Hydrolyzed Fish Collagen Inhibits Inflammatory Cytokines Secretion in Lipopolysaccharide-Induced HUVECs

2014 ◽  
Vol 1025-1026 ◽  
pp. 570-573 ◽  
Author(s):  
Chao Liu ◽  
Yang Xue ◽  
Jiao Sun

The present study was designed to investigate the effects of hydrolyzed fish collagen (HFC) on the cytokine production of lipopolysaccharide (LPS)-induced human umbilical vein endothelial cells (HUVECs). The in vitro inflammation model was established using LPS-induced HUVECs.The cell viability of HUVECs and the secretion level of inflammatory cytokines,IL-6, IL-8, and TNF-α from LPS-treated HUVECs exposed to HFC were determined respectively using MTT and ELISA assays. Our results indicate that HFC promoted HUVECs proliferation, and significantly reduced IL-6, IL-8, and TNF-α production in LPS-stimulated HUVECs. It suggested that HFC had a prominent anti-inflammatory property, HFC could be considered as a promising candidate for anti-inflammatory wound dressing.

Author(s):  
Reza Afrisham ◽  
Sahar Sadegh-Nejadi ◽  
Reza Meshkani ◽  
Solaleh Emamgholipour ◽  
Molood Bagherieh ◽  
...  

Introduction: Obesity is a disorder with low-grade chronic inflammation that plays a key role in the hepatic inflammation and steatosis. Moreover, there are studies to support the role of exosomes in the cellular communications, the regulation of metabolic homeostasis and immunomodulatory activity. Accordingly, we aimed to evaluate the influence of plasma circulating exosomes derived from females with normal-weight and obesity on the secretion of inflammatory cytokines in human liver cells. Methods: Plasma circulating exosomes were isolated from four normal (N-Exo) and four obese (O-Exo) women. The exosomes were characterized and approved for CD63 expression (common exosomal protein marker) and morphology/size using the western blot and TEM methods, respectively. The exosomes were used for stimulation of HepG2 cells in vitro. After 24 h incubation, the protein levels of TNF-α,IL-6, and IL-1β were measured in the culture supernatant of HepG2 cells using the ELISA kit. Results: The protein levels of IL-6 and TNF-α in the cells treated with O-Exo and N-Exo reduced significantly in comparison with control group (P=0.039 and P<0.001 respectively), while significance differences were not found between normal and obese groups (P=0.808, and P=0.978 respectively). However, no significant differences were found between three groups in term of IL-1β levels (P=0.069). Based on the correlation analysis, the protein levels of IL-6 were positively correlated with TNF-α (r 0.978, P<0.001). Conclusion: These findings suggest that plasma circulating exosomes have probably anti-inflammatory properties independently from body mass index and may decrease the secretion of inflammatory cytokines in liver. However, further investigations in vitro and in vivo are needed to address the anti-inflammatory function of N-Exo and O-Exo in human liver cells and/or other cells.


2015 ◽  
Vol 43 (01) ◽  
pp. 183-198 ◽  
Author(s):  
Wenjuan Yao ◽  
Chengjing Gu ◽  
Haoran Shao ◽  
Guoliang Meng ◽  
Huiming Wang ◽  
...  

Endothelial dysfunction plays an important role in the pathogenesis of atherogenesis. 2,3,5,4′-tetrahydroxystilbene-2-O-β-D-glucoside (TSG), an active component of the rhizome extract from Polygonum multiflorum (PM), exhibits significant anti-atherosclerotic activity. Here, we used human umbilical vein endothelial cells (HUVECs) induced by tumor necrosis factor-α (TNF-α) in vitro to investigate the cytoprotective effects of TSG on TNF-α-induced endothelial injury and the related mechanisms. Pretreatment with 50 and 100 μM TSG markedly attenuated TNF-α-induced loss of cell viability and release of lactate dehydrogenase (LDH) and inhibited TNF-α-induced cell apoptosis. The inhibition of vimentin expression was involved in the cytoprotection afforded by TSG. Using inhibitors for PI3K and TGFβ or siRNA for Akt and Smad2, we found that vimentin production in HUVECs is regulated by TGFβ/Smad signaling, but not by PI3K–Akt–mTOR signaling. Meanwhile, TSG inhibited both the expression of TGFβ1 and the phosphorylation of Smad2 and Smad3, and TSG suppressed the nuclear translocation of Smad4 induced by TNF-α. These results suggest that TSG protects HUVECs against TNF-α-induced cell damage by inhibiting vimentin expression via the interruption of the TGFβ/Smad signaling pathway.


