Regulation of angiopoietin expression by bacterial lipopolysaccharide

2008 ◽  
Vol 294 (5) ◽  
pp. L955-L963 ◽  
Author(s):  
Mahroo Mofarrahi ◽  
Thamir Nouh ◽  
Salman Qureshi ◽  
Loic Guillot ◽  
Dominique Mayaki ◽  
...  

Angiopoietins are ligands for Tie-2 receptors and play important roles in angiogenesis and inflammation. While angiopoietin-1 (Ang-1) inhibits inflammatory responses, angiopoietin-2 (Ang-2) promotes cytokine production and vascular leakage. In this study, we evaluated in vivo and in vitro effects of Escherichia coli lipopolysaccharides (LPS) on angiopoietin expression. Wild-type C57/BL6 mice were injected with saline (control) or E. coli LPS (20 mg/ml ip) and killed 6, 12, and 24 h later. The diaphragm, lung, and liver were excised and assayed for mRNA and protein expression of Ang-1, Ang-2, and Tie-2 protein and tyrosine phosphorylation. LPS injection elicited a severalfold rise in Ang-2 mRNA and protein levels in the three organs. By comparison, both Ang-1 and Tie-2 levels in the diaphragm, liver, and lung were significantly attenuated by LPS administration. In addition, Tie-2 tyrosine phosphorylation in the lung was significantly reduced in response to LPS injection. In vitro exposure to E. coli LPS elicited cell-specific changes in Ang-1 expression, with significant induction in Ang-1 expression being observed in cultured human epithelial cells, whereas significant attenuation of Ang-1 expression was observed in response to E. coli LPS exposure in primary human skeletal myoblasts. In both cell types, E. coli LPS elicited substantial induction of Ang-2 mRNA, a response that was mediated in part through NF-κB. We conclude that in vivo endotoxemia triggers functional inhibition of the Ang-1/Tie-2 receptor pathway by reducing Ang-1 and Tie-2 expression and inducing Ang-2 levels and that this response may contribute to enhanced vascular leakage in sepsis.

2020 ◽  
Vol 11 (5) ◽  
Author(s):  
Yi Peng ◽  
Ji-Ling Zhao ◽  
Zhi-Yong Peng ◽  
Wei-Fang Xu ◽  
Guo-Long Yu

Abstract Mesenchymal stem cell (MSC) therapy is a promising approach against myocardial infarction (MI). Studies have demonstrated that MSCs can communicate with other cells by secreting exosomes. In the present study, we aimed to identify exosomal microRNAs that might contribute to MSC-mediated cardioprotective effects. Primary cardiomyocytes were deprived of oxygen and glucose to mimic MI in vitro. For the animal model of MI, the left anterior descending artery was ligated for 1 h, followed by reperfusion for 12 h. MSC-derived exosomes were used to treat primary cardiomyocytes or mice. Cardioprotection-related microRNAs were determined, followed by target gene identification and functional studies with quantitative PCR, western blotting, MTT assay, flow cytometry assay, chromatin immunoprecipitation and dual-luciferase assay. We found that MSC co-culture reduced OGD-induced cardiomyocyte apoptosis and inflammatory responses. Cardioprotection was also observed upon treatment with MSC-derived exosomes in vitro and in vivo. In line with this, exosome uptake led to a significant increase in miR-25-3p in cardiomyocytes. Depletion of miR-25-3p in MSCs abolished the protective effects of exosomes. Mechanistically, miR-25-3p directly targeted the pro-apoptotic genes FASL and PTEN and reduced their protein levels. Moreover, miR-25-3p decreased the levels of EZH2 and H3K27me3, leading to derepression of the cardioprotective gene eNOS as well as the anti-inflammatory gene SOCS3. Inhibition of EZH2 or overexpression of miR-25-3p in cardiomyocytes was sufficient to confer cardioprotective effects in vitro and in vivo. We concluded that exosomal miR-25-3p from MSCs alleviated MI by targeting pro-apoptotic proteins and EZH2.


2007 ◽  
Vol 75 (4) ◽  
pp. 1895-1903 ◽  
Author(s):  
Jeffrey Tessier ◽  
Candace Green ◽  
Diana Padgett ◽  
Wei Zhao ◽  
Lawrence Schwartz ◽  
...  

