scholarly journals PTX-3 Secreted by Intra-Articular-Injected SMUP-Cells Reduces Pain in an Osteoarthritis Rat Model

Cells ◽  
2021 ◽  
Vol 10 (9) ◽  
pp. 2420
Author(s):  
Minju Lee ◽  
Gee-Hye Kim ◽  
Miyeon Kim ◽  
Ji Min Seo ◽  
Yu Mi Kim ◽  
...  

Mesenchymal stem cells (MSCs) are accessible, abundantly available, and capable of regenerating; they have the potential to be developed as therapeutic agents for diseases. However, concerns remain in their further application. In this study, we developed a SMall cell+Ultra Potent+Scale UP cell (SMUP-Cell) platform to improve whole-cell processing, including manufacturing bioreactors and xeno-free solutions for commercialization. To confirm the superiority of SMUP-Cell improvements, we demonstrated that a molecule secreted by SMUP-Cells is capable of polarizing inflammatory macrophages (M1) into their anti-inflammatory phenotype (M2) at the site of injury in a pain-associated osteoarthritis (OA) model. Lipopolysaccharide-stimulated macrophages co-cultured with SMUP-Cells expressed low levels of M1-phenotype markers (CD11b, tumor necrosis factor-α, interleukin-1α, and interleukin-6), but high levels of M2 markers (CD163 and arginase-1). To identify the paracrine action underlying the anti-inflammatory effect of SMUP-Cells, we employed a cytokine array and detected increased levels of pentraxin-related protein-3 (PTX-3). Additionally, PTX-3 mRNA silencing was applied to confirm PTX-3 function. PTX-3 silencing in SMUP-Cells significantly decreased their therapeutic effects against monosodium iodoacetate (MIA)-induced OA. Thus, PTX-3 expression in injected SMUP-Cells, applied as a therapeutic strategy, reduced pain in an OA model.

2012 ◽  
Vol 2012 ◽  
pp. 1-11 ◽  
Author(s):  
Joon-Ki Kim ◽  
Sang-Won Park ◽  
Jung-Woo Kang ◽  
Yu-Jin Kim ◽  
Sung Youl Lee ◽  
...  

Therapeutic effects of GCSB-5 on osteoarthritis were measured by the amount of glycosaminoglycan in rabbit articular cartilage explantsin vitro, in experimental osteoarthritis induced by intra-articular injection of monoiodoacetate in ratsin vivo. GCSB-5 was orally administered for 28 days.In vitro, GCSB-5 inhibited proteoglycan degradation. GCSB-5 significantly suppressed the histological changes in monoiodoacetate-induced osteoarthritis. Matrix metalloproteinase (MMP) activity, as well as, the levels of serum tumor necrosis factor-α, cyclooxygenase-2, inducible nitric oxide synthase protein, and mRNA expressions were attenuated by GCSB-5, whereas the level of interleukin-10 was potentiated. By GCSB-5, the level of nuclear factor-κB p65 protein expression was significantly attenuated but, on the other hand, the level of inhibitor of κB-α protein expression was increased. These results indicate that GCSB-5 is a potential therapeutic agent for the protection of articular cartilage against progression of osteoarthritis through inhibition of MMPs activity, inflammatory mediators, and NF-κB activation.


2019 ◽  
Vol 7 (4) ◽  
pp. 17-33
Author(s):  
Alsadek H Bogzil ◽  
Gamal Shams ◽  
Sohair Abd El-Latif

The present study was designed to compare the anti-inflammatory effect of sodium hyaluronate, which is similar to the lubricant fluid that found naturally in the capsule of the healthy joint with diclofenac sodium, a member of NSAIDs commonly used in treatment of Osteoarthritis (OA), separately and in combination on an experimental model of osteoarthritis in rats induced by monosodium-iodoacetate (MIA). Twenty-five male albino rats weighing at the beginning of the experiment 160± 20 gm were used in this study. Rats were housed in cages at 25± 0.5°C. The rats were divided into 5 main groups.  