2020 ◽  
Vol 35 (Supplement_3) ◽  
Author(s):  
Dan Li ◽  
Chenyu Li ◽  
Yan Xu

Abstract Background and Aims Acute kidney injury (AKI), commonly appeared in cardiac arrest, surgery and kidney transplantation which involved in ischemia-reperfusion (IR) injury of kidney. However, the mechanisms underlying inflammatory response in IR AKI is still unclear. Method Public dataset showed kruppel-like factor 6 (KLF6) was significantly highly expressed (P&lt;0.05) in AKI, implies KLF6 might be associated with AKI. To evaluate the mechanism of KLF6 on IR AKI, 30 rats were randomly divided into sham and IR group, and were sacrificed at 0 h, 3 h, 6 h, 12 h or 24 h after IR. Results The results showed KLF6 expression was peaking at 6 h after IR, and the expression of pro-inflammatory cytokines MCP-1 and TNF-α were increased both in serum and kidney tissues after IR, while anti-inflammatory cytokine IL-10 was decreased after IR. Furthermore, in vitro results showed KLF6 knock-down reduced the pro-inflammatory cytokines expression and increased the anti-inflammatory cytokines expression. Conclusion These results suggest that (1) KLF6 might be a novel biomarker for early diagnosis of AKI and (2) targeting KLF6 expression may offer novel strategies to protect kidneys from IR AKI Figure KLF6, AKI, Control Inflammation


2013 ◽  
Vol 110 (07) ◽  
pp. 141-152 ◽  
Author(s):  
Yaw Asare ◽  
Erdenechimeg Shagdarsuren ◽  
Johannes Schmid ◽  
Pathricia Tilstam ◽  
Jochen Grommes ◽  
...  

SummaryThe COP9 signalosome (CSN), a multifunctional protein complex involved in the regulation of cullin-RING-E3 ubiquitin ligases (CRLs), has emerged as a regulator of NF-κB signalling. As NF-κB drives the expression of pro-inflammatory and pro-atherosclerotic genes, we probed the yet unknown role of the CSN, in particular CSN5, on NF-KB-mediated atherogenic responses in endothelial cells. Co-immunoprecipitation in human umbilical vein endothelial cells (HUVECs) revealed the presence of a super-complex between IKK and CSN, which dissociates upon TNF-α stimulation. Furthermore, CSN5 silencing enhanced TNF-α-induced IKB-α degradation and NF-κB activity in luci-ferase reporter assays. This was paralleled by an increased NF-KB-driven upregulation of atherogenic chemokines and adhesion molecules, as measured by qPCR and flow cytometry, and translated into an enhanced arrest of THP-1 monocytes on TNF-α-stimulated, CSN5-depleted HUVECs. Reverse effects on NF-κB activity and THP-1 arrest were seen upon CSN5 overexpression. Finally, double-immunostaining confirmed the expression of CSN subunits in the endothelium of human atherosclerotic lesions, and revealed an increased expression of CSN5 which correlated with atheroprogression. In conclusion, endothelial CSN5 attenuates NF-KB-dependent pro-inflammatory gene expression and monocyte arrest on stimulated endothelial cells in vitro, suggesting that CSN5 might serve as a negative regulator of atherogenesis.Note: The review process for this manuscript was fully handled by G. Y. H. Lip, Editor in Chief.


2014 ◽  
Vol 884-885 ◽  
pp. 446-449
Author(s):  
Fu Jiang Chu ◽  
Hong Yan Ma ◽  
Xiao Bao Jin ◽  
Jia Yong Zhu

House fly maggot, Musca domestica (Linnaeus) (Diptera: Muscidae) is one of the traditional Chinese medicine (TCM). In our earlier studies, the anti-inflammatory and anti-atherosclerotic functions of the housefly maggot have been found and also the anti-inflammatory effective parts have been acquired. In this study, the effect of housefly maggot anti-inflammatory parts on proliferation and migration of TNF-α-stimulated human umbilical vein endothelial cells (HUVEC) were investigated. And the results showed that the proliferation index and the migration rates of HUVEC which stimulated by TNF-α were decreased significantly in housefly maggot anti-inflammatory parts treatment group. And also the secretion of vascular endothelial growth factor (VEGF) was decreased too compared with only TNF-α treatment group. Based on the above, the housefly maggot anti-inflammatory parts could regulate the endothelial cell dysfunction through decreasing cell proliferation and migration and a reduction in VEGF expression might plays a key role in this process.


