1
|
Lisboa FSS, Benevento EM, Kaneko LO, Bertolucci V, Rosini Silva ÁA, Sardim AC, Ruiz VF, Dos Reis IGM, Porcari AM, Messias LHD. Plasma metabolites associated with biopsychosocial parameters in overweight/obese women with severe knee osteoarthritis. Front Cell Dev Biol 2024; 12:1454084. [PMID: 39296935 PMCID: PMC11408288 DOI: 10.3389/fcell.2024.1454084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 08/23/2024] [Indexed: 09/21/2024] Open
Abstract
Introduction Obesity aligned with quadriceps muscle weakness contributes to the high incidence of knee osteoarthritis (KOA), which is prevalent in women. Although molecular signatures of KOA have been suggested, the association between biopsychosocial responses and the plasma metabolomic profile in overweight/ obese women with KOA remains in its early stages of investigation. This study aims to associate the plasma metabolome with biopsychosocial parameters of overweight/obese women diagnosed with KOA. Methods Twenty-eight overweight/obese women (Control-n = 14; KOA-n = 14) underwent two visits to the laboratory. Functional tests and questionnaires assessing biopsychosocial parameters were administered during the first visit. After 48 h, the participants returned to the laboratory for blood collection. Specific to the KOA condition, the Numerical Pain Rating Scale (NPRS), Tampa Scale for Kinesiophobia (TSK), and Knee injury and Osteoarthritis Outcome Score (KOOS) were applied. Results Thirteen molecules were different between groups, and four correlated with KOA's biopsychosocial parameters. DG 22:4-2OH and gamma-Glutamylvaline were inversely associated with KOSS leisure and TSK score, respectively. LysoPE 18:0 and LysoPE 20:5 were positively associated with KOSS symptoms and TSK score, respectively. Discussion While the correlations of LysoPE 18:0 and gamma-Glutamylvaline are supported by existing literature, this is not the case for DG 22:4-2OH and LysoPE 20:5. Further studies are recommended to better elucidate these correlations before dismissing their potential involvement in the biopsychosocial factors of the disease.
Collapse
Affiliation(s)
- Fabiola Socorro Silva Lisboa
- Research Group on Technology Applied to Exercise Physiology-GTAFE, Health Sciences Postgraduate Program, São Francisco University, Bragança Paulista, Brazil
- Research Group on Musculoskeletal Rehabilitation, São Francisco University, Bragança Paulista, Brazil
| | - Enzo Martins Benevento
- Research Group on Technology Applied to Exercise Physiology-GTAFE, Health Sciences Postgraduate Program, São Francisco University, Bragança Paulista, Brazil
- Research Group on Musculoskeletal Rehabilitation, São Francisco University, Bragança Paulista, Brazil
| | - Luisa Oliveira Kaneko
- Research Group on Technology Applied to Exercise Physiology-GTAFE, Health Sciences Postgraduate Program, São Francisco University, Bragança Paulista, Brazil
| | - Vanessa Bertolucci
- Research Group on Technology Applied to Exercise Physiology-GTAFE, Health Sciences Postgraduate Program, São Francisco University, Bragança Paulista, Brazil
| | - Álex Ap Rosini Silva
- MS4Life Laboratory of Mass Spectrometry, Health Sciences Postgraduate Program, São Francisco University, Bragança Paulista, Brazil
| | - André Cabral Sardim
- Research Group on Musculoskeletal Rehabilitation, São Francisco University, Bragança Paulista, Brazil
| | - Valter Ferreira Ruiz
- Research Group on Musculoskeletal Rehabilitation, São Francisco University, Bragança Paulista, Brazil
| | - Ivan Gustavo Masseli Dos Reis
- Research Group on Technology Applied to Exercise Physiology-GTAFE, Health Sciences Postgraduate Program, São Francisco University, Bragança Paulista, Brazil
| | - Andreia M Porcari
- MS4Life Laboratory of Mass Spectrometry, Health Sciences Postgraduate Program, São Francisco University, Bragança Paulista, Brazil
| | - Leonardo Henrique Dalcheco Messias
- Research Group on Technology Applied to Exercise Physiology-GTAFE, Health Sciences Postgraduate Program, São Francisco University, Bragança Paulista, Brazil
| |
Collapse
|
2
|
Komaravolu RK, Mehta-D'souza P, Conner T, Allen M, Lumry J, Batushansky A, Pezant NP, Montgomery CG, Griffin TM. Sex-specific effects of injury and beta-adrenergic activation on metabolic and inflammatory mediators in a murine model of post-traumatic osteoarthritis. Osteoarthritis Cartilage 2024; 32:1097-1112. [PMID: 38527663 PMCID: PMC11330734 DOI: 10.1016/j.joca.2024.03.109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 02/09/2024] [Accepted: 03/19/2024] [Indexed: 03/27/2024]
Abstract
OBJECTIVE Metabolic processes are intricately linked to the resolution of innate inflammation and tissue repair, two critical steps for treating post-traumatic osteoarthritis (PTOA). Based on lipolytic and immunoregulatory actions of norepinephrine, we hypothesized that intra-articular β-adrenergic receptor (βAR) stimulation would suppress PTOA-associated inflammation in the infrapatellar fat pad (IFP) and synovium. DESIGN We used the βAR agonist isoproterenol to perturb intra-articular metabolism 3.5 weeks after applying a non-invasive single-load compression injury to knees of 12-week-old male and female mice. We examined the acute effects of intra-articular isoproterenol treatment relative to saline on IFP histology, multiplex gene expression of synovium-IFP tissue, synovial fluid metabolomics, and mechanical allodynia. RESULTS Injured knees developed PTOA pathology characterized by heterotopic ossification, articular cartilage loss, and IFP atrophy and fibrosis. Isoproterenol suppressed the upregulation of pro-fibrotic genes and downregulated the expression of adipose genes and pro-inflammatory genes (Adam17, Cd14, Icam1, Csf1r, and Casp1) in injured joints of female (but not male) mice. Analysis of published single-cell RNA-seq data identified elevated catecholamine-associated gene expression in resident-like synovial-IFP macrophages after injury. Injury substantially altered synovial fluid metabolites by increasing amino acids, peptides, sphingolipids, phospholipids, bile acids, and dicarboxylic acids, but these changes were not appreciably altered by isoproterenol. Intra-articular injection of either isoproterenol or saline increased mechanical allodynia in female mice, whereas neither substance affected male mice. CONCLUSIONS Acute βAR activation altered synovial-IFP transcription in a sex and injury-dependent manner, suggesting that women with PTOA may be more sensitive than men to treatments targeting sympathetic neural signaling pathways.
Collapse
MESH Headings
- Animals
- Female
- Male
- Mice
- Isoproterenol/pharmacology
- Adrenergic beta-Agonists/pharmacology
- Disease Models, Animal
- Sex Factors
- Synovial Membrane/metabolism
- Adipose Tissue/metabolism
- Inflammation Mediators/metabolism
- Receptors, Adrenergic, beta/metabolism
- Injections, Intra-Articular
- Knee Injuries/complications
- Knee Injuries/metabolism
- Osteoarthritis, Knee/metabolism
- Osteoarthritis, Knee/etiology
- Cartilage, Articular/metabolism
- Cartilage, Articular/drug effects
- Cartilage, Articular/pathology
- Mice, Inbred C57BL
Collapse
Affiliation(s)
- Ravi K Komaravolu
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA.
| | - Padmaja Mehta-D'souza
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA.
| | - Taylor Conner
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA.
| | - Madeline Allen
- Department of Health and Exercise Science, University of Oklahoma, Norman, OK 73019, USA.
| | - Jessica Lumry
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA.
| | - Albert Batushansky
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA.
| | - Nathan P Pezant
- Center for Biomedical Data Sciences, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA.
| | - Courtney G Montgomery
- Center for Biomedical Data Sciences, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA; Genes and Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA.
| | - Timothy M Griffin
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA; Oklahoma City VA Health Care System, Oklahoma City, OK 73104, USA; Oklahoma Center for Geroscience and the Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA.
| |
Collapse
|
3
|
Stanciugelu SI, Patrascu JM, Patrascu JM, Socaciu C, Socaciu AI, Nitusca D, Marian C. Lipidomic Signature of Plasma and Synovial Fluid in Patients with Osteoarthritis: Putative Biomarkers Determined by UHPLC-QTOF-ESI+MS. Diagnostics (Basel) 2024; 14:1834. [PMID: 39202323 PMCID: PMC11354166 DOI: 10.3390/diagnostics14161834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 08/19/2024] [Accepted: 08/20/2024] [Indexed: 09/03/2024] Open
Abstract
BACKGROUND Osteoarthritis (OA) is a prevalent joint condition causing pain and disability, especially in the elderly. Currently, OA diagnosis relies on clinical data and imaging, but recent interest in metabolomics suggests that early biochemical changes in biofluids, particularly synovial fluid (SF), could enable an earlier diagnosis and understanding of the disease. METHODS In this regard, we conducted a lipidomics study in 33 plasma and SF samples from OA patients and 20 OA-free controls to assess the diagnostic value of various lipid metabolites, using UHPLC-QTOF-ESI+MS. RESULTS In plasma samples, 25 metabolites had area-under-the-curve (AUC) values higher than 0.9, suggesting a very good diagnostic potential for phosphatidic acid PA (16:0/16:0), PA (34:0), phosphatidylethanolamine PE (34:2), glucosylceramide, phosphatidylcholine PC (32:1), and other metabolites while in SF 20, metabolites had AUC values higher than 0.8, the vast majority belonging to lipid metabolism as well. CONCLUSIONS Although the results align with the previous literature, larger cohort studies are necessary to confirm the diagnostic value of the lipid metabolites.
Collapse
Affiliation(s)
- Stefan Iulian Stanciugelu
- Doctoral School, Department of Biochemistry and Pharmacology, Victor Babes University of Medicine and Pharmacy, Pta Eftimie Murgu Nr. 2, 300041 Timisoara, Romania;
- Orthopedic and Traumatology Clinic, Timisoara County Emergency Clinical Hospital, B-dul L Rebreanu Nr. 156, 300723 Timisoara, Romania; (J.M.P.); (J.M.P.J.)
| | - Jenel Marian Patrascu
- Orthopedic and Traumatology Clinic, Timisoara County Emergency Clinical Hospital, B-dul L Rebreanu Nr. 156, 300723 Timisoara, Romania; (J.M.P.); (J.M.P.J.)
- Department of Orthopedics and Trauma, Victor Babes University of Medicine and Pharmacy, Pta Eftimie Murgu Nr. 2, 300041 Timisoara, Romania
| | - Jenel Marian Patrascu
- Orthopedic and Traumatology Clinic, Timisoara County Emergency Clinical Hospital, B-dul L Rebreanu Nr. 156, 300723 Timisoara, Romania; (J.M.P.); (J.M.P.J.)
- Department of Orthopedics and Trauma, Victor Babes University of Medicine and Pharmacy, Pta Eftimie Murgu Nr. 2, 300041 Timisoara, Romania
| | - Carmen Socaciu
- BIODIATECH, Research Center for Applied Biotechnology in Diagnosis and Molecular Therapy, 400478 Cluj-Napoca, Romania;
| | - Andreea Iulia Socaciu
- Department of Occupational Health, Iuliu Hateganu University of Medicine and Pharmacy, Str. Victor Babes Nr. 8, 400347 Cluj-Napoca, Romania;
| | - Diana Nitusca
- Department of Biochemistry and Pharmacology, Victor Babes University of Medicine and Pharmacy, Pta Eftimie Murgu Nr. 2, 300041 Timisoara, Romania;
- Center for Complex Networks Science, Victor Babes University of Medicine and Pharmacy, Pta Eftimie Murgu Nr. 2, 300041 Timisoara, Romania
| | - Catalin Marian
- Department of Biochemistry and Pharmacology, Victor Babes University of Medicine and Pharmacy, Pta Eftimie Murgu Nr. 2, 300041 Timisoara, Romania;
- Center for Complex Networks Science, Victor Babes University of Medicine and Pharmacy, Pta Eftimie Murgu Nr. 2, 300041 Timisoara, Romania
| |
Collapse
|
4
|
Timm T, Hild C, Liebisch G, Rickert M, Lochnit G, Steinmeyer J. Functional Insights into the Sphingolipids C1P, S1P, and SPC in Human Fibroblast-like Synoviocytes by Proteomic Analysis. Int J Mol Sci 2024; 25:8363. [PMID: 39125932 PMCID: PMC11313292 DOI: 10.3390/ijms25158363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 07/25/2024] [Accepted: 07/29/2024] [Indexed: 08/12/2024] Open
Abstract
The (patho)physiological function of the sphingolipids ceramide-1-phosphate (C1P), sphingosine-1-phosphate (S1P), and sphingosylphosphorylcholine (SPC) in articular joints during osteoarthritis (OA) is largely unknown. Therefore, we investigated the influence of these lipids on protein expression by fibroblast-like synoviocytes (FLSs) from OA knees. Cultured human FLSs (n = 7) were treated with 1 of 3 lipid species-C1P, S1P, or SPC-IL-1β, or with vehicle. The expression of individual proteins was determined by tandem mass tag peptide labeling followed by high-resolution electrospray ionization (ESI) mass spectrometry after liquid chromatographic separation (LC-MS/MS/MS). The mRNA levels of selected proteins were analyzed using RT-PCR. The 3sphingolipids were quantified in the SF of 18 OA patients using LC-MS/MS. A total of 4930 proteins were determined using multiplex MS, of which 136, 9, 1, and 0 were regulated both reproducibly and significantly by IL-1β, C1P, S1P, and SPC, respectively. In the presence of IL-1ß, all 3 sphingolipids exerted ancillary effects. Only low SF levels of C1P and SPC were found. In conclusion, the 3 lipid species regulated proteins that have not been described in OA. Our results indicate that charged multivesicular body protein 1b, metal cation symporter ZIP14, glutamine-fructose-6-P transaminase, metallothionein-1F and -2A, ferritin, and prosaposin are particularly interesting proteins due to their potential to affect inflammatory, anabolic, catabolic, and apoptotic mechanisms.
Collapse
Affiliation(s)
- Thomas Timm
- Protein Analytics Group, Institute of Biochemistry, Justus Liebig University Giessen, 35392 Giessen, Germany
| | - Christiane Hild
- Laboratory for Experimental Orthopaedics, Department of Orthopaedics, Justus Liebig University Giessen, 35392 Giessen, Germany
| | - Gerhard Liebisch
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Markus Rickert
- Laboratory for Experimental Orthopaedics, Department of Orthopaedics, Justus Liebig University Giessen, 35392 Giessen, Germany
| | - Guenter Lochnit
- Protein Analytics Group, Institute of Biochemistry, Justus Liebig University Giessen, 35392 Giessen, Germany
| | - Juergen Steinmeyer
- Laboratory for Experimental Orthopaedics, Department of Orthopaedics, Justus Liebig University Giessen, 35392 Giessen, Germany
| |
Collapse
|
5
|
Leopold J, Schiller J. (Chemical) Roles of HOCl in Rheumatic Diseases. Antioxidants (Basel) 2024; 13:921. [PMID: 39199167 PMCID: PMC11351306 DOI: 10.3390/antiox13080921] [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: 03/27/2024] [Revised: 07/16/2024] [Accepted: 07/26/2024] [Indexed: 09/01/2024] Open
Abstract
Chronic rheumatic diseases such as rheumatoid arthritis (RA) are characterized by a dysregulated immune response and persistent inflammation. The large number of neutrophilic granulocytes in the synovial fluid (SF) from RA patients leads to elevated enzyme activities, for example, from myeloperoxidase (MPO) and elastase. Hypochlorous acid (HOCl), as the most important MPO-derived product, is a strong reactive oxygen species (ROS) and known to be involved in the processes of cartilage destruction (particularly regarding the glycosaminoglycans). This review will discuss open questions about the contribution of HOCl in RA in order to improve the understanding of oxidative tissue damaging. First, the (chemical) composition of articular cartilage and SF and the mechanisms of cartilage degradation will be discussed. Afterwards, the products released by neutrophils during inflammation will be summarized and their effects towards the individual, most abundant cartilage compounds (collagen, proteoglycans) and selected cellular components (lipids, DNA) discussed. New developments about neutrophil extracellular traps (NETs) and the use of antioxidants as drugs will be outlined, too. Finally, we will try to estimate the effects induced by these different agents and their contributions in RA.