ABSTRACT Bacillus anthracis edema toxin (ET), composed of protective antigen and an adenylate cyclase edema factor (EF), elicits edema in host tissues, but the target cells and events leading from EF-mediated cyclic-AMP production to edema are unknown. We evaluated the direct effect of ET on several cell types in vitro and tested the possibility that mediators of vascular leakage, such as histamine, contribute to edema in rabbits given intradermal ET. ET increased the transendothelial electrical resistance of endothelial monolayers, a response that is mechanistically inconsistent with the in vivo vascular leakage induced by ET. Screening of several drugs by intradermal treatment prior to toxin injection demonstrated reduced ET-induced vascular leakage with a cyclo-oxygenase inhibitor (indomethacin), agents that interfere with histamine (pyrilamine or cromolyn), or a neurokinin antagonist (spantide). Systemic administration of indomethacin or celecoxib (cyclo-oxygenase inhibitors), pyrilamine, aprepitant (a neurokinin 1 receptor antagonist), or indomethacin with pyrilamine significantly reduced vascular leakage associated with ET. Although the effects of pyrilamine, cromolyn, or aprepitant on ET-induced vascular leakage suggest a possible role for mast cells (MC) and sensory neurons in ET-induced edema, ET did not elicit degranulation of human skin MC or substance P release from NT2N cells in vitro. Our results indicate that ET, acting indirectly or directly on a target yet to be identified, stimulates the production/release of multiple inflammatory mediators, specifically neurokinins, prostanoids, and histamine. These mediators, individually and through complex interactions, increase vascular permeability, and interventions directed at these mediators may benefit hosts infected with B. anthracis.


2007 ◽  
Vol 75 (11) ◽  
pp. 5148-5157 ◽  
Author(s):  
Laetitia Breuilh ◽  
François Vanhoutte ◽  
Josette Fontaine ◽  
Caroline M. W. van Stijn ◽  
Isabelle Tillie-Leblond ◽  
...  

ABSTRACT Galectin-3 (Gal-3) is a multifunctional β-galactoside-binding lectin that senses self-derived and microbial glycoconjugates. Although Gal-3 is important in immune reactions and host defense in some experimental models, the function of Gal-3 during helminthic diseases (e.g., schistosomiasis) is still elusive. We show that, compared to wild-type Schistosoma mansoni-infected mice, infected Gal-3−/− mice have a reduced number of T and B lymphocytes in the spleen, develop reduced liver granulomas at 7 weeks (acute phase) and 14 weeks (chronic phase) postinfection, and mount a biased cellular and humoral Th1 response. In an attempt to understand this latter phenomenon, we studied the role of endogenous Gal-3 in dendritic cells (DCs), the most potent antigen-presenting cells, both in vitro and in vivo. Although Gal-3 deficiency in DCs does not impact their differentiation and maturation processes, it greatly influences the strength (but not the nature) of the adaptive immune response that they trigger, suggesting that Gal-3 deficiency in some other cell types may be important during murine schistosomiasis. As a whole, this study implies that Gal-3 is a modulator of the immune/inflammatory responses during helminthic infection and reveals for the first time that Gal-3 expression in DCs is pivotal to control the magnitude of T-lymphocyte priming.


2008 ◽  
Vol 295 (2) ◽  
pp. F446-F453 ◽  
Author(s):  
Erika I. Boesen ◽  
Jennifer M. Sasser ◽  
Mohamed A. Saleh ◽  
William A. Potter ◽  
Mandy Woods ◽  
...  

The inflammatory cytokines IL-1β and IL-6 have been shown to stimulate production of endothelin-1 (ET-1) by several cell types in vitro, but their effects on renal ET-1 production in vivo are not known. To test whether IL-1β and IL-6 stimulate renal ET-1 production and release in vivo, urine was collected from male C57BL/6 mice over 24-h periods at baseline and on days 7 and 14 of a 14-day subcutaneous infusion of IL-1β (10 ng/h), IL-6 (16 ng/h), or vehicle. By day 14, plasma ET-1 was significantly increased by IL-1β infusion (1.7 ± 0.1 vs. 0.8 ± 0.1 pg/ml for vehicle, P < 0.001). Compared with vehicle infusion, IL-1β infusion induced significant increases in urinary ET-1 excretion rate and urine flow but did not affect conscious mean arterial pressure (telemetry). IL-1β infusion significantly increased renal cortical and medullary IL-1β content (ELISA) and prepro-ET-1 mRNA expression (quantitative real-time PCR). In contrast, 14 days of IL-6 infusion had no significant effect on plasma ET-1 or urinary ET-1 excretion rate. To determine whether IL-1β stimulates ET-1 release via activation of NF-κB, inner medullary collecting duct (IMCD-3) cells were incubated for 24 h with IL-1β, and ET-1 release and NF-κB activation were measured (ELISA). IL-1β activated NF-κB and increased ET-1 release in a concentration-dependent manner. The effect of IL-1β on ET-1 release could be partially inhibited by pretreatment of IMCD-3 cells with an inhibitor of NF-κB activation (BAY 11-7082). These results indicate that IL-1β stimulates renal and systemic ET-1 production in vivo, providing further evidence that ET-1 participates in inflammatory responses.


2021 ◽  
Vol 14 ◽  
Author(s):  
Kai Zhang ◽  
Qingyao Wang ◽  
Yiyao Liang ◽  
Yu Yan ◽  
Haiqiong Wang ◽  
...  

Nerve injury induces profound and complex changes at molecular and cellular levels, leading to axonal self-destruction as well as immune and inflammatory responses that may further promote neurodegeneration. To better understand how neural injury changes the proteome within the injured nerve, we set up a mouse model of sciatic nerve injury (SNI) and conducted an unbiased, quantitative proteomic study followed by biochemical assays to confirm some of the changed proteins. Among them, the protein levels of ADP-dependent glucokinase (ADPGK) were significantly increased in the injured sciatic nerve. Further examination indicated that ADPGK was specifically expressed and upregulated in macrophages but not neurons or Schwann cells upon injury. Furthermore, culturing immortalized bone marrow-derived macrophages (iBMDMs) in vitro with the conditioned media from transected axons of mouse dorsal root ganglion (DRG) neurons induced ADPGK upregulation in iBMDMs, suggesting that injured axons could promote ADPGK expression in macrophages non-cell autonomously. Finally, we showed that overexpression of ADPGK per se did not activate macrophages but promoted the phagocytotic activity of lipopolysaccharides (LPS)-treated macrophages. Together, this proteomic analysis reveals interesting changes of many proteins within the injured nerve and our data identify ADPGK as an important in vivo booster of injury-induced macrophage phagocytosis.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 1256-1256 ◽  
Author(s):  
Angelica A. Silveira ◽  
Clare Cunningham ◽  
Emma Corr ◽  
Wilson Alves Ferreira ◽  
Fernando F. Costa ◽  
...  

Abstract Intravascular hemolysis results in the release of damaging hemoglobin and free heme into the circulation. A role for heme as a danger associated molecular pattern (DAMP), with a function in sterile inflammatory responses, is becoming increasingly recognized. Whilst heme has known effects on leukocytes, activating their migration, adhesion molecule expression and cytokine expression, more recent data demonstrate that this molecule can induce NLRP3 inflammasome formation in murine bone marrow macrophages, with consequent interleukin (IL)-1β processing and neutrophil recruitment (Dutra et al., Proc. Natl Acad Sci. 111: E4110, 2014). We aimed to investigate whether heme can also induce inflammasome activation in primary human macrophages (hMACs) and to further characterize the pathways by which heme-induced inflammatory responses may be amplified under sterile conditions. CD14+ cells were separated from human peripheral blood (using anti-CD14 magnetic beads) and differentiated into hMACs under M-CSF media supplementation and in the presence of 10% fetal bovine serum. In vitro results are expressed as means ± SEM for triplicate cultures and are representative of three independent experiments. Priming of hMACs with lipopolysaccharide (LPS; 100 ng/mL; 3h) alone induced low level secretion of IL-1β (14.11±9.2 pg/106 cells, as measured by ELISA), while heme (50 µM), in the absence of pre-stimulation with LPS, was unable to induce significant IL-1β secretion within 3h (2.46±1.4 pg/106 cells). In contrast, co-incubation of hMACs with both LPS and heme for 3h significantly enhanced hMAC IL-1β release (490.3±36.3 pg/106 cells; P<0.05 compared to LPS alone). The inflamassome pathway inhibitors, MCC950 (5 µM; a specific inhibitor of NLRP3) and YVAD (40 µM; a caspase-1 inhibitor) significantly inhibited IL-1β secretion in LPS-primed hMACs stimulated with heme (reduced to 35.12±3.9; 184±30.4 pg/106 cells, respectively; 3h; P<0.05 compared to LPS/heme). Co-incubation of the LPS-primed cells with varying concentrations of heme, under the conditions employed, did not induce TNF-α production (data not shown), consistent with the hypothesis that IL-1β processing in heme-induced LPS-primed hMAC was mediated by inflammasome formation. Interestingly, qPCR showed that incubation of hMACs (1x106 cells/mL) with heme (50 µM) for 24h stimulated an approximately 10-fold increase (P<0.01) in the expression of the gene encoding, S100A8, another DAMP known to act as a TLR-4 agonist and to contribute to ischemia/reperfusion injury. Priming of hMACs with 1 µg/ml recombinant S100A8 for 3h and subsequent activation with heme (50 or 100 µM, 14h) significantly augmented the release of IL-1β (42.1±0.4 and 89.4±32.4 pg/106 cells for 50 and 100 µM heme, respectively; P<0.05), compared with S100A8 alone (20.6±3.5 pg/106 cells), without any modulation in TNF-α secretion (P>0.05). Using a model of acute intravascular hemolysis, we confirmed an association between heme release and S100A8 secretion, in vivo. Plasma heme levels increased significantly from 26.3±5 µM (i.v. saline control; N=4) to 87±18 µM in C57BL/6 mice at 1h after receiving i.v. water (150 µl; N=4, P=0.04). A concomitant increase in plasma S100A8 levels was also observed within 1h of the hemolytic stimulus (986±102 pg/mL, compared to 694.2±102 pg/ml in control mice; N=4, P=0.05), which was maintained for 3h (P<0.05). Thus, we present data to demonstrate that heme can induce IL-1β processing in LPS-primed human macrophages under in vitro conditions, probably via formation of the NLRP3/caspase-1 inflammasome machinery. In the absence of LPS, heme-stimulated hMACs can express the S100A8 DAMP; furthermore, a hemolytic stimulus induced mouse S100A8 production in vivo. As such, S100A8 may amplify heme-dependent inflammasome formation in an autocrine fashion, even under sterile conditions. Data provide new insights into the mechanisms by which heme may induce and potentiate inflammatory responses in hemolytic diseases, such as sickle cell disease, and suggest S100A8, together with heme, as potential therapeutic targets for reducing inflammation in these diseases. Disclosures Ferreira: Bayer AG: Research Funding. Almeida:Jassen & Cilag: Other: Currently employed with. Conran:Bayer AG: Research Funding.


2005 ◽  
Vol 187 (16) ◽  
pp. 5782-5789 ◽  
Author(s):  
Junsang Ko ◽  
Insook Kim ◽  
Seokho Yoo ◽  
Bumchan Min ◽  
Kyungmin Kim ◽  
...  

ABSTRACT Methylglyoxal (MG) is a toxic metabolite known to accumulate in various cell types. We detected in vivo conversion of MG to acetol in MG-accumulating Escherichia coli cells by use of 1H nuclear magnetic resonance (1H-NMR) spectroscopy. A search for homologs of the mammalian aldo-keto reductases (AKRs), which are known to exhibit activity to MG, revealed nine open reading frames from the E. coli genome. Based on both sequence similarities and preliminary characterization with 1H-NMR for crude extracts of the corresponding mutant strains, we chose five genes, yafB, yqhE, yeaE, yghZ, and yajO, for further study. Quantitative assessment of the metabolites produced in vitro from the crude extracts of these mutants and biochemical study with purified AKRs indicated that the yafB, yqhE, yeaE, and yghZ genes are involved in the conversion of MG to acetol in the presence of NADPH. When we assessed their in vivo catalytic activities by creating double mutants, all of these genes except for yqhE exhibited further sensitivities to MG in a glyoxalase-deficient strain. The results imply that the glutathione-independent detoxification of MG can occur through multiple pathways, consisting of yafB, yqhE, yeaE, and yghZ genes, leading to the generation of acetol.


2002 ◽  
Vol 80 (10) ◽  
pp. 1015-1021 ◽  
Author(s):  
Ju-Feng Wang ◽  
Xinhua Yan ◽  
Jiangyong Min ◽  
Matthew F Sullivan ◽  
Thomas G Hampton ◽  
...  

Cocaine has been shown to depress myocardial function, which may be linked to abnormal Ca2+ handling by the sarcoplasmic reticulum (SR). To examine whether cocaine affects Ca2+-handling proteins and myocardial performance, we injected BALB/c mice with cocaine daily (30 mg/kg, i.p.) for 14 d. Sarcoplasmic reticulum Ca2+-ATPase (SERCA2a) levels, phospholamban (PLB) protein levels, and hemodynamic parameters were measured. After cocaine exposure, myocardial function was significantly decreased both in vivo and in vitro. Also, SERCA2a protein levels were significantly decreased in all cocaine-treated hearts (p < 0.05 compared with saline control). Normalized SERCA2a levels were 1.2 ± 0.2 (densitometric units) in the cocaine groups (p < 0.05 compared with saline control). However, there was no statistical difference in PLB protein levels between the cocaine and the saline groups. In isolated papillary muscle studies, cocaine did not block the response to extracellular Ca2+ but it did prolong the relaxation time of the muscle. These results indicate that cocaine does not block extracellular Ca2+ entrance across the cell membrane, but that it decreases SERCA2a protein levels. In conclusion, our study demonstrates that cocaine decreases SERCA2a protein levels and depresses myocardial function.Key words: cocaine, SERCA2a, phospholamban, ventricular function, mouse.


2016 ◽  
Vol 64 (4) ◽  
pp. 963.1-963
Author(s):  
E Letsiou ◽  
H Wang ◽  
P Belvitch ◽  
S Dudek ◽  
S Sammani

IntroductionAcute lung injury (ALI) and its more severe form, the Acute Respiratory Distress Syndrome (ARDS), are serious conditions resulting from direct or indirect lung injury that occur in critically ill patients and are associated with an unacceptable mortality of up to 40%. A key biological event in the pathogenesis of ALI/ARDS is the dysfunction of the lung endothelium (EC), which is triggered by a variety of inflammatory insults leading to damaged EC, vascular leak, and excessive inflammation. Recently, we demonstrated that an Abl family tyrosine kinase inhibitor, imatinib, protects against LPS-induced endothelial dysfunction by inhibiting c-Abl kinase through mechanisms that remain largely unknown. In the present study, we identified parkin, a novel c-Abl substrate, as a critical mediator of endothelial dysfunction in ALI.MethodsIn vitro Human pulmonary artery endothelial cells (EC) were transfected with siRNA for parkin and then challenged with LPS (1 µg/ml, 3 hrs). Inflammatory mediators were determined in cell lysates and supernatants by Western blotting and ELISA respectively. In vivo C57BL/6 (WT) and parkin deficient (PARK2 KO) male mice (8–12 wks, n=5–8) were subjected to LPS (intratracheally, 1 mg/kg) or PBS (controls), and allowed to recover prior to harvest 18 hrs later. Leakage of proteins into the alveolar space was assessed by measuring the protein levels in the bronchoalveolar lavage (BAL). To assess lung inflammation, neutrophil cell counts, myeloperoxidase (MPO) activity, and IL-6 levels were determined in BAL.ResultsIn human lung EC, down-regulation of parkin by siRNA reduces LPS-induced VCAM-1 expression (adhesion molecule involved in neutrophil adhesion to EC) (by 35%, p<0.05), IL-8 (neutrophil chemoattractant) (by 59%, p<0.01), and IL-6 (inflammatory cytokine) release (by 79%, p<0.01). PARK2 KO mice exhibit less ALI after LPS compared to WT. In PARK2 KO, BAL protein levels were reduced by 27% (p=0.0024) compared to WT mice. LPS-induced neutrophil recruitment into the alveoli of PARK2 KO was attenuated by 47% compared to WT (p=0.0019). BAL MPO activity (marker of neutrophil activation) and BAL IL-6 levels were also significantly lower in PARK2 KO by 52% (p=0.03) and 28% (p=0.0061) respectively.ConclusionThese results suggest that endothelial parkin mediates EC activation and neutrophil adhesion/migration after LPS, and therefore it may represent a new potential therapeutic target in ALI/ARDS.


2016 ◽  
Vol 48 (8) ◽  
pp. 626-632 ◽  
Author(s):  
Marina M. Hanson ◽  
Fengming Liu ◽  
Shen Dai ◽  
Alison Kearns ◽  
Xuebin Qin ◽  
...  

Effective methods for cell ablation are important tools for examining the anatomical, functional, and behavioral consequences of selective loss of specific cell types in animal models. We have developed an ablation system based on creating genetically modified animals that express human CD59 (hCD59), a membrane receptor, and administering intermedilysin (ILY), a toxin produced by Streptococcus intermedius, which binds specifically to hCD59 to induce cell lysis. As proof-of-concept in the rat, we generated an anemia model, SD-Tg(CD59-HBA1)Bryd, which expresses hCD59 on erythrocytes. Hemolysis is a common complication of inherited or acquired blood disorders, which can result in cardiovascular compromise and death. A rat model that can replicate hemolysis through specific ablation of erythrocytes would allow further study of disease and novel treatments. In vitro, complete lysis of erythrocytes expressing hCD59 was observed at and above 250 pM ILY, while no lysis was observed in wild-type erythrocytes at any ILY concentration (8-1,000 pM). In vivo, ILY intravenous injection (100 ng/g body wt) dramatically reduced the hematocrit within 10 min, with a mean hematocrit reduction of 43% compared with 1.4% in the saline control group. Rats injected with ILY at 500 ng/g intraperitoneally developed gross signs of anemia. Histopathology confirmed anemia and revealed hepatic necrosis, with microthrombi present. These studies validate the hCD59-ILY cell ablation technology in the rat and provide the scientific community with a new rapid conditional targeted ablation model for hemolytic anemia and hemolysis-associated sequelae.


Sign in / Sign up

Export Citation Format

Share Document