to provide a comprehensive overview of the current understanding of the pathogenesis, diagnosis, and treatment strategies for OA. In particular, the roles of EVs as biomarkers in synovial fluid, chondrocytes, and plasma for the early detection of equine OA are discussed.
Collapse
Affiliation(s)
- Mohamed I. Elashry
- Institute of Veterinary Anatomy, Histology and Embryology, Justus-Liebig-University of Giessen, 35392 Giessen, Germany; (J.S.); (M.C.K.); (S.A.)
| | - Julia Speer
- Institute of Veterinary Anatomy, Histology and Embryology, Justus-Liebig-University of Giessen, 35392 Giessen, Germany; (J.S.); (M.C.K.); (S.A.)
| | - Isabelle De Marco
- Clinic of Small Animals, c/o Institute of Veterinary Anatomy, Histology and Embryology, Justus-Liebig-University of Giessen, 35392 Giessen, Germany; (I.D.M.); (S.W.)
| | - Michele C. Klymiuk
- Institute of Veterinary Anatomy, Histology and Embryology, Justus-Liebig-University of Giessen, 35392 Giessen, Germany; (J.S.); (M.C.K.); (S.A.)
| | - Sabine Wenisch
- Clinic of Small Animals, c/o Institute of Veterinary Anatomy, Histology and Embryology, Justus-Liebig-University of Giessen, 35392 Giessen, Germany; (I.D.M.); (S.W.)
| | - Stefan Arnhold
- Institute of Veterinary Anatomy, Histology and Embryology, Justus-Liebig-University of Giessen, 35392 Giessen, Germany; (J.S.); (M.C.K.); (S.A.)
| |
Collapse
|
3
|
Xu Y, Hu X, Cai J, Li Y, Zou Y, Wang Y, Xie C, Xu S, Wang Y, Zheng Y, Mahamat DA, Xu Y, Wang X, Li X, Liu A, Chen D, Zhu L, Guo J. Atractylenolide-III alleviates osteoarthritis and chondrocyte senescence by targeting NF-κB signaling. Phytother Res 2023; 37:4607-4620. [PMID: 37380363 DOI: 10.1002/ptr.7929] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 06/01/2023] [Accepted: 06/10/2023] [Indexed: 06/30/2023]
Abstract
Atractylenolide-III (AT-III) is well known as its role in antioxidant and anti-inflammatory. Present study was aimed to figure out its effects on osteoarthritis and potential mechanisms. Rat model, human osteoarthritis cartilage explants as well as rat/human chondrocyte cultures were prepared to test AT-III's effects on osteoarthritis progression and chondrocyte senescence. Potential targeted molecules of AT-III were predicted using network pharmacology and molecular docking, assessed by Western blotting and then verified with rescue experiments. AT-III treatment alleviated osteoarthritis severity (shown by OARSI grading score and micro-CT) and chondrocyte senescence (indexed by levels of SA-β-gal, P16, P53, MMP13, ROS and ratio of healthy/collapsed mitochondrial membrane potentials). Network pharmacology and molecular docking suggested that AT-III might play role through NF-κB pathway. Further experiments revealed that AT-III reduced phosphorylation of IKKα/β, IκBα and P65 in NF-κB pathway. As well as nuclear translocation of p65. Both in vivo and in vitro experiments indicated that AT-III's effects on osteoarthritis and anti-senescence were reversed by an NF-κB agonist. AT-III could alleviate osteoarthritis by inhibiting chondrocyte senescence through NF-κB pathway, which indicated that AT-III is a prospective drug for osteoarthritis treatment.
Collapse
Affiliation(s)
- Yizhou Xu
- Department of Histology and Embryology, Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, National Demonstration Center for Experimental Education, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
- Department of Spinal Surgery, Orthopedic Medical Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaofang Hu
- Department of Histology and Embryology, Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, National Demonstration Center for Experimental Education, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Jiale Cai
- Department of Histology and Embryology, Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, National Demonstration Center for Experimental Education, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Yunlun Li
- Department of Histology and Embryology, Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, National Demonstration Center for Experimental Education, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Ying Zou
- Department of Histology and Embryology, Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, National Demonstration Center for Experimental Education, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Yihan Wang
- Department of Spinal Surgery, Orthopedic Medical Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Changnan Xie
- Department of Spinal Surgery, Orthopedic Medical Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Shuyi Xu
- Department of Histology and Embryology, Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, National Demonstration Center for Experimental Education, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Yanqing Wang
- Research Center of Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yuli Zheng
- Department of Histology and Embryology, Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, National Demonstration Center for Experimental Education, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Djibril Adam Mahamat
- Department of Histology and Embryology, Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, National Demonstration Center for Experimental Education, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Yuantao Xu
- Department of Histology and Embryology, Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, National Demonstration Center for Experimental Education, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
- Department of Spinal Surgery, Orthopedic Medical Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xianghai Wang
- Department of Histology and Embryology, Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, National Demonstration Center for Experimental Education, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Xican Li
- School of Chinese Herbal Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Aijun Liu
- Research Center of Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Dongfeng Chen
- Research Center of Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lixin Zhu
- Department of Spinal Surgery, Orthopedic Medical Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jiasong Guo
- Department of Histology and Embryology, Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, National Demonstration Center for Experimental Education, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
- Department of Spinal Surgery, Orthopedic Medical Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China
| |
Collapse
|
4
|
Wakale S, Wu X, Sonar Y, Sun A, Fan X, Crawford R, Prasadam I. How are Aging and Osteoarthritis Related? Aging Dis 2023; 14:592-604. [PMID: 37191424 PMCID: PMC10187698 DOI: 10.14336/ad.2022.0831] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 08/31/2022] [Indexed: 05/17/2023] Open
Abstract
Osteoarthritis is the most prevalent degenerative joint disease and one of the leading causes of physical impairment in the world's aging population. The human lifespan has significantly increased as a result of scientific and technological advancements. According to estimates, the world's elderly population will increase by 20% by 2050. Aging and age-related changes are discussed in this review in relation to the development of OA. We specifically discussed the cellular and molecular changes that occur in the chondrocytes during aging and how these changes may make synovial joints more susceptible to OA development. These changes include chondrocyte senescence, mitochondrial dysfunction, epigenetic modifications, and decreased growth factor response. The age-associated changes occur not only in the chondrocytes but also in the matrix, subchondral bone, and synovium. This review aims to provide an overview of the interplay between chondrocytes and matrix and how age-related changes affect the normal function of cartilage and contribute to OA development. Understanding the alterations that affect the function of chondrocytes will emerge new possibilities for prospective therapeutic options for the treatment of OA.
Collapse
Affiliation(s)
- Shital Wakale
- Centre for Biomedical Technologies, Faculty of Engineering, Queensland University of Technology, Brisbane, Queensland, Australia.
| | - Xiaoxin Wu
- Centre for Biomedical Technologies, Faculty of Engineering, Queensland University of Technology, Brisbane, Queensland, Australia.
| | - Yogita Sonar
- Centre for Biomedical Technologies, Faculty of Engineering, Queensland University of Technology, Brisbane, Queensland, Australia.
| | - Antonia Sun
- Centre for Biomedical Technologies, Faculty of Engineering, Queensland University of Technology, Brisbane, Queensland, Australia.
| | - Xiwei Fan
- Centre for Biomedical Technologies, Faculty of Engineering, Queensland University of Technology, Brisbane, Queensland, Australia.
| | - Ross Crawford
- Centre for Biomedical Technologies, Faculty of Engineering, Queensland University of Technology, Brisbane, Queensland, Australia.
- Orthopaedic Department, The Prince Charles Hospital, Brisbane, Queensland, Australia.
| | - Indira Prasadam
- Centre for Biomedical Technologies, Faculty of Engineering, Queensland University of Technology, Brisbane, Queensland, Australia.
| |
Collapse
|
5
|
Kovács P, Pushparaj PN, Takács R, Mobasheri A, Matta C. The clusterin connectome: Emerging players in chondrocyte biology and putative exploratory biomarkers of osteoarthritis. Front Immunol 2023; 14:1103097. [PMID: 37033956 PMCID: PMC10081159 DOI: 10.3389/fimmu.2023.1103097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Accepted: 02/23/2023] [Indexed: 03/17/2023] Open
Abstract
IntroductionClusterin is amoonlighting protein that hasmany functions. It is amultifunctional Q6 holdase chaperone glycoprotein that is present intracellularly and extracellularly in almost all bodily fluids. Clusterin is involved in lipid transport, cell differentiation, regulation of apoptosis, and clearance of cellular debris, and plays a protective role in ensuring cellular survival. However, the possible involvement of clusterin in arthritic disease remains unclear. Given the significant potential of clusterin as a biomarker of osteoarthritis (OA), a more detailed analysis of its complex network in an inflammatory environment, specifically in the context of OA, is required. Based on the molecular network of clusterin, this study aimed to identify interacting partners that could be developed into biomarker panels for OA.MethodsThe STRING database and Cytoscape were used to map and visualize the clusterin connectome. The Qiagen Ingenuity Pathway Analysis (IPA) software was used to analyze and study clusterinassociated signaling networks in OA. We also analyzed transcription factors known to modulate clusterin expression, which may be altered in OA.ResultsThe top hits in the clusterin network were intracellular chaperones, aggregate-forming proteins, apoptosis regulators and complement proteins. Using a text-mining approach in Cytoscape, we identified additional interacting partners, including serum proteins, apolipoproteins, and heat shock proteins.DiscussionBased on known interactions with proteins, we predicted potential novel components of the clusterin connectome in OA, including selenoprotein R, semaphorins, and meprins, which may be important for designing new prognostic or diagnostic biomarker panels.
Collapse
Affiliation(s)
- Patrik Kovács
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Peter Natesan Pushparaj
- Center of Excellence in Genomic Medicine Research (CEGMR), Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Center for Transdisciplinary Research, Department of Pharmacology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Chennai, India
| | - Roland Takács
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Ali Mobasheri
- FibroHealth Interdisciplinary Research Programme, Fibrobesity Cluster, Research Unit of Health Sciences and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania
- Department of Joint Surgery, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
- World Health Organization Collaborating Center for Public Health Aspects of Musculoskeletal Health and Aging, Université de Liège, Liège, Belgium
- *Correspondence: Csaba Matta, ; Ali Mobasheri,
| | - Csaba Matta
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- *Correspondence: Csaba Matta, ; Ali Mobasheri,
| |
Collapse
|
6
|
Liu Y, Zhang Z, Li T, Xu H, Zhang H. Senescence in osteoarthritis: from mechanism to potential treatment. Arthritis Res Ther 2022; 24:174. [PMID: 35869508 PMCID: PMC9306208 DOI: 10.1186/s13075-022-02859-x] [Citation(s) in RCA: 87] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Accepted: 07/05/2022] [Indexed: 12/12/2022] Open
Abstract
Osteoarthritis (OA) is an age-related cartilage degenerative disease, and chondrocyte senescence has been extensively studied in recent years. Increased numbers of senescent chondrocytes are found in OA cartilage. Selective clearance of senescent chondrocytes in a post-traumatic osteoarthritis (PTOA) mouse model ameliorated OA development, while intraarticular injection of senescent cells induced mouse OA. However, the means and extent to which senescence affects OA remain unclear. Here, we review the latent mechanism of senescence in OA and propose potential therapeutic methods to target OA-related senescence, with an emphasis on immunotherapies. Natural killer (NK) cells participate in the elimination of senescent cells in multiple organs. A relatively comprehensive discussion is presented in that section. Risk factors for OA are ageing, obesity, metabolic disorders and mechanical overload. Determining the relationship between known risk factors and senescence will help elucidate OA pathogenesis and identify optimal treatments.
Collapse
|
7
|
Discovery proteomics for the detection of putative markers for eradication of infection in an experimental model of equine septic arthritis using LC-MS/MS. J Proteomics 2022; 261:104571. [DOI: 10.1016/j.jprot.2022.104571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 02/28/2022] [Accepted: 03/16/2022] [Indexed: 11/22/2022]
|
8
|
Kalvaityte U, Matta C, Bernotiene E, Pushparaj PN, Kiapour AM, Mobasheri A. Exploring the translational potential of clusterin as a biomarker of early osteoarthritis. J Orthop Translat 2022; 32:77-84. [PMID: 34976733 PMCID: PMC8671091 DOI: 10.1016/j.jot.2021.10.001] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 10/01/2021] [Accepted: 10/03/2021] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Clusterin (CLU; also known as apolipoprotein J) is an ATP-independent holdase chaperone that prevents proteotoxicity as a consequence of protein aggregation. It is a ∼60 kDa disulfide-linked heterodimeric protein involved in the clearance of cellular debris and the regulation of apoptosis. CLU has been proposed to protect cells from cytolysis by complement components and has been implicated in Alzheimer's disease due to its ability to bind amyloid-β peptides and prevent aggregate formation in the brain. Recent studies suggest that CLU performs moonlighting functions. CLU exists in two major forms: an intracellular form and a secreted extracellular form. The intracellular form of CLU may suppress stress-induced apoptosis by forming complexes with misfolded proteins and facilitates their degradation. The secreted form of CLU functions as an extracellular chaperone that prevents protein aggregation. METHODS In this review, we discuss the published literature on the biology of CLU in cartilage, chondrocytes, and other synovial joint tissues. We also review clinical studies that have examined the potential for using this protein as a biomarker in synovial and systemic fluids of patients with rheumatoid arthritis (RA) or osteoarthritis (OA). RESULTS Since CLU functions as an extracellular chaperone, we propose that it may be involved in cytoprotective functions in osteoarticular tissues. The secreted form of CLU can be measured in synovial and systemic fluids and may have translational potential as a biomarker of early repair responses in OA. CONCLUSION There is significant potential for investigating synovial and systemic CLU as biomarkers of OA. Future translational and clinical orthopaedic studies should carefully consider the diverse roles of this protein and its involvement in other comorbidities. Therefore, future biomarker studies should not correlate circulating CLU levels exclusively to the process of OA pathogenesis and progression. Special attention should be paid to CLU levels in synovial fluid. THE TRANSLATIONAL POTENTIAL OF THIS ARTICLE There is significant potential for investigating synovial and systemic CLU as a predictive biomarker of osteoarthritis (OA) progression and response to novel treatments and interventions. Given that CLU plays diverse roles in other comorbidities such as rheumatoid arthritis (RA) and obesity, future translational and clinical orthopaedic biomarker studies should not directly correlate circulating CLU levels to the process of OA pathogenesis and progression. However, special attention should be paid to CLU levels in synovial fluid. The cytoprotective properties of CLU may support the implementation of regenerative strategies and new approaches for developing targeted therapeutics for OA.
