1
|
Mustonen AM, Palviainen M, Säisänen L, Karttunen L, Tollis S, Esrafilian A, Reijonen J, Julkunen P, Siljander PRM, Kröger H, Mäki J, Arokoski J, Nieminen P. Tetraspanin profiles of serum extracellular vesicles reflect functional limitations and pain perception in knee osteoarthritis. Arthritis Res Ther 2024; 26:33. [PMID: 38254142 PMCID: PMC10801950 DOI: 10.1186/s13075-023-03234-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 12/11/2023] [Indexed: 01/24/2024] Open
Abstract
BACKGROUND Emerging evidence suggests that extracellular vesicles (EVs) can play roles in inflammatory processes and joint degradation in primary osteoarthritis (OA), a common age-associated joint disease. EV subpopulations express tetraspanins and platelet markers that may reflect OA pathogenesis. The present study investigated the associations between these EV surface markers and articular cartilage degradation, subjectively and objectively assessed pain, and functional limitations in primary knee OA (KOA). METHODS Serum EVs were determined by high-sensitivity flow cytometry (large CD61+ EVs) and single particle interferometric reflectance imaging sensor (small CD41+, CD63+, CD81+, and CD9+ EVs) from end-stage KOA patients and controls (n = 8 per group). Knee pain and physical functions were assessed with several health- and pain-related questionnaires, established measurements of physical medicine, and neuromuscular examination. The obtained data were analyzed using supervised and unsupervised univariate and multivariate models. RESULTS With the combined dataset of cartilage thickness, knee function, pain, sensation, and EV molecular signatures, we identified highly correlated groups of variables and found several EV markers that were statistically significant predictors of pain, physical limitations, and other aspects of well-being for KOA patients, for instance CD41+/CD63+/CD9+ small EVs associated with the range of motion of the knee, physical performance, and pain sensitivity. CONCLUSIONS Particular serum EV subpopulations showed clear associations with KOA pain and functional limitations, suggesting that their implications in OA pathophysiology warrant further study.
Collapse
Affiliation(s)
- Anne-Mari Mustonen
- Institute of Biomedicine, School of Medicine, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland.
- Department of Environmental and Biological Sciences, Faculty of Science, Forestry and Technology, University of Eastern Finland, Joensuu, Finland.
| | - Mari Palviainen
- EV core and EV group, Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences University of Helsinki, Helsinki, Finland
| | - Laura Säisänen
- Department of Clinical Neurophysiology, Kuopio University Hospital, Kuopio, Finland
- Department of Technical Physics, Faculty of Science, Forestry and Technology, University of Eastern Finland, Kuopio, Finland
| | - Lauri Karttunen
- Department of Physical and Rehabilitation Medicine, Central Finland Hospital Nova, Jyväskylä, Finland
| | - Sylvain Tollis
- Institute of Biomedicine, School of Medicine, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland.
| | - Amir Esrafilian
- Department of Technical Physics, Faculty of Science, Forestry and Technology, University of Eastern Finland, Kuopio, Finland
| | - Jusa Reijonen
- Department of Clinical Neurophysiology, Kuopio University Hospital, Kuopio, Finland
- Department of Technical Physics, Faculty of Science, Forestry and Technology, University of Eastern Finland, Kuopio, Finland
| | - Petro Julkunen
- Department of Clinical Neurophysiology, Kuopio University Hospital, Kuopio, Finland
- Department of Technical Physics, Faculty of Science, Forestry and Technology, University of Eastern Finland, Kuopio, Finland
| | - Pia R-M Siljander
- EV core and EV group, Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences University of Helsinki, Helsinki, Finland
| | - Heikki Kröger
- Department of Orthopaedics, Traumatology and Hand Surgery, Kuopio University Hospital, Kuopio, Finland
- Kuopio Musculoskeletal Research Unit, University of Eastern Finland, Kuopio, Finland
| | - Jussi Mäki
- Department of Rehabilitation, Kuopio University Hospital, Kuopio, Finland
| | - Jari Arokoski
- Department of Physical and Rehabilitation Medicine, Helsinki University Hospital, Helsinki, Finland and University of Helsinki, Helsinki, Finland
| | - Petteri Nieminen
- Institute of Biomedicine, School of Medicine, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
2
|
Yu SP, Deveza LA, Kraus VB, Karsdal M, Bay-Jensen AC, Collins JE, Guermazi A, Roemer FW, Ladel C, Bhagavath V, Hunter DJ. Association of biochemical markers with bone marrow lesion changes on imaging-data from the Foundation for the National Institutes of Health Osteoarthritis Biomarkers Consortium. Arthritis Res Ther 2024; 26:30. [PMID: 38238803 PMCID: PMC10795356 DOI: 10.1186/s13075-023-03253-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 12/27/2023] [Indexed: 01/22/2024] Open
Abstract
BACKGROUND To assess the prognostic value of short-term change in biochemical markers as it relates to bone marrow lesions (BMLs) on MRI in knee osteoarthritis (OA) over 24 months and, furthermore, to assess the relationship between biochemical markers involved with tissue turnover and inflammation and BMLs on MRI. METHODS Data from the Foundation for the National Institutes of Health OA Biomarkers Consortium within the Osteoarthritis Initiative (n = 600) was analyzed. BMLs were measured according to the MRI Osteoarthritis Knee Score (MOAKS) system (0-3), in 15 knee subregions. Serum and urinary biochemical markers assessed were as follows: serum C-terminal crosslinked telopeptide of type I collagen (CTX-I), serum crosslinked N-telopeptide of type I collagen (NTX-I), urinary CTX-Iα and CTX-Iβ, urinary NTX-I, urinary C-terminal cross-linked telopeptide of type II collagen (CTX-II), serum matrix metalloproteinase (MMP)-degraded type I, II, and III collagen (C1M, C2M, C3M), serum high sensitivity propeptide of type IIb collagen (hsPRO-C2), and matrix metalloproteinase-generated neoepitope of C-reactive protein (CRPM). The association between change in biochemical markers over 12 months and BMLs over 24 months was examined using regression models adjusted for covariates. The relationship between C1M, C2M, C3M, hsPRO-C2, and CRPM and BMLs at baseline and over 24 months was examined. RESULTS Increases in serum CTX-I and urinary CTX-Iβ over 12 months were associated with increased odds of changes in the number of subregions affected by any BML at 24 months. Increase in hsPRO-C2 was associated with decreased odds of worsening in the number of subregions affected by any BML over 24 months. C1M and C3M were associated with BMLs affected at baseline. CONCLUSIONS Short-term changes in serum CTX-I, hsPRO-C2, and urinary CTX-Iβ hold the potential to be prognostic of BML progression on MRI. The association of C1M and C3M with baseline BMLs on MRI warrants further investigation.
Collapse
Affiliation(s)
- Shirley P Yu
- Department of Rheumatology, Royal North Shore Hospital, Reserve Road, St Leonards, Sydney, NSW, 2065, Australia.
