Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 188
Filtrar
1.
Eur J Clin Invest ; : e14298, 2024 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-39105347

RESUMO

BACKGROUND: Hypocortisolemia is associated with increased expression of NR3C1 (glucocorticoid receptor, GR) in blood cells. As endogenous cortisol production is decreased in some RA patients, we tested the hypothesis that GR may be aberrantly expressed in rheumatoid synovium. METHODS: We defined the cellular pattern of NR3C1 synovial expression using human and mouse single-cell RNA-sequencing data. Bulk synovial RNA-sequencing data from early (n = 57) or established (n = 94) RA were compared to osteoarthritis (n = 22) and healthy synovium (n = 28). RESULTS: GR was expressed in all synovial cell types in both human and experimental arthritis. GR synovial expression, as well as 11ß-HSD1/11ß-HSD2 enzyme ratio, were higher in RA than healthy and osteoarthritic tissue, regardless of disease duration or treatment. Given that GR expression varied across samples, we searched for differences between RA patients with higher versus lower GR expression. Indeed, the synovial transcriptome of RA patients with high versus low GR expression (1st quartile, 30,517 ± 4876 vs. 4th quartile, 19,382 ± 2523 normalized counts) was enriched for proinflammatory gene-sets, including 'inflammatory response', 'IFN-γ response' and 'IL6/JAK/STAT3 signalling'. High synovial GR expression was also associated with increased JAK2 and PTPRK expression, denoting activation of the proinflammatory sublining fibroblasts. In contrast, low GR expression was associated with increased COMP and COL6A2 expression, denoting a resting synovial state. CONCLUSIONS: GR is overexpressed in the synovium of some RA patients in association with proinflammatory gene expression and activated sublining fibroblast status. Further studies should examine whether GR overexpression may act as a compensatory mechanism sensitizing synovial tissue to glucocorticoid action in RA.

2.
Artigo em Inglês | MEDLINE | ID: mdl-38960140

RESUMO

OBJECTIVE: Synovitis is a widely accepted sign of osteoarthritis (OA), characterised by tissue hyperplasia, where increased infiltration of immune cells and proliferation of resident fibroblasts adopt a pro-inflammatory phenotype, and increased the production of pro-inflammatory mediators that are capable of sensitising and activating sensory nociceptors, which innervate the joint tissues. As such, it is important to understand the cellular composition of synovium and their involvement in pain sensitisation to better inform the development of effective analgesics. METHODS: Studies investigating pain sensitisation in OA with a focus on immune cells and fibroblasts were identified using PubMed, Web of Science and SCOPUS. RESULTS: In this review, we comprehensively assess the evidence that cellular crosstalk between resident immune cells or synovial fibroblasts with joint nociceptors in inflamed OA synovium contributes to peripheral pain sensitisation. Moreover, we explore whether the elucidation of common mechanisms identified in similar joint conditions may inform the development of more effective analgesics specifically targeting OA joint pain. CONCLUSION: The concept of local environment and cellular crosstalk within the inflammatory synovium as a driver of nociceptive joint pain presents a compelling opportunity for future research and therapeutic advancements.

3.
J Pers Med ; 14(7)2024 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-39063955

RESUMO

Pulsed electromagnetic field stimulation (PEMF) is gaining more attention as a non-invasive arthritis treatment. In our study, immortalized synovial fibroblasts (K4IM) derived from a non-arthritic donor were exposed to MAGCELL® ARTHRO, a PEMF device, with 105 mT intensity, 8 Hz frequency, and 2 × 2.5 min sessions conducted thrice with a 1 h interval, to understand the underlying mechanism in regard to the complement system. Additionally, tumor necrosis factor (TNFα, 10 ng/mL) pre-treatment prior to PEMF stimulation, as well as 3-day versus 6-day stimulation, were compared. Gene expression of C4b binding protein-alpha and -beta (C4BPα, C4BPß), complement factor (CF)-H, CFI, CD55, CD59, Interleukin (IL-6) and TNFα was analyzed. Immunofluorescence staining of CD55, CD59, and Ki67 was conducted. Results showed the absence of C4BPα gene expression, but C4BPß was present. One and three days of PEMF stimulation caused no significant changes. However, after six days, there was a significant increase in CD55, CFH, and CD59 gene expression, indicating cytoprotective effects. Conversely, IL-6 gene expression increased after six days of stimulation and even after a single session in TNFα pre-stimulated cells, indicating a pro-inflammatory effect. PEMF's ambivalent, i.e., enhancing complement regulatory proteins and pro-inflammatory cytokines, highlights its complexity at the molecular level.