Collapse
Key Words
- ACL, anterior cruciate ligament
- ACR, American College of Rheumatology
- ApoJ, apolipoprotein J
- Apoptosis
- CLU, clusterin
- CMC-I, carpometacarpal joint
- COMP, cartilage oligomeric matrix protein
- Clusterin (CLU)
- ECM, extracellular matrix
- ELISA, enzyme-linked immunosorbent assay
- ESCEO, The European Society for Clinical and Economic Aspects of Osteoporosis: Osteoarthritis and Musculoskeletal Diseases
- Inflammation
- OA, osteoarthritis
- OARSI, Osteoarthritis Research Society International
- Osteoarthritis (OA)
- PsA, psoriatic arthritis
- RA, rheumatoid arthritis
- Rheumatoid arthritis (RA)
- SF, synovial fluid
- TNF-α, tumor necrosis factor-α
- Translational biomarker
- hsCRP, high sensitivity C-reactive protein
- qRT-PCR, quantitative reverse transcription polymerase chain reaction
- sCLU, secreted clusterin
Collapse
Affiliation(s)
- Ursule Kalvaityte
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, LT, 08406, Vilnius, Lithuania
| | - Csaba Matta
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, Debrecen, H, 4032, Hungary
| | - Eiva Bernotiene
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, LT, 08406, Vilnius, Lithuania
| | - Peter Natesan Pushparaj
- Center of Excellence in Genomic Medicine Research (CEGMR), Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Ata M. Kiapour
- Department of Orthopedic Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA, 021115, USA
| | - Ali Mobasheri
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, LT, 08406, Vilnius, Lithuania
- Research Unit of Medical Imaging, Physics and Technology, Faculty of Medicine, University of Oulu, FI, 90014, Oulu, Finland
- Department of Orthopedics, Rheumatology and Clinical Immunology, University Medical Center Utrecht, 508, GA, Utrecht, the Netherlands
- Department of Joint Surgery, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
- World Health Organization Collaborating Center for Public Health Aspects of Musculoskeletal Health and Aging, Université de Liège, Liège, Belgium
| |
Collapse
|
9
|
Arabiyat AS, Chen H, Erndt-Marino J, Burkhard K, Scola L, Fleck A, Wan LQ, Hahn MS. Hyperosmolar Ionic Solutions Modulate Inflammatory Phenotype and sGAG Loss in a Cartilage Explant Model. Cartilage 2021; 13:713S-721S. [PMID: 32975437 PMCID: PMC8804856 DOI: 10.1177/1947603520961167] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
OBJECTIVE The objective of this study was to compare the effects of hyperosmolar sodium (Na+), lithium (Li+) and potassium (K+) on catabolic and inflammatory osteoarthritis (OA) markers and sulfated glycosaminoglycan (sGAG) loss in TNF-α-stimulated cartilage explants. METHODS Explants from bovine stifle joints were stimulated with TNF-α for 1 day to induce cartilage degradation followed by supplementation with 50 mM potassium chloride (KCl), 50 mM lithium chloride (LiCl), 50 mM sodium chloride (NaCl), or 100 nM dexamethasone for an additional 6 days. We assessed the effect of TNF-α stimulation and hyperosmolar ionic treatment on sGAG loss and expression of OA-associated proteins: ADAMTS-5, COX-2, MMP-1, MMP-13, and VEGF. RESULTS TNF-α treatment increased sGAG loss (P < 0.001) and expression of COX-2 (P = 0.018), MMP-13 (P < 0.001), and VEGF (P = 0.017) relative to unstimulated controls. Relative to activated controls, LiCl and dexamethasone treatment attenuated sGAG loss (P = 0.008 and P = 0.042, respectively) and expression of MMP-13 (P = 0.005 and P = 0.036, respectively). In contrast, KCl treatment exacerbated sGAG loss (P = 0.032) and MMP-1 protein expression (P = 0.010). NaCl treatment, however, did not alter sGAG loss or expression of OA-related proteins. Comparing LiCl and KCl treatment shows a potent reduction (P < 0.05) in catabolic and inflammatory mediators following LiCl treatment. CONCLUSION These results suggest that these ionic species elicit varying responses in TNF-α-stimulated explants. Cumulatively, these findings support additional studies of hyperosmolar ionic solutions for potential development of novel intraarticular injections targeting OA.
Collapse
Affiliation(s)
- Ahmad S. Arabiyat
- Department of Biomedical Engineering,
Rensselaer Polytechnic Institute (RPI), Troy, NY, USA
- Center for Biotechnology and
Interdisciplinary Studies, Rensselaer Polytechnic Institute (RPI), Troy, NY,
USA
| | - Hongyu Chen
- Department of Biomedical Engineering,
Rensselaer Polytechnic Institute (RPI), Troy, NY, USA
- Center for Biotechnology and
Interdisciplinary Studies, Rensselaer Polytechnic Institute (RPI), Troy, NY,
USA
| | - Josh Erndt-Marino
- Department of Biomedical Engineering,
Tufts University, Medford, MA, USA
| | - Katie Burkhard
- Department of Biomedical Engineering,
Rensselaer Polytechnic Institute (RPI), Troy, NY, USA
| | - Lisa Scola
- Department of Biomedical Engineering,
Rensselaer Polytechnic Institute (RPI), Troy, NY, USA
| | - Allison Fleck
- Department of Biomedical Engineering,
Rensselaer Polytechnic Institute (RPI), Troy, NY, USA
- Center for Biotechnology and
Interdisciplinary Studies, Rensselaer Polytechnic Institute (RPI), Troy, NY,
USA
| | - Leo Q. Wan
- Department of Biomedical Engineering,
Rensselaer Polytechnic Institute (RPI), Troy, NY, USA
- Center for Biotechnology and
Interdisciplinary Studies, Rensselaer Polytechnic Institute (RPI), Troy, NY,
USA
| | - Mariah S. Hahn
- Department of Biomedical Engineering,
Rensselaer Polytechnic Institute (RPI), Troy, NY, USA
- Center for Biotechnology and
Interdisciplinary Studies, Rensselaer Polytechnic Institute (RPI), Troy, NY,
USA
| |
Collapse
|
10
|
Herger S, Vach W, Liphardt AM, Nüesch C, Egloff C, Mündermann A. Experimental-analytical approach to assessing mechanosensitive cartilage blood marker kinetics in healthy adults: dose-response relationship and interrelationship of nine candidate markers. F1000Res 2021; 10:490. [PMID: 35284064 PMCID: PMC8907551 DOI: 10.12688/f1000research.52159.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/06/2022] [Indexed: 11/20/2022] Open
Abstract
Purpose: To determine the suitability of selected blood biomarkers of articular cartilage as mechanosensitive markers and to investigate the dose-response relationship between ambulatory load magnitude and marker kinetics in response to load. Methods: Serum samples were collected from 24 healthy volunteers before and at three time points after a 30-minute walking stress test performed on three test days. In each experimental session, one of three ambulatory loads was applied: 100% body weight (BW); 80%BW; 120%BW. Serum concentrations of COMP, MMP-3, MMP-9, ADAMTS-4, PRG-4, CPII, C2C and IL-6 were assessed using commercial enzyme-linked immunosorbent assays. A two-stage analytical approach was used to determine the suitability of a biomarker by testing the response to the stress test (criterion I) and the dose-response relationship between ambulatory load magnitude and biomarker kinetics (criterion II). Results. COMP, MMP-3 and IL-6 at all three time points after, MMP-9 at 30 and 60 minutes after, and ADAMTS-4 and CPII at immediately after the stress test showed an average response to load or an inter-individual variation in response to load of up to 25% of pre-test levels. The relation to load magnitude on average or an inter-individual variation in this relationship was up to 8% from load level to load level. There was a positive correlation for the slopes of the change-load relationship between COMP and MMP-3, and a negative correlation for the slopes between COMP, MMP-3 and IL-6 with MMP-9, and COMP with IL6. Conclusions: COMP, MMP-3, IL-6, MMP-9, and ADAMTS-4 warrant further investigation in the context of articular cartilage mechanosensitivity and its role in joint degeneration and OA. While COMP seems to be able to reflect a rapid response, MMP-3 seems to reflect a slightly longer lasting, but probably also more distinct response. MMP-3 showed also the strongest association with the magnitude of load.
Collapse
Affiliation(s)
- Simon Herger
- Department of Orthopaedics and Traumatology, University Hospital Basel, Basel, BS, 4031, Switzerland
- Department of Spine Surgery, University Hospital Basel, Basel, BS, 4031, Switzerland
- Department of Biomedical Engineering, University of Basel, Allschwil, BL, 4123, Switzerland
| | - Werner Vach
- Department of Orthopaedics and Traumatology, University Hospital Basel, Basel, BS, 4031, Switzerland
- Department of Clinical Research, University of Basel, Basel, BS, 4031, Switzerland
- Basel Academy for Quality and Research in Medicine, Basel, Switzerland
| | - Anna-Maria Liphardt
- Department of Internal Medicine 3 – Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nuremberg (FAU), Universitätsklinikum Erlangen, Erlangen, Germany
| | - Corina Nüesch
- Department of Orthopaedics and Traumatology, University Hospital Basel, Basel, BS, 4031, Switzerland
- Department of Spine Surgery, University Hospital Basel, Basel, BS, 4031, Switzerland
- Department of Biomedical Engineering, University of Basel, Allschwil, BL, 4123, Switzerland
- Department of Clinical Research, University of Basel, Basel, BS, 4031, Switzerland
| | - Christian Egloff
- Department of Orthopaedics and Traumatology, University Hospital Basel, Basel, BS, 4031, Switzerland
| | - Annegret Mündermann
- Department of Orthopaedics and Traumatology, University Hospital Basel, Basel, BS, 4031, Switzerland
- Department of Spine Surgery, University Hospital Basel, Basel, BS, 4031, Switzerland
- Department of Biomedical Engineering, University of Basel, Allschwil, BL, 4123, Switzerland
- Department of Clinical Research, University of Basel, Basel, BS, 4031, Switzerland
| |
Collapse
|
11
|
Herger S, Vach W, Liphardt AM, Nüesch C, Egloff C, Mündermann A. Experimental-analytical approach to assessing mechanosensitive cartilage blood marker kinetics in healthy adults: dose-response relationship and interrelationship of nine candidate markers. F1000Res 2021; 10:490. [PMID: 35284064 PMCID: PMC8907551 DOI: 10.12688/f1000research.52159.1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/15/2021] [Indexed: 11/23/2023] Open
Abstract
Purpose: To determine the suitability of selected blood biomarkers of articular cartilage as mechanosensitive markers and to investigate the dose-response relationship between ambulatory load magnitude and marker kinetics in response to load. Methods: Serum samples were collected from 24 healthy volunteers before and at three time points after a 30-minute walking stress test performed on three test days. In each experimental session, one of three ambulatory loads was applied: 100% body weight (BW); 80%BW; 120%BW. Serum concentrations of COMP, MMP-3, MMP-9, ADAMTS-4, PRG-4, CPII, C2C and IL-6 were assessed using commercial enzyme-linked immunosorbent assays. A two-stage analytical approach was used to determine the suitability of a biomarker by testing the response to the stress test (criterion I) and the dose-response relationship between ambulatory load magnitude and biomarker kinetics (criterion II). Results. COMP, MMP-3 and IL-6 at all three time points after, MMP-9 at 30 and 60 minutes after, and ADAMTS-4 and CPII at immediately after the stress test showed an average response to load or an inter-individual variation in response to load of up to 25% of pre-test levels. The relation to load magnitude on average or an inter-individual variation in this relationship was up to 8% from load level to load level. There was a positive correlation for the slopes of the change-load relationship between COMP and MMP-3, and a negative correlation for the slopes between COMP, MMP-3 and IL-6 with MMP-9, and COMP with IL6. Conclusions: COMP, MMP-3, IL-6, MMP-9, and ADAMTS-4 warrant further investigation in the context of articular cartilage mechanosensitivity and its role in joint degeneration and OA. While COMP seems to be able to reflect a rapid response, MMP-3 seems to reflect a slightly longer lasting, but probably also more distinct response. MMP-3 showed also the strongest association with the magnitude of load.
Collapse
Affiliation(s)
- Simon Herger
- Department of Orthopaedics and Traumatology, University Hospital Basel, Basel, BS, 4031, Switzerland
- Department of Spine Surgery, University Hospital Basel, Basel, BS, 4031, Switzerland
- Department of Biomedical Engineering, University of Basel, Allschwil, BL, 4123, Switzerland
| | - Werner Vach
- Department of Orthopaedics and Traumatology, University Hospital Basel, Basel, BS, 4031, Switzerland
- Department of Clinical Research, University of Basel, Basel, BS, 4031, Switzerland
- Basel Academy for Quality and Research in Medicine, Basel, Switzerland
| | - Anna-Maria Liphardt
- Department of Internal Medicine 3 – Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nuremberg (FAU), Universitätsklinikum Erlangen, Erlangen, Germany
| | - Corina Nüesch
- Department of Orthopaedics and Traumatology, University Hospital Basel, Basel, BS, 4031, Switzerland
- Department of Spine Surgery, University Hospital Basel, Basel, BS, 4031, Switzerland
- Department of Biomedical Engineering, University of Basel, Allschwil, BL, 4123, Switzerland
- Department of Clinical Research, University of Basel, Basel, BS, 4031, Switzerland
| | - Christian Egloff
- Department of Orthopaedics and Traumatology, University Hospital Basel, Basel, BS, 4031, Switzerland
| | - Annegret Mündermann
- Department of Orthopaedics and Traumatology, University Hospital Basel, Basel, BS, 4031, Switzerland
- Department of Spine Surgery, University Hospital Basel, Basel, BS, 4031, Switzerland
- Department of Biomedical Engineering, University of Basel, Allschwil, BL, 4123, Switzerland
- Department of Clinical Research, University of Basel, Basel, BS, 4031, Switzerland
| |
Collapse
|
12
|
Matta C, Fellows CR, Quasnichka H, Williams A, Jeremiasse B, Allaway D, Mobasheri A. Clusterin secretion is attenuated by the proinflammatory cytokines interleukin-1β and tumor necrosis factor-α in models of cartilage degradation. J Orthop Res 2021; 39:1017-1029. [PMID: 32725904 DOI: 10.1002/jor.24814] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 06/17/2020] [Accepted: 07/27/2020] [Indexed: 02/04/2023]
Abstract
The protein clusterin has been implicated in the molecular alterations that occur in articular cartilage during osteoarthritis (OA). Clusterin exists in two isoforms with opposing functions, and their roles in cartilage have not been explored. The secreted form of clusterin (sCLU) is a cytoprotective extracellular chaperone that prevents protein aggregation, enhances cell proliferation and promotes viability, whereas nuclear clusterin acts as a pro-death signal. Therefore, these two clusterin isoforms may be putative molecular markers of repair and catabolic responses in cartilage and the ratio between them may be important. In this study, we focused on sCLU and used established, pathophysiologically relevant, in vitro models to understand its role in cytokine-stimulated cartilage degradation. The secretome of equine cartilage explants, osteochondral biopsies and isolated unpassaged chondrocytes was analyzed by western blotting for released sCLU, cartilage oligomeric protein (COMP) and matrix metalloproteinases (MMP) 3 and 13, following treatment with the proinflammatory cytokines interleukin-1β (IL-1β) and tumor necrosis factor-α. Release of sulfated glycosaminoglycans (sGAG) was determined using the dimethylmethylene blue assay. Clusterin messenger RNA (mRNA) expression was quantified by quantitative real-time polymerase chain reaction. MMP-3, MMP-13, COMP, and sGAG release from explants and osteochondral biopsies was elevated with cytokine treatment, confirming cartilage degradation in these models. sCLU release was attenuated with cytokine treatment in all models, potentially limiting its cytoprotective function. Clusterin mRNA expression was down-regulated 7-days post cytokine stimulation. These observations implicate sCLU in catabolic responses of chondrocytes, but further studies are required to evaluate its role in OA and its potential as an investigative biomarker.
