Apoptosis of Proliferating Endothelial Cells Induced by Cleaved High Molecular Weight Kininogen (HKa) Involves an Oxidation-Dependent Pathway.

Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 3926-3926
Author(s):  
Danyu Sun ◽  
Keith McCrae

High molecular weight kininogen (HK) is a single-chain glycoprotein that plays a central role in contact activation. We previously reported that the cleaved form of high molecular weight kininogen (HKa), which lacks bradykinin, induces apoptosis of proliferating endothelial cells and inhibits angiogenesis in vivo. This activity involves binding of HKa to tropomyosin exposed on the endothelial cell surface. We have also demonstrated that endothelial cells cultured on types I or IV collagen are endowed with relative resistance to the apoptotic effects of HKa. We hypothesized that one mechanism that might account for this specific effect could be the ability of collagen to inhibit hydroxyl radical induced apoptosis through scavenging of hydroxyl radicals and inhibition of cellular lipid peroxidation. To address this issue, we assessed the ability of HKa to inhibit endothelial cell proliferation in the absence or presence of glutathione (GSH), an intracellular thiol antioxidant which scavenges reactive oxygen species (ROS) and prevents ROS-induced cell damage. The results of these experiments demonstrated that GSH blocked the ability of HKa to inhibit endothelial proliferation and induce endothelial cell apoptosis on all ECM proteins tested, including gelatin, laminin, vitronectin, fibronectin, and collagens I and IV. Similar results were obtained when N-acetylcysteine (NAC), a cell membrane permeable GSH precursor, was employed. However, neither GSH nor NAC protected endothelial cells from apoptosis induced by 2-methoxyestradiol, which also induces selective apoptosis of proliferating endothelial cells, suggesting that the ability of GSH to block HKa-induced endothelial cell apoptosis is specific. To further examine the role of ROS in HKa induced endothelial cell apoptosis, we measured the cellular GSH level and content of lipid peroxidation products MDA and 4-HNE following exposure to HKa. These studies demonstrated that exposure of endothelial cells cultured on gelatin to HKa led to a rapid fall in intracellular GSH, accompanied by a three fold increase in MDA and 4-HNE. In contrast, little change in the levels of oxidation products were observed when cells were cultured on collagen type I or IV. These results demonstrate that the ability of HKa to induce apoptosis of proliferating endothelial cells is associated with the production of reactive oxygen species (ROS) and a change in intracellular redox status. To further test this conclusion, we examined another redox sensitive signaling regulator, thioredoxin (TRX), which normally occurs primarily in the cytoplasm but translocates to the nucleus in response to oxidative stress. In response to HKa, thioredoxin quickly translocated from the cytoplasm to the nucleus of endothelial cells cultured on gelatin, though nuclear translocation of thioredoxin did not occur when cells were cultured on type I collagen. These results suggest that nuclear accumulation of TRX may be intimately involved in HKa induced endothelial cell apoptosis by sensing oxidative stress. In conclusion, our data suggests that HKa induces endothelial cell apoptosis through an oxidation dependent pathway, which is co-regulated through additional signals emanating from the extracellular matrix. Ongoing studies are focused on defining the mechanisms by which ROS are generated, and the role of HKa-tropomyosin interactions in stimulating this oxidant pathway.

Blood ◽  
2006 ◽  
Vol 107 (12) ◽  
pp. 4714-4720 ◽  
Author(s):  
Danyu Sun ◽  
Keith R. McCrae

AbstractHigh–molecular-weight kininogen (HK) is an abundant plasma protein that plays a central role in activation of the kallikrein-kinin system. Cleavage of HK by plasma kallikrein results in release of the nonapeptide bradykinin (BK), leaving behind cleaved high–molecular-weight kininogen (HKa). Previous studies have demonstrated that HKa induces apoptosis of proliferating endothelial cells and inhibits angiogenesis in vivo, activities mediated primarily through its domain 5. However, the mechanisms by which these effects occur are not well understood. Here, we demonstrate that HKa induces apoptosis of endothelial cells cultured on gelatin, vitronectin, fibronectin, or laminin but not collagen type I or IV. The ability of HKa to induce endothelial-cell apoptosis is dependent on the generation of intracellular reactive oxygen species and associated with depletion of glutathione and peroxidation of endothelial-cell lipids, effects that occur only in cells cultured on matrix proteins permissive for HKa-induced apoptosis. Finally, the ability of HKa to induce endothelial-cell apoptosis is blocked by the addition of reduced glutathione or N-acetylcysteine. These studies demonstrate a unique role for oxidant stress in mediating the activity of an antiangiogenic polypeptide and highlight the importance of the extracellular matrix in regulating endothelial-cell survival.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 3305-3305
Author(s):  
Venkaiah Betapudi ◽  
Keith R. McCrae

Abstract Abstract 3305 Background and objective: High molecular weight kininogen (HK) is an abundant plasma protein that serves as an important component of the intrinsic pathway of coagulation. HK normally circulates as in the single chain form, but may be cleaved by plasma kallikrein to release the nonapeptide bradykinin, resulting in the formation cleaved high molecular weight kininogen (HKa) that consists of a heavy and light chain linked by a single disulfide bond. Conformational changes occurring after kallikrein cleavage result in increased exposure of histidine and glycine-rich regions with kininogen domain 5 that impart HKa with unique properties, including the ability to inhibit angiogenesis by causing selective apoptosis of proliferating endothelial cells. However, the receptors that mediate the antiangiogenic activity of HKa remain controversial, and the signaling pathways that lead to apoptosis have not been defined. Previous studies suggested possible involvement of SRC family kinases (SFK) in this process, and the purpose of this work was to further define the activation of SFKs and their downstream targets during HKa-induced endothelial cell apoptosis. Results: We first assessed the activation of SFKs in proliferating endothelial cells stimulated with bFGF before and after incubation with HKa (6–20 nM). SFKs are maintained in an inactive state through tyrosine phosphorylation of their C-terminal region mediated by the negative regulator C-terminal Src kinase (Csk). Exposure of endothelial cells to HKa caused downregulation of Csk in a dose-dependent manner within 60 minutes. In parallel, we observed a significant increase in expression of the proapoptotic SFK Lck in endothelial cells exposed to HKa, though expression of other SFKs including Lyn, Fyn, Src, Hck and Blk were not significantly altered. Increased expression of Lck was associated with activation of p53 and increased expression of the pro-apoptotic Bcl-2 family members Bax and Bak. Endothelial cell lysates prepared within 60 minutes of exposure to HKa demonstrated significant increases in the activity of caspases 3 and 7, as well as depletion of DNA fragmentation factors (DFF) 45 and 35, which cleave and inactivate DFF40, a major endonuclease involved in apoptosis. In parallel studies, endothelial cells depleted of Lck by treatment with Lck siRNA displayed loss of p53 phosphorylation, caspase 3 and 7 activity, and expression of Bax and Bad with no effects on the expression of Bad and Bid. Conclusion: These findings demonstrate a critical role for Csk in regulation of SFK activation and endothelial homeostasis, and demonstrate that downregulation of Csk by HKa leads to activation of a Lck-dependent, p53-mediated apoptotic pathway. Increasing the expression of Lck may represent a novel mechanism for regulation of aberrant angiogenesis. Disclosures: No relevant conflicts of interest to declare.


2002 ◽  
Vol 80 (2) ◽  
pp. 85-90 ◽  
Author(s):  
Jing-Chuan Zhang ◽  
Xiaoping Qi ◽  
Jose' Juarez ◽  
Marian Plunkett ◽  
Fernando Donaté ◽  
...  

We recently reported that the two-chain form of human high molecular weight kininogen (HKa) inhibits angiogenesis by inducing endothelial cell apoptosis (Zhang et al. 2000). This property appears to be primarily conferred by HKa domain 5 (HKa D5). In this manuscript, we further characterize the activity of these polypeptides toward proliferating endothelial cells, as well as their in vivo anti-angiogenic activity in the chick chorioallantoic membrane (CAM). We also demonstrate that short peptides derived from endothelial cell binding regions in HKa domains 3 and 5 inhibit endothelial cell proliferation and induce endothelial cell apoptosis. Like HKa and HKa D5, peptides derived from the latter domain induce endothelial cell apoptosis in a Zn2+-dependent manner, while those derived from domain 3 function independently of Zn2+. The implications of these findings to the regulation of angiogenesis and development of anti-angiogenic therapeutics are discussed.Key words: angiogenesis, kininogen, endothelial cells, apoptosis, peptides.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 3937-3937
Author(s):  
Danyu Sun ◽  
Keith R. McCrae

Abstract Previous studies from our laboratory and others have demonstrated that cleaved high molecular weight kininogen (HKa) induces selective apoptosis of proliferating endothelial cells and inhibits angiogenesis. Recently we reported that the induction of endothelial cell apoptosis by HKa was matrix dependent and requires the generation of reactive oxygen species. To further define the mechanisms of HKa-induced endothelial cell apoptosis, we have investigated the pathway through which apoptosis occurs. HKa-induced endothelial cell apoptosis was caspase-dependent, as demonstrated by the cleavage of pro-caspase 3 and the inhibition of apoptosis by the broad spectrum caspase inhibitor z-VAD-fmk. Activation of the intrinsic pathway of apoptosis was also confirmed by demonstrating cleavage of pro-caspase 9 as well as the release of cytochrome C from mitochondria. These results are consistent with our previous work, as apoptosis induced through oxidative stress is a well known activator of the intrinsic apoptotic pathway. Given the resistance to HKa-induced endothelial cell apoptosis conferred by culture of endothelial cells on collagen I or IV, we questioned whether MMPs may be involved in this process. Interestingly, we observed that the broad spectrum MMP inhibitor GM6001 completely protected endothelial cells from HKa-induced apoptosis. Moreover, we observed that HKa-induced apoptosis was associated with PECAM-1 cleavage within 1 hour of exposure, and that inhibition of apoptosis by either GM6001 or z-VAD-fmk also inhibited the cleavage of PECAM-1. These findings are likely to be relevant to the apoptotic pathway, as intact PECAM-1 has been previously reported to inhibit cytochrome c release after exposure of cells to cytotoxic stimuli, while a cleaved cytoplasmic domain of PECAM-1 enhances the susceptibility of cells to apoptosis. In summary, our studies demonstrate that the antiangiogenic activity of HKa may be critically dependent on MMP activation, which in turn leads to ROS generation and caspase activation. The subsequent cleavage of PECAM-1, presumably by caspases, may be another important event in HKa-induced apoptosis.


2000 ◽  
Vol 14 (15) ◽  
pp. 2589-2600 ◽  
Author(s):  
Jing‐Chuan Zhang ◽  
Kevin Claffey ◽  
Ramasamy Sakthivel ◽  
Zbigniev Darzynkiewicz ◽  
David Elliot Shaw ◽  
...  

2020 ◽  
Vol 26 (1) ◽  
Author(s):  
Zhimin Zhang ◽  
Mingzhu Lv ◽  
Xiang Wang ◽  
Zheng Zhao ◽  
Daolong Jiang ◽  
...  

Abstract Background The oncogenic role of the newly identified lncRNA LUADT1 has been revealed in lung adenocarcinoma. It was reported that LUADT1 plays a critical role in multiple human diseases. This study was carried out to investigate the role of LUADT1 in sepsis. Methods Sixty patients with sepsis and sixty healthy volunteers were recruited for this study. Plasma samples were collected from all participants. Human primary coronary artery endothelial cells were also used in this study. The expression of Pim-1, miR-195 and LUADT1 were detected by RT-qPCR. The interaction between miR-195 and LUADT1 was determined by overexpression experiments and luciferase activity assay. Cell apoptosis was detected by flow cytometry. The expression of apoptosis-related protein was detected by Western blotting. Results Bioinformatics analysis revealed the potential interaction between LUADT1 and miR-195, which was confirmed by dual luciferase reporter assay. LUADT1 was downregulated in patients with sepsis. Moreover, LPS treatment downregulated the expression of LUADT1 in primary cardiac endothelial cells. Overexpression of LUADT1 and miR-195 did not affect the expression of each other in primary cardiac endothelial cells. Interestingly, overexpression of LUADT1 was found to upregulate the expression of Pim-1, a target of miR-195. In addition, it was found that overexpression of LUADT1 and Pim-1 reduced the enhancement effects of miR-195 on LPS-induced cardiac endothelial cell apoptosis. Conclusion In summary, LUADT1 may protect cardiac endothelial cells against apoptosis in sepsis by regulating the miR-195/Pim-1 axis.


1987 ◽  
Author(s):  
Freek van Iwaarden ◽  
G Philip ◽  
de Groot ◽  
Bonno N Bouma

The presence of High Molecular Weight kininogen (HMWK) was demonstrated in cultured human endothelial cells (EC) by immunofluorescence techniques. Using an enzyme linked immunosorbent assay a concentration of 58 ng HMWK/10 cells was determined. Immunoprecipitation studies performed with lysed metabolically labelled endothelial cells and mono-specific antisera directed against HMWK suggested that HMWK is not synthesized by the endothelial cells. Endothelial cells cultured in the presence of HMWK-depleted serum did not contain HMWK. This, suggests that endothelial cells can internalize HMWK. Using 125I-HMWK it was demonstrated that cultured endothelial cells bind HMWK in a time-dependent, specific and saturable.way. The cells were found to internalize 125I-HMWK, since I-HMWK was detected in solubilized endothelial cells after the cell bound 125I-HMWK had been eluted with dextran sulphate.The binding of I-HMWK required the presence of zinc ions. Optimal binding of 125I-HMWK was observed at 50 μM Zn++ . Calcium ions inhibited the Zn++ dependent binding of 125I-HMWK |25EC. In the presence of 3 mM CaCl2 the total binding of 125I-HMWK was significantly decreased, and a .concentration of 200 μM Zn++ was Required for the binding of 125I-HMWK to thecells. Higher,. Ca concentrations did not further decrease the binding of 125I-HMWK. Analysis of tl^e binding data by the ligand computer program indicated 3.2 x 10 binding sites per cell for HMWK with a Kd of 35 nM at 50 μM ZnCl2 and 1 mM CaCl2. Specify binding of HMWK did also occur at physiological plasma Zn++ concentrations. Half maximal binding was observed at HMWK concentrations of ± 105 nM at 10 μM ZnCl2 and 45 nM at 25 μM ZnCl2. The HMWK binding sites were saturatecT at HMWK concentrations of 130 nM with 1.6 x 10 molecules of HMWK bound per cell and at 80 nM with 2.8 x 10 molecules of HMWK bound per cell at 10 and 25 pM ZnCl2 respectively. These results suggest that at physiological zinc, calcium and HMWK concentrations the HMWK binding sites on the endothelial cell are saturated. The presence of HMWK on the endothelial cell surface may play a role in the initiation of the intrinsic coagulation pathway. M ZnCl2 and 45 nM at 25 μM ZnCl2. The HMWK binding sites were saturatecT at HMWK concentrations of 130 nM with 1.6 x 10 molecules of HMWK bound per cell and at 80 nM with 2.8 x 10 molecules of HMWK bound per cell at 10 and 25 μM ZnCl2 respectively. These results suggest that at physiological zinc, calcium and HMWK concentrations the HMWK binding sites on the endothelial cell are saturated. The presence of HMWK on the endothelial cell surface may play a role in the initiation of the intrinsic coagulation pathway. M ZnCl2 and 45 nM at 25 μM ZnCl2. The HMWK binding sites were saturatecT at HMWK concentrations of 130 nM with 1.6 x 10 molecules of HMWK bound per cell and at 80 nM with 2.8 x 10 molecules of HMWK bound per cell at 10 and 25 μM ZnCl2 respectively. These results suggest that at physiological zinc, calcium and HMWK concentrations the HMWK binding sites on the endothelial cell are saturated. The presence of HMWK on the endothelial cell surface may play a role in the initiation of the intrinsic coagulation pathway. M ZnCl2 and 45 nM at 25 μM ZnCl2. The HMWK binding sites were saturatecT at HMWK concentrations of 130 nM with 1.6 x 10 molecules of HMWK bound per cell and at 80 nM with 2.8 x 10 molecules of HMWK bound per cell at 10 and 25 μM ZnCl2 respectively. These results suggest that at physiological zinc, calcium and HMWK concentrations the HMWK binding sites on the endothelial cell are saturated. The presence of HMWK on the endothelial cell surface may play a role in the initiation of the intrinsic coagulation pathway.M ZnCl2 and 45 nM at 25 μM ZnCl2. The HMWK binding sites were saturatecT at HMWK concentrations of 130 nM with 1.6 x 16 molecules of HMWK bound per cell and at 80 nM with 2.8 x 106 molecules of HMWK bound per cell at 10 and 25 μM ZnCl2 respectively. These results suggest that at physiological zinc, calcium and HMWK concentrations the HMWK binding sites on the endothelial cell are saturated. The presence of HMWK on the endothelial cell surface may play a role in the initiation of the intrinsic coagulation pathway.


2009 ◽  
Vol 296 (2) ◽  
pp. C273-C284 ◽  
Author(s):  
Tsung-Chuan Ho ◽  
Show-Li Chen ◽  
Yuh-Cheng Yang ◽  
Tzu-Hsiu Lo ◽  
Jui-Wen Hsieh ◽  
...  

Pigment epithelium-derived factor (PEDF) is an intrinsic antiangiogenic factor and a potential therapeutic agent. Previously, we discovered the mechanism of PEDF-induced apoptosis of human umbilical vein endothelial cells (HUVECs) as sequential induction/activation of p38 mitogen-activated protein kinase (MAPK), peroxisome proliferator-activated receptor gamma (PPAR-γ), and p53. In the present study, we investigated the signaling role of cytosolic calcium-dependent phospholipase A2-α (cPLA2-α) to bridge p38 MAPK and PPAR-γ activation. PEDF induced cPLA2-α activation in HUVECs and in endothelial cells in chemical burn-induced vessels on mouse cornea. The cPLA2-α activation is evident from the phosphorylation and nuclear translocation of cPLA2-α as well as arachidonic acid release and the cleavage of PED6, a synthetic PLA2 substrate. Such activation can be abolished by p38 MAPK inhibitor. The PEDF-induced PPAR-γ activation, p53 expression, caspase-3 activity, and apoptosis can be abolished by both cPLA2 inhibitor and small interfering RNA targeting cPLA2-α. Our observation not only establishes the signaling role of cPLA2-α but also for the first time demonstrates the sequential activation of p38 MAPK, cPLA2-α, PPAR-γ, and p53 as the mechanism of PEDF-induced endothelial cell apoptosis.


Sign in / Sign up

Export Citation Format

Share Document