- Sydney Musculoskeletal Health, The Kolling Institute, School of Medicine, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia.
| | - Leticia A Deveza
- Department of Rheumatology, Royal North Shore Hospital, Reserve Road, St Leonards, Sydney, NSW, 2065, Australia
- Sydney Musculoskeletal Health, The Kolling Institute, School of Medicine, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Virginia B Kraus
- Department of Medicine, Duke University School of Medicine, Durham, NC, USA
| | | | | | - Jamie E Collins
- Orthopaedic and Arthritis Centre for Outcomes Research, Brigham and Women's Hospital, Boston, MA, USA
| | - Ali Guermazi
- Chobanian & Avedisian School of Medicine, Boston University, Boston, MA, USA
| | - Frank W Roemer
- Chobanian & Avedisian School of Medicine, Boston University, Boston, MA, USA
- Department of Radiology, Universitätsklinikum Erlangen & Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | | | - Venkatesha Bhagavath
- Sydney Musculoskeletal Health, The Kolling Institute, School of Medicine, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
- Northern Sydney Local Health District, Royal North Shore Hospital, St Leonards, Sydney, NSW, Australia
| | - David J Hunter
- Department of Rheumatology, Royal North Shore Hospital, Reserve Road, St Leonards, Sydney, NSW, 2065, Australia
- Sydney Musculoskeletal Health, The Kolling Institute, School of Medicine, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
3
|
Wang Y, Han X, Shi J, Liao Z, Zhang Y, Li Y, Jiang M, Liu M. Distinct Metabolites in Osteopenia and Osteoporosis: A Systematic Review and Meta-Analysis. Nutrients 2023; 15:4895. [PMID: 38068753 PMCID: PMC10708105 DOI: 10.3390/nu15234895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 11/04/2023] [Accepted: 11/12/2023] [Indexed: 12/18/2023] Open
Abstract
Multiple studies have indicated that distinct metabolites are involved in the occurrence and development of osteopenia (ON) and osteoporosis (OP); however, these metabolites in OP and ON have not yet been classified and standardized. This systematic review and meta-analysis included 21 articles aiming to investigate the distinct metabolites in patients with ON and OP. The quality of the included articles was generally high; seventeen studies had >7 stars, and the remaining four received 6 stars. This systematic review showed that three metabolites (phosphatidylcholine (PC) (lipid metabolites), galactose (carbohydrate metabolites), and succinic acid (other metabolites)) increased, four (glycylglycine (gly-gly), cystine (amino acids), sphingomyelin (SM) (lipid metabolites) and glucose (carbohydrate metabolites)) decreased, and five (glutamine, hydroxyproline, taurine (amino acids), lysophosphatidylcholine (LPC) (lipid metabolites), and lactate (other metabolites)) had conflicting directions in OP/ON. The results of the meta-analysis show that gly-gly (MD = -0.77, 95%CI -1.43 to -0.11, p = 0.02) and cystine (MD = -5.52, 95%CI -7.35 to -3.68, p < 0.00001) decreased in the OP group compared with the healthy control group. Moreover, LPC (MD = 1.48, 95%CI 0.11 to 2.86, p = 0.03) increased in the OP group compared with the healthy control group. These results indicate that distinct metabolites were associated with ON and OP, which could be considered a predictor for OP.
Collapse
Affiliation(s)
- Yuhe Wang
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China; (Y.W.); (J.S.); (Z.L.); (Y.Z.); (Y.L.)
| | - Xu Han
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing 100700, China;
| | - Jingru Shi
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China; (Y.W.); (J.S.); (Z.L.); (Y.Z.); (Y.L.)
| | - Zeqi Liao
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China; (Y.W.); (J.S.); (Z.L.); (Y.Z.); (Y.L.)
| | - Yuanyue Zhang
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China; (Y.W.); (J.S.); (Z.L.); (Y.Z.); (Y.L.)
| | - Yuanyuan Li
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China; (Y.W.); (J.S.); (Z.L.); (Y.Z.); (Y.L.)
| | - Miao Jiang
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing 100700, China;
| | - Meijie Liu
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China; (Y.W.); (J.S.); (Z.L.); (Y.Z.); (Y.L.)
| |
Collapse
|
4
|
Sluka KA, Wager TD, Sutherland SP, Labosky PA, Balach T, Bayman EO, Berardi G, Brummett CM, Burns J, Buvanendran A, Caffo B, Calhoun VD, Clauw D, Chang A, Coffey CS, Dailey DL, Ecklund D, Fiehn O, Fisch KM, Frey Law LA, Harris RE, Harte SE, Howard TD, Jacobs J, Jacobs JM, Jepsen K, Johnston N, Langefeld CD, Laurent LC, Lenzi R, Lindquist MA, Lokshin A, Kahn A, McCarthy RJ, Olivier M, Porter L, Qian WJ, Sankar CA, Satterlee J, Swensen AC, Vance CG, Waljee J, Wandner LD, Williams DA, Wixson RL, Zhou XJ. Predicting chronic postsurgical pain: current evidence and a novel program to develop predictive biomarker signatures. Pain 2023; 164:1912-1926. [PMID: 37326643 PMCID: PMC10436361 DOI: 10.1097/j.pain.0000000000002938] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 03/02/2023] [Accepted: 03/02/2023] [Indexed: 06/17/2023]
Abstract
ABSTRACT Chronic pain affects more than 50 million Americans. Treatments remain inadequate, in large part, because the pathophysiological mechanisms underlying the development of chronic pain remain poorly understood. Pain biomarkers could potentially identify and measure biological pathways and phenotypical expressions that are altered by pain, provide insight into biological treatment targets, and help identify at-risk patients who might benefit from early intervention. Biomarkers are used to diagnose, track, and treat other diseases, but no validated clinical biomarkers exist yet for chronic pain. To address this problem, the National Institutes of Health Common Fund launched the Acute to Chronic Pain Signatures (A2CPS) program to evaluate candidate biomarkers, develop them into biosignatures, and discover novel biomarkers for chronification of pain after surgery. This article discusses candidate biomarkers identified by A2CPS for evaluation, including genomic, proteomic, metabolomic, lipidomic, neuroimaging, psychophysical, psychological, and behavioral measures. Acute to Chronic Pain Signatures will provide the most comprehensive investigation of biomarkers for the transition to chronic postsurgical pain undertaken to date. Data and analytic resources generatedby A2CPS will be shared with the scientific community in hopes that other investigators will extract valuable insights beyond A2CPS's initial findings. This article will review the identified biomarkers and rationale for including them, the current state of the science on biomarkers of the transition from acute to chronic pain, gaps in the literature, and how A2CPS will address these gaps.
