scholarly journals Hepatocyte tissue factor activates the coagulation cascade in mice

Blood ◽  
2013 ◽  
Vol 121 (10) ◽  
pp. 1868-1874 ◽  
Author(s):  
Bradley P. Sullivan ◽  
Anna K. Kopec ◽  
Nikita Joshi ◽  
Holly Cline ◽  
Juliette A. Brown ◽  
...  

Key Points Mouse hepatocytes express cell surface tissue factor. Hepatocyte tissue factor activates the coagulation cascade in mice.

Blood ◽  
2006 ◽  
Vol 107 (12) ◽  
pp. 4746-4753 ◽  
Author(s):  
Samir K. Mandal ◽  
Usha R. Pendurthi ◽  
L. Vijaya Mohan Rao

AbstractTissue factor (TF) is the cellular receptor for clotting factor VIIa (FVIIa). The formation of TF-FVIIa complexes on cell surfaces triggers the activation of coagulation cascade and cell signaling. In the present study, we characterized the subcellular distribution of TF and its transport in fibroblasts by dual immunofluorescence confocal microscopy and biochemical methods. Our data show that a majority of TF resides in various intracellular compartments, predominantly in the Golgi. Tissue factor at the cell surface is localized in cholesterol-rich lipid rafts and extensively colocalized with caveolin-1. FVIIa binding to TF induces the internalization of TF. Of interest, we found that TF-FVIIa complex formation at the cell surface leads to TF mobilization from the Golgi with a resultant increase in TF expression at the cell surface. This process is dependent on FVIIa protease activity. Overall, the present data suggest a novel mechanism for TF expression at the cell surface by FVIIa. This mechanism could play an important role in hemostasis in response to vascular injury by increasing TF activity where and when it is needed.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 1924-1924
Author(s):  
Gourab Bhattacharjee ◽  
Jasimuddin Ahamed ◽  
Brian Pedersen ◽  
Amr El-Sheikh ◽  
Cheng Liu ◽  
...  

Abstract In vivo biopanning with phage displayed peptide libraries has generated a group of peptide probes which bind selectively to the surface of atherosclerotic plaque endothelium. The highest affinity peptide, EKO130, binds to the 78 kDa glucose regulated protein (Grp78). Grp78 has been demonstrated to play a role in numerous pathological processes as well as a possible role in the local cell surface regulation of the coagulation cascade. The goal of this study is to determine the role of Grp78 in coagulation including plasma clotting, factor Xa (Xa) generation, and tissue factor (TF) gene expression. siRNA mediated inhibition of Grp78 results in a marked increase in TF gene expression in bEND.3 endothelial cells and RAW macrophage-like cells. Antibody mediated inhibition of cell surface Grp78 results in increased TF procoagulant activity and TF-dependent Xa generation in both the endothelial and macrophage cell types. These studies are consistent with results from another laboratory demonstrating that Grp78 over-expression inhibits TF mediated initiation and support of the coagulation protease cascade. Thus, our work indicates that Grp78 suppresses TF at both the functional and molecular level by inhibiting both its thrombogenic potential and gene expression.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 1747-1747 ◽  
Author(s):  
Usha Pendurthi ◽  
Samit Ghosh ◽  
Samir Mandal ◽  
L. Vijaya Mohan Rao

Abstract Tissue factor (TF) is the cellular receptor for plasma clotting factor VIIa, and the formation of TF-VIIa complexes on cell surfaces trigger the coagulation cascade and cell signaling. It is a well-known fact that only a small fraction of TF at the cell surface is coagulantly active whereas a majority of TF on the cell surface is non-functional (cryptic). However, it is unclear, at present, how the coagulant active TF differs from the cryptic form, and mechanisms involved in TF activation. Recent studies show that a thiol oxidizing agent, HgCl2, increases TF coagulant activity on the surface of HL-60 cells by several fold (Chen et al., Blood vol 106, abstract #684, 2005). Further, TF is shown to associate with protein disulfide isomerase (PDI) in HaCaT cells (Ahamed et al., Blood vol 106, abstract #685, 2005). Based on these and other observations, it has been proposed that switching between cryptic and coagulant TF involves cleavage and formation of allosteric disulfide bond (Cys186-Cys209) and PDI has been implicated in controlling the conversion of cryptic TF to the coagulant form and to act as a switch between TF-mediated signaling and coagulation. Although these data are interesting and novel, there is no fail-proof evidence that disulfide switching alone and not other potential changes, such as exposure of anionic phospholipids, at the cell surface is responsible for the TF activation associated with various treatments. Therefore we have examined the effect of HgCl2 and other treatments on TF activation in MDA 231 cells in relation to anionic phospholipids and also characterized the cellular expression of PDI in this and other cell types. As reported earlier, the HgCl2 treatment increased the cell surface TF coagulant activity (5-fold or higher). However, the HgCl2 treatment also increased the prothombinase activity by 3-fold. More importantly, annexin V, which binds to anionic phospholipids, markedly reduced the increased TF coagulant activity associated with the HgCl2 treatment whereas it had only minimal and insignificant effect on TF activity of the control cells. Further, pretreatment of cells with 5,5′-dithio-bis(2-nitronezoic acid) (DTNB), a sulfhydryl reagent that reacts with thiol groups and thus can block disulfide switching, failed to prevent the increase in TF activity associated with the HgCl2 treatment. Interestingly, we found that treatment of MDA 231 cells with glutathione (5 to 100 mM), a cell impermeable reducing agent, also increased the surface TF activity by about 2- to 3-fold. In additional studies, we found that PDI antibodies had no effect on either the TF coagulant activity or TF-mediated cell signaling. Further, we found no evidence for the expression of PDI at the cell surface in immunofluorescence confocal microscopy as both monoclonal and polyclonal PDI antibodies failed to stain nonpermeabilized cells whereas they brightly stained intracellular PDI in permeabilized cells. In contrast, TF antibodies stained intensely the surface of both nonpermeabilized and permeabilized cells. Exposure of tumor cells to various proteases failed to transport the intracellular PDI to the cell surface. The present data raise a valid question whether disulfide switching by PDI plays the predominant and general regulatory role in controlling the TF coagulant activity and signaling functions. Our data also suggest that other cellular changes, including increase in anionic phospholipids, may be responsible for increased TF coagulant activity associated with the thiol oxidizers and other treatments.


Blood ◽  
2007 ◽  
Vol 110 (1) ◽  
pp. 161-170 ◽  
Author(s):  
Samir K. Mandal ◽  
Usha R. Pendurthi ◽  
L. Vijaya Mohan Rao

Tissue factor (TF) is the cellular receptor for clotting factor VIIa (FVIIa), and the formation of TF-FVIIa complexes on cell surfaces triggers the activation of the coagulation cascade and the cell signaling. Our recent studies have shown that a majority of TF resides in various intracellular compartments, predominantly in the Golgi, and that FVIIa binding to cell surface TF induces TF endocytosis and mobilizes the Golgi TF pool to translocate it to the cell surface. This present study is aimed to elucidate the mechanisms involved in TF endocytosis and its mobilization from the Golgi. Activation of protease-activated receptor 1 (PAR1) and PAR2 by specific peptide agonists and proteases, independent of FVIIa, mobilized TF from the Golgi store and increased the cell surface expression of TF. Blocking PAR2 activation, but not PAR1, with neutralizing antibodies fully attenuated the FVIIa-induced TF mobilization. Consistent with these data, silencing the PAR2 receptor, and not PAR1, abrogated the FVIIa-mediated TF mobilization. In contrast to their effect on TF mobilization, PAR1 and PAR2 activation, in the absence of FVIIa, had no effect on TF endocytosis. However, PAR2 activation is found to be critical for the FVIIa-induced TF endocytosis. Overall the data herein provide novel insights into the role of PARs in regulating cell surface TF expression.


1996 ◽  
Vol 133 (2) ◽  
pp. 293-304 ◽  
Author(s):  
J R Sevinsky ◽  
L V Rao ◽  
W Ruf

The ability to regulate proteolytic functions is critical to cell biology. We describe events that regulate the initiation of the coagulation cascade on endothelial cell surfaces. The transmembrane protease receptor tissue factor (TF) triggers coagulation by forming an enzymatic complex with the serine protease factor VIIa (VIIa) that activates substrate factor X to the protease factor Xa (Xa). Feedback inhibition of the TF-VIIa enzymatic complex is achieved by the formation of a quaternary complex of TF-VIIa, Xa, and the Kunitz-type inhibitor tissue factor pathway inhibitor (TFPI). Concomitant with the downregulation of TF-VIIa function on endothelial cells, we demonstrate by immunogold EM that TF redistributes to caveolae. Consistently, TF translocates from the Triton X-100-soluble membrane fractions to low-density, detergent-insoluble microdomains that inefficiently support TF-VIIa proteolytic function. Downregulation of TF-VIIa function is dependent on quaternary complex formation with TFPI that is detected predominantly in detergent-insoluble microdomains. Partitioning of TFPI into low-density fractions results from the association of the inhibitor with glycosyl phosphatidylinositol anchored binding sites on external membranes. Free Xa is not efficiently bound by cell-associated TFPI; hence, we propose that the transient ternary complex of TF-VIIa with Xa supports translocation and assembly with TFPI in glycosphingolipid-rich microdomains. The redistribution of TF provides evidence for an assembly-dependent translocation of the inhibited TF initiation complex into caveolae, thus implicating caveolae in the regulation of cell surface proteolytic activity.


Blood ◽  
1990 ◽  
Vol 76 (2) ◽  
pp. 361-367 ◽  
Author(s):  
SD Carson ◽  
DR Johnson

Abstract Complement activation at the cell surface initiates cell damage through a series of reactions occurring at the cell membrane and, after assembly of the terminal membrane attack complex, produces leakage of cytoplasmic contents from the cell. It has been documented that chemical or physical damage to cell membranes can cause a rapid increase in the expression of tissue factor procoagulant activity. In this study, antibody-mediated complement activation at the cell surface resulted in increased tissue factor activity, which correlated with cytolysis, as measured by 51-chromium release. Therefore, complement fixation on the cell surface can have a direct and immediate stimulatory effect on the coagulation cascade at the point of its initiation, with formation of a fibrin clot requiring only three consecutive proteolytic reactions after immunologically mediated cell damage.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 541-541
Author(s):  
Samir K. Mandal ◽  
Usha R. Pendurthi ◽  
L. Vijaya Mohan Rao

Abstract Tissue factor (TF) is the cellular receptor for clotting factor VIIa (FVIIa) and the formation of TF-FVIIa complexes on cell surfaces triggers the activation of coagulation cascade and cell signaling. TF is constitutively expressed in many extravascular cells, including fibroblasts and pericytes in and surrounding blood vessel walls, and lung epithelial cells. Our recent studies (Blood2006; 107:4746–4753) show that a majority of TF resides in various intracellular compartments, predominantly in the Golgi. FVIIa binding to cell surface TF induces the internalization of TF, and interestingly, mobilizes the Golgi TF pool and transports it to the outer cell surface. This process is dependent on FVIIa protease activity. This present study is aimed to elucidate potential mechanisms involved in TF internalization and the mobilization from the Golgi. Since studies from our laboratory and others showed that TF-FVIIa could activate protease-activated receptor (PAR)-mediated cell signaling and FVIIa protease activity is required for FVIIa-dependent internalization and trafficking of TF, we hypothesize that TF-VIIa activation of PAR1 or PAR2 plays a role in TF internalization and trafficking. To test this hypothesis, we first examined the role of PAR activation in TF-internalization and trafficking. Lung fibroblasts (WI-38 cells) were exposed to a variety of PAR activators, PAR activating peptide agonists (AP) and various proteases, and TF internalization and trafficking was evaluated by measuring the cell surface TF antigen and activity levels, internalization of cell surface TF (by using biotinylation of cell surface receptors and immunoprecipitation techniques) and mobilization of TF from the Golgi (by immunofluorescence confocal microscopy). PAR1 AP and PAR2 AP treatments increased the TF activity and antigen levels at the cell surface by 20 to 50% whereas PAR3 AP and PAR4 AP had no effect on cell surface TF activity and antigen levels. Cell surface TF activity and antigen levels were also increased slightly in fibroblasts exposed to thrombin and trypsin. Confocal microscopic image analysis of distribution of TF and the Golgi protein (golgin-97) revealed that about 85% of the untreated cells possess intact Golgi TF pool with high degree of colocalization with golgin-97 whereas as only 20–30% of FVIIa, thrombin, trypsin, PAR1 AP or PAR2 AP-treated cells had TF pool in the Golgi. Plasmin and FXa had moderate effect on TF mobilization from the Golgi. No detectable differences were found between control (untreated) cells and cells treated with either FFR-FVIIa, APC, PAR3 AP or PAR4 AP. Next, we investigated the role of PAR1 and PAR2 activation in FVIIa-mediated TF internalization and trafficking. Pretreatment of fibroblasts with PAR2 but not PAR1 activation blocking antibodies attenuated FVIIa-mediated Golgi TF mobilization. Consistent with these data, silencing PAR2 receptor by siRNA technique completely blocked FVIIa-mediated Golgi TF mobilization whereas PAR1 siRNA transfection had no effect (in control studies, we showed PAR1 antibodies or PAR1si RNA transfection blocked thrombin-mediated TF mobilization). Additional studies showed a significant internalization of TF in cells exposed to FVIIa which was completely blocked by silencing PAR2 but not PAR1. Overall the data provided herein suggest a novel mechanism by which tissue factor expression is regulated at the cell surface.


2005 ◽  
Vol 25 (8) ◽  
pp. 1737-1743 ◽  
Author(s):  
Gourab Bhattacharjee ◽  
Jasimuddin Ahamed ◽  
Brian Pedersen ◽  
Amr El-Sheikh ◽  
Nigel Mackman ◽  
...  

Blood ◽  
1990 ◽  
Vol 76 (2) ◽  
pp. 361-367
Author(s):  
SD Carson ◽  
DR Johnson

Complement activation at the cell surface initiates cell damage through a series of reactions occurring at the cell membrane and, after assembly of the terminal membrane attack complex, produces leakage of cytoplasmic contents from the cell. It has been documented that chemical or physical damage to cell membranes can cause a rapid increase in the expression of tissue factor procoagulant activity. In this study, antibody-mediated complement activation at the cell surface resulted in increased tissue factor activity, which correlated with cytolysis, as measured by 51-chromium release. Therefore, complement fixation on the cell surface can have a direct and immediate stimulatory effect on the coagulation cascade at the point of its initiation, with formation of a fibrin clot requiring only three consecutive proteolytic reactions after immunologically mediated cell damage.


1994 ◽  
Vol 72 (06) ◽  
pp. 848-855 ◽  
Author(s):  
Dzung The Le ◽  
Samuel I Rapaport ◽  
L Vijaya Mohan Rao

SummaryFibroblast monolayers constitutively expressing surface membrane tissue factor (TF) were treated with 0.1 mM N-ethylmaleimide (NEM) for 1 min to inhibit aminophospholipid translocase activity without inducing general cell damage. This resulted in increased anionic phospholipid in the outer leaflet of the cell surface membrane as measured by the binding of 125I-annexin V and by the ability of the monolayers to support the generation of prothrombinase. Specific binding of 125I-rVIIa to TF on NEM-treated monolayers was increased 3- to 4-fold over control monolayers after only brief exposure to 125I-rVIIa, but this difference progressively diminished with longer exposure times. A brief exposure of NEM-treated monolayers to rVIIa led to a maximum 3- to 4-fold enhancement of VIIa/TF catalytic activity towards factor X over control monolayers, but, in contrast to the binding studies, this 3- to 4-fold difference persisted despite increasing time of exposure to rVIIa. Adding prothrombin fragment 1 failed to diminish the enhanced VIIa/TF activation of factor X of NEM-treated monolayers. Moreover, adding annexin V, which was shown to abolish the ability of NEM to enhance factor X binding to the fibroblast monolayers, also failed to diminish the enhanced VIIa/TF activation of factor X. These data provide new evidence for a possible mechanism by which availability of anionic phospholipid in the outer layer of the cell membrane limits formation of functional VIIa/TF complexes on cell surfaces.


Sign in / Sign up

Export Citation Format

Share Document