Collapse
Affiliation(s)
- Jenny Leopold
- Institute for Medical Physics and Biophysics, Medical Faculty, Leipzig University, 04103 Leipzig, Germany;
| | | |
Collapse
|
6
|
Zhang W, Yang Q, Song Y, Liu W, Li Y. Exploratory metabolomic analysis for characterizing the metabolic profile of the urinary bladder under estrogen deprivation. Front Endocrinol (Lausanne) 2024; 15:1384115. [PMID: 38883607 PMCID: PMC11176512 DOI: 10.3389/fendo.2024.1384115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 05/20/2024] [Indexed: 06/18/2024] Open
Abstract
Background Estrogen homeostasis is crucial for bladder function, and estrogen deprivation resulting from menopause, ovariectomy or ovarian dysfunction may lead to various bladder dysfunctions. However, the specific mechanisms are not fully understood. Methods We simulated estrogen deprivation using a rat ovariectomy model and supplemented estrogen through subcutaneous injections. The metabolic characteristics of bladder tissue were analyzed using non-targeted metabolomics, followed by bioinformatics analysis to preliminarily reveal the association between estrogen deprivation and bladder function. Results We successfully established a rat model with estrogen deprivation and, through multivariate analysis and validation, identified several promising biomarkers represented by 3, 5-tetradecadiencarnitine, lysoPC (15:0), and cortisol. Furthermore, we explored estrogen deprivation-related metabolic changes in the bladder primarily characterized by amino acid metabolism imbalance. Conclusion This study, for the first time, depicts the metabolic landscape of bladder resulting from estrogen deprivation, providing an important experimental basis for future research on bladder dysfunctions caused by menopause.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Qingbo Yang
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yingying Song
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Wenheng Liu
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yao Li
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
7
|
Zhang Z, Shen C, Zhang P, Xu S, Kong L, Liang X, Li C, Qiu X, Huang J, Cui X. Fundamental, mechanism and development of hydration lubrication: From bio-inspiration to artificial manufacturing. Adv Colloid Interface Sci 2024; 327:103145. [PMID: 38615561 DOI: 10.1016/j.cis.2024.103145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 03/26/2024] [Accepted: 03/30/2024] [Indexed: 04/16/2024]
Abstract
Friction and lubrication are ubiquitous in all kinds of movements and play a vital role in the smooth operation of production machinery. Water is indispensable both in the lubrication systems of natural organisms and in hydration lubrication systems. There exists a high degree of similarity between these systems, which has driven the development of hydration lubrication from biomimetic to artificial manufacturing. In particular, significant advancements have been made in the understanding of the mechanisms of hydration lubrication over the past 30 years. This enhanced understanding has further stimulated the exploration of biomimetic inspiration from natural hydration lubrication systems, to develop novel artificial hydration lubrication systems that are cost-effective, easily transportable, and possess excellent capability. This review summarizes the recent experimental and theoretical advances in the understanding of hydration-lubrication processes. The entire paper is divided into three parts. Firstly, surface interactions relevant to hydration lubrication are discussed, encompassing topics such as hydrogen bonding, hydration layer, electric double layer force, hydration force, and Stribeck curve. The second part begins with an introduction to articular cartilage in biomaterial lubrication, discussing its compositional structure and lubrication mechanisms. Subsequently, three major categories of bio-inspired artificial manufacturing lubricating material systems are presented, including hydrogels, polymer brushes (e.g., neutral, positive, negative and zwitterionic brushes), hydration lubricant additives (e.g., nano-particles, polymers, ionic liquids), and their related lubrication mechanism is also described. Finally, the challenges and perspectives for hydration lubrication research and materials development are presented.
Collapse
Affiliation(s)
- Zekai Zhang
- Center for Advanced Jet Engineering Technologies (CaJET), Key Laboratory of High Efficiency and Clean Mechanical Manufacture of Ministry of Education, School of Mechanical Engineering, Shandong University, Jinan, Shandong 25006, China
| | - Chaojie Shen
- Center for Advanced Jet Engineering Technologies (CaJET), Key Laboratory of High Efficiency and Clean Mechanical Manufacture of Ministry of Education, School of Mechanical Engineering, Shandong University, Jinan, Shandong 25006, China
| | - Peipei Zhang
- Advanced Interdisciplinary Technology Research Center, National Innovation Institute of Defense Technology, Beijing 100071, China
| | - Shulei Xu
- Center for Advanced Jet Engineering Technologies (CaJET), Key Laboratory of High Efficiency and Clean Mechanical Manufacture of Ministry of Education, School of Mechanical Engineering, Shandong University, Jinan, Shandong 25006, China
| | - Lingchao Kong
- Advanced Interdisciplinary Technology Research Center, National Innovation Institute of Defense Technology, Beijing 100071, China
| | - Xiubing Liang
- Advanced Interdisciplinary Technology Research Center, National Innovation Institute of Defense Technology, Beijing 100071, China
| | - Chengcheng Li
- Advanced Interdisciplinary Technology Research Center, National Innovation Institute of Defense Technology, Beijing 100071, China
| | - Xiaoyong Qiu
- Center for Advanced Jet Engineering Technologies (CaJET), Key Laboratory of High Efficiency and Clean Mechanical Manufacture of Ministry of Education, School of Mechanical Engineering, Shandong University, Jinan, Shandong 25006, China
| | - Jun Huang
- Center for Advanced Jet Engineering Technologies (CaJET), Key Laboratory of High Efficiency and Clean Mechanical Manufacture of Ministry of Education, School of Mechanical Engineering, Shandong University, Jinan, Shandong 25006, China.
| | - Xin Cui
- Advanced Interdisciplinary Technology Research Center, National Innovation Institute of Defense Technology, Beijing 100071, China.
| |
Collapse
|
8
|
Li H, Cui Y, Wang J, Zhang W, Chen Y, Zhao J. Identification and validation of biomarkers related to lipid metabolism in osteoarthritis based on machine learning algorithms. Lipids Health Dis 2024; 23:111. [PMID: 38637751 PMCID: PMC11025229 DOI: 10.1186/s12944-024-02073-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 03/07/2024] [Indexed: 04/20/2024] Open
Abstract
BACKGROUND Osteoarthritis and lipid metabolism are strongly associated, although the precise targets and regulatory mechanisms are unknown. METHODS Osteoarthritis gene expression profiles were acquired from the GEO database, while lipid metabolism-related genes (LMRGs) were sourced from the MigSB database. An intersection was conducted between these datasets to extract gene expression for subsequent differential analysis. Following this, functional analyses were performed on the differentially expressed genes (DEGs). Subsequently, machine learning was applied to identify hub genes associated with lipid metabolism in osteoarthritis. Immune-infiltration analysis was performed using CIBERSORT, and external datasets were employed to validate the expression of these hub genes. RESULTS Nine DEGs associated with lipid metabolism in osteoarthritis were identified. UGCG and ESYT1, which are hub genes involved in lipid metabolism in osteoarthritis, were identified through the utilization of three machine learning algorithms. Analysis of the validation dataset revealed downregulation of UGCG in the experimental group compared to the normal group and upregulation of ESYT1 in the experimental group compared to the normal group. CONCLUSIONS UGCG and ESYT1 were considered as hub LMRGs in the development of osteoarthritis, which were regarded as candidate diagnostic markers. The effects are worth expected in the early diagnosis and treatment of osteoarthritis.
Collapse
Affiliation(s)
- Hang Li
- Wuxi Medical Center, Nanjing Medical University, No. 299 Qing Yang Road, Wuxi, Jiangsu, 214023, China
| | - Yubao Cui
- Clinical Research Center, The Affiliated Wuxi People's Hospital of Nanjing Medical University, No. 299 Qing Yang Road, Wuxi, 214023, Jiangsu, China
| | - Jian Wang
- Wuxi Medical Center, Nanjing Medical University, No. 299 Qing Yang Road, Wuxi, Jiangsu, 214023, China
| | - Wei Zhang
- Wuxi Medical Center, Nanjing Medical University, No. 299 Qing Yang Road, Wuxi, Jiangsu, 214023, China
| | - Yuhao Chen
- Wuxi Medical Center, Nanjing Medical University, No. 299 Qing Yang Road, Wuxi, Jiangsu, 214023, China
| | - Jijun Zhao
- Department of Orthopedic, The Affiliated Wuxi People's Hospital of Nanjing Medical University, No. 299 Qing Yang Road, Wuxi, Jiangsu, 214023, China.
| |
Collapse
|
9
|
Fu C, Wang Z, Zhou X, Hu B, Li C, Yang P. Protein-based bioactive coatings: from nanoarchitectonics to applications. Chem Soc Rev 2024; 53:1514-1551. [PMID: 38167899 DOI: 10.1039/d3cs00786c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Protein-based bioactive coatings have emerged as a versatile and promising strategy for enhancing the performance and biocompatibility of diverse biomedical materials and devices. Through surface modification, these coatings confer novel biofunctional attributes, rendering the material highly bioactive. Their widespread adoption across various domains in recent years underscores their importance. This review systematically elucidates the behavior of protein-based bioactive coatings in organisms and expounds on their underlying mechanisms. Furthermore, it highlights notable advancements in artificial synthesis methodologies and their functional applications in vitro. A focal point is the delineation of assembly strategies employed in crafting protein-based bioactive coatings, which provides a guide for their expansion and sustained implementation. Finally, the current trends, challenges, and future directions of protein-based bioactive coatings are discussed.
Collapse
Affiliation(s)
- Chengyu Fu
- Key Laboratory of Applied Surface and Colloid Chemistry, Ministry of Education, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an 710119, China.
- Xi'an Key Laboratory of Polymeric Soft Matter, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an 710119, China
- International Joint Research Center on Functional Fiber and Soft Smart Textile, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an 710119, China
| | - Zhengge Wang
- Key Laboratory of Applied Surface and Colloid Chemistry, Ministry of Education, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an 710119, China.
- Xi'an Key Laboratory of Polymeric Soft Matter, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an 710119, China
- International Joint Research Center on Functional Fiber and Soft Smart Textile, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an 710119, China
| | - Xingyu Zhou
- Key Laboratory of Applied Surface and Colloid Chemistry, Ministry of Education, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an 710119, China.
- Xi'an Key Laboratory of Polymeric Soft Matter, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an 710119, China
- International Joint Research Center on Functional Fiber and Soft Smart Textile, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an 710119, China
| | - Bowen Hu
- Key Laboratory of Applied Surface and Colloid Chemistry, Ministry of Education, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an 710119, China.
- Xi'an Key Laboratory of Polymeric Soft Matter, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an 710119, China
- International Joint Research Center on Functional Fiber and Soft Smart Textile, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an 710119, China
| | - Chen Li
- School of Chemistry and Chemical Engineering, Henan Institute of Science and Technology, Eastern HuaLan Avenue, Xinxiang, Henan 453003, China
| | - Peng Yang
- Key Laboratory of Applied Surface and Colloid Chemistry, Ministry of Education, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an 710119, China.
- Xi'an Key Laboratory of Polymeric Soft Matter, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an 710119, China
- International Joint Research Center on Functional Fiber and Soft Smart Textile, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an 710119, China
| |
Collapse
|
10
|
Wei G, Lu K, Umar M, Zhu Z, Lu WW, Speakman JR, Chen Y, Tong L, Chen D. Risk of metabolic abnormalities in osteoarthritis: a new perspective to understand its pathological mechanisms. Bone Res 2023; 11:63. [PMID: 38052778 PMCID: PMC10698167 DOI: 10.1038/s41413-023-00301-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 10/11/2023] [Accepted: 10/27/2023] [Indexed: 12/07/2023] Open
Abstract
Although aging has traditionally been viewed as the most important risk factor for osteoarthritis (OA), an increasing amount of epidemiological evidence has highlighted the association between metabolic abnormalities and OA, particularly in younger individuals. Metabolic abnormalities, such as obesity and type II diabetes, are strongly linked to OA, and they affect both weight-bearing and non-weight-bearing joints, thus suggesting that the pathogenesis of OA is more complicated than the mechanical stress induced by overweight. This review aims to explore the recent advances in research on the relationship between metabolic abnormalities and OA risk, including the impact of abnormal glucose and lipid metabolism, the potential pathogenesis and targeted therapeutic strategies.
Collapse
Affiliation(s)
- Guizheng Wei
- Department of Bone and Joint Surgery, the First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
- Research Center for Computer-aided Drug Discovery, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- Faculty of Pharmaceutical Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Ke Lu
- Research Center for Computer-aided Drug Discovery, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- Faculty of Pharmaceutical Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Muhammad Umar
- Research Center for Computer-aided Drug Discovery, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- Faculty of Pharmaceutical Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Zhenglin Zhu
- Department of Orthopedic Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - William W Lu
- Faculty of Pharmaceutical Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - John R Speakman
- Center for Energy Metabolism and Reproduction, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Yan Chen
- Department of Bone and Joint Surgery, the First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China.
| | - Liping Tong
- Research Center for Computer-aided Drug Discovery, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| | - Di Chen
- Research Center for Computer-aided Drug Discovery, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- Faculty of Pharmaceutical Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| |
Collapse
|
11
|
Haartmans MJJ, Claes BSR, Eijkel GB, Emanuel KS, Tuijthof GJM, Heeren RMA, Emans PJ, Cillero-Pastor B. Matrix-assisted laser desorption/ionization mass spectrometry imaging (MALDI-MSI) reveals potential lipid markers between infrapatellar fat pad biopsies of osteoarthritis and cartilage defect patients. Anal Bioanal Chem 2023; 415:5997-6007. [PMID: 37505238 PMCID: PMC10556153 DOI: 10.1007/s00216-023-04871-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 07/12/2023] [Accepted: 07/14/2023] [Indexed: 07/29/2023]
Abstract
The incidence of osteoarthritis (OA) has been expected to increase due to an aging population, as well as an increased incidence of intra-articular (osteo-) chondral damage. Lipids have already been shown to be involved in the inflammatory process of OA. This study aims at revealing region-specific lipid profiles of the infrapatellar fat pad (IPFP) of OA or cartilage defect patients by matrix-assisted laser desorption/ionization mass spectrometry imaging (MALDI-MSI), which could be used as biomarkers for early OA detection. A higher presence of phospholipids was found in OA patients compared with cartilage defect patients. In addition, a higher abundance of ether-linked phosphatidylethanolamines (PE O-s) containing arachidonic acid was specifically found in OA patients compared with cartilage defect patients. These lipids were mainly found in the connective tissue of the IPFP. Specific lipid species were associated to OA patients compared with cartilage defect patients. PE O-s have been suggested as possible biomarkers for OA. As these were found more abundantly in the connective tissue, the IPFP's intra-tissue heterogeneity might play an important role in biomarker discovery, implying that the amount of fibrous tissue is associated with OA.
Collapse
Affiliation(s)
- Mirella J J Haartmans
- Division of Imaging Mass Spectrometry, Maastricht MultiModal Molecular Imaging Institute (M4i), Maastricht University, Maastricht, the Netherlands
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Joint Preserving Clinic, CAPHRI Care and Public Health Research Institute, Maastricht University Medical Center+, Maastricht, the Netherlands
| | - Britt S R Claes
- Division of Imaging Mass Spectrometry, Maastricht MultiModal Molecular Imaging Institute (M4i), Maastricht University, Maastricht, the Netherlands
| | - Gert B Eijkel
- Division of Imaging Mass Spectrometry, Maastricht MultiModal Molecular Imaging Institute (M4i), Maastricht University, Maastricht, the Netherlands
| | - Kaj S Emanuel
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Joint Preserving Clinic, CAPHRI Care and Public Health Research Institute, Maastricht University Medical Center+, Maastricht, the Netherlands
- Department of Orthopedic Surgery and Sport Medicine, Amsterdam Movement Sciences, Amsterdam UMC, Amsterdam, the Netherlands
| | - Gabrielle J M Tuijthof
- Biomedical Device Design and Production Technology, Faculty of Engineering Technology, University of Twente, Enschede, the Netherlands
| | - Ron M A Heeren
- Division of Imaging Mass Spectrometry, Maastricht MultiModal Molecular Imaging Institute (M4i), Maastricht University, Maastricht, the Netherlands
| | - Pieter J Emans
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Joint Preserving Clinic, CAPHRI Care and Public Health Research Institute, Maastricht University Medical Center+, Maastricht, the Netherlands
| | - Berta Cillero-Pastor
- Division of Imaging Mass Spectrometry, Maastricht MultiModal Molecular Imaging Institute (M4i), Maastricht University, Maastricht, the Netherlands.
- MERLN Institute for Technology-Inspired Regenerative Medicine, Department of Cell Biology-Inspired Tissue Engineering (cBITE), Maastricht University, Universiteitssingel 40, 6229 ER, Maastricht, the Netherlands.
| |
Collapse
|
12
|
Mielke S, Sorkin R, Klein J. Effect of cholesterol on the mechanical stability of gel-phase phospholipid bilayers studied by AFM force spectroscopy. THE EUROPEAN PHYSICAL JOURNAL. E, SOFT MATTER 2023; 46:77. [PMID: 37672138 DOI: 10.1140/epje/s10189-023-00338-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 08/24/2023] [Indexed: 09/07/2023]
Abstract
The remarkably low sliding friction of articular cartilage in the major joints such as hips and knees, which is crucial for its homeostasis and joint health, has been attributed to lipid bilayers forming lubricious boundary layers at its surface. The robustness of such layers, and thus their lubrication efficiency at joint pressures, depends on the lipids forming them, including cholesterol which is a ubiquitous component, and which may act to strengthen of weaken the bilayer. In this work, a systematic study using an atomic force microscope (AFM) was carried out to understand the effect of cholesterol on the nanomechanical stability of two saturated phospholipids, DSPC (1,2-distearoyl-sn-glycero-3-phosphatidlycholine) and DPPC (1,2-dipalmitoyl-sn-glycero- phosphatidylcholine), that differ in acyl chain lengths. Measurements were carried out both in water and in phosphate buffer solution (PBS). The nanomechanical stability of the lipid bilayers was quantitatively evaluated by measuring the breakthrough force needed to puncture the bilayer by the AFM tip. The molar fractions of cholesterol incorporated in the bilayers were 10% and 40%. We found that for both DSPC and DPPC, cholesterol significantly decreases the mechanical stability of the bilayers in solid-ordered (SO) phase. In accordance with the literature, the strengthening effect of salt on the lipid bilayers was also observed. For DPPC with 10 mol % cholesterol, the effect of tip properties and the experimental procedure parameters on the breakthrough forces were also studied. Tip radius (2-42 nm), material (Si, Si3N4, Au) and loading rate (40-1000 nm/s) were varied systematically. The values of the breakthrough forces measured were not significantly affected by any of these parameters, showing that the weakening effect of cholesterol does not result from such changes in experimental conditions. As we have previously demonstrated that mechanical robustness improves the tribological performance of lipid layers, this study helps to shed light on the mechanism of physiological lubrication. Nanoindentation of SDPC bilayers.
