scholarly journals OP0236 RELEVANCE OF BIASED PAR2 INHIBITORS IN REDUCING INFLAMMATION AND CARTILAGE DEGRADATION IN IN VITRO AND IN VIVO MODELS OF RHEUMATOID ARTHRITIS

2020 ◽  
Vol 79 (Suppl 1) ◽  
pp. 149.2-150
Author(s):  
T. Brugat ◽  
B. Rugeri ◽  
G. Hommet ◽  
A. Dumont ◽  
L. Baron ◽  
...  

Background:Protease-activated receptor-2 (PAR2) is a member of a family of G-protein-coupled receptors involved in multiple physiological mechanisms. Compelling evidences have unravelled the key roles of PAR2 in the pathology of both rheumatoid arthritis (RA) and osteoarthritis (OA)1. Indeed, in vitro, in vivo and ex vivo experiments showed that this receptor promotes inflammation, cartilage erosion (and subsequent bone degradation), and pain. However, the signalling pathways involved in these functions are not well understood2. This is of importance as some pathways can promote the pathogenesis3while others prevent it4. We developed a new series of small molecules as novel biased PAR2 inhibitors to treat rheumatic diseases.Objectives:To evaluate the efficacy and mechanism of action of new biased PAR2 inhibitors on cartilage erosion and inflammation.Methods:The potency of compounds to inhibit human PAR2 signalling was evaluated in vitro by FLIPR calcium assay in HEK293 cells. The same assay was used to determine their selectivity over human PAR1 and PAR4 as well as murine versions of PAR2. The effect of several PAR2 inhibitors on 9 signalling pathways (Gi2, GoB, Gz, Gq, G13, G14, G15, B arrestin 2, EPAC) was evaluated by the BRET-based bioSens-All™ technology. In vitro anti-hypertrophic effect was determined by measuring the mRNA level of type II collagen, aggrecan and MMP13 in rat chondrocytes after IL1β stimulation. In vitro anti-inflammatory effect was determined by measuring the secretion of IL6, IL8, IL1β, TNFα and IFNγ by human monocytes. In vivo, the pharmacodynamic of our small molecules was assessed after intravenous and oral administration. Therapeutic efficacy of a compound was then evaluated in a collagen-induced arthritis model in DBA1/J mice. In this model, measures of the arthritis index score, body weight, plasma level of TNFα, IL6, IL8 and IL1β and histological evaluation of cartilage erosion were performed.Results:Our new series of small molecules are potent PAR2 inhibitors (IC50<1nM in calcium assay) with some selectivity over PAR1 and PAR4. Our compounds significantly inhibited PAR2 mediated recruitment of Gz, Gq, G13, G14 and G15. However, surprisingly, these small molecules had no effect on B arrestin 2, EPAC, Gi2 and GoB demonstrating that they are biased inhibitors. The effect of our compounds on PAR2 signalling was clearly different from 3 already existing PAR2 inhibitors described in the literature (I-117, AZ3451 and P2pal-18s). We compared the in vitro anti-hypertrophic effect on chondrocyte and anti-inflammatory effect on monocytes of these compounds to determine the importance of PAR2 signalling pathways in these cellular functions. In vivo, our small molecules had good bioavailability after oral administration of 10mg/kg in mice (clearance = 0.038L/h/kg; T½ = 9.9h; AUC= 162564 ng.h/mL; Cmax = 9005 ng/mL). The in vivo therapeutic efficacy of a biased PAR2 inhibitor in a model of collagen-induced arthritis will be presented.Conclusion:Our results show the potency of biased PAR2 inhibitors to reduce both the inflammation and cartilage erosion in rheumatoid arthritis. They confirm the huge potential of PAR2 as a therapeutic target and unravel the relevance of biased antagonism of this receptor to treat rheumatic diseases.References:[1]McCulloch et al., Frontiers in Endocrinology, 2018;2Hollenberg et al., British Journal of Pharmacology, 2014;3Sharma et al., Genes and Immunity, 2015;4Rayees et al., Cell Reports, 2019Disclosure of Interests:Thibaut Brugat Employee of: Domain Therapeutics, Baptiste Rugeri Employee of: Domain Therapeutics, Gaël Hommet Employee of: Domain Therapeutics, Alexia Dumont Employee of: Domain Therapeutics, Luc Baron Employee of: Domain Therapeutics, Célia Halter Employee of: Domain Therapeutics, Meriem Sémache Employee of: Domain Therapeutics, Arturo Mancini Employee of: Domain Therapeutics, Camille Amalric Employee of: Domain Therapeutics, Marie Giambelluco Employee of: Domain Therapeutics, Nathalie Lenne Employee of: Domain Therapeutics, Marjorie Sidhoum Employee of: Domain Therapeutics, Christel Franchet Employee of: Domain Therapeutics, Stanislas Mayer Employee of: Domain Therapeutics, Xavier Leroy Employee of: Domain Therapeutics, Stephan Schann Employee of: Domain Therapeutics

2021 ◽  
Vol 12 (1) ◽  
Author(s):  
Wei Zhang ◽  
Guoyu Yin ◽  
Heping Zhao ◽  
Hanzhi Ling ◽  
Zhen Xie ◽  
...  

AbstractIn inflamed joints, enhanced hyaluronic acid (HA) degradation is closely related to the pathogenesis of rheumatoid arthritis (RA). KIAA1199 has been identified as a hyaladherin that mediates the intracellular degradation of HA, but its extracellular function remains unclear. In this study, we found that the serum and synovial levels of secreted KIAA1199 (sKIAA1199) and low-molecular-weight HA (LMW-HA, MW < 100 kDa) in RA patients were significantly increased, and the positive correlation between them was shown for the first time. Of note, treatment with anti-KIAA1199 mAb effectively alleviated the severity of arthritis and reduced serum LMW-HA levels and cytokine secretion in collagen-induced arthritis (CIA) mice. In vitro, sKIAA1199 was shown to mediate exogenous HA degradation by attaching to the cell membrane of RA fibroblast-like synoviosytes (RA FLS). Furthermore, the HA-degrading activity of sKIAA1199 depended largely on its adhesion to the membrane, which was achieved by its G8 domain binding to ANXA1. In vivo, kiaa1199-KO mice exhibited greater resistance to collagen-induced arthritis. Interestingly, this resistance could be partially reversed by intra-articular injection of vectors encoding full-length KIAA1199 instead of G8-deleted KIAA119 mutant, which further confirmed the indispensable role of G8 domain in KIAA1199 involvement in RA pathological processes. Mechanically, the activation of NF-κB by interleukin-6 (IL-6) through PI3K/Akt signaling is suggested to be the main pathway to induce KIAA1199 expression in RA FLS. In conclusion, our study supported the contribution of sKIAA1199 to RA pathogenesis, providing a new therapeutic target for RA by blocking sKIAA1199-mediated HA degradation.


2021 ◽  
Vol 11 (1) ◽  
Author(s):  
F. N. Novikov ◽  
M. V. Panova ◽  
I. Y. Titov ◽  
V. S. Stroylov ◽  
O. V. Stroganov ◽  
...  

AbstractThe pathophysiology of osteoarthritis (OA) includes the destruction of subchondral bone tissue and inflammation of the synovium. Thus, an effective disease-modifying treatment should act on both of these pathogenetic components. It is known that cSrc kinase is involved in bone and cartilage remodeling, and SYK kinase is associated with the inflammatory component. Thus the aim of this study was to characterize the mechanism of action and efficacy of a small molecule multikinase inhibitor MT-SYK-03 targeting SYK and cSrc kinases among others in different in vitro and in vivo arthritis models. The selectivity of MT-SYK-03 kinase inhibition was assayed on a panel of 341 kinases. The compound was evaluated in a set of in vitro models of OA and in vivo OA and RA models: surgically-induced arthritis (SIA), monosodium iodoacetate-induced arthritis (MIA), collagen-induced arthritis (CIA), adjuvant-induced arthritis (AIA). MT-SYK-03 inhibited cSrc and SYK with IC50 of 14.2 and 23 nM respectively. Only five kinases were inhibited > 90% at 500 nM of MT-SYK-03. In in vitro OA models MT-SYK-03 reduced hypertrophic changes of chondrocytes, bone resorption, and inhibited SYK-mediated inflammatory signaling. MT-SYK-03 showed preferential distribution to joint and bone tissue (in rats) and revealed disease-modifying activity in vivo by halving the depth of cartilage erosion in rat SIA model, and increasing the pain threshold in rat MIA model. Chondroprotective and antiresorptive effects were shown in a monotherapy regime and in combination with methotrexate (MTX) in murine and rat CIA models; an immune-mediated inflammation in rat AIA model was decreased. The obtained preclinical data support inhibition of cSrc and SYK as a viable strategy for disease-modifying treatment of OA. A Phase 2 clinical study of MT-SYK-03 is to be started.


2019 ◽  
Vol 15 (11) ◽  
pp. 2281-2290 ◽  
Author(s):  
Yao Zhao ◽  
Zhesheng He ◽  
Ruoping Wang ◽  
Pengju Cai ◽  
Xiangchun Zhang ◽  
...  

Rheumatoid arthritis (RA) is a chronic autoimmune disease characterized by synovial inflammation and progressive cartilage and bone damage. In our previous studies, we found that Au clusters using glutathione as a template (GACs) produced profound anti-inflammatory effects in vitro on lipopolysaccharide (LPS)-induced inflammation in mouse macrophage RAW 264.7 cells and type II collagen-induced rat RA in vivo. In this study, we examined whether the template for Au clusters synthesis has an effect on its anti-inflammatory effect and whether Au nanoparticles with larger particle diameter produce the same anti-inflammatory effect. We synthesized Au clusters with bovine serum albumin (BSA) as a template (BACs), Au clusters with glutathione (GSH) as a template (GACs), and Au nanoparticles with glutathione as a template (GANs) and compared their anti-inflammatory effects in vitro and in vivo. These three Au nanomaterials can inhibit the production of lipopolysaccharide (LPS)-induced proinflammatory mediators and ameliorate type II collagen-induced rat RA. However, although the three Au nanomaterials produced similar anti-inflammatory effects, the GANs with larger particle sizes were less stable in vivo and accumulated in the peritoneum after intraperitoneal injection, resulting in poor absorption in vivo. The BACs showed relatively high liver accumulation due to the larger molecular weight of the outer shell. Therefore, we believe that the GACs are potential reliable nanodrugs for the treatment of RA.


2020 ◽  
Vol 21 (2) ◽  
pp. 378
Author(s):  
Kyung Eun Kim ◽  
Suwon Jeon ◽  
Jisun Song ◽  
Tae Sung Kim ◽  
Min Kyung Jung ◽  
...  

Rheumatoid arthritis (RA) is a chronic autoimmune disease that is associated with systemic inflammation and results in the destruction of joints and cartilage. The pathogenesis of RA involves a complex inflammatory process resulting from the action of various proinflammatory cytokines and, therefore, many novel therapeutic agents to block cytokines or cytokine-mediated signaling have been developed. Here, we tested the preventive effects of a small peptide, AESIS-1, in a mouse model of collagen-induced arthritis (CIA) with the aim of identifying a novel safe and effective biological for treating RA. This novel peptide significantly suppressed the induction and development of CIA, resulting in the suppression of synovial inflammation and cartilage degradation in vivo. Moreover, AESIS-1 regulated JAK/STAT3-mediated gene expression in vitro. In particular, the gene with the most significant change in expression was suppressor of cytokine signaling 3 (Socs3), which was enhanced 8-fold. Expression of the STAT3-specific inhibitor, Socs3, was obviously enhanced dose-dependently by AESIS-1 at both the mRNA and protein levels, resulting in a significant reduction of STAT3 phosphorylation in splenocytes from severe CIA mice. This indicated that AESIS-1 regulated STAT3 activity by upregulation of SOCS3 expression. Furthermore, IL-17 expression and the frequency of Th17 cells were considerably decreased by AESIS-1 in vivo and in vitro. Collectively, our data suggest that the novel synthetic peptide AESIS-1 could be an effective therapeutic for treating RA via the downregulation of STAT3 signaling.


2022 ◽  
Author(s):  
Chenxi Li ◽  
Rui Liu ◽  
Yurong Song ◽  
Dongjie Zhu ◽  
Liuchunyang Yu ◽  
...  

Abstract Triptolide (TP) is a DMARD highly effective in patients with RA. Hyaluronic acid (HA) hydrogels loaded RGD-attached gold nanoparticles containing TP were synthesized to alleviate the toxicity and increase therapeutic specificity. The hydrogels can be applied for targeted photothermal-chemo therapy, and in vivo imaging of RA. Heat was locally generated at the inflammation site after degradation of HA chains due to near-infrared resonance (NIR) irradiation of gold nanoparticles (AuNPs), and TP was released. Administration of the hybrid hydrogels containing low dosage of TP combined with NIR irradiation alleviated arthritic conditions and improved the inflamed joint in collagen-induced arthritis (CIA) mice. In vitro effect of the hydrogel was mediated through decrease of phosphorylation of mTOR and its substrate, p70S6K1, thus inhibiting mTOR pathway.


2021 ◽  
Vol 12 ◽  
Author(s):  
Yunhui Feng ◽  
Liyan Mei ◽  
Maojie Wang ◽  
Qingchun Huang ◽  
Runyue Huang

18β-Glycyrrhetinic acid (18β-GA), an active component from Glycyrrhiza glabra L. root (licorice), has been demonstrated to be able to protect against inflammatory response and reduce methotrexate (MTX)-derived toxicity. This study was therefore designed to test the therapeutic possibility of 18β-GA on rheumatoid arthritis (RA) and to explore the underlying mechanism. LPS or TNF-α-induced inflammatory cell models and collagen-induced arthritis (CIA) animal models were applied in this study. Real-time quantitative PCR (RT-qPCR) was used to measure the mRNA levels of various cytokines and FOXO family members. The protein levels of molecules in the MAPK/NF-κB signaling pathway were analyzed using western blot. The cell proliferation assay and colony-forming assay were used to test the influence of 18β-GA on cell viability. The cell apoptosis assay and cell cycle assay were performed to detect the effect of 18β-GA on cell proliferative capacity by using flow cytometry. Hematoxylin and eosin (H&amp;E) staining was performed to evaluate pathological changes after drug administration. The enzyme-linked immunosorbent assay (ELISA) was carried out for the detection of cytokines in serum. In vitro, we found that 18β-GA decreased the mRNA levels of IL-1β, IL-6, and COX-2 by inhibiting the MAPK/NF-κB signaling pathway in MH7A and RAW264.7 cell lines. Moreover, 18β-GA was able to suppress cell viability, trigger cell apoptosis, and G1 phase cell cycle arrest in our in vitro studies. 18β-GA dramatically enhanced the mRNA level of FOXO3 in both TNF-α- and LPS-induced inflammation models in vitro. Interestingly, after analyzing GEO datasets, we found that the FOXO3 gene was significantly decreased in the RA synovial tissue as compared to healthy donors in multiple microarray studies. In vivo, 18β-GA exhibited a promising therapeutic effect in a collagen-induced arthritis mouse model by alleviating joint pathological changes and declining serum levels of TNF-α, IL-1β, and IL-6. Finally, we observed that 18β-GA administration could mitigate liver damage caused by collagen or MTX. Collectively, the current study demonstrates for the first time that 18β-GA can inhibit inflammation and proliferation of synovial cells, and the underlying mechanism may be associated with its inhibition of MAPK/NF-κB signaling and promotion of FOXO3 signaling. Therefore, 18β-GA is expected to be a new drug candidate for RA therapy.


Arthritis ◽  
2013 ◽  
Vol 2013 ◽  
pp. 1-13 ◽  
Author(s):  
Christopher Rashidifard ◽  
Christopher Vercollone ◽  
Scott Martin ◽  
Bin Liu ◽  
Mark E. Brezinski

Many musculoskeletal disorders (MDs) are associated with irreversible bone and cartilage damage; this is particularly true for osteoarthritis (OA). Therefore, a clinical need exists for modalities which can detect OA and other MDs at early stages. Optical coherence tomography (OCT) is an infrared-based imaging, currently FDA approved in cardiology and ophthalmology, which has a resolution greater than 10 microns and acquisition rate of 120 frames/second. It has shown feasibility for imaging early OA, identifying changes prior to cartilage thinning both in vitro and in vivo in patients and in OA animal models. In addition, OCT has shown an ability to identify early rheumatoid arthritis (RA) and guide tendon repair, but has the potential for an even greater impact. Clinical trials in OA are currently underway, as well as in several other MDs.


Author(s):  
Silpa Gampala ◽  
Fenil Shah ◽  
Chi Zhang ◽  
Steven D. Rhodes ◽  
Olivia Babb ◽  
...  

Abstract Background MPNST is a rare soft-tissue sarcoma that can arise from patients with NF1. Existing chemotherapeutic and targeted agents have been unsuccessful in MPNST treatment, and recent findings implicate STAT3 and HIF1-α in driving MPNST. The DNA-binding and transcriptional activity of both STAT3 and HIF1-α is regulated by Redox factor-1 (Ref-1) redox function. A first-generation Ref-1 inhibitor, APX3330, is being tested in cancer clinical trials and could be applied to MPNST. Methods We characterised Ref-1 and p-STAT3 expression in various MPNST models. Tumour growth, as well as biomarkers of apoptosis and signalling pathways, were measured by qPCR and western blot following treatment with inhibitors of Ref-1 or STAT3. Results MPNSTs from Nf1-Arfflox/floxPostnCre mice exhibit significantly increased positivity of p-STAT3 and Ref-1 expression when malignant transformation occurs. Inhibition of Ref-1 or STAT3 impairs MPNST growth in vitro and in vivo and induces apoptosis. Genes highly expressed in MPNST patients are downregulated following inhibition of Ref-1 or STAT3. Several biomarkers downstream of Ref-1 or STAT3 were also downregulated following Ref-1 or STAT3 inhibition. Conclusions Our findings implicate a unique therapeutic approach to target important MPNST signalling nodes in sarcomas using new first-in-class small molecules for potential translation to the clinic.


2019 ◽  
Vol 26 (30) ◽  
pp. 5609-5624
Author(s):  
Dijana Saftić ◽  
Željka Ban ◽  
Josipa Matić ◽  
Lidija-Marija Tumirv ◽  
Ivo Piantanida

: Among the most intensively studied classes of small molecules (molecular weight < 650) in biomedical research are small molecules that non-covalently bind to DNA/RNA, and another intensively studied class is nucleobase derivatives. Both classes have been intensively elaborated in many books and reviews. However, conjugates consisting of DNA/RNA binder covalently linked to nucleobase are much less studied and have not been reviewed in the last two decades. Therefore, this review summarized reports on the design of classical DNA/RNA binder – nucleobase conjugates, as well as data about their interactions with various DNA or RNA targets, and even in some cases protein targets are involved. According to these data, the most important structural aspects of selective or even specific recognition between small molecule and target are proposed, and where possible related biochemical and biomedical aspects were discussed. The general conclusion is that this, rather new class of molecules showed an amazing set of recognition tools for numerous DNA or RNA targets in the last two decades, as well as few intriguing in vitro and in vivo selectivities. Several lead research lines show promising advancements toward either novel, highly selective markers or bioactive, potentially druggable molecules.


Sign in / Sign up

Export Citation Format

Share Document