Faculty Opinions recommendation of A critical role for extracellular protein disulfide isomerase during thrombus formation in mice.

Author(s):  
Steve Watson
Toxins ◽  
2019 ◽  
Vol 11 (8) ◽  
pp. 458 ◽  
Author(s):  
Jessica Guyette ◽  
Patrick Cherubin ◽  
Albert Serrano ◽  
Michael Taylor ◽  
Faisal Abedin ◽  
...  

Protein disulfide isomerase (PDI) is mainly located in the endoplasmic reticulum (ER) but is also secreted into the bloodstream where its oxidoreductase activity is involved with thrombus formation. Quercetin-3-rutinoside (Q3R) blocks this activity, but its inhibitory mechanism against PDI is not fully understood. Here, we examined the potential inhibitory effect of Q3R on another process that requires PDI: disassembly of the multimeric cholera toxin (CT). In the ER, PDI physically displaces the reduced CTA1 subunit from its non-covalent assembly in the CT holotoxin. This is followed by CTA1 dislocation from the ER to the cytosol where the toxin interacts with its G protein target for a cytopathic effect. Q3R blocked the conformational change in PDI that accompanies its binding to CTA1, which, in turn, prevented PDI from displacing CTA1 from its holotoxin and generated a toxin-resistant phenotype. Other steps of the CT intoxication process were not affected by Q3R, including PDI binding to CTA1 and CT reduction by PDI. Additional experiments with the B chain of ricin toxin found that Q3R could also disrupt PDI function through the loss of substrate binding. Q3R can thus inhibit PDI function through distinct mechanisms in a substrate-dependent manner.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 691-691 ◽  
Author(s):  
Reema Jasuja ◽  
Jaehyung Cho ◽  
Bruce Furie ◽  
Barbara Furie

Abstract We have previously reported that protein disulfide isomerase is required in wild-type mice for platelet thrombus formation and fibrin generation in an in vivo laser injury model of thrombosis (Cho et al. J. Clin. Invest., 2008; 118:1123–31). Fibrin deposition after laser injury to the vessel wall in Par4−/− mice, lacking the G protein-coupled platelet thrombin receptor, is independent of platelets or requires minimal platelet activation or accumulation (Vandendries et al. Proc. Natl. Acad. Sci., 2007; 104:288–92). However, protein disulfide isomerase inhibitors have a dramatic effect on fibrin accumulation in Par4− mice, suggesting that these inhibitors may function by a platelet independent mechanism. Here, we compare the contributions of endothelium and platelet-derived protein disulfide isomerase to fibrin generation in the mouse laser injury model of thrombosis. In vitro studies using cultured human umbilical vein endothelial cells and human aortic endothelial cells show that protein disulfide isomerase can be secreted rapidly into the culture medium from these cells upon thrombin stimulation. Using intravital microscopy, we observe that protein disulfide isomerase is not detectable on the vessel wall prior to laser injury but can be detected on the injured cremaster arteriolar wall and in the developing thrombus very rapidly after laser induced injury in the live mouse. The median integrated fluorescence intensity for protein disulfide isomerase in wild-type mice was compared to wild-type mice injected with 10ug/g mouse of Integrilin, an inhibitor of platelet activation and platelet thrombus formation, and thus, an inhibitor of the contribution of platelet derived protein disulfide isomerase to thrombus formation. Protein disulfide isomerase expression was similar in both treated and untreated animals upto 30 seconds post-laser injury. After 30 seconds, the expression of protein disulfide isomerase in integrilin treated mice was significantly decreased compared to that in untreated mice, indicating that the initial protein disulfide isomerase was derived from the endothelium and later additional protein disulfide isomerase was derived from the platelets following their accumulation in the developing thrombus. Fibrin deposition, a measure of thrombin generation was comparable in wild-type mice that had been treated with Integrilin or treated with a control buffer, suggesting that endothelial-derived protein disulfide isomerase was sufficient for fibrin generation. The rate and amount of fibrin generation was indistinguishable in both groups. Furthermore, inhibition of the protein disulfide isomerase with the function blocking monoclonal antibody RL-90 (3ug/g mouse) eliminated any fibrin deposition in wild-type mice that had been treated with Integrilin. Taken together, these data indicate that endothelium-derived protein disulfide isomerase is necessary to support fibrin deposition in vivo in our laser injury model of thrombus formation. The initial protein disulfide isomerase expressed at the site of injury is derived from endothelial cells but platelets activated at the site of thrombus formation contribute, amplify and sustain protein disulfide isomerase expression.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 2168-2168
Author(s):  
Lu Wang ◽  
Yi Wu ◽  
Junsong Zhou ◽  
Syed S. Ahmad ◽  
Bulent Mutus ◽  
...  