Collapse
Affiliation(s)
- Kathleen A. Sluka
- Department of Physical Therapy and Rehabilitation Science, Carver College of Medicine, University of Iowa, Iowa City, IA
| | - Tor D. Wager
- Department of Psychological and Brain Sciences, Dartmouth College, Hanover, NH
| | - Stephani P. Sutherland
- Department of Biostatistics, Johns Hopkins Bloomberg Schools of Public Health, Baltimore, MD
| | - Patricia A. Labosky
- Office of Strategic Coordination, Division of Program Coordination, Planning and Strategic Initiatives, Office of the Director, National Institutes of Health, Bethesda, MD
| | - Tessa Balach
- Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago, Chicago, IL
| | - Emine O. Bayman
- Clinical Trials and Data Management Center, Department of Biostatistics, University of Iowa, Iowa City, IA
| | - Giovanni Berardi
- Department of Physical Therapy and Rehabilitation Science, Carver College of Medicine, University of Iowa, Iowa City, IA
| | - Chad M. Brummett
- Department of Anesthesiology, University of Michigan Medical School, Ann Arbor, MI
| | - John Burns
- Division of Behavioral Sciences, Rush Medical College, Chicago, IL
| | | | - Brian Caffo
- Department of Biostatistics, Johns Hopkins Bloomberg Schools of Public Health, Baltimore, MD
| | - Vince D. Calhoun
- Tri-Institutional Center for Translational Research in Neuroimaging and Data Science (TReNDS), Georgia State, Georgia Tech, and Emory University, Atlanta, GA
| | - Daniel Clauw
- Department of Anesthesiology, University of Michigan Medical School, Ann Arbor, MI
| | - Andrew Chang
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI
| | - Christopher S. Coffey
- Clinical Trials and Data Management Center, Department of Biostatistics, University of Iowa, Iowa City, IA
| | - Dana L. Dailey
- Department of Physical Therapy and Rehabilitation Science, Carver College of Medicine, University of Iowa, Iowa City, IA
| | - Dixie Ecklund
- Clinical Trials and Data Management Center, Department of Biostatistics, University of Iowa, Iowa City, IA
| | - Oliver Fiehn
- University of California, Davis, Davis, CA, United States
| | - Kathleen M. Fisch
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California San Diego, San Diego, CA, United States
- Center for Computational Biology and Bioinformatics, University of California San Diego, San Diego, CA, United States
| | - Laura A. Frey Law
- Department of Physical Therapy and Rehabilitation Science, Carver College of Medicine, University of Iowa, Iowa City, IA
| | - Richard E. Harris
- Department of Anesthesiology, University of Michigan Medical School, Ann Arbor, MI
| | - Steven E. Harte
- Department of Anesthesiology, University of Michigan Medical School, Ann Arbor, MI
| | - Timothy D. Howard
- Department of Biochemistry, Center for Precision Medicine, Wake Forest School of Medicine, Winstom-Salem, NC
- Center for Precision Medicine, Wake Forest School of Medicine, Winstom-Salem, NC
| | - Joshua Jacobs
- Department of Orthopedic Surgery, Rush Medical College, CHicago, IL
| | - Jon M. Jacobs
- Environmental and Molecular Sciences Laboratory, Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA
| | | | | | - Carl D. Langefeld
- Center for Precision Medicine, Wake Forest School of Medicine, Winstom-Salem, NC
- Department of Biostatistics and Data Science, Center for Precision Medicine, Wake Forest School of Medicine, Winstom-Salem, NC
| | - Louise C. Laurent
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California San Diego, San Diego, CA, United States
| | - Rebecca Lenzi
- Office of Strategic Coordination, Division of Program Coordination, Planning and Strategic Initiatives, Office of the Director, National Institutes of Health, Bethesda, MD
| | - Martin A. Lindquist
- Department of Biostatistics, Johns Hopkins Bloomberg Schools of Public Health, Baltimore, MD
| | | | - Ari Kahn
- Texas Advanced Computing Center, University of Texas, AUstin, TX
| | | | - Michael Olivier
- Center for Precision Medicine, Wake Forest School of Medicine, Winstom-Salem, NC
- Department of Internal Medicine, Center for Precision Medicine, Wake Forest School of Medicine, Winstom-Salem, NC
| | - Linda Porter
- National Institute of Neurological Disorders and Stroke, Bethesda, MD
- Office of Pain Policy and Planning National Institutes of Health, Bethesda, MD
| | - Wei-Jun Qian
- Environmental and Molecular Sciences Laboratory, Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA
| | - Cheryse A. Sankar
- National Institute of Neurological Disorders and Stroke, Bethesda, MD
| | | | - Adam C. Swensen
- Environmental and Molecular Sciences Laboratory, Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA
| | - Carol G.T. Vance
- Department of Physical Therapy and Rehabilitation Science, Carver College of Medicine, University of Iowa, Iowa City, IA
| | - Jennifer Waljee
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI
| | - Laura D. Wandner
- National Institute of Neurological Disorders and Stroke, Bethesda, MD
| | - David A. Williams
- Department of Anesthesiology, University of Michigan Medical School, Ann Arbor, MI
| | | | - Xiaohong Joe Zhou
- Center for MR Research and Departments of Radiology, Neurosurgery, and Bioengineering, University of Illinois College of Medicine at Chicago, Chicago, IL, United States
| |
Collapse
|
5
|
Kalogera S, Jansen MP, Bay-Jensen AC, Frederiksen P, Karsdal MA, Thudium CS, Mastbergen SC. Relevance of Biomarkers in Serum vs. Synovial Fluid in Patients with Knee Osteoarthritis. Int J Mol Sci 2023; 24:ijms24119483. [PMID: 37298434 DOI: 10.3390/ijms24119483] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 05/25/2023] [Accepted: 05/26/2023] [Indexed: 06/12/2023] Open
Abstract
The association between structural changes and pain sensation in osteoarthritis (OA) remains unclear. Joint deterioration in OA leads to the release of protein fragments that can either systemically (serum) or locally (synovial fluid; SF) be targeted as biomarkers and describe structural changes and potentially pain. Biomarkers of collagen type I (C1M), type II (C2M), type III (C3M), type X (C10C), and aggrecan (ARGS) degradation were measured in the serum and SF of knee OA patients. Spearman's rank correlation was used to assess the correlation of the biomarkers' levels between serum and SF. Linear regression adjusted for confounders was used to evaluate the associations between the biomarkers' levels and clinical outcomes. The serum C1M levels were negatively associated with subchondral bone density. The serum C2M levels were negatively associated with KL grade and positively associated with minimum joint space width (minJSW). The C10C levels in SF were negatively associated with minJSW and positively associated with KL grade and osteophyte area. Lastly, the serum C2M and C3M levels were negatively associated with pain outcomes. Most of the biomarkers seemed to mainly be associated with structural outcomes. The overall biomarkers of extracellular matrix (ECM) remodeling in serum and SF may provide different information and reflect different pathogenic processes.
Collapse
Affiliation(s)
- Stefania Kalogera
- Nordic Bioscience, Biomarkers and Research, 2730 Herlev, Denmark
- Department of Drug Design and Pharmacology, Copenhagen University, 1165 Copenhagen, Denmark
| | - Mylène P Jansen
- Department of Rheumatology & Clinical Immunology, UMC Utrecht, Utrecht University, 3584 CX Utrecht, The Netherlands
| | | | | | - Morten A Karsdal
- Nordic Bioscience, Biomarkers and Research, 2730 Herlev, Denmark
| | | | - Simon C Mastbergen
- Department of Rheumatology & Clinical Immunology, UMC Utrecht, Utrecht University, 3584 CX Utrecht, The Netherlands
- Regenerative Medicine Center, Utrecht University, 3584 CS Utrecht, The Netherlands
| |
Collapse
|
6
|
Afzali MF, Pannone SC, Martinez RB, Campbell MA, Sanford JL, Pezzanite L, Kurihara J, Johnson V, Dow SW, Santangelo KS. Intravenous injection of adipose-derived mesenchymal stromal cells benefits gait and inflammation in a spontaneous osteoarthritis model. J Orthop Res 2023; 41:902-912. [PMID: 36030381 PMCID: PMC9968820 DOI: 10.1002/jor.25431] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 07/05/2022] [Accepted: 08/22/2022] [Indexed: 02/04/2023]
Abstract
Osteoarthritis (OA) is a leading cause of morbidity among aging populations, yet symptom and/or disease-modification remains elusive. Adipose-derived mesenchymal stromal cells (adMSCs) have demonstrated immunomodulatory and anti-inflammatory properties that may alleviate clinical signs and interrupt disease onset and progression. Indeed, multiple manuscripts have evaluated intra-articular administration of adMSCs as a therapeutic; however, comparatively few evaluations of systemic delivery methods have been published. Therefore, the aim of this study was to evaluate the short-term impact of intravenous (IV) delivery of allogeneic adMSCs in an established model of spontaneous OA, the Hartley guinea pig. Animals with moderate OA received once weekly injections of 2 × 106 adMSCs or vehicle control for 4 weeks in peripheral veins; harvest occurred 2 weeks after the final injection. Systemic administration of adMSCs resulted in no adverse effects and was efficacious in reducing clinical signs of OA (as assessed by computer-aided gait analysis) compared to control injected animals. Further, there were significant decreases in key inflammatory mediators (including monocyte chemoattractant protein-1, tumor necrosis factor, and prostaglandin E2 ) both systemically (liver, kidney, and serum) and locally in the knee (joint tissues and synovial fluid) in animals treated with IV adMSCs relative to controls (as per enzyme-linked immunosorbent assay and/or immunohistochemistry, dictated by tissue sample). Thus, systemic administration of adMSCs by IV injection significantly improved gait parameters and reduced both systemic and intra-articular inflammatory mediators in animals with OA. These findings demonstrate the potential utility of alternative delivery approaches for cellular therapy of OA, particularly for patients with multiple affected joints.