2021 ◽  
Vol 2021 ◽  
pp. 1-8
Author(s):  
Xubin Bao ◽  
Cai Chen ◽  
Liyong Yuan

Triptolide (T10) is a common anti-inflammatory and analgesic drug. However, the activation of microglia and elimination of the corresponding inflammatory response are new targets for the treatment of neuropathic pain. Chemokine CCL (CCL2) is a key mediator for activating microglia. In this study, the effects of triptolide on the activation and polarization of microglia cells and CCL2 and its corresponding receptor, chemokine receptor 2 (CCR2), were mainly discussed. Microglia were stimulated with 1 μg/mL lipopolysaccharide (LPS) and pretreated with 10, 20, and 40 nM T10 and CCR2 antagonist (RS102895), respectively. The quantitative polymerase chain reaction (QPCR) and western blot results showed that T10 could obviously inhibit the upregulation of CCL2 and CCR2 induced by LPS stimulation in microglia cells, inhibit the fluorescence intensity of glial fibrillary acidic protein (GFAP) and inducible nitric oxide synthase (iNOS) antibody immunostaining in cells, and upregulate the fluorescence intensity of arginase 1 antibody in cells. The expression of interleukin-6 (IL-6), interleukin-1β (IL-1β), and tumor necrosis factor-α (TNF-α) was inhibited in a dose-dependent manner. RS102895 can significantly reverse the activation and M2 polarization of microglia pretreated with 40 nM T10 and weaken the anti-inflammatory effect of T10. The addition of CCL2 did not extremely affect the function of RS102895. T10 may inhibit microglia activation and M1 polarization by inhibiting the expression of CCL2 and CCR2, promoting M2 polarization, reducing the level of inflammatory factors in cells, and exerting its analgesic effect, which is worthy of clinical promotion as a drug for neuropathic pain.


2019 ◽  
Author(s):  
Lei Li ◽  
Xiang-Hui Wu ◽  
Xiao-Jing Zhao ◽  
Lu Xu ◽  
Cai-Long Pan ◽  
...  

Abstract Background: Alzheimer’s disease (AD) is a major clinical problem, but there is a distinct lack of effective therapeutic drugs for this disease. We investigated the potential therapeutic effects of zerumbone, a subtropical ginger sesquiterpene, in transgenic APP/PS1 mice, rodent models of AD which exhibit cerebral amyloidosis and neuroinflammation. Methods: The N9 microglial cell line and primary microglial cells were cultured to investigate the effects of zerumbone on microglia. APP/PS1 mice were treated with zerumbone, and non-cognitive and cognitive behavioral impairments were assessed and compared between the treatment and control groups. The animals were then sacrificed, and tissues were collected for further analysis. The potential therapeutic mechanism of zerumbone and the signaling pathways involved were also investigated. Results: Zerumbone suppressed the expression of pro-inflammatory cytokines and induced a switch in microglial phenotype from the classic inflammatory phenotype to the alternative anti-inflammatory phenotype by inhibiting the mitogen-activated protein kinase (MAPK)/nuclear factor-kappa B signaling pathway in vitro. After a treatment period of 20 days, zerumbone significantly ameliorated deficits in both non-cognitive and cognitive behaviors in transgenic APP/PS1 mice. Zerumbone significantly reduced β-amyloid deposition and attenuated pro-inflammatory microglial activation in the cortex and hippocampus. Interestingly, zerumbone significantly increased the proportion of anti-inflammatory microglia among all activated microglia, potentially contributing to reduced β-amyloid deposition by enhancing phagocytosis. Meanwhile, zerumbone also reduced the expression of key molecules of the MAPK pathway, such as p38 and extracellular signal-regulated kinase. Conclusions: Overall, zerumbone effectively ameliorated behavioral impairments, attenuated neuroinflammation, and reduced β-amyloid deposition in transgenic APP/PS1 mice. Zerumbone exhibited substantial anti-inflammatory activity in microglial cells and induced a phenotypic switch in microglia from the pro-inflammatory phenotype to the anti-inflammatory phenotype by inhibiting the MAPK signaling pathway, which may play an important role in its neuroprotective effects. Our results suggest that zerumbone is a potential therapeutic agent for human neuroinflammatory and neurodegenerative diseases, in particular AD.


Plasma ◽  
2018 ◽  
Vol 1 (2) ◽  
pp. 261-276 ◽  
Author(s):  
Letizia Crestale ◽  
Romolo Laurita ◽  
Anna Liguori ◽  
Augusto Stancampiano ◽  
Maria Talmon ◽  
...  