Collapse
Affiliation(s)
- Salomé Mielke
- Department of Molecular Chemistry and Materials Science, Weizmann Institute of Science, 76100, Rehovot, Israel
| | - Raya Sorkin
- School of Chemistry, Raymond and Beverly Sackler Faculty of Exact Sciences, Tel Aviv University, 6997801, Tel Aviv, Israel.
| | - Jacob Klein
- Department of Molecular Chemistry and Materials Science, Weizmann Institute of Science, 76100, Rehovot, Israel.
| |
Collapse
|
13
|
Chabronova A, van den Akker G, Housmans BAC, Caron MMJ, Cremers A, Surtel DAM, Peffers MJ, van Rhijn LW, Marchand V, Motorin Y, Welting TJM. Depletion of SNORA33 Abolishes ψ of 28S-U4966 and Affects the Ribosome Translational Apparatus. Int J Mol Sci 2023; 24:12578. [PMID: 37628759 PMCID: PMC10454564 DOI: 10.3390/ijms241612578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 08/04/2023] [Accepted: 08/07/2023] [Indexed: 08/27/2023] Open
Abstract
Eukaryotic ribosomes are complex molecular nanomachines translating genetic information from mRNAs into proteins. There is natural heterogeneity in ribosome composition. The pseudouridylation (ψ) of ribosomal RNAs (rRNAs) is one of the key sources of ribosome heterogeneity. Nevertheless, the functional consequences of ψ-based ribosome heterogeneity and its relevance for human disease are yet to be understood. Using HydraPsiSeq and a chronic disease model of non-osteoarthritic primary human articular chondrocytes exposed to osteoarthritic synovial fluid, we demonstrated that the disease microenvironment is capable of instigating site-specific changes in rRNA ψ profiles. To investigate one of the identified differential rRNA ψ sites (28S-ψ4966), we generated SNORA22 and SNORA33 KO SW1353 cell pools using LentiCRISPRv2/Cas9 and evaluated the ribosome translational capacity by 35S-Met/Cys incorporation, assessed the mode of translation initiation and ribosomal fidelity using dual luciferase reporters, and assessed cellular and ribosomal proteomes by LC-MS/MS. We uncovered that the depletion of SNORA33, but not SNORA22, reduced 28S-ψ4966 levels. The resulting loss of 28S-ψ4966 affected ribosomal protein composition and function and led to specific changes in the cellular proteome. Overall, our pioneering findings demonstrate that cells dynamically respond to disease-relevant changes in their environment by altering their rRNA pseudouridylation profiles, with consequences for ribosome function and the cellular proteome relevant to human disease.
Collapse
Affiliation(s)
- Alzbeta Chabronova
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University, 6229 HX Maastricht, The Netherlands
| | - Guus van den Akker
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University, 6229 HX Maastricht, The Netherlands
| | - Bas A C Housmans
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University, 6229 HX Maastricht, The Netherlands
| | - Marjolein M J Caron
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University, 6229 HX Maastricht, The Netherlands
| | - Andy Cremers
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University, 6229 HX Maastricht, The Netherlands
| | - Don A M Surtel
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University, 6229 HX Maastricht, The Netherlands
| | - Mandy J Peffers
- Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L8 7TX, UK
| | - Lodewijk W van Rhijn
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University, 6229 HX Maastricht, The Netherlands
| | - Virginie Marchand
- UAR2008 IBSLor CNRS-INSERM-Université de Lorraine, F54000 Nancy, France
| | - Yuri Motorin
- UAR2008 IBSLor CNRS-INSERM-Université de Lorraine, F54000 Nancy, France
- UMR7365 IMOPA, CNRS-Université de Lorraine, F54000 Nancy, France
| | - Tim J M Welting
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University, 6229 HX Maastricht, The Netherlands
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University Medical Center+ (MUMC+), 6229 HX Maastricht, The Netherlands
| |
Collapse
|
14
|
Timm T, Hild C, Liebisch G, Rickert M, Lochnit G, Steinmeyer J. Functional Characterization of Lysophospholipids by Proteomic and Lipidomic Analysis of Fibroblast-like Synoviocytes. Cells 2023; 12:1743. [PMID: 37443777 PMCID: PMC10340184 DOI: 10.3390/cells12131743] [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: 06/01/2023] [Revised: 06/23/2023] [Accepted: 06/27/2023] [Indexed: 07/15/2023] Open
Abstract
Synovial fluid (SF) from human knee joints with osteoarthritis (OA) has elevated levels of lysophosphatidylcholine (LPC) species, but their functional role is not well understood. This in vitro study was designed to test the hypothesis that various LPCs found elevated in OA SF and their metabolites, lysophosphatidic acids (LPAs), modulate the abundance of proteins and phospholipids (PLs) in human fibroblast-like synoviocytes (FLSs), with even minute chemical variations in lysophospholipids determining the extent of regulation. Cultured FLSs (n = 5-7) were treated with one of the LPC species, LPA species, IL-1β, or a vehicle. Tandem mass tag peptide labeling coupled with LC-MS/MS/MS was performed to quantify proteins. The expression of mRNA from regulated proteins was analyzed using RT-PCR. PL synthesis was determined via ESI-MS/MS, and the release of radiolabeled PLs was determined by means of liquid scintillation counting. In total, 3960 proteins were quantified using multiplexed MS, of which 119, 8, and 3 were significantly and reproducibly regulated by IL-1β, LPC 16:0, and LPC 18:0, respectively. LPC 16:0 significantly inhibited the release of PLs and the synthesis of phosphatidylcholine, LPC, and sphingomyelin. Neither LPC metabolite-LPA 16:0 nor LPA 18:0-had any reproducible effect on the levels of each protein. In conclusion, small chemical variations in LPC species can result in the significantly altered expression and secretion of proteins and PLs from FLSs. IL-1β influenced all proteins that were reproducibly regulated by LPC 16:0. LPC species are likely to modulate FLS protein expression only in more advanced OA stages with low IL-1β levels. None of the eight proteins being significantly regulated by LPC 16:0 have been previously reported in OA. However, our in vitro findings show that the CD81 antigen, calumenin, and B4E2C1 are promising candidates for further study, focusing in particular on their potential ability to modulate inflammatory and catabolic mechanisms.
Collapse
Affiliation(s)
- Thomas Timm
- Protein Analytics Group, Institute of Biochemistry, Justus Liebig University Giessen, 35392 Giessen, Germany
| | - Christiane Hild
- Laboratory for Experimental Orthopedics, Department of Orthopedics, Justus Liebig University Giessen, 35392 Giessen, Germany
| | - Gerhard Liebisch
- Department for Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Markus Rickert
- Laboratory for Experimental Orthopedics, Department of Orthopedics, Justus Liebig University Giessen, 35392 Giessen, Germany
| | - Guenter Lochnit
- Protein Analytics Group, Institute of Biochemistry, Justus Liebig University Giessen, 35392 Giessen, Germany
| | - Juergen Steinmeyer
- Laboratory for Experimental Orthopedics, Department of Orthopedics, Justus Liebig University Giessen, 35392 Giessen, Germany
| |
Collapse
|
15
|
Mancino C, Pasto A, De Rosa E, Dolcetti L, Rasponi M, McCulloch P, Taraballi F. Immunomodulatory biomimetic nanoparticles target articular cartilage trauma after systemic administration. Heliyon 2023; 9:e16640. [PMID: 37313169 PMCID: PMC10258364 DOI: 10.1016/j.heliyon.2023.e16640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 05/19/2023] [Accepted: 05/23/2023] [Indexed: 06/15/2023] Open
Abstract
Post-traumatic osteoarthritis (PTOA) is one of the leading causes of disability in developed countries and accounts for 12% of all osteoarthritis cases in the United States. After trauma, inflammatory cells (macrophages amongst others) are quickly recruited within the inflamed synovium and infiltrate the joint space, initiating dysregulation of cartilage tissue homeostasis. Current therapeutic strategies are ineffective, and PTOA remains an open clinical challenge. Here, the targeting potential of liposome-based nanoparticles (NPs) is evaluated in a PTOA mouse model, during the acute phase of inflammation, in both sexes. NPs are composed of biomimetic phospholipids or functionalized with macrophage membrane proteins. Intravenous administration of NPs in the acute phase of PTOA and advanced in vivo imaging techniques reveal preferential accumulation of NPs within the injured joint for up to 7 days post injury, in comparison to controls. Finally, imaging mass cytometry uncovers an extraordinary immunomodulatory effect of NPs that are capable of decreasing the amount of immune cells infiltrating the joint and conditioning their phenotype. Thus, biomimetic NPs could be a powerful theranostic tool for PTOA as their accumulation in injury sites allows their identification and they have an intrinsic immunomodulatory effect.
Collapse
Affiliation(s)
- Chiara Mancino
- Center for Musculoskeletal Regeneration, Houston Methodist Academic Institute, Houston, TX, USA
- Orthopedics and Sports Medicine, Houston Methodist Hospital, Houston, TX, USA
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milano, Italy
| | - Anna Pasto
- Richard Dimbleby Laboratory of Cancer Research, School of Cancer & Pharmaceutical Sciences, King's College London, London, UK
| | - Enrica De Rosa
- Center for Musculoskeletal Regeneration, Houston Methodist Academic Institute, Houston, TX, USA
- Orthopedics and Sports Medicine, Houston Methodist Hospital, Houston, TX, USA
| | - Luigi Dolcetti
- Richard Dimbleby Laboratory of Cancer Research, School of Cancer & Pharmaceutical Sciences, King's College London, London, UK
| | - Marco Rasponi
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milano, Italy
| | - Patrick McCulloch
- Orthopedics and Sports Medicine, Houston Methodist Hospital, Houston, TX, USA
| | - Francesca Taraballi
- Center for Musculoskeletal Regeneration, Houston Methodist Academic Institute, Houston, TX, USA
- Orthopedics and Sports Medicine, Houston Methodist Hospital, Houston, TX, USA
| |
Collapse
|
16
|
Li Q, Xu JY, Hu X, Li J, Huang XJ, Wu ZY, Wang DG, Ge YB. The protective effects and mechanism of Ruyi Zhenbao Pill, a Tibetan medicinal compound, in a rat model of osteoarthritis. JOURNAL OF ETHNOPHARMACOLOGY 2023; 308:116255. [PMID: 36809823 DOI: 10.1016/j.jep.2023.116255] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 02/06/2023] [Accepted: 02/07/2023] [Indexed: 06/18/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Ruyi Zhenbao Pill (RZP) is a prescribed Tibetan formulation for the treatment of white-pulse-disease, yellow-water-disease as well as pain-related disease. RZP is composed of 30 medicinal materials including herbal medicine, animal medicine and mineral medicine. They are widely used in the Tibetan area to treat cerebrovascular disease, hemiplegia, rheumatism, and pain diseases for centuries. AIM OF THE STUDY The aim of the present study was to evaluate the anti-osteoarthritis function of RZP and to clarify the underlying mechanisms. MATERIALS AND METHODS The active components in RZP were identified using HPLC methods. Osteoarthritis (OA) animal model was established via intra-articular injection of papain in rat knees. After the administration of RZP (0.45, 0.9 g/kg) for 28 days, the clinical observation was conducted, and pathological changes as well as serum biochemical indexes were detected. Moreover, therapeutic targets and pathways of RZP were discussed. RESULTS The results showed that RZP could suppress knee joint swelling and arthralgia, thus relieving joint pain and inflammation in OA rats. Microcomputed tomography (μCT)-based physiological imaging and staining pictures confirmed the therapeutic effects of RZP on OA symptoms including knee joint swelling and structural changes with progressive inflammation in OA rats. RZP could promote the synthesis or inhibit the degradation of COLⅡ, attenuate OA-induced OPN up-regulation and thus relieve the OA symptom. Furthermore, RZP (0.45-0.9 g/kg) could all ameliorate the imbalance of biomarkers related to OA such as MMP1, TNF-α, COX2, IL-1β and iNOS in knee joints or serum. CONCLUSION In conclusion, RZP could effectively relieve inflammatory reaction induced by OA injury and the formulation could be applied to the treatment of OA therapy.
Collapse
Affiliation(s)
- Qien Li
- Tibetan Medical College, Qinghai University, Xining, PR China
| | - Jing-Yi Xu
- College of Pharmaceutical Science, South-Central Minzu University, Wuhan, PR China
| | - Xin Hu
- College of Pharmaceutical Science, South-Central Minzu University, Wuhan, PR China
| | - Jun Li
- College of Pharmaceutical Science, South-Central Minzu University, Wuhan, PR China
| | - Xian-Ju Huang
- College of Pharmaceutical Science, South-Central Minzu University, Wuhan, PR China; Ethnopharmacology Level 3 Laboratory, National Administration of Traditional Chinese Medicine, South-Central Minzu University, Wuhan, 430074, PR China.
| | - Zhou-Yang Wu
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China.
| | - Da-Gui Wang
- College of Pharmaceutical Science, South-Central Minzu University, Wuhan, PR China
| | - Yue-Bin Ge
- College of Pharmaceutical Science, South-Central Minzu University, Wuhan, PR China
| |
Collapse
|
17
|
Mustonen AM, Tollis S, Käkelä R, Sihvo SP, Palosaari S, Pohjanen VM, Yli-Hallila A, Lehenkari P, Nieminen P. Increased n-6 Polyunsaturated Fatty Acids Indicate Pro- and Anti-Inflammatory Lipid Modifications in Synovial Membranes with Rheumatoid Arthritis. Inflammation 2023:10.1007/s10753-023-01816-3. [PMID: 37140681 PMCID: PMC10359413 DOI: 10.1007/s10753-023-01816-3] [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: 01/16/2023] [Revised: 03/31/2023] [Accepted: 04/04/2023] [Indexed: 05/05/2023]
Abstract
Emerging evidence suggests that fatty acids (FAs) and their lipid mediator derivatives can induce both beneficial and detrimental effects on inflammatory processes and joint degradation in osteoarthritis (OA) and autoimmune-driven rheumatoid arthritis (RA). The present study characterized the detailed FA signatures of synovial membranes collected during knee replacement surgery of age- and gender-matched OA and RA patients (n = 8/diagnosis). The FA composition of total lipids was determined by gas chromatography and analyzed with univariate and multivariate methods supplemented with hierarchical clustering (HC), random forest (RF)-based classification of FA signatures, and FA metabolism pathway analysis. RA synovium lipids were characterized by reduced proportions of shorter-chain saturated FAs (SFAs) and elevated percentages of longer-chain SFAs and monounsaturated FAs, alkenyl chains, and C20 n-6 polyunsaturated FAs compared to OA synovium lipids. In HC, FAs and FA-derived variables clustered into distinct groups, which preserved the discriminatory power of the individual variables in predicting the RA and OA inflammatory states. In RF classification, SFAs and 20:3n-6 were among the most important FAs distinguishing RA and OA. Pathway analysis suggested that elongation reactions of particular long-chain FAs would have increased relevance in RA. The present study was able to determine the individual FAs, FA groups, and pathways that distinguished the more inflammatory RA from OA. The findings suggest modifications of FA elongation and metabolism of 20:4n-6, glycerophospholipids, sphingolipids, and plasmalogens in the chronically inflamed RA synovium. These FA alterations could have implications in lipid mediator synthesis and potential as novel diagnostic and therapeutic tools.
Collapse
Affiliation(s)
- Anne-Mari Mustonen
- Institute of Biomedicine, School of Medicine, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland.