Collapse
Affiliation(s)
- Maryam F. Afzali
- Department of Microbiology, Immunology & Pathology, Colorado State University, Fort Collins, CO
| | - Stephen C. Pannone
- Department of Microbiology, Immunology & Pathology, Colorado State University, Fort Collins, CO
| | - Richard B. Martinez
- Department of Microbiology, Immunology & Pathology, Colorado State University, Fort Collins, CO
| | - Margaret A Campbell
- Department of Microbiology, Immunology & Pathology, Colorado State University, Fort Collins, CO
| | - Joseph L. Sanford
- Department of Microbiology, Immunology & Pathology, Colorado State University, Fort Collins, CO
| | - Lynn Pezzanite
- Department of Clinical Sciences College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO
| | - Jade Kurihara
- Department of Clinical Sciences College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO
| | - Valerie Johnson
- Department of Clinical Sciences College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, East Lansing, MI
| | - Steven W. Dow
- Department of Clinical Sciences College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO
| | - Kelly S. Santangelo
- Department of Microbiology, Immunology & Pathology, Colorado State University, Fort Collins, CO
| |
Collapse
|
7
|
Duarte FCK, Chien R, Ghazinour G, Murnaghan K, West DWD, Kumbhare DA. Myofascial Pain as an Unseen Comorbidity in Osteoarthritis: A Scoping Review. Clin J Pain 2023; 39:188-201. [PMID: 36943163 DOI: 10.1097/ajp.0000000000001102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 01/25/2023] [Indexed: 03/23/2023]
Abstract
OBJECTIVE This review aimed to identify, summarize, and appraise the evidence supporting the coexistence of myofascial pain (MPS) and trigger points (MTrP) in osteoarthritis (OA), and the effectiveness of MTrPs treatments in OA-related pain and physical function outcomes. METHODS Three databases were searched from inception to June 2022. We included observational and experimental studies to fulfill our 2 study aims. Two independent reviewers conducted 2-phase screening procedures and risk of bias using checklist tools for cross-sectional, quasi-experimental, and randomized control trials. Patient characteristics, findings of active and latent MTrPs in relevant muscles, treatments, and pain and physical function outcomes were extracted from low-risk bias studies. RESULTS The literature search yielded 2898 articles, of which 6 observational and 7 experimental studies had a low bias risk and the data extracted. Active MTrPs in knee OA patients was more evident in the quadriceps and hamstring muscles than in healthy individuals. Dry needling on active MTrPs improved pain and physical function in the short term compared with sham treatment in hip OA patients. In knee OA, dry needling on latent or active MTrPs improved pain and functional outcomes compared with sham needling but did not result in better pain and physical outcomes when combined with a physical exercise program. DISCUSSION The presence of active versus latent MTrPs seems to be a more sensitive discriminating feature of OA given that latent is often present in OA and healthy individuals. Dry needling on active MTrPs improved pain and physical function in the short term compared with sham treatment in hip OA patients. However, the small sample size and the few number of studies limit any firm recommendation on the treatment. REGISTRY The study protocol was prospectively registered in Open Science Framework (https://doi.org/10.17605/OSF.IO/8DVU3).
Collapse
Affiliation(s)
- Felipe C K Duarte
- Canadian Memorial Chiropractic College
- KITE Research, Toronto Rehabilitation Institute, University Health Network
- Discipline of Chiropractic, School of Health, Medical and Applied Sciences, CQUniversity, Brisbane, Australia
| | | | - Golnaz Ghazinour
- Faculty of Kinesiology and Physical Education, University of Toronto
| | | | - Daniel W D West
- Faculty of Kinesiology and Physical Education, University of Toronto
- Discipline of Chiropractic, School of Health, Medical and Applied Sciences, CQUniversity, Brisbane, Australia
| | - Dinesh A Kumbhare
- Faculty of Kinesiology and Physical Education, University of Toronto
- Division of Physical Medicine and Rehabilitation, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
- Discipline of Chiropractic, School of Health, Medical and Applied Sciences, CQUniversity, Brisbane, Australia
| |
Collapse
|
8
|
An Observational Study on Chronic Pain Biomarkers in Fibromyalgia and Osteoarthritis Patients: Which Role for Mu Opioid Receptor’s Expression on NK Cells? Biomedicines 2023; 11:biomedicines11030931. [PMID: 36979910 PMCID: PMC10046119 DOI: 10.3390/biomedicines11030931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 02/28/2023] [Accepted: 03/14/2023] [Indexed: 03/19/2023] Open
Abstract
The evaluation of chronic pain is challenging because of the lack of specific biomarkers. We identified the Mu opioid receptor-positive (Mu+) B cell percentage of expression, named Mu-Lympho-Marker (MLM), as a candidate marker for chronic pain in fibromyalgia (FM) and osteoarthritis (OA) patients. Here, we investigate the role of MLM on natural killer (NK) cells in the same patients. Twenty-nine FM and twelve OA patients were analyzed, and twenty-three pain-free subjects were considered as the control group. Blood samples were collected to perform immunophenotyping and Western blot analysis. Biological and clinical data were statistically analyzed. The final results showed that the percentage of NK cells expressing Mu was statistically lower in FM and OA patients than in pain-free subjects, as already demonstrated for B cells. A Western blot analysis was performed in order to detect NK cells’ functional status. Moreover, the correlation analysis of MLM expression with pharmacological therapy did not show any significant results. In conclusion, here, we confirm the role of MLM as a suitable marker for chronic pain and underline NK cells as a new possible immune cell type involved in the “Mu opioid receptor reserve theory”.
Collapse
|
9
|
Biomarkers for Osteoarthritis Diseases. Life (Basel) 2022; 12:life12111799. [PMID: 36362955 PMCID: PMC9697481 DOI: 10.3390/life12111799] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 11/01/2022] [Accepted: 11/04/2022] [Indexed: 11/09/2022] Open
Abstract
Growing evidence has revealed the pivotal role of inflammatory biomarkers in the pathogenesis of osteoarthritis. There is significant interest in the prognostic value of select biomarkers, given the potential for early identification and treatment of patients at risk of osteoarthritis prior to the development of irreversible clinical disease. Clinical trials of novel therapeutics that disrupt the inflammatory pathways of osteoarthritis are also ongoing. The purpose of this review is to summarize the current literature on key biomarkers within the context of osteoarthritis pathogenesis, clinical symptom development, and treatment capabilities. Multiple recent studies have established biomarkers that signal the existence of osteoarthritis pathology and the development of clinical symptomology. However, prior to implementation in clinical practice, additional research is required to precisely define the prognostic value for numerous biomarkers and standardize their measurement. Biomarker-driven investigations represent a promising avenue for the early diagnosis and treatment of osteoarthritis.
Collapse
|
10
|
Szilagyi IA, Vallerga CL, Boer CG, Schiphof D, Ikram MA, Bierma-Zeinstra SMA, van Meurs JBJ. Plasma proteomics identifies CRTAC1 as a biomarker for osteoarthritis severity and progression. Rheumatology (Oxford) 2022; 62:1286-1295. [PMID: 35924962 PMCID: PMC9977119 DOI: 10.1093/rheumatology/keac415] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 07/07/2022] [Accepted: 07/07/2022] [Indexed: 11/14/2022] Open
Abstract
OBJECTIVES The aim of this study was to identify biomarkers for radiographic OA severity and progression acting within the inflammation and metabolic pathways. METHODS For 3517 Rotterdam Study participants, 184 plasma protein levels were measured using Olink inflammation and cardiometabolic panels. We studied associations with severity and progression of knee, hip and hand OA and a composite overall OA burden score by multivariable regression models, adjusting for age, sex, cell counts and BMI. RESULTS We found 18 significantly associated proteins for overall OA burden, of which 5 stayed significant after multiple testing correction: circulating cartilage acidic protein 1 (CRTAC1), cartilage oligomeric matrix protein (COMP), thrombospondin 4, IL-18 receptor 1 (IL-18R1) and TNF ligand superfamily member 14. These proteins were also associated with progression of knee OA, with the exception of IL-18R1. The strongest association was found for the level of CRTAC1, with 1 s.d. increase in protein level resulting in an increase of 0.09 (95% CI 0.06, 0.12) in the overall OA Kellgren-Lawrence sum score (P = 2.9 × 10-8) in the model adjusted for age, sex, BMI and cell counts. This association was also present with the severity of OA in all three joints and progression of knee OA and was independent of BMI. We observed a stronger association for CRTAC1 with OA than for the well-known OA biomarker COMP. CONCLUSION We identified several compelling biomarkers reflecting the overall OA burden and the increased risk for OA progression. CRTAC1 was the most compelling and robust biomarker for OA severity and progression. Such a biomarker may be used for disease monitoring.
Collapse
Affiliation(s)
| | | | | | | | - M Arfan Ikram
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | | | - Joyce B J van Meurs
- Correspondence to: Prof. dr. Joyce B. J. van Meurs, Department of Internal Medicine, Erasmus MC University Medical Center, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands. E-mail:
| |
Collapse
|
11
|
Zhang Y, Wang Q, Liang J, Liu L, Liu P, Zhao H. Serum proteomic analysis of differentially expressed proteins and pathways involved in the mechanism of endemic osteoarthritis. Mol Omics 2022; 18:745-753. [PMID: 35820143 DOI: 10.1039/d2mo00154c] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Kashin-Beck disease (KBD) is a chronic and endemic osteochondral disease and the etiology and pathogenic mechanism of KBD are still unknown. This study aimed to elucidate and screen KBD-associated proteins, which were differentially expressed between KBD patients and healthy controls. We combined protein fractionation and liquid chromatography-tandem mass spectrometry (LC-MS/MS) with a high-resolution mass spectrometer coupled with tandem mass tags (TMTs) to quantitatively analyze and screen KBD-associated proteins, which were differentially expressed between KBD patients and healthy controls. In addition, we used parallel reaction monitoring (PRM) to quantify proteins in serum from patients with KBD and healthy controls in order to verify the differentially expressed proteins in patients with KBD. We identified 224 differentially expressed proteins, including 11 up-regulated and 213 down-regulated proteins. Catalase (CAT) was observed to be significantly elevated in patients with KBD compared with control patients. Further, the fold difference of CAT is significantly elevated in PRM compared with label-free quantification. The results in this study suggest that CAT may be the reflection of the dynamic nature of KBD and could be considered as a novel pathogenic indicator for patients with KBD.
Collapse
Affiliation(s)
- Yan Zhang
- Foot and ankle surgery department, Honghui Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China.
| | - Qiong Wang
- Foot and ankle surgery department, Honghui Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China.
| | - Jingqi Liang
- Foot and ankle surgery department, Honghui Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China.
| | - Liang Liu
- Foot and ankle surgery department, Honghui Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China.
| | - Peilong Liu
- Foot and ankle surgery department, Honghui Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China.
| | - Hongmou Zhao
- Foot and ankle surgery department, Honghui Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China.
| |
Collapse
|
12
|
Wu C, Liang C, Sun J, Zhang Z, Pan X. Effects of Diosmin on IL-1β- Induced Inflammatory Response in Primary Rat Chondrocytes. INT J PHARMACOL 2022. [DOI: 10.3923/ijp.2022.667.672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
13
|
Kalogera S, He Y, Bay-Jensen AC, Gantzel T, Sun S, Manon-Jensen T, Karsdal MA, Thudium CS. The activation fragment of PAR2 is elevated in serum from patients with rheumatoid arthritis and reduced in response to anti-IL6R treatment. Sci Rep 2021; 11:24285. [PMID: 34930943 PMCID: PMC8688421 DOI: 10.1038/s41598-021-03346-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 11/30/2021] [Indexed: 12/29/2022] Open
Abstract
AbstractOsteoarthritis (OA) and rheumatoid arthritis (RA) are serious and painful diseases. Protease-activated receptor 2 (PAR2) is involved in the pathology of both OA and RA including roles in synovial hyperplasia, cartilage destruction, osteophyogenesis and pain. PAR2 is activated via cleavage of its N-terminus by serine proteases. In this study a competitive ELISA assay was developed targeting the 36-amino acid peptide that is cleaved and released after PAR2 activation (PRO-PAR2). Technical assay parameters including antibody specificity, intra- and inter-assay variation (CV%), linearity, accuracy, analyte stability and interference were evaluated. PRO-PAR2 release was confirmed after in vitro cleavage of PAR2 recombinant protein and treatment of human synovial explants with matriptase. Serum levels of 22 healthy individuals, 23 OA patients and 15 RA patients as well as a subset of RA patients treated with tocilizumab were evaluated. The PRO-PAR2 antibody was specific for the neo-epitope and intra-inter assay CV% were 6.4% and 5.8% respectively. In vitro cleavage and matriptase treated explants showed increased PRO-PAR2 levels compared to controls. In serum, PRO-PAR2 levels were increased in RA patients and decreased in RA patients treated with tocilizumab. In conclusion, PRO-PAR2 may be a potential biomarker for monitoring RA disease and pharmacodynamics of treatment.
Collapse
|
14
|
Jeon M, Jagodnik KM, Kropiwnicki E, Stein DJ, Ma'ayan A. Prioritizing Pain-Associated Targets with Machine Learning. Biochemistry 2021; 60:1430-1446. [PMID: 33606503 DOI: 10.1021/acs.biochem.0c00930] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
While hundreds of genes have been associated with pain, much of the molecular mechanisms of pain remain unknown. As a result, current analgesics are limited to few clinically validated targets. Here, we trained a machine learning (ML) ensemble model to predict new targets for 17 categories of pain. The model utilizes features from transcriptomics, proteomics, and gene ontology to prioritize targets for modulating pain. We focused on identifying novel G-protein-coupled receptors (GPCRs), ion channels, and protein kinases because these proteins represent the most successful drug target families. The performance of the model to predict novel pain targets is 0.839 on average based on AUROC, while the predictions for arthritis had the highest accuracy (AUROC = 0.929). The model predicts hundreds of novel targets for pain; for example, GPR132 and GPR109B are highly ranked GPCRs for rheumatoid arthritis. Overall, gene-pain association predictions cluster into three groups that are enriched for cytokine, calcium, and GABA-related cell signaling pathways. These predictions can serve as a foundation for future experimental exploration to advance the development of safer and more effective analgesics.
Collapse
Affiliation(s)
- Minji Jeon
- Department of Pharmacological Sciences, Knowledge Management Center for Illuminating the Druggable Genome (KMC-IDG), Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, P.O. Box 1603, New York, New York 10029, United States
| | - Kathleen M Jagodnik
- Department of Pharmacological Sciences, Knowledge Management Center for Illuminating the Druggable Genome (KMC-IDG), Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, P.O. Box 1603, New York, New York 10029, United States
| | - Eryk Kropiwnicki
- Department of Pharmacological Sciences, Knowledge Management Center for Illuminating the Druggable Genome (KMC-IDG), Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, P.O. Box 1603, New York, New York 10029, United States
| | - Daniel J Stein
- Department of Pharmacological Sciences, Knowledge Management Center for Illuminating the Druggable Genome (KMC-IDG), Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, P.O. Box 1603, New York, New York 10029, United States
| | - Avi Ma'ayan
- Department of Pharmacological Sciences, Knowledge Management Center for Illuminating the Druggable Genome (KMC-IDG), Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, P.O. Box 1603, New York, New York 10029, United States
| |
Collapse
|
15
|
Sang W, Xue S, Jiang Y, Lu H, Zhu L, Wang C, Ma J. METTL3 involves the progression of osteoarthritis probably by affecting ECM degradation and regulating the inflammatory response. Life Sci 2021; 278:119528. [PMID: 33894271 DOI: 10.1016/j.lfs.2021.119528] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 04/08/2021] [Accepted: 04/11/2021] [Indexed: 12/18/2022]
Abstract
We aimed to identify RNA N6-methyladenosine methylation associated genes in osteoarthritis (OA), and to explore possible regulatory mechanisms of these RNA methylation associated genes. Bioinformatics analyses, including differential expression analysis, functional enrichment analysis, verification analysis, and box plot analysis, were conducted based on different datasets from OA and non-OA patients. Gene expression at mRNA and protein levels was determined by quantitative reverse transcription PCR, western blot and immunofluorescence. Interleukin 1β (IL-1β)-treated SW1353 cells was used as cell model. Lentiviral vector was used for over-expression METTL3 in vitro. CCK-8 assay kit was used to determine cell viability and inflammatory cytokines (IL-1α, IL-6, IL-8, IL-10 and TNF-α) was detected using ELISA kits. Bioinformatics analysis showed that METTL3 expression was decreased in OA group, which was confirmed in clinical samples. Expression of METTL3 was also reduced in IL-1β-treated cells. Levels of inflammatory cytokines were obviously reduced in the METTL3 overexpression group, while IL-1β treatment reversed such decrease caused by METTL3 overexpression (p < 0.05). Both METTL3 overexpression and IL-1β treatment promoted expression of p65 protein and p-ERK (p < 0.01). Additionally, increased expression of MMP1 and MMP3, and decreased expression of MMP13, TIMP-1, and TIMP-2 at both mRNA and protein levels were observed in the METTL3 overexpression group when compared with the control group (p < 0.01). Expression of m6A methylation gene METTL3 was reduced in OA. METTL3 is involved in OA probably by regulating the inflammatory response. METTL3 overexpression may affect extracellular matrix degradation in OA by adjusting the balance between TIMPs and MMPs.
Collapse
Affiliation(s)
- Weilin Sang
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 301620, China
| | - Song Xue
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 301620, China
| | - Yafei Jiang
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 301620, China
| | - Haiming Lu
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 301620, China
| | - Libo Zhu
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 301620, China
| | - Cong Wang
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 301620, China
| | - Jinzhong Ma
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 301620, China.
| |
Collapse
|
16
|
Bishnoi M, Jain A, Singla Y, Shrivastava B. Sublingual delivery of chondroitin sulfate conjugated tapentadol loaded nanovesicles for the treatment of osteoarthritis. J Liposome Res 2021; 31:30-44. [PMID: 32064982 DOI: 10.1080/08982104.2020.1730400] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 01/25/2020] [Accepted: 02/02/2020] [Indexed: 10/25/2022]
Abstract
Recent treatment approaches of osteoarthritis (OA) face a number of obstacles due to the progressive multitude of pain generators, nociceptive mechanisms, first pass mechanism, less efficacy and compromised safety. The present study was aimed to bring a novel approach for the effective management of OA, by developing sublingual targeted nanovesicles (NVs) bearing tapentadol HCl (TAP), surface modified with chondroitin sulfate (CS). Optimized nontargeted nanovesicle formulation (MB-NV) was developed by an ultrasound method, characterized as spherical in shape, nanometric in size (around 150 nm) with narrow size distribution (polydispersity index <0.5), and good entrapment efficiency (around 50%). MB-NV conjugated with CS which was confirmed by IR and 1H NMR spectroscopy. C-MB-NV showed improved pharmacokinetics parameters i.e. increased t1/2 (9.7 h), AUC (159.725 μg/mL*h), and MRT (14.99 h) of TAP than nontargeted formulation and plain drug soln. C-MB-NV in in vitro release studies proved sustained drug release pattern for more than 24 h following Higuchi model kinetics with Fickian diffusion (n ≤ 0.5).Targeted nanovesicles exhibited an improved bioavailability and enhanced analgesic activity in a disease-induced Wistar rat model which indicated the superior targeting potential of C-MB-NV exploiting CD44 receptors as mediators, overexpressed at the affected joints in the OA model. It could be a propitious approach to accustomed therapies for methodical and efficient management in advanced OA therapy.
Collapse
Affiliation(s)
- Mamta Bishnoi
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Jaipur National University, Jaipur, India
| | - Ankit Jain
- Department of Pharmaceutical Sciences, Pharmaceutics Research Projects Laboratory, Dr. Harisingh Gour University, Sagar, India
- Department of Materials Engineering, Indian Institute of Science, Bangalore, India
| | - Yashpaul Singla
- Department of Pharmaceutical Sciences, Guru Jambheshwar University of Science and Technology, Hisar, India
| | - Birendra Shrivastava
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Jaipur National University, Jaipur, India
| |
Collapse
|
17
|
Dong J, Li L, Fang X, Zang M. Exosome-Encapsulated microRNA-127-3p Released from Bone Marrow-Derived Mesenchymal Stem Cells Alleviates Osteoarthritis Through Regulating CDH11-Mediated Wnt/β-Catenin Pathway. J Pain Res 2021; 14:297-310. [PMID: 33574696 PMCID: PMC7871222 DOI: 10.2147/jpr.s291472] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 12/29/2020] [Indexed: 12/24/2022] Open
Abstract
Objective Exosome-encapsulated microRNAs (miRNAs) are being considered as either diagnostic or predictive markers in different types of diseases. Here, we discussed the effects of exosome-encapsulated miR-127-3p from bone marrow-derived mesenchymal stem cells (BM-MSCs) on osteoarthritis (OA). Methods BM-MSCs and primary chondrocytes were isolated from Sprague Dawley rats. IL-1β was utilized to treat chondrocytes to mimic an OA in vitro model, and exosomes extracted from BM-MSCs were utilized to treat chondrocytes so as to verify their protective effects on OA. Through online website prediction and experiments confirmation, we found the most significantly enriched miRNA in exosomes and elucidated the effect of this miRNA on the therapeutic effect of exosomes by interfering with its expression. Also, the genes targeted by the miRNA and the involved pathway were also found through bioinformatics analysis and experimental research, thereby probing into the protective mechanism of exosomes on chondrocytes. Results Exosomes derived from BM-MSCs restricted the IL-1β-induced chondrocytes damage. miR-127-3p was found to be enriched in exosomes, and the protective effect of exosomes was reversed by miR-127-3p inhibition. miR-127-3p targeted CDH11, and overexpressed CDH11 in chondrocytes weakened the therapeutic effect of exosomes. IL-1β treatment resulted in the activation of the Wnt/β-catenin pathway in chondrocytes. Exosomes treatment could inhibit the activation of this pathway, and overexpressed CDH11 reversed the inhibitory effect of exosomes on this pathway. Conclusion This study suggests that exosomal miR-127-3p derived from BM-MSCs inhibits CDH11 in chondrocytes, thereby blocking the Wnt/β-catenin pathway activation and relieving chondrocyte damage in OA.