Collapse
Affiliation(s)
- Csaba Matta
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.,Department of Veterinary Preclinical Sciences, School of Veterinary Medicine, Faculty of Health and Medical Sciences, University of Surrey, Surrey, UK
| | - Christopher R Fellows
- Department of Veterinary Preclinical Sciences, School of Veterinary Medicine, Faculty of Health and Medical Sciences, University of Surrey, Surrey, UK
| | - Helen Quasnichka
- Department of Veterinary Preclinical Sciences, School of Veterinary Medicine, Faculty of Health and Medical Sciences, University of Surrey, Surrey, UK
| | | | - Bernadette Jeremiasse
- Departments of Rheumatology and Clinical Immunology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - David Allaway
- Biomarkers Division, WALTHAM Petcare Science Institute, Waltham-on-the-Wolds, Leicestershire, UK
| | - Ali Mobasheri
- Departments of Rheumatology and Clinical Immunology, University Medical Centre Utrecht, Utrecht, The Netherlands.,Research Unit of Medical Imaging, Physics and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland.,Department of Regenerative Medicine, State Research Institute, Centre for Innovative Medicine, Vilnius, Lithuania.,Department of Orthopedics, University Medical Centre Utrecht, Utrecht, The Netherlands.,Centre for Sport, Exercise and Osteoarthritis Versus Arthritis, Queen's Medical Centre, Nottingham, UK
| |
Collapse
|
13
|
Khella CM, Asgarian R, Horvath JM, Rolauffs B, Hart ML. An Evidence-Based Systematic Review of Human Knee Post-Traumatic Osteoarthritis (PTOA): Timeline of Clinical Presentation and Disease Markers, Comparison of Knee Joint PTOA Models and Early Disease Implications. Int J Mol Sci 2021; 22:1996. [PMID: 33671471 PMCID: PMC7922905 DOI: 10.3390/ijms22041996] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 02/05/2021] [Accepted: 02/08/2021] [Indexed: 12/15/2022] Open
Abstract
Understanding the causality of the post-traumatic osteoarthritis (PTOA) disease process of the knee joint is important for diagnosing early disease and developing new and effective preventions or treatments. The aim of this review was to provide detailed clinical data on inflammatory and other biomarkers obtained from patients after acute knee trauma in order to (i) present a timeline of events that occur in the acute, subacute, and chronic post-traumatic phases and in PTOA, and (ii) to identify key factors present in the synovial fluid, serum/plasma and urine, leading to PTOA of the knee in 23-50% of individuals who had acute knee trauma. In this context, we additionally discuss methods of simulating knee trauma and inflammation in in vivo, ex vivo articular cartilage explant and in vitro chondrocyte models, and answer whether these models are representative of the clinical inflammatory stages following knee trauma. Moreover, we compare the pro-inflammatory cytokine concentrations used in such models and demonstrate that, compared to concentrations in the synovial fluid after knee trauma, they are exceedingly high. We then used the Bradford Hill Framework to present evidence that TNF-α and IL-6 cytokines are causal factors, while IL-1β and IL-17 are credible factors in inducing knee PTOA disease progresssion. Lastly, we discuss beneficial infrastructure for future studies to dissect the role of local vs. systemic inflammation in PTOA progression with an emphasis on early disease.
Collapse
Affiliation(s)
| | | | | | | | - Melanie L. Hart
- G.E.R.N. Center for Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Medical Center—Albert-Ludwigs-University of Freiburg, 79085 Freiburg im Breisgau, Germany; (C.M.K.); (R.A.); (J.M.H.); (B.R.)
| |
Collapse
|
14
|
Liu L, He J, Liu C, Yang M, Fu J, Yi J, Ai X, Liu M, Zhuang Y, Zhang Y, Huang B, Li C, Zhou Y, Feng C. Cartilage intermediate layer protein affects the progression of intervertebral disc degeneration by regulating the extracellular microenvironment (Review). Int J Mol Med 2021; 47:475-484. [PMID: 33416131 PMCID: PMC7797476 DOI: 10.3892/ijmm.2020.4832] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Accepted: 11/27/2020] [Indexed: 12/25/2022] Open
Abstract
Intervertebral disc degeneration (IDD), which is caused by multiple factors, affects the health of individuals and contributes to low back pain. The pathology of IDD is complicated, and changes in the extracellular microenvironment play an important role in promoting the process of degeneration. Cartilage intermediate layer protein (CILP) is a matrix protein that resides in the middle of human articular cartilage and is involved in numerous diseases that affect cartilage. However, there is no detailed review of the relationship between CILP and degenerative disc disease. Growing evidence has revealed the presence of CILP in the extracellular microenvironment of intervertebral discs (IVDs) and has suggested that there is a gradual increase in CILP in degenerative discs. Specifically, CILP plays an important role in regulating the metabolism of the extracellular matrix (ECM), an important component of the extracellular microenvironment. CILP can combine with transforming growth factor‑β or insulin‑like growth factor‑1 to regulate the ECM synthesis of IVDs and influence the balance of ECM metabolism, which leads to changes in the extracellular microenvironment to promote the process of IDD. It may be possible to show the correlation of CILP with IDD and to target CILP to interfere with IDD. For this purpose, in the present study, the current knowledge on CILP was summarized and a detailed description of CILP in discs was provided.
Collapse
Affiliation(s)
- Libangxi Liu
- Department of Orthopedics, Xinqiao Hospital, Army Medical University
| | - Jinyue He
- Department of Orthopedics, Xi'nan Hospital, Army Medical University, Chongqing 400037
| | - Chang Liu
- Department of Orthopedics, Xinqiao Hospital, Army Medical University
| | - Minghui Yang
- Department of Orthopedics, Xinqiao Hospital, Army Medical University
| | - Jiawei Fu
- Department of Orthopedics, Xinqiao Hospital, Army Medical University
| | - Jiarong Yi
- Department of Orthopedics, Xinqiao Hospital, Army Medical University
| | - Xuezheng Ai
- Department of Orthopedics, Xinqiao Hospital, Army Medical University
| | - Miao Liu
- Department of Orthopedics, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| | - Yong Zhuang
- Department of Orthopedics, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| | - Yaqing Zhang
- Department of Orthopedics, Xinqiao Hospital, Army Medical University
| | - Bo Huang
- Department of Orthopedics, Xinqiao Hospital, Army Medical University
| | - Changqing Li
- Department of Orthopedics, Xinqiao Hospital, Army Medical University
| | - Yue Zhou
- Department of Orthopedics, Xinqiao Hospital, Army Medical University
| | - Chencheng Feng
- Department of Orthopedics, Xinqiao Hospital, Army Medical University
| |
Collapse
|
15
|
Miran I, Scherer D, Ostyn P, Mazouni C, Drusch F, Bernard M, Louvet E, Adam J, Mathieu MC, Haffa M, Antignac JP, Le Bizec B, Vielh P, Dessen P, Perdry H, Delaloge S, Feunteun J. Adipose Tissue Properties in Tumor-Bearing Breasts. Front Oncol 2020; 10:1506. [PMID: 32974182 PMCID: PMC7472783 DOI: 10.3389/fonc.2020.01506] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 07/14/2020] [Indexed: 12/11/2022] Open
Abstract
The tissue stroma plays a major role in tumors' natural history. Most programs for tumor progression are not activated as cell-autonomous processes but under the conditions of cross-talks between tumor and stroma. Adipose tissue is a major component of breast stroma. This study compares adipose tissues in tumor-bearing breasts to those in tumor-free breasts with the intention of defining a signature that could translate into markers of cancer risk. In tumor-bearing breasts, we sampled adipose tissues adjacent to, or distant from the tumor. Parameters studied included: adipocytes size and density, immune cell infiltration, vascularization, secretome and gene expression. Adipose tissues from tumor-bearing breasts, whether adjacent to or distant from the tumor, do not differ from each other by any of these parameters. By contrast, adipose tissues from tumor-bearing breasts have the capacity to secrete twice as much interleukin 8 (IL-8) than those from tumor-free breasts and differentially express a set of 137 genes of which a significant fraction belongs to inflammation, integrin and wnt signaling pathways. These observations show that adipose tissues from tumor-bearing breasts have a distinct physiological status from those from tumor-free breasts. We propose that this constitutive status contributes as a non-cell autonomous process to determine permissiveness for tumor growth.
Collapse
Affiliation(s)
- Isabelle Miran
- Translational Research Lab, INSERM U981, Université Paris-Saclay, Villejuif, France
| | - Dominique Scherer
- Division of Preventive Oncology, National Center for Tumor Diseases (NCT) and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Institute of Medical Biometry and Informatics, University of Heidelberg, Heidelberg, Germany
| | - Pauline Ostyn
- UMR 9019 Genome Integrity and Cancers, Université Paris-Saclay, Villejuif, France
| | - Chafika Mazouni
- Breast Cancer Group, Université Paris-Saclay, Villejuif, France
| | - Françoise Drusch
- Biology and Pathology Department, Université Paris-Saclay, Villejuif, France
| | - Marine Bernard
- Biology and Pathology Department, Université Paris-Saclay, Villejuif, France
| | - Emilie Louvet
- Biology and Pathology Department, Université Paris-Saclay, Villejuif, France
| | - Julien Adam
- Biology and Pathology Department, Université Paris-Saclay, Villejuif, France
| | - Marie-Christine Mathieu
- Laboratoire d'Etude des Résidus et Contaminants dans les Aliments (LABERCA), UMR 1329 Oniris-INRA, Nantes, France
| | - Mariam Haffa
- Division of Preventive Oncology, National Center for Tumor Diseases (NCT) and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Translational Functional Cancer Genomics, National Center for Tumor Diseases (NCT) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jean-Philippe Antignac
- Laboratoire d'Etude des Résidus et Contaminants dans les Aliments (LABERCA), UMR 1329 Oniris-INRA, Nantes, France
| | - Bruno Le Bizec
- Laboratoire d'Etude des Résidus et Contaminants dans les Aliments (LABERCA), UMR 1329 Oniris-INRA, Nantes, France
| | - Philippe Vielh
- Biology and Pathology Department, Université Paris-Saclay, Villejuif, France
| | - Philippe Dessen
- Bioinformatics Core Facility, Université Paris-Saclay, Villejuif, France
| | - Hervé Perdry
- INSERM U669 - Equipe GGS Génomique & Génétique Statistique, Villejuif, France
| | | | - Jean Feunteun
- UMR 9019 Genome Integrity and Cancers, Université Paris-Saclay, Villejuif, France
| |
Collapse
|
16
|
Jeremiasse B, Matta C, Fellows CR, Boocock DJ, Smith JR, Liddell S, Lafeber F, van Spil WE, Mobasheri A. Alterations in the chondrocyte surfaceome in response to pro-inflammatory cytokines. BMC Mol Cell Biol 2020; 21:47. [PMID: 32586320 PMCID: PMC7318434 DOI: 10.1186/s12860-020-00288-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 06/03/2020] [Indexed: 02/07/2023] Open
Abstract
Background Chondrocytes are exposed to an inflammatory micro-environment in the extracellular matrix (ECM) of articular cartilage in joint diseases such as osteoarthritis (OA) and rheumatoid arthritis (RA). In OA, degenerative changes and low-grade inflammation within the joint transform the behaviour and metabolism of chondrocytes, disturb the balance between ECM synthesis and degradation, and alter the osmolality and ionic composition of the micro-environment. We hypothesize that chondrocytes adjust their physiology to the inflammatory microenvironment by modulating the expression of cell surface proteins, collectively referred to as the ‘surfaceome’. Therefore, the aim of this study was to characterize the surfaceome of primary equine chondrocytes isolated from healthy joints following exposure to the pro-inflammatory cytokines interleukin-1-beta (IL-1β) and tumour necrosis factor-alpha (TNF-α). We employed combined methodology that we recently developed for investigating the surfaceome in stem cells. Membrane proteins were isolated using an aminooxy-biotinylation technique and analysed by mass spectrometry using high throughput shotgun proteomics. Selected proteins were validated by western blotting. Results Amongst the 431 unique cell surface proteins identified, a high percentage of low-abundance proteins, such as ion channels, receptors and transporter molecules were detected. Data are available via ProteomeXchange with identifier PXD014773. A high number of proteins exhibited different expression patterns following chondrocyte stimulation with pro-inflammatory cytokines. Low density lipoprotein related protein 1 (LPR-1), thrombospondin-1 (TSP-1), voltage dependent anion channel (VDAC) 1–2 and annexin A1 were considered to be of special interest and were analysed further by western blotting. Conclusions Our results provide, for the first time, a repository for proteomic data on differentially expressed low-abundance membrane proteins on the surface of chondrocytes in response to pro-inflammatory stimuli.
Collapse
Affiliation(s)
- Bernadette Jeremiasse
- Department of Rheumatology & Clinical Immunology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Csaba Matta
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.
| | - Christopher R Fellows
- Department of Veterinary Pre-Clinical Sciences, School of Veterinary Science and Medicine, University of Surrey, Guildford, UK
| | - David J Boocock
- John van Geest Cancer Research Centre, Nottingham Trent University, Nottingham, NG11 8NS, UK
| | | | | | - Floris Lafeber
- Department of Rheumatology & Clinical Immunology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Willem E van Spil
- Department of Rheumatology & Clinical Immunology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Ali Mobasheri
- Department of Rheumatology & Clinical Immunology, University Medical Centre Utrecht, Utrecht, The Netherlands. .,Research Unit of Medical Imaging, Physics and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland. .,Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania. .,Centre for Sport, Exercise and Osteoarthritis Research Versus Arthritis, Queen's Medical Centre, Nottingham, UK. .,Department of Orthopedics, UMC Utrecht, Utrecht, The Netherlands.
| |
Collapse
|
17
|
Fernández-Puente P, González-Rodríguez L, Calamia V, Picchi F, Lourido L, Camacho-Encina M, Oreiro N, Rocha B, Paz-González R, Marina A, García C, Blanco FJ, Ruiz-Romero C. Analysis of Endogenous Peptides Released from Osteoarthritic Cartilage Unravels Novel Pathogenic Markers. Mol Cell Proteomics 2019; 18:2018-2028. [PMID: 31352363 PMCID: PMC6773562 DOI: 10.1074/mcp.ra119.001554] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 07/08/2019] [Indexed: 12/31/2022] Open
Abstract
Osteoarthritis (OA) is a pathology characterized by the loss of articular cartilage. In this study, we performed a peptidomic strategy to identify endogenous peptides (neopeptides) that are released from human osteoarthritic tissue, which may serve as disease markers. With this aim, secretomes of osteoarthritic and healthy articular cartilages obtained from knee and hip were analyzed by shotgun peptidomics. This discovery step led to the identification of 1175 different peptides, corresponding to 101 proteins, as products of the physiological or pathological turnover of cartilage extracellular matrix. Then, a targeted multiple reaction monitoring-mass spectrometry method was developed to quantify the panel of best marker candidates on a larger set of samples (n = 62). Statistical analyses were performed to evaluate the significance of the observed differences and the ability of the neopeptides to classify the tissue. Eight of them were differentially abundant in the media from wounded zones of OA cartilage compared with the healthy tissue (p < 0.05). Three neopeptides belonging to Clusterin and one from Cartilage Oligomeric Matrix Protein showed a disease-dependent decrease specifically in hip OA, whereas two from Prolargin (PRELP) and one from Cartilage Intermediate Layer Protein 1 were significantly increased in samples from knee OA. The release of one peptide from PRELP showed the best metrics for tissue classification (AUC = 0.834). The present study reveals specific neopeptides that are differentially released from knee or hip human osteoarthritic cartilage compared with healthy tissue. This evidences the intervention of characteristic pathogenic pathways in OA and provides a novel panel of peptidic candidates for biomarker development.