- Department of Environmental and Biological Sciences, Faculty of Science, Forestry and Technology, University of Eastern Finland, P.O. Box 111, FI-80101, Joensuu, Finland.
| | - Sylvain Tollis
- Institute of Biomedicine, School of Medicine, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland
| | - Reijo Käkelä
- Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, P.O. Box 65, FI-00014, Helsinki, Finland
- Helsinki University Lipidomics Unit (HiLIPID), Helsinki Institute of Life Science (HiLIFE) and Biocenter Finland, University of Helsinki, P.O. Box 65, FI-00014, Helsinki, Finland
| | - Sanna P Sihvo
- Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, P.O. Box 65, FI-00014, Helsinki, Finland
- Helsinki University Lipidomics Unit (HiLIPID), Helsinki Institute of Life Science (HiLIFE) and Biocenter Finland, University of Helsinki, P.O. Box 65, FI-00014, Helsinki, Finland
| | - Sanna Palosaari
- Cancer and Translational Medicine Research Unit, Faculty of Medicine, University of Oulu, P.O. Box 5000, FI-90014, Oulu, Finland
- Medical Research Center, University of Oulu and Oulu University Hospital, P.O. Box 5000, FI-90014, Oulu, Finland
| | - Vesa-Matti Pohjanen
- Cancer and Translational Medicine Research Unit, Faculty of Medicine, University of Oulu, P.O. Box 5000, FI-90014, Oulu, Finland
- Medical Research Center, University of Oulu and Oulu University Hospital, P.O. Box 5000, FI-90014, Oulu, Finland
| | - Aaron Yli-Hallila
- Cancer and Translational Medicine Research Unit, Faculty of Medicine, University of Oulu, P.O. Box 5000, FI-90014, Oulu, Finland
| | - Petri Lehenkari
- Cancer and Translational Medicine Research Unit, Faculty of Medicine, University of Oulu, P.O. Box 5000, FI-90014, Oulu, Finland
- Medical Research Center, University of Oulu and Oulu University Hospital, P.O. Box 5000, FI-90014, Oulu, Finland
- Department of Surgery, Oulu University Hospital, P.O. Box 21, FI-90029, OYS, Oulu, Finland
| | - Petteri Nieminen
- Institute of Biomedicine, School of Medicine, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland
| |
Collapse
|
18
|
Cedeno M, Murillo-Saich J, Coras R, Cedola F, Brandy A, Prior A, Pedersen A, Mateo L, Martinez-Morillo M, Guma M. Serum metabolomic profiling identifies potential biomarkers in arthritis in older adults: an exploratory study. Metabolomics 2023; 19:37. [PMID: 37022535 PMCID: PMC11449491 DOI: 10.1007/s11306-023-02004-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 03/29/2023] [Indexed: 04/07/2023]
Abstract
BACKGROUND Seronegative elderly-onset rheumatoid arthritis (EORA)neg and polymyalgia rheumatica (PMR) have similar clinical characteristics making them difficult to distinguish based on clinical features. We hypothesized that the study of serum metabolome could identify potential biomarkers of PMR vs. EORAneg. METHODS Arthritis in older adults (ARTIEL) is an observational prospective cohort with patients older than 60 years of age with newly diagnosed arthritis. Patients' blood samples were compared at baseline with 18 controls. A thorough clinical examination was conducted. A Bruker Avance 600 MHz spectrometer was used to acquire Nuclear Magnetic Resonance (NMR) spectra of serum samples. Chenomx NMR suite 8.5 was used for metabolite identification and quantification.Student t-test, one-way ANOVA, binary linear regression and ROC curve, Pearson's correlation along with pathway analyses were conducted. RESULTS Twenty-eight patients were diagnosed with EORAneg and 20 with PMR. EORAneg patients had a mean disease activity score (DAS)-Erythrocyte Sedimentation Rate (ESR) of 6.21 ± 1.00. All PMR patients reported shoulder pain, and 90% reported pelvic pain. Fifty-eight polar metabolites were identified. Of these, 3-hydroxybutyrate, acetate, glucose, glycine, lactate, and o-acetylcholine (o-ACh), were significantly different between groups. Of interest, IL-6 correlated with different metabolites in PMR and EORAneg suggesting different inflammatory activated pathways. Finally, lactate, o-ACh, taurine, and sex (female) were identified as distinguishable factors of PMR from EORAneg with a sensitivity of 90%, specificity of 92.3%, and an AUC of 0.925 (p < 0.001). CONCLUSION These results suggest that EORAneg and PMR have different serum metabolomic profiles that might be related to their pathobiology and can be used as biomarker to discriminate between both diseases.
Collapse
Affiliation(s)
- Martha Cedeno
- Department of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Jessica Murillo-Saich
- Department of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Roxana Coras
- Department of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
- Department of Medicine, Autonomous University of Barcelona, Plaça Cívica, Bellaterra, Barcelona, 08193, Spain
| | - Francesca Cedola
- Department of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Anahy Brandy
- Department of Rheumatology, Germans Trias i Pujol, University Hospital, Carretera de Canyet, Badalona, 08916, Spain
| | - Agueda Prior
- Department of Rheumatology, Germans Trias i Pujol, University Hospital, Carretera de Canyet, Badalona, 08916, Spain
| | - Anders Pedersen
- Swedish NMR Centre, University of Gothenburg, Gothenburg, 41390, Sweden
| | - Lourdes Mateo
- Department of Rheumatology, Germans Trias i Pujol, University Hospital, Carretera de Canyet, Badalona, 08916, Spain
| | - Melania Martinez-Morillo
- Department of Rheumatology, Germans Trias i Pujol, University Hospital, Carretera de Canyet, Badalona, 08916, Spain.
| | - Monica Guma
- Department of Medicine, University of California San Diego, La Jolla, CA, 92093, USA.
- Department of Medicine, Autonomous University of Barcelona, Plaça Cívica, Bellaterra, Barcelona, 08193, Spain.
- VA Healthcare Service, San Diego, CA, 92161, USA.
| |
Collapse
|
19
|
Mustonen AM, Lehmonen N, Paakkonen T, Raekallio M, Käkelä R, Niemelä T, Mykkänen A, Sihvo SP, Nieminen P. Equine osteoarthritis modifies fatty acid signatures in synovial fluid and its extracellular vesicles. Arthritis Res Ther 2023; 25:39. [PMID: 36895037 PMCID: PMC9996872 DOI: 10.1186/s13075-023-02998-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 01/27/2023] [Indexed: 03/11/2023] Open
Abstract
BACKGROUND Individual fatty acids (FAs) and their derivatives (lipid mediators) with pro-inflammatory or dual anti-inflammatory and pro-resolving properties have potential to influence the health of joint tissues. Osteoarthritis (OA) is an age-associated chronic joint disease that can be featured with altered FA composition in the synovial fluid (SF) of human patients. The counts and cargo of extracellular vesicles (EVs), membrane-bound particles released by synovial joint cells and transporting bioactive lipids, can also be modified by OA. The detailed FA signatures of SF and its EVs have remained unexplored in the horse - a well-recognized veterinary model for OA research. METHODS The aim of the present study was to compare the FA profiles in equine SF and its ultracentrifuged EV fraction between control, contralateral, and OA metacarpophalangeal joints (n = 8/group). The FA profiles of total lipids were determined by gas chromatography and the data compared with univariate and multivariate analyses. RESULTS The data revealed distinct FA profiles in SF and its EV-enriched pellet that were modified by naturally occurring equine OA. Regarding SFs, linoleic acid (generalized linear model, p = 0.0006), myristic acid (p = 0.003), palmitoleic acid (p < 0.0005), and n-3/n-6 polyunsaturated FA ratio (p < 0.0005) were among the important variables that separated OA from control samples. In EV-enriched pellets, saturated FAs palmitic acid (p = 0.020), stearic acid (p = 0.002), and behenic acid (p = 0.003) indicated OA. The observed FA modifications are potentially detrimental and could contribute to inflammatory processes and cartilage degradation in OA. CONCLUSIONS Equine OA joints can be distinguished from normal joints based on their FA signatures in SF and its EV-enriched pellet. Clarifying the roles of SF and EV FA compositions in the pathogenesis of OA and their potential as joint disease biomarkers and therapeutic targets warrants future studies.
Collapse
Affiliation(s)
- Anne-Mari Mustonen
- grid.9668.10000 0001 0726 2490Institute of Biomedicine, School of Medicine, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland
- grid.9668.10000 0001 0726 2490Department of Environmental and Biological Sciences, Faculty of Science, Forestry and Technology, University of Eastern Finland, P.O. Box 111, FI-80101 Joensuu, Finland
| | - Nina Lehmonen
- grid.7737.40000 0004 0410 2071Department of Equine and Small Animal Medicine, Faculty of Veterinary Medicine, University of Helsinki, P.O. Box 57, FI-00014 Helsinki, Finland
| | - Tommi Paakkonen
- grid.9668.10000 0001 0726 2490Institute of Biomedicine, School of Medicine, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland
| | - Marja Raekallio
- grid.7737.40000 0004 0410 2071Department of Equine and Small Animal Medicine, Faculty of Veterinary Medicine, University of Helsinki, P.O. Box 57, FI-00014 Helsinki, Finland
| | - Reijo Käkelä
- grid.7737.40000 0004 0410 2071Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, P.O. Box 65, FI-00014 Helsinki, Finland
- grid.484023.9Helsinki University Lipidomics Unit (HiLIPID), Helsinki Institute of Life Science (HiLIFE) and Biocenter Finland, P.O. Box 65, FI-00014 Helsinki, Finland
| | - Tytti Niemelä
- grid.7737.40000 0004 0410 2071Department of Equine and Small Animal Medicine, Faculty of Veterinary Medicine, University of Helsinki, P.O. Box 57, FI-00014 Helsinki, Finland
| | - Anna Mykkänen
- grid.7737.40000 0004 0410 2071Department of Equine and Small Animal Medicine, Faculty of Veterinary Medicine, University of Helsinki, P.O. Box 57, FI-00014 Helsinki, Finland
| | - Sanna P. Sihvo
- grid.7737.40000 0004 0410 2071Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, P.O. Box 65, FI-00014 Helsinki, Finland
- grid.484023.9Helsinki University Lipidomics Unit (HiLIPID), Helsinki Institute of Life Science (HiLIFE) and Biocenter Finland, P.O. Box 65, FI-00014 Helsinki, Finland
| | - Petteri Nieminen
- grid.9668.10000 0001 0726 2490Institute of Biomedicine, School of Medicine, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland
| |
Collapse
|
20
|
Cao L, Zheng X, Han P, Ren L, Hu F, Li Z. Raman spectroscopy as a promising diagnostic method for rheumatoid arthritis. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2023; 15:709-718. [PMID: 36598183 DOI: 10.1039/d2ay01904c] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Background: Diagnosis of rheumatoid arthritis (RA) basically relies on clinical symptoms and autoantibodies, especially anti-citrullinated protein antibodies (ACPAs) and rheumatoid factor (RF). However, the lack of autoantibodies is still a dilemma clinically in seronegative RA, especially in the early stage of the disease. This study aimed to provide a unique disease fingerprint with high diagnostic value to discriminate RA based on Raman spectroscopy. Methods: Raman spectroscopy provides a repertoire of biomolecules in serum from RA. Multivariate dimension-reducing methods and machine-learning algorithms were exploited to reveal the intrinsic differences and the potential discrimination power. The underlying differential biomolecules were retrieved by the assignment of Raman peaks. Moreover, the correlations between the spectral differences and RA patient's clinical and immunological manifestations were also analyzed. Results: RA patients exhibited unique Raman spectra characterized by biomolecular alterations during the disease progression. The discrimination power yielded 97.3% sensitivity and 94.8% specificity for RA diagnosis. In the recognition of ACPA-negative RA, the sensitivity and specificity also reached 95.6% and 92.8%, respectively. In particular, the differential Raman spectrum peaks of RA patients mainly represented lipids, amino acids, glycogen, and fatty acids. Further analysis showed that the different serum Raman spectra correlated with the clinical features of RA, including disease duration, RF, anticyclic citrullinated peptide antibodies (anti-CCPs), IgA, IgM, IgG, tender joint count, and swollen joint count (|rs| = 0.15-0.52, p < 0.05). Conclusions: Raman spectroscopy was revealed to be a promising diagnostic method for RA, especially for ACPA-negative patients.
Collapse
Affiliation(s)
- Lulu Cao
- Department of Rheumatology and Immunology, Peking University People's Hospital and Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China.
| | - Xi Zheng
- Department of Rheumatology and Immunology, Peking University People's Hospital and Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China.
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Peng Han
- Department of Rheumatology and Immunology, Peking University People's Hospital and Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China.
| | - Limin Ren
- Department of Rheumatology and Immunology, Peking University People's Hospital and Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China.
| | - Fanlei Hu
- Department of Rheumatology and Immunology, Peking University People's Hospital and Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China.
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Zhanguo Li
- Department of Rheumatology and Immunology, Peking University People's Hospital and Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China.
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| |
Collapse
|
21
|
Khoury S, Colas J, Breuil V, Kosek E, Ahmed AS, Svensson CI, Marchand F, Deval E, Ferreira T. Identification of Lipid Biomarkers for Chronic Joint Pain Associated with Different Joint Diseases. Biomolecules 2023; 13:biom13020342. [PMID: 36830710 PMCID: PMC9953120 DOI: 10.3390/biom13020342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 02/04/2023] [Accepted: 02/06/2023] [Indexed: 02/12/2023] Open
Abstract
Lipids, especially lysophosphatidylcholine LPC16:0, have been shown to be involved in chronic joint pain through the activation of acid-sensing ion channels (ASIC3). The aim of the present study was to investigate the lipid contents of the synovial fluids from controls and patients suffering from chronic joint pain in order to identify characteristic lipid signatures associated with specific joint diseases. For this purpose, lipids were extracted from the synovial fluids and analyzed by mass spectrometry. Lipidomic analyses identified certain choline-containing lipid classes and molecular species as biomarkers of chronic joint pain, regardless of the pathology, with significantly higher levels detected in the patient samples. Moreover, correlations were observed between certain lipid levels and the type of joint pathologies. Interestingly, LPC16:0 levels appeared to correlate with the metabolic status of patients while other choline-containing lipids were more specifically associated with the inflammatory state. Overall, these data point at selective lipid species in synovial fluid as being strong predictors of specific joint pathologies which could help in the selection of the most adapted treatment.
Collapse
Affiliation(s)
- Spiro Khoury
- Université de Poitiers, Laboratoire Lipotoxicity and Channelopathies (LiTch)—ConicMeds, 86073 Poitiers, France
- Correspondence:
| | - Jenny Colas
- Université de Poitiers, Laboratoire Lipotoxicity and Channelopathies (LiTch)—ConicMeds, 86073 Poitiers, France
- Université de Poitiers, Laboratoire PRéTI, 86073 Poitiers, France
| | - Véronique Breuil
- Université Côte d’Azur (UCA), UMR E-4320 MATOs CEA/iBEB/SBTN, Faculté de Médecine, CEDEX 2, 06107 Nice, France
- Service de Rhumatologie, Hôpital Pasteur, CHU de Nice, 06000 Nice, France
| | - Eva Kosek
- Department of Clinical Neuroscience, Karolinska Institutet, 17165 Solna, Sweden
- Department of Surgical Sciences, Uppsala University, 75185 Uppsala, Sweden
| | - Aisha S. Ahmed
- Department of Molecular Medicine and Surgery, Karolinska Institutet, 17176 Stockholm, Sweden
| | - Camilla I. Svensson
- Department of Physiology and Pharmacology, Center for Molecular Medicine, Karolinska Institutet, 17165 Solna, Sweden
| | - Fabien Marchand
- Université Clermont Auvergne, Inserm U1107 Neuro-Dol, Pharmacologie Fondamentale et Clinique de la Douleur, 63001 Clermont-Ferrand, France
| | - Emmanuel Deval
- Université Côte d’Azur, CNRS, IPMC, LabEx ICST, FHU InovPain, 06560 Valbonne, France
| | - Thierry Ferreira
- Université de Poitiers, Laboratoire Lipotoxicity and Channelopathies (LiTch)—ConicMeds, 86073 Poitiers, France
- Université de Poitiers, Laboratoire PRéTI, 86073 Poitiers, France
| |
Collapse
|
22
|
Sarvilina IV, Danilov AB, Tkacheva ON, Gromova OA, Solovieva EY, Dudinskaya EN, Rozanov AV, Kartashova EA. [Influence of chronic pain in osteoarthritis on the risk of cardiovascular diseases and modern methods of drug prevention]. Zh Nevrol Psikhiatr Im S S Korsakova 2023; 123:20-30. [PMID: 37315238 DOI: 10.17116/jnevro202312305120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
The purpose of the review of scientific medical literature was to evaluate the data of the epidemiology of osteoarthritis (OA) and cardiovascular diseases (CVD) with the analysis of risk factors, pathophysiological and pathobiochemical mechanisms of the relationship between OA and the risk of developing CVD in the presence of chronic pain, modern strategies for screening and management of this cohort of patients, the mechanism of action and pharmacological effects of chondroitin sulfate (CS). Conclusions were drawn about the need for additional clinical and observational studies of the efficacy and safety of the parenteral form of CS (Chondroguard) in patients with chronic pain in OA and CVD, improvement of clinical recommendations for the treatment of chronic pain in patients with OA and cardiovascular risk, with special attention to interventions that eliminate mobility restrictions in patients and the inclusion of basic and adjuvant therapy with DMOADs to achieve the goals of multipurpose monotherapy in patients with contraindications to standard therapy drugs.
Collapse
Affiliation(s)
- I V Sarvilina
- Medical Center «Novomedicina» LLC, Rostov-on-Don, Russia
| | - Al B Danilov
- Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - O N Tkacheva
- Russian Clinical and Research Center of Gerontology - Pirogov Russian National Research Medical University, Moscow, Russia
| | - O A Gromova
- Federal Research Center «Computer Science and Control», Moscow, Russia
| | - E Yu Solovieva
- Pirogov Russian National Research Medical University, Moscow, Russia
| | - E N Dudinskaya
- Russian Clinical and Research Center of Gerontology - Pirogov Russian National Research Medical University, Moscow, Russia
| | - A V Rozanov
- Russian Clinical and Research Center of Gerontology - Pirogov Russian National Research Medical University, Moscow, Russia
| | | |
Collapse
|
23
|
Liu H, Witzigreuter L, Sathiaseelan R, Agbaga MP, Brush RS, Stout MB, Zhu S. Obesity promotes lipid accumulation in mouse cartilage-A potential role of acetyl-CoA carboxylase (ACC) mediated chondrocyte de novo lipogenesis. J Orthop Res 2022; 40:2771-2779. [PMID: 35279877 PMCID: PMC9647658 DOI: 10.1002/jor.25322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Revised: 01/27/2022] [Accepted: 03/01/2022] [Indexed: 02/04/2023]
Abstract
Obesity promotes the development of osteoarthritis (OA). It is also well-established that obesity leads to excessive lipid deposition in nonadipose tissues, which often induces lipotoxicity. The objective of this study was to investigate changes in the levels of various lipids in mouse cartilage in the context of obesity and determine if chondrocyte de novo lipogenesis is altered. We used Oil Red O to determine the accumulation of lipid droplets in cartilage from mice fed high-fat diet (HFD) or low-fat diet (LFD). We further used mass spectrometry-based lipidomic analyses to quantify levels of different lipid species. Expression of genes involving in fatty acid (FA) uptake, synthesis, elongation, and desaturation were examined using quantitative polymerase chain reaction. To further study the potential mechanisms, we cultured primary mouse chondrocytes under high-glucose and high-insulin conditions to mimic the local microenvironment associated with obesity and subsequently examined the abundance of cellular lipid droplets. The acetyl-CoA carboxylase (ACC) inhibitor, ND-630, was added to the culture medium to examine the effect of inhibiting de novo lipogenesis on lipid accumulation in chondrocytes. When compared to the mice receiving LFD, the HFD group displayed more chondrocytes with visible intracellular lipid droplets. Significantly higher amounts of total FAs were also detected in the HFD group. Five out of six significantly upregulated FAs were ω-6 FAs, while the two significantly downregulated FAs were ω-3 FAs. Consequently, the HFD group displayed a significantly higher ω-6/ω-3 FA ratio. Ether linked phosphatidylcholine was also found to be higher in the HFD group. Fatty acid desaturase (Fad1-3), fatty acid-binding protein 4 (Fabp4), and fatty acid synthase (Fasn) transcripts were not found to be different between the treatment groups and fatty acid elongase (Elovl1-7) transcripts were undetectable in cartilage. Ceramide synthase 2 (Cers-2), the only transcript found to be changed in these studies, was significantly upregulated in the HFD group. In vitro, chondrocytes upregulated de novo lipogenesis when cultured under high-glucose, high-insulin conditions, and this observation was associated with the activation of ACC, which was attenuated by the addition of ND-630. This study provides the first evidence that lipid deposition is increased in cartilage with obesity and that this is associated with the upregulation of ACC-mediated de novo lipogenesis. This was supported by our observation that ACC inhibition ameliorated lipid accumulation in chondrocytes, thereby suggesting that ACC could potentially be targeted to treat obesity-associated OA.
Collapse
Affiliation(s)
- Huanhuan Liu
- Department of Biomedical Sciences, Ohio University, OH, 45701, USA
- Ohio Musculoskeletal and Neurological Institute (OMNI), Ohio University, OH, 45701, USA
| | - Luke Witzigreuter
- Department of Biological Sciences, Ohio University, Athens, OH, 45701, USA
| | - Roshini Sathiaseelan
- Department of Nutritional Sciences, University of Oklahoma Health Sciences Center, OK, 73117, USA
| | - Martin-Paul Agbaga
- Department of Ophthalmology, University of Oklahoma Health Sciences Center, OK, 73104, USA
- Dean A. McGee Eye Institute, OK, 73104, USA
| | - Richard S. Brush
- Department of Ophthalmology, University of Oklahoma Health Sciences Center, OK, 73104, USA
- Dean A. McGee Eye Institute, OK, 73104, USA
| | - Michael B. Stout
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA
| | - Shouan Zhu
- Department of Biomedical Sciences, Ohio University, OH, 45701, USA
- Ohio Musculoskeletal and Neurological Institute (OMNI), Ohio University, OH, 45701, USA
| |
Collapse
|
24
|
Ben-Trad L, Matei CI, Sava MM, Filali S, Duclos ME, Berthier Y, Guichardant M, Bernoud-Hubac N, Maniti O, Landoulsi A, Blanchin MG, Miossec P, Granjon T, Trunfio-Sfarghiu AM. Synovial Extracellular Vesicles: Structure and Role in Synovial Fluid Tribological Performances. Int J Mol Sci 2022; 23:ijms231911998. [PMID: 36233300 PMCID: PMC9570016 DOI: 10.3390/ijms231911998] [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] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 09/30/2022] [Accepted: 10/04/2022] [Indexed: 11/06/2022] Open
Abstract
The quality of the lubricant between cartilaginous joint surfaces impacts the joint’s mechanistic properties. In this study, we define the biochemical, ultrastructural, and tribological signatures of synovial fluids (SF) from patients with degenerative (osteoarthritis-OA) or inflammatory (rheumatoid arthritis-RA) joint pathologies in comparison with SF from healthy subjects. Phospholipid (PL) concentration in SF increased in pathological contexts, but the proportion PL relative to the overall lipids decreased. Subtle changes in PL chain composition were attributed to the inflammatory state. Transmission electron microscopy showed the occurrence of large multilamellar synovial extracellular vesicles (EV) filled with glycoprotein gel in healthy subjects. Synovial extracellular vesicle structure was altered in SF from OA and RA patients. RA samples systematically showed lower viscosity than healthy samples under a hydrodynamic lubricating regimen whereas OA samples showed higher viscosity. In turn, under a boundary regimen, cartilage surfaces in both pathological situations showed high wear and friction coefficients. Thus, we found a difference in the biochemical, tribological, and ultrastructural properties of synovial fluid in healthy people and patients with osteoarthritis and arthritis of the joints, and that large, multilamellar vesicles are essential for good boundary lubrication by ensuring a ball-bearing effect and limiting the destruction of lipid layers at the cartilage surface.
Collapse
Affiliation(s)
- Layth Ben-Trad
- Laboratory of Contact and Structural Mechanics, University of Lyon, CNRS, INSA Lyon, UMR5259, Villeurbanne, 69100 Lyon, France
- Institute de Chimie et Biochimie Moléculaires et Supramoléculaires, ICBMS UMR 5246, University of Lyon, Université Lyon 1, CNRS, 69622 Lyon, France
- Faculty of Sciences of Bizerte, University of Carthage, Laboratory of Risques Liés aux Stress Environnementaux: Lutte et Prévention, Zarzouna 1054, Tunisia
- Institut Multidisciplinaire de Biochimie des Lipides, 69621 Villeurbanne, France
| | - Constantin Ionut Matei
- Laboratory of Contact and Structural Mechanics, University of Lyon, CNRS, INSA Lyon, UMR5259, Villeurbanne, 69100 Lyon, France
- Institut Multidisciplinaire de Biochimie des Lipides, 69621 Villeurbanne, France
- Institute Lumiere Mat, University of Lyon, CNRS, UCBL, ILM, UMR5506, 69622 Villeurbanne, France
| | - Mirela Maria Sava
- Laboratory of Contact and Structural Mechanics, University of Lyon, CNRS, INSA Lyon, UMR5259, Villeurbanne, 69100 Lyon, France
- Institut Multidisciplinaire de Biochimie des Lipides, 69621 Villeurbanne, France
| | - Samira Filali
- Unit of Immunogenetics & Inflammation EA-4130 & Department of Clinical Immunology and Rheumatology, University of Lyon, Hôpital Edouard Herriot, 69437 Lyon, France
| | - Marie-Eve Duclos
- Charles River Laboratories, 13, Allée de Nudlingen, 27950 Saint-Marcel, France
| | - Yves Berthier
- Laboratory of Contact and Structural Mechanics, University of Lyon, CNRS, INSA Lyon, UMR5259, Villeurbanne, 69100 Lyon, France
- Institut Multidisciplinaire de Biochimie des Lipides, 69621 Villeurbanne, France
| | - Michel Guichardant
- Laboratory of Contact and Structural Mechanics, University of Lyon, CNRS, INSA Lyon, UMR5259, Villeurbanne, 69100 Lyon, France
- Institut Multidisciplinaire de Biochimie des Lipides, 69621 Villeurbanne, France
| | - Nathalie Bernoud-Hubac
- Laboratory of Contact and Structural Mechanics, University of Lyon, CNRS, INSA Lyon, UMR5259, Villeurbanne, 69100 Lyon, France
- Institut Multidisciplinaire de Biochimie des Lipides, 69621 Villeurbanne, France
| | - Ofelia Maniti
- Institute de Chimie et Biochimie Moléculaires et Supramoléculaires, ICBMS UMR 5246, University of Lyon, Université Lyon 1, CNRS, 69622 Lyon, France
- Institut Multidisciplinaire de Biochimie des Lipides, 69621 Villeurbanne, France
| | - Ahmed Landoulsi
- Faculty of Sciences of Bizerte, University of Carthage, Laboratory of Risques Liés aux Stress Environnementaux: Lutte et Prévention, Zarzouna 1054, Tunisia
| | | | - Pierre Miossec
- Unit of Immunogenetics & Inflammation EA-4130 & Department of Clinical Immunology and Rheumatology, University of Lyon, Hôpital Edouard Herriot, 69437 Lyon, France
- Correspondence: (P.M.); (T.G.); Tel.: +33-472-431-503 (T.G.)
| | - Thierry Granjon
- Institute de Chimie et Biochimie Moléculaires et Supramoléculaires, ICBMS UMR 5246, University of Lyon, Université Lyon 1, CNRS, 69622 Lyon, France
- Institut Multidisciplinaire de Biochimie des Lipides, 69621 Villeurbanne, France
- Correspondence: (P.M.); (T.G.); Tel.: +33-472-431-503 (T.G.)
| | - Ana-Maria Trunfio-Sfarghiu
- Laboratory of Contact and Structural Mechanics, University of Lyon, CNRS, INSA Lyon, UMR5259, Villeurbanne, 69100 Lyon, France
- Institut Multidisciplinaire de Biochimie des Lipides, 69621 Villeurbanne, France
| |
Collapse
|
25
|
Lysophosphatidylcholine 16:0 mediates chronic joint pain associated to rheumatic diseases through acid-sensing ion channel 3. Pain 2022; 163:1999-2013. [PMID: 35086123 PMCID: PMC9479040 DOI: 10.1097/j.pain.0000000000002596] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 12/08/2021] [Indexed: 02/04/2023]
Abstract
ABSTRACT Rheumatic diseases are often associated to debilitating chronic pain, which remains difficult to treat and requires new therapeutic strategies. We had previously identified lysophosphatidylcholine (LPC) in the synovial fluids from few patients and shown its effect as a positive modulator of acid-sensing ion channel 3 (ASIC3) able to induce acute cutaneous pain in rodents. However, the possible involvement of LPC in chronic joint pain remained completely unknown. Here, we show, from 2 independent cohorts of patients with painful rheumatic diseases, that the synovial fluid levels of LPC are significantly elevated, especially the LPC16:0 species, compared with postmortem control subjects. Moreover, LPC16:0 levels correlated with pain outcomes in a cohort of osteoarthritis patients. However, LPC16:0 do not appear to be the hallmark of a particular joint disease because similar levels are found in the synovial fluids of a second cohort of patients with various rheumatic diseases. The mechanism of action was next explored by developing a pathology-derived rodent model. Intra-articular injections of LPC16:0 is a triggering factor of chronic joint pain in both male and female mice, ultimately leading to persistent pain and anxiety-like behaviors. All these effects are dependent on ASIC3 channels, which drive sufficient peripheral inputs to generate spinal sensitization processes. This study brings evidences from mouse and human supporting a role for LPC16:0 via ASIC3 channels in chronic pain arising from joints, with potential implications for pain management in osteoarthritis and possibly across other rheumatic diseases.
Collapse
|
26
|
Lin W, Kampf N, Klein J. Neutral polyphosphocholine-modified liposomes as boundary superlubricants. Colloids Surf A Physicochem Eng Asp 2022. [DOI: 10.1016/j.colsurfa.2022.129218] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
27
|
Petan T, Manček-Keber M. Half is enough: Oxidized lysophospholipids as novel bioactive molecules. Free Radic Biol Med 2022; 188:351-362. [PMID: 35779690 DOI: 10.1016/j.freeradbiomed.2022.06.228] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 06/01/2022] [Accepted: 06/13/2022] [Indexed: 10/17/2022]
Abstract
Studies in the last decade have established the roles of oxidized phospholipids as modulators of various cellular processes, from inflammation and immunity to cell death. Oxidized lysophospholipids, formed through the activity of phospholipases and oxidative enzymes and lacking an acyl chain in comparison with parent phospholipids, are now emerging as novel bioactive lipid mediators. Their detection and structural characterization have been limited in the past due to low amounts and the complexity of their biosynthetic and removal pathways, but recent studies have unequivocally demonstrated their formation under inflammatory conditions. The involvement of oxidized lysophospholipids in immune regulation classifies them as damage-associated molecular patterns (DAMPs), which can promote sterile inflammation and contribute to autoimmune and chronic diseases as well as aging-related diseases. Their signaling pathways are just beginning to be revealed. As the first publications indicate that oxidized lysophospholipids use the same receptors as pathogen-associated molecular patterns (PAMPs), it is likely that the inhibition of signaling pathways activated by oxidized lysophospholipids would affect innate immunity per se. On the other hand, inhibition or modulation of their enzymatic formation, which would not interfere with the response to pathogens, might be beneficial and is potentially a promising new field of research.
Collapse
Affiliation(s)
- Toni Petan
- Department of Molecular and Biomedical Sciences, Jožef Stefan Institute, 1000, Ljubljana, Slovenia.
| | - Mateja Manček-Keber
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, 1000, Ljubljana, Slovenia.
| |
Collapse
|
28
|
Jurczak A, Delay L, Barbier J, Simon N, Krock E, Sandor K, Agalave NM, Rudjito R, Wigerblad G, Rogóż K, Briat A, Miot-Noirault E, Martinez-Martinez A, Brömme D, Grönwall C, Malmström V, Klareskog L, Khoury S, Ferreira T, Labrum B, Deval E, Jiménez-Andrade JM, Marchand F, Svensson CI. Antibody-induced pain-like behavior and bone erosion: links to subclinical inflammation, osteoclast activity, and acid-sensing ion channel 3-dependent sensitization. Pain 2022; 163:1542-1559. [PMID: 34924556 PMCID: PMC9341234 DOI: 10.1097/j.pain.0000000000002543] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Revised: 09/28/2021] [Accepted: 09/30/2021] [Indexed: 11/27/2022]
Abstract
ABSTRACT Several bone conditions, eg, bone cancer, osteoporosis, and rheumatoid arthritis (RA), are associated with a risk of developing persistent pain. Increased osteoclast activity is often the hallmark of these bony pathologies and not only leads to bone remodeling but is also a source of pronociceptive factors that sensitize the bone-innervating nociceptors. Although historically bone loss in RA has been believed to be a consequence of inflammation, both bone erosion and pain can occur years before the symptom onset. Here, we have addressed the disconnection between inflammation, pain, and bone erosion by using a combination of 2 monoclonal antibodies isolated from B cells of patients with RA. We have found that mice injected with B02/B09 monoclonal antibodies (mAbs) developed a long-lasting mechanical hypersensitivity that was accompanied by bone erosion in the absence of joint edema or synovitis. Intriguingly, we have noted a lack of analgesic effect of naproxen and a moderate elevation of few inflammatory factors in the ankle joints suggesting that B02/B09-induced pain-like behavior does not depend on inflammatory processes. By contrast, we found that inhibiting osteoclast activity and acid-sensing ion channel 3 signaling prevented the development of B02/B09-mediated mechanical hypersensitivity. Moreover, we have identified secretory phospholipase A2 and lysophosphatidylcholine 16:0 as critical components of B02/B09-induced pain-like behavior and shown that treatment with a secretory phospholipase A2 inhibitor reversed B02/B09-induced mechanical hypersensitivity and bone erosion. Taken together, our study suggests a potential link between bone erosion and pain in a state of subclinical inflammation and offers a step forward in understanding the mechanisms of bone pain in diseases such as RA.