Collapse
Affiliation(s)
- Jisheng Dong
- Department of Orthopedics, The Second People's Hospital of Hefei, The Affiliated Hefei Hospital of Anhui Medical University, Hefei, Anhui, 230011, People's Republic of China
| | - Li Li
- Department of Orthopedics, The Second People's Hospital of Hefei, The Affiliated Hefei Hospital of Anhui Medical University, Hefei, Anhui, 230011, People's Republic of China
| | - Xing Fang
- Department of Orthopedics, The Second People's Hospital of Hefei, The Affiliated Hefei Hospital of Anhui Medical University, Hefei, Anhui, 230011, People's Republic of China
| | - Mousheng Zang
- Department of Orthopedics, The Second People's Hospital of Hefei, The Affiliated Hefei Hospital of Anhui Medical University, Hefei, Anhui, 230011, People's Republic of China
| |
Collapse
|
18
|
Fayet M, Hagen M. Pain characteristics and biomarkers in treatment approaches for osteoarthritis pain. Pain Manag 2021; 11:59-73. [DOI: 10.2217/pmt-2020-0055] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Osteoarthritis (OA) is a progressive disease and OA pain intensity is related to ongoing pathophysiological changes. However, OA pain is complex and multimodal; its characteristics, including severity, localization and the stimuli that elicit it, can change as the disease progresses and differ greatly among patients. Understanding mechanisms underlying specific pain characteristics may help guide clinicians in choosing appropriate treatments, targeting treatments to those patients most likely to benefit. Associations have been demonstrated between biomarkers and some characteristics of OA pain, and to processes linked to the shift in pain characteristics over the course of OA. This article examines how understanding OA pain characteristics and their relation to the disease process could inform treatment choice when applying well-established treatment guidelines.
Collapse
Affiliation(s)
- Marina Fayet
- GSK Consumer Healthcare S.A., Route de l'Etraz 2, 1260, Nyon, Switzerland
| | - Martina Hagen
- GSK Consumer Healthcare S.A., Route de l'Etraz 2, 1260, Nyon, Switzerland
| |
Collapse
|
19
|
Identification of MOR-Positive B Cell as Possible Innovative Biomarker (Mu Lympho-Marker) for Chronic Pain Diagnosis in Patients with Fibromyalgia and Osteoarthritis Diseases. Int J Mol Sci 2020; 21:ijms21041499. [PMID: 32098316 PMCID: PMC7073128 DOI: 10.3390/ijms21041499] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 02/19/2020] [Accepted: 02/20/2020] [Indexed: 01/08/2023] Open
Abstract
Fibromyalgia (FM) diagnosis follows the American College of Rheumatology (ACR) criteria, based on clinical evaluation and written questionnaires without any objective diagnostic tool. The lack of specific biomarkers is a tragic aspect for FM and chronic pain diseases in general. Interestingly, the endogenous opioid system is close to the immune one because of the expression of opioid receptors on lymphocytes membrane. Here we analyzed the role of the Mu opioid receptor on B lymphocytes as a specific biomarker for FM and osteoarthritis (OA) patients. We enrolled three groups of females: FM patients, OA patients (chronic pain control group) and healthy subjects (pain-free negative control group). We collected blood samples to apply immunophenotyping analysis. Written tests were administrated for psychological analysis. Data were statistically analyzed. Final results showed that the percentage of Mu-positive B cells were statistically lower in FM and OA patients than in pain-free subjects. A low expression of Mu-positive B cell was not associated with the psychological characteristics investigated. In conclusion, here we propose the percentage of Mu-positive B cells as a biological marker for an objective diagnosis of chronic pain suffering patients, also contributing to the legitimacy of FM as a truly painful disease.
Collapse
|
20
|
Löfvall H, Katri A, Dąbrowska A, Karsdal MA, Luo Y, He Y, Manon-Jensen T, Dziegiel MH, Bay-Jensen AC, Thudium CS, Henriksen K. GPDPLQ 1237-A Type II Collagen Neo-Epitope Biomarker of Osteoclast- and Inflammation-Derived Cartilage Degradation in vitro. Sci Rep 2019; 9:3050. [PMID: 30816326 PMCID: PMC6395810 DOI: 10.1038/s41598-019-39803-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 01/31/2019] [Indexed: 01/21/2023] Open
Abstract
C-telopeptide of type II collagen (CTX-II) has been shown to be a highly relevant biomarker of cartilage degradation in human rheumatic diseases, if measured in synovial fluid or urine. However, serum or plasma CTX-II have not been demonstrated to have any clinical utility to date. Here, we describe the GPDPLQ1237 ELISA which targets the EKGPDPLQ↓ neo-epitope, an elongated version of the CTX-II neo-epitope (EKGPDP↓), speculated to be a blood-precursor of CTX-II generated by the cysteine protease cathepsin K. Human osteoclast cartilage resorption cultures as well as oncostatin M and tumour necrosis factor α-stimulated bovine cartilage explant cultures were used to validate GPDPLQ1237 biologically by treating the cultures with the cysteine protease inhibitor E-64 and/or the matrix metalloproteinase (MMP) inhibitor GM6001 to assess the potential contributions of these two protease classes to GPDPLQ1237 release. Cartilage resorption-derived GPDPLQ1237 release was inhibited by E-64 (72.1% inhibition), GM6001 (75.5%), and E-64/GM6001 (91.5%), whereas CTX-II release was inhibited by GM6001 (87.0%) but not by E-64 (5.5%). Cartilage explant GPDPLQ1237 and CTX-II release were both fully inhibited by GM6001 but were not inhibited by E-64. No clinically relevant GPDPLQ1237 reactivity was identified in human serum, plasma, or urine from healthy donors or arthritis patients. In conclusion, the GPDPLQ1237 biomarker is released during osteoclast-derived cysteine protease- and MMP-mediated cartilage degradation in vitro, whereas CTX-II release is mediated by MMPs and not by cysteine proteases, as well as from MMP-mediated cartilage degradation under a pro-inflammatory stimulus. These findings suggest that GPDPLQ1237 may be relevant in diseases with pathological osteoclast activity and cartilage degradation. Further studies are required to validate the neo-epitope in human samples.
Collapse
Affiliation(s)
- Henrik Löfvall
- Nordic Bioscience, Herlev, Denmark.,Division of Molecular Medicine and Gene Therapy, Lund Strategic Center for Stem Cell Biology, Lund, Sweden
| | - Anna Katri
- Nordic Bioscience, Herlev, Denmark.,Department of Drug Design and Pharmacology, Copenhagen University, Copenhagen, Denmark
| | | | | | | | - Yi He
- Nordic Bioscience, Herlev, Denmark
| | | | - Morten H Dziegiel
- Department of Clinical Immunology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | | | | | | |
Collapse
|
21
|
Zhang R, Guo H, Yang X, Li Z, Zhang D, Li B, Zhang D, Li Q, Xiong Y. Potential candidate biomarkers associated with osteoarthritis: Evidence from a comprehensive network and pathway analysis. J Cell Physiol 2019; 234:17433-17443. [PMID: 30820958 DOI: 10.1002/jcp.28365] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 01/21/2019] [Accepted: 01/24/2019] [Indexed: 12/16/2022]
Abstract
Osteoarthritis (OA) is one of the most common forms of arthritis world widely. Some key genes and diagnostic markers have been reported due to the development of modern molecular biology technologies. However, the etiology and pathogenesis of OA remains unknown. In this study, an integrated network and pathway analysis towards the biological function of OA-associated genes was conducted to provide valuable information to further explore the etiology and pathogenesis of OA. A total of 2,548 genes which reported a statistically significant association with OA were screened. An integrated network and pathway analysis was performed to identify the pathways and genes most associated to OA. Moreover, OA-specific protein-protein interaction (PPI) network was constructed by cytocluster based on the Molecular Complex Detection Algorithm (MCODE) to screen its candidate biomarkers. Quantitative real-time polymerase chain reaction was used to confirm the expression levels and to validate the results of MCODE cluster analysis by six genes. The pathway networks suggested that extracellular matrix (ECM) organization, collagen degradation and collagen formation showed important associations with OA. In top two PPI clusters, 61 of the OA-associated genes were included in the OA-specific PPI network, which also included 23 candidate genes that are likely to be highly associated with OA based on MCODE clusters. Analysis of mRNA showed that the expression levels of COL9A1, COL9A2, ITGA3, COL9A3, ITGA2, and LAMA1 in the peripheral blood mononuclear cells of OA patients were significantly lower than those of the normal controls (p<0.005). To our knowledge, this is the first comprehensive and systematic report based on OA-related genes demonstrating that the functional destruction of collagen in cartilage may be a very important contributing factor to OA. Quantitative detection of collagen synthesis may be of great help in early identification and prediction of OA. Maintaining the quality and quantity of collagen can be a potential target for clinical treatment of OA in the future practice.
Collapse
Affiliation(s)
- Rongqiang Zhang
- School of Public Health, Xi'an Jiaotong University Health Science Center, Key Laboratory of Trace Elements and Endemic Diseases of National Health Commission of the People's Republic of China, Xi'an, China.,Shaanxi University of Chinese Medicine, Xianyang, China
| | - Hao Guo
- School of Public Health, Xi'an Jiaotong University Health Science Center, Key Laboratory of Trace Elements and Endemic Diseases of National Health Commission of the People's Republic of China, Xi'an, China
| | - Xiaoli Yang
- School of Public Health, Xi'an Jiaotong University Health Science Center, Key Laboratory of Trace Elements and Endemic Diseases of National Health Commission of the People's Republic of China, Xi'an, China
| | - Zhaofang Li
- School of Public Health, Xi'an Jiaotong University Health Science Center, Key Laboratory of Trace Elements and Endemic Diseases of National Health Commission of the People's Republic of China, Xi'an, China
| | - Dandan Zhang
- School of Public Health, Xi'an Jiaotong University Health Science Center, Key Laboratory of Trace Elements and Endemic Diseases of National Health Commission of the People's Republic of China, Xi'an, China
| | - Baorong Li
- School of Public Health, Xi'an Jiaotong University Health Science Center, Key Laboratory of Trace Elements and Endemic Diseases of National Health Commission of the People's Republic of China, Xi'an, China
| | - Di Zhang
- School of Public Health, Xi'an Jiaotong University Health Science Center, Key Laboratory of Trace Elements and Endemic Diseases of National Health Commission of the People's Republic of China, Xi'an, China
| | - Qiang Li
- School of Public Health, Xi'an Jiaotong University Health Science Center, Key Laboratory of Trace Elements and Endemic Diseases of National Health Commission of the People's Republic of China, Xi'an, China
| | - Yongmin Xiong
- School of Public Health, Xi'an Jiaotong University Health Science Center, Key Laboratory of Trace Elements and Endemic Diseases of National Health Commission of the People's Republic of China, Xi'an, China
| |
Collapse
|
22
|
Duarte FCK, Hurtig M, Clark A, Simpson J, Srbely JZ. Association between naturally occurring spine osteoarthritis in geriatric rats and neurogenic inflammation within neurosegmentally linked skeletal muscle. Exp Gerontol 2019; 118:31-38. [PMID: 30615897 DOI: 10.1016/j.exger.2019.01.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 12/12/2018] [Accepted: 01/02/2019] [Indexed: 12/15/2022]
Abstract
OBJECTIVE This study aimed to investigate the association between naturally occurring spinal osteoarthritis (OA) (L3-L5), the expression of substance P (SP) centrally (L4-L5) and the presence of neurogenic inflammation within the neurosegmentally linked quadriceps (L2-L5) in elderly rats versus young controls. DESIGN Eight aged (27 ± 3.2 months) and six young (4 ± 0.0 months) male Wistar Kyoto rats were euthanized and submitted to micro-computerized tomography for determination of spine OA. SP expression (% area) at the dorsal horn of the spinal cord as well as the relative expression of SP and protease-activated receptor 2 (PAR2) to alpha-tubulin within quadriceps muscle were determined by immunohistochemistry and Western Blot. RESULTS Spine osteoarthritis was confirmed in all aged rats but no young controls. Aged rats expressed significant increase of SP protein expression within the dorsal horn (MD = 0.086; 95% CI [0.026 to 0.145]; p = 0.0094) and quadriceps (MD = 1.209; 95% CI [0.239 to 2.179]; p = 0.0191) and PAR2 (MD = 0.797; 95% CI [0.160 to 1.435]; p = 0.0187) compared to young controls. CONCLUSION These observations provide novel insight into the potential role of neurogenic inflammation in the pathophysiology of myofascial pain syndrome in the naturally occurring spinal OA in elderly population.
Collapse
Affiliation(s)
- Felipe C K Duarte
- Department of Human Health and Nutritional Science, University of Guelph, 50 Stone Road East, Guelph, ON N1G 2W1, Canada.
| | - Mark Hurtig
- Department of Clinical Studies, Ontario Veterinary College, University of Guelph, 50 McGilvray Lane, Guelph, ON N1G 2W1, Canada.
| | - Andrea Clark
- Department of Human Health and Nutritional Science, University of Guelph, 50 Stone Road East, Guelph, ON N1G 2W1, Canada.
| | - Jeremy Simpson
- Department of Human Health and Nutritional Science, University of Guelph, 50 Stone Road East, Guelph, ON N1G 2W1, Canada.
| | - John Z Srbely
- Department of Human Health and Nutritional Science, University of Guelph, 50 Stone Road East, Guelph, ON N1G 2W1, Canada.
| |
Collapse
|