Collapse
MESH Headings
- Aged
- Aged, 80 and over
- Biomarkers/metabolism
- Cartilage, Articular/cytology
- Cartilage, Articular/metabolism
- Cartilage, Articular/pathology
- Case-Control Studies
- Cells, Cultured
- Chromatography, Liquid
- Culture Media, Conditioned/chemistry
- Extracellular Matrix/metabolism
- Female
- Humans
- Male
- Organ Specificity
- Osteoarthritis, Hip/metabolism
- Osteoarthritis, Hip/pathology
- Osteoarthritis, Knee/metabolism
- Osteoarthritis, Knee/pathology
- Peptides/metabolism
- Proteomics/methods
- Tandem Mass Spectrometry
Collapse
Affiliation(s)
- Patricia Fernández-Puente
- Proteomics Unit-PBR2-ProteoRed/ISCIII, Grupo de Investigación de Reumatología (GIR). Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), SERGAS. As Xubias, 84, 15006 A Coruña, Spain.; Agrupación Estratégica CICA - INIBIC, Universidade da Coruña, 15071 A Coruña, Spain
| | - Lucía González-Rodríguez
- Proteomics Unit-PBR2-ProteoRed/ISCIII, Grupo de Investigación de Reumatología (GIR). Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), SERGAS. As Xubias, 84, 15006 A Coruña, Spain
| | - Valentina Calamia
- Proteomics Unit-PBR2-ProteoRed/ISCIII, Grupo de Investigación de Reumatología (GIR). Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), SERGAS. As Xubias, 84, 15006 A Coruña, Spain
| | - Florencia Picchi
- Proteomics Unit-PBR2-ProteoRed/ISCIII, Grupo de Investigación de Reumatología (GIR). Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), SERGAS. As Xubias, 84, 15006 A Coruña, Spain
| | - Lucía Lourido
- Proteomics Unit-PBR2-ProteoRed/ISCIII, Grupo de Investigación de Reumatología (GIR). Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), SERGAS. As Xubias, 84, 15006 A Coruña, Spain
| | - María Camacho-Encina
- Proteomics Unit-PBR2-ProteoRed/ISCIII, Grupo de Investigación de Reumatología (GIR). Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), SERGAS. As Xubias, 84, 15006 A Coruña, Spain
| | - Natividad Oreiro
- Proteomics Unit-PBR2-ProteoRed/ISCIII, Grupo de Investigación de Reumatología (GIR). Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), SERGAS. As Xubias, 84, 15006 A Coruña, Spain
| | - Beatriz Rocha
- Proteomics Unit-PBR2-ProteoRed/ISCIII, Grupo de Investigación de Reumatología (GIR). Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), SERGAS. As Xubias, 84, 15006 A Coruña, Spain
| | - Rocío Paz-González
- Proteomics Unit-PBR2-ProteoRed/ISCIII, Grupo de Investigación de Reumatología (GIR). Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), SERGAS. As Xubias, 84, 15006 A Coruña, Spain
| | - Anabel Marina
- Centro de Biología Molecular Severo Ochoa, CSIC. Nicolás Cabrera, 1, 28049 Madrid, Spain
| | - Carlos García
- Centro de Biología Molecular Severo Ochoa, CSIC. Nicolás Cabrera, 1, 28049 Madrid, Spain
| | - Francisco J Blanco
- Proteomics Unit-PBR2-ProteoRed/ISCIII, Grupo de Investigación de Reumatología (GIR). Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), SERGAS. As Xubias, 84, 15006 A Coruña, Spain.; Departamento de Medicina, Fisioterapia y Ciencias Biomédicas. Universidade da Coruña, 15006 A Coruña, Spain.; RIER-RED de Inflamación y Enfermedades Reumáticas, INIBIC-CHUAC, As Xubias 84, 15006 A Coruña, Spain.
| | - Cristina Ruiz-Romero
- Proteomics Unit-PBR2-ProteoRed/ISCIII, Grupo de Investigación de Reumatología (GIR). Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), SERGAS. As Xubias, 84, 15006 A Coruña, Spain.; CIBER-BBN Instituto de Salud Carlos III INIBIC-CHUAC As Xubias 84, 15006 A Coruna, Spain.
| |
Collapse
|
18
|
Song P, Kwon Y, Joo JY, Kim DG, Yoon JH. Secretomics to Discover Regulators in Diseases. Int J Mol Sci 2019; 20:ijms20163893. [PMID: 31405033 PMCID: PMC6720857 DOI: 10.3390/ijms20163893] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 08/01/2019] [Accepted: 08/08/2019] [Indexed: 01/03/2023] Open
Abstract
Secretory proteins play important roles in the cross-talk of individual functional units, including cells. Since secretory proteins are essential for signal transduction, they are closely related with disease development, including metabolic and neural diseases. In metabolic diseases, adipokines, myokines, and hepatokines are secreted from respective organs under specific environmental conditions, and play roles in glucose homeostasis, angiogenesis, and inflammation. In neural diseases, astrocytes and microglia cells secrete cytokines and chemokines that play roles in neurotoxic and neuroprotective responses. Mass spectrometry-based secretome profiling is a powerful strategy to identify and characterize secretory proteins. This strategy involves stepwise processes such as the collection of conditioned medium (CM) containing secretome proteins and concentration of the CM, peptide preparation, mass analysis, database search, and filtering of secretory proteins; each step requires certain conditions to obtain reliable results. Proteomic analysis of extracellular vesicles has become a new research focus for understanding the additional extracellular functions of intracellular proteins. Here, we provide a review of the insights obtained from secretome analyses with regard to disease mechanisms, and highlight the future prospects of this technology. Continued research in this field is expected to provide valuable information on cell-to-cell communication and uncover new pathological mechanisms.
Collapse
Affiliation(s)
- Parkyong Song
- Department of Convergence Medicine, Pusan National University School of Medicine, Yangsan 50612, Korea
| | - Yonghoon Kwon
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Korea
| | - Jae-Yeol Joo
- Neurodegenerative Disease Research Group, Korea Brain Research Institute, Daegu 41062, Korea
| | - Do-Geun Kim
- Dementia Research Group, Korea Brain Research Institute, Daegu 41062, Korea
| | - Jong Hyuk Yoon
- Neurodegenerative Disease Research Group, Korea Brain Research Institute, Daegu 41062, Korea.
| |
Collapse
|
19
|
Rezuș E, Cardoneanu A, Burlui A, Luca A, Codreanu C, Tamba BI, Stanciu GD, Dima N, Bădescu C, Rezuș C. The Link Between Inflammaging and Degenerative Joint Diseases. Int J Mol Sci 2019; 20:614. [PMID: 30708978 PMCID: PMC6386892 DOI: 10.3390/ijms20030614] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Revised: 01/21/2019] [Accepted: 01/29/2019] [Indexed: 12/12/2022] Open
Abstract
Aging is an inevitable process in the human body that is associated with a multitude of systemic and localized changes. All these conditions have a common pathogenic mechanism characterized by the presence of a low-grade proinflammatory status. Inflammaging refers to all the processes that contribute to the occurrence of various diseases associated with aging such as frailty, atherosclerosis, Alzheimer's disease, sarcopenia, type 2 diabetes, or osteoarthritis. Inflammaging is systemic, chronic, and asymptomatic. Osteoarthritis and many age-related degenerative joint diseases are correlated with aging mechanisms such as the presence of an inflammatory microenvironment and the impaired link between inflammasomes and autophagy. There is a close relationship between chondrocyte activity and local articular environment changes due to cell senescence, followed by secretion of inflammatory mediators. In addition, systemic inflammaging can lead to cartilage destruction, pain, disability, and an impaired quality of life. The purpose of this review is to summarize the main mechanisms implicated in inflammaging and the connection it has with degenerative joint diseases.
Collapse
Affiliation(s)
- Elena Rezuș
- Department of Rheumatology and Physiotherapy, "Grigore T. Popa'' University of Medicine and Pharmacy Iași, University Street no. 16, 700115 Iași, Romania.
| | - Anca Cardoneanu
- Department of Rheumatology and Physiotherapy, "Grigore T. Popa'' University of Medicine and Pharmacy Iași, University Street no. 16, 700115 Iași, Romania.
| | - Alexandra Burlui
- Department of Rheumatology and Physiotherapy, "Grigore T. Popa'' University of Medicine and Pharmacy Iași, University Street no. 16, 700115 Iași, Romania.
| | - Andrei Luca
- Advanced Center for Research and Development in Experimental Medicine (CEMEX), "Grigore T. Popa'' University of Medicine and Pharmacy Iași, Mihail Kogălniceanu Street no. 9-13, 700454 Iasi, Romania.
| | - Cătălin Codreanu
- Center for Rheumatic Diseases, "Carol Davila" University of Medicine and Pharmacy Bucharest, Eroii Sanitari no.8 Boulevard, 050474 Bucharest, Romania.
| | - Bogdan Ionel Tamba
- Advanced Center for Research and Development in Experimental Medicine (CEMEX), "Grigore T. Popa'' University of Medicine and Pharmacy Iași, Mihail Kogălniceanu Street no. 9-13, 700454 Iasi, Romania.
| | - Gabriela-Dumitrița Stanciu
- Advanced Center for Research and Development in Experimental Medicine (CEMEX), "Grigore T. Popa'' University of Medicine and Pharmacy Iași, Mihail Kogălniceanu Street no. 9-13, 700454 Iasi, Romania.
| | - Nicoleta Dima
- Department of Internal Medicine, "Grigore T. Popa'' University of Medicine and Pharmacy Iași, University Street no. 16, zip code 700115 Iași, Romania.
| | - Codruța Bădescu
- Department of Internal Medicine, "Grigore T. Popa'' University of Medicine and Pharmacy Iași, University Street no. 16, zip code 700115 Iași, Romania.
| | - Ciprian Rezuș
- Department of Internal Medicine, "Grigore T. Popa'' University of Medicine and Pharmacy Iași, University Street no. 16, zip code 700115 Iași, Romania.
| |
Collapse
|
20
|
Mobasheri A, Bay-Jensen AC, Gualillo O, Larkin J, Levesque MC, Henrotin Y. Soluble biochemical markers of osteoarthritis: Are we close to using them in clinical practice? Best Pract Res Clin Rheumatol 2018; 31:705-720. [PMID: 30509415 DOI: 10.1016/j.berh.2018.07.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 05/17/2018] [Accepted: 07/06/2018] [Indexed: 02/07/2023]
Abstract
Osteoarthritis (OA) is the most common form of arthritis and a major cause of pain and disability. Recent work suggests that the global burden of OA is increasing, and costs associated with treatment are expected to increase dramatically as the aging human population expands. OA is currently diagnosed using radiography, but this technique is an indirect and insensitive measure of alterations in articular cartilage and fails to measure dynamic inflammatory processes in the joint. Radiographic changes detected overtime are small and occur in only a subset (progressors) of patients with OA. Therefore, we diagnose patients with OA on the basis of a diagnostic classification that is outdated. We also use the same tools and approaches for assessing the efficacy of new pharmacological and nonpharmacological interventions. In this review, we discuss the utility of soluble biochemical markers as biomarkers of OA and discuss whether we are close to using them in clinical practice. Combining patient information, functional imaging and carefully selected panels of biomarkers can help in achieving enhanced patient stratification and lead to better designed clinical trials. Biomarkers can be used for molecular endotyping and for developing more effective and more personalized treatments that will enhance clinical care for patients with OA.
Collapse
Affiliation(s)
- Ali Mobasheri
- The D-BOARD FP7 Consortium(1), European Union; The APPROACH IMI Consortium(2), European Union; Faculty of Health and Medical Sciences, University of Surrey, Guildford, Surrey, United Kingdom; Arthritis Research UK Centre for Sport, Exercise and Osteoarthritis, Queen's Medical Centre, Nottingham, United Kingdom; Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania.
| | - Anne-Christine Bay-Jensen
- The D-BOARD FP7 Consortium(1), European Union; The APPROACH IMI Consortium(2), European Union; Rheumatology, Biomarkers and Research, Nordic Bioscience A/S, Herlev, Denmark
| | - Oreste Gualillo
- SERGAS (Servizo Galego de Saude) and IDIS (Instituto de Investigación Sanitaria de Santiago), Research Laboratory 9, The NEIRID Lab (Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases), Santiago University Clinical Hospital, Santiago de Compostela, 15706, Spain
| | - Jonanthan Larkin
- The APPROACH IMI Consortium(2), European Union; C3 DPU, Immunoinflammation Therapeutic Area, GlaxoSmithKline, King of Prussia, PA, 19406, United States
| | - Marc C Levesque
- The APPROACH IMI Consortium(2), European Union; AbbVie Bioresearch Center, 100 Research Drive, Worcester, MA, 01605, United States
| | - Yves Henrotin
- The D-BOARD FP7 Consortium(1), European Union; The APPROACH IMI Consortium(2), European Union; Bone and Cartilage Research Unit, Arthropôle Liege, University of Liège, Liège, Belgium; Physical Therapy and Rehabilitation Department, Princess Paola Hospital, Vivalia, Marche-en-Famenne, Belgium
| |
Collapse
|
21
|
Abstract
INTRODUCTION Osteoarthritis (OA), a chronic, debilitating and degenerative disease of the joints, is the most common form of arthritis. The seriousness of this prevalent and chronic disease is often overlooked. Disease modifying OA drug development is hindered by the lack of soluble biomarkers to detect OA early. The objective of OA biomarker research is to identify early OA prior to the appearance of radiographic signs and the development of pain. Areas covered: This review has focused on extracellular genomic material that could serve as biomarkers of OA. Recent studies have examined the expression of extracellular genomic material such as miRNA, lncRNA, snoRNA, mRNA and cell-free DNA, which are aberrantly expressed in the body fluids of OA patients. Changes in genomic content of peripheral blood mononuclear cells in OA could also function as biomarkers of OA. Expert commentary: There is an unmet need for soluble biomarkers for detecting and then monitoring OA disease progression. Extracellular genomic material research may also reveal more about the underlying pathophysiology of OA. Minimally-invasive liquid biopsies such as synovial fluid and blood sampling of genomic material may be more sensitive over radiography in the detection, diagnosis and monitoring of OA in the future.
Collapse
Affiliation(s)
- Emma Budd
- a The D-BOARD European Consortium for Biomarker Discovery, School of Veterinary Medicine , University of Surrey , Guildford , UK.,b Department of Veterinary Pre-Clinical Sciences, School of Veterinary Medicine, Faculty of Health and Medical Sciences , University of Surrey , Guildford , UK
| | - Giovanna Nalesso
- b Department of Veterinary Pre-Clinical Sciences, School of Veterinary Medicine, Faculty of Health and Medical Sciences , University of Surrey , Guildford , UK
| | - Ali Mobasheri
- a The D-BOARD European Consortium for Biomarker Discovery, School of Veterinary Medicine , University of Surrey , Guildford , UK.,b Department of Veterinary Pre-Clinical Sciences, School of Veterinary Medicine, Faculty of Health and Medical Sciences , University of Surrey , Guildford , UK.,c Arthritis Research UK Centre for Sport, Exercise and Osteoarthritis , Queen's Medical Centre , Nottingham , UK
| |
Collapse
|
22
|
Sanchez C, Bay-Jensen AC, Pap T, Dvir-Ginzberg M, Quasnichka H, Barrett-Jolley R, Mobasheri A, Henrotin Y. Chondrocyte secretome: a source of novel insights and exploratory biomarkers of osteoarthritis. Osteoarthritis Cartilage 2017; 25:1199-1209. [PMID: 28232143 DOI: 10.1016/j.joca.2017.02.797] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 01/31/2017] [Accepted: 02/14/2017] [Indexed: 02/02/2023]
Abstract
The extracellular matrix (ECM) of articular cartilage is comprised of complex networks of proteins and glycoproteins, all of which are expressed by its resident cell, the chondrocyte. Cartilage is a unique tissue given its complexity and ability to resist repeated load and deformation. The mechanisms by which articular cartilage maintains its integrity throughout our lifetime is not fully understood, however there are numerous regulatory pathways known to govern ECM turnover in response to mechanical stimuli. To further our understanding of this field, we envision that proteomic analysis of the secretome will provide information on how the chondrocyte remodels the surrounding ECM in response to load, in addition to providing information on the metabolic state of the cell. In this review, we attempt to summarize the recent mass spectrometry-based proteomic discoveries in healthy and diseased cartilage and chondrocytes, to facilitate the discovery of novel biomarkers linked to degenerative pathologies, such as osteoarthritis (OA).