Collapse
Affiliation(s)
- Alexandra Jurczak
- Department of Physiology and Pharmacology, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Lauriane Delay
- Université Clermont Auvergne, Inserm U1107 Neuro-Dol, Pharmacologie Fondamentale et Clinique de la Douleur, Clermont-Ferrand, France
| | - Julie Barbier
- Université Clermont Auvergne, Inserm U1107 Neuro-Dol, Pharmacologie Fondamentale et Clinique de la Douleur, Clermont-Ferrand, France
| | - Nils Simon
- Department of Physiology and Pharmacology, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Emerson Krock
- Department of Physiology and Pharmacology, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Katalin Sandor
- Department of Physiology and Pharmacology, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Nilesh M. Agalave
- Department of Physiology and Pharmacology, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Resti Rudjito
- Department of Physiology and Pharmacology, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Gustaf Wigerblad
- Department of Physiology and Pharmacology, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Katarzyna Rogóż
- Department of Physiology and Pharmacology, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Arnaud Briat
- Université Clermont Auvergne, Inserm UMR 1240, IMoST, Imagerie Moléculaire et Stratégies Théranostiques, Clermont-Ferrand, France
| | - Elisabeth Miot-Noirault
- Université Clermont Auvergne, Inserm UMR 1240, IMoST, Imagerie Moléculaire et Stratégies Théranostiques, Clermont-Ferrand, France
| | - Arisai Martinez-Martinez
- Unidad Academica Multidisciplinaria Reynosa Aztlan, Universidad Autonoma de Tamaulipas, Reynosa, Tamaulipas, Mexico
| | - Dieter Brömme
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| | - Caroline Grönwall
- Department of Medicine, Division of Rheumatology, Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Vivianne Malmström
- Department of Medicine, Division of Rheumatology, Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Lars Klareskog
- Department of Medicine, Division of Rheumatology, Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Spiro Khoury
- Lipotoxicity and Channelopathies (LiTch)—ConicMeds, Université de Poitiers, Poitiers, France
| | - Thierry Ferreira
- Lipotoxicity and Channelopathies (LiTch)—ConicMeds, Université de Poitiers, Poitiers, France
| | - Bonnie Labrum
- Université Côte d’Azur, CNRS, IPMC, LabEx ICST, FHU InovPain, France
| | - Emmanuel Deval
- Université Côte d’Azur, CNRS, IPMC, LabEx ICST, FHU InovPain, France
| | - Juan Miguel Jiménez-Andrade
- Unidad Academica Multidisciplinaria Reynosa Aztlan, Universidad Autonoma de Tamaulipas, Reynosa, Tamaulipas, Mexico
| | - Fabien Marchand
- Université Clermont Auvergne, Inserm U1107 Neuro-Dol, Pharmacologie Fondamentale et Clinique de la Douleur, Clermont-Ferrand, France
| | - Camilla I. Svensson
- Department of Physiology and Pharmacology, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
29
|
Kim M, Koyama E, Saunders CM, Querido W, Pleshko N, Pacifici M. Synovial joint cavitation initiates with microcavities in interzone and is coupled to skeletal flexion and elongation in developing mouse embryo limbs. Biol Open 2022; 11:bio059381. [PMID: 35608281 PMCID: PMC9212078 DOI: 10.1242/bio.059381] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 05/16/2022] [Indexed: 11/20/2022] Open
Abstract
The synovial cavity and its fluid are essential for joint function and lubrication, but their developmental biology remains largely obscure. Here, we analyzed E12.5 to E18.5 mouse embryo hindlimbs and discovered that cavitation initiates around E15.0 with emergence of multiple, discrete, µm-wide tissue discontinuities we term microcavities in interzone, evolving into a single joint-wide cavity within 12 h in knees and within 72-84 h in interphalangeal joints. The microcavities were circumscribed by cells as revealed by mTmG imaging and exhibited a carbohydrate and protein content based on infrared spectral imaging at micro and nanoscale. Accounting for differing cavitation kinetics, we found that the growing femur and tibia anlagen progressively flexed at the knee over time, with peak angulation around E15.5 exactly when the full knee cavity consolidated; however, interphalangeal joint geometry changed minimally over time. Indeed, cavitating knee interzone cells were elongated along the flexion angle axis and displayed oblong nuclei, but these traits were marginal in interphalangeal cells. Conditional Gdf5Cre-driven ablation of Has2 - responsible for production of the joint fluid component hyaluronic acid (HA) - delayed the cavitation process. Our data reveal that cavitation is a stepwise process, brought about by sequential action of microcavities, skeletal flexion and elongation, and HA accumulation. This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Minwook Kim
- Translational Research Program in Pediatric Orthopaedics, Division of Orthopaedic Surgery, Department of Surgery, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Eiki Koyama
- Translational Research Program in Pediatric Orthopaedics, Division of Orthopaedic Surgery, Department of Surgery, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Cheri M. Saunders
- Translational Research Program in Pediatric Orthopaedics, Division of Orthopaedic Surgery, Department of Surgery, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - William Querido
- Department of Bioengineering, Temple University, Philadelphia, PA 19122, USA
| | - Nancy Pleshko
- Department of Bioengineering, Temple University, Philadelphia, PA 19122, USA
| | - Maurizio Pacifici
- Translational Research Program in Pediatric Orthopaedics, Division of Orthopaedic Surgery, Department of Surgery, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| |
Collapse
|
30
|
Di Francesco M, Fragassi A, Pannuzzo M, Ferreira M, Brahmachari S, Decuzzi P. Management of osteoarthritis: From drug molecules to nano/micromedicines. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2022; 14:e1780. [PMID: 35253405 PMCID: PMC9285805 DOI: 10.1002/wnan.1780] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 12/29/2021] [Accepted: 01/21/2022] [Indexed: 12/12/2022]
Abstract
With the change in lifestyle and aging of the population, osteoarthritis (OA) is emerging as a major medical burden globally. OA is a chronic inflammatory and degenerative disease initially manifesting with joint pain and eventually leading to permanent disability. To date, there are no drugs available for the definitive treatment of osteoarthritis and most therapies have been palliative in nature by alleviating symptoms rather than curing the disease. This coupled with the vague understanding of the early symptoms and methods of diagnosis so that the disease continues as a global problem and calls for concerted research efforts. A cascade of events regulates the onset and progression of osteoarthritis starting with the production of proinflammatory cytokines, including interleukin (IL)‐1β, IL‐6, tumor necrosis factor (TNF)‐α; catabolic enzymes, such as matrix metalloproteinases (MMPs)‐1, ‐3, and ‐13, culminating into cartilage breakdown, loss of lubrication, pain, and inability to load the joint. Although intra‐articular injections of small and macromolecules are often prescribed to alleviate symptoms, low residence times within the synovial cavity severely impair their efficacy. This review will briefly describe the factors dictating the onset and progression of the disease, present the current clinically approved methods for its treatment and diagnosis, and finally elaborate on the main challenges and opportunities for the application of nano/micromedicines in the treatment of osteoarthritis. Thus, future treatment regimens will benefit from simultaneous consideration of the mechanobiological, the inflammatory, and tissue degradation aspects of the disease. This article is categorized under:Nanotechnology Approaches to Biology > Nanoscale Systems in Biology Implantable Materials and Surgical Technologies > Nanotechnology in Tissue Repair and Replacement
Collapse
Affiliation(s)
- Martina Di Francesco
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Genoa, Italy
| | - Agnese Fragassi
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Genoa, Italy.,Department of Chemistry and Industrial Chemistry, University of Genova, Genoa, Italy
| | - Martina Pannuzzo
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Genoa, Italy
| | - Miguel Ferreira
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Genoa, Italy
| | - Sayanti Brahmachari
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Genoa, Italy
| | - Paolo Decuzzi
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Genoa, Italy
| |
Collapse
|
31
|
Batushansky A, Zhu S, Komaravolu RK, South S, Mehta-D'souza P, Griffin TM. Fundamentals of OA. An initiative of Osteoarthritis and Cartilage. Obesity and metabolic factors in OA. Osteoarthritis Cartilage 2022; 30:501-515. [PMID: 34537381 PMCID: PMC8926936 DOI: 10.1016/j.joca.2021.06.013] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 05/14/2021] [Accepted: 06/07/2021] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Obesity was once considered a risk factor for knee osteoarthritis (OA) primarily for biomechanical reasons. Here we provide an additional perspective by discussing how obesity also increases OA risk by altering metabolism and inflammation. DESIGN This narrative review is presented in four sections: 1) metabolic syndrome and OA, 2) metabolic biomarkers of OA, 3) evidence for dysregulated chondrocyte metabolism in OA, and 4) metabolic inflammation: joint tissue mediators and mechanisms. RESULTS Metabolic syndrome and its components are strongly associated with OA. However, evidence for a causal relationship is context dependent, varying by joint, gender, diagnostic criteria, and demographics, with additional environmental and genetic interactions yet to be fully defined. Importantly, some aspects of the etiology of obesity-induced OA appear to be distinct between men and women, especially regarding the role of adipose tissue. Metabolomic analyses of serum and synovial fluid have identified potential diagnostic biomarkers of knee OA and prognostic biomarkers of disease progression. Connecting these biomarkers to cellular pathophysiology will require future in vivo studies of joint tissue metabolism. Such studies will help reveal when a metabolic process or a metabolite itself is a causal factor in disease progression. Current evidence points towards impaired chondrocyte metabolic homeostasis and metabolic-immune dysregulation as likely factors connecting obesity to the increased risk of OA. CONCLUSIONS A deeper understanding of how obesity alters metabolic and inflammatory pathways in synovial joint tissues is expected to provide new therapeutic targets and an improved definition of "metabolic" and "obesity" OA phenotypes.
Collapse
Affiliation(s)
- A Batushansky
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA.
| | - S Zhu
- Department of Biomedical Sciences, Ohio Musculoskeletal and Neurological Institute (OMNI), Ohio University, Athens, OH, 45701, USA.
| | - R K Komaravolu
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA.
| | - S South
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA.
| | - P Mehta-D'souza
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA.
| | - T M Griffin
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA; Reynolds Oklahoma Center on Aging, Department of Biochemistry and Molecular Biology, Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA; Veterans Affairs Medical Center, Oklahoma City, OK, 73104, USA.
| |
Collapse
|
32
|
Cao Y, Klein J. Lipids and lipid mixtures in boundary layers: From hydration lubrication to osteoarthritis. Curr Opin Colloid Interface Sci 2022. [DOI: 10.1016/j.cocis.2021.101559] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
33
|
Thottakkattumana Parameswaran V, Hild C, Eichner G, Ishaque B, Rickert M, Steinmeyer J. Interleukin-1 Induces the Release of Lubricating Phospholipids from Human Osteoarthritic Fibroblast-like Synoviocytes. Int J Mol Sci 2022; 23:2409. [PMID: 35269552 PMCID: PMC8910712 DOI: 10.3390/ijms23052409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 02/09/2022] [Accepted: 02/19/2022] [Indexed: 12/04/2022] Open
Abstract
(1) Background: Synovial fluid (SF) from knee joints with osteoarthritis (OA) has increased levels of phospholipids (PL). We have reported earlier that TGF-ß and IGF-1 stimulate fibroblast-like synoviocytes (FLS) to synthesize increased amounts of PLs. The current study examined whether IL-1ß induces the release of PLs in FLS and the underlying mechanism. (2) Methods: Cultured human OA FLS were treated with IL-1ß alone and with pathway inhibitors or with synthetic liver X receptor (LXR) agonists. Cholesterol hydroxylases, ABC transporters, apolipoproteins (APO), LXR, sterol regulatory binding proteins (SREBPs), and 3-hydroxy-3-methylglutaryl-coenzyme A reductase (HMGCR) were analyzed by RT-PCR, Western blot, and ELISA. The release of radiolabeled PLs from FLS was determined, and statistical analysis was performed using R (N = 5-9). (3) Results: Like synthetic LXR agonists, IL-1ß induced a 1.4-fold greater release of PLs from FLS. Simultaneously, IL-1ß upregulated the level of the PL transporter ABCA1 and of cholesterol hydroxylases CH25H and CYP7B1. IL-1ß and T0901317 stimulated the expression of SREBP1c, whereas only T0901317 enhanced SREBP2, HMGCR, APOE, LXRα, and ABCG1 additionally. (4) Conclusions: IL-1ß partially controls PL levels in OA-SF by affecting the release of PLs from FLS. Our data show that IL-1ß upregulates cholesterol hydroxylases and thus the formation of oxysterols, which, as natural agonists of LXR, increase the level of active ABCA1, in turn enhancing the release of PLs.
Collapse
Affiliation(s)
- Vishnu Thottakkattumana Parameswaran
- Laboratory for Experimental Orthopaedics, Department of Orthopaedics, Justus Liebig University Giessen, 35392 Giessen, Germany; (V.T.P.); (C.H.); (B.I.); (M.R.)
| | - Christiane Hild
- Laboratory for Experimental Orthopaedics, Department of Orthopaedics, Justus Liebig University Giessen, 35392 Giessen, Germany; (V.T.P.); (C.H.); (B.I.); (M.R.)
| | - Gerrit Eichner
- Mathematical Institute, Justus Liebig University Giessen, 35392 Giessen, Germany;
| | - Bernd Ishaque
- Laboratory for Experimental Orthopaedics, Department of Orthopaedics, Justus Liebig University Giessen, 35392 Giessen, Germany; (V.T.P.); (C.H.); (B.I.); (M.R.)
| | - Markus Rickert
- Laboratory for Experimental Orthopaedics, Department of Orthopaedics, Justus Liebig University Giessen, 35392 Giessen, Germany; (V.T.P.); (C.H.); (B.I.); (M.R.)
| | - Juergen Steinmeyer
- Laboratory for Experimental Orthopaedics, Department of Orthopaedics, Justus Liebig University Giessen, 35392 Giessen, Germany; (V.T.P.); (C.H.); (B.I.); (M.R.)
| |
Collapse
|
34
|
Metabolomics of Synovial Fluid and Infrapatellar Fat Pad in Patients with Osteoarthritis or Rheumatoid Arthritis. Inflammation 2022; 45:1101-1117. [PMID: 35041143 PMCID: PMC9095531 DOI: 10.1007/s10753-021-01604-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 11/17/2021] [Accepted: 11/19/2021] [Indexed: 12/13/2022]
Abstract
Osteoarthritis (OA) and autoimmune-driven rheumatoid arthritis (RA) are inflammatory joint diseases with complex and insufficiently understood pathogeneses. Our objective was to characterize the metabolic fingerprints of synovial fluid (SF) and its adjacent infrapatellar fat pad (IFP) obtained during the same surgical operation from OA and RA knees. Non-targeted metabolite profiling was performed for 5 non-inflammatory trauma controls, 10 primary OA (pOA) patients, and 10 seropositive RA patients with high-resolution mass spectrometry-based techniques, and metabolites were matched with known metabolite identities. Groupwise differences in metabolic features were analyzed with the univariate Welch’s t-test and the multivariate linear discriminant analysis (LDA) and principal component analysis (PCA). Significant discrimination of metabolite profiles was discovered by LDA for both SF and IFP and by PCA for SF based on diagnosis. In addition to a few drug-derived substances, there were 16 and 13 identified metabolites with significant differences between the diagnoses in SF and IFP, respectively. The pathways downregulated in RA included androgen, bile acid, amino acid, and histamine metabolism, and those upregulated included biotin metabolism in pOA and purine metabolism in RA and pOA. The RA-induced downregulation of androgen and bile acid metabolism was observed for both SF and IFP. The levels of 11 lipid metabolites, mostly glycerophospholipids and fatty acid amides, were also altered by these inflammatory conditions. The identified metabolic pathways could be utilized in the future to deepen our understanding of the pathogeneses of OA and RA and to develop not only biomarkers for their early diagnosis but also therapeutic targets.
Collapse
|
35
|
Lin W, Goldberg R, Klein J. Poly-phosphocholination of liposomes leads to highly-extended retention time in mice joints. J Mater Chem B 2022; 10:2820-2827. [PMID: 35099493 PMCID: PMC9007059 DOI: 10.1039/d1tb02346b] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Surface-attached layers of phosphatidylcholine (PC) lipid vesicles (liposomes) may reduce the friction coefficient μ (= force-to-slide/load) between the sliding surfaces down to μ ≈ 10−3–10−4 up to tens of atm contact pressures, as high as those in the major joints (hips or knees). Such friction reduction is attributed to hydration lubrication by the highly-hydrated phosphocholine head-groups exposed at the outer vesicle surfaces. It has been suggested therefore that intra-articular (IA) administration of liposomes as potential boundary lubricants may alleviate degenerative, friction-associated joint conditions such as osteoarthritis (OA), which is associated with insufficient lubrication at the articular cartilage surface. To overcome the problem, common to all nanoparticles, of rapid removal by the mononuclear phagocyte system, as well as to ensure long-term colloidal stability during storage, functionalizing liposomes with poly(ethylene glycol) moieties, PEGylation, is often used. Here we describe a different liposome functionalization approach, using poly(2-methacryloyloxyethyl phosphorylcholine), PMPC, moieties (strictly, lipid–PMPC conjugates), and compare the retention time in mice joints of such PMPCylated liposomes with otherwise-identical but PEGylated vesicles following IA administration. We find, using fluorescence labeling and in vivo optical imaging, that when PMPC-stabilized liposomes are injected into mice knee joints, there is a massive increase of the vesicles’ retention half-life in the joints of about (4–5)-fold (ca. 300–400% increase in retention time) compared with the PEGylated liposomes (and some 100-fold longer than the retention time of intra-articularly injected hyaluronan or HA). Such PMPCylated liposomes are therefore promising candidates as potential long-lived boundary lubricants at the articular cartilage surface, with implication for friction-associated pathologies. Moreover, as lipid vesicles are well known to be efficient drug carriers, such long retention in the joints may enable analgesic or anti-inflammatory agents for joint pathologies to be more efficiently delivered via IA administration using PMPCylated liposomal vehicles relative to PEGylated ones. PMPCylated liposomes injected into mice joints show a massive increase in retention half-life compared with PEGylated liposomes (or hyaluronan, HA), making them promising candidates as boundary lubricants at articular cartilage, or as drug carriers.![]()
Collapse
Affiliation(s)
- Weifeng Lin
- Department of Molecular Chemistry and Materials Science, Weizmann Institute of Science, Rehovot 76100, Israel.
| | - Ronit Goldberg
- Department of Molecular Chemistry and Materials Science, Weizmann Institute of Science, Rehovot 76100, Israel.
| | - Jacob Klein
- Department of Molecular Chemistry and Materials Science, Weizmann Institute of Science, Rehovot 76100, Israel.
| |
Collapse
|
36
|
Intra-Articular Drug Delivery for Osteoarthritis Treatment. Pharmaceutics 2021; 13:pharmaceutics13122166. [PMID: 34959445 PMCID: PMC8703898 DOI: 10.3390/pharmaceutics13122166] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 12/13/2021] [Accepted: 12/13/2021] [Indexed: 02/07/2023] Open
Abstract
Osteoarthritis (OA) is the most prevalent degenerative joint disease affecting millions of people worldwide. Currently, clinical nonsurgical treatments of OA are only limited to pain relief, anti-inflammation, and viscosupplementation. Developing disease-modifying OA drugs (DMOADs) is highly demanded for the efficient treatment of OA. As OA is a local disease, intra-articular (IA) injection directly delivers drugs to synovial joints, resulting in high-concentration drugs in the joint and reduced side effects, accompanied with traditional oral or topical administrations. However, the injected drugs are rapidly cleaved. By properly designing the drug delivery systems, prolonged retention time and targeting could be obtained. In this review, we summarize the drugs investigated for OA treatment and recent advances in the IA drug delivery systems, including micro- and nano-particles, liposomes, and hydrogels, hoping to provide some information for designing the IA injected formulations.
Collapse
|
37
|
Rocha B, Illiano A, Calamia V, Pinto G, Amoresano A, Ruiz-Romero C, Blanco FJ. Targeted phospholipidomic analysis of synovial fluid as a tool for osteoarthritis deep phenotyping. OSTEOARTHRITIS AND CARTILAGE OPEN 2021; 3:100219. [DOI: 10.1016/j.ocarto.2021.100219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 08/14/2021] [Accepted: 10/04/2021] [Indexed: 10/20/2022] Open
|
38
|
Loef M, Faquih TO, von Hegedus JH, Ghorasaini M, Ioan-Facsinay A, Kroon FP, Giera M, Kloppenburg M. The lipid profile for the prediction of prednisolone treatment response in patients with inflammatory hand osteoarthritis: The HOPE study. OSTEOARTHRITIS AND CARTILAGE OPEN 2021; 3:100167. [DOI: 10.1016/j.ocarto.2021.100167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Revised: 04/03/2021] [Accepted: 04/09/2021] [Indexed: 10/21/2022] Open
|
39
|
Wang J, Fan Q, Yu T, Zhang Y. Identifying the hub genes and immune cell infiltration in synovial tissue between osteoarthritic and rheumatoid arthritic patients by bioinformatic approach. Curr Pharm Des 2021; 28:497-509. [PMID: 34736376 DOI: 10.2174/1381612827666211104154459] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 06/28/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Osteoarthritis (OA) and rheumatoid arthritis (RA) are two common diseases that result in limb disability and a decrease in quality of life. The major symptoms of OA and RA are pain, swelling, stiffness, and malformation of joints, and each disease also has unique characteristics. OBJECTIVE To compare the pathological mechanisms of OA and RA via weighted correlation network analysis (WGCNA) and immune infiltration analysis and find potential diagnostic and pharmaceutical targets for the treatment of OA and RA. METHODS The gene expression profiles of ten OA and ten RA synovial tissue samples were downloaded from the Gene Expression Omnibus (GEO) database (GSE55235). After obtaining differentially expressed genes (DEGs) via GEO2R, WGCNA was conducted using an R package, and modules and genes that were highly correlated with OA and RA were identified. Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment, and protein-protein interaction (PPI) network analyses were also conducted. Hub genes were identified using the Search Tool for the Retrieval of Interacting Genes (STRING) and Cytoscape software. Immune infiltration analysis was conducted using the Perl program and CIBERSORT software. RESULTS Two hundred ninety-nine DEGs, 24 modules, 16 GO enrichment terms, 6 KEGG pathway enrichment terms, 10 hub genes (CXCL9, CXCL10, CXCR4, CD27, CD69, CD3D, IL7R, STAT1, RGS1, and ISG20), and 8 kinds of different infiltrating immune cells (plasma cells, CD8 T cells, activated memory CD4 T cells, T helper follicular cells, M1 macrophages, Tregs, resting mast cells, and neutrophils) were found to be involved in the different pathological mechanisms of OA and RA. CONCLUSION Inflammation-associated genes were the top differentially expressed hub genes between OA and RA, and their expression was downregulated in OA. Genes associated with lipid metabolism may have upregulated expression in OA. In addition, immune cells that participate in the adaptive immune response play an important role in RA. OA mainly involves immune cells that are associated with the innate immune response.
Collapse
Affiliation(s)
- Junjie Wang
- Qingdao University, Qingdao, Shandong 266000. China
| | - Qin Fan
- Qingdao University, Qingdao, Shandong 266000. China
| | - Tengbo Yu
- Qingdao University, Qingdao, Shandong 266000. China
| | - Yingze Zhang
- Qingdao University, Qingdao, Shandong 266000. China
| |
Collapse
|
40
|
Alterations in the gut microbiota and metabolite profiles of patients with Kashin-Beck disease, an endemic osteoarthritis in China. Cell Death Dis 2021; 12:1015. [PMID: 34711812 PMCID: PMC8553765 DOI: 10.1038/s41419-021-04322-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 10/12/2021] [Accepted: 10/18/2021] [Indexed: 12/13/2022]
Abstract
Kashin-Beck disease (KBD) is a severe osteochondral disorder that may be driven by the interaction between genetic and environmental factors. We aimed to improve our understanding of the gut microbiota structure in KBD patients of different grades and the relationship between the gut microbiota and serum metabolites. Fecal and serum samples collected from KBD patients and normal controls (NCs) were used to characterize the gut microbiota using 16S rDNA gene and metabolomic sequencing via liquid chromatography-mass spectrometry (LC/MS). To identify whether gut microbial changes at the species level are associated with the genes or functions of the gut bacteria in the KBD patients, metagenomic sequencing of fecal samples from grade I KBD, grade II KBD and NC subjects was performed. The KBD group was characterized by elevated levels of Fusobacteria and Bacteroidetes. A total of 56 genera were identified to be significantly differentially abundant between the two groups. The genera Alloprevotella, Robinsoniella, Megamonas, and Escherichia_Shigella were more abundant in the KBD group. Consistent with the 16S rDNA analysis at the genus level, most of the differentially abundant species in KBD subjects belonged to the genus Prevotella according to metagenomic sequencing. Serum metabolomic analysis identified some differentially abundant metabolites among the grade I and II KBD and NC groups that were involved in lipid metabolism metabolic networks, such as that for unsaturated fatty acids and glycerophospholipids. Furthermore, we found that these differences in metabolite levels were associated with altered abundances of specific species. Our study provides a comprehensive landscape of the gut microbiota and metabolites in KBD patients and provides substantial evidence of a novel interplay between the gut microbiome and metabolome in KBD pathogenesis.
Collapse
|
41
|
Zhou G, Lu J, Xu T, Lu Y, Chen W, Wang J, Ke M, Shen Q, Zhu Y, Shan J, Liu S. Clinical lipidomics analysis reveals biomarkers of lipid peroxidation in serum from patients with rheumatoid arthritis. Microchem J 2021. [DOI: 10.1016/j.microc.2021.106607] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
42
|
Bhagavatham SKS, Khanchandani P, Kannan V, Potikuri D, Sridharan D, Pulukool SK, Naik AA, Dandamudi RB, Divi SM, Pargaonkar A, Ray R, Santha SSR, Seshagiri PB, Narasimhan K, Gumdal N, Sivaramakrishnan V. Adenosine deaminase modulates metabolic remodeling and orchestrates joint destruction in rheumatoid arthritis. Sci Rep 2021; 11:15129. [PMID: 34301999 PMCID: PMC8302689 DOI: 10.1038/s41598-021-94607-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Accepted: 07/14/2021] [Indexed: 02/07/2023] Open
Abstract
Rheumatoid Arthritis (RA) is a chronic autoimmune disease associated with inflammation and joint remodeling. Adenosine deaminase (ADA), a risk factor in RA, degrades adenosine, an anti-inflammatory molecule, resulting in an inflammatory bias. We present an integrative analysis of clinical data, cytokines, serum metabolomics in RA patients and mechanistic studies on ADA-mediated effects on in vitro cell culture models. ADA activity differentiated patients into low and high ADA sets. The levels of the cytokines TNFα, IFNγ, IL-10, TGFβ and sRANKL were elevated in RA and more pronounced in high ADA sets. Serum metabolomic analysis shows altered metabolic pathways in RA which were distinct between low and high ADA sets. Comparative analysis with previous studies shows similar pathways are modulated by DMARDs and biologics. Random forest analysis distinguished RA from control by methyl-histidine and hydroxyisocaproic acid, while hexose-phosphate and fructose-6-phosphate distinguished high ADA from low ADA. The deregulated metabolic pathways of High ADA datasets significantly overlapped with high ADA expressing PBMCs GEO transcriptomics dataset. ADA induced the death of chondrocytes, synoviocyte proliferation, both inflammation in macrophages and their differentiation into osteoclasts and impaired differentiation of mesenchymal stem cells to osteoblasts and mineralization. PBMCs expressing elevated ADA had increased expression of cytokines and P2 receptors compared to synovial macrophages which has low expression of ADA. Our data demonstrates increased cytokine levels and distinct metabolic signatures of RA based on the ADA activity, suggests an important role for ADA in the pathophysiology of RA joints and as a potential marker and therapeutic target in RA patients.
Collapse
Affiliation(s)
- Sai Krishna Srimadh Bhagavatham
- grid.444651.60000 0004 0496 6988Disease Biology Lab, Department of Biosciences, Sri Sathya Sai Institute of Higher Learning, Puttaparthi, 515134 India
| | - Prakash Khanchandani
- grid.496668.30000 0004 1767 3076Department of Orthopedics, Sri Sathya Sai Institute of Higher Medical Sciences, PG, Puttaparthi, 515134 India
| | - Vishnu Kannan
- grid.444651.60000 0004 0496 6988Disease Biology Lab, Department of Biosciences, Sri Sathya Sai Institute of Higher Learning, Puttaparthi, 515134 India ,grid.411552.60000 0004 1766 4022Present Address: Department of Botany/Biotechnology, CMS College, Kottayam, 686001 India
| | | | - Divya Sridharan
- grid.34980.360000 0001 0482 5067Molecular Reproduction and Developmental Genetics, Indian Institute of Science, Bengaluru, 560012 India
| | - Sujith Kumar Pulukool
- grid.444651.60000 0004 0496 6988Disease Biology Lab, Department of Biosciences, Sri Sathya Sai Institute of Higher Learning, Puttaparthi, 515134 India
| | - Ashwin Ashok Naik
- grid.444651.60000 0004 0496 6988Disease Biology Lab, Department of Biosciences, Sri Sathya Sai Institute of Higher Learning, Puttaparthi, 515134 India
| | - Rajesh Babu Dandamudi
- grid.444651.60000 0004 0496 6988Department of Chemistry, Sri Sathya Sai Institute of Higher Learning, Prasanthi Nilayam, India ,Present Address: Phenomenex India, Hyderabad, Telangana 500084 India
| | - Sai Mangala Divi
- grid.496668.30000 0004 1767 3076Department of Biochemistry, Sri Sathya Sai Institute of Higher Medical Sciences, PG, Puttaparthi, 515134 India
| | - Ashish Pargaonkar
- grid.464737.50000 0004 1775 153XAgilent Technologies India Pvt Ltd, Bengaluru, 560048 India
| | - Rahul Ray
- grid.496668.30000 0004 1767 3076Department of Orthopedics, Sri Sathya Sai Institute of Higher Medical Sciences, PG, Puttaparthi, 515134 India
| | - Saibharath Simha Reddy Santha
- grid.444651.60000 0004 0496 6988Disease Biology Lab, Department of Biosciences, Sri Sathya Sai Institute of Higher Learning, Puttaparthi, 515134 India
| | - Polani B. Seshagiri
- grid.34980.360000 0001 0482 5067Molecular Reproduction and Developmental Genetics, Indian Institute of Science, Bengaluru, 560012 India
| | - K. Narasimhan
- Sri Sathya Sai General Hospital, Puttaparthi, 515134 India
| | | | - Venketesh Sivaramakrishnan
- grid.444651.60000 0004 0496 6988Disease Biology Lab, Department of Biosciences, Sri Sathya Sai Institute of Higher Learning, Puttaparthi, 515134 India
| |
Collapse
|
43
|
Haartmans MJJ, Emanuel KS, Tuijthof GJM, Heeren RMA, Emans PJ, Cillero-Pastor B. Mass Spectrometry-based Biomarkers for Knee Osteoarthritis: A Systematic Review. Expert Rev Proteomics 2021; 18:693-706. [PMID: 34228576 DOI: 10.1080/14789450.2021.1952868] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Introduction: Knee osteoarthritis (OA) is a joint disease, affecting multiple tissues in the joint. Early detection and intervention may delay OA development and avoid total knee arthroplasty. Specific biomarker profiles for early detection and guiding clinical decision-making of OA have not yet been identified. One technique that can contribute to the finding of this "OA biomarker" is mass spectrometry (MS), which offers the possibility to analyze different molecules in tissues or fluids. Several proteomic, lipidomic, metabolomic and other -omic approaches aim to identify these molecular profiles; however, variation in methods and techniques complicate the finding of promising candidate biomarkers.Areas covered: In this systematic review, we aim to provide an overview of molecules in OA knee patients. Possible biomarkers in several tissue types of OA and non-OA patients, as well as current limitations and possible future suggestions will be discussed.Expert opinion: According to this review, we do not believe one specific biomarker will function as predictive molecule for OA. Likely, a group of molecules will give insight in OA development and possible therapeutic targets. For clinical implementation of MS-analysis in clinical decision-making, standardized procedures, large cohort studies and sharing protocols and data is necessary.
Collapse
Affiliation(s)
- Mirella J J Haartmans
- Maastricht MultiModal Molecular Imaging Institute (M4i), Division of Imaging Mass Spectrometry, Maastricht University Universiteitssingel 50, 6229 ER Maastricht, The Netherlands.,Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, CAPHRI Care and Public Health Research Institute, Maastricht University Medical Center+, P. Debyelaan 25, 6229 HX Maastricht, The Netherlands
| | - Kaj S Emanuel
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, CAPHRI Care and Public Health Research Institute, Maastricht University Medical Center+, P. Debyelaan 25, 6229 HX Maastricht, The Netherlands
| | - Gabrielle J M Tuijthof
- Department of Research Engineering, Maastricht University Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| | - Ron M A Heeren
- Maastricht MultiModal Molecular Imaging Institute (M4i), Division of Imaging Mass Spectrometry, Maastricht University Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| | - Pieter J Emans
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, CAPHRI Care and Public Health Research Institute, Maastricht University Medical Center+, P. Debyelaan 25, 6229 HX Maastricht, The Netherlands
| | - Berta Cillero-Pastor
- Maastricht MultiModal Molecular Imaging Institute (M4i), Division of Imaging Mass Spectrometry, Maastricht University Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| |
Collapse
|
44
|
Li JT, Zeng N, Yan ZP, Liao T, Ni GX. A review of applications of metabolomics in osteoarthritis. Clin Rheumatol 2021; 40:2569-2579. [PMID: 33219452 DOI: 10.1007/s10067-020-05511-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 11/10/2020] [Accepted: 11/15/2020] [Indexed: 02/08/2023]
Abstract
Osteoarthritis (OA) represents the most prevalent and disabling arthritis worldwide due to its heterogeneous and progressive articular degradation. However, effective and timely diagnosis and fundamental treatment for this disorder are lacking. Metabolomics, a growing field in life science research in recent years, has the potential to detect many metabolites and thus explains the underlying pathophysiological processes. Hence, new specific metabolic markers and related metabolic pathways can be identified for OA. In this review, we aimed to provide an overview of studies related to the metabolomics of OA in animal models and humans to describe the metabolic changes and related pathways for OA. The present metabolomics studies reveal that the pathogenesis of OA may be significantly related to perturbations of amino acid metabolism. These altered amino acids (e.g., branched-chain amino acids, arginine, and alanine), as well as phospholipids, were identified as potential biomarkers to distinguish patients with OA from healthy individuals.
