Inhibition of endothelial cell activation by bHLH protein E2-2 and its impairment of angiogenesis

Blood ◽  
2010 ◽  
Vol 115 (20) ◽  
pp. 4138-4147 ◽  
Author(s):  
Aya Tanaka ◽  
Fumiko Itoh ◽  
Koichi Nishiyama ◽  
Toshiaki Takezawa ◽  
Hiroki Kurihara ◽  
...  

E2-2 belongs to the basic helix-loop-helix (bHLH) family of transcription factors. E2-2 associates with inhibitor of DNA binding (Id) 1, which is involved in angiogenesis. In this paper, we demonstrate that E2-2 interacts with Id1 and provide evidence that this interaction potentiates angiogenesis. Mutational analysis revealed that the HLH domain of E2-2 is required for the interaction with Id1 and vice versa. In addition, Id1 interfered with E2-2–mediated effects on luciferase reporter activities. Interestingly, injection of E2-2–expressing adenoviruses into Matrigel plugs implanted under the skin blocked in vivo angiogenesis. In contrast, the injection of Id1-expressing adenoviruses rescued E2-2–mediated inhibition of in vivo angiogenic reaction. Consistent with the results of the Matrigel plug assay, E2-2 could inhibit endothelial cell (EC) migration, network formation, and proliferation. On the other hand, knockdown of E2-2 in ECs increased EC migration. The blockade of EC migration by E2-2 was relieved by exogenous expression of Id1. We also demonstrated that E2-2 can perturb VEGFR2 expression via inhibition of VEGFR2 promoter activity. This study suggests that E2-2 can maintain EC quiescence and that Id1 can counter this effect.

Hypertension ◽  
2020 ◽  
Vol 76 (Suppl_1) ◽  
Author(s):  
Justin P Van Beusecum ◽  
Natalia R Barbaro ◽  
Charles D Smart ◽  
David M Patrick ◽  
Cyndya A Shibao ◽  
...  

We have shown that dendritic cells (DCs) from hypertensive mice convey hypertension when adoptively transferred to recipients. Recently a novel subset of DCs in humans that express Axl and Sigelc-6 + (AS DCs) have been identified which drive T cell proliferation and produce IL-1β, IL-6 and IL-23, consistent with DCs we have observed in hypertension. We hypothesized that AS cells are increased in hypertension and contribute to immune activation in this disease. We quantified circulating AS DCs by flow cytometry in normotensive (n=23) and hypertensive (n=11) subjects and found a more than 2-fold increase in circulating AS DCs in hypertensive compared to normotensive subjects (297 ± 73 vs. 108 ± 26/ml; p =0.0304). To investigate the mechanism by which AS DCs are formed in hypertension, we co-cultured human aortic endothelial cells (HAECs) undergoing either normotensive (5%) or hypertensive (10%) cyclical stretch for 48 hours with CD14 + monocytes from normotensive donors. Co-culture of monocytes with HAECs exposed to 10% stretch significantly increased AS DCs and AS DC IL-1β production when compared to 5% stretch alone as assessed by flow cytometry (21 ± 5 vs. 131 ± 32 IL-1β + AS DCs). Moreover, inhibition of Axl signaling with R248, completely abolished the production of IL-1β in AS DCs (34 ± 8 IL-1β + AS DCs). In additional experiments we found that 10% stretch caused a 50% increase in release of growth arrest 6 (GAS6), the ligand for Axl, from HAECs compared to 5% stretch. Treatment of human monocytes with GAS6 mimicked the effect of 10% stretch in promoting AS cell formation and IL-1β production. Based on the increased secretion of GAS6 from HAECs, we used a J-wire to harvest human endothelial cells from 23 additional volunteers to assess endothelial cell activation and GAS6 secretion in vivo. We found a positive association between pulse pressure and plasma GAS6 (R 2 =0.25, p =0.0079) and a striking positive association between GAS6 and ICAM-1 (R 2 =0.39, p =0.0012). These data show that secretion of GAS6 by an activated endothelial seems to promote the formation and activation of AS DCs. Thus, the interplay between endothelial-derived GAS6 and AS DCs seem to be an important mechanism in human hypertension and might be a novel therapeutic target for this disease.


1994 ◽  
Vol 72 (01) ◽  
pp. 028-032 ◽  
Author(s):  
P Collins ◽  
A Roderick ◽  
D O’Brien ◽  
E Tuddenham ◽  
A O’Driscoll ◽  
...  

SummaryHepatic venocclusive disease causes considerable morbidity and mortality following bone marrow transplantation. There are two hypotheses regarding the aetiology of this syndrome; firstly that changes in plasma coagulation factors and natural anticoagulants lead to a prothrombotic state and secondly that endothelial cell activation stimulates intravascular deposition of fibrin. We have investigated these mechanisms by measuring the changes in proteins C and S and factors VII and X in the post transplant period and by using the plasma concentration of factor Vila as an in vivo marker of potential endothelial cell tissue factor expression. Protein C fell in both allograft and autograft patients but more so in the allografts. Similar results were found for factors VII and X. These changes were predominantly due to hepatic dysfunction induced by the chemo-radiotherapy. Factor Vila levels were unchanged in both the allograft and autograft patients. We conclude that there is no convincing evidence for a procoagulant state following BMT as there are both anticoagulant and procoagulant changes. The absence of any changes in factor Vila levels suggests that tissue factor was not exposed to the general circulation following BMT but does not exclude focal expression at the sites of thrombosis.


Cancers ◽  
2020 ◽  
Vol 12 (4) ◽  
pp. 1040
Author(s):  
Mitra Nair ◽  
Maninder Khosla ◽  
Yoshihiro Otani ◽  
Margaret Yeh ◽  
Flora Park ◽  
...  

Vascularization is a common pathology for many solid tumors, and therefore anti-angiogenic strategies are being investigated as a therapeutic target for treatment. Numerous studies are also being conducted regarding the effects of oncolytic viruses, including ImlygicTM, an FDA approved oncolytic herpes simplex virus-1 (oHSV) for the treatment of highly vascularized tumors such as Kaposi sarcoma (NCT04065152), and brain tumors. To our knowledge, the effects of combining oncolytic HSV with angiogenesis inhibition on endothelial cell activation has not been previously described. Here, we tested the effects of Rapid Antiangiogenesis Mediated By Oncolytic Virus (RAMBO), an oHSV which expresses a potent anti-angiogenic gene Vasculostatin on endothelial cell activation in heavily vascularized solid tumors. oHSV treatment induces endothelial cell activation, which inhibits virus propagation and oncolysis in adjacent tumor cells in vitro. Consistently, this was also observed in intravital imaging of intracranial tumor-bearing mice in vivo where infected tumor endothelial cells could efficiently clear the virus without cell lysis. Quantitative real-time PCR (Q-PCR), leukocyte adhesion assay, and fluorescent microscopy imaging data, however, revealed that RAMBO virus significantly decreased expression of endothelial cell activation markers and leukocyte adhesion, which in turn increased virus replication and cytotoxicity in endothelial cells. In vivo RAMBO treatment of subcutaneously implanted sarcoma tumors significantly reduced tumor growth in mice bearing sarcoma compared to rHSVQ. In addition, histological analysis of RAMBO-treated tumor tissues revealed large areas of necrosis and a statistically significant reduction in microvessel density (MVD). This study provides strong preclinical evidence of the therapeutic benefit for the use of RAMBO virus as a treatment option for highly vascularized tumors.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 133-133
Author(s):  
Silvia S. Pierangeli ◽  
Mariano E. Vega-Ostertag ◽  
Elena Raschi ◽  
Xiaowei Liu ◽  
Maria O. Borghi ◽  
...  

Abstract Background: Antiphospholipid antibodies (aPL) are associated with thrombosis and pregnancy loss in patients with Antiphospholipid Syndrome (APS). aPL and bacterial lipopolysaccharide (LPS) bind to and activate endothelial cells (EC) through NFκB and p38 MAPK pathways. Recent studies suggest that aPL might interact with toll-like receptor-4 (TLR-4), the receptor for LPS. Aim: to investigate the role of TLR-4 in antiphospholipid syndrome (APS). Methods: we examined: i) the aPL effects on thrombosis and EC activation in LPS non-responsive (LPS−/−) mice that display a spontaneous mutation of TLR-4 vs LPS-responsive (LPS+/+) mice displaying wild type TLR-4, ii) the prevalence of TLR-4 Asp299gly and Thr399Ile polymorphisms - both associated with decreased response to LPS - in 110 APS patients (with arterial and/or venous thrombosis) vs 220 controls (of same ethnic background). IgGs were purified from a patient with APS (IgG-APS) and from control serum (IgG-NHS). LPS −/− and LPS +/+ mice, in groups of nine, were treated with IgG-APS or with IgG-NHS twice intraperitoneally. Size of induced thrombi and # of leukocyte (WBC) adhering to endothelial cells in the microcirculation of endothelium of the cremaster muscle of mice (as a means to measure endothelial cell activation) were determined in vivo, seventy-two hours after the first injection. TLR-4 Asp299gly & Thr399Ile polymorphisms were evaluated by Allele-Specific PCR. Results: LPS +/+ mice treated with IgG-APS produced significantly larger thrombi when compared to mice treated with IgG-NHS (2166 ± 1419 μm2 vs 1176 ± 841 μm2) and significantly larger number of WBC adherence to ECs (4.5 ± 1.9 vs 2.2 ±1.1). Thrombus size and number of adhering WBC to ECs were significantly lower in LPS −/− mice treated with IgG-APS compared to LPS +/+ mice treated with IgG-APS [thrombus size: 779 ± 628 μm2 vs 2166 ± 1419 μm2 (p<0.05) and number of adherent WBC to EC: 1.0 ± 0.5 vs 4.5 1.9 (p<0.05)], respectively. The titer of anticardiolipin antibodies in the sera of mice injected with aPL was 48.2 ± 17.1 GPL (for LPS −/− mice) and 50.8 ± 11.2 GPL (for LPS +/+ mice), respectively (NS). A significant reduction in TLR-4 Asp 299gly & Thr399Ile polymorphisms was observed in APS patients (5%) compared to controls (11.4%) (p<0.05). Conclusions: These findings strongly suggest that TLR-4 is involved in aPL interaction with endothelial cells and mediates their pathogenic effects.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 294-294
Author(s):  
Ben T. Atkinson ◽  
Prathima Nandivada ◽  
Bruce Furie ◽  
Barbara C. Furie

Abstract The endothelium serves as a metabolically active interface between the blood and underlying tissues and offers response to changes in its microenvironment. The endothelium is rapidly activated in response to stimuli associated with fluctuations in shear stress, physical trauma, oxidative stress, and thrombotic and inflammatory mediators. To study endothelial cell activation we have monitored calcium mobilization in vitro in cultured endothelial cells and also in situ in the living mouse, following vessel wall injury. The addition of exogenous ADP (10 μM) or thrombin (1 U/mL) to Human Umbilical Vein Endothelial Cells (HUVECs) loaded with the calcium sensitive dye, Fluo-4 AM, is followed by rapid elevation of intracellular calcium, with a sustained peak observed within 10 sec. To further investigate the ability of the endothelium to activate in response to mediators potentially localized in the microenvironment of the endothelium during thrombus formation, we investigated the ability of IL-6 (0.1 ng/mL), IL-8 (80 ng/mL), MCP-1 (15 ng/mL), NAP-2 (10 ng/mL) and SDF-α (60 ng/mL) to mobilize calcium in HUVECs. These potential agonists were selected based on their primary roles in inflammation and thrombosis, and proposed roles in atherosclerosis and tumor angiogenesis. SDF-α and NAP-2 rapidly mobilized calcium in HUVECs, with similar maximum responses, but NAP-2 initiated a more prolonged (4-fold longer) rise in intracellular calcium. IL-6, IL-8, and MCP-1 also mobilized calcium, but the rise in intracellular calcium was almost 4-fold lower than that observed with SDF-α and NAP-2. Comparable calcium mobilization occurs in HUVECs subjected to a single pulse of a nitrogen dye-tuned laser. In addition, targeting of single cells within a confluent culture of endothelial cells initiated calcium elevation in the targeted cell and was followed by a wave of calcium elevation in surrounding cells. To determine whether this endothelial cell activation, and more specifically the calcium elevation, occurs in vivo, we conducted experiments using the laser-induced model of thrombus formation to look for calcium elevation in the arteriolar endothelium in live mice. Calcium elevation was monitored by Fluo-4AM introduced intravenously into the mouse circulation via the jugular vein. Fluo-4AM is non-specific in its uptake among cell types, and endothelial cell uptake of the dye in the cremaster muscle microcirculation was observed. Upon laser-induced injury, calcium elevation in the endothelium could be monitored by intravital microscopy. The observed calcium elevation was rapid (within 30 s) and preceded detection of platelets in the developing thrombus. These results add to the evidence of a dynamic endothelium and demonstrate that the endothelium activates rapidly prior to thrombus formation in the laser induced thrombosis model.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 5192-5192
Author(s):  
Richard Robins ◽  
Catherine A. Lemarie ◽  
Mark D. Blostein

Abstract Abstract 5192 Forkhead proteins play a broad role in endothelial cell biology. These factors mediate cell adhesion to extracellular matrix, regulate the expression of pro-inflammatory and pro-thrombotic genes, and participate in cell repair, proliferation and apoptosis. FoxOs are known downstream targets of the PI3K/Akt signaling pathway. Phosphorylation of FoxO transcription factors results in their translocation from the nucleus to the cytoplasm, thereby inhibiting their transcriptional activity. It has recently been shown that the deletion of the three FoxO isoforms in endothelial cells protects mice from vascular dysfunction. Gas6, a member of the vitamin K-dependent family of proteins, has been shown to protect endothelial cells from apoptosis and promote endothelial cell activation in vivo. It has been shown that the expression of ICAM-1 and VCAM-1 were blunted in the absence of gas6. Interestingly, a role for VCAM-1 in the pathogenesis of venous thrombosis has been proposed. Elevated levels of the soluble form of VCAM-1 have been detected in the serum of patients with venous thrombosis. We previously demonstrated that the anti-apoptotic effect of gas6 was mediated partially through FoxO1, but overall, the signalling mechanisms occurring downstream of gas6 remain largely unknown. We hypothesize that gas6 promotes thrombin-induced VCAM-1 expression through the regulation of FoxO1 in endothelial cells. Western blot analysis demonstrated that thrombin induced time dependent phosphorylation of FoxO1 with a maximum at 30 minutes in WT (p<0. 05) but not in gas6 deficient (−/−) cells. In addition, thrombin reduced the nuclear content of FoxO in WT (p<0. 05) but not in gas6−/− endothelial cells. Using qPCR, we found that mRNA expression of VCAM-1 was increased after 30 minutes of stimulation with thrombin in WT cells (p<0. 05). More importantly, thrombin-mediated induction of VCAM-1 was blunted in gas6−/− endothelial cells. We found that FoxO1 siRNA increased basal VCAM-1 expression in WT endothelial cells. Taken together, our data demonstrate that gas6 is a crucial mediator of FoxO1 that regulates thrombin-induced VCAM-1 expression. This pathway may explain the pro-thrombotic and pro-inflammatory role of gas6. Disclosures: No relevant conflicts of interest to declare.


2013 ◽  
Vol 32 (2) ◽  
pp. 154-161 ◽  
Author(s):  
Paul O. Zamora ◽  
Yi Liu ◽  
Henry Guo ◽  
Xinhua Lin

The biocompatibility/inflammation profile of B2A-coated ceramic granules was evaluated using a panel of standard biocompatibility protocols (International Organization for Standardization-10993) including skin irritation and delayed-type hypersensitivity (Kligman maximization test), as well as acute, subacute, and chronic toxicity. Additionally, the potential of B2A-coated granules to elicit inflammatory reactions was also assessed using in vivo air pouch models, and B2A was evaluated using in vitro models of leukocyte recruitment and endothelial cell activation. Overall, the findings demonstrate that B2A-coated ceramic granules exhibit good biocompatibility profiles in the murine air pouch model and in standard subcutaneous implant models, and B2A did not demonstrate evidence of leukocyte recruitment or endothelial cell activation. These findings suggest that B2A and B2A-coated granules have little, if any, propensity to initiate inflammation reactions based on leukocyte recruitment. Thus, traditional biocompatibility and specially designed inflammation models indicate a high degree of biocompatibility and a low possibility of toxicity, inflammation, or edema following the implant of B2A-coated granules.


Author(s):  
Indranil Biswas ◽  
Sumith R Panicker ◽  
Hemant Giri ◽  
Xiaofeng S Cai ◽  
Alireza R Rezaie

Plasmodium falciparum (Pf)-derived histidine-rich protein II (HRPII) has been shown to inhibit heparin-dependent anticoagulant activity of antithrombin (AT) and induce inflammation in vitro and in vivo. In a recent study, we showed that HRPII interacts with the AT-binding vascular glycosaminoglycans (GAGs) to not only disrupt the barrier-permeability function of endothelial cells but also inhibit the anti-inflammatory signaling function of AT. Here we investigated the mechanisms of the pro-inflammatory function of HRPII and the protective activity of AT in cellular and animal models. We found that AT competitively inhibits the GAG-dependent HRPII-mediated activation of NF-κB and expression of intercellular cell adhesion molecule 1 (ICAM1) in endothelial cells. Furthermore, AT inhibits HRPII-mediated histone H3 citrullination and neutrophil extracellular trap (NET) formation in HL60 cells and freshly isolated human neutrophils. In vivo, HRPII induced Mac1 expression on blood neutrophils, MPO release in plasma, neutrophil infiltration and histone H3 citrullination in the lung tissues. HRPII also induced endothelial cell activation as measured by increased ICAM1 expression and elevated vascular permeability in the lungs. AT effectively inhibited HRPII-mediated neutrophil infiltration, NET formation and endothelial cell activation in vivo. AT also inhibited HRPII-meditated deposition of platelets and fibrin(ogen) in the lungs and circulating level of von Willebrand factor in the plasma. We conclude that AT exerts protective effects against pathogenic effects of Pf-derived HRPII in both cellular and animal models.


2008 ◽  
Vol 20 (9) ◽  
pp. 112
Author(s):  
Q. Chen ◽  
C. Viall ◽  
P. R. Stone ◽  
L. W. Chamley

Preeclampsia is characterised by elevated maternal blood pressure which is preceded by endothelial activation. The cause of this endothelial cell dysfunction is unclear but it appears to be triggered by a placental factor. One of the risk factors for developing preeclampsia is the presence of antiphospholipid antibodies (aPL) in the maternal blood but exactly how aPL predispose women to developing preeclampsia is unclear. A second feature known to be associated with preeclampsia is excessive shedding and deportation of dead trophoblasts. We have previously shown that shed trophoblasts are phagocytosed by endothelial cells and that phagocytosis of necrotic trophoblasts leads to endothelial cell activation1. In this study we examined the hypothesis that aPL alter the number or nature of trophoblasts shed from the placenta resulting in endothelial cell activation. Using our published model of trophoblast shedding 2 human first trimester placental explants were treated with monoclonal aPL, IIC5 or ID2, or control antibody CD45 for 72 h. Shed trophoblasts then were harvested and counted using a Cellometer AutoT4 automated cell counter. The activity of caspases 3&7 was analysed in all treated shed trophoblasts using a FLICA™ kit. The treated shed trophoblasts also were exposed to the endothelial cell line HMEC-1 for 24 h. The level of ICAM-1 by HMEC-1 was determined by cell-based ELISA. The number of trophoblasts shed from placental explants was increased 2 fold following aPL treatment whereas, treatment with CD45 resulted in only a 1.3 fold increase in shedding. Trophoblasts shed from aPL-treated explants contained less active caspases 3 & 7 compared with control shed trophoblasts. Moreover, phagocytosis of trophoblasts shed from aPL-treated explants induced significantly increased expression of ICAM-1 compared with controls. aPL treatment affected the number and nature of trophoblasts shed from placentae in such a way that phagocytosing endothelium become activated. These findings suggest that aPL treatment may have shifted the type of cell death that shed trophoblasts are undergoing from apoptosis to a more necrotic or aponecrotic mechanism. This type of shedding of trophoblasts in vivo might contribute to the endothelial cell activation which is a hallmark feature of preeclampsia. (1) Chen Q, Stone PR, McCowan LM et al. Phagocytosis of necrotic but not apoptotic trophoblasts induces endothelial cell activation. Hypertension. 2006;47:116–121. (2) Abumaree MH, Stone PR, Chamley LW. An in vitro model of human placental trophoblast deportation/shedding. Mol Hum Reprod. 2006;12:687–694.


Sign in / Sign up

Export Citation Format

Share Document