Collapse
Affiliation(s)
- C Sanchez
- Bone and Cartilage Research Unit, Arthropôle Liège, University of Liège, CHU Sart-Tilman, Belgium; The D-BOARD European Consortium for Biomarker Discovery.
| | - A-C Bay-Jensen
- The D-BOARD European Consortium for Biomarker Discovery; Department of Rheumatology, Biomarkers and Research, Nordic Bioscience, Herlev Hovedgade 207, 2730, Herlev, Denmark.
| | - T Pap
- The D-BOARD European Consortium for Biomarker Discovery; Institute of Experimental Musculoskeletal Medicine, University Hospital Munster, Domagkstrasse 3, D-48149, Munster, Germany.
| | - M Dvir-Ginzberg
- The D-BOARD European Consortium for Biomarker Discovery; Institute of Dental Sciences, Faculty of Dental Medicine, Hebrew University of Jerusalem, P.O. Box 12272, Jerusalem, 91120, Israel.
| | - H Quasnichka
- The D-BOARD European Consortium for Biomarker Discovery; Department of Veterinary Pre-Clinical Sciences, School of Veterinary Medicine, University of Surrey, Guildford, GU2 7AL, United Kingdom.
| | - R Barrett-Jolley
- The D-BOARD European Consortium for Biomarker Discovery; Department of Musculoskeletal Biology, Institute of Ageing and Chronic Disease, Faculty of Health & Life Sciences, University of Liverpool, Liverpool, United Kingdom.
| | - A Mobasheri
- The D-BOARD European Consortium for Biomarker Discovery; Department of Veterinary Pre-Clinical Sciences, School of Veterinary Medicine, University of Surrey, Guildford, GU2 7AL, United Kingdom; Faculty of Health and Medical Sciences, Duke of Kent Building, University of Surrey, Guildford, Surrey, GU2 7XH, United Kingdom; Arthritis Research UK Centre for Sport, Exercise and Osteoarthritis, Queen's Medical Centre, Nottingham, NG7 2UH, United Kingdom; Center of Excellence in Genomic Medicine Research (CEGMR), King Fahd Medical Research Center (KFMRC), Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, 21589, Saudi Arabia.
| | - Y Henrotin
- Bone and Cartilage Research Unit, Arthropôle Liège, University of Liège, CHU Sart-Tilman, Belgium; The D-BOARD European Consortium for Biomarker Discovery.
| |
Collapse
|
23
|
Schneider MC, Barnes CA, Bryant SJ. Characterization of the chondrocyte secretome in photoclickable poly(ethylene glycol) hydrogels. Biotechnol Bioeng 2017; 114:2096-2108. [PMID: 28436002 DOI: 10.1002/bit.26320] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 12/28/2016] [Accepted: 04/17/2017] [Indexed: 12/30/2022]
Abstract
Poly(ethylene glycol) (PEG) hydrogels are highly tunable platforms that are promising cell delivery vehicles for chondrocytes and cartilage tissue engineering. In addition to characterizing the type of extracellular matrix (ECM) that forms, understanding the types of proteins that are secreted by encapsulated cells may be important. Thus, the objectives for this study were to characterize the secretome of chondrocytes encapsulated in PEG hydrogels and determine whether the secretome varies as a function of hydrogel stiffness and culture condition. Bovine chondrocytes were encapsulated in photoclickable PEG hydrogels with a compressive modulus of 8 and 46 kPa and cultured under free swelling or dynamic compressive loading conditions. Cartilage ECM deposition was assessed by biochemical assays and immunohistochemistry. The conditioned medium was analyzed by liquid chromatography-tandem mass spectrometry. Chondrocytes maintained their phenotype within the hydrogels and deposited cartilage-specific ECM that increased over time and included aggrecan and collagens II and VI. Analysis of the secretome revealed a total of 64 proteins, which were largely similar among all experimental conditions. The identified proteins have diverse functions such as biological regulation, response to stress, and collagen fibril organization. Notably, many of the proteins important to the assembly of a collagen-rich cartilage ECM were identified and included collagen types II(α1), VI (α1, α2, and α3), IX (α1), XI (α1 and α2), and biglycan. In addition, many of the other identified proteins have been reported to be present within cell-secreted exosomes. In summary, chondrocytes encapsulated within photoclickable PEG hydrogels secrete many types of proteins that diffuse out of the hydrogel and which have diverse functions, but which are largely preserved across different hydrogel culture environments. Biotechnol. Bioeng. 2017;114: 2096-2108. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Margaret C Schneider
- Department of Chemical and Biological Engineering, University of Colorado, 3415 Colorado Ave, Campus Box 596, Boulder 80309, Colorado.,Biofrontiers Institute, University of Colorado, Boulder, Colorado
| | | | - Stephanie J Bryant
- Department of Chemical and Biological Engineering, University of Colorado, 3415 Colorado Ave, Campus Box 596, Boulder 80309, Colorado.,Biofrontiers Institute, University of Colorado, Boulder, Colorado.,Material Science and Engineering Program, University of Colorado, Boulder, Colorado
| |
Collapse
|
24
|
Rocha B, Cillero-Pastor B, Blanco FJ, Ruiz-Romero C. MALDI mass spectrometry imaging in rheumatic diseases. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2016; 1865:784-794. [PMID: 27742553 DOI: 10.1016/j.bbapap.2016.10.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2016] [Revised: 09/29/2016] [Accepted: 10/04/2016] [Indexed: 01/15/2023]
Abstract
Mass spectrometry imaging (MSI) is a technique used to visualize the spatial distribution of biomolecules such as peptides, proteins, lipids or other organic compounds by their molecular masses. Among the different MSI strategies, MALDI-MSI provides a sensitive and label-free approach for imaging of a wide variety of protein or peptide biomarkers from the surface of tissue sections, being currently used in an increasing number of biomedical applications such as biomarker discovery and tissue classification. In the field of rheumatology, MALDI-MSI has been applied to date for the analysis of joint tissues such as synovial membrane or cartilage. This review summarizes the studies and key achievements obtained using MALDI-MSI to increase understanding on rheumatic pathologies and to describe potential diagnostic or prognostic biomarkers of these diseases. This article is part of a Special Issue entitled: MALDI Imaging, edited by Dr. Corinna Henkel and Prof. Peter Hoffmann.
Collapse
Affiliation(s)
- Beatriz Rocha
- Proteomics Unit-ProteoRed/ISCIII, Rheumatology Group, INIBIC - Hospital Universitario de A Coruña, SERGAS, A Coruña, Spain
| | | | - Francisco J Blanco
- Proteomics Unit-ProteoRed/ISCIII, Rheumatology Group, INIBIC - Hospital Universitario de A Coruña, SERGAS, A Coruña, Spain; RIER-RED de Inflamación y Enfermedades Reumáticas, INIBIC-CHUAC, A Coruña, Spain.
| | - Cristina Ruiz-Romero
- Proteomics Unit-ProteoRed/ISCIII, Rheumatology Group, INIBIC - Hospital Universitario de A Coruña, SERGAS, A Coruña, Spain; CIBER-BBN Instituto de Salud Carlos III, INIBIC-CHUAC, A Coruña, Spain.
| |
Collapse
|
25
|
Wilson R, Golub SB, Rowley L, Angelucci C, Karpievitch YV, Bateman JF, Fosang AJ. Novel Elements of the Chondrocyte Stress Response Identified Using an in Vitro Model of Mouse Cartilage Degradation. J Proteome Res 2016; 15:1033-50. [PMID: 26794603 DOI: 10.1021/acs.jproteome.5b01115] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The destruction of articular cartilage in osteoarthritis involves chondrocyte dysfunction and imbalanced extracellular matrix (ECM) homeostasis. Pro-inflammatory cytokines such as interleukin-1α (IL-1α) contribute to osteoarthritis pathophysiology, but the effects of IL-1α on chondrocytes within their tissue microenvironment have not been fully evaluated. To redress this we used label-free quantitative proteomics to analyze the chondrocyte response to IL-1α within a native cartilage ECM. Mouse femoral heads were cultured with and without IL-1α, and both the tissue proteome and proteins released into the media were analyzed. New elements of the chondrocyte response to IL-1α related to cellular stress included markers for protein misfolding (Armet, Creld2, and Hyou1), enzymes involved in glutathione biosynthesis and regeneration (Gstp1, Gsto1, and Gsr), and oxidative stress proteins (Prdx2, Txn, Atox1, Hmox1, and Vnn1). Other proteins previously not associated with the IL-1α response in cartilage included ECM components (Smoc2, Kera, and Crispld1) and cysteine proteases (cathepsin Z and legumain), while chondroadherin and cartilage-derived C-type lectin (Clec3a) were identified as novel products of IL-1α-induced cartilage degradation. This first proteome-level view of the cartilage IL-1α response identified candidate biomarkers of cartilage destruction and novel targets for therapeutic intervention in osteoarthritis.
Collapse
Affiliation(s)
- Richard Wilson
- Central Science Laboratory, University of Tasmania , Hobart, Tasmania 7001, Australia.,Murdoch Childrens Research Institute, Royal Children's Hospital , Parkville, Melbourne, Victoria 3052, Australia
| | - Suzanne B Golub
- Murdoch Childrens Research Institute, Royal Children's Hospital , Parkville, Melbourne, Victoria 3052, Australia.,Department of Pediatrics, University of Melbourne , Parkville, Victoria 3052, Australia
| | - Lynn Rowley
- Murdoch Childrens Research Institute, Royal Children's Hospital , Parkville, Melbourne, Victoria 3052, Australia
| | - Constanza Angelucci
- Murdoch Childrens Research Institute, Royal Children's Hospital , Parkville, Melbourne, Victoria 3052, Australia
| | - Yuliya V Karpievitch
- School of Physical Sciences, University of Tasmania , Hobart, Tasmania 7001, Australia.,Centre of Excellence in Plant Energy Biology, University of Western Australia and Harry Perkins Institute of Medical Research , Perth, Western Australia 6009, Australia
| | - John F Bateman
- Murdoch Childrens Research Institute, Royal Children's Hospital , Parkville, Melbourne, Victoria 3052, Australia.,Department of Biochemistry and Molecular Biology, University of Melbourne , Parkville, Victoria 3052, Australia
| | - Amanda J Fosang
- Murdoch Childrens Research Institute, Royal Children's Hospital , Parkville, Melbourne, Victoria 3052, Australia.,Department of Pediatrics, University of Melbourne , Parkville, Victoria 3052, Australia
| |
Collapse
|
26
|
Zhang L, Liu YZ, Zeng Y, Zhu W, Zhao YC, Zhang JG, Zhu JQ, He H, Shen H, Tian Q, Deng FY, Papasian CJ, Deng HW. Network-based proteomic analysis for postmenopausal osteoporosis in Caucasian females. Proteomics 2015; 16:12-28. [DOI: 10.1002/pmic.201500005] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Revised: 09/06/2015] [Accepted: 10/28/2015] [Indexed: 01/18/2023]
Affiliation(s)
- Lan Zhang
- Center for Bioinformatics and Genomics, Department of Biostatistics and Bioinformatics, School of Public Health and Tropical Medicine; Tulane University; New Orleans LA USA
| | - Yao-Zhong Liu
- Center for Bioinformatics and Genomics, Department of Biostatistics and Bioinformatics, School of Public Health and Tropical Medicine; Tulane University; New Orleans LA USA
| | - Yong Zeng
- Center for Bioinformatics and Genomics, Department of Biostatistics and Bioinformatics, School of Public Health and Tropical Medicine; Tulane University; New Orleans LA USA
- College of Life Sciences and Bioengineering; Beijing Jiaotong University; Beijing P. R. China
| | - Wei Zhu
- Center for Bioinformatics and Genomics, Department of Biostatistics and Bioinformatics, School of Public Health and Tropical Medicine; Tulane University; New Orleans LA USA
| | - Ying-Chun Zhao
- Center for Bioinformatics and Genomics, Department of Biostatistics and Bioinformatics, School of Public Health and Tropical Medicine; Tulane University; New Orleans LA USA
| | - Ji-Gang Zhang
- Center for Bioinformatics and Genomics, Department of Biostatistics and Bioinformatics, School of Public Health and Tropical Medicine; Tulane University; New Orleans LA USA
| | - Jia-Qiang Zhu
- Center for Bioinformatics and Genomics, Department of Biostatistics and Bioinformatics, School of Public Health and Tropical Medicine; Tulane University; New Orleans LA USA
| | - Hao He
- Center for Bioinformatics and Genomics, Department of Biostatistics and Bioinformatics, School of Public Health and Tropical Medicine; Tulane University; New Orleans LA USA
| | - Hui Shen
- Center for Bioinformatics and Genomics, Department of Biostatistics and Bioinformatics, School of Public Health and Tropical Medicine; Tulane University; New Orleans LA USA
| | - Qing Tian
- Center for Bioinformatics and Genomics, Department of Biostatistics and Bioinformatics, School of Public Health and Tropical Medicine; Tulane University; New Orleans LA USA
| | - Fei-Yan Deng
- Center for Bioinformatics and Genomics, Department of Biostatistics and Bioinformatics, School of Public Health and Tropical Medicine; Tulane University; New Orleans LA USA
- Laboratory of Proteins and Proteomics, Department of Epidemiology; Soochow University School of Public Health; Suzhou P. R. China
| | - Christopher J. Papasian
- Department of Basic Medical Sciences, School of Medicine; University of Missouri - Kansas City; MO USA
| | - Hong-Wen Deng
- Center for Bioinformatics and Genomics, Department of Biostatistics and Bioinformatics, School of Public Health and Tropical Medicine; Tulane University; New Orleans LA USA
- College of Life Sciences and Bioengineering; Beijing Jiaotong University; Beijing P. R. China
| |
Collapse
|
27
|
Aging-related inflammation in osteoarthritis. Osteoarthritis Cartilage 2015; 23:1966-71. [PMID: 26521742 PMCID: PMC4630808 DOI: 10.1016/j.joca.2015.01.008] [Citation(s) in RCA: 359] [Impact Index Per Article: 35.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Revised: 01/05/2015] [Accepted: 01/09/2015] [Indexed: 02/07/2023]
Abstract
It is well accepted that aging is an important contributing factor to the development of osteoarthritis (OA). The mechanisms responsible appear to be multifactorial and may include an age-related pro-inflammatory state that has been termed "inflamm-aging." Age-related inflammation can be both systemic and local. Systemic inflammation can be promoted by aging changes in adipose tissue that result in increased production of cytokines such as interleukin (IL)-6 and tumor necrosis factor-α (TNFα). Numerous studies have shown an age-related increase in blood levels of IL-6 that has been associated with decreased physical function and frailty. Importantly, higher levels of IL-6 have been associated with an increased risk of knee OA progression. However, knockout of IL-6 in male mice resulted in worse age-related OA rather than less OA. Joint tissue cells, including chondrocytes and meniscal cells, as well as the neighboring infrapatellar fat in the knee joint, can be a local source of inflammatory mediators that increase with age and contribute to OA. An increased production of pro-inflammatory mediators that include cytokines and chemokines, as well as matrix-degrading enzymes important in joint tissue destruction, can be the result of cell senescence and the development of the senescence-associated secretory phenotype (SASP). Further studies are needed to better understand the basis for inflamm-aging and its role in OA with the hope that this work will lead to new interventions targeting inflammation to reduce not only joint tissue destruction but also pain and disability in older adults with OA.
