1
|
Bergström B, Selldén T, Bollmann M, Svensson MND, Ekwall AKH. Methotrexate promotes the release of granulocyte-macrophage colony-stimulating factor from rheumatoid arthritis fibroblast-like synoviocytes via autocrine interleukin-1 signaling. Arthritis Res Ther 2024; 26:178. [PMID: 39394168 PMCID: PMC11468154 DOI: 10.1186/s13075-024-03406-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 09/23/2024] [Indexed: 10/13/2024] Open
Abstract
BACKGROUND Activated fibroblast-like synoviocytes (FLS) are drivers of synovitis and structural joint damage in rheumatoid arthritis (RA). Despite the use of disease-modifying drugs, only about 50% of RA patients reach remission in real-world settings. We used an unbiased approach to investigate the effects of standard-of-care methotrexate (MTX) and a Janus kinase inhibitor, tofacitinib (TOFA), on gene expression in RA-FLS, in order to identify untargeted disease mediators. METHODS Primary RA-FLS were activated by stimulation with interleukin-1β (IL-1β) or platelet-derived growth factor + IL-1β in the presence or absence of MTX or TOFA, with or without additional inhibitors. Co-cultures of synovial cells were performed in direct and indirect systems. Cells were collected for RNA sequencing or qPCR, and supernatants were analyzed for protein concentrations. RESULTS Six thousand three hundred fifty genes were differentially expressed, the majority being upregulated, in MTX-treated activated RA-FLS and 970 genes, the majority being downregulated, in TOFA-treated samples. Pathway analysis showed that MTX had largest effects on 'Molecular mechanisms of cancer' and TOFA on 'Interferon signaling'. Targeted analysis of disease-associated genes revealed that MTX increased the expression of cell cycle-regulating genes but also of pro-inflammatory mediators like IL-1α (IL1A) and granulocyte-macrophage colony-stimulating factor, GM-CSF (CSF2). The MTX-promoted expression of CSF2 in activated RA-FLS peaked at 48 h, could be mediated via either NF-κB or AP-1 transcription factors, and was abrogated by IL-1 inhibitors (IRAK4 inhibitor and anakinra). In a co-culture setting, MTX-treatment of activated RA-FLS induced IL1B expression in macrophages. CONCLUSIONS MTX treatment induces secretion of IL-1 from activated RA-FLS which by autocrine signaling augments their release of GM-CSF. This unexpected effect of MTX might contribute to the persistence of synovitis.
Collapse
Affiliation(s)
- Beatrice Bergström
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Tilia Selldén
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Miriam Bollmann
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- SciLifeLab, University of Gothenburg, Gothenburg, Sweden
| | - Mattias N D Svensson
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- SciLifeLab, University of Gothenburg, Gothenburg, Sweden
| | - Anna-Karin Hultgård Ekwall
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
- Department of Rheumatology, Division 3, Sahlgrenska University Hospital, Gothenburg, Sweden.
| |
Collapse
|
2
|
Mangoni AA, Zinellu A. Diagnostic accuracy of the neutrophil-to-lymphocyte ratio and the platelet-to-lymphocyte ratio in rheumatoid arthritis: a systematic review and meta-analysis. Clin Exp Med 2024; 24:207. [PMID: 39230596 PMCID: PMC11374877 DOI: 10.1007/s10238-024-01478-x] [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: 04/28/2024] [Accepted: 08/23/2024] [Indexed: 09/05/2024]
Abstract
Existing challenges with the early diagnosis of rheumatoid arthritis (RA) and active disease, mainly by non-rheumatologists, have prompted the search for novel biomarkers. Elevations in indices derived from blood cell counts, e.g., the neutrophil-to-lymphocyte ratio (NLR) and the platelet-to-lymphocyte ratio (PLR), have been reported in RA patients. However, their diagnostic accuracy has not been comprehensively assessed. Therefore, we conducted a systematic review and meta-analysis of studies reporting the sensitivity and specificity of the NLR and PLR, obtained by receiver operating characteristic (ROC) curve analysis, for the presence of RA and active disease. We searched electronic databases from inception to 15 March 2024 and assessed the risk of bias using the JBI Critical Appraisal Checklist (PROSPERO registration number: CRD42024533546). In 15 studies, the NLR exhibited acceptable accuracy for the presence of RA (area under the curve, AUC = 0.76, 95% CI 0.72 to 0.80) and active disease (AUC = 0.70, 95% CI 0.66 to 0.74). The PLR exhibited good accuracy for the presence of RA (AUC = 0.80, 95% CI 0.76 to 0.83). There were insufficient studies to assess the accuracy of the PLR for the presence of active disease. Our systematic review and meta-analysis suggests that the NLR and the PLR are promising biomarkers of RA (NLR and PLR) and active disease (NLR). Further research is required to investigate whether the NLR and PLR can significantly enhance the capacity to diagnose RA and active disease in clinical practice.
Collapse
Affiliation(s)
- Arduino A Mangoni
- Discipline of Clinical Pharmacology, College of Medicine and Public Health, Flinders University, Adelaide, Australia.
- Department of Clinical Pharmacology, Flinders Medical Centre, Southern Adelaide Local Health Network, Adelaide, Australia.
- Department of Clinical Pharmacology, College of Medicine and Public Health, Flinders University and Flinders Medical Centre, Bedford Park, SA, 5042, Australia.
| | - Angelo Zinellu
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| |
Collapse
|
3
|
Zheng X, Yang H, Zhang Z, Liang X, Liu Y, Wang C, Yang X, Tang J, Mao J, Nie Y, Zhou X, Li C. pH-responsive size-adjustable liposomes induce apoptosis of fibroblasts and macrophages for rheumatoid arthritis treatment. Acta Biomater 2024; 179:256-271. [PMID: 38484831 DOI: 10.1016/j.actbio.2024.03.006] [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: 12/06/2023] [Revised: 02/19/2024] [Accepted: 03/07/2024] [Indexed: 03/25/2024]
Abstract
In rheumatoid arthritis (RA), macrophages infiltrate joints, while fibroblast-like synovial cells proliferate abnormally, forming a barrier against drug delivery, which hinders effective drug delivery to joint focus. Here we firstly designed a pH-responsive size-adjustable nanoparticle, composed by methotrexate (MTX)-human serum albumin (HSA) complex coating with pH-responsive liposome (Lipo/MTX-HSA) for delivering drugs specifically to inflamed joints in acidic environments. We showed in vitro that the nanoparticles can induce mitochondrial dysfunction, promote apoptosis of fibroblast-like synoviocytes and macrophages, further reduce the secretion of inflammatory factors (TNF-α, IL-1β, MMP-9), and regulate the inflammatory microenvironment. We also demonstrated similar effects in a rat model of arthritis, in which Lipo/MTX-HSA accumulated in arthritic joints, and at low pH, liposome phospholipid bilayer cleavage released small-sized MTX-HSA, which effectively reduced the number of fibroblast-synoviocytes and macrophages in joints, alleviated joint inflammation, and repaired bone erosion. These findings suggest that microenvironment-responsive size-adjustable nanoparticles show promise as a treatment against rheumatoid arthritis. STATEMENT OF SIGNIFICANCE: Abnormal proliferation of fibroblast synoviocytes poses a physical barrier to effective nanoparticle delivery. We designed size-adjustable nano-delivery systems by preparing liposomes with cholesterol hemisuccinate (CHEM), which were subsequently loaded with small-sized albumin nanoparticles encapsulating the cytotoxic drug MTX (MTX-HSA), termed Lipo/MTX-HSA. Upon tail vein injection, Lipo/MTX-HSA could be aggregated at the site of inflammation via the ELVIS effect in the inflamed joint microenvironment. Specifically, intracellular acidic pH-triggered dissociation of liposomes promoted the release of MTX-HSA, which was further targeted to fibroblasts or across fibroblasts to macrophages to exert anti-inflammatory effects. The results showed that liposomes with adjustable particle size achieved efficient drug delivery, penetration and retention in joint sites; the strategy exerted significant anti-inflammatory effects in the treatment of rheumatoid arthritis by inducing mitochondrial dysfunction to promote apoptosis in fibrosynoviocytes and macrophages.
Collapse
Affiliation(s)
- Xiu Zheng
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China; Drug laboratory, Department of pharmacy, Chengdu Xinhua Hospital affiliated to North Sichuan Medical College, Chengdu, Sichuan, 610000, China
| | - Hong Yang
- Department of Pediatrics, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Zongquan Zhang
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Xiaoya Liang
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Yan Liu
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Chenglong Wang
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Xi Yang
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Jun Tang
- Analysis and Testing Center, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Jingying Mao
- Department of Thyroid Surgery, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Yu Nie
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan, 610064, China.
| | - Xiangyu Zhou
- Department of Thyroid Surgery, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, China; Basic Medicine Research Innovation Center for Cardiometabolic Disease, Ministry of Education, Southwest Medical University, Luzhou, Sichuan, 646000, China.
| | - Chunhong Li
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China; Basic Medicine Research Innovation Center for Cardiometabolic Disease, Ministry of Education, Southwest Medical University, Luzhou, Sichuan, 646000, China.
| |
Collapse
|
4
|
Aripova N, Duryee MJ, England BR, Hunter CD, Mordeson JE, Ryan EM, Daubach EC, Romberger DJ, Thiele GM, Mikuls TR. Citrullinated and malondialdehyde-acetaldehyde modified fibrinogen activates macrophages and promotes an aggressive synovial fibroblast phenotype in patients with rheumatoid arthritis. Front Immunol 2023; 14:1203548. [PMID: 37654483 PMCID: PMC10467288 DOI: 10.3389/fimmu.2023.1203548] [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: 04/10/2023] [Accepted: 07/27/2023] [Indexed: 09/02/2023] Open
Abstract
Objective Post-translational protein modifications with malondialdehyde-acetaldehyde (MAA) and citrulline (CIT) are implicated in the pathogenesis of rheumatoid arthritis (RA). Although precise mechanisms have not been elucidated, macrophage-fibroblast interactions have been proposed to play a central role in the development and progression of RA. The purpose of our study was to evaluate the downstream effects of macrophage released soluble mediators, following stimulation with fibrinogen (FIB) modified antigens, on human fibroblast-like synoviocytes (HFLS). Methods PMA-treated U-937 monocytes (Mϕ) and macrophage-differentiated peripheral blood mononuclear cells (MP) were stimulated with FIB, FIB-MAA, FIB-CIT, or FIB-MAA-CIT. HFLS-RA cells were stimulated directly with FIB antigens or with supernatants (SN) from macrophages (Mϕ-SN or MP-SN) stimulated with FIB antigens. Genes associated with an aggressive HFLS phenotype, extracellular matrix proteins, and activated signaling pathways were evaluated. Results HFLS-RA cells treated with Mϕ-SNFIB-CIT and Mϕ-SNFIB-MAA-CIT demonstrated significant increases in mRNA expression of genes associated with an aggressive phenotype at 24-h as compared to direct stimulation with the same antigens. Similar results were obtained using MP-SN. Cellular morphology was altered and protein expression of vimentin (p<0.0001 vs. Mϕ-SNFIB) and type II collagen (p<0.0001) were significantly increased in HFLS-RA cells treated with any of the Mϕ-SN generated following stimulation with modified antigens. Phosphorylation of JNK, Erk1/2, and Akt were increased most substantially in HFLS-RA treated with Mϕ-SNFIB-MAA-CIT (p<0.05 vs Mϕ-SNFIB). These and other data suggested the presence of PDGF-BB in Mϕ-SN. Mϕ-SNFIB-MAA-CIT contained the highest concentration of PDGF-BB (p<0.0001 vs. Mϕ-SNFIB) followed by Mϕ-SNFIB-CIT then Mϕ-SNFIB-MAA. HFLS-RA cells treated with PDGF-BB showed similar cellular morphology to the Mϕ-SN generated following stimulation with modified FIB, as well as the increased expression of vimentin, type II collagen, and the phosphorylation of JNK, Erk1/2 and Akt signaling molecules. Conclusion Together, these findings support the hypothesis that in response to MAA-modified and/or citrullinated fibrinogen, macrophages release soluble factors including PDGF-BB that induce fibroblast activation and promote an aggressive fibroblast phenotype. These cellular responses were most robust following macrophage activation with dually modified fibrinogen, compared to single modification alone, providing novel insights into the combined role of multiple post-translational protein modifications in the development of RA.
Collapse
Affiliation(s)
- Nozima Aripova
- Department of Internal Medicine, Division of Rheumatology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Michael J. Duryee
- Department of Internal Medicine, Division of Rheumatology, University of Nebraska Medical Center, Omaha, NE, United States
- Department of Research Services 151, Veteran Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, United States
| | - Bryant R. England
- Department of Internal Medicine, Division of Rheumatology, University of Nebraska Medical Center, Omaha, NE, United States
- Department of Research Services 151, Veteran Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, United States
| | - Carlos D. Hunter
- Department of Internal Medicine, Division of Rheumatology, University of Nebraska Medical Center, Omaha, NE, United States
- Department of Research Services 151, Veteran Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, United States
| | - Jack E. Mordeson
- Department of Internal Medicine, Division of Rheumatology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Evan M. Ryan
- Department of Internal Medicine, Division of Rheumatology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Eric C. Daubach
- Department of Internal Medicine, Division of Rheumatology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Debra J. Romberger
- Department of Internal Medicine, Division of Rheumatology, University of Nebraska Medical Center, Omaha, NE, United States
- Department of Internal Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, Omaha, NE, United States
| | - Geoffrey M. Thiele
- Department of Internal Medicine, Division of Rheumatology, University of Nebraska Medical Center, Omaha, NE, United States
- Department of Research Services 151, Veteran Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, United States
| | - Ted R. Mikuls
- Department of Internal Medicine, Division of Rheumatology, University of Nebraska Medical Center, Omaha, NE, United States
- Department of Research Services 151, Veteran Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, United States
| |
Collapse
|
5
|
Singh V, Naldi A, Soliman S, Niarakis A. A large-scale Boolean model of the rheumatoid arthritis fibroblast-like synoviocytes predicts drug synergies in the arthritic joint. NPJ Syst Biol Appl 2023; 9:33. [PMID: 37454172 DOI: 10.1038/s41540-023-00294-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 06/29/2023] [Indexed: 07/18/2023] Open
Abstract
Rheumatoid arthritis (RA) is a complex autoimmune disease with an unknown aetiology. However, rheumatoid arthritis fibroblast-like synoviocytes (RA-FLS) play a significant role in initiating and perpetuating destructive joint inflammation by expressing immuno-modulating cytokines, adhesion molecules, and matrix remodelling enzymes. In addition, RA-FLS are primary drivers of inflammation, displaying high proliferative rates and an apoptosis-resistant phenotype. Thus, RA-FLS-directed therapies could become a complementary approach to immune-directed therapies by predicting the optimal conditions that would favour RA-FLS apoptosis, limit inflammation, slow the proliferation rate and minimise bone erosion and cartilage destruction. In this paper, we present a large-scale Boolean model for RA-FLS that consists of five submodels focusing on apoptosis, cell proliferation, matrix degradation, bone erosion and inflammation. The five-phenotype-specific submodels can be simulated independently or as a global model. In silico simulations and perturbations reproduced the expected biological behaviour of the system under defined initial conditions and input values. The model was then used to mimic the effect of mono or combined therapeutic treatments and predict novel targets and drug candidates through drug repurposing analysis.
Collapse
Affiliation(s)
- Vidisha Singh
- Université Paris-Saclay, Laboratoire Européen de Recherche pour la Polyarthrite rhumatoïde-Genhotel, Univ Evry, Evry, France
| | - Aurelien Naldi
- Lifeware Group, Inria, Saclay-île de France, 91120, Palaiseau, France
| | - Sylvain Soliman
- Lifeware Group, Inria, Saclay-île de France, 91120, Palaiseau, France
| | - Anna Niarakis
- Université Paris-Saclay, Laboratoire Européen de Recherche pour la Polyarthrite rhumatoïde-Genhotel, Univ Evry, Evry, France.
- Lifeware Group, Inria, Saclay-île de France, 91120, Palaiseau, France.
| |
Collapse
|
6
|
Liu Y, Jiang H, Kang T, Shi X, Liu X, Li C, Hou X, Li M. Platelets-related signature based diagnostic model in rheumatoid arthritis using WGCNA and machine learning. Front Immunol 2023; 14:1204652. [PMID: 37426641 PMCID: PMC10327425 DOI: 10.3389/fimmu.2023.1204652] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 06/12/2023] [Indexed: 07/11/2023] Open
Abstract
Background and aim Rheumatoid arthritis (RA) is an autoinflammatory disease that may lead to severe disability. The diagnosis of RA is limited due to the need for biomarkers with both reliability and efficiency. Platelets are deeply involved in the pathogenesis of RA. Our study aims to identify the underlying mechanism and screening for related biomarkers. Methods We obtained two microarray datasets (GSE93272 and GSE17755) from the GEO database. We performed Weighted correlation network analysis (WGCNA) to analyze the expression modules in differentially expressed genes identified from GSE93272. We used KEGG, GO and GSEA enrichment analysis to elucidate the platelets-relating signatures (PRS). We then used the LASSO algorithm to develop a diagnostic model. We then used GSE17755 as a validation cohort to assess the diagnostic performance by operating Receiver Operating Curve (ROC). Results The application of WGCNA resulted in the identification of 11 distinct co-expression modules. Notably, Module 2 exhibited a prominent association with platelets among the differentially expressed genes (DEGs) analyzed. Furthermore, a predictive model consisting of six genes (MAPK3, ACTB, ACTG1, VAV2, PTPN6, and ACTN1) was constructed using LASSO coefficients. The resultant PRS model demonstrated excellent diagnostic accuracy in both cohorts, as evidenced by area under the curve (AUC) values of 0.801 and 0.979. Conclusion We elucidated the PRSs occurred in the pathogenesis of RA and developed a diagnostic model with excellent diagnostic potential.
Collapse
Affiliation(s)
- Yuchen Liu
- School of Clinical Medicine, Peking Union Medical College, Beijing, China
- Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Haixu Jiang
- Department of Rheumatology, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Tianlun Kang
- Department of Rheumatology, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaojun Shi
- Department of Rheumatology, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaoping Liu
- Department of Rheumatology, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Chen Li
- Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Rheumatology, Fangshan Hospital Beijing University of Chinese Medicine, Beijing, China
| | - Xiujuan Hou
- Department of Rheumatology, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Meiling Li
- Department of Rheumatology, Fuyang Hospital of Anhui Medical University, Fuyang, Anhui, China
- Department of Rheumatology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| |
Collapse
|
7
|
Comparison of the effects of peficitinib and tofacitinib in the adjuvant-induced arthritis rat model. Eur J Pharmacol 2023; 941:175490. [PMID: 36608862 DOI: 10.1016/j.ejphar.2023.175490] [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: 09/23/2022] [Revised: 12/22/2022] [Accepted: 01/03/2023] [Indexed: 01/05/2023]
Abstract
We investigated and compared the pharmacologic properties of two Janus kinase (JAK) inhibitors, peficitinib and tofacitinib, in an adjuvant-induced arthritis rat model. Repeated administration of peficitinib (3 - 30 mg/kg) or tofacitinib (1 - 10 mg/kg) exhibited a dose-related and significant attenuation of arthritis score, paw swelling, pain threshold, grip strength and histopathologic injuries in the model; peficitinib 10 mg/kg and tofacitinib 3 mg/kg demonstrated comparable efficacy. Equivalent Cmax and AUC0-12h values were observed with peficitinib 10 mg/kg and tofacitinib 3 mg/kg, suggesting that the two drugs may demonstrate comparable efficacy on arthritis-associated symptoms at comparable plasma concentration levels. However, peficitinib 10 mg/kg had greater efficacy than tofacitinib 3 mg/kg on some inflammation- and bone destruction-associated parameters in the paw fluid, including the production of vascular endothelial growth factor (VEGF), platelet-derived growth factor (PDGF), receptor activator of nuclear factor kappa-B ligand, and matrix metalloproteinase-3, which are associated with arthritis exacerbation. Peficitinib 10 mg/kg also showed significantly greater inhibitory effects than tofacitinib 3 mg/kg on loss of bone mineral density and synovial thickening score, which might be a result of the VEGF and PDGF receptor kinase inhibitory effects of peficitinib, in addition to JAK inhibition. In conclusion, both tofacitinib and peficitinib potently improved arthritis and associated symptoms in adjuvant-induced arthritis rats; moreover, owing to possible differences in the mechanism of action of the two drugs, peficitinib may have exerted its effects through JAK inhibition and additional unique off-target properties.
Collapse
|
8
|
Machado CRL, Dias FF, Resende GG, Oliveira PGD, Xavier RM, Andrade MVMD, Kakehasi AM. Morphofunctional analysis of fibroblast-like synoviocytes in human rheumatoid arthritis and mouse collagen-induced arthritis. Adv Rheumatol 2023; 63:1. [PMID: 36597166 DOI: 10.1186/s42358-022-00281-0] [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: 07/15/2022] [Accepted: 11/28/2022] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Fibroblast-like synoviocytes (FLS) play a prominent role in rheumatoid synovitis and degradation of the extracellular matrix through the production of inflammatory cytokines and metalloproteinases (MMPs). Since animal models are frequently used for elucidating the disease mechanism and therapeutic development, it is relevant to study the ultrastructural characteristics and functional responses in human and mouse FLS. The objective of the study was to analyze ultrastructural characteristics, Interleukin-6 (IL-6) and Metalloproteinase-3 (MMP-3) production and the activation of intracellular pathways in Fibroblast like synoviocytes (FLS) cultures obtained from patients with rheumatoid arthritis (RA) and from mice with collagen-induced arthritis (CIA). METHODS FLSs were obtained from RA patients (RA-FLSs) (n = 8) and mice with CIA (CIA-FLSs) (n = 4). Morphology was assessed by transmission and scanning electron microscopy. IL-6 and MMP-3 production was measured by ELISA, and activation of intracellular signaling pathways (NF-κB and MAPK: p-ERK1/2, p-P38 and p-JNK) was measured by Western blotting in cultures of RA-FLSs and CIA-FLSs stimulated with tumor necrosis factor-alpha (TNF-α) and IL-1β. RESULTS RA-FLS and CIA-FLS cultures exhibited rich cytoplasm, rough endoplasmic reticula and prominent and well-developed Golgi complexes. Transmission electron microscopy demonstrated the presence of lamellar bodies, which are cytoplasmic structures related to surfactant production, in FLSs from both sources. Increased levels of pinocytosis and numbers of pinocytotic vesicles were observed in RA-FLSs (p < 0.05). Basal production of MMP-3 and IL-6 was present in RA-FLSs and CIA-FLSs. Regarding the production of MMP-3 and IL-6 and the activation of signaling pathways, the present study demonstrated a lower response to IL-1β by CIA-FLSs than by RA-FLSs. CONCLUSION This study provides a comprehensive understanding of the biology of RA-FLS and CIA-FLS. The differences and similarities in ultrastructural morphology and important inflammatory cytokines shown, contribute to future in vitro studies using RA-FLS and CIA-FLS, in addition, they indicate that the adoption of CIA-FLS for studies should take careful and be well designed, since they do not completely resemble human diseases.
Collapse
Affiliation(s)
- Camilla Ribeiro Lima Machado
- Laboratory of Scientific Research - Professor Lineu Freire-Maia, Faculty of Medicine, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.
| | - Felipe Ferraz Dias
- Laboratory of Scientific Research, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | | | - Ricardo Machado Xavier
- Internal Medicine Department, Faculty of Medicine, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Marcus Vinicius Melo de Andrade
- Laboratory of Scientific Research - Professor Lineu Freire-Maia, Faculty of Medicine, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Adriana Maria Kakehasi
- Post Graduate Program in Sciences Applied to Adult Health Care, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
9
|
Aghakhani S, Soliman S, Niarakis A. Metabolic reprogramming in Rheumatoid Arthritis Synovial Fibroblasts: A hybrid modeling approach. PLoS Comput Biol 2022; 18:e1010408. [PMID: 36508473 PMCID: PMC9779668 DOI: 10.1371/journal.pcbi.1010408] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 12/22/2022] [Accepted: 11/11/2022] [Indexed: 12/15/2022] Open
Abstract
Rheumatoid Arthritis (RA) is an autoimmune disease characterized by a highly invasive pannus formation consisting mainly of Synovial Fibroblasts (RASFs). This pannus leads to cartilage, bone, and soft tissue destruction in the affected joint. RASFs' activation is associated with metabolic alterations resulting from dysregulation of extracellular signals' transduction and gene regulation. Deciphering the intricate mechanisms at the origin of this metabolic reprogramming may provide significant insight into RASFs' involvement in RA's pathogenesis and offer new therapeutic strategies. Qualitative and quantitative dynamic modeling can address some of these features, but hybrid models represent a real asset in their ability to span multiple layers of biological machinery. This work presents the first hybrid RASF model: the combination of a cell-specific qualitative regulatory network with a global metabolic network. The automated framework for hybrid modeling exploits the regulatory network's trap-spaces as additional constraints on the metabolic network. Subsequent flux balance analysis allows assessment of RASFs' regulatory outcomes' impact on their metabolic flux distribution. The hybrid RASF model reproduces the experimentally observed metabolic reprogramming induced by signaling and gene regulation in RASFs. Simulations also enable further hypotheses on the potential reverse Warburg effect in RA. RASFs may undergo metabolic reprogramming to turn into "metabolic factories", producing high levels of energy-rich fuels and nutrients for neighboring demanding cells through the crucial role of HIF1.
Collapse
Affiliation(s)
- Sahar Aghakhani
- GenHotel–Laboratoire Européen de Recherche pour la Polyarthrite Rhumatoïde, Univ. Evry, Univ. Paris-Saclay, Evry, France
- Lifeware Group, Inria Saclay Île-de-France, Palaiseau, France
| | - Sylvain Soliman
- Lifeware Group, Inria Saclay Île-de-France, Palaiseau, France
| | - Anna Niarakis
- GenHotel–Laboratoire Européen de Recherche pour la Polyarthrite Rhumatoïde, Univ. Evry, Univ. Paris-Saclay, Evry, France
- Lifeware Group, Inria Saclay Île-de-France, Palaiseau, France
- * E-mail:
| |
Collapse
|
10
|
Jiang SZ, To JL, Hughes MR, McNagny KM, Kim H. Platelet signaling at the nexus of innate immunity and rheumatoid arthritis. Front Immunol 2022; 13:977828. [PMID: 36505402 PMCID: PMC9732516 DOI: 10.3389/fimmu.2022.977828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 10/03/2022] [Indexed: 11/26/2022] Open
Abstract
Rheumatoid arthritis (RA) is a debilitating autoimmune disorder characterized by chronic inflammation of the synovial tissues and progressive destruction of bone and cartilage. The inflammatory response and subsequent tissue degradation are orchestrated by complex signaling networks between immune cells and their products in the blood, vascular endothelia and the connective tissue cells residing in the joints. Platelets are recognized as immune-competent cells with an important role in chronic inflammatory diseases such as RA. Here we review the specific aspects of platelet function relevant to arthritic disease, including current knowledge of the molecular crosstalk between platelets and other innate immune cells that modulate RA pathogenesis.
Collapse
Affiliation(s)
- Steven Z. Jiang
- Centre for Blood Research, University of British Columbia, Vancouver, BC, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| | - Jeffrey L. To
- Centre for Blood Research, University of British Columbia, Vancouver, BC, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| | - Michael R. Hughes
- Centre for Blood Research, University of British Columbia, Vancouver, BC, Canada
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
| | - Kelly M. McNagny
- Centre for Blood Research, University of British Columbia, Vancouver, BC, Canada
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Hugh Kim
- Centre for Blood Research, University of British Columbia, Vancouver, BC, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
- Department of Oral Biological and Medical Sciences, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
11
|
Sahu N, Grandi FC, Bhutani N. A single-cell mass cytometry platform to map the effects of preclinical drugs on cartilage homeostasis. JCI Insight 2022; 7:160702. [PMID: 36194485 PMCID: PMC9744259 DOI: 10.1172/jci.insight.160702] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 09/12/2022] [Indexed: 02/04/2023] Open
Abstract
No disease-modifying drug exists for osteoarthritis (OA). Despite success in animal models, candidate drugs continue to fail in clinical trials owing to the unmapped interpatient heterogeneity and disease complexity. We used a single-cell platform based on cytometry by time-of-flight (cyTOF) to precisely outline the effects of candidate drugs on human OA chondrocytes. OA chondrocytes harvested from patients undergoing total knee arthroplasty were treated with 2 drugs, an NF-κB pathway inhibitor, BMS-345541, and a chondroinductive small molecule, kartogenin, that showed preclinical success in animal models for OA. cyTOF conducted with 30 metal isotope-labeled antibodies parsed the effects of the drugs on inflammatory, senescent, and chondroprogenitor cell populations. The NF-κB pathway inhibition decreased the expression of p-NF-κB, HIF2A, and inducible NOS in multiple chondrocyte clusters and significantly depleted 4 p16ink4a-expressing senescent populations, including NOTCH1+STRO1+ chondroprogenitor cells. While kartogenin also affected select p16ink4a-expressing senescent clusters, there was a less discernible effect on chondroprogenitor cell populations. Overall, BMS-345541 elicited a uniform drug response in all patients, while only a few responded to kartogenin. These studies demonstrate that a single-cell cyTOF-based drug screening platform can provide insights into patient response assessment and patient stratification.
Collapse
|
12
|
Dąbkowska K, Wojdas M, Kuźnik-Trocha K, Wisowski G, Gruenpeter A, Komosińska-Vassev K, Olczyk K, Winsz-Szczotka K. GAAGs, COMP, and YKL-40 as Potential Markers of Cartilage Turnover in Blood of Children with Juvenile Idiopathic Arthritis Treated with Etanercept—Relationship with ADAMTS4, ADAMTS5, and PDGF-BB. J Clin Med 2022; 11:jcm11175069. [PMID: 36079004 PMCID: PMC9457057 DOI: 10.3390/jcm11175069] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 08/21/2022] [Accepted: 08/26/2022] [Indexed: 11/28/2022] Open
Abstract
We quantified galactosaminoglycans (GAAGs), oligomeric cartilage matrix protein (COMP), and human cartilage glycoprotein 39 (YKL-40) in blood obtained from juvenile idiopathic arthritis (JIA) before and during 2-year treatment with etanercept (ETA), as potential biomarkers of cartilage extracellular matrix (ECM) dysfunction and indicators of efficacy of biologic therapy. We also evaluated the relationship of the mentioned markers with the factors that regulate their metabolism, disintegrin and thrombospondin motif metalloproteinases 4 (ADAMTS4), ADAMTS5, and platelet-derived growth factor BB (PDGF-BB). Methods: We studied 38 children diagnosed with JIA and 45 healthy children. We quantified GAAGs by assessing the concentration of unsaturated disaccharide units formed by digestion of isolated glycosaminoglycans with chondroitinase ABC, while COMP, YKL-40, and PDGF-BB were quantified using immunoenzymatic methods. Results: Compared to the control group, GAAGs and COMP levels were significantly lower, while YKL-40 levels were higher in the blood of patients with aggressive JIA, qualified for ETA treatment. ETA therapy leading to clinical improvement simultaneously promoted normalization of COMP and YKL-40 levels, but not GAAGs. After 24 months of taking ETA, glycan levels were still significantly lower, relative to controls. GAAGs, COMP, and YKL-40 levels were significantly influenced by ADAMTS4, ADAMTS5, and PDGF-BB levels both before and during ETA treatment. Conclusions: The dynamics of changes in marker concentrations during treatment seem to indicate that measurement of COMP and YKL-40 levels can be used to assess the chondroprotective biological efficacy of therapy. In contrast, changes in GAAGs concentrations reflect systemic extracellular matrix transformations in the course of JIA.
Collapse
Affiliation(s)
- Klaudia Dąbkowska
- Department of Clinical Chemistry and Laboratory Diagnostics, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, ul. Jedności 8, 41-200 Sosnowiec, Poland
- Correspondence:
| | - Magdalena Wojdas
- Department of Clinical Chemistry and Laboratory Diagnostics, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, ul. Jedności 8, 41-200 Sosnowiec, Poland
| | - Kornelia Kuźnik-Trocha
- Department of Clinical Chemistry and Laboratory Diagnostics, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, ul. Jedności 8, 41-200 Sosnowiec, Poland
| | - Grzegorz Wisowski
- Department of Clinical Chemistry and Laboratory Diagnostics, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, ul. Jedności 8, 41-200 Sosnowiec, Poland
| | - Anna Gruenpeter
- Department of Rheumatology, The John Paul II Pediatric Center in Sosnowiec, ul. G. Zapolskiej 3, 41-218 Sosnowiec, Poland
| | - Katarzyna Komosińska-Vassev
- Department of Clinical Chemistry and Laboratory Diagnostics, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, ul. Jedności 8, 41-200 Sosnowiec, Poland
| | - Krystyna Olczyk
- Department of Clinical Chemistry and Laboratory Diagnostics, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, ul. Jedności 8, 41-200 Sosnowiec, Poland
| | - Katarzyna Winsz-Szczotka
- Department of Clinical Chemistry and Laboratory Diagnostics, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, ul. Jedności 8, 41-200 Sosnowiec, Poland
| |
Collapse
|
13
|
Nathan J, Shameera R, Palanivel G. Studying molecular signaling in major angiogenic diseases. Mol Cell Biochem 2022; 477:2433-2450. [PMID: 35581517 DOI: 10.1007/s11010-022-04452-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 04/24/2022] [Indexed: 10/18/2022]
Abstract
The growth of blood vessels from already existing vasculature is angiogenesis and it is one of the fundamental processes in fetal development, tissue damage or repair, and the reproductive cycle. In a healthy person, angiogenesis is regulated by the balance between pro- and anti-angiogenic factors. However, when the balance is disturbed, it results in various diseases or disorders. The angiogenesis pathway is a sequential cascade and differs based on the stimuli. Therefore, targeting one of the factors involved in the process can help us find a therapeutic strategy to treat irregular angiogenesis. In the past three decades of cancer research, angiogenesis has been at its peak, where an anti-angiogenic agent inhibiting vascular endothelial growth factor acts as a promising substance to treat cancer. In addition, cancer can be assessed based on the expression of angiogenic factors and its response to therapies. Angiogenesis is important for all tissues, which might be normal or pathologically changed and occur through ages. In clinical therapeutics, target therapy focusing on discovery of novel anti-angiogenic agents like bevacizumab, cetuximab, sunitinib, imatinib, lenvatinib, thalidomide, everolimus etc., to block or inhibit the angiogenesis pathway is well explored in recent times. In this review, we will discuss about the molecular signaling pathways involved in major angiogenic diseases in detail.
Collapse
Affiliation(s)
- Jhansi Nathan
- Zebrafish Developmental Biology Laboratory, AUKBC Research Centre, Anna University, Chennai, Tamil Nadu, 600044, India.
| | - Rabiathul Shameera
- Zebrafish Developmental Biology Laboratory, AUKBC Research Centre, Anna University, Chennai, Tamil Nadu, 600044, India
| | - Gajalakshmi Palanivel
- Zebrafish Developmental Biology Laboratory, AUKBC Research Centre, Anna University, Chennai, Tamil Nadu, 600044, India
| |
Collapse
|
14
|
Shin MJ, Park JY, Lee DH, Khang D. Stem Cell Mimicking Nanoencapsulation for Targeting Arthritis. Int J Nanomedicine 2022; 16:8485-8507. [PMID: 35002240 PMCID: PMC8725870 DOI: 10.2147/ijn.s334298] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Accepted: 12/05/2021] [Indexed: 12/12/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are considered a promising regenerative therapy due to their ability to migrate toward damaged tissues. The homing ability of MSCs is unique compared with that of non-migrating cells and MSCs are considered promising therapeutic vectors for targeting major cells in many pathophysiological sites. MSCs have many advantages in the treatment of malignant diseases, particularly rheumatoid arthritis (RA). RA is a representative autoimmune disease that primarily affects joints, and secreted chemokines in the joints are well recognized by MSCs following their migration to the joints. Furthermore, MSCs can regulate the inflammatory process and repair damaged cells in the joints. However, the functionality and migration ability of MSCs injected in vivo still show insufficient. The targeting ability and migration efficiency of MSCs can be enhanced by genetic engineering or modification, eg, overexpressing chemokine receptors or migration-related genes, thus maximizing their therapeutic effect. However, there are concerns about genetic changes due to the increased probability of oncogenesis resulting from genome integration of the viral vector, and thus, clinical application is limited. Furthermore, it is suspected that administering MSCs can promote tumor growth and metastasis in xenograft and orthotopic models. For this reason, MSC mimicking nanoencapsulations are an alternative strategy that does not involve using MSCs or bioengineered MSCs. MSC mimicking nanoencapsulations consist of MSC membrane-coated nanoparticles, MSC-derived exosomes and artificial ectosomes, and MSC membrane-fused liposomes with natural or genetically engineered MSC membranes. MSC mimicking nanoencapsulations not only retain the targeting ability of MSCs but also have many advantages in terms of targeted drug delivery. Specifically, MSC mimicking nanoencapsulations are capable of encapsulating drugs with various components, including chemotherapeutic agents, nucleic acids, and proteins. Furthermore, there are fewer concerns over safety issues on MSC mimicking nanoencapsulations associated with mutagenesis even when using genetically engineered MSCs, because MSC mimicking nanoencapsulations use only the membrane fraction of MSCs. Genetic engineering is a promising route in clinical settings, where nano-encapsulated technology strategies are combined. In this review, the mechanism underlying MSC homing and the advantages of MSC mimicking nanoencapsulations are discussed. In addition, genetic engineering of MSCs and MSC mimicking nanoencapsulation is described as a promising strategy for the treatment of immune-related diseases.
Collapse
Affiliation(s)
- Min Jun Shin
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, 21999, South Korea.,Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, 21999, South Korea
| | - Jun Young Park
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, 21999, South Korea.,Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, 21999, South Korea
| | - Dae Ho Lee
- Department of Internal Medicine, Gachon University Gil Medical Center, Incheon, 21999, South Korea.,Department of Internal Medicine, Gachon University College of Medicine, Incheon, 21999, South Korea
| | - Dongwoo Khang
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, 21999, South Korea.,Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, 21999, South Korea.,Department of Physiology, School of Medicine, Gachon University, Incheon, 21999, South Korea
| |
Collapse
|
15
|
Chhabra S, Dogra S, Sharma K, Raychaudhuri SK, Raychaudhuri SP. Recent Update on Immunopathogenesis of Psoriasis. Indian J Dermatol 2022; 67:360-373. [PMID: 36578729 PMCID: PMC9792009 DOI: 10.4103/ijd.ijd_569_22] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Psoriasis is a chronic disabling complex inflammatory disorder prevalent worldwide with environmental and genetic components that involve predominantly skin in addition to nails and joints associated with various systemic comorbidities having periods of exacerbations and remissions. Psoriasis is characterized by hyper-proliferation as well as abnormal differentiation of epidermal keratinocytes and lymphocyte infiltration (mainly T cells) with resultant inflammatory cytokines and chemokines. Immunological and genetic studies over the last decade have identified genetic susceptibility risk alleles, molecular, cellular and immunological mechanisms involved in immunopathogenesis of psoriasis. The current disease model emphasizes the role of aberrant Th1 and Th17 responses regulated by a complex network of different cytokines, including TNF-α, IL-17 and IL-23; signal transduction pathways downstream to the cytokine receptors; and various activated transcription factors, including NF-κB, interferon regulatory factors and signal transducer and activator of transcriptions. Cytokines targeting biologics (IL-17, IL-23 and TNFα) therapies have revolutionized the management of severe skin disease having beneficial effects on joints and systemic inflammation of psoriasis as well. Further better understanding of immunopathogenesis of psoriasis will pave way for precision medicine based on specific immunopathogenic targets in a given phenotype of disease. Complex interplay of psoriasis with associated comorbidities is also a future area of research for overall better patient management and to improve their quality of life.
Collapse
Affiliation(s)
- Seema Chhabra
- From the Department of Immunopathology, Venereology and Leprology, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Sunil Dogra
- Department of Dermatology, Venereology and Leprology, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Keshav Sharma
- From the Department of Immunopathology, Venereology and Leprology, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Smriti K. Raychaudhuri
- Department of Medicine and Infectious Diseases, VA Northern California Health Care System, 10535 Hospital Way, Mather, CA, United States
| | - Siba P. Raychaudhuri
- Department of Medicine and Infectious Diseases, VA Northern California Health Care System, 10535 Hospital Way, Mather, CA, United States,Department of Dermatology, Division of Rheumatology, Allergy and Clinical Immunology, University of California Davis, School of Medicine, Davis, CA, United States,Address for correspondence: Dr. Siba P. Raychaudhuri, Professor, Division of Rheumatology, Allergy and Clinical Immunology, University of California Davis, School of Medicine, Program Director Rheumatology, Chief of Rheumatology, VA Northern California Health Care System, 10535 Hospital Way, Mather, CA - 95655, United States. E-mail:
| |
Collapse
|
16
|
Wenzel D, Haddadi N, Afshari K, Richmond JM, Rashighi M. Upcoming treatments for morphea. Immun Inflamm Dis 2021; 9:1101-1145. [PMID: 34272836 PMCID: PMC8589364 DOI: 10.1002/iid3.475] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 05/29/2021] [Indexed: 11/25/2022] Open
Abstract
Morphea (localized scleroderma) is a rare autoimmune connective tissue disease with variable clinical presentations, with an annual incidence of 0.4-2.7 cases per 100,000. Morphea occurs most frequently in children aged 2-14 years, and the disease exhibits a female predominance. Insights into morphea pathogenesis are often extrapolated from studies of systemic sclerosis due to their similar skin histopathologic features; however, clinically they are two distinct diseases as evidenced by different demographics, clinical features, disease course and prognosis. An interplay between genetic factors, epigenetic modifications, immune and vascular dysfunction, along with environmental hits are considered as the main contributors to morphea pathogenesis. In this review, we describe potential new therapies for morphea based on both preclinical evidence and ongoing clinical trials. We focus on different classes of therapeutics, including antifibrotic, anti-inflammatory, cellular and gene therapy, and antisenolytic approaches, and how these target different aspects of disease pathogenesis.
Collapse
Affiliation(s)
- Dan Wenzel
- Department of DermatologyUniversity of Massachusetts Medical SchoolWorcesterMassachusettsUSA
| | - Nazgol‐Sadat Haddadi
- Department of DermatologyUniversity of Massachusetts Medical SchoolWorcesterMassachusettsUSA
| | - Khashayar Afshari
- Department of DermatologyUniversity of Massachusetts Medical SchoolWorcesterMassachusettsUSA
| | - Jillian M. Richmond
- Department of DermatologyUniversity of Massachusetts Medical SchoolWorcesterMassachusettsUSA
| | - Mehdi Rashighi
- Department of DermatologyUniversity of Massachusetts Medical SchoolWorcesterMassachusettsUSA
| |
Collapse
|
17
|
Chellamuthu G, Muthu S, Khanna M, Khanna V. "Platelet-rich plasma holds promise in management of rheumatoid arthritis"-systematic review. Rheumatol Int 2021; 41:1895-1903. [PMID: 33834280 DOI: 10.1007/s00296-021-04849-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 03/24/2021] [Indexed: 02/05/2023]
Abstract
The treatment of rheumatoid arthritis (RA) has been closely evolving with an understanding of disease pathogenesis with disease modifying anti-rheumatoid drugs (DMARDS) and Biologic DMARDS being the main stay. platelet rich plasma (PRP) has been the center of research in many specialties in the past decade. Its ability to stop and reverse inflammation have attracted researchers to try PRP in RA. A systematic review of studies on PRP in RA is lacking. The study protocol was prospectively registered in PROSPERO. Detailed search of Cochrane, Scopus, Medline, Embase, and Web of science databases were made to identify the relevant articles till Sep 2020 following Cochrane and PRISMA guidelines. Number of subjects, Animal model used, cell lines used for the study, method of induction of arthritis, PRP dose, concentration used, frequency of administration and clinical, histologic, and molecular changes from baseline following PRP use were extracted and analysed. Eight studies were included for the review. Four of these were in-vitro studies. Two were exclusive animal studies. One study analysed the effects of PRP in RA in both animal models (mice) and Hela cell lines. One study was a report of a series of patients of resistant RA treated with PRP. In the in vitro studies while platelets increase the migration and invasion of RA-FLS, they suppressed the inflammation on the whole. Available animal studies and the Human study have shown encouraging results. There has been no evidence of exacerbation of inflammation in these studies. The quantity and quality of literature on the effects of PRP in treating joint pathologies in RA is limited. Preclinical studies show decrease in disease activity with good safety profile. Invitro studies show suppression of inflammation. Thus, the available literature is encouraging towards the use of PRP in RA. Larger trials and molecular studies to understand the exact role of platelets in disease pathogenesis and treatment mechanisms are needed to decide the future course of PRP in RA.
Collapse
Affiliation(s)
| | - Sathish Muthu
- Orthopaedic Research Group, Coimbatore, India
- Indian Stem Cell Study Group, Lucknow, India
| | - Manish Khanna
- Indian Stem Cell Study Group, Lucknow, India
- Prasad Institute of Medical Sciences, Lucknow, India
| | - Venus Khanna
- Prasad Institute of Medical Sciences, Lucknow, India
| |
Collapse
|
18
|
Huang R, Zhang C, Bu Y, Li Z, Zheng X, Qiu S, Machuki JO, Zhang L, Yang Y, Guo K, Gao F. A multifunctional nano-therapeutic platform based on octahedral yolk-shell Au NR@CuS: Photothermal/photodynamic and targeted drug delivery tri-combined therapy for rheumatoid arthritis. Biomaterials 2021; 277:121088. [PMID: 34464824 DOI: 10.1016/j.biomaterials.2021.121088] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 08/23/2021] [Accepted: 08/24/2021] [Indexed: 12/16/2022]
Abstract
Rheumatoid arthritis (RA) is a common chronic autoimmune disease that results from synovial hyperplasia. The hyperplasia of synovium directly degrades cartilage by secreting matrix-degrading enzymes and inducing cartilage degradation and even loss of joint function. In this study, a metal/semiconductor composite, octahedral copper sulfide shell, and gold nanorod core (Au NR@CuS) is designed for, photothermal therapy (PTT), photodynamic therapy (PDT), and chemotherapy (CT) combination therapy for RA to remove hyperplasia of the synovium. Upon laser irradiation, the coupling of the local surface electromagnetic field improves the electromagnetic field of the Au NR core and the absorption of light of the CuS shell, whereby the photothermal effect is enhanced. Due to the Fenton-like reactions and the integration of Au NR and CuS semiconductor photocatalyst inhibits hole recombination and provides a reaction site for photocatalysis, which introduces additional •OH to photodynamics therapy. In addition, the large octahedral void space in Au NR@CuS NPs can be used for loading a high quantity of drugs for chemotherapy, and modified with vasoactive intestinal peptide and hyaluronic acid (HA) formation VIP-HA-Au NR@CuS NPs to target synovial cells in RA. Under combination therapy, VIP-HA-Au NR@CuS NPs were shown to effectively inhibit the synovial cells and the edema degree of the CIA mouse was alleviated apparently. Both in vitro and in vivo studies indicate that the VIP-HA-Au NR@CuS NPs can provide a potential possibility for the treatment of RA.
Collapse
Affiliation(s)
- Ruqi Huang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 221004, Xuzhou, China; Department of Orthopedics, Affiliated Hospital of Xuzhou Medical University, 221002, Xuzhou, China
| | - Caiyi Zhang
- The Affiliated Xuzhou Oriental Hospital of Xuzhou Medical University, 221004, Xuzhou, China
| | - Yeyang Bu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 221004, Xuzhou, China; Department of Orthopedics, Affiliated Hospital of Xuzhou Medical University, 221002, Xuzhou, China
| | - Zheng Li
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 221004, Xuzhou, China; Department of Orthopedics, Affiliated Hospital of Xuzhou Medical University, 221002, Xuzhou, China
| | - Xin Zheng
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 221004, Xuzhou, China; Department of Orthopedics, Affiliated Hospital of Xuzhou Medical University, 221002, Xuzhou, China
| | - Shang Qiu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 221004, Xuzhou, China; Department of Orthopedics, Affiliated Hospital of Xuzhou Medical University, 221002, Xuzhou, China
| | - Jeremiah Ong'achwa Machuki
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 221004, Xuzhou, China
| | - Lijie Zhang
- Nanomaterials and Chemistry Key Laboratory, Wenzhou University, 325027, Wenzhou, China
| | - Yun Yang
- Nanomaterials and Chemistry Key Laboratory, Wenzhou University, 325027, Wenzhou, China
| | - Kaijin Guo
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 221004, Xuzhou, China; Department of Orthopedics, Affiliated Hospital of Xuzhou Medical University, 221002, Xuzhou, China.
| | - Fenglei Gao
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 221004, Xuzhou, China.
| |
Collapse
|
19
|
Malhotra H, Garg V, Singh G. Biomarker Approach Towards Rheumatoid Arthritis Treatment. Curr Rheumatol Rev 2021; 17:162-175. [PMID: 33327920 DOI: 10.2174/1573397116666201216164013] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 10/02/2020] [Accepted: 10/19/2020] [Indexed: 01/08/2023]
Abstract
Rheumatoid arthritis is an auto-immune disorder, recognized by cartilage as well as bone destruction, which causes irreversible joint deformities, which further results in functional limitations in the patient. Genes like HLA-DRB1 and PTPN22 are likely implicated in the genetic predisposition of rheumatoid arthritis pathology. The first and foremost clinical manifestation in a person with rheumatoid arthritis is joint destruction followed by cartilage and bone destruction caused by cell-cell interactions. The cell-cell interactions are thought to be initialized through the contact of antigen-presenting cells (APC) with CD4+ cells, leading to the progression of the disease. APC includes a complex of class ІІ major histocompatibility complex molecules along with peptide antigens and binds to the receptors present on the surface of T-cells. Further, the activation of macrophages is followed by the release of various pro-inflammatory cytokines such as IL-1 and TNF-α, which lead to the secretion of enzymes that degrade proteoglycan and collagen, which in turn, increase tissue degradation. Biomarkers like IL-6, IL-12, IL-8 and IL-18, 14-3-3η, RANKL, IFN-γ, IFN-β and TGF-β have been designated as key biomarkers in disease development and progression. The study of these biomarkers is very important as they act as a molecular indicator of pathological processes that aggravate the disease.
Collapse
Affiliation(s)
- Hitesh Malhotra
- Chandigarh College of Pharmacy Landran, Mohali, Punjab, India
| | - Vandana Garg
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, Haryana, India
| | - Govind Singh
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, Haryana, India
| |
Collapse
|
20
|
Wojdas M, Dąbkowska K, Winsz-Szczotka K. Alterations of Extracellular Matrix Components in the Course of Juvenile Idiopathic Arthritis. Metabolites 2021; 11:132. [PMID: 33668781 PMCID: PMC7996267 DOI: 10.3390/metabo11030132] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 02/19/2021] [Accepted: 02/22/2021] [Indexed: 12/12/2022] Open
Abstract
Juvenile idiopathic arthritis (JIA) is the most common group of chronic connective tissue diseases in children that is accompanied by joint structure and function disorders. Inflammation underlying the pathogenic changes in JIA, caused by hypersecretion of proinflammatory cytokines, leads to the destruction of articular cartilage. The degradation which progresses with the duration of JIA is not compensated by the extent of repair processes. These disorders are attributed in particular to changes in homeostasis of extracellular matrix (ECM) components, including proteoglycans, that forms articular cartilage. Changes in metabolism of matrix components, associated with the disturbance of their degradation and biosynthesis processes, are the basis of the progressive wear of joint structures observed in the course of JIA. Clinical evaluation and radiographic imaging are current methods to identify the destruction. The aim of this paper is to review enzymatic and non-enzymatic factors involved in catabolism of matrix components and molecules stimulating their biosynthesis. Therefore, we discuss the changes in these factors in body fluids of children with JIA and their potential diagnostic use in the assessment of disease activity. Understanding the changes in ECM components in the course of the child-hood arthritis may provide the introduction of both new diagnostic tools and new therapeutic strategies in children with JIA.
Collapse
Affiliation(s)
- Magdalena Wojdas
- Department of Clinical Chemistry and Laboratory Diagnostics, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, ul. Jedności 8, 41-200 Sosnowiec, Poland; (K.D.); (K.W.-S.)
| | | | | |
Collapse
|
21
|
Kvacskay P, Yao N, Schnotz JH, Scarpone R, Carvalho RDA, Klika KD, Merkt W, Tretter T, Lorenz HM, Tykocinski LO. Increase of aerobic glycolysis mediated by activated T helper cells drives synovial fibroblasts towards an inflammatory phenotype: new targets for therapy? Arthritis Res Ther 2021; 23:56. [PMID: 33588937 PMCID: PMC7883459 DOI: 10.1186/s13075-021-02437-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 02/02/2021] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND A dysregulated glucose metabolism in synovial fibroblasts (SF) has been associated with their aggressive phenotype in rheumatoid arthritis (RA). Even though T helper (Th) cells are key effector cells in the propagation and exacerbation of synovitis in RA, little is known about their influence on the metabolism of SF. Thus, this study investigates the effect of Th cells on the glucose metabolism and phenotype of SF and how this is influenced by the blockade of cytokines, janus kinases (JAKs) and glycolysis. METHODS SF from patients with RA or osteoarthritis (OA) were cultured in the presence of a stable glucose isotopomer ([U-13C]-glucose) and stimulated with the conditioned media of activated Th cells (ThCM). Glucose consumption and lactate production were measured by proton nuclear magnetic resonance (1H NMR) spectroscopy. Cytokine secretion was quantified by ELISA. The expression of glycolytic enzymes was analysed by PCR, western blot and immunofluorescence. JAKs were blocked using either baricitinib or tofacitinib and glycolysis by using either 3-bromopyruvate or FX11. RESULTS Quiescent RASF produced significantly higher levels of lactate, interleukin (IL)-6 and matrix metalloproteinase (MMP) 3 than OASF. Stimulation by ThCM clearly changed the metabolic profile of both RASF and OASF by inducing a shift towards aerobic glycolysis with strongly increased lactate production together with a rise in IL-6 and MMP3 secretion. Interestingly, chronic stimulation of OASF by ThCM triggered an inflammatory phenotype with significantly increased glycolytic activity compared to unstimulated, singly stimulated or re-stimulated OASF. Finally, in contrast to cytokine-neutralizing biologics, inhibition of JAKs or glycolytic enzymes both significantly reduced lactate production and cytokine secretion by Th cell-stimulated SF. CONCLUSIONS Soluble mediators released by Th cells drive SF towards a glycolytic and pro-inflammatory phenotype. Targeting of JAKs or glycolytic enzymes both potently modulate SF's glucose metabolism and decrease the release of IL-6 and MMP3. Thus, manipulation of glycolytic pathways could represent a new therapeutic strategy to decrease the pro-inflammatory phenotype of SF.
Collapse
Affiliation(s)
- Peter Kvacskay
- Department of Medicine V, Division of Rheumatology, University of Heidelberg, INF 410, 69120, Heidelberg, Germany
| | - Nina Yao
- Department of Medicine V, Division of Rheumatology, University of Heidelberg, INF 410, 69120, Heidelberg, Germany
| | - Jürgen-Heinz Schnotz
- Department of Medicine V, Division of Rheumatology, University of Heidelberg, INF 410, 69120, Heidelberg, Germany
| | - Roberta Scarpone
- Department of Medicine V, Division of Rheumatology, University of Heidelberg, INF 410, 69120, Heidelberg, Germany
| | - Rui de Albuquerque Carvalho
- Department of Life Sciences, Faculty of Sciences and Technology, University of Coimbra, Coimbra, Portugal
- REQUIMTE/LAQV, Group of Pharmaceutical Technology, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
| | - Karel D Klika
- Molecular Structure Analysis, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Wolfgang Merkt
- Department of Medicine V, Division of Rheumatology, University of Heidelberg, INF 410, 69120, Heidelberg, Germany
| | - Theresa Tretter
- Department of Medicine V, Division of Rheumatology, University of Heidelberg, INF 410, 69120, Heidelberg, Germany
| | - Hanns-Martin Lorenz
- Department of Medicine V, Division of Rheumatology, University of Heidelberg, INF 410, 69120, Heidelberg, Germany
| | - Lars-Oliver Tykocinski
- Department of Medicine V, Division of Rheumatology, University of Heidelberg, INF 410, 69120, Heidelberg, Germany.
| |
Collapse
|
22
|
Mousavi MJ, Karami J, Aslani S, Tahmasebi MN, Vaziri AS, Jamshidi A, Farhadi E, Mahmoudi M. Transformation of fibroblast-like synoviocytes in rheumatoid arthritis; from a friend to foe. AUTO- IMMUNITY HIGHLIGHTS 2021; 12:3. [PMID: 33546769 PMCID: PMC7863458 DOI: 10.1186/s13317-020-00145-x] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 10/20/2020] [Indexed: 02/07/2023]
Abstract
Swelling and the progressive destruction of articular cartilage are major characteristics of rheumatoid arthritis (RA), a systemic autoimmune disease that directly affects the synovial joints and often causes severe disability in the affected positions. Recent studies have shown that type B synoviocytes, which are also called fibroblast-like synoviocytes (FLSs), as the most commonly and chiefly resident cells, play a crucial role in early-onset and disease progression by producing various mediators. During the pathogenesis of RA, the FLSs' phenotype is altered, and represent invasive behavior similar to that observed in tumor conditions. Modified and stressful microenvironment by FLSs leads to the recruitment of other immune cells and, eventually, pannus formation. The origins of this cancerous phenotype stem fundamentally from the significant metabolic changes in glucose, lipids, and oxygen metabolism pathways. Moreover, the genetic abnormalities and epigenetic alterations have recently been implicated in cancer-like behaviors of RA FLSs. In this review, we will focus on the mechanisms underlying the transformation of FLSs to a cancer-like phenotype during RA. A comprehensive understanding of these mechanisms may lead to devising more effective and targeted treatment strategies.
Collapse
Affiliation(s)
- Mohammad Javad Mousavi
- Rheumatology Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Department of Hematology, Faculty of Allied Medicine, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Jafar Karami
- Rheumatology Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Department of Laboratory Sciences, Khomein University of Medical Sciences, Khomein, Iran
| | - Saeed Aslani
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Arash Sharafat Vaziri
- Joint Reconstruction Reseach Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Ahmadreza Jamshidi
- Rheumatology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Elham Farhadi
- Rheumatology Research Center, Tehran University of Medical Sciences, Tehran, Iran.
- Inflammation Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| | - Mahdi Mahmoudi
- Rheumatology Research Center, Tehran University of Medical Sciences, Tehran, Iran.
- Inflammation Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
23
|
Lee WS, Kato M, Sugawara E, Kono M, Kudo Y, Kono M, Fujieda Y, Bohgaki T, Amengual O, Oku K, Yasuda S, Onodera T, Iwasaki N, Atsumi T. Protective Role of Optineurin Against Joint Destruction in Rheumatoid Arthritis Synovial Fibroblasts. Arthritis Rheumatol 2020; 72:1493-1504. [DOI: 10.1002/art.41290] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 04/14/2020] [Indexed: 01/02/2023]
Affiliation(s)
- Wen Shi Lee
- Hokkaido University, Sapporo, Japan, and Tokyo Medical and Dental University Tokyo Japan
| | | | | | | | | | | | | | | | | | | | - Shinsuke Yasuda
- Hokkaido University, Sapporo, Japan, and Tokyo Medical and Dental University Tokyo Japan
| | | | | | | |
Collapse
|
24
|
CXCL4 triggers monocytes and macrophages to produce PDGF-BB, culminating in fibroblast activation: Implications for systemic sclerosis. J Autoimmun 2020; 111:102444. [DOI: 10.1016/j.jaut.2020.102444] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 03/28/2020] [Accepted: 03/29/2020] [Indexed: 12/26/2022]
|
25
|
Wang BH, Lu YH, Wu LF, Lu X, Guo W, Deng FY, Lei SF. Evaluation of plasma cytokine protein array profile: the highlighted PDGF-BB in rheumatoid arthritis. Clin Rheumatol 2020; 39:3323-3330. [PMID: 32385763 DOI: 10.1007/s10067-020-05109-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 03/11/2020] [Accepted: 04/16/2020] [Indexed: 10/24/2022]
Abstract
OBJECTIVES The cytokines play critical roles in the complex pathogenesis of rheumatoid arthritis (RA), but the specific cytokines are still in need of being discovered. This multi-stage study was performed to identify novel RA cytokines in plasma and further understand the pathological mechanism of the identified cytokines. METHOD The plasma cytokine protein profile was evaluated by using Human Cytokine Antibody Array 440 in 18 subjects (RA: healthy control = 9:9). Then, enzyme-linked immunosorbent assay (ELISA) was used to validate the highlighted cytokines in 80 subjects (RA: healthy control = 40:40). Further functional experiments on fibroblast-like synoviocytes were performed to identify the pathological mechanisms of the highlighted cytokines for RA. RESULTS A total of seven significant cytokines have differential expressions between RA patients and controls (fold change (FC) > 2, P value < 0.05). The difference in plasma for the highlighted platelet-derived growth factor (PDGF)-BB was validated in an independent validation sample (P = 0.005). Further, the PDGF-BB obviously promotes cell proliferation of MH7A cell, probably by inhibiting cell apoptosis and accelerating the cell cycle. The PDGF-BB can also promote MH7A cell migration. CONCLUSIONS This study evaluated the plasma cytokine protein array profile associated with RA and highlighted the importance of PDGF-BB. PDGF-BB has an important role in RA-FLS proliferation and migration. These results suggest that PDGF-BB might contribute to occurrence and development of RA. Key Points • The levels of plasma cytokines were systemically tested using Human Cytokine Antibody Arrays. • The expression difference of PDGF-BB was validated in an independent sample. • PDGF-BB obviously promotes cell proliferation and migration in RA-FLS.
Collapse
Affiliation(s)
- Bing-Hua Wang
- Center for Genetic Epidemiology and Genomics, School of Public Health, Medical College of Soochow University, Suzhou, Jiangsu, People's Republic of China.,Department of Epidemiology and Health Statistics, School of Public Health, Xuzhou Medical University, Xuzhou, Jiangsu, People's Republic of China.,Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, Jiangsu, People's Republic of China
| | - Yi-Hua Lu
- Center for Genetic Epidemiology and Genomics, School of Public Health, Medical College of Soochow University, Suzhou, Jiangsu, People's Republic of China.,Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, Jiangsu, People's Republic of China
| | - Long-Fei Wu
- Center for Genetic Epidemiology and Genomics, School of Public Health, Medical College of Soochow University, Suzhou, Jiangsu, People's Republic of China.,Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, Jiangsu, People's Republic of China
| | - Xin Lu
- Center for Genetic Epidemiology and Genomics, School of Public Health, Medical College of Soochow University, Suzhou, Jiangsu, People's Republic of China.,Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, Jiangsu, People's Republic of China
| | - Wei Guo
- Center for Genetic Epidemiology and Genomics, School of Public Health, Medical College of Soochow University, Suzhou, Jiangsu, People's Republic of China.,Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, Jiangsu, People's Republic of China
| | - Fei-Yan Deng
- Center for Genetic Epidemiology and Genomics, School of Public Health, Medical College of Soochow University, Suzhou, Jiangsu, People's Republic of China.,Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, Jiangsu, People's Republic of China
| | - Shu-Feng Lei
- Center for Genetic Epidemiology and Genomics, School of Public Health, Medical College of Soochow University, Suzhou, Jiangsu, People's Republic of China. .,Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, Jiangsu, People's Republic of China.
| |
Collapse
|
26
|
Mariani E, Roffi A, Cattini L, Pulsatelli L, Assirelli E, Krishnakumar GS, Cenacchi A, Kon E, Filardo G. Release kinetic of pro- and anti-inflammatory biomolecules from platelet-rich plasma and functional study on osteoarthritis synovial fibroblasts. Cytotherapy 2020; 22:344-353. [PMID: 32327304 DOI: 10.1016/j.jcyt.2020.02.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 02/24/2020] [Accepted: 02/24/2020] [Indexed: 01/17/2023]
Abstract
BACKGROUND AIMS This study evaluated the release kinetics of numerous representative and less studied platelet-rich plasma (PRP) cytokines/chemokines with regard to the effects of various cellular compositions and incubation times. In addition, the biological effects of different PRPs on osteoarthritis synovial fibroblasts in vitro were tested. METHODS Peripheral whole blood was collected from healthy donors, and pure platelet-rich plasma (P-PRP), leukocyte-rich platelet-rich plasma (L-PRP) and platelet-poor plasma (PPP) were prepared for the analysis of the following biomolecules: IL-1β, IL-4, IL-6, IL-10, IL-17a, IL-22, MIP-1α/CCL-3, RANTES/CCL-5, MCP-3/CCL-7, Gro-α/CXCL-1, PF-4/CXCL-4, ENA-78/CXCL-5, NAP-2/CXCL-7, IL-8/CXCL-8, Fractalkine/CX3CL-1, s-CD40L P-PRP, L-PRP and PPP. Their effect on osteoarthritis synovial fibroblasts in vitro was tested by analyzing changes induced in both gene expression on a panel of representative molecules involved in physiopathology of joint environment and synthesis of IL-1β, IL-8 and hyaluronic acid. RESULTS This study demonstrated that among the 16 analyzed biomolecules, four were undetectable, whereas most of the detected biomolecules were more concentrated in L-PRP even when concentrations were normalized to platelet number. Despite the pro-inflammatory boost, the various PRP preparations did not alter synovial fibroblast gene expression of specific factors that play a pivotal role in joint tissue homeostasis and are able to induce anti-inflammatory (TIMP-1) biomolecules. DISCUSSION This study provides a set of reference data on the concentration and release kinetics of some less explored biomolecules that could represent potential specific effectors in the modulation of inflammatory processes and in tissue repair after treatment with PRP.
Collapse
Affiliation(s)
- Erminia Mariani
- Laboratorio di Immunoreumatologia e rigenerazione tissutale, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy; Dipartimento di Scienze Mediche e Chirurgiche, Alma Mater Studiorum-Università di Bologna, Bologna, Italy.
| | - Alice Roffi
- Applied and Translational Research Center, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Luca Cattini
- Laboratorio di Immunoreumatologia e rigenerazione tissutale, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Lia Pulsatelli
- Laboratorio di Immunoreumatologia e rigenerazione tissutale, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Elisa Assirelli
- Laboratorio di Immunoreumatologia e rigenerazione tissutale, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Gopal Shankar Krishnakumar
- Department of Biotechnology, Bannari Amman Institute of Technology, Sathyamangalam, Erode, Tamil Nadu, India
| | - Annarita Cenacchi
- Servizio di Immunoematologia e Medicina trasfusionale, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Elizaveta Kon
- Humanitas University Department of Biomedical Sciences, Humanitas Clinical and Research Center, Milan, Italy
| | - Giuseppe Filardo
- Applied and Translational Research Center, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| |
Collapse
|
27
|
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease in which a variety of circulating pro-inflammatory cells and dysregulated molecules are involved in disease aetiology and progression. Platelets are an important cellular element in the circulation that can bind several dysregulated molecules (such as collagen, thrombin and fibrinogen) that are present both in the synovium and the circulation of patients with RA. Platelets not only respond to dysregulated molecules in their environment but also transport and express their own inflammatory mediators, and serve as regulators at the boundary between haemostasis and immunity. Activated platelets also produce microparticles, which further convey signalling molecules and receptors to the synovium and circulation, thereby positioning these platelet-derived particles as strategic regulators of inflammation. These diverse functions come together to make platelets facilitators of cellular crosstalk in RA. Thus, the receptor functions, ligand binding potential and dysregulated signalling pathways in platelets are becoming increasingly important for treatment in RA. This Review aims to highlight the role of platelets in RA and the need to closely examine platelets as health indicators when designing effective pharmaceutical targets in this disease.
Collapse
|
28
|
Pap T, Dankbar B, Wehmeyer C, Korb-Pap A, Sherwood J. Synovial fibroblasts and articular tissue remodelling: Role and mechanisms. Semin Cell Dev Biol 2020; 101:140-145. [PMID: 31956018 DOI: 10.1016/j.semcdb.2019.12.006] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 12/12/2019] [Accepted: 12/12/2019] [Indexed: 02/06/2023]
Abstract
Synovial joints are unique functional elements of the body and provide the ability for locomotion and for physical interaction with the environment. They are composed of different connective tissue structures, of which the synovial membrane is one central component. It shows a number of peculiarities that makes it different from other membranes in our body, while several lines of evidence suggest that synovial fibroblasts, also termed fibroblast-like synoviocytes (FLS) critically contribute to these peculiarities. This becomes evident particularly under disease conditions such as in rheumatoid arthritis and osteoarthritis, where the synovium is a key pathophysiological component. Therefore, an in-depth knowledge of FLS biology is not only important for understanding key features of articular function but also provides explanations for important characteristics of both degenerative and inflammatory joint diseases. This article reviews the structure, biochemical composition and functions of the synovial membrane and by focusing on the role of synovial fibroblasts explains key features of articular tissue remodelling particularly under disease conditions.
Collapse
Affiliation(s)
- Thomas Pap
- Institute of Musculoskeletal Medicine (IMM), Westfalian Wilhelms-University Münster, Germany.
| | - Berno Dankbar
- Institute of Musculoskeletal Medicine (IMM), Westfalian Wilhelms-University Münster, Germany
| | - Corinna Wehmeyer
- Institute of Musculoskeletal Medicine (IMM), Westfalian Wilhelms-University Münster, Germany
| | - Adelheid Korb-Pap
- Institute of Musculoskeletal Medicine (IMM), Westfalian Wilhelms-University Münster, Germany
| | - Joanna Sherwood
- Institute of Musculoskeletal Medicine (IMM), Westfalian Wilhelms-University Münster, Germany
| |
Collapse
|
29
|
Singh V, Kalliolias GD, Ostaszewski M, Veyssiere M, Pilalis E, Gawron P, Mazein A, Bonnet E, Petit-Teixeira E, Niarakis A. RA-map: building a state-of-the-art interactive knowledge base for rheumatoid arthritis. Database (Oxford) 2020; 2020:baaa017. [PMID: 32311035 PMCID: PMC7170216 DOI: 10.1093/database/baaa017] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Revised: 01/21/2020] [Accepted: 02/13/2020] [Indexed: 02/07/2023]
Abstract
Rheumatoid arthritis (RA) is a progressive, inflammatory autoimmune disease of unknown aetiology. The complex mechanism of aetiopathogenesis, progress and chronicity of the disease involves genetic, epigenetic and environmental factors. To understand the molecular mechanisms underlying disease phenotypes, one has to place implicated factors in their functional context. However, integration and organization of such data in a systematic manner remains a challenging task. Molecular maps are widely used in biology to provide a useful and intuitive way of depicting a variety of biological processes and disease mechanisms. Recent large-scale collaborative efforts such as the Disease Maps Project demonstrate the utility of such maps as versatile tools to organize and formalize disease-specific knowledge in a comprehensive way, both human and machine-readable. We present a systematic effort to construct a fully annotated, expert validated, state-of-the-art knowledge base for RA in the form of a molecular map. The RA map illustrates molecular and signalling pathways implicated in the disease. Signal transduction is depicted from receptors to the nucleus using the Systems Biology Graphical Notation (SBGN) standard representation. High-quality manual curation, use of only human-specific studies and focus on small-scale experiments aim to limit false positives in the map. The state-of-the-art molecular map for RA, using information from 353 peer-reviewed scientific publications, comprises 506 species, 446 reactions and 8 phenotypes. The species in the map are classified to 303 proteins, 61 complexes, 106 genes, 106 RNA entities, 2 ions and 7 simple molecules. The RA map is available online at ramap.elixir-luxembourg.org as an open-access knowledge base allowing for easy navigation and search of molecular pathways implicated in the disease. Furthermore, the RA map can serve as a template for omics data visualization.
Collapse
Affiliation(s)
- Vidisha Singh
- Laboratoire Européen de Recherche pour la Polyarthrite Rhumatoïde - Genhotel, Univ Evry, Université Paris-Saclay, 2, rue Gaston Crémieux, 91057 EVRY-GENOPOLE cedex, Evry, France
| | - George D Kalliolias
- Arthritis and Tissue Degeneration Program, Hospital for Special Surgery, 535 East 70th Street, New York, NY 10021, USA
- Weill Cornell Medical Center, Weill Department of Medicine, 525 East 68th Street, New York, NY 10065, USA
| | - Marek Ostaszewski
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, 6 Avenue du Swing, L-4367 Belvaux, Luxembourg
| | - Maëva Veyssiere
- Laboratoire Européen de Recherche pour la Polyarthrite Rhumatoïde - Genhotel, Univ Evry, Université Paris-Saclay, 2, rue Gaston Crémieux, 91057 EVRY-GENOPOLE cedex, Evry, France
| | - Eleftherios Pilalis
- eNIOS Applications P.C., R&D department, Alexandrou Pantou 25, 17671, Kallithea-Athens, Greece
| | - Piotr Gawron
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, 6 Avenue du Swing, L-4367 Belvaux, Luxembourg
| | - Alexander Mazein
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, 6 Avenue du Swing, L-4367 Belvaux, Luxembourg
| | - Eric Bonnet
- Centre National de Recherche en Génomique Humaine (CNRGH), CEA, 2 rue Gaston Crémieux, CP5706 91057 EVRY-GENOPOLE cedex, Evry, France
| | - Elisabeth Petit-Teixeira
- Laboratoire Européen de Recherche pour la Polyarthrite Rhumatoïde - Genhotel, Univ Evry, Université Paris-Saclay, 2, rue Gaston Crémieux, 91057 EVRY-GENOPOLE cedex, Evry, France
| | - Anna Niarakis
- Laboratoire Européen de Recherche pour la Polyarthrite Rhumatoïde - Genhotel, Univ Evry, Université Paris-Saclay, 2, rue Gaston Crémieux, 91057 EVRY-GENOPOLE cedex, Evry, France
| |
Collapse
|
30
|
Sung PJ, Rama N, Imbach J, Fiore S, Ducarouge B, Neves D, Chen HW, Bernard D, Yang PC, Bernet A, Depil S, Mehlen P. Cancer-Associated Fibroblasts Produce Netrin-1 to Control Cancer Cell Plasticity. Cancer Res 2019; 79:3651-3661. [DOI: 10.1158/0008-5472.can-18-2952] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 02/06/2019] [Accepted: 05/10/2019] [Indexed: 11/16/2022]
|
31
|
Melisi D, Garcia-Carbonero R, Macarulla T, Pezet D, Deplanque G, Fuchs M, Trojan J, Kozloff M, Simionato F, Cleverly A, Smith C, Wang S, Man M, Driscoll KE, Estrem ST, Lahn MMF, Benhadji KA, Tabernero J. TGFβ receptor inhibitor galunisertib is linked to inflammation- and remodeling-related proteins in patients with pancreatic cancer. Cancer Chemother Pharmacol 2019; 83:975-991. [PMID: 30887178 DOI: 10.1007/s00280-019-03807-4] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 03/01/2019] [Indexed: 12/26/2022]
Abstract
PURPOSE Galunisertib, the first small molecule transforming growth factor beta (TGFβ) receptor inhibitor, plus gemcitabine resulted in the improvement of survival in patients with unresectable pancreatic cancer, but markers to identify patients likely to respond are lacking. METHODS In the Phase 1b/2 JBAJ study, 156 patients were randomized 2:1 to galunisertib + gemcitabine (N = 104) or placebo + gemcitabine (N = 52). Clinical outcome data were integrated with baseline markers and pharmacodynamic markers while patients were on treatment, including circulating proteins using a multi-analyte panel, T cell subset evaluation, and miRNA profiling. RESULTS Baseline biomarkers associated with overall prognosis regardless of treatment included CA19-9 and TGF-β1. In addition, IP-10, FSH, MIP-1α, and PAI-1 were potential predictive proteins. Baseline proteins that were changed during treatment included amphiregulin, CA15-3, cathepsin D, P-selectin, RAGE, sortilin, COMP, eotaxin-2, N-BNP, osteopontin, and thrombospondin-4. Plasma miRNA with potential prognostic value included miR-21-5p, miR-301a-3p, miR-210-3p, and miR-141-3p, while those with potential predictive value included miR-424-5p, miR-483-3p, and miR-10b-5p. CONCLUSIONS Galunisertib + gemcitabine resulted in improvement of overall survival, and 4 proteins (IP-10, FSH, MIP-1α, PAI-1) were potentially predictive for this combination treatment. Future studies should also include baseline evaluation of miR-424-5p, miR-483-3p, and miR-10b-5p. TRIAL REGISTRATION Clinicaltrials.gov NCT01373164.
Collapse
Affiliation(s)
- Davide Melisi
- Digestive Molecular Clinical Oncology Research Unit, Section of Medical Oncology, Department of Medicine, Università degli studi di Verona, Piazzale L.A. Scuro, 10, 37134, Verona, Italy.
| | - Rocio Garcia-Carbonero
- University Hospital Doce de Octubre, Institute of Health Research Hospital 12 de Octubre (imas12), UCM, CNIO, CIBERONC, Madrid, Spain
| | - Teresa Macarulla
- Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology, Autonomous University of Barcelona, CIBERONC, Barcelona, Spain
| | - Denis Pezet
- Digestive Surgery Service, CHU Clermont-Ferrand, University Clermont Auvergne, Clermont-Ferrand, France
| | | | - Martin Fuchs
- Hospital Bogenhausen, Municipal Hospital Munich GmbH, Munich, Germany
| | - Jorg Trojan
- Goethe University Medical Center, Frankfurt, Germany
| | | | - Francesca Simionato
- Digestive Molecular Clinical Oncology Research Unit, Section of Medical Oncology, Department of Medicine, Università degli studi di Verona, Piazzale L.A. Scuro, 10, 37134, Verona, Italy
| | | | | | | | - Michael Man
- Eli Lilly and Company, Indianapolis, IN, USA
| | | | | | | | | | - Josep Tabernero
- Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology, Autonomous University of Barcelona, CIBERONC, Barcelona, Spain
| |
Collapse
|
32
|
Madarampalli B, Watts GFM, Panipinto PM, Nguygen HN, Brenner MB, Noss EH. Interactions between cadherin-11 and platelet-derived growth factor receptor-alpha signaling link cell adhesion and proliferation. Biochim Biophys Acta Mol Basis Dis 2019; 1865:1516-1524. [PMID: 30876808 DOI: 10.1016/j.bbadis.2019.03.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 02/25/2019] [Accepted: 03/11/2019] [Indexed: 12/26/2022]
Abstract
Cadherins are homophilic cell-to-cell adhesion molecules that help cells respond to environmental changes. Newly formed cadherin junctions are associated with increased cell phosphorylation, but the pathways driving this signaling response are largely unknown. Since cadherins have no intrinsic signaling activity, this phosphorylation must occur through interactions with other signaling molecules. We previously reported that cadherin-11 engagement activates joint synovial fibroblasts, promoting inflammatory and degradative pathways important in rheumatoid arthritis (RA) pathogenesis. Our objective in this study was to discover interacting partners that mediate cadherin-11 signaling. Protein array screening showed that cadherin-11 extracellular binding domains linked to an Fc domain (cad11Fc) induced platelet-derived growth factor (PDGFR)-α phosphorylation in synovial fibroblasts and glioblastoma cells. PDGFRs are growth factor receptor tyrosine kinases that promote cell proliferation, survival, and migration in mesodermally derived cells. Increased PDGFR activity is implicated in RA pathology and associates with poor prognosis in several cancers, including sarcoma and glioblastoma. PDGFRα activation by cadherin-11 signaling promoted fibroblast proliferation, a signaling pathway independent from cadherin-11-stimulated IL-6 or matrix metalloproteinase (MMP)-3 release. PDGFRα phosphorylation mediated most of the cad11Fc-induced phosphatidyl-3-kinase (PI3K)/Akt activation, but only part of the mitogen-activated protein kinase (MAPK) response. PDGFRα-dependent signaling did not require cell cadherin-11 expression. Rather, cad11Fc immunoprecipitated PDGFRα, indicating a direct interaction between cadherin-11 and PDGFRα extracellular domains. This study is the first to report an interaction between cadherin-11 and PDGFRα and adds to our growing understanding that cadherin-growth factor receptor interactions help balance the interplay between tissue growth and adhesion.
Collapse
Affiliation(s)
- Bhanupriya Madarampalli
- Division of Rheumatology, Department of Medicine, University of Washington, 750 Republican St, Seattle, WA 98019, USA.
| | - Gerald F M Watts
- Division of Rheumatology, Immunology, and Allergy, Brigham and Women's Hospital and Harvard Medical School, 75 Francis St, Boston, MA 02115, USA.
| | - Paul M Panipinto
- Division of Rheumatology, Department of Medicine, University of Washington, 750 Republican St, Seattle, WA 98019, USA.
| | - Hung N Nguygen
- Division of Rheumatology, Immunology, and Allergy, Brigham and Women's Hospital and Harvard Medical School, 75 Francis St, Boston, MA 02115, USA.
| | - Michael B Brenner
- Division of Rheumatology, Immunology, and Allergy, Brigham and Women's Hospital and Harvard Medical School, 75 Francis St, Boston, MA 02115, USA.
| | - Erika H Noss
- Division of Rheumatology, Department of Medicine, University of Washington, 750 Republican St, Seattle, WA 98019, USA; Division of Rheumatology, Immunology, and Allergy, Brigham and Women's Hospital and Harvard Medical School, 75 Francis St, Boston, MA 02115, USA.
| |
Collapse
|
33
|
Bottini A, Wu DJ, Ai R, Le Roux M, Bartok B, Bombardieri M, Doody KM, Zhang V, Sacchetti C, Zoccheddu M, Lonic A, Li X, Boyle DL, Hammaker D, Meng TC, Liu L, Corr M, Stanford SM, Lewis M, Wang W, Firestein GS, Khew-Goodall Y, Pitzalis C, Bottini N. PTPN14 phosphatase and YAP promote TGFβ signalling in rheumatoid synoviocytes. Ann Rheum Dis 2019; 78:600-609. [PMID: 30808624 DOI: 10.1136/annrheumdis-2018-213799] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 01/28/2019] [Accepted: 01/30/2019] [Indexed: 11/04/2022]
Abstract
OBJECTIVE We aimed to understand the role of the tyrosine phosphatase PTPN14-which in cancer cells modulates the Hippo pathway by retaining YAP in the cytosol-in fibroblast-like synoviocytes (FLS) from patients with rheumatoid arthritis (RA). METHODS Gene/protein expression levels were measured by quantitative PCR and/or Western blotting. Gene knockdown in RA FLS was achieved using antisense oligonucleotides. The interaction between PTPN14 and YAP was assessed by immunoprecipitation. The cellular localisation of YAP and SMAD3 was examined via immunofluorescence. SMAD reporter studies were carried out in HEK293T cells. The RA FLS/cartilage coimplantation and passive K/BxN models were used to examine the role of YAP in arthritis. RESULTS RA FLS displayed overexpression of PTPN14 when compared with FLS from patients with osteoarthritis (OA). PTPN14 knockdown in RA FLS impaired TGFβ-dependent expression of MMP13 and potentiation of TNF signalling. In RA FLS, PTPN14 formed a complex with YAP. Expression of PTPN14 or nuclear YAP-but not of a non-YAP-interacting PTPN14 mutant-enhanced SMAD reporter activity. YAP promoted TGFβ-dependent SMAD3 nuclear localisation in RA FLS. Differences in epigenetic marks within Hippo pathway genes, including YAP, were found between RA FLS and OA FLS. Inhibition of YAP reduced RA FLS pathogenic behaviour and ameliorated arthritis severity. CONCLUSION In RA FLS, PTPN14 and YAP promote nuclear localisation of SMAD3. YAP enhances a range of RA FLS pathogenic behaviours which, together with epigenetic evidence, points to the Hippo pathway as an important regulator of RA FLS behaviour.
Collapse
Affiliation(s)
- Angel Bottini
- Dept. of Medicine, University of California San Diego, La Jolla, California, USA.,Division of Cellular Biology, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Dennis J Wu
- Dept. of Medicine, University of California San Diego, La Jolla, California, USA
| | - Rizi Ai
- Dept. of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA, USA
| | - Michelle Le Roux
- Division of Cellular Biology, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Beatrix Bartok
- Dept. of Medicine, University of California San Diego, La Jolla, California, USA
| | - Michele Bombardieri
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Karen M Doody
- Division of Cellular Biology, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Vida Zhang
- Dept. of Medicine, University of California San Diego, La Jolla, California, USA.,Division of Cellular Biology, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Cristiano Sacchetti
- Dept. of Medicine, University of California San Diego, La Jolla, California, USA.,Division of Cellular Biology, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Martina Zoccheddu
- Dept. of Medicine, University of California San Diego, La Jolla, California, USA
| | - Ana Lonic
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, South Australia, Australia
| | - Xiaochun Li
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, South Australia, Australia
| | - David L Boyle
- Dept. of Medicine, University of California San Diego, La Jolla, California, USA
| | - Deepa Hammaker
- Dept. of Medicine, University of California San Diego, La Jolla, California, USA
| | - Tzu-Ching Meng
- Institute for Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Lin Liu
- Dept. of Family Medicine and Public Health, University of California San Diego, La Jolla, CA, USA
| | - Maripat Corr
- Dept. of Medicine, University of California San Diego, La Jolla, California, USA
| | - Stephanie M Stanford
- Dept. of Medicine, University of California San Diego, La Jolla, California, USA.,Division of Cellular Biology, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Myles Lewis
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Wei Wang
- Dept. of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA, USA.,Dept. of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
| | - Gary S Firestein
- Dept. of Medicine, University of California San Diego, La Jolla, California, USA
| | - Yeesim Khew-Goodall
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, South Australia, Australia
| | - Costantino Pitzalis
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Nunzio Bottini
- Dept. of Medicine, University of California San Diego, La Jolla, California, USA .,Division of Cellular Biology, La Jolla Institute for Immunology, La Jolla, CA, USA
| |
Collapse
|
34
|
Matsumura T, Saito Y, Suzuki T, Teramoto A, Ozasa Y, Yamashita T, Fujimiya M, Saito-Chikenji T. Phosphorylated Platelet-Derived Growth Factor Receptor-Positive Cells With Anti-apoptotic Properties Accumulate in the Synovium of Patients With Rheumatoid Arthritis. Front Immunol 2019; 10:241. [PMID: 30828336 PMCID: PMC6384265 DOI: 10.3389/fimmu.2019.00241] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 01/28/2019] [Indexed: 12/29/2022] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease caused by inflammation of the synovium and characterized by chronic polyarthritis that destroys bone and cartilage. Fibroblast-like synoviocytes (FLSs) in the synovium of patients with RA can promote cartilage and bone destruction by producing proteins such as matrix metalloproteinases and receptor activator of NF-κB ligand, thereby representing an important therapeutic target for RA. FLSs have several phenotypes depending on which cell surface proteins and adhesion factors are expressed. Identifying the cellular functions associated with different phenotypes and methods of controlling them are considered essential for developing therapeutic strategies for RA. In this study, synovial tissue was collected from patients with RA and control subjects who required surgery due to ligament injury or fracture. Immunohistological analysis was used to investigate the rates of positivity for phosphorylated platelet-derived growth factor receptor-αβ (pPDGFRαβ) and cadherin-11 (CDH11) expression, and apoptosis-related markers were assessed for each cell phenotype. Next, FLSs were isolated in vitro and stimulated with tumor necrosis factor-α (TNF-α) in addition to a combination of PDGF and transforming growth factor (2GF) to investigate pPDGFRαβ and CDH11 expression and the effects of the inhibition of TNF and cyclin-dependent kinase (CDK) 4/6 on FLSs. Immunohistological analysis showed a large percentage of pPDGFRαβ+CDH11– cells in the sub-lining layer (SL) of patients with RA. These cells exhibited increased B-cell lymphoma-2 expression, reduced TNF receptor-1 expression, resistance to cell death, and abnormal proliferation, suggesting a tendency to accumulate in the synovium. Further, in vitro 2GF stimulation of FLSs lowered, whereas 2GF + TNF stimulation increased the pPDGFRαβ/CDH11 ratio. Hypothesizing that FLSs stimulated with 2GF + TNF would accumulate in vivo in RA, we determined the therapeutic effects of TNF and CDK4/6 inhibitors. The TNF inhibitor lowered the pPDGFRαβ/CDH11 ratio, whereas the CDK4/6 inhibitor suppressed cell proliferation. However, a synergistic effect was not observed by combining both the drugs. We observed an increase in pPDGFRαβ+CDH11– cells in the SL of the RA synovium and accumulation of these cells in the synovium. We found that the TNF inhibitor suppressed FLS activity and the CDK4/6 inhibitor reduced cell proliferation.
Collapse
Affiliation(s)
- Takashi Matsumura
- Department of Orthopaedic Surgery, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Yuki Saito
- Department of Anatomy, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Tomoyuki Suzuki
- Department of Orthopaedic Surgery, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Atsushi Teramoto
- Department of Orthopaedic Surgery, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Yasuhiro Ozasa
- Department of Orthopaedic Surgery, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Toshihiko Yamashita
- Department of Orthopaedic Surgery, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Mineko Fujimiya
- Department of Anatomy, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Takako Saito-Chikenji
- Department of Anatomy, Sapporo Medical University School of Medicine, Sapporo, Japan
| |
Collapse
|
35
|
Bergström B, Carlsten H, Ekwall AKH. Methotrexate inhibits effects of platelet-derived growth factor and interleukin-1β on rheumatoid arthritis fibroblast-like synoviocytes. Arthritis Res Ther 2018; 20:49. [PMID: 29554943 PMCID: PMC5859417 DOI: 10.1186/s13075-018-1554-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 02/25/2018] [Indexed: 11/23/2022] Open
Abstract
Background A key feature of joints in rheumatoid arthritis (RA) is the formation of hyperplastic destructive pannus tissue, which is orchestrated by activated fibroblast-like synoviocytes (FLS). We have demonstrated that the RA risk gene and tumor suppressor Limb bud and heart development (LBH) regulates cell cycle progression in FLS. Methotrexate (MTX) is the first-line treatment for RA, but its mechanisms of action remain incompletely understood. Here, we studied the effects of MTX on mitogen-induced FLS proliferation and expression of cell cycle regulators in vitro. Methods Primary FLS from patients with RA or osteoarthritis were stimulated with the mitogen platelet-derived growth factor (PDGF) and the cytokine interleukin-1β (IL-1β) in the presence or absence of MTX. Cells were then subjected to qPCR for gene expression and cell cycle analysis by flow cytometry. Results Stimulation with PDGF and IL-1β increased the percentage of FLS in the G2/M phase and shifted the cell morphology to a dendritic shape. These effects were inhibited by MTX. Furthermore, PDGF + IL-1β reduced LBH mRNA expression. However, MTX treatment yielded significantly higher transcript levels of LBH, and of CDKN1A (p21) and TP53 (p53), compared to untreated samples upon mitogen stimulation. The expression of DNA methyltransferase-1 (DNMT1) was also higher in the presence of MTX and there was strong correlation between DNMT1 and LBH expression. Conclusions Therapeutic concentrations of MTX abolish the effects of PDGF and IL-1β on tumor suppressor expression and inhibit mitogen-promoted FLS proliferation. These data demonstrate novel and important effects of MTX on pathogenic effector cells in the joint, which might involve epigenetic mechanisms.
Collapse
Affiliation(s)
- Beatrice Bergström
- Department of Rheumatology and Inflammation Research, Institute of Medicine, The Sahlgrenska Academy, University of Gothenburg, Box 480, 405 30, Gothenburg, Sweden.,Centre for Bone and Arthritis Research, University of Gothenburg, Gothenburg, Sweden
| | - Hans Carlsten
- Department of Rheumatology and Inflammation Research, Institute of Medicine, The Sahlgrenska Academy, University of Gothenburg, Box 480, 405 30, Gothenburg, Sweden.,Centre for Bone and Arthritis Research, University of Gothenburg, Gothenburg, Sweden
| | - Anna-Karin Hultgård Ekwall
- Department of Rheumatology and Inflammation Research, Institute of Medicine, The Sahlgrenska Academy, University of Gothenburg, Box 480, 405 30, Gothenburg, Sweden. .,Centre for Bone and Arthritis Research, University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
36
|
Huhtakangas JA, Veijola J, Turunen S, Karjalainen A, Valkealahti M, Nousiainen T, Yli-Luukko S, Vuolteenaho O, Lehenkari P. 1,25(OH) 2D 3 and calcipotriol, its hypocalcemic analog, exert a long-lasting anti-inflammatory and anti-proliferative effect in synoviocytes cultured from patients with rheumatoid arthritis and osteoarthritis. J Steroid Biochem Mol Biol 2017; 173:13-22. [PMID: 28167299 DOI: 10.1016/j.jsbmb.2017.01.017] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 01/08/2017] [Accepted: 01/24/2017] [Indexed: 01/01/2023]
Abstract
OBJECTIVES We investigated the effects of 1,25-dihydroxy vitamin D3 (1,25(OH)2D3), i.e. biologically active vitamin D and calcipotriol, a vitamin D analog, on growth and secretion of inflammatory mediators in synovial stromal cells (SSC) of patients with rheumatoid arthritis (RA) or osteoarthritis (OA). METHODS Synovial stromal cells (SSC) isolated during knee prosthesis surgery from four patients with RA and four with OA were exposed to 1,25(OH)2D3 or calcipotriol with or without stimulation of cells with IL-1β or TNF-α. The proliferation of cells was studied by MTT assay. Levels of cytokines were analyzed by a magnetic bead-based multiplex assay (a panel of 27 important cytokines and IL-6 alone) and RT-PCR was used to validate the concentrations of the key cytokines secreted by SSC. The vitamin D receptor (VDR) was visualized by immunofluorescence in SSC and by immunohistochemistry in the synovial tissues of three RA and three OA patients. RESULTS We detected intense staining for VDR in the synovial lining and vascular endothelium in tissue sections from all our RA and OA patients. Both 1,25(OH)2D3 and calcipotriol inhibited SSC proliferation for a prolonged time (up to 23 days with calcipotriol), but dexamethasone tended to increase SSC proliferation in a 4-day culture. 1,25(OH)2D3, calcipotriol and dexamethasone reduced the secretion of most inflammatory factors. Calcipotriol and dexamethasone additively reduced the secretions of IL-6, IFN-γ, basic FGF and VEGF in TNF-α stimulated SSC. The level of IL-6 was still diminished at 10 days after exposure, emphasizing the long-term impact of calcipotriol on SSC. CONCLUSIONS Exposure for 24-48h to 1,25(OH)2D3 or calcipotriol causes a long-lasting inhibition of cell proliferation and cytokine production in SSC in vitro.
Collapse
Affiliation(s)
- Johanna A Huhtakangas
- Cancer and Translational Medicine Research Unit, Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland; Rheumatology Unit, Department of Medicine, Oulu University Hospital and University of Oulu, Oulu, Finland.
| | - Johanna Veijola
- Biochemistry and Molecular Medicine Research Unit, Medical Research Center Oulu, Oulu University, Oulu, Finland.
| | - Sanna Turunen
- Cancer and Translational Medicine Research Unit, Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland.
| | - Anna Karjalainen
- Rheumatology Unit, Department of Medicine, Oulu University Hospital and University of Oulu, Oulu, Finland.
| | - Maarit Valkealahti
- Division of Operative Care, Oulu University Hospital and University of Oulu, Oulu, Finland.
| | - Tomi Nousiainen
- Division of Operative Care, Oulu University Hospital and University of Oulu, Oulu, Finland.
| | - Susanna Yli-Luukko
- Division of Operative Care, Oulu University Hospital and University of Oulu, Oulu, Finland.
| | - Olli Vuolteenaho
- Biomedicine Unit, Department of Physiology, Medical Research Center Oulu, Oulu University, Oulu, Finland.
| | - Petri Lehenkari
- Cancer and Translational Medicine Research Unit, Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland; Division of Operative Care, Oulu University Hospital and University of Oulu, Oulu, Finland.
| |
Collapse
|
37
|
Mei HE, Hahne S, Redlin A, Hoyer BF, Wu K, Baganz L, Lisney AR, Alexander T, Rudolph B, Dörner T. Plasmablasts With a Mucosal Phenotype Contribute to Plasmacytosis in Systemic Lupus Erythematosus. Arthritis Rheumatol 2017. [DOI: 10.1002/art.40181] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Henrik E. Mei
- Charité University Medicine Berlin and German Rheumatism Research Center Berlin; Berlin Germany
| | - Stefanie Hahne
- Charité University Medicine Berlin and German Rheumatism Research Center Berlin; Berlin Germany
| | - Andreas Redlin
- Charité University Medicine Berlin and German Rheumatism Research Center Berlin; Berlin Germany
| | - Bimba F. Hoyer
- Charité University Medicine Berlin and German Rheumatism Research Center Berlin; Berlin Germany
| | - Kaiyin Wu
- Charité University Medicine Berlin; Berlin Germany
| | - Lisa Baganz
- German Rheumatism Research Center Berlin; Berlin Germany
| | - Anna R. Lisney
- Charité University Medicine Berlin and German Rheumatism Research Center Berlin; Berlin Germany
| | - Tobias Alexander
- Charité University Medicine Berlin and German Rheumatism Research Center Berlin; Berlin Germany
| | | | - Thomas Dörner
- Charité University Medicine Berlin and German Rheumatism Research Center Berlin; Berlin Germany
| |
Collapse
|
38
|
Zhao M, Hu Y, Jin J, Yu Y, Zhang S, Cao J, Zhai Y, Wei R, Shou J, Cai W, Liu S, Yang X, Xu GT, Yang J, Corry DB, Su SB, Liu X, Yang T. Interleukin 37 promotes angiogenesis through TGF-β signaling. Sci Rep 2017; 7:6113. [PMID: 28733640 PMCID: PMC5522482 DOI: 10.1038/s41598-017-06124-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 05/30/2017] [Indexed: 01/10/2023] Open
Abstract
IL-37 is a novel pro-angiogenic cytokine that potently promotes endothelial cell activation and pathological angiogenesis in our previous study, but the mechanisms behind the pro-angiogenic effect of IL-37 are less well understood. Extending our observations, we found that TGF-β interacts with IL-37, and potently enhances the binding affinity of IL-37 to the ALK1 receptor complex, thus allowing IL-37 to signal through ALK1 to activate pro-angiogenic responses. We further show that TGF-β and ALK1 are required in IL-37 induced pro-angiogenic response in ECs and in the mouse model of Matrigel plug and oxygen-induced retinopathy. The result suggests that IL-37 induces pro-angiogenic responses through TGF-β, which may act as the bridging molecule that mediates IL-37 binding to the TGF-β receptor complex.
Collapse
Affiliation(s)
- Mengmeng Zhao
- Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yongguang Hu
- Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jiayi Jin
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Ying Yu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Shanshan Zhang
- Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jingjing Cao
- Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yuanfen Zhai
- Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Rongbin Wei
- Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Juanjuan Shou
- Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Wenping Cai
- Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Shangfeng Liu
- Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xiaoping Yang
- Johns Hopkins University School of Medicine, Baltimore, United States
| | - Guo-Tong Xu
- Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jianhua Yang
- Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - David B Corry
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, United States
| | - Shao Bo Su
- Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.,State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Xialin Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China.
| | - Tianshu Yang
- Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.
| |
Collapse
|
39
|
Lefèvre S, Schwarz M, Meier FMP, Zimmermann-Geller B, Tarner IH, Rickert M, Steinmeyer J, Sauerbier M, Rehart S, Müller-Ladner U, Neumann E. Disease-Specific Effects of Matrix and Growth Factors on Adhesion and Migration of Rheumatoid Synovial Fibroblasts. THE JOURNAL OF IMMUNOLOGY 2017; 198:4588-4595. [PMID: 28500074 DOI: 10.4049/jimmunol.1600989] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Accepted: 04/10/2017] [Indexed: 01/15/2023]
Abstract
In rheumatoid arthritis (RA), cartilage and bone matrix are degraded, and extracellular matrix (ECM) proteins, acting as cellular activators, are liberated. Similar to ECM proteins, matrix-bound chemokines, cytokines, and growth factors (GFs) influence functional properties of key cells in RA, especially synovial fibroblasts. The role of these molecules on attachment, migration, and proinflammatory and prodestructive activation of RASFs was analyzed. Adhesion/migration of RASFs were examined under GF-enriched (GF+) or -reduced (GF-) conditions with or without addition of matrix-associated GFs, TGF-β, and platelet-derived GF to GF- or culture supernatants. Fibroblast adhesion and alterations in proinflammatory/prodestructive properties (e.g., IL-6/matrix metalloproteinase 3-release) in response to matrix-associated molecules were compared. Effects of GF+, GF-, and other ECM components on human RASF-mediated cartilage invasion were examined in the SCID mouse model. RASF adhesion under GF- conditions was significantly lower compared with GF+ conditions (6.8- versus 8.3-fold). This effect was specific for RA because control cells showed opposite effects (e.g., osteoarthritis synovial fibroblasts [SF]; GF- versus GF+: 10.7- versus 8-fold). Addition of TGF-β to GF- increased RASF attachment (12.7-fold) compared with other matrices and components. RASF adhesion to GF+ matrix resulted in the strongest IL-6 and matrix metalloproteinase-3 release, and was even more pronounced compared with supplementation of single GFs. In vivo, GF- matrix decreased RASF-mediated cartilage invasion compared with GF+ matrix. ECM components and especially GFs when bound within ECM actively enhance RASF attraction and cartilage adhesion. This observation was specific for RASFs as a reverse behavior was observed for controls.
Collapse
Affiliation(s)
- Stephanie Lefèvre
- Department of Internal Medicine and Rheumatology, Justus-Liebig-University of Giessen, Kerckhoff Clinic, 61231 Bad Nauheim, Germany
| | - Maria Schwarz
- Department of Internal Medicine and Rheumatology, Justus-Liebig-University of Giessen, Kerckhoff Clinic, 61231 Bad Nauheim, Germany
| | - Florian M P Meier
- Department of Internal Medicine and Rheumatology, Justus-Liebig-University of Giessen, Kerckhoff Clinic, 61231 Bad Nauheim, Germany
| | - Birgit Zimmermann-Geller
- Department of Internal Medicine and Rheumatology, Justus-Liebig-University of Giessen, Kerckhoff Clinic, 61231 Bad Nauheim, Germany
| | - Ingo H Tarner
- Department of Internal Medicine and Rheumatology, Justus-Liebig-University of Giessen, Kerckhoff Clinic, 61231 Bad Nauheim, Germany
| | - Markus Rickert
- Department of Orthopaedics and Orthopaedic Surgery, University Hospital Giessen and Marburg, 35392 Giessen, Germany
| | - Jürgen Steinmeyer
- Department of Orthopaedics and Orthopaedic Surgery, University Hospital Giessen and Marburg, 35392 Giessen, Germany
| | - Michael Sauerbier
- Department of Plastic, Hand and Reconstructive Surgery, BG Trauma Center, 60389 Frankfurt, Germany; and
| | - Stefan Rehart
- Department of Orthopaedics and Trauma Surgery, Agaplesion Markus-Hospital, 60431 Frankfurt, Germany
| | - Ulf Müller-Ladner
- Department of Internal Medicine and Rheumatology, Justus-Liebig-University of Giessen, Kerckhoff Clinic, 61231 Bad Nauheim, Germany
| | - Elena Neumann
- Department of Internal Medicine and Rheumatology, Justus-Liebig-University of Giessen, Kerckhoff Clinic, 61231 Bad Nauheim, Germany;
| |
Collapse
|
40
|
Hydrogel is Superior to Fibrin Gel as Matrix of Stem Cells in Alleviating Antigen-Induced Arthritis. Polymers (Basel) 2016; 8:polym8050182. [PMID: 30979276 PMCID: PMC6431989 DOI: 10.3390/polym8050182] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Revised: 03/31/2016] [Accepted: 04/28/2016] [Indexed: 01/08/2023] Open
Abstract
Recently, therapy with bone marrow mesenchymal stem cells (BMMSCs) has been attempted to relieve rheumatoid arthritis (RA) and reconstruct cartilage injury. However, treatment has been unsuccessful in complete prevention of persistent cartilage destruction and resulted in inferior outcomes of cartilage regeneration. Scaffolds are an important construct in the field of cartilage tissue engineering, but their role in arthritis treatment has not yet been fully examined. Here, we transplanted two types of scaffold-assisted BMMSCs: fibrin gel- and poly(l-lactide-co-glycolide)-poly(ethylene glycol)-poly(l-lactide-co-glycolide) (PLGA-PEG-PLGA) hydrogel-assisted BMMSCs referred as FGB and HGB groups, respectively, into subchondral defects for the treatment of antigen-induced arthritis. The administration of exogenous BMMSCs ameliorated joint swelling and decreased both joint surface temperature and inflammatory cytokine levels in both groups. Immune cell composition of the inflammation of surrounding synovium, protection of adjacent cartilage, and improved cartilage repair were also observed. Overall, the HGB group had a better therapeutic efficacy than the FGB group. In conclusion, local transplantation of BMMSCs in subchondral defects presents a novel approach in inducing RA remission and recovery of RA-induced cartilage injury. To induce these changes, the selection of scaffold for cell support is exceedingly important. Further studies are needed regarding the treatment options of subchondral defects in arthritis based on modified scaffold development, application of defined MSCs sources, combination of pharmacotherapeutics, and the addition of factors that inhibit the processes of RA remission, promote the recovery of RA-induced cartilage injury and the relationship between these factors.
Collapse
|
41
|
Charbonneau M, Lavoie RR, Lauzier A, Harper K, McDonald PP, Dubois CM. Platelet-Derived Growth Factor Receptor Activation Promotes the Prodestructive Invadosome-Forming Phenotype of Synoviocytes from Patients with Rheumatoid Arthritis. THE JOURNAL OF IMMUNOLOGY 2016; 196:3264-75. [DOI: 10.4049/jimmunol.1500502] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 02/15/2016] [Indexed: 11/19/2022]
|
42
|
Lauzier A, Lavoie RR, Charbonneau M, Gouin-Boisvert B, Harper K, Dubois CM. Snail Is a Critical Mediator of Invadosome Formation and Joint Degradation in Arthritis. THE AMERICAN JOURNAL OF PATHOLOGY 2015; 186:359-74. [PMID: 26704941 DOI: 10.1016/j.ajpath.2015.10.021] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Revised: 08/31/2015] [Accepted: 10/07/2015] [Indexed: 01/08/2023]
Abstract
Progressive cartilage destruction, mediated by invasive fibroblast-like synoviocytes, is a central feature in the pathogenesis of rheumatoid arthritis (RA). Members of the Snail family of transcription factors are required for cell migration and invasion, but their role in joint destruction remains unknown. Herein, we demonstrate that Snail is essential for the formation of extracellular matrix-degrading invadosomal structures by synovial cells from collagen-induced arthritis (CIA) rats and RA patients. Mechanistically, Snail induces extracellular matrix degradation in synovial cells by repressing PTEN, resulting in increased phosphorylation of platelet-derived growth factor receptor and activation of the phosphatidylinositol 3-kinase/AKT pathway. Of significance, Snail is overexpressed in synovial cells and tissues of CIA rats and RA patients, whereas knockdown of Snail in CIA joints prevents cartilage invasion and joint damage. Furthermore, Snail expression is associated with an epithelial-mesenchymal transition gene signature characteristic of transglutaminase 2/transforming growth factor-β activation. Transforming growth factor-β and transglutaminase 2 stimulate Snail-dependent invadosome formation in rat and human synoviocytes. Our results identify the Snail-PTEN platelet-derived growth factor receptor/phosphatidylinositol 3-kinase axis as a novel regulator of the prodestructive invadosome-forming phenotype of synovial cells. New therapies for RA target inflammation, and are only partly effective in preventing joint damage. Blocking Snail and/or its associated gene expression program may provide an additional tool to improve the efficacy of treatments to prevent joint destruction.
Collapse
Affiliation(s)
- Annie Lauzier
- Immunology Division, Department of Pediatrics, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Roxane R Lavoie
- Immunology Division, Department of Pediatrics, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Martine Charbonneau
- Immunology Division, Department of Pediatrics, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Béatrice Gouin-Boisvert
- Immunology Division, Department of Pediatrics, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Kelly Harper
- Immunology Division, Department of Pediatrics, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Claire M Dubois
- Immunology Division, Department of Pediatrics, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada.
| |
Collapse
|
43
|
Winsz-Szczotka K, Komosińska-Vassev K, Kuźnik-Trocha K, Siwiec A, Żegleń B, Olczyk K. Circulating keratan sulfate as a marker of metabolic changes of cartilage proteoglycan in juvenile idiopathic arthritis; influence of growth factors as well as proteolytic and prooxidative agents on aggrecan alterations. Clin Chem Lab Med 2015; 53:291-7. [PMID: 25153398 DOI: 10.1515/cclm-2014-0441] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Accepted: 07/04/2014] [Indexed: 11/15/2022]
Abstract
BACKGROUND The aim of this study was to evaluate the plasma keratan sulfate (KS) level as a potential marker of joint damage in children with juvenile idiopathic arthritis (JIA). The influence of growth factors as well as proteolytic and prooxidative agents on aggrecan alterations were evaluated in this study. METHODS Plasma levels of KS, transforming growth factor β1 (TGF-β1), platelet-derived growth factor BB (PDGF-BB), a disintegrin and metalloproteinase with thrombospondin motifs 4 and 5 (ADAMTS-4 and ADAMTS-5), and thiol groups (TG) were quantified in samples obtained from 30 healthy subjects and 30 patients with JIA before and after treatment. RESULTS Increased (p<0.01) plasma KS was observed in JIA patients before treatment. Therapy resulted in a decrease in KS level. However, plasma KS level remained higher (p<0.05) than in controls. Increased levels of TGF-β1 (p<0.01) and PDGF-BB (p<0.05) in untreated JIA patients were recorded. Clinical improvement was accompanied by significant decrease in TGF-β1 and PDGF-BB, compared with a pretreatment condition and a control group. The concentrations of proteinases were characterized by different trends of alterations. When the ADAMTS-4 level increased (p<0.01) in the blood of untreated patients, the concentration of ADAMTS-5 was found to be reduced (p<0.0001), compared with controls. JIA treatment resulted in the normalization of ADAMTS-4 level. Plasma TG concentration was decreased only in untreated patients (p<0.05). We have revealed a significant correlation between plasma KS level and ADAMTS-4, TGF-β1, TG, C-reactive protein, and erythrocyte sedimentation rate levels. CONCLUSIONS Plasma KS level in JIA patients, reflecting the aggrecan structure, indicates that treatment that modifies inflammation simultaneously does not contribute to total regeneration of articular matrix components and signalizes the need for further treatment.
Collapse
|
44
|
Galimberti S, Fontanelli G, Barsotti S, Ricci F, Guerrini F, Baratè C. Increased values of the circulating PDGFβ sustains the “withdrawal syndrome” after tyrosine kinase inhibitor discontinuation in patients affected by chronic myeloid leukemia. Blood Cells Mol Dis 2015; 55:211-2. [DOI: 10.1016/j.bcmd.2015.06.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Accepted: 06/22/2015] [Indexed: 10/23/2022]
|
45
|
Kuzmina LA, Petinati NA, Shipounova IN, Sats NV, Bigildeev AE, Zezina EA, Popova MD, Drize NJ, Parovichnikova EN, Savchenko VG. Analysis of multipotent mesenchymal stromal cells used for acute graft-versus-host disease prophylaxis. Eur J Haematol 2015; 96:425-34. [PMID: 26115424 DOI: 10.1111/ejh.12613] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/22/2015] [Indexed: 12/15/2022]
Abstract
BACKGROUND Multipotent mesenchymal stromal cells (MSCs) are used for prophylaxis of acute graft-versus-host disease (aGvHD) after allogeneic hematopoietic cell transplantation (allo-HCT). Not all samples of MSC are efficient for aGvHD prevention. The suitability of MSCs for aGvHD prophylaxis was studied. METHODS MSCs were derived from the bone marrow (BM) of HCT donor and cultivated for no more than three passages. The characteristics of donor BM samples including colony-forming unit fibroblast (CFU-F) concentration, growth parameters of MSCs, and the relative expression levels (REL) of different genes were analyzed. MSCs were injected intravenously precisely at the moment of blood cell reconstitution. RESULTS MSCs infusion induced a significant threefold decrease in aGvHD development and improved overall survival compared with the standard prophylaxis group. In ineffective MSC samples (9.4%), a significant decrease in total cell production and the REL of CSF1, FGFR1, and PDGFRB was observed. In all studied BM samples, the cumulative MSC production and CFU-F concentrations decreased with age. The expression levels of FGFR2, PPARG, and VEGF differed by age. CONCLUSIONS A universal single indicator for the prediction of MSC eligibility for aGvHD prophylaxis was not identified. A multiparameter mathematical model for selecting MSC samples effective for the prevention of aGvHD was proposed.
Collapse
Affiliation(s)
- Larisa A Kuzmina
- Federal Government Budget Institution National Research Center for Hematology, Ministry of Health, Moscow, Russia
| | - Nataliya A Petinati
- Federal Government Budget Institution National Research Center for Hematology, Ministry of Health, Moscow, Russia
| | - Irina N Shipounova
- Federal Government Budget Institution National Research Center for Hematology, Ministry of Health, Moscow, Russia
| | - Natalia V Sats
- Federal Government Budget Institution National Research Center for Hematology, Ministry of Health, Moscow, Russia
| | - Alexey E Bigildeev
- Federal Government Budget Institution National Research Center for Hematology, Ministry of Health, Moscow, Russia
| | - Ekaterina A Zezina
- Department of Molecular Immunology, Faculty of Biology, Moscow State University, Moscow, Russia
| | - Maria D Popova
- Department of Molecular Immunology, Faculty of Biology, Moscow State University, Moscow, Russia
| | - Nina J Drize
- Federal Government Budget Institution National Research Center for Hematology, Ministry of Health, Moscow, Russia
| | - Elena N Parovichnikova
- Federal Government Budget Institution National Research Center for Hematology, Ministry of Health, Moscow, Russia
| | - Valery G Savchenko
- Federal Government Budget Institution National Research Center for Hematology, Ministry of Health, Moscow, Russia
| |
Collapse
|
46
|
Suárez-Causado A, Caballero-Díaz D, Bertrán E, Roncero C, Addante A, García-Álvaro M, Fernández M, Herrera B, Porras A, Fabregat I, Sánchez A. HGF/c-Met signaling promotes liver progenitor cell migration and invasion by an epithelial-mesenchymal transition-independent, phosphatidyl inositol-3 kinase-dependent pathway in an in vitro model. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1853:2453-63. [PMID: 26001768 DOI: 10.1016/j.bbamcr.2015.05.017] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Revised: 05/08/2015] [Accepted: 05/13/2015] [Indexed: 12/11/2022]
Abstract
Oval cells constitute an interesting hepatic cell population. They contribute to sustain liver regeneration during chronic liver damage, but in doing this they can be target of malignant conversion and become tumor-initiating cells and drive hepatocarcinogenesis. The molecular mechanisms beneath either their pro-regenerative or pro-tumorigenic potential are still poorly understood. In this study, we have investigated the role of the HGF/c-Met pathway in regulation of oval cell migratory and invasive properties. Our results show that HGF induces c-Met-dependent oval cell migration both in normal culture conditions and after in vitro wounding. HGF-triggered migration involves F-actin cytoskeleton reorganization, which is also evidenced by activation of Rac1. Furthermore, HGF causes ZO-1 translocation from cell-cell contact sites to cytoplasm and its concomitant activation by phosphorylation. However, no loss of expression of cell-cell adhesion proteins, including E-cadherin, ZO-1 and Occludin-1, is observed. Additionally, migration does not lead to cell dispersal but to a characteristic organized pattern in rows, in turn associated with Golgi compaction, providing strong evidence of a morphogenic collective migration. Besides migration, HGF increases oval cell invasion through extracellular matrix, a process that requires PI3K activation and is at least partly mediated by expression and activation of metalloproteases. Altogether, our findings provide novel insights into the cellular and molecular mechanisms mediating the essential role of HGF/c-Met signaling during oval cell-mediated mouse liver regeneration.
Collapse
Affiliation(s)
- A Suárez-Causado
- Dep. Bioquímica y Biología Molecular II, Facultad de Farmacia, Universidad Complutense, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid, Spain.
| | - D Caballero-Díaz
- Dep. Bioquímica y Biología Molecular II, Facultad de Farmacia, Universidad Complutense, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid, Spain.
| | - E Bertrán
- Laboratori d'Oncologia Molecular, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Universitat de Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain.
| | - C Roncero
- Dep. Bioquímica y Biología Molecular II, Facultad de Farmacia, Universidad Complutense, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid, Spain.
| | - A Addante
- Dep. Bioquímica y Biología Molecular II, Facultad de Farmacia, Universidad Complutense, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid, Spain.
| | - M García-Álvaro
- Dep. Bioquímica y Biología Molecular II, Facultad de Farmacia, Universidad Complutense, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid, Spain.
| | - M Fernández
- Dep. Bioquímica y Biología Molecular II, Facultad de Farmacia, Universidad Complutense, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid, Spain.
| | - B Herrera
- Dep. Bioquímica y Biología Molecular II, Facultad de Farmacia, Universidad Complutense, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid, Spain.
| | - A Porras
- Dep. Bioquímica y Biología Molecular II, Facultad de Farmacia, Universidad Complutense, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid, Spain.
| | - I Fabregat
- Laboratori d'Oncologia Molecular, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Universitat de Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain; Departament de Ciències Fisiològiques II, Universitat de Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain.
| | - A Sánchez
- Dep. Bioquímica y Biología Molecular II, Facultad de Farmacia, Universidad Complutense, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid, Spain.
| |
Collapse
|
47
|
Ekwall AKH, Whitaker JW, Hammaker D, Bugbee WD, Wang W, Firestein GS. The Rheumatoid Arthritis Risk Gene LBH Regulates Growth in Fibroblast-like Synoviocytes. Arthritis Rheumatol 2015; 67:1193-202. [PMID: 25707478 PMCID: PMC4490933 DOI: 10.1002/art.39060] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Accepted: 01/29/2015] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Fibroblast-like synoviocytes (FLS) are key players in the synovial pathology of rheumatoid arthritis (RA). Currently, there is no treatment that specifically targets these aggressive cells. By combining 3 different "omics" data sets, i.e., 1) risk genes in RA, 2) differentially expressed genes, and 3) differential DNA methylation in RA versus osteoarthritis (OA) FLS, we identified LBH (limb bud and heart development) as a candidate gene in RA. The present study was undertaken to define the role of this gene in FLS. METHODS Synovial tissue specimens from RA and OA patients were collected at the time of joint replacement surgery. LBH expression was silenced using small interfering RNA or overexpressed using an LBH expression vector in primary FLS. Gene expression profiles were determined by microarray and assessed using Ingenuity Pathway Analysis. Effects of modified LBH expression were investigated in functional assays. RESULTS LBH was expressed in the synovial lining layer in patients with RA. Transforming growth factor β1 significantly increased LBH expression in primary FLS, and platelet-derived growth factor BB decreased it. Pathway analysis of the transcriptome of LBH-deficient FLS compared to control FLS identified "cellular growth and proliferation" as the most significantly enriched pathway. In growth assays, LBH deficiency increased FLS proliferation. Conversely, LBH overexpression significantly inhibited cell growth. Cell cycle analysis demonstrated a marked increase in cells entering the cell cycle in LBH-deficient FLS compared to controls. LBH did not alter apoptosis. CONCLUSION LBH is a candidate gene for synovial pathology in RA. It is regulated by growth factors and modulates cell growth in primary FLS. Our data suggest a novel mechanism for synovial intimal hyperplasia and joint damage in RA.
Collapse
Affiliation(s)
- Anna-Karin H. Ekwall
- Anna-Karin H. Ekwall, MD, MSc, PhD (current address: University of Gothenburg, Gothenburg, Sweden), John W. Whitaker, PhD (current address: Discovery Science, Janssen Pharmaceuticals, San Diego, California), Deepa Hammaker, PhD, Wei Wang, PhD, Gary S. Firestein, MD: University of California at San Diego, La Jolla
| | - John W. Whitaker
- Anna-Karin H. Ekwall, MD, MSc, PhD (current address: University of Gothenburg, Gothenburg, Sweden), John W. Whitaker, PhD (current address: Discovery Science, Janssen Pharmaceuticals, San Diego, California), Deepa Hammaker, PhD, Wei Wang, PhD, Gary S. Firestein, MD: University of California at San Diego, La Jolla
| | - Deepa Hammaker
- Anna-Karin H. Ekwall, MD, MSc, PhD (current address: University of Gothenburg, Gothenburg, Sweden), John W. Whitaker, PhD (current address: Discovery Science, Janssen Pharmaceuticals, San Diego, California), Deepa Hammaker, PhD, Wei Wang, PhD, Gary S. Firestein, MD: University of California at San Diego, La Jolla
| | | | - Wei Wang
- Anna-Karin H. Ekwall, MD, MSc, PhD (current address: University of Gothenburg, Gothenburg, Sweden), John W. Whitaker, PhD (current address: Discovery Science, Janssen Pharmaceuticals, San Diego, California), Deepa Hammaker, PhD, Wei Wang, PhD, Gary S. Firestein, MD: University of California at San Diego, La Jolla
| | - Gary S. Firestein
- Anna-Karin H. Ekwall, MD, MSc, PhD (current address: University of Gothenburg, Gothenburg, Sweden), John W. Whitaker, PhD (current address: Discovery Science, Janssen Pharmaceuticals, San Diego, California), Deepa Hammaker, PhD, Wei Wang, PhD, Gary S. Firestein, MD: University of California at San Diego, La Jolla
| |
Collapse
|
48
|
Winsz-Szczotka K, Kuźnik-Trocha K, Komosińska-Vassev K, Wisowski G, Gruenpeter A, Lachór-Motyka I, Żegleń B, Lemski W, Olczyk K. Plasma and urinary glycosaminoglycans in the course of juvenile idiopathic arthritis. Biochem Biophys Res Commun 2015; 458:639-643. [PMID: 25684189 DOI: 10.1016/j.bbrc.2015.02.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Accepted: 02/04/2015] [Indexed: 11/16/2022]
Abstract
OBJECTIVES The aim of the study was to perform analyses of plasma and urinary glycosaminoglycan isolated from juvenile idiopathic arthritis (JIA). METHODS, RESULTS Chondroitin/dermatan sulfate (CS/DS), heparan sulfate/heparin (HS/H) and hyaluronic acid (HA) were evaluated in samples obtained from JIA patients before and after treatment. Electrophoretic analysis of GAGs identified the presence of CS, DS and HS/H in plasma of healthy subjects and JIA patients. CS were the predominant plasma GAGs constituent in all investigated subject. The plasma CS level in untreated patients was significantly decreased. Therapy resulted in an increase in this glycan level. However, plasma CS concentration still remained higher than in controls. Increased levels of DS and HA in untreated JIA patients were recorded. Anti-inflammatory treatment led to normalization of these parameters concentrations. Plasma and urinary concentrations of HS/H were similar in all groups of individuals. Urinary CS/DS and HA were decreased only in untreated patients. CONCLUSIONS The data presented indicate that changes in plasma and urinary glycosaminoglycan occur in the course of JIA. There are probably the expression of both local articular cartilage matrix and systemic changes in connective tissue remodeling.
Collapse
Affiliation(s)
- Katarzyna Winsz-Szczotka
- Department of Clinical Chemistry and Laboratory Diagnostics, Faculty of Pharmacy with the Division of Laboratory Medicine, Medical University of Silesia, Jedności 8, 41-200 Sosnowiec, Poland.
| | - Kornelia Kuźnik-Trocha
- Department of Clinical Chemistry and Laboratory Diagnostics, Faculty of Pharmacy with the Division of Laboratory Medicine, Medical University of Silesia, Jedności 8, 41-200 Sosnowiec, Poland
| | - Katarzyna Komosińska-Vassev
- Department of Clinical Chemistry and Laboratory Diagnostics, Faculty of Pharmacy with the Division of Laboratory Medicine, Medical University of Silesia, Jedności 8, 41-200 Sosnowiec, Poland
| | - Grzegorz Wisowski
- Department of Clinical Chemistry and Laboratory Diagnostics, Faculty of Pharmacy with the Division of Laboratory Medicine, Medical University of Silesia, Jedności 8, 41-200 Sosnowiec, Poland
| | - Anna Gruenpeter
- Department of Rheumatology, The Pediatric Center in Sosnowiec, Zapolskiej 3, 41-218 Sosnowiec, Poland
| | - Iwona Lachór-Motyka
- Department of Rheumatology, The Pediatric Center in Sosnowiec, Zapolskiej 3, 41-218 Sosnowiec, Poland
| | - Bogusław Żegleń
- Department of Clinical Chemistry and Laboratory Diagnostics, Faculty of Pharmacy with the Division of Laboratory Medicine, Medical University of Silesia, Jedności 8, 41-200 Sosnowiec, Poland
| | - Wojciech Lemski
- Department of Clinical Chemistry and Laboratory Diagnostics, Faculty of Pharmacy with the Division of Laboratory Medicine, Medical University of Silesia, Jedności 8, 41-200 Sosnowiec, Poland
| | - Krystyna Olczyk
- Department of Clinical Chemistry and Laboratory Diagnostics, Faculty of Pharmacy with the Division of Laboratory Medicine, Medical University of Silesia, Jedności 8, 41-200 Sosnowiec, Poland
| |
Collapse
|
49
|
Assirelli E, Filardo G, Mariani E, Kon E, Roffi A, Vaccaro F, Marcacci M, Facchini A, Pulsatelli L. Effect of two different preparations of platelet-rich plasma on synoviocytes. Knee Surg Sports Traumatol Arthrosc 2015; 23:2690-703. [PMID: 24942296 PMCID: PMC4541703 DOI: 10.1007/s00167-014-3113-3] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Accepted: 05/29/2014] [Indexed: 01/15/2023]
Abstract
PURPOSE To analyse the modifications induced by two different platelet-rich plasma (PRP) preparations on osteoarthritis (OA) synoviocytes, by documenting changes in gene expression of factors involved in joint physiopathology. METHODS OA synoviocytes were cultured for 7 days in medium with different concentrations of either P-PRP (a pure platelet concentrate without leucocytes but with a limited number of platelets), L-PRP (a higher platelet concentrate containing leucocytes) or platelet-poor plasma (PPP). Gene expression of interleukin (IL)-1beta, IL-6, IL-8/CXCL8, tumour necrosis factor alpha, IL-10, IL-4, IL-13, metalloproteinase-13, tissue inhibitor of metalloproteinase (TIMP)-1, (TIMP)-3, (TIMP)-4, vascular endothelial growth factor, transforming growth factor beta1, fibroblast growth factor (FGF)-2, hepatocyte growth factor (HGF), hyaluronic acid (HA) synthases (HAS)-1, (HAS)-2, and (HAS)-3 was analysed by RT-PCR. HA production was determined in culture supernatants by ELISA. RESULTS IL-1β, IL-8 and FGF-2 were significantly induced by L-PRP compared to both P-PRP and PPP; HGF was down-modulated by L-PRP versus both P-PRP and PPP, and an inverse dose-response influence was shown for all preparations. Expression level of TIMP-4 was lower in the presence of L-PRP compared with P-PRP. HA production and HAS gene expression did not seem to be modulated by PRP. CONCLUSIONS L-PRP is able to sustain the up-regulation of proinflammatory factors, (IL-1beta, IL-8 and FGF-2), together with a down-modulation of HGF and TIMP-4 expression, two factors that have been recognized as anti-catabolic mediators in cartilage, thus supporting the need to further optimize the PRP preparations to be applied in clinical practice.
Collapse
Affiliation(s)
- Elisa Assirelli
- Laboratory of Immunorheumatology and Tissue Regeneration/RAMSES, Rizzoli Orthopaedic Institute, Via di Barbiano 1/10, 40136, Bologna, Italy,
| | - Giuseppe Filardo
- Laboratory of Biomechanics and Technology Innovation/NABI, 2nd Orthopaedic and Traumatologic Clinic, Rizzoli Orthopaedic Institute, Via di Barbiano 1/10, Bologna, Italy
| | - Erminia Mariani
- Laboratory of Immunorheumatology and Tissue Regeneration/RAMSES, Rizzoli Orthopaedic Institute, Via di Barbiano 1/10, 40136 Bologna, Italy ,Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Elizaveta Kon
- Laboratory of Biomechanics and Technology Innovation/NABI, 2nd Orthopaedic and Traumatologic Clinic, Rizzoli Orthopaedic Institute, Via di Barbiano 1/10, Bologna, Italy
| | - Alice Roffi
- Laboratory of Biomechanics and Technology Innovation/NABI, 2nd Orthopaedic and Traumatologic Clinic, Rizzoli Orthopaedic Institute, Via di Barbiano 1/10, Bologna, Italy
| | - Franca Vaccaro
- Immunohematology and Transfusion Medicine Service, San Pietro Hospital, Via Cassia 600, Rome, Italy
| | - Maurilio Marcacci
- Laboratory of Biomechanics and Technology Innovation/NABI, 2nd Orthopaedic and Traumatologic Clinic, Rizzoli Orthopaedic Institute, Via di Barbiano 1/10, Bologna, Italy
| | - Andrea Facchini
- Laboratory of Immunorheumatology and Tissue Regeneration/RAMSES, Rizzoli Orthopaedic Institute, Via di Barbiano 1/10, 40136 Bologna, Italy ,Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Lia Pulsatelli
- Laboratory of Immunorheumatology and Tissue Regeneration/RAMSES, Rizzoli Orthopaedic Institute, Via di Barbiano 1/10, 40136 Bologna, Italy
| |
Collapse
|
50
|
Lee HP, Lin YY, Duh CY, Huang SY, Wang HM, Wu SF, Lin SC, Jean YH, Wen ZH. Lemnalol attenuates mast cell activation and osteoclast activity in a gouty arthritis model. ACTA ACUST UNITED AC 2014; 67:274-85. [PMID: 25557511 DOI: 10.1111/jphp.12331] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Accepted: 09/21/2014] [Indexed: 12/14/2022]
Abstract
OBJECTIVES In this study, we investigated the effects of a soft coral-derived anti-inflammatory compound, lemnalol, on mast cell (MC) function and osteoclast activity in rats with monosodium urate (MSU) crystal-induced gouty arthritis. METHODS In this study, we examined the therapeutic effects of lemnalol on intra-articular injection of MSU induces gouty arthritis with the measurement of ankle oedema. Toluidine blue staining were used to analyse the infiltration and the percentage degranulation MCs. Immunohistochemical analysis showed CD117, transforming growth factor beta 1 (TGF-β1), matrix metalloproteinase 9 (MMP-9), the osteoclast markers cathepsin K and tartrate-resistant acid phosphatase (TRAP) protein expression in ankle tissue. KEY FINDINGS We found that both infiltration and degranulation of MCs increased at 24 h after MSU injection in the ankle joint. Immunohistochemical analysis showed that MSU induced upregulation of TGF-β1, MMP-9, the osteoclast markers cathepsin K and TRAP in ankle tissues. Administration of lemnalol ameliorated MSU-induced TGF-β1, MMP-9, cathepsin K and TRAP protein expression. CONCLUSIONS Taken together, our results show that MSU-induced gouty arthritis is accompanied by osteoclast-related protein upregulation and that lemnalol treatment may be beneficial for the attenuation of MC infiltration and degranulation and for suppressing osteoclast activation in gouty arthritis.
Collapse
Affiliation(s)
- Hsin-Pai Lee
- Department of Marine Biotechnology and Resources, Asia-Pacific Ocean Research Center, National Sun Yat-sen University, Kaohsiung, Taiwan; Department of Orthopaedic Surgery, Ping-Tung Christian Hospital, Ping-Tung, Taiwan
| | | | | | | | | | | | | | | | | |
Collapse
|