Collapse
Affiliation(s)
- Jie-Ting Li
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, People's Republic of China
| | - Ni Zeng
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, People's Republic of China
| | - Zhi-Peng Yan
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, People's Republic of China
| | - Tao Liao
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, People's Republic of China
| | - Guo-Xin Ni
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing, 100084, People's Republic of China.
| |
Collapse
|
45
|
Synovial Fluid Fatty Acid Profiles Are Differently Altered by Inflammatory Joint Pathologies in the Shoulder and Knee Joints. BIOLOGY 2021; 10:biology10050401. [PMID: 34064447 PMCID: PMC8147852 DOI: 10.3390/biology10050401] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 04/26/2021] [Accepted: 04/29/2021] [Indexed: 12/26/2022]
Abstract
Anomalies of fatty acid (FA) metabolism characterize osteoarthritis (OA) and rheumatoid arthritis (RA) in the knee joint. No previous study has investigated the synovial fluid (SF) FA manifestations in these aging-related inflammatory diseases in the shoulder. The present experiment compared the FA alterations between the shoulder and knee joints in patients with end-stage OA or end-stage RA. SF samples were collected during glenohumeral or knee joint surgery from trauma controls and from OA and RA patients (n = 42). The FA composition of SF total lipids was analyzed by gas chromatography with flame ionization and mass spectrometric detection and compared across cohorts. The FA signatures of trauma controls were mostly uniform in both anatomical locations. RA shoulders were characterized by elevated percentages of 20:4n-6 and 22:6n-3 and with reduced proportions of 18:1n-9. The FA profiles of OA and RA knees were relatively uniform and displayed lower proportions of 18:2n-6, 22:6n-3 and total n-6 polyunsaturated FAs (PUFAs). The results indicate location- and disease-dependent differences in the SF FA composition. These alterations in FA profiles and their potential implications for the production of PUFA-derived lipid mediators may affect joint lubrication, synovial inflammation and pannus formation as well as cartilage and bone degradation and contribute to the pathogeneses of inflammatory joint diseases.
Collapse
|
46
|
Rocha B, Cillero-Pastor B, Ruiz-Romero C, Paine MRL, Cañete JD, Heeren RMA, Blanco FJ. Identification of a distinct lipidomic profile in the osteoarthritic synovial membrane by mass spectrometry imaging. Osteoarthritis Cartilage 2021; 29:750-761. [PMID: 33582239 DOI: 10.1016/j.joca.2020.12.025] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 11/12/2020] [Accepted: 12/16/2020] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Synovial inflammation is one of the most characteristic events in different types of arthritis, including Osteoarthritis (OA). Emerging evidence also suggests the involvement of lipids in the regulation of inflammatory processes. The aim of this study was to elucidate the heterogeneity and spatial distribution of lipids in the OA synovial membrane and explore their putative involvement in inflammation. METHOD The abundance and distribution of lipids were examined in human synovial membranes. To this end, histological cuts from this tissue were analysed by matrix-assisted laser desorption ionization - mass spectrometry imaging (MALDI-MSI). The lipidomic profile of OA synovium was characterized and compared with healthy and other forms of inflammatory arthropathies as Rheumatoid Arthritis (RA) and Psoriatic Arthritis (PsA) using principal component analysis and discriminant analysis methods. Lipid identification was undertaken by tandem MS analyses and database queries. RESULTS Our results reveal differential and characteristic lipidomic profiles between OA and control samples. Specifically, we unveiled that OA synovium presents elevated levels of phosphatidylcholines, fatty acids and lysophosphatidic acids and lower levels of lysophosphatidylcholines compared to control tissues. The spatial distribution of particular glycerophospholipids was also correlated with hypertrophic, inflamed or vascularized synovial areas. Compared with other inflammatory arthritis, the OA tissue showed lower amounts of phosphatidylethanolamine-based plasmalogens. CONCLUSIONS This study provides a novel insight into the lipid profiles of synovial membrane and differences in abundance between OA and control tissues. The lipidomic alterations improves understanding of the pathogenic mechanisms of OA and may be important for its diagnosis.
Collapse
Affiliation(s)
- B Rocha
- Grupo de Unidad de Proteómica, Grupo de Investigación de Reumatología (GIR), Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas, Universidade da Coruña (UDC), C/ As Xubias de Arriba 84, 15006, A Coruña, Spain
| | - B Cillero-Pastor
- The Maastricht Multimodal Molecular Imaging Institute (M4I), Division of Imaging Mass Spectrometry, Maastricht University, the Netherlands
| | - C Ruiz-Romero
- Grupo de Unidad de Proteómica, Grupo de Investigación de Reumatología (GIR), Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas, Universidade da Coruña (UDC), C/ As Xubias de Arriba 84, 15006, A Coruña, Spain; Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Av. Monforte de Lemos, 3-5. Pabellón 11, 28029, Madrid, Spain.
| | - M R L Paine
- The Maastricht Multimodal Molecular Imaging Institute (M4I), Division of Imaging Mass Spectrometry, Maastricht University, the Netherlands
| | - J D Cañete
- Unidad de Artritis. Servicio de Reumatología. Hospital Clínico de Barcelona, Barcelona, Spain
| | - R M A Heeren
- The Maastricht Multimodal Molecular Imaging Institute (M4I), Division of Imaging Mass Spectrometry, Maastricht University, the Netherlands
| | - F J Blanco
- Grupo de Unidad de Proteómica, Grupo de Investigación de Reumatología (GIR), Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas, Universidade da Coruña (UDC), C/ As Xubias de Arriba 84, 15006, A Coruña, Spain; Universidade da Coruña (UDC), Grupo de Investigación de Reumatología y Salud (GIR-S), Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Facultad de Fisioterapia, Campus de Oza, 15008, A Coruña, Spain.
| |
Collapse
|
47
|
Lin W, Klein J. Recent Progress in Cartilage Lubrication. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2005513. [PMID: 33759245 DOI: 10.1002/adma.202005513] [Citation(s) in RCA: 154] [Impact Index Per Article: 51.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 11/23/2020] [Indexed: 05/18/2023]
Abstract
Healthy articular cartilage, covering the ends of bones in major joints such as hips and knees, presents the most efficiently-lubricated surface known in nature, with friction coefficients as low as 0.001 up to physiologically high pressures. Such low friction is indeed essential for its well-being. It minimizes wear-and-tear and hence the cartilage degradation associated with osteoarthritis, the most common joint disease, and, by reducing shear stress on the mechanotransductive, cartilage-embedded chondrocytes (the only cell type in the cartilage), it regulates their function to maintain homeostasis. Understanding the origins of such low friction of the articular cartilage, therefore, is of major importance in order to alleviate disease symptoms, and slow or even reverse its breakdown. This progress report considers the relation between frictional behavior and the cellular mechanical environment in the cartilage, then reviews the mechanism of lubrication in the joints, in particular focusing on boundary lubrication. Following recent advances based on hydration lubrication, a proposed synergy between different molecular components of the synovial joints, acting together in enabling the low friction, has been proposed. Additionally, recent development of natural and bio-inspired lubricants is reviewed.
Collapse
Affiliation(s)
- Weifeng Lin
- Department of Materials and Interfaces, Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Jacob Klein
- Department of Materials and Interfaces, Weizmann Institute of Science, Rehovot, 76100, Israel
| |
Collapse
|
48
|
Fatty Acids and Oxylipins in Osteoarthritis and Rheumatoid Arthritis-a Complex Field with Significant Potential for Future Treatments. Curr Rheumatol Rep 2021; 23:41. [PMID: 33913032 PMCID: PMC8081702 DOI: 10.1007/s11926-021-01007-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/23/2021] [Indexed: 02/08/2023]
Abstract
Purpose of Review Osteoarthritis (OA) and rheumatoid arthritis (RA) are characterized by abnormal lipid metabolism manifested as altered fatty acid (FA) profiles of synovial fluid and tissues and in the way dietary FA supplements can influence the symptoms of especially RA. In addition to classic eicosanoids, the potential roles of polyunsaturated FA (PUFA)-derived specialized pro-resolving lipid mediators (SPM) have become the focus of intensive research. Here, we summarize the current state of knowledge of the roles of FA and oxylipins in the degradation or protection of synovial joints. Recent Findings There exists discordance between the large body of literature from cell culture and animal experiments on the adverse and beneficial effects of individual FA and the lack of effective treatments for joint destruction in OA and RA patients. Saturated 16:0 and 18:0 induce mostly deleterious effects, while long-chain n-3 PUFA, especially 20:5n-3, have positive influence on joint health. The situation can be more complex for n-6 PUFA, such as 18:2n-6, 20:4n-6, and its derivative prostaglandin E2, with a combination of potentially adverse and beneficial effects. SPM analogs have future potential as analgesics for arthritic pain. Summary Alterations in FA profiles and their potential implications in SPM production may affect joint lubrication, synovial inflammation, pannus formation, as well as cartilage and bone degradation and contribute to the pathogeneses of inflammatory joint diseases. Further research directions include high-quality randomized controlled trials on dietary FA supplements and investigations on the significance of lipid composition of microvesicle membrane and cargo in joint diseases.
Collapse
|
49
|
Agnihotri P, Monu, Ramani S, Chakraborty D, Saquib M, Biswas S. Differential Metabolome in Rheumatoid Arthritis: a Brief Perspective. Curr Rheumatol Rep 2021; 23:42. [PMID: 33913028 DOI: 10.1007/s11926-021-00989-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/20/2021] [Indexed: 12/17/2022]
Abstract
PURPOSE OF REVIEW Rheumatoid arthritis (RA) is a chronic autoimmune, inflammatory disease of the synovium that affects the movable joints. It develops due to the infiltration and invasion of the synovial joints by immune cells. Metabolism is anabolic or catabolic chemical reactions occurring in a cell. The biochemical pathways in synovial and immune cells are altered affecting the downstream metabolite formation. Changes in the metabolite levels alter signaling cascades which further intensify the disease. Despite current knowledge of metabolomics, there remain certain features that need to be elucidated to correlate the differential metabolite levels with RA. RECENT FINDINGS Metabolite profiling can be used to find altered patterns of metabolites in RA. Glucose, lipid, amino acid, and estrogen metabolism are the key pathways that are altered and contribute to the aggravation of RA. The altered metabolic pathways involved in different cells in RA results in complex interactions between metabolites and biomacromolecules; thus, it generates autoantigens. Moreover, understanding the correlation between differential metabolites and disease severity might help reveal potential new biomarkers and therapeutic targets for RA pathogenesis. So, considering the multi-faceted role of altered metabolites in the pathogenesis of RA, metabolic pathways of different cells are needed to be studied for a better understanding of their functions in the disease and thus, improving the present therapeutic strategies.
Collapse
Affiliation(s)
- Prachi Agnihotri
- Council of Industrial Research (CSIR)-Institute of Genomics and Integrative Biology, Mall Road, Delhi University Campus, Delhi, 110007, India
| | - Monu
- Council of Industrial Research (CSIR)-Institute of Genomics and Integrative Biology, Mall Road, Delhi University Campus, Delhi, 110007, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Sheetal Ramani
- Council of Industrial Research (CSIR)-Institute of Genomics and Integrative Biology, Mall Road, Delhi University Campus, Delhi, 110007, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Debolina Chakraborty
- Council of Industrial Research (CSIR)-Institute of Genomics and Integrative Biology, Mall Road, Delhi University Campus, Delhi, 110007, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Mohd Saquib
- Council of Industrial Research (CSIR)-Institute of Genomics and Integrative Biology, Mall Road, Delhi University Campus, Delhi, 110007, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Sagarika Biswas
- Council of Industrial Research (CSIR)-Institute of Genomics and Integrative Biology, Mall Road, Delhi University Campus, Delhi, 110007, India.
| |
Collapse
|
50
|
Christmann U, Hancock CL, Poole CM, Emery AL, Poovey JR, Hagg C, Mattson EA, Scarborough JJ, Christopher JS, Dixon AT, Craney DJ, Wood PL. Dynamics of DHA and EPA supplementation: incorporation into equine plasma, synovial fluid, and surfactant glycerophosphocholines. Metabolomics 2021; 17:41. [PMID: 33866431 DOI: 10.1007/s11306-021-01792-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 04/05/2021] [Indexed: 01/14/2023]
Abstract
INTRODUCTION Horses with asthma or osteoarthritis frequently receive ω-3 fatty acid supplements. Docosahexaenoic (DHA; 22:6) and eicosapentaenoic (EPA; 20:5) acids are essential ω-3 fatty acid precursors of anti-inflammatory mediators and components of structural glycerophospholipids (GPL) that act as reservoirs of these fatty acids. Analysis of the incorporation of dietary DHA + EPA into GPL pools in different body compartments has not been undertaken in horses. OBJECTIVES We undertook a detailed study of dietary supplementation with DHA + EPA in horses and monitored incorporation into DHA- and EPA-containing glycerophosphocholines (GPC) 38:5, 38:6, 40:5, and 40:6 in plasma, synovial fluid (SF), and surfactant. METHODS Horses (n = 20) were randomly assigned to the supplement or control group and evaluated on days 0, 30, 60, and 90. GPC in plasma, SF, and surfactant were measured by high-resolution mass spectrometry with less than 3 ppm mass error. Validation of DHA and EPA incorporation into these GPC was conducted utilizing MS2 of the [M + Cl]- adducts of GPC. RESULTS Dietary supplementation resulted in augmented levels of GPC 38:5, 38:6, 40:5, and 40:6 in all compartments. Maximum incorporation into GPCs was delayed until 60 days. Significant increases in the levels of GPC 38:5, 40:5, and 40:6, containing docosapentaenoic acid (DPA; 22:5), also was noted. CONCLUSIONS DHA and EPA supplementation results in augmented storage pools of ω-3 essential fatty acids in SF and surfactant GPC. This has the potential to improve the ability of anti-inflammatory mechanisms to resolve inflammatory pathways in these critical compartments involved in arthritis and asthma.
Collapse
Affiliation(s)
- Undine Christmann
- Metabolomics Unit, College of Veterinary Medicine, Lincoln Memorial University, 6965 Cumberland Gap Pkwy, Harrogate, TN, 37752, USA.
| | - Courtney L Hancock
- Metabolomics Unit, College of Veterinary Medicine, Lincoln Memorial University, 6965 Cumberland Gap Pkwy, Harrogate, TN, 37752, USA
| | - Cathleen M Poole
- Metabolomics Unit, College of Veterinary Medicine, Lincoln Memorial University, 6965 Cumberland Gap Pkwy, Harrogate, TN, 37752, USA
| | - Audrey L Emery
- Metabolomics Unit, College of Veterinary Medicine, Lincoln Memorial University, 6965 Cumberland Gap Pkwy, Harrogate, TN, 37752, USA
| | - Jesse R Poovey
- Metabolomics Unit, College of Veterinary Medicine, Lincoln Memorial University, 6965 Cumberland Gap Pkwy, Harrogate, TN, 37752, USA
| | - Casey Hagg
- Metabolomics Unit, College of Veterinary Medicine, Lincoln Memorial University, 6965 Cumberland Gap Pkwy, Harrogate, TN, 37752, USA
| | - Eric A Mattson
- Metabolomics Unit, College of Veterinary Medicine, Lincoln Memorial University, 6965 Cumberland Gap Pkwy, Harrogate, TN, 37752, USA
| | - Jon J Scarborough
- DeBusk College of Osteopathic Medicine, Lincoln Memorial University, 6965 Cumberland Gap Pkwy, Harrogate, TN, 37752, USA
| | - Jordan S Christopher
- DeBusk College of Osteopathic Medicine, Lincoln Memorial University, 6965 Cumberland Gap Pkwy, Harrogate, TN, 37752, USA
| | - Alexander T Dixon
- DeBusk College of Osteopathic Medicine, Lincoln Memorial University, 6965 Cumberland Gap Pkwy, Harrogate, TN, 37752, USA
| | - Dustin J Craney
- DeBusk College of Osteopathic Medicine, Lincoln Memorial University, 6965 Cumberland Gap Pkwy, Harrogate, TN, 37752, USA
| | - Paul L Wood
- Metabolomics Unit, College of Veterinary Medicine, Lincoln Memorial University, 6965 Cumberland Gap Pkwy, Harrogate, TN, 37752, USA
| |
Collapse
|