Collapse
|
28
|
Veronesi F, Fini M, Giavaresi G, Ongaro A, De Mattei M, Pellati A, Setti S, Tschon M. Experimentally induced cartilage degeneration treated by pulsed electromagnetic field stimulation; an in vitro study on bovine cartilage. BMC Musculoskelet Disord 2015; 16:308. [PMID: 26480822 PMCID: PMC4616002 DOI: 10.1186/s12891-015-0760-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2015] [Accepted: 10/07/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Osteoarthritis (OA) is the final result of progressive alterations to articular cartilage structure, composition and cellularity, followed by an increase in the concentration of pro-inflammatory cytokines in joint synovial fluid. Even though the effect of pulsed electromagnetic field (PEMF) stimulation in counteracting OA progression and inflammation is of increasing interest, because of its anabolic and anti-inflammatory properties, the present study aimed to improve the knowledge on cartilage extracellular matrix (ECM) and chondrocyte changes related to the exposure of PEMF, from a histological and histomorphometric point of view. METHODS An in vitro OA model was realized, culturing bovine cartilage explants with a high dose of interleukin 1β (IL1β, 50 ng/ml) at different experimental times (24 h, and 7 and 21 days). The effects of PEMFs (75 Hz, 1.5 mT) were evaluated in cartilage explants treated with IL1β or not (control), in terms of cartilage structure, cellularity and proteoglycans, glycosaminoglycans, collagen II and transforming growth factor β1 synthesis by using histology, histomorphometry and immunohistochemistry. RESULTS Making a comparison with control cartilage, IL1β-treated explants showed a decrease in cartilage matrix, structure and cellularity parameters. PEMFs were able to counteract the progression of OA acting on both cartilage cellularity and ECM in cartilage previously treated with IL1β. Normal distribution (Kolmogroc-Smirnov test) and homoscedasticity (Levene test) of data were verified, then, the non-parametric Kruskal Wallis test followed by Mann-Whiteny U test for pairwise comparisons were performed. The p-value was adjusted according to the Dunn-Sidak correction. CONCLUSIONS These results, obtained by culturing and treating cartilage explants from two different joints, confirmed that PEMF stimulation can be used as adjuvant therapy to preserve cartilage from detrimental effects of high inflammatory cytokine levels during OA.
Collapse
Affiliation(s)
- Francesca Veronesi
- Department Rizzoli RIT, Rizzoli Orthopedic Institute, Laboratory of Biocompatibility, Innovative Technologies and Advanced Therapies, Bologna, 40136, Italy.
| | - Milena Fini
- Department Rizzoli RIT, Rizzoli Orthopedic Institute, Laboratory of Biocompatibility, Innovative Technologies and Advanced Therapies, Bologna, 40136, Italy. .,Laboratory of Preclinical and Surgical Studies, Rizzoli Orthopedic Institute, Bologna, 40136, Italy.
| | - Gianluca Giavaresi
- Department Rizzoli RIT, Rizzoli Orthopedic Institute, Laboratory of Biocompatibility, Innovative Technologies and Advanced Therapies, Bologna, 40136, Italy. .,Laboratory of Preclinical and Surgical Studies, Rizzoli Orthopedic Institute, Bologna, 40136, Italy.
| | - Alessia Ongaro
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, 44121, Italy.
| | - Monica De Mattei
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, 44121, Italy.
| | - Agnese Pellati
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, 44121, Italy.
| | - Stefania Setti
- IGEA - Clinical Biophysic, Carpi (Modena), 41012, Italy.
| | - Matilde Tschon
- Department Rizzoli RIT, Rizzoli Orthopedic Institute, Laboratory of Biocompatibility, Innovative Technologies and Advanced Therapies, Bologna, 40136, Italy. .,Laboratory of Preclinical and Surgical Studies, Rizzoli Orthopedic Institute, Bologna, 40136, Italy.
| |
Collapse
|
29
|
Hanzawa H, Sakamoto T, Kaneko A, Manri N, Zhao Y, Zhao S, Tamaki N, Kuge Y. Combined Plasma and Tissue Proteomic Study of Atherogenic Model Mouse: Approach To Elucidate Molecular Determinants in Atherosclerosis Development. J Proteome Res 2015; 14:4257-69. [PMID: 26323832 DOI: 10.1021/acs.jproteome.5b00405] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Atherogenic cardiovascular diseases are the major cause of mortality. Prevention and prediction of incidents is important; however, biomarkers that directly reflect the disease progression remain poorly investigated. To elucidate molecular determinants of atherogenesis, proteomic approaches are advantageous by using model animals for comparing changes occurring systematically (bloodstream) and locally (lesion) in accordance with the disease progression stages. We conducted differential mass spectrometric analysis between apolipoprotein E deficient (apoED) and wild-type (wt) mice using the plasma and arterial tissue of both types of mice obtained at four pathognomonic time points of the disease. A total of 100 proteins in the plasma and 390 in the arterial tissues were continuously detected throughout the four time points; 29 were identified in common. Of those, 13 proteins in the plasma and 36 in the arterial tissues showed significant difference in abundance between the apoED and wt mice at certain time points. Importantly, we found that quantitative variation patterns regarding the pathognomonic time points did not always correspond between the plasma and arterial tissues, resulting in gaining insight into atherosclerotic plaque progression. These characteristic proteins were found to be components of inflammation, thrombus formation, and vascular remodeling, suggesting drastic and integrative alteration in accordance with atherosclerosis development.
Collapse
Affiliation(s)
- Hiroko Hanzawa
- Center for Exploratory Research, Research & Development Group, Hitachi, Ltd. , 350-0395 Hatoyama, Saitama Japan.,Central Institute of Isotope Science, Hokkaido University , 060-0814 Sapporo, Japan
| | - Takeshi Sakamoto
- Center for Technology Innovation - Healthcare, Research & Development Group, Hitachi, Ltd. , 185-8601 Kokubunji, Japan.,Department of Nuclear Medicine, Graduate School of Medicine, Hokkaido University , 060-8638 Sapporo, Japan
| | - Akihito Kaneko
- Center for Technology Innovation - Healthcare, Research & Development Group, Hitachi, Ltd. , 185-8601 Kokubunji, Japan
| | - Naomi Manri
- Center for Technology Innovation - Healthcare, Research & Development Group, Hitachi, Ltd. , 185-8601 Kokubunji, Japan.,Central Institute of Isotope Science, Hokkaido University , 060-0814 Sapporo, Japan
| | - Yan Zhao
- Department of Nuclear Medicine, Graduate School of Medicine, Hokkaido University , 060-8638 Sapporo, Japan
| | - Songji Zhao
- Department of Tracer Kinetics & Bio-analysis, Graduate School of Medicine, Hokkaido University , 060-8638 Sapporo, Japan
| | - Nagara Tamaki
- Department of Nuclear Medicine, Graduate School of Medicine, Hokkaido University , 060-8638 Sapporo, Japan
| | - Yuji Kuge
- Central Institute of Isotope Science, Hokkaido University , 060-0814 Sapporo, Japan.,Department of Integrated Molecular Imaging, Graduate School of Medicine, Hokkaido University , 060-8638 Sapporo, Japan
| |
Collapse
|
30
|
McIlwraith CW, Clegg PD. Science in brief: Report on the Havemeyer Foundation workshop on equine musculoskeletal biomarkers--current knowledge and future needs. Equine Vet J 2015; 46:651-3. [PMID: 25319159 DOI: 10.1111/evj.12339] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- C W McIlwraith
- Gail Holmes Equine Orthopaedic Research Center, Colorado State University, Fort Collins, USA
| | | |
Collapse
|
31
|
Svala E, Löfgren M, Sihlbom C, Rüetschi U, Lindahl A, Ekman S, Skiöldebrand E. An inflammatory equine model demonstrates dynamic changes of immune response and cartilage matrix molecule degradation in vitro. Connect Tissue Res 2015; 56:315-25. [PMID: 25803623 DOI: 10.3109/03008207.2015.1027340] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The molecular aspects of inflammation were investigated in equine articular cartilage explants using quantitative proteomics. Articular cartilage explants were stimulated with interleukin (IL)-1β in vitro for 25 days, and proteins released into cell culture media were chemically labeled with isobaric mass tags and analyzed by liquid chromatography-tandem mass spectrometry. A total of 127 proteins were identified and quantified in media from explants. IL-1β-stimulation resulted in an abundance of proteins related to inflammation, including matrix metalloproteinases, acute phase proteins, complement components and IL-6. Extracellular matrix (ECM) molecules were released at different time points, and fragmentation of aggrecan and cartilage oligomeric matrix protein was observed at days 3 and 6, similar to early-stage OA in vivo. Degradation products of the collagenous network were observed at days 18 and 22, similar to late-stage OA. This model displays a longitudinal quantification of released molecules from the ECM of articular cartilage. Identification of dynamic changes of extracellular matrix molecules in the secretome of equine explants stimulated with IL-1β over time may be useful for identifying components released at different time points during the spontaneous OA process.
Collapse
Affiliation(s)
- Emilia Svala
- Section of Pathology, Department of Biomedical Sciences and Veterinary Public Health, Swedish University of Agricultural Sciences , Uppsala , Sweden
| | | | | | | | | | | | | |
Collapse
|
32
|
Stoddart MJ, Bara J, Alini M. Cells and secretome--towards endogenous cell re-activation for cartilage repair. Adv Drug Deliv Rev 2015; 84:135-45. [PMID: 25174306 DOI: 10.1016/j.addr.2014.08.007] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Revised: 06/26/2014] [Accepted: 08/20/2014] [Indexed: 01/01/2023]
Abstract
Regenerative medicine approaches to cartilage tissue repair have mainly been concerned with the implantation of a scaffold material containing monolayer expanded cells into the defect, with the aim to differentiate the cells into chondrocytes. While this may be a valid approach, the secretome of the implanted cells and its effects on the endogenous resident cells, is gaining in interest. This review aims to summarize the knowledge on the secretome of mesenchymal stem cells, including knowledge from other tissues, in order to indicate how these mechanisms may be of value in repairing articular cartilage defects. Potential therapies and their effects on the repair of articular cartilage defects will be discussed, with a focus on the transition from classical cell therapy to the implantation of cell free matrices releasing specific cytokines.
Collapse
|
33
|
Swan AL, Stekel DJ, Hodgman C, Allaway D, Alqahtani MH, Mobasheri A, Bacardit J. A machine learning heuristic to identify biologically relevant and minimal biomarker panels from omics data. BMC Genomics 2015; 16 Suppl 1:S2. [PMID: 25923811 PMCID: PMC4315157 DOI: 10.1186/1471-2164-16-s1-s2] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Investigations into novel biomarkers using omics techniques generate large amounts of data. Due to their size and numbers of attributes, these data are suitable for analysis with machine learning methods. A key component of typical machine learning pipelines for omics data is feature selection, which is used to reduce the raw high-dimensional data into a tractable number of features. Feature selection needs to balance the objective of using as few features as possible, while maintaining high predictive power. This balance is crucial when the goal of data analysis is the identification of highly accurate but small panels of biomarkers with potential clinical utility. In this paper we propose a heuristic for the selection of very small feature subsets, via an iterative feature elimination process that is guided by rule-based machine learning, called RGIFE (Rule-guided Iterative Feature Elimination). We use this heuristic to identify putative biomarkers of osteoarthritis (OA), articular cartilage degradation and synovial inflammation, using both proteomic and transcriptomic datasets. RESULTS AND DISCUSSION Our RGIFE heuristic increased the classification accuracies achieved for all datasets when no feature selection is used, and performed well in a comparison with other feature selection methods. Using this method the datasets were reduced to a smaller number of genes or proteins, including those known to be relevant to OA, cartilage degradation and joint inflammation. The results have shown the RGIFE feature reduction method to be suitable for analysing both proteomic and transcriptomics data. Methods that generate large 'omics' datasets are increasingly being used in the area of rheumatology. CONCLUSIONS Feature reduction methods are advantageous for the analysis of omics data in the field of rheumatology, as the applications of such techniques are likely to result in improvements in diagnosis, treatment and drug discovery.
Collapse
Affiliation(s)
- Anna L Swan
- School of Biosciences, Faculty of Science, University of Nottingham, Sutton Bonington Campus, Leicestershire, LE12 5RD, United Kingdom
| | - Dov J Stekel
- School of Biosciences, Faculty of Science, University of Nottingham, Sutton Bonington Campus, Leicestershire, LE12 5RD, United Kingdom
| | - Charlie Hodgman
- School of Biosciences, Faculty of Science, University of Nottingham, Sutton Bonington Campus, Leicestershire, LE12 5RD, United Kingdom
- The D-BOARD European Consortium for Biomarker Discovery, The Universities of Surrey, Nottingham and Newcastle, United Kingdom
| | - David Allaway
- WALTHAM® Centre for Pet Nutrition, Waltham-on-the-Wolds, Melton Mowbray, Leicestershire, LE14 4RT, United Kingdom
| | - Mohammed H Alqahtani
- Center of Excellence in Genomic Medicine Research (CEGMR), King AbdulAziz University, Jeddah, 21589, Kingdom of Saudi Arabia
| | - Ali Mobasheri
- The D-BOARD European Consortium for Biomarker Discovery, The Universities of Surrey, Nottingham and Newcastle, United Kingdom
- School of Veterinary Medicine, Faculty of Health and Medical Sciences, University of Surrey, Duke of Kent Building, Guildford, Surrey, GU2 7XH, United Kingdom
- Center of Excellence in Genomic Medicine Research (CEGMR), King AbdulAziz University, Jeddah, 21589, Kingdom of Saudi Arabia
- Arthritis Research UK Centre for Sport, Exercise, and Osteoarthritis, Arthritis Research UK Pain Centre, Medical Research Council-Arthritis Research UK Centre for Musculoskeletal Ageing Research, Faculty of Medicine and Health Sciences, University of Nottingham, University Park, Nottingham, NG7 2RD, United Kingdom
| | - Jaume Bacardit
- The D-BOARD European Consortium for Biomarker Discovery, The Universities of Surrey, Nottingham and Newcastle, United Kingdom
- The Interdisciplinary Computing and Complex BioSystems (ICOS) research group, School of Computing Science, Newcastle University, Claremont Tower, Newcastle-upon-Tyne, NE1 7RU, United Kingdom
| |
Collapse
|
34
|
Abstract
Tissue engineering holds promise for the treatment of damaged and diseased tissues, especially for those tissues that do not undergo repair and regeneration readily in situ. Many techniques are available for cell and tissue culturing and differentiation of chondrocytes using a variety of cell types, differentiation methods, and scaffolds. In each case, it is critical to demonstrate the cellular phenotype and tissue composition, with particular attention to the extracellular matrix molecules that play a structural role and that contribute to the mechanical properties of the resulting tissue construct. Mass spectrometry provides an ideal analytical method with which to characterize the full spectrum of proteins produced by tissue-engineered cartilage. Using normal cartilage tissue as a standard, tissue-engineered cartilage can be optimized according to the entire proteome. Proteomic analysis is a complementary approach to biochemical, immunohistochemical, and mechanical testing of cartilage constructs. Proteomics is applicable as an analysis approach to most cartilage constructs generated from a variety of cellular sources including primary chondrocytes, mesenchymal stem cells from bone marrow, adipose tissue, induced pluripotent stem cells, and embryonic stem cells. Additionally, proteomics can be used to optimize novel scaffolds and bioreactor applications, yielding cartilage tissue with the proteomic profile of natural cartilage.
Collapse
Affiliation(s)
- Xinzhu Pu
- Department of Biological Sciences, Biomolecular Research Center, Boise State University, Boise, ID, USA
| | - Julia Thom Oxford
- Department of Biological Sciences, Biomolecular Research Center, Boise State University, 1910 University Drive, Mail Stop 1511, Boise, ID, USA.
| |
Collapse
|
35
|
Cillero-Pastor B, Eijkel GB, Blanco FJ, Heeren RMA. Protein classification and distribution in osteoarthritic human synovial tissue by matrix-assisted laser desorption ionization mass spectrometry imaging. Anal Bioanal Chem 2014; 407:2213-22. [DOI: 10.1007/s00216-014-8342-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Revised: 11/07/2014] [Accepted: 11/12/2014] [Indexed: 12/26/2022]
|
36
|
Lourido L, Calamia V, Mateos J, Fernández-Puente P, Fernández-Tajes J, Blanco FJ, Ruiz-Romero C. Quantitative proteomic profiling of human articular cartilage degradation in osteoarthritis. J Proteome Res 2014; 13:6096-106. [PMID: 25383958 DOI: 10.1021/pr501024p] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Osteoarthritis (OA) is the most common rheumatic pathology and is characterized primarily by articular cartilage degradation. Despite its high prevalence, there is no effective therapy to slow disease progression or regenerate the damaged tissue. Therefore, new diagnostic and monitoring tests for OA are urgently needed, which would also promote the development of alternative therapeutic strategies. In the present study, we have performed an iTRAQ-based quantitative proteomic analysis of secretomes from healthy human articular cartilage explants, comparing their protein profile to those from unwounded (early disease) and wounded (advanced disease) zones of osteoarthritic tissue. This strategy allowed us to identify a panel of 76 proteins that are distinctively released by the diseased tissue. Clustering analysis allowed the classification of proteins according to their different profile of release from cartilage. Among these proteins, the altered release of osteoprotegerin (decreased in OA) and periostin (increased in OA), both involved in bone remodelling processes, was verified in further analyses. Moreover, periostin was also increased in the synovial fluid of OA patients. Altogether, the present work provides a novel insight into the mechanisms of human cartilage degradation and a number of new cartilage-characteristic proteins with possible biomarker value for early diagnosis and prognosis of OA.
Collapse
Affiliation(s)
- Lucía Lourido
- Proteomics Group-PBR2-ProteoRed/ISCIII, Rheumatology Division, §RIER-RED de Inflamación y Enfermedades Reumáticas, ∥CIBER-BBN Instituto de Salud Carlos III, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas, Universidade da Coruña (UDC) , As Xubias, 84, 15006-A Coruña, Spain
| | | | | | | | | | | | | |
Collapse
|
37
|
Williams A, Smith JR, Allaway D, Harris P, Liddell S, Mobasheri A. Carprofen inhibits the release of matrix metalloproteinases 1, 3, and 13 in the secretome of an explant model of articular cartilage stimulated with interleukin 1β. Arthritis Res Ther 2014; 15:R223. [PMID: 24373218 PMCID: PMC3978949 DOI: 10.1186/ar4424] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2013] [Accepted: 12/05/2013] [Indexed: 12/24/2022] Open
Abstract
INTRODUCTION Arthritic diseases are characterized by the degradation of collagenous and noncollagenous extracellular matrix (ECM) components in articular cartilage. The increased expression and activity of matrix metalloproteinases (MMPs) is partly responsible for cartilage degradation. This study used proteomics to identify inflammatory proteins and catabolic enzymes released in a serum-free explant model of articular cartilage stimulated with the pro-inflammatory cytokine interleukin 1β (IL-1β). Western blotting was used to quantify the release of selected proteins in the presence or absence of the cyclooxygenase-2 specific nonsteroidal pro-inflammatory drug carprofen. METHODS Cartilage explant cultures were established by using metacarpophalangeal joints from horses euthanized for purposes other than research. Samples were treated as follows: no treatment (control), IL-1β (10 ng/ml), carprofen (100 μg/ml), and carprofen (100 μg/ml) + IL-1β (10 ng/ml). Explants were incubated (37°C, 5% CO2) over twelve day time courses. High-throughput nano liquid chromatography/mass spectrometry/mass spectrometry uncovered candidate proteins for quantitative western blot analysis. Proteoglycan loss was assessed by using the dimethylmethylene blue (DMMB) assay, which measures the release of sulfated glycosaminoglycans (GAGs). RESULTS Mass spectrometry identified MMP-1, -3, -13, and the ECM constituents thrombospondin-1 (TSP-1) and fibronectin-1 (FN1). IL-1β stimulation increased the release of all three MMPs. IL-1β also stimulated the fragmentation of FN1 and increased chondrocyte cell death (as assessed by β-actin release). Addition of carprofen significantly decreased MMP release and the appearance of a 60 kDa fragment of FN1 without causing any detectable cytotoxicity to chondrocytes. DMMB assays suggested that carprofen initially inhibited IL-1β-induced GAG release, but this effect was transient. Overall, during the two time courses, GAG release was 58.67% ± 10.91% (SD) for IL-1β versus 52.91% ± 9.35% (SD) with carprofen + IL-1β. CONCLUSIONS Carprofen exhibits beneficial anti-inflammatory and anti-catabolic effects in vitro without causing any detectable cytotoxicity. Combining proteomics with this explant model provides a sensitive screening system for anti-inflammatory compounds.
Collapse
|
38
|
Abstract
Our friend and colleague, Dr. Dick Heinegård, contributed greatly to the understanding of joint tissue biochemistry, the discovery and validation of arthritis-related biomarkers and the establishment of methodology for proteomic studies in osteoarthritis (OA). To date, discovery of OA-related biomarkers has focused on cartilage, synovial fluid and serum. Methods, such as affinity depletion and hyaluronidase treatment have facilitated proteomics discovery research from these sources. Osteoarthritis usually involves multiple joints; this characteristic makes it easier to detect OA with a systemic biomarker but makes it hard to delineate abnormalities of individual affected joints. Although the abundance of cartilage proteins in urine may generally be lower than other tissue/sample sources, the protein composition of urine is much less complex and its collection is non-invasive thereby facilitating the development of patient friendly biomarkers. To date however, relatively few proteomics studies have been conducted in OA urine. Proteomics strategies have identified many proteins that may relate to pathological mechanisms of OA. Further targeted approaches to validate the role of these proteins in OA are needed. Herein we summarize recent proteomic studies related to joint tissues and the cohorts used; a clear understanding of the cohorts is important for this work as we expect that the decisive discoveries of OA-related biomarkers rely on comprehensive phenotyping of healthy non-OA and OA subjects. Besides the common phenotyping criteria that include, gender, age, and body mass index (BMI), it is essential to collect data on symptoms and signs of OA outside the index joints and to bolster this with objective imaging data whenever possible to gain the most precise appreciation of the total burden of disease. Proteomic studies on systemic biospecimens, such as serum and urine, rely on comprehensive phenotyping data to unravel the true meaning of the proteomic results.
Collapse
|
39
|
Ceciliani F, Restelli L, Lecchi C. Proteomics in farm animals models of human diseases. Proteomics Clin Appl 2014; 8:677-88. [PMID: 24595991 DOI: 10.1002/prca.201300080] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Revised: 11/18/2013] [Accepted: 01/14/2014] [Indexed: 12/21/2022]
Abstract
The need to provide in vivo complex environments to understand human diseases strongly relies on the use of animal models, which traditionally include small rodents and rabbits. It is becoming increasingly evident that the few species utilised to date cannot be regarded as universal. There is a great need for new animal species that are naturally endowed with specific features relevant to human diseases. Farm animals, including pigs, cows, sheep and horses, represent a valid alternative to commonly utilised rodent models. There is an ample scope for the application of proteomic techniques in farm animals, and the establishment of several proteomic maps of plasma and tissue has clearly demonstrated that farm animals provide a disease environment that closely resembles that of human diseases. The present review offers a snapshot of how proteomic techniques have been applied to farm animals to improve their use as biomedical models. Focus will be on specific topics of biomedical research in which farm animal models have been characterised through the application of proteomic techniques.
Collapse
Affiliation(s)
- Fabrizio Ceciliani
- Department of Veterinary Sciences and Public Health, Università di Milano, Milan, Italy; Interdepartmental Centre for Studies on Mammary Gland, Università di Milano, Milan, Italy
| | | | | |
Collapse
|
40
|
Bundgaard L, Jacobsen S, Sørensen MA, Sun Z, Deutsch EW, Moritz RL, Bendixen E. The Equine PeptideAtlas: a resource for developing proteomics-based veterinary research. Proteomics 2014; 14:763-73. [PMID: 24436130 DOI: 10.1002/pmic.201300398] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2013] [Revised: 12/13/2013] [Accepted: 12/15/2013] [Indexed: 11/11/2022]
Abstract
Progress in MS-based methods for veterinary research and diagnostics is lagging behind compared to the human research, and proteome data of domestic animals is still not well represented in open source data repositories. This is particularly true for the equine species. Here we present a first Equine PeptideAtlas encompassing high-resolution tandem MS analyses of 51 samples representing a selection of equine tissues and body fluids from healthy and diseased animals. The raw data were processed through the Trans-Proteomic Pipeline to yield high quality identification of proteins and peptides. The current release comprises 24 131 distinct peptides representing 2636 canonical proteins observed at false discovery rates of 0.2% at the peptide level and 1.4% at the protein level. Data from the Equine PeptideAtlas are available for experimental planning, validation of new datasets, and as a proteomic data mining resource. The advantages of the Equine PeptideAtlas are demonstrated by examples of mining the contents for information on potential and well-known equine acute phase proteins, which have extensive general interest in the veterinary clinic. The extracted information will support further analyses, and emphasizes the value of the Equine PeptideAtlas as a resource for the design of targeted quantitative proteomic studies.
Collapse
Affiliation(s)
- Louise Bundgaard
- Department of Large Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Molecular Biology and Genetics, Faculty of Science and Technology, Aarhus University, Aarhus, Denmark
| | | | | | | | | | | | | |
Collapse
|
41
|
Xu J, Zhang C, Song W. Screening of differentially expressed genes associated with non-union skeletal fractures and analysis with a DNA microarray. Exp Ther Med 2014; 7:609-614. [PMID: 24520254 PMCID: PMC3919922 DOI: 10.3892/etm.2014.1478] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2013] [Accepted: 12/02/2013] [Indexed: 12/15/2022] Open
Abstract
The purpose of this study was to identify the feature genes that are associated with non-union skeletal fractures using samples of normal union and non-union skeletal fracture microarray data. The gene expression profile GSE494 was downloaded from the Gene Expression Omnibus database and included 12 samples based on three different platforms (GPL92, GPL93 and GPL8300). Each of the platforms had four sets of expression data, two from normal union skeletal fracture samples and two from non-union skeletal fracture samples. The differentially expressed genes within the three platforms of expression data were identified using packages in R language and the differentially expressed genes common to the three platforms were selected. The selected common differentially expressed genes were further analyzed using bioinformatic methods. The software HitPredict was used to search interactions of the common differentially expressed genes and then FuncAssociate was used to conduct a functional analysis of the genes in the interaction network. Further, the associated pathways were identified using the software WebGestalt. Under the three different platforms, GPL92, GPL93 and GPL8300, the numbers of differentially expressed genes identified were 531, 418 and 914, respectively. The common gene CLU and its interacting genes were most significantly associated with the regulation of sterol transport and the osteoclast differentiation pathway. Upregulation of the gene CLU was identified by comparing data for normal union and non-union skeletal fracture samples. According to the function of CLU and its interacting genes, it was concluded that they inhibit the normal healing process following a fracture, and result in non-union skeletal fractures through the regulation of sterol transport and the pathways of differentiation in osteoclasts.
Collapse
Affiliation(s)
- Jiaming Xu
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University, Xuhui, Shanghai 200233, P.R. China
| | - Changqing Zhang
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University, Xuhui, Shanghai 200233, P.R. China
| | - Wenqi Song
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University, Xuhui, Shanghai 200233, P.R. China
| |
Collapse
|
42
|
Swan AL, Hillier KL, Smith JR, Allaway D, Liddell S, Bacardit J, Mobasheri A. Analysis of mass spectrometry data from the secretome of an explant model of articular cartilage exposed to pro-inflammatory and anti-inflammatory stimuli using machine learning. BMC Musculoskelet Disord 2013; 14:349. [PMID: 24330474 PMCID: PMC3878677 DOI: 10.1186/1471-2474-14-349] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2013] [Accepted: 10/09/2013] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Osteoarthritis (OA) is an inflammatory disease of synovial joints involving the loss and degeneration of articular cartilage. The gold standard for evaluating cartilage loss in OA is the measurement of joint space width on standard radiographs. However, in most cases the diagnosis is made well after the onset of the disease, when the symptoms are well established. Identification of early biomarkers of OA can facilitate earlier diagnosis, improve disease monitoring and predict responses to therapeutic interventions. METHODS This study describes the bioinformatic analysis of data generated from high throughput proteomics for identification of potential biomarkers of OA. The mass spectrometry data was generated using a canine explant model of articular cartilage treated with the pro-inflammatory cytokine interleukin 1 β (IL-1β). The bioinformatics analysis involved the application of machine learning and network analysis to the proteomic mass spectrometry data. A rule based machine learning technique, BioHEL, was used to create a model that classified the samples into their relevant treatment groups by identifying those proteins that separated samples into their respective groups. The proteins identified were considered to be potential biomarkers. Protein networks were also generated; from these networks, proteins pivotal to the classification were identified. RESULTS BioHEL correctly classified eighteen out of twenty-three samples, giving a classification accuracy of 78.3% for the dataset. The dataset included the four classes of control, IL-1β, carprofen, and IL-1β and carprofen together. This exceeded the other machine learners that were used for a comparison, on the same dataset, with the exception of another rule-based method, JRip, which performed equally well. The proteins that were most frequently used in rules generated by BioHEL were found to include a number of relevant proteins including matrix metalloproteinase 3, interleukin 8 and matrix gla protein. CONCLUSIONS Using this protocol, combining an in vitro model of OA with bioinformatics analysis, a number of relevant extracellular matrix proteins were identified, thereby supporting the application of these bioinformatics tools for analysis of proteomic data from in vitro models of cartilage degradation.
Collapse
Affiliation(s)
- Anna L Swan
- School of Biosciences, Faculty of Science, University of Nottingham, Sutton Bonington Campus, Leicestershire, LE12 5RD, UK
| | - Kirsty L Hillier
- Musculoskeletal Research Group, School of Veterinary Medicine and Science, Faculty of Medicine and Health Science, University of Nottingham, Sutton Bonington Campus, Leicestershire, LE12 5RD, UK
| | | | - David Allaway
- WALTHAM® Centre for Pet Nutrition, Waltham-on-the-Wolds, Melton Mowbray, Leicestershire, LE14 4RT, UK
| | - Susan Liddell
- School of Biosciences, Faculty of Science, University of Nottingham, Sutton Bonington Campus, Leicestershire, LE12 5RD, UK
- Proteomics Laboratory, School of Biosciences, University of Nottingham, Sutton Bonington Campus, Leicestershire, LE12 5RD, UK
- The D-BOARD European Consortium for Biomarker Discovery, University of Nottingham, University Park, Nottingham, NG7 2RD, UK
| | - Jaume Bacardit
- School of Computer Science, University of Nottingham, Jubilee Campus, Nottingham, NG8 1BB, UK
- The D-BOARD European Consortium for Biomarker Discovery, University of Nottingham, University Park, Nottingham, NG7 2RD, UK
- School of Computing Science, Newcastle University, Claremont Tower, Newcastle-upon-Tyne, NE1 7RU, UK
| | - Ali Mobasheri
- Musculoskeletal Research Group, School of Veterinary Medicine and Science, Faculty of Medicine and Health Science, University of Nottingham, Sutton Bonington Campus, Leicestershire, LE12 5RD, UK
- The D-BOARD European Consortium for Biomarker Discovery, University of Nottingham, University Park, Nottingham, NG7 2RD, UK
- Arthritis Research UK Centre for Sport, Exercise and Osteoarthritis, Nottingham University Hospitals, Nottingham, NG7 2UH, UK
- Arthritis Research UK Pain Centre, The University of Nottingham, Queen's Medical Centre, Nottingham, NG7 2UH, UK
- Medical Research Council and Arthritis Research UK Centre for Musculoskeletal Ageing Research, The University of Nottingham, Queen’s Medical Centre, Nottingham, NG7 2UH, UK
- Center of Excellence in Genomic Medicine Research (CEGMR), King Fahad Medical Research Center (KFMRC), King AbdulAziz University, Jeddah, 21589, Kingdom of Saudi Arabia
- Schools of Pharmacy and Life Sciences, University of Bradford, Richmond Road, Bradford, BD7 1DP, UK
- Comparative Physiology, Medical Research Council-Arthritis Research UK Centre for Musculoskeletal Ageing Research, Arthritis Research UK Pain Centre, Arthritis Research UK Centre for Sport, Exercise, and Osteoarthritis, Faculty of Medicine and Health Sciences, The University of Nottingham, Sutton Bonington Campus, Leicestershire, LE12 5RD, UK
- Faculty of Medicine and Health Sciences, The University of Nottingham, Sutton Bonington Campus, Leicestershire, LE12 5RD, UK
| |
Collapse
|
43
|
Peffers MJ, Beynon RJ, Clegg PD. Absolute quantification of selected proteins in the human osteoarthritic secretome. Int J Mol Sci 2013; 14:20658-81. [PMID: 24132152 PMCID: PMC3821636 DOI: 10.3390/ijms141020658] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2013] [Revised: 08/19/2013] [Accepted: 09/23/2013] [Indexed: 12/27/2022] Open
Abstract
Osteoarthritis (OA) is characterized by a loss of extracellular matrix which is driven by catabolic cytokines. Proteomic analysis of the OA cartilage secretome enables the global study of secreted proteins. These are an important class of molecules with roles in numerous pathological mechanisms. Although cartilage studies have identified profiles of secreted proteins, quantitative proteomics techniques have been implemented that would enable further biological questions to be addressed. To overcome this limitation, we used the secretome from human OA cartilage explants stimulated with IL-1β and compared proteins released into the media using a label-free LC-MS/MS-based strategy. We employed QconCAT technology to quantify specific proteins using selected reaction monitoring. A total of 252 proteins were identified, nine were differentially expressed by IL-1 β stimulation. Selected protein candidates were quantified in absolute amounts using QconCAT. These findings confirmed a significant reduction in TIMP-1 in the secretome following IL-1β stimulation. Label-free and QconCAT analysis produced equivocal results indicating no effect of cytokine stimulation on aggrecan, cartilage oligomeric matrix protein, fibromodulin, matrix metalloproteinases 1 and 3 or plasminogen release. This study enabled comparative protein profiling and absolute quantification of proteins involved in molecular pathways pertinent to understanding the pathogenesis of OA.
Collapse
Affiliation(s)
- Mandy J. Peffers
- Department of Musculoskeletal Biology, Institute of Ageing and Chronic Disease, University of Liverpool, Leahurst, Chester High Road, Neston, Cheshire, CH64 7TE, UK; E-Mail:
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +44-151-795-6006; Fax: +44-151-795-6101
| | - Robert J. Beynon
- Protein Function Group, Institute of Integrative Biology, University of Liverpool, Biosciences Building, Crown Street, Liverpool, L69 7ZB, UK; E-Mail:
| | - Peter D. Clegg
- Department of Musculoskeletal Biology, Institute of Ageing and Chronic Disease, University of Liverpool, Leahurst, Chester High Road, Neston, Cheshire, CH64 7TE, UK; E-Mail:
| |
Collapse
|
44
|
Abstract
Advancement in electrophoresis and mass spectrometry techniques along with the recent progresses in genomics, culminating in bovine and pig genome sequencing, widened the potential application of proteomics in the field of veterinary medicine. The aim of the present review is to provide an in-depth perspective about the application of proteomics to animal disease pathogenesis, as well as its utilization in veterinary diagnostics. After an overview on the various proteomic techniques that are currently applied to veterinary sciences, the article focuses on proteomic approaches to animal disease pathogenesis. Included as well are recent achievements in immunoproteomics (ie, the identifications through proteomic techniques of antigen involved in immune response) and histoproteomics (ie, the application of proteomics in tissue processed for immunohistochemistry). Finally, the article focuses on clinical proteomics (ie, the application of proteomics to the identification of new biomarkers of animal diseases).
Collapse
|
45
|
Cillero-Pastor B, Eijkel GB, Kiss A, Blanco FJ, Heeren RMA. Matrix-assisted laser desorption ionization-imaging mass spectrometry: a new methodology to study human osteoarthritic cartilage. ACTA ACUST UNITED AC 2013; 65:710-20. [PMID: 23280504 DOI: 10.1002/art.37799] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2012] [Accepted: 11/08/2012] [Indexed: 01/08/2023]
Abstract
OBJECTIVE Information about the distribution of proteins and the modulation that they undergo in the different phases of rheumatic pathologies is essential to understanding the development of these diseases. We undertook this study to demonstrate the utility of mass spectrometry (MS)-based molecular imaging for studying the spatial distribution of different components in human articular cartilage sections. METHODS We compared the distribution of peptides and proteins in human control and osteoarthritic (OA) cartilage. Human control and OA cartilage slices were cut and deposited on conductive slides. After tryptic digestion, we performed matrix-assisted laser desorption ionization-imaging MS (MALDI-IMS) experiments in a MALDI-quadrupole time-of-flight mass spectrometer. Protein identification was undertaken with a combination of multivariate statistical methods and Mascot protein database queries. Hematoxylin and eosin staining and immunohistochemistry were performed to validate the results. RESULTS We created maps of peptide distributions at 150-μm raster size from control and OA human cartilage. Proteins such as biglycan, prolargin, decorin, and aggrecan core protein were identified and localized. Specific protein markers for cartilage oligomeric matrix protein and fibronectin were found exclusively in OA cartilage samples. Their distribution displayed a stronger intensity in the deep area than in the superficial area. New tentative OA markers were found in the deep area of the OA cartilage. CONCLUSION MALDI-IMS identifies and localizes disease-specific peptides and proteins in cartilage. All the OA-related peptides and proteins detected display a stronger intensity in the deep cartilage. MS-based molecular imaging is demonstrated to be an innovative method for studying OA pathology.
Collapse
|
46
|
Furse S, Liddell S, Ortori CA, Williams H, Neylon DC, Scott DJ, Barrett DA, Gray DA. The lipidome and proteome of oil bodies from Helianthus annuus (common sunflower). J Chem Biol 2013; 6:63-76. [PMID: 23532185 PMCID: PMC3606697 DOI: 10.1007/s12154-012-0090-1] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2012] [Accepted: 12/28/2012] [Indexed: 12/02/2022] Open
Abstract
In this paper we report the molecular profiling, lipidome and proteome, of the plant organelle known as an oil body (OB). The OB is remarkable in that it is able to perform its biological role (storage of triglycerides) whilst resisting the physical stresses caused by changes during desiccation (dehydration) and germination (rehydration). The molecular profile that confers such extraordinary physical stability on OBs was determined using a combination of 31P/1H nuclear magnetic resonance (NMR), high-resolution mass spectrometry and nominal mass-tandem mass spectrometry for the lipidome, and gel-electrophoresis-chromatography-tandem mass spectrometry for the proteome. The integrity of the procedure for isolating OBs was supported by physical evidence from small-angle neutron-scattering experiments. Suppression of lipase activity was crucial in determining the lipidome. There is conclusive evidence that the latter is dominated by phosphatidylcholine (∼60 %) and phosphatidylinositol (∼20 %), with a variety of other head groups (∼20 %). The fatty acid profile of the surface monolayer comprised palmitic, linoleic and oleic acids (2:1:0.25, 1H NMR) with only traces of other fatty acids (C24:0, C22:0, C18:0, C18:3, C16:2; by MS). The proteome is rich in oleosins (78 %) with the remainder being made up of caleosins and steroleosins. These data are sufficiently detailed to inform an update of the understood model of this organelle and can be used to inform the use of such components in a range of molecular biological, biotechnological and food industry applications. The techniques used in this study for profiling the lipidome throw a new light on the lipid profile of plant cellular compartments.
Collapse
Affiliation(s)
- Samuel Furse
- School of Biosciences, University of Nottingham, College Lane, Sutton Bonington, Nottinghamshire, LE12 5RD UK
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Brown KJ, Formolo CA, Seol H, Marathi RL, Duguez S, An E, Pillai D, Nazarian J, Rood BR, Hathout Y. Advances in the proteomic investigation of the cell secretome. Expert Rev Proteomics 2013; 9:337-45. [PMID: 22809211 DOI: 10.1586/epr.12.21] [Citation(s) in RCA: 90] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Studies of the cell secretome have greatly increased in recent years owing to improvements in proteomic platforms, mass spectrometry instrumentation and to the increased interaction between analytical chemists, biologists and clinicians. Several secretome studies have been implemented in different areas of research, leading to the generation of a valuable secretome catalogs. Secreted proteins continue to be an important source of biomarkers and therapeutic target discovery and are equally valuable in the field of microbiology. Several discoveries have been achieved in vitro using cell culture systems, ex vivo using human tissue specimens and in vivo using animal models. In this review, some of the most recent advances in secretome studies and the fields that have benefited the most from this evolving technology are highlighted.
Collapse
Affiliation(s)
- Kristy J Brown
- Children's National Medical Center, Center for Genetic Medicine Research, 111 Michigan Avenue NW, Washington, DC 20010, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Abstract
PURPOSE OF REVIEW Aging is a primary risk factor for the development of osteoarthritis and the understanding of how aging processes contribute to the development of osteoarthritis is an important area of active research. The most recent literature in this area was reviewed in order to update investigators on the status of the field. RECENT FINDINGS The field is beginning to move beyond a cartilage focus to include other joint tissues relevant to osteoarthritis such as ligaments, meniscus, and bone. Synovitis also appears to play a role in osteoarthritis but has not been a focus of aging studies. Studies in small animals, including mice and rats, demonstrate age-related changes that can contribute to osteoarthritis and show that animal age is a key factor to be considered in interpreting the results of studies using surgically induced models of osteoarthritis. There is accumulating evidence that cellular processes such as damage-induced cell senescence contribute to osteoarthritis and a growing body of literature on the role of epigenetic regulation of gene expression in aging and osteoarthritis. SUMMARY Not all osteoarthritis is due to aging processes in joint tissues, but the age-related changes being discovered certainly could play a major contributing role.
Collapse
Affiliation(s)
- Richard F Loeser
- Department of Internal Medicine, Section of Molecular Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157, USA.
| |
Collapse
|
49
|
Calamia V, Lourido L, Fernández-Puente P, Mateos J, Rocha B, Montell E, Vergés J, Ruiz-Romero C, Blanco FJ. Secretome analysis of chondroitin sulfate-treated chondrocytes reveals anti-angiogenic, anti-inflammatory and anti-catabolic properties. Arthritis Res Ther 2012; 14:R202. [PMID: 23031212 PMCID: PMC3580514 DOI: 10.1186/ar4040] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2012] [Accepted: 10/02/2012] [Indexed: 02/06/2023] Open
Abstract
INTRODUCTION Chondroitin sulfate (CS) is a symptomatic slow-acting drug for osteoarthritis (OA) widely used in the clinic. The aim of this work is to find proteins whose secretion from cartilage cells under proinflammatory stimuli (IL-1β) is regulated by CS, employing a novel quantitative proteomic approach. METHODS Human articular chondrocytes released from three normal cartilages were grown in SILAC medium. When complete incorporation of the heavy isotope was achieved, chondrocytes were stimulated with IL-1β 5 ng/ml with or without CS pretreatment (200 µg/ml). Forty-eight hours later, chondrocyte secretomes were analyzed by nano-scale liquid chromatography-mass spectrometry. Real-time PCR, western blot and immunohistochemistry analyses were employed to confirm some of the results. RESULTS We could identify 75 different proteins in the secretome of human articular chondrocytes. Eighteen of these were modulated by CS with statistical significance (six increased and 12 decreased). In normal chondrocytes stimulated with IL-1β, CS reduces inflammation directly by decreasing the presence of several complement components (CFAB, C1S, CO3, and C1R) and also indirectly by increasing proteins such as TNFα-induced protein (TSG6). TSG6 overexpression correlates with a decrease in pro-matrix metalloproteinase activation (observed in MMP1 and MMP3 levels). Finally, we observed a strong CS-dependent increase of an angiogenesis inhibitor, thrombospondin-1. CONCLUSION We have generated a quantitative profile of chondrocyte extracellular protein changes driven by CS in the presence of IL-1β. We have also provided novel evidences of its anti-angiogenic, anti-inflammatory, and anti-catabolic properties. Demonstration of the anti-angiogenic action of CS might provide a novel therapeutic approach for OA targeting.
Collapse
|
50
|
Gharbi M, Deberg M, Henrotin Y. Application for proteomic techniques in studying osteoarthritis: a review. Front Physiol 2011; 2:90. [PMID: 22144964 PMCID: PMC3228966 DOI: 10.3389/fphys.2011.00090] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2011] [Accepted: 11/11/2011] [Indexed: 01/12/2023] Open
Abstract
After the genomic era, proteomic corresponds to a wide variety of techniques that study the protein content of cells, tissue, or organism and that allow the isolation of protein of interest. It offers the choice between gel-based and gel-free methods or shotgun proteomics. Applications of proteomic technology may concern three principal objectives in several biomedical or clinical domains of research as in osteoarthritis: (i) to understand the physiopathology or underlying mechanisms leading to a disease or associated with a particular model, (ii), to find disease-specific biomarker, and (iii) to identify new therapeutic targets. This review aimed at gathering most of the data regarding the proteomic techniques and their applications to osteoarthritis research. It also reported technical limitations and solutions, as for example for sample preparation. Proteomics open wide perspectives in biochemical research but many technical matters still remain to be solved.
Collapse
|