Pharmaceutics ◽  
2019 ◽  
Vol 11 (10) ◽  
pp. 500 ◽  
Author(s):  
Denise Beconcini ◽  
Francesca Felice ◽  
Ylenia Zambito ◽  
Angela Fabiano ◽  
Anna Maria Piras ◽  
...  

This study aimed at evaluating the anti-inflammatory effect of natural cherry extract (CE), either free or encapsulated in nanoparticles (NPs) based on chitosan derivatives (Ch-der) or poly(lactic-co-glycolic acid) (PLGA), on human umbilical vein endothelial cells (HUVEC). CE from Prunus avium L. was characterized for total polyphenols, flavonoids, and anthocyanins content. CE and CE-loaded NP cytotoxicity and protective effect on lipopolysaccharide (LPS)-stressed HUVEC were tested by water-soluble tetrazolium salt (WST-1) assay. Pro- and anti-inflammatory cytokines (TNF-α, IL-6, IL-10, and PGE2) released by HUVEC were quantified by enzyme-linked immunosorbent assay (ELISA). All NP types were internalized into HUVEC after 2 h incubation and promoted the anti-inflammatory effect of free CE at the concentration of 2 µg gallic acid equivalents (GAE)/mL. CE-loaded Ch-der NPs showed the highest in vitro uptake and anti-inflammatory activity, blunting the secretion of IL-6, TNF-α, and PGE2 cytokines. Moreover, all NPs reduced the production of nitric oxide and NLRP3 inflammasome, and had a stronger anti-inflammatory effect than the major corticosteroid dexamethasone. In particular, the results demonstrate that natural CE protects endothelial cells from inflammatory stress when encapsulated in NPs based on quaternary ammonium chitosan. The CE beneficial effects were directly related with in vitro internalization of CE-loaded NPs.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 3274-3274
Author(s):  
Lionel Loubaki ◽  
Renée Bazin

Abstract Abstract 3274 Background: Cells from the monocytic lineage are known to play a central role in the immune defense against pathogens. In the adaptive immune response, they act as antigen presenting cells to trigger T and B cell responses. Monocytic cells also participate in innate immunity following recognition of pathogen-associated molecular patterns (PAMPs) such as bacterial lipopolysaccharides (LPS), which leads to their activation and release of very potent inflammatory mediators. The innate immune response thus needs to be tightly regulated to control not only its onset, but also its termination in order to avoid excessive inflammation. Recent studies have shown that the differentiation and functions of monocytic cells involve small RNA species, named microRNAs (miRNAs). MiRNAs are 21–23 nucleotide long single strand RNAs, which mainly cause gene silencing by degradation of target mRNAs or by inhibition of translation. Among them, miR-146a has captivated interest as it plays an important role in the negative regulation of acute inflammatory responses during activation of the innate immune system. In fact, it has been shown that miR-146a expression is gradually increased in THP-1 monocytic cells following stimulation with LPS or cytokines (e.g. IL-1β and TNF-α) via a NF-κB dependent pathway. MiR-146a inhibits the expression of IRAK1 and TRAF6 leading to the subsequent suppression of NF-κB activity. Consequently, the expression of NF-κB target genes such as IL-1β, TNF-α and PU.1 is suppressed. Therefore, miR146a controls NF-κB signaling via a negative feedback regulation loop and thus can be considered as an anti-inflammatory mediator. IVIg is a therapeutic preparation of polyclonal human IgG isolated from the plasma of thousands of healthy donors. IVIg is well known for its anti-inflammatory effects on a variety of immune cells and processes. More precisely, it has been shown to abrogate the capacity of monocyte-derived dendritic cells to secrete pro-inflammatory cytokines while increasing the expression of anti-inflammatory cytokines such as IL-10. We thus hypothesize that at least some of the anti-inflammatory effects of IVIg on monocytic cells could be triggered through the modulation of miR-146a expression. Objectives: To evaluate the involvement of miR-146a in the anti-inflammatory effects of IVIg following LPS stimulation of human monocytes. Methods: Human monocytes were obtained from the blood of healthy volunteers and treated with LPS (1 mg/mL) or IVIg (15 mg/mL) alone or alternatively, pretreated with LPS followed by addition of IVIg. Pre-treatment with LPS was done during for 4 h prior to addition of IVIg for 3, 6, 12 and 24 hours. Cells were then recovered and separated in two parts. The first part was used to extract the small RNA fraction of total RNA for miRNA analysis and the second part was used for protein isolation. The miR-146a level was measured by real time PCR while NF-kB and IRF4 protein levels were evaluated by western blotting. Finally, the expression of the transcription factor PU.1 was evaluated by flow cytometry. Results: Our preliminary data revealed that addition of IVIg to LPS-pretreated human monocytes resulted in a significant upregulation of miR-146a expression associated with a significant reduction in NF-κB expression. Furthermore, the expression of the PU.1/IRF4 transcriptional activator complex involved in the stimulation of inflammatory cytokine production was modulated. Indeed, we found that the expression PU.1 was reduced in IVIg-treated cells whereas IRF4 expression was increased, thus promoting the IRF4-mediated cytokine production inhibitory pathway. Conclusion: Our preliminary data suggest that in human monocytes, the anti-inflammatory effects of IVIg may involve miR-146a negative feedback loop regulation of NF-κB activity. Disclosures: No relevant conflicts of interest to declare.


PeerJ ◽  
2020 ◽  
Vol 8 ◽  
pp. e9533 ◽  
Author(s):  
Zhiyu Wang ◽  
Yanfei Wang ◽  
Prachi Vilekar ◽  
Seung-Pil Yang ◽  
Mayuri Gupta ◽  
...  

The novel coronavirus SARS-CoV-2 has become a global health concern. The morbidity and mortality of the potentially lethal infection caused by this virus arise from the initial viral infection and the subsequent host inflammatory response. The latter may lead to excessive release of pro-inflammatory cytokines, IL-6 and IL-8, as well as TNF-α ultimately culminating in hypercytokinemia (“cytokine storm”). To address this immuno-inflammatory pathogenesis, multiple clinical trials have been proposed to evaluate anti-inflammatory biologic therapies targeting specific cytokines. However, despite the obvious clinical utility of such biologics, their specific applicability to COVID-19 has multiple drawbacks, including they target only one of the multiple cytokines involved in COVID-19’s immunopathy. Therefore, we set out to identify a small molecule with broad-spectrum anti-inflammatory mechanism of action targeting multiple cytokines of innate immunity. In this study, a library of small molecules endogenous to the human body was assembled, subjected to in silico molecular docking simulations and a focused in vitro screen to identify anti-pro-inflammatory activity via interleukin inhibition. This has enabled us to identify the loop diuretic furosemide as a candidate molecule. To pre-clinically evaluate furosemide as a putative COVID-19 therapeutic, we studied its anti-inflammatory activity on RAW264.7, THP-1 and SIM-A9 cell lines stimulated by lipopolysaccharide (LPS). Upon treatment with furosemide, LPS-induced production of pro-inflammatory cytokines was reduced, indicating that furosemide suppresses the M1 polarization, including IL-6 and TNF-α release. In addition, we found that furosemide promotes the production of anti-inflammatory cytokine products (IL-1RA, arginase), indicating M2 polarization. Accordingly, we conclude that furosemide is a reasonably potent inhibitor of IL-6 and TNF-α that is also safe, inexpensive and well-studied. Our pre-clinical data suggest that it may be a candidate for repurposing as an inhaled therapy against COVID-19.


Author(s):  
W.G. Jerome ◽  
S. Handt ◽  
R.R. Hantgan

In an effort to reestablish blood flow in blocked coronary arteries, heart attack victims are now routinely treated with thrombolytic (clot busting) drugs. However, thrombolytic therapy fails to restore sufficient flow in approximately 20% of cases. The reasons for these failures are undoubtedly multifactorial and difficult to study in vivo. We have developed an “in vitro” model blood vessel which allows controlled study of factors influencing thrombolysis. In the studies reported here, we investigated the 3-D distribution of HUVEC secreted PAI-1 within the clot using immuno-gold cytochemistry and intermediate voltage electron microscopy (IVEM).Human umbilical vein endothelial cells (HUVEC) were grown on the inside surface of fibronectin-coated glass tubes. The cells are stimulated for 4 hours with 1000 U/ml of TNF-α. Clot formation was initiated by addition of 2.3 μM fibrinogen, 100 nM plasminogen, and then 2 nM thrombin in HEPES buffered Gey's salts + 0.5% human serum albumin. After aging the clot for up to 4 hours (to mimic the time delay between heart attack and treatment), 0.5 nM rt-PA (thrombolytic agent) was perfused through the clot for 10 min. at 5 ml/hr. Formation and dissolution of the clot were monitored by laser light scattering.


Sign in / Sign up

Export Citation Format

Share Document