4.
Intractable Rare Dis Res ; 13(2): 110-116, 2024 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-38836181

RESUMO

Rheumatoid arthritis (RA) is an autoimmune disease with complex etiology, and its pathological mechanism remains unclear. Our aim was to explore the effect of protein succinylation on RA by silencing Sirt5, sequencing succinylated proteins, and analyzing the sequencing results to identify potential biomarkers. We wanted to gain a clearer understanding of RA pathogenesis, quantitative assessment of succinylated proteins in Fibroblast-like synoviocytes (FLS) from RA patients using liquid chromatography- tandem mass spectrometry and enrichment analysis investigated using Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG). A total of 679 proteins and 2,471 lysine succinylation sites were found in RA patients, and 436 differentially expressed proteins and 1,548 differentially expressed succinylation sites were identified. Among them, 48 succinylation sites were upregulated in 38 proteins and 144 succinylation sites were downregulated in 82 proteins. Bioinformatics showed that succinylated proteins were significantly enriched in amino and fatty acid metabolisms. Results indicated that Sirt5 can affect various biological processes involved in RA FLSs, and succinylation caused by silencing Sirt5 plays a major role in RA progression. This study provides further understanding of RA pathogenesis and may facilitate searching for potential RA biomarkers.

5.
Pharmaceuticals (Basel) ; 17(6)2024 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-38931405

RESUMO

Calcium pyrophosphate dehydrate (CPPD) crystals are found in the synovial fluid of patients with articular chondrocalcinosis or sometimes with osteoarthritis. In inflammatory conditions, the synovial membrane (SM) is subjected to transient hypoxia, especially during movement. CPPD formation is supported by an increase in extracellular inorganic pyrophosphate (ePPi) levels, which are mainly controlled by the transporter Ank and ectonucleotide pyrophosphatase/phosphodiesterase 1 (ENPP1). We demonstrated previously that transforming growth factor (TGF)-ß1 increased ePPi production by inducing Ank and Enpp1 expression in chondrocytes. As the TGF-ß1 level raises in synovial fluid under hypoxic conditions, we investigated whether hypoxia may transform SM as a major source of ePPi production. Synovial fibroblasts and SM explants were exposed to 10 ng/mL of TGF-ß1 in normoxic or hypoxic (5% O2) culture conditions. Ank and Enpp1 expression were assessed by quantitative PCR, Western blot and immunohistochemistry. ePPi was quantified in culture supernatants. RNA silencing was used to define the respective roles of Ank and Enpp1 in TGF-ß1-induced ePPi generation. The molecular mechanisms involved in hypoxia were investigated using an Ank promoter reporter plasmid for transactivation studies, as well as gene overexpression and RNA silencing, the respective role of hypoxia-induced factor (HIF)-1 and HIF-2. Our results showed that TGF-ß1 increased Ank, Enpp1, and therefore ePPi production in synovial fibroblasts and SM explants. Ank was the major contributor in ePPi production compared to ENPP1. Hypoxia increased ePPi levels on its own and enhanced the stimulating effect of TGF-ß1. Hypoxic conditions enhanced Ank promoter transactivation in an HIF-1-dependent/HIF-2-independent fashion. We demonstrated that under hypoxia, SM is an important contributor to ePPi production in the joint through the induction of Enpp1 and Ank. These findings are of interest as a rationale for the beneficial effect of anti-inflammatory drugs on SM in crystal depositions.

6.
Inflamm Regen ; 44(1): 29, 2024 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-38831367

RESUMO

BACKGROUND: Current strategies that target cytokines (e.g., tumor necrosis factor (TNF)-α), or signaling molecules (e.g., Janus kinase (JAK)) have advanced the management for allergies and autoimmune diseases. Nevertheless, the molecular mechanism that underpins its clinical efficacy have largely remained elusive, especially in the local tissue environment. Here, we aimed to identify the genetic, epigenetic, and immunological targets of JAK inhibitors (JAKis), focusing on their effects on synovial fibroblasts (SFs), the major local effectors associated with destructive joint inflammation in rheumatoid arthritis (RA). METHODS: SFs were activated by cytokines related to inflammation in RA, and were treated with three types of JAKis or a TNF-α inhibitor (TNFi). Dynamic changes in transcriptome and chromatin accessibility were profiled across samples to identify drug targets. Furthermore, the putative targets were validated using luciferase assays and clustered regularly interspaced short palindromic repeat (CRISPR)-based genome editing. RESULTS: We found that both JAKis and the TNFi targeted the inflammatory module including IL6. Conversely, specific gene signatures that were preferentially inhibited by either of the drug classes were identified. Strikingly, RA risk enhancers for CD40 and TRAF1 were distinctively regulated by JAKis and the TNFi. We performed luciferase assays and CRISPR-based genome editing, and successfully fine-mapped the single causal variants in these loci, rs6074022-CD40 and rs7021049-TRAF1. CONCLUSIONS: JAKis and the TNFi had a direct impact on different RA risk enhancers, and we identified nucleotide-resolution targets for both drugs. Distinctive targets of clinically effective drugs could be useful for tailoring the application of these drugs and future design of more efficient treatment strategies.