Abstract Abstract 2168 Several members of the protein disulfide isomerase family of enzymes are important in platelet function and in thrombosis. Platelet protein disulfide isomerase (PDI) has been shown to have an important role in platelet function but is reported to not be required for thrombus formation in vivo. A novel platelet PDI called ERp57 mediates platelet aggregation but its role in thrombus formation is unknown. To determine the specific role of platelet-derived ERp57 in hemostasis and thrombosis we generated a megakaryocyte/platelet specific knockout. Despite normal platelet counts and platelet glycoprotein expression, mice with ERp57-deficient platelets had prolonged tail-bleeding times and thrombus occlusion times, and defective activation of the αIIbβ3 integrin and platelet aggregation. The aggregation defect was corrected by addition of exogenous ERp57 implicating surface ERp57 in platelet aggregation. Platelet surface ERp57 protein and activity increased substantially with platelet activation. We conclude that platelet-derived ERp57 is required for hemostasis and thrombosis and platelet function. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 2880-2880
Author(s):  
Pavan K Bendapudi ◽  
Roelof H Bekendam ◽  
Lin Lin ◽  
Mingdong Huang ◽  
Bruce Furie ◽  
...  

Abstract Vascular thiol isomerases comprise a family of enzymes including protein disulfide isomerase (PDI), ERp5, and ERp57 that are important in the process of thrombus formation. PDI is secreted at sites of vascular injury, and antibody-mediated PDI inhibition prevents thrombus formation in a mouse laser injury model. Our group has previously reported on the discovery of the small molecule PDI inhibitors quercetin-3-rutinoside and ML359. Identified as part of a high-throughput screen, ML359 is a second-generation PDI inhibitor that selectively blocks PDI oxidoreductase activity with approximately ten-fold the potency of quercetin-3-rutinoside. To better understand the mechanism of allosteric modulation of PDI by small molecules, we evaluated the association of ML359 with isolated domains of PDI, determined the effects of ML359 on a variety of PDI functions, and compared the activity of ML359 to that of quercetin-3-rutinoside. PDI is composed of four thioredoxin-like domains and an x-linker region in the sequence a-b-b’-x-a’. Major substrate binding is thought to occur in the b-b’ region while the a and a’ domains contain catalytically active cysteine motifs (CGHC) that mediate the oxidoreducase, nitrosylase, and thiol isomerase functions of PDI. In order to identify potential binding sites of ML359 on PDI, we constructed and expressed the domain fragments a, ab, abb’, b’xa’, and a’. These fragments were tested in the presence of 10 µM ML359 using an insulin turbidometric assay that measures the oxidoreductase activity of PDI. ML359 demonstrated full inhibition of oxidoreductase activity when full-length PDI and the b’xa’ fragment were used whereas no inhibition was observed with the other fragments assayed. These results are consistent with docking studies showing that ML359 likely binds in a pocket formed at the b’x interface. In contrast, when the same experiment was performed in the presence of 30 µM of quercetin-3-rutinoside, inhibition was only noted with full-length PDI and the abb’ and b’xa’ fragments, suggesting that binding was dependent on the b’ and not the x-linker region. To determine if ML359 has differential effects on the oxidoreductase and nitrosylase functions of PDI, we utilized a platelet-based assay in which fluorescence intensity stemming from the NO-sensitive intracellular dye DAF-FM was measured as an indicator of PDI-mediated translocation of NO from the extracellular surface into the cytosol (transnitrosylation). While quercetin-3-rutinoside potently inhibited PDI-mediated transnitrosylation activity, ML359 had no effect. These results are consistent with the idea that the transnitrosylase and oxidoreducase functions of PDI are separable and inhibition of either is specific to the small molecule used. We evaluated the ability of ML359 to inhibit thrombosis in a mouse laser injury model. Intravital microscopy was used to follow thrombus formation in mouse cremaster arterioles after laser-induced vascular injury. Infusion of ML359 resulted in inhibition of thrombus formation, in contrast to thrombosis seen after infusion of vehicle alone. In summary, ML359 is a second generation small molecule inhibitor of PDI that likely binds at the b’x interface of the enzyme. Furthermore, ML359 is able to selectively inhibit PDI oxidoreductase activity without affecting transnitrosylase activity. ML359 may prove a useful molecular probe to better understand the different functions of PDI relative to thrombus formation in vivo. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 15-15
Author(s):  
Chao Fang ◽  
Sheryl R. Bowley ◽  
Barbara C. Furie ◽  
Bruce Furie

Abstract Protein disulfide isomerase (PDI), secreted by platelets and endothelial cells upon vascular injury, is required for thrombus formation. However, the precise mechanism by which PDI regulates thrombosis remains elusive. Using PDI variants that form stable mixed disulfide complexes with their substrates, we performed kinetic trapping experiment in platelet rich plasma and identified multiple substrate proteins for PDI, including vitronectin. Importantly, when using variants of endoplasmic reticulum protein 57 (ERp57), a thiol isomerase that has a similar domain structure as PDI and is also important for thrombus formation, the trapping mutants of ERp57 do not interact with vitronectin. This result has demonstrated the substrate specificity of PDI during our kinetic trapping experiment. Further study using polyethylene glycol (PEG)-based gel mobility shift assay combined with mass spectrometry has identified the redox reaction between PDI and vitronectin occurs on two disulfide bonds Cys 137-161 and Cys 274-453 in the hemopexin-like domains of plasma vitronectin. Vitronectin, as a substrate of extracellular PDI, has been shown to be important for thrombus formation. Vitronectin null mice have reduced platelet accumulation and fibrin deposition in the cremaster arterioles following laser injury. Vitronectin null mice also have significantly prolonged large-vessel thrombosis in the carotid artery using the ferric chloride thrombosis model. Using intravital microscopy we showed that vitronectin rapidly accumulates in a growing thrombus following vessel injury. When mice are treated with eptifibatide to eliminate platelet accumulation, we still observe significant amount of vitronectin accumulation on the vessel wall in the absence of platelet thrombus. This observation was further confirmed using confocal intravital microscopy. After 3D reconstruction of a growing thrombus in mouse cremaster arteriole, vitronectin was identified to locate primarily on the CD31 stained vessel wall. These combined studies suggest that plasma-derived vitronectin and not platelet-derived vitronectin is the primary substrate of PDI. Our study further showed that the indispensable role of vitronectin to a growing thrombus depends on extracellular PDI. Native plasma vitronectin does not bind to αvβ3 or αIIbβ3-integrins on endothelial cells and platelets. On solid phase binding assay, plasma sample pre-treated with wild-type PDI showed significantly increased binding of vitronectin to its ligand αvβ3 or αIIbβ3-integrins. However, this increase was not observed in plasma pre-treated with dead-mutant PDI or ERp57. In addition, using immunofluorescent staining, PDI treated plasma sample also showed significantly increased binding of vitronectin to activated human umbilical vein endothelial cells (HUVECs) and this binding was abrogated by RGD peptides or an αvβ3 blocking antibody. The critical role of extracellular PDI for the regulation of vitronectin in a growing thrombus was further confirmed in our in vivo studies. When mice were treated with quecetin-3-rutinoside or two different inhibitory antibodies that selectively block PDI activity, the accumulation of vitronectin and platelets was significantly reduced. These combined results demonstrate that extracellular PDI regulates vitronectin in a growing thrombus to promote platelet accumulation and fibrin generation. In summary, our studies have revealed a novel regulatory mechanism during the initiation of thrombus formation. Under normal physiologic conditions in the absence of secreted PDI, thrombus formation is suppressed and maintains a quiescent, patent vasculature. The release of PDI during vascular injury serves as a novel regulatory switch that allows activation of proteins, including vitronectin, which are critical for the following platelet accumulation and fibrin generation. Figure. Figure. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (6) ◽  
pp. 1052-1061 ◽  
Author(s):  
Kyungho Kim ◽  
Eunsil Hahm ◽  
Jing Li ◽  
Lisa-Marie Holbrook ◽  
Parvathy Sasikumar ◽  
...  

Key Points Platelet PDI regulates αIIbβ3 integrin activation without affecting platelet activation and inside-out integrin signaling. Platelet PDI is essential for platelet