Monocytes are involved in innate immune surveillance, establishment and resolution on inflammation, and can polarize versus M1 (pro-inflammatory) or M2 (anti-inflammatory) macrophages. The possibility to control and drive immune cells activity through plasma stimulation is therefore attractive. We focused on the effects induced by cold-atmospheric plasma on human primary monocytes and monocyte-derived macrophages. Monocytes resulted more susceptible than monocyte-derived macrophages to the plasma treatment as demonstrated by the increase in reactive oxygen (ROS) production and reduction of viability. Macrophages instead were not induced to produce ROS and presented a stable viability. Analysis of macrophage markers demonstrated a time-dependent decrease of the M1 population and a correspondent increase of M2 monocyte-derived macrophages (MDM). These findings suggest that plasma treatment may drive macrophage polarization towards an anti-inflammatory phenotype.


2021 ◽  
Vol 35 ◽  
pp. 205873842110267
Author(s):  
Ping Ni ◽  
Yue-Qin Liu ◽  
Jin-Yu Man ◽  
Wang Li ◽  
Shan-Shan Xue ◽  
...  

Macrophage plays a critical part in host defense, tissue repair, and anti-inflammation; Macrophage reprogramming is responsible for disease development or regression. We aimed to clarify the effect of sinomenine-4-hydroxy-palmitate (C16), on macrophage reprogramming and anti-inflammatory in endotoxemia model. According to a structure modification of SIN (Sinomenine), C16 was found. Then, based on the endotoxin model, the mice liver and kidney toxicity was evaluated and serum cytokines level of IL-6 (Interleukin-6), TNF-α (Tumor necrosis factor-α), and IL-1β (Interleukin-1β) were measured by ELISA (Enzyme linked immunosorbent assay). Then, we confirmed the effect of C16 on macrophages reprogramming, we used the flow cytometry to test the effect of C16 on macrophages apoptosis in vitro. Then, iNOS (Inducible nitric oxide synthase), M1-type related cytokines, such as IL-1β, TNF-α, and M2-type related cytokines, such as Arg-1 (Arginase-1), CD206, Fizz1, and Ym1 was detected, which expressed in ANA-1 and primary peritoneal macrophages. To further explore the molecular mechanism of C16 in reprogramming of macrophages from M1 toward M2 phenotype, the expression of STAT1 (signal transducer and activator of Transcription 1), STAT3, ERK1/2 (extracellular signal regulated kinase1/2), AKT, p38, and its corresponding phosphorylation were determined by western blot. Our results demonstrated that C16 improved the survival rate of LPS- (lipopolysaccharide) challenged mice and decreased the inflammatory cytokines expression; After C16 treatment, the expression of M1 phenotype correlation factors decreased significantly, while the expression of M2 phenotype correlation factors increased significantly at different levels compared with normal group. It indicated that C16 reprogram macrophages phenotype from M1 toward M2 following LPS stimulus. Furthermore, the results also showed that C16 showed anti-inflammatory effect by inhibiting LPS-induced p38, AKT and STAT1 phosphorylation and contributing ERK1/2 activation. C16 promoted macrophage reprogramming toward M2-like phenotype via p-p38/p-AKT or STAT1 signals pathway and C16 might be a valid candidate for inflammatory disease.


2021 ◽  
Vol 12 ◽  
Author(s):  
Daniela C. Ivan ◽  
Sabrina Walthert ◽  
Giuseppe Locatelli

In multiple sclerosis (MS) and other neuroinflammatory diseases, monocyte-derived cells (MoCs) traffic through distinct central nervous system (CNS) barriers and gain access to the organ parenchyma exerting detrimental or beneficial functions. How and where these MoCs acquire their different functional commitments during CNS invasion remains however unclear, thus hindering the design of MS treatments specifically blocking detrimental MoC actions. To clarify this issue, we investigated the distribution of iNOS+ pro-inflammatory and arginase-1+ anti-inflammatory MoCs at the distinct border regions of the CNS in a mouse model of MS. Interestingly, MoCs within perivascular parenchymal spaces displayed a predominant pro-inflammatory phenotype compared to MoCs accumulating at the leptomeninges and at the intraventricular choroid plexus (ChP). Furthermore, in an in vitro model, we could observe the general ability of functionally-polarized MoCs to migrate through the ChP epithelial barrier, together indicating the ChP as a potential CNS entry and polarization site for MoCs. Thus, pro- and anti-inflammatory MoCs differentially accumulate at distinct CNS barriers before reaching the parenchyma, but the mechanism for their phenotype acquisition remains undefined. Shedding light on this process, we observed that endothelial (BBB) and epithelial (ChP) CNS barrier cells can directly regulate transcription of Nos2 (coding for iNOS) and Arg1 (coding for arginase-1) in interacting MoCs. More specifically, while TNF-α+IFN-γ stimulated BBB cells induced Nos2 expression in MoCs, IL-1β driven activation of endothelial BBB cells led to a significant upregulation of Arg1 in MoCs. Supporting this latter finding, less pro-inflammatory MoCs could be found nearby IL1R1+ vessels in the mouse spinal cord upon neuroinflammation. Taken together, our data indicate differential distribution of pro- and anti-inflammatory MoCs at CNS borders and highlight how the interaction of MoCs with CNS barriers can significantly affect the functional activation of these CNS-invading MoCs during autoimmune inflammation.


2017 ◽  
Vol 474 (4) ◽  
pp. 521-537 ◽  
Author(s):  
Nicola J. Darling ◽  
Rachel Toth ◽  
J. Simon C. Arthur ◽  
Kristopher Clark

The salt-inducible kinases (SIKs) control a novel molecular switch regulating macrophage polarization. Pharmacological inhibition of the SIKs induces a macrophage phenotype characterized by the secretion of high levels of anti-inflammatory cytokines, including interleukin (IL)-10, and the secretion of very low levels of pro-inflammatory cytokines, such as tumour necrosis factor α. The SIKs, therefore, represent attractive new drug targets for the treatment of macrophage-driven diseases, but which of the three isoforms, SIK1, SIK2 or SIK3, would be appropriate to target remains unknown. To address this question, we developed knock-in (KI) mice for SIK1, SIK2 and SIK3, in which we introduced a mutation that renders the enzymes catalytically inactive. Characterization of primary macrophages from the single and double KI mice established that all three SIK isoforms, and in particular SIK2 and SIK3, contribute to macrophage polarization. Moreover, we discovered that inhibition of SIK2 and SIK3 during macrophage differentiation greatly enhanced the production of IL-10 compared with their inhibition in mature macrophages. Interestingly, macrophages differentiated in the presence of SIK inhibitors, MRT199665 and HG-9-91-01, still produced very large amounts of IL-10, but very low levels of pro-inflammatory cytokines, even after the SIKs had been reactivated by removal of the drugs. Our data highlight an integral role for SIK2 and SIK3 in innate immunity by preventing the differentiation of macrophages into a potent and stable anti-inflammatory phenotype.


2021 ◽  
Vol 41 (1) ◽  
Author(s):  
Shunsuke Nojiri ◽  
Atsunori Tsuchiya ◽  
Kazuki Natsui ◽  
Suguru Takeuchi ◽  
Takayuki Watanabe ◽  
...  

AbstractThe liver has a high regenerative ability and can induce spontaneous regression of fibrosis when early liver damage occurs; however, these abilities are lost when chronic liver damage results in decompensated cirrhosis. Cell therapies, such as mesenchymal stem cell (MSC) and macrophage therapies, have attracted attention as potential strategies for mitigating liver fibrosis. Here, we evaluated the therapeutic effects of HMGB1 peptide synthesized from box A of high mobility group box 1 protein. Liver damage and fibrosis were evaluated using a carbon tetrachloride (CCl4)-induced cirrhosis mouse model. The effects of HMGB1 peptide against immune cells were evaluated by single-cell RNA-seq using liver tissues, and those against monocytes/macrophages were further evaluated by in vitro analyses. Administration of HMGB1 peptide did not elicit a rapid response within 36 h, but attenuated liver damage after 1 week and suppressed fibrosis after 2 weeks. Fibrosis regression developed over time, despite continuous liver damage, suggesting that administration of this peptide could induce fibrolysis. In vitro analyses could not confirm a direct effect of HMGB1 peptide against monocyte/macrophages. However, macrophages were the most affected immune cells in the liver, and the number of scar-associated macrophages (Trem2+Cd9+ cells) with anti-inflammatory markers increased in the liver following HMGB1 treatment, suggesting that indirect effects of monocytes/macrophages were important for therapeutic efficacy. Overall, we established a new concept for cell-free therapy using HMGB1 peptide for cirrhosis through the induction of anti-inflammatory macrophages.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 2006-2006
Author(s):  
Rena Feinman ◽  
Iriana Colorado ◽  
Jenny Zilberberg ◽  
Thobekile T Ndlovu ◽  
Moshe Z Miller ◽  
...  

Abstract The intestinal epithelium is a primary target of graft-versus-host disease (GVHD). The hypoxia-signaling pathway has been implicated as an adaptive response in the intestinal epithelium in numerous models of inflammatory bowel disease, such as colitis and T-cell induced diarrhea. The transcription factor family, hypoxia-inducible factor (HIF) has emerged as master regulators of the transcriptional response to hypoxic stress in normal and transformed cells. HIF heterodimers consist of an oxygen-labile α subunit (HIF-1α, HIF-2α) and a constitutively expressed HIF-1β subunit that mediate a wide spectrum of physiological and cellular adaptive responses, including angiogenesis, metabolic adaption, and erythropoiesis. HIF-1 has recently been implicated as a gut-protective factor in inflammatory bowel disease models by maintaining intestinal homeostasis. HIF-1 can also skew the differentiation of T cells to regulatory T cells (Treg) via the induction of FoxP3, thereby attenuating T-cell driven colitis. Although HIF-1 and HIF-2 share many overlapping functions, HIF-1 has been implicated in the inflammatory phenotype of M1 macrophages via inducible nitric oxide synthase (iNOS) whereas HIF-2 is involved in the anti-inflammatory phenotype of M2 macrophages via arginase-1 (Arg1). Based on these findings and that mucosal inflamed tissues are hypoxic, we hypothesized that the induction of the hypoxia-signaling pathway may limit GVHD-induced mucosal inflammation and injury. To determine the adaptive roles of HIF-1 and HIF-2 in GVHD, we first tested the major histocompatibility complex (MHC)-haploidentical C567BL/6 (B6,H2b) -> B6xDBA/2 (B6D2)F1 (H2b/d) model in which donor spenocytes (2x107) and anti-Thy1 + C’ treated (T- cell depleted) bone marrow (ATBM) cells (5x106) are transplanted into B6D2F1 recipients after exposure to lethal irradiation (11Gy, split dose). B6 ATBM cells transplanted alone into B6D2F1 mice served as a negative control for all comparisons. Realtime PCR analysis demonstrated a modest increase in ileal mucosal HIF-2α expression 8 days (d) post-transplant (p<0.027). In contrast, HIF-1α mRNA levels were not induced. However, both HIF-1α and HIF-2α protein levels were upregulated 2-fold and 5-fold, respectively, in the ileal mucosa of B6D2F1 recipients receiving ATBM plus splenocytes, as determined by western blotting. Notably, Arg1 mRNA levels (HIF-2 target) were markedly upregulated during GVHD (p<0.018), whereas iNOS mRNA levels (HIF-1α target) were downregulated (p<0.01). Increased HIF-2α and Arg1 expression in the ileum as a consequence of GVHD was also observed in two MHC H2b-matched, minor histocompatability antigen (miHA)-mismatched models. BALB.B and CXB-2 mice were exposed to lethal irradiation (9Gy, split dose) and transplanted with B6 ATBM cells alone or along with host-presensitized B6 CD4+ T cells. After 8d, HIF-2α and HIF-1α mRNA ileal levels were increased and decreased, respectively, in both models undergoing GVHD. Similarly, Arg1 transcripts were increased by 12-fold (p<0.03) and 6.1 fold (p<0.007), respectively in B6->BALB.B and B6->CXB-2 models. However, after 20d post-transplant, a 4- and 3-fold decrease in Arg1 mRNA levels occurred in both models. Likewise, two anti-inflammatory, Treg-associated cytokines, interleukin-10 (IL-10) and transforming growth factor beta (TGFβ) mRNA expression were elevated by 2-and 7-fold, respectively, after d8 in B6->BALB.B mice. TGFβ levels returned to baseline (p<0.05 vs d8) after 20d and IL-10 mRNA levels were reduced by 2.5 fold (p<0.029 vs d8). Lastly, in an ELISpot assay, the addition of a prolyl hydroxylase inhibitor, dimethyloxaloylgylcine (DMOG), a HIF activator, reduced the alloreactive interferon-γ response to vehicle levels (p>0.001) in a B6 anti-B6D2F1 mixed lymphocyte reaction. Taken together, our data suggest that HIF-2/Arg1 axis confers an anti-inflammatory response in the ileum after 8d of GVHD. However, after 20d, this response is inversely correlated with the lethality of the GVHD response. The amelioration of alloreactivity by DMOG suggests that the persistent activation of HIF may be necessary to dampen GVHD. Further studies will delineate the contribution of the HIF-2 response in the maintenance of intestinal homeostasis and limiting T cell alloreactivity. Disclosures: Zilberberg: Onyx Pharmaceuticals: Research Funding. Dziopa:Onyx Pharmaceuticals: Research Funding.


Sign in / Sign up

Export Citation Format

Share Document