7.
J Nanobiotechnology ; 22(1): 188, 2024 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-38632657

RESUMO

Rheumatoid arthritis (RA) is a progressive autoimmune disease accompanied by joint swelling, cartilage erosion and bone damage. Drug therapy for RA has been restricted due to poor therapeutic effect, recurrence and adverse effects. Macrophages and synovial fibroblasts both play important roles in the pathology of RA. Macrophages secrete large amount of pro-inflammatory cytokines, while synovial fibroblasts are tightly correlated with hypoxia synovium microenvironment, cytokine release, recruitment of pro-inflammatory cells, bone and cartilage erosion. Therefore, in this timely research, an injectable and pH-sensitive peptide hydrogel loading methotrexate (MTX) and bismuthene nanosheet/polyethyleneimine (BiNS/PEI) has been developed to reduce the activity of macrophages and eliminate over-proliferated synovial fibroblasts simultaneously. MTX can reduce the cytokine secretion of macrophages/anti-apoptosis property of synovial fibroblasts and BiNS/PEI can eliminate synovial fibroblasts via photodynamic therapy (PDT) and photothermal therapy (PTT) routes. The hydrogel was injected into the acidic inflammatory synovium for precise targeting and served as a drug reservoir for pH responsive and sustained drug release, while improving the bioavailability and reducing the toxicity of MTX. Excellent therapeutic efficacy has been achieved in both in vivo and in vitro studies, and this unique drug delivery system provides a new and robust strategy to eliminate synovial fibroblasts and modulate immune system for RA treatment in clinical.


Assuntos
Artrite Reumatoide , Hidrogéis , Humanos , Hidrogéis/farmacologia , Membrana Sinovial/patologia , Macrófagos , Metotrexato/farmacologia , Citocinas , Fibroblastos
8.
Environ Toxicol ; 39(6): 3283-3291, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38380842

RESUMO

Rheumatoid arthritis (RA) is a well-known autoimmune disorder related with joint pain, joint swelling, cartilage and bone degradation as well as deformity. Fibroblast growth factor 23 (FGF23) is an endocrine factor of the FGF family primarily produced by osteocytes and osteoblasts, involves an essential effect in pathogenesis of RA. IL-1ß is a vital proinflammatory factor in the development of RA. However, the role of FGF23 on IL-1ß synthesis in RA has not been fully explored. Our analysis of database revealed higher levels of FGF23 and IL-1ß in RA samples compared with healthy controls. High-throughput screening demonstrated that IL-1ß is a potential candidate factor after FGF23 treatment in RA synovial fibroblasts (RASFs). FGF23 concentration dependently promotes IL-1ß synthesis in RASFs. FGF23 enhances IL-1ß expression by activating the PI3K, Akt, and NF-κB pathways. Our findings support the notion that FGF23 is a promising target in the remedy of RA.


Assuntos
Artrite Reumatoide , Fator de Crescimento de Fibroblastos 23 , Fibroblastos , Interleucina-1beta , Transdução de Sinais , Feminino , Humanos , Masculino , Artrite Reumatoide/metabolismo , Células Cultivadas , Fatores de Crescimento de Fibroblastos/metabolismo , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Interleucina-1beta/metabolismo , NF-kappa B/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos , Membrana Sinovial/efeitos dos fármacos , Membrana Sinovial/metabolismo
9.
Biochem Pharmacol ; 221: 116037, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38301965

RESUMO

Rheumatoid arthritis (RA) is a well-known autoimmune disorder associated with joint pain, joint swelling, cartilage and bone degradation as well as deformity. The chemokine (C-X-C motif) ligand 13 (CXCL13) plays a crucial role in multiple cellular pathogenesis processes, including RA. TNF-α is a vital proinflammatory factor in the progression of RA. However, the role of CXCL13 in TNF-α production in RA has not been fully explored. Our analysis of both database and clinical samples revealed higher levels of CXCL13 and TNF-α in RA samples compared to healthy controls. CXCL13 concentration-dependently induces TNF-α synthesis in RA synovial fibroblasts. CXCL13 enhances TNF-α expression by interacting with the CXCR5 receptor, activating the ERK/p38 pathways, and inhibiting miR-330-3p generation. Importantly, treatment with CXCL13 shRNA counteracted the upregulation of TNF-α production induced by collagen-induced arthritis. Our findings support the notion that CXCL13 is a promising target in the treatment of RA.


Assuntos
Artrite Experimental , Artrite Reumatoide , Doenças Autoimunes , MicroRNAs , Animais , Fator de Necrose Tumoral alfa/farmacologia , Artrite Reumatoide/genética , MicroRNAs/genética
10.
Int Immunopharmacol ; 128: 111487, 2024 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-38183911

RESUMO

Rheumatoid arthritis (RA) is an autoimmune disease that affects joints, causing inflammation, synovitis, and erosion of cartilage and bone. Periplogenin is an active ingredient in the anti-rheumatic and anti-inflammatory herb, cortex periplocae. We conducted a study using a CIA model and an in vitro model of fibroblast-like synoviocytes (FLS) induced by Tumor Necrosis Factor-alpha (TNF-α) stimulation. We evaluated cell activity, proliferation, and migration using the CCK8 test, EDU kit, and transwell assays, as well as network pharmacokinetic analysis of periplogenin targets and RA-related effects. Furthermore, we measured inflammatory factors and matrix metalloproteinases (MMPs) expression using ELISA and qRT-PCR assays. We also evaluated joint destruction using HE and Safranin O-Fast Green Staining and examined the changes in the JAK2/3-STAT3 pathway using western blot. The results indicated that periplogenin can effectively inhibit the secretion of inflammatory factors, suppress the JAK2/3-STAT3 pathway, and impede the proliferation and migration of RA FLS. Thus, periplogenin alleviated the Synovial inflammatory infiltration of RA.


Assuntos
Artrite Experimental , Artrite Reumatoide , Digitoxigenina/análogos & derivados , Sinoviócitos , Humanos , Animais , Inflamação/metabolismo , Proliferação de Células , Fibroblastos , Membrana Sinovial/patologia , Células Cultivadas , Janus Quinase 2/metabolismo , Fator de Transcrição STAT3/metabolismo
11.
Connect Tissue Res ; 65(1): 73-87, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38090785

RESUMO

PURPOSE: We aimed to investigate the heterogeneity of synovial fibroblasts and their potential to undergo cell state transitions at the resolution of single cells. MATERIALS AND METHODS: We employed the single-cell RNA sequencing (scRNA-seq) approach to comprehensively map the cellular landscape of the shoulder synovium in individuals with chronic rotator cuff tears (RCTs) and acute proximal humerus fractures (PHFs). Utilizing unbiased clustering analysis, we successfully identified distinct subpopulations of fibroblasts within the synovial environment. We utilized Monocle 3 to delineate the trajectory of synovial fibroblast state transition. And we used CellPhone DB v2.1.0 to predict cell-cell communication patterns within the synovial microenvironment. RESULTS: We identified eight main cell clusters in the shoulder synovium. Unbiased clustering analysis identified four synovial fibroblast subpopulations, with diverse biological functions associated with protein secretion, ECM remodeling, inflammation regulation and cell division. Lining, mesenchymal, pro-inflammatory and proliferative fibroblasts subsets were identified. Combining the results from StemID and characteristic gene features, mesenchymal fibroblasts exhibited characteristics of fibroblast progenitor cells. The trajectory of synovial fibroblast state transition showed a transition from mesenchymal to pro-inflammatory and lining phenotypes. In addition, the cross talk between fibroblast subclusters increased in degenerative shoulder diseases compared to acute trauma. CONCLUSION: We successfully generated the scRNA-seq transcriptomic atlas of the shoulder synovium, which provides a comprehensive understanding of the heterogeneity of synovial fibroblasts, their potential to undergo state transitions, and their intercellular communication in the context of chronic degenerative and acute traumatic shoulder diseases.


Assuntos
Artrite Reumatoide , Lesões do Manguito Rotador , Humanos , Membrana Sinovial/metabolismo , Comunicação Celular , Fibroblastos/metabolismo , Perfilação da Expressão Gênica
12.
Int Immunopharmacol ; 127: 111365, 2024 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-38104370

RESUMO

OBJECTIVE: Rheumatoid arthritis (RA) is an autoimmune disease characterized by chronic joint inflammation, with synovial fibroblasts (SFs) playing a pivotal role in its pathogenesis. Dysregulation of microRNA (miRNA) expression in SFs contributes to RA development. Exosomes (Exos) have emerged as effective carriers for therapeutic molecules, facilitating miRNA transfer between cells. This study explores the therapeutic potential of Exos derived from human umbilical cord mesenchymal stem cells (hUCMSCs), loaded with miR-451a, to modulate ATF2 expression, aiming to address RA in both in vivo and in vitro settings. METHODS: In this study, hUCMSC and RA SFs were isolated and identified, and hUCMSC-Exos were extracted and characterized. The influence of hUCMSC-Exos on RA SFs was detected. And hUCMSC-Exos targeting RA SFs was traced. HUCMSCKD-AGO2 was prepared by knocking down AGO2 in hUCMSC. HUCMSCKD-AGO2-Exos was extracted and characterized,and their influence on RA SFs was detected. The miRNA profiles before and after hUCMSC-Exos intervention in RA SFs were mapped to identify differential miRNAs. RT-qPCR was used to verify the differential miRNAs, with hsa-miR-451a finally selected as the target gene. The effect of miR-451a on SFs was detected. The latent binding of miR-451a to activating transcription factor 2 (ATF2) was analyzed. The effect of hUCMSC-ExosmiR-451a on SFs was detected, and the expression of miR-451a and ATF2 was measured by RT-PCR. In vivo, hUCMSC-ExosmiR-451a was injected into the ankle joint of CIA rats, and arthritis index, joint imaging and synovial pathology were assessed. The expression of miR-451a and ATF2 in synovial tissue was detected. Finally, the safety of hUCMSC-ExosmiR-451a in CIA rats was evaluated. RESULTS: This study revealed that hUCMSC-Exos can inhibit RA SFs proliferation, migration and invasion through miRNAs. High throughput sequencing detected 13 miRNAs that could be transmitted from hUCMSCs to RA SFs via hUCMSC-Exos. miR-451a inhibited RA SFs proliferation, migration and invasion by regulating ATF2. hUCMSC-Exos loaded with miR-451a targeted ATF2 to inhibit RA SFs proliferation, migration and invasion, and improve joint inflammation and imaging findings in CIA rats. CONCLUSIONS: This study demonstrates that miR-451a carried by hUCMSC-Exos can play a role in inhibiting RA SFs biological traits and improving arthritis in CIA rats by inhibiting ATF2. The findings suggest a promising treatment for RA and provide insights into the mechanism of action of hUCMSC-Exos in RA. Future research directions will continue to explore the potential in this field.


Assuntos
Artrite Reumatoide , Exossomos , Células-Tronco Mesenquimais , MicroRNAs , Animais , Humanos , Ratos , Fator 2 Ativador da Transcrição/genética , Fator 2 Ativador da Transcrição/metabolismo , Artrite Reumatoide/genética , Artrite Reumatoide/terapia , Artrite Reumatoide/metabolismo , Exossomos/genética , Exossomos/metabolismo , Inflamação/metabolismo , Células-Tronco Mesenquimais/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Cordão Umbilical
13.
Arthritis Res Ther ; 25(1): 238, 2023 12 07.
Artigo em Inglês | MEDLINE | ID: mdl-38062469

RESUMO

BACKGROUNDS: Rheumatoid arthritis (RA) is a chronic inflammatory autoimmune disease characterized by synovial hyperplasia. Maintaining a balance between the proliferation and apoptosis of rheumatoid arthritis synovial fibroblasts (RASFs) is crucial for preventing the erosion of bone and cartilage and, ultimately, mitigating the progression of RA. We found that the lncRNA LEF1-AS1 was expressed at low levels in the RASFs and inhibited their abnormal proliferation by targeting PIK3R2 protein and regulating the PI3K/AKT signal pathway through its interaction with miR-30-5p. In this study, we fabricated a nano-drug delivery system for LEF1-AS1 using Zn-Adenine nanoparticles (NPs) as a novel therapeutic strategy against RA. METHODS: The expression levels of LEF1-AS1, miR-30-5p, PIK3R2, p-PI3K, and p-AKT were detected in the primary RASFs and a human fibroblast-like synovial cell line (HFLS). Zn-Adenine nanoparticles (NPs) were functionalized with anti-CD305 antibody to construct (Zn-Adenine)@Ab. These NPs were then loaded with LEF1-AS1 to form (Zn-Adenine)@Ab@lncRNA LEF1-AS1. Finally, the (Zn-Adenine)@Ab@lncRNA LEF1-AS1 NPs were locally injected into a rat model with collagen-induced arthritis (CIA). The arthritic injuries in each group were evaluated by HE staining and other methods. RESULTS: LEF1-AS1 was expressed at low levels in the primary RASFs. High expression levels of LEF1-AS1 were detected in the HFLS cells, which corresponded to a significant downregulation of miR-30-5p. In addition, the expression level of PIK3R2 was significantly increased, and that of p-PI3K and p-AKT were significantly downregulated in these cells. The (Zn-Adenine)@Ab@lncRNA LEF1-AS1 NPs significantly inhibited the proliferation of RASFs and decreased the production of inflammatory cytokines (IL-1ß, IL-6, TNF-α). Intra-articular injection (IAI) of (Zn-Adenine)@Ab@lncRNA LEF1-AS1 NPs significantly alleviated cartilage destruction and joint injury in the CIA-modeled rats. CONCLUSIONS: LEF1-AS1 interacts with miR-30-5p to inhibit the abnormal proliferation of RASFs by regulating the PI3K/AKT signal pathway. The (Zn-Adenine)@Ab NPs achieved targeted delivery of the loaded LEF1-AS1 into the RASFs, which improved the cellular internalization rate and therapeutic effects. Thus, LEF1-AS1 is a potential target for the treatment of RA.


Assuntos
Artrite Experimental , Artrite Reumatoide , MicroRNAs , RNA Longo não Codificante , Humanos , Ratos , Animais , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , MicroRNAs/genética , Membrana Sinovial/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proliferação de Células/fisiologia , Artrite Reumatoide/tratamento farmacológico , Artrite Reumatoide/genética , Artrite Reumatoide/metabolismo , Anticorpos/metabolismo , Artrite Experimental/tratamento farmacológico , Artrite Experimental/genética , Artrite Experimental/metabolismo , Fibroblastos/metabolismo , Inflamação/metabolismo , Zinco/metabolismo , Fator 1 de Ligação ao Facilitador Linfoide/metabolismo
14.
Heliyon ; 9(11): e21412, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37920489

RESUMO

Our previous study has confirmed that miR338-3p/TRAP-1 axis was involved in regulation of hyperactivation in human synovial fibroblasts (HFLS) of patients with osteoarthritis (OA). Here, we aim to further investigate the underlying causes of the abnormal activation miR338-3p/TRAP-1 at the molecular level. Our results showed that the decrease of NF-E2-related factor 2(Nrf2) was the direct cause of downregulation of miR338-3p and upregulation of TRAP-1 protein expression in HFLS of OA patients. Furthermore, we also found that the phosphorylation and nuclear entry of Nrf2 protein were significantly reduced in HFLS of OA patients than that of normal individuals, and both of them were positively correlated with miR338-3p levels. Bioinformatics analysis, luciferase assay, and CHIP experiment together indicated that Nrf2 could positively regulate the transcription of miR338-3p by binding to the Transcription Factor Binding Sites (TFBS) on its promoter. It was confirmed by in vitro assays that oltipraz (agonists of Nrf2) treatment effectively inhibited the hyperactivation of HFLS induced by TGF-ß1, and the effects of oltipraz could be reversed by the exogenous TRAP-1. In short, our research has revealed for the first time that Nrf2/miR338-3p/TRAP-1 pathway was involved in hyperactivation of HFLS in OA patients, Nrf2 has the potential to be used as therapy and new drug target of OA.

15.
Stem Cells Transl Med ; 12(12): 849-862, 2023 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-37934808

RESUMO

Currently available therapies for rheumatoid arthritis (RA) are inadequate to alleviate the inflammation and reduce joint damage. While the immune-regulatory effect of human mesenchymal/stromal stem cells (MSCs) extracellular vesicles (EVs) has been tested in many inflammation-related diseases, little is known regarding their effect on patients with RA. Thus, we assessed the effect of human MSCs and MSC-EVs (from naïve or IFN-ß-primed MSCs) on CD4+ T cells from patients with RA. Moreover, we investigated the effect of MSC-EVs on RA patients-derived synovial fibroblasts (FLS). MSC-EVs were prepared using a PEG precipitation followed by ultracentrifugation-based protocol. Applied to RA CD4+ T cells, EVs from IFN-ß-primed MSCs, suppressed the expression of more key RA-associated cytokines (IL-4, GM-CSF IFN-γ, IL-2, TNF-α), and decreased CD4+ T-cell polyfunctionality than MSCs or EVs from naïve MSCs. MSCs mediated a slight decrease in the frequency of T-regulatory cells, while MSC-EVs rescued the frequency of T-regulatory cells. MSCs significantly inhibited CD4+ T-cell proliferation (P < .05), while no inhibition was observed in response to EV preparations. EVs from IFN-ß-primed MSCs inhibited (P < .01) RA FLS migration and downregulated (P < .05) RA FLS surface markers CD34 and HLA-DR. Collectively, we demonstrated the immune-modulatory function of MSCs and their derived EVs in RA CD4+ T cells, which could be further enhanced by priming MSCs with IFN-ß. Moreover, EVs from IFN-ß-primed MSCs more efficiently inhibit RA FLS migration, and expression of RA FLS-related surface markers, suggesting these EVs as a potent therapy for RA.


Assuntos
Artrite Reumatoide , Vesículas Extracelulares , Humanos , Vesículas Extracelulares/metabolismo , Artrite Reumatoide/terapia , Citocinas/metabolismo , Inflamação/metabolismo , Células-Tronco/metabolismo
16.
Arthritis Res Ther ; 25(1): 211, 2023 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-37885040

RESUMO

BACKGROUND: Rheumatoid arthritis is a chronic systemic autoimmune disease that involves transformation of the lining of synovial joints into an invasive and destructive tissue. Synovial fibroblasts become transformed, invading and destroying the bone and cartilage of the affected joint(s). Due to the significant role these cells play in the progression of the disease process, developing a therapeutic strategy to target and inhibit their invasive destructive nature could help patients who are afflicted with this debilitating disease. Gingival-derived mesenchymal stem cells are known to possess immunomodulatory properties and have been studied extensively as potential cell-based therapeutics for several autoimmune disorders. METHODS: A chimeric human/mouse model of synovitis was created by surgically implanting SCID mice with a piece of human articular cartilage surrounded by RASF. Mice were injected once with either GMSC or GMSCExo at 5-7 days post-implantation. Histology and IHC were used to assess RASF invasion of the cartilage. Flow cytometry was used to understand the homing ability of GMSC in vivo and the incidence of apoptosis of RASF in vitro. RESULTS: We demonstrate that both GMSC and GMSCExo are potent inhibitors of the deleterious effects of RASF. Both treatments were effective in inhibiting the invasive destructive properties of RASF as well as the potential for these cells to migrate to secondary locations and attack the cartilage. GMSC home to the site of the implant and induce programmed cell death of the RASF. CONCLUSIONS: Our results indicate that both GMSC and GMSCExo can block the pathological effects of RASF in this chimeric model of RA. A single dose of either GMSC or GMSCExo can inhibit the deleterious effects of RASF. These treatments can also block the invasive migration of the RASF, suggesting that they can inhibit the spread of RA to other joints. Because the gingival tissue is harvested with little difficulty, relatively small amounts of tissue are required to expand the cells, the simple in vitro expansion process, and the increasing technological advances in the production of therapeutic exosomes, we believe that GMSCExo are excellent candidates as a potential therapeutic for RA.


Assuntos
Artrite Reumatoide , Exossomos , Células-Tronco Mesenquimais , Humanos , Animais , Camundongos , Membrana Sinovial/metabolismo , Exossomos/metabolismo , Células Cultivadas , Camundongos SCID , Artrite Reumatoide/metabolismo , Células-Tronco Mesenquimais/metabolismo , Fibroblastos/metabolismo
17.
Endocrinology ; 164(12)2023 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-37864848

RESUMO

The close association between rheumatoid arthritis (RA), sex, reproductive state, and stress has long linked prolactin (PRL) to disease progression. PRL has both proinflammatory and anti-inflammatory outcomes in RA, but responsible mechanisms are not understood. Here, we show that PRL modifies in an opposite manner the proinflammatory actions of IL-1ß and TNF-α in mouse synovial fibroblasts in culture. Both IL-1ß and TNF-α upregulated the metabolic activity and the expression of proinflammatory factors (Il1b, Inos, and Il6) via the activation of the nuclear factor-κB (NF-κB) signaling pathway. However, IL-1ß increased and TNF-α decreased the levels of the long PRL receptor isoform in association with dual actions of PRL on synovial fibroblast inflammatory response. PRL reduced the proinflammatory effect and activation of NF-κB by IL-1ß but increased TNF-α-induced inflammation and NF-κB signaling. The double-faceted role of PRL against the 2 cytokines manifested also in vivo. IL-1ß or TNF-α with or without PRL were injected into the knee joints of healthy mice, and joint inflammation was monitored after 24 hours. IL-1ß and TNF-α increased the joint expression of proinflammatory factors and the infiltration of immune cells. PRL prevented the actions of IL-1ß but was either inactive or further increased the proinflammatory effect of TNF-α. We conclude that PRL exerts opposite actions on joint inflammation in males and females that depend on specific proinflammatory cytokines, the level of the PRL receptor, and the activation of NF-κB signaling. Dual actions of PRL may help balance joint inflammation in RA and provide insights for development of new treatments.


Assuntos
Artrite Reumatoide , Citocinas , Masculino , Feminino , Camundongos , Animais , Citocinas/metabolismo , NF-kappa B/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Fator de Necrose Tumoral alfa/metabolismo , Prolactina/farmacologia , Prolactina/metabolismo , Membrana Sinovial/metabolismo , Células Cultivadas , Artrite Reumatoide/metabolismo , Inflamação/metabolismo , Fibroblastos/metabolismo
18.
Arthritis Res Ther ; 25(1): 180, 2023 09 25.
Artigo em Inglês | MEDLINE | ID: mdl-37749630

RESUMO

BACKGROUND: Dimerization of the myeloid differentiation primary response 88 protein (MyD88) plays a pivotal role in the exacerbated response to innate immunity-dependent signaling in rheumatoid arthritis (RA). ST2825 is a highly specific inhibitor of MyD88 dimerization, previously shown to inhibit the pro-inflammatory gene expression in peripheral blood mononuclear cells from RA patients (RA PBMC). In this study, we elucidated the effect of disrupting MyD88 dimerization by ST2825 on the pathological properties of synovial fibroblasts from RA patients (RA SFs). METHODS: RA SFs were treated with varying concentrations of ST2825 in the presence or absence of bacterial lipopolysaccharides (LPS) to activate innate immunity-dependent TLR signaling. The DNA content of the RA SFs was quantified by imaging cytometry to investigate the effect of ST2825 on different phases of the cell cycle and apoptosis. RNA-seq was used to assess the global response of the RA SF toward ST2825. The invasiveness of RA SFs in Matrigel matrices was measured in organoid cultures. SFs from osteoarthritis (OA SFs) patients and healthy dermal fibroblasts were used as controls. RESULTS: ST2825 reduced the proliferation of SFs by arresting the cells in the G0/G1 phase of the cell cycle. In support of this finding, transcriptomic analysis by RNA-seq showed that ST2825 may have induced cell cycle arrest by primarily inhibiting the expression of critical cell cycle regulators Cyclin E2 and members of the E2F family transcription factors. Concurrently, ST2825 also downregulated the genes encoding for pain, inflammation, and joint catabolism mediators while upregulating the genes required for the translocation of nuclear proteins into the mitochondria and members of the mitochondrial respiratory complex 1. Finally, we demonstrated that ST2825 inhibited the invasiveness of RA SFs, by showing decreased migration of LPS-treated RA SFs in spheroid cultures. CONCLUSIONS: The pathological properties of the RA SFs, in terms of their aberrant proliferation, increased invasiveness, upregulation of pain and inflammation mediators, and disruption of mitochondrial homeostasis, were attenuated by ST2825 treatment. Taken together with the previously reported anti-inflammatory effects of ST2825 in RA PBMC, this study strongly suggests that targeting MyD88 dimerization could mitigate both systemic and synovial pathologies in a variety of inflammatory arthritic diseases.


Assuntos
Artrite Reumatoide , Osteoartrite , Humanos , Dimerização , Leucócitos Mononucleares , Fator 88 de Diferenciação Mieloide/genética , Lipopolissacarídeos , Artrite Reumatoide/tratamento farmacológico , Proteínas Adaptadoras de Transdução de Sinal , Fibroblastos , Osteoartrite/tratamento farmacológico
19.
Int Immunopharmacol ; 124(Pt B): 110909, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37722260

RESUMO

Rheumatoid arthritis (RA) is the most common autoimmune disease, affecting the joints of the hands and feet. Several chemokines and their receptors are crucial in RA pathogenesis through immune cell recruitment. C-X-C Motif Chemokine Ligand 1 (CXCL1), a chemokine for the recruitment of various immune cells, can be upregulated in patients with RA. However, the discussion on the role of CXCL1 in RA pathogenesis is insufficient. Here, we found that CXCL1 promoted cyclooxygenase-2 (COX-II) expression in a dose- and time-dependent manner in rheumatoid arthritis synovial fibroblasts (RASFs). CXCL1 overexpression in RASFs led to a significant increase in COX-II expression, while the transfection of RASFs with the shRNA plasmid resulted in a noticeable decrease in COX-II expression. Next, we delineated the molecular mechanism underlying CXCL1-promoted COX-II expression and noted that CXC chemokine receptor 2 (CXCR2), phospholipase C (PLC), and protein kinase C (PKC) signal transduction were responsible for COX-II expression after CXCL1 incubation for RASFs. Finally, we confirmed the transcriptional activation of nuclear factor κB (NF-κB) in RASFs after incubation with CXCL1. In conclusion, the current study provided a novel insight into the role of CXCL1 in RA pathogenesis.


Assuntos
Artrite Reumatoide , NF-kappa B , Humanos , NF-kappa B/metabolismo , Receptores de Interleucina-8B/metabolismo , Membrana Sinovial/patologia , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/metabolismo , Fosfolipases Tipo C/metabolismo , Transdução de Sinais , Quimiocinas/metabolismo , Fibroblastos/metabolismo , Células Cultivadas , Quimiocina CXCL1/metabolismo
20.
J Orthop Surg Res ; 18(1): 718, 2023 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-37737195

RESUMO

BACKGROUND: Circular RNAs (circRNAs) are important regulators on the onset and progression of rheumatoid arthritis (RA). Our purpose is to explore the role and underpin mechanism of circ_0000396 in RA progression. METHODS: RA patients (n = 39) and healthy volunteers (n = 33) were recruited from the Affiliated Hospital of Shaanxi University of Chinese Medicine for the present work. Circ_0000396, microRNA-574-5p (miR-574-5p) and R-spondin 1 (RSPO1) RNA levels were analyzed by reverse transcription-quantitative polymerase chain reaction. Cell proliferation was analyzed by 3-(4,5-dimethyl-thiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, colony formation assay, and 5-ethynyl-2'-deoxyuridine (EDU) assay. Cell apoptosis was assessed by flow cytometry. Protein expression levels of proliferating cell nuclear antigen (PCNA), Cyclin D1, Cyclin E1, BCL2-associated × protein (Bax), B-cell lymphoma-2 (Bcl2), interleukin-1ß (IL-1ß), tumor necrosis factor-α (TNF-α) and RSPO1 were detected by western blot assay. Enzyme-linked immunosorbent assay (ELISA) was conducted to analyze the secretion of pro-inflammatory cytokines including IL-1ß and TNF-α. The interaction between miR-574-5p and circ_0000396 or RSPO1 was confirmed by dual-luciferase reporter assay and RNA-pull down assay. RESULTS: Circ_0000396 expression was notably down-regulated in RA patients compared with healthy controls. Circ_0000396 overexpression suppressed the proliferation and inflammatory response and triggered the apoptosis of RA synovial fibroblasts (RASFs), accompanied by decreases in PCNA, Cyclin D1, Cyclin E1, Bcl2, IL-1ß and TNF-α protein expression and an increase in Bax protein expression. Circ_0000396 acted as a molecular sponge for miR-574-5p, and circ_0000396 overexpression-mediated protective effects on RASFs dysfunction were largely reversed by the introduction of miR-574-5p mimics. miR-574-5p interacted with the 3' untranslated region (3'UTR) of RSPO1, and miR-574-5p negatively regulated RSPO1 expression in RASFs. Circ_0000396 could up-regulate the expression of RSPO1 by sponging miR-574-5p in RASFs. RSPO1 interference largely overturned circ_0000396 overexpression-mediated effects in RASFs. CONCLUSION: Circ_0000396 restrained the proliferation and inflammation and induced the apoptosis of RASFs by mediating miR-574-5p/RSPO1 axis, which provided novel potential targets for RA treatment.


Assuntos
Artrite Reumatoide , MicroRNAs , RNA Circular , Humanos , Regiões 3' não Traduzidas , Artrite Reumatoide/genética , Proliferação de Células/genética , Ciclina D1 , Inflamação/genética , MicroRNAs/genética , Antígeno Nuclear de Célula em Proliferação , Trombospondinas , Fator de Necrose Tumoral alfa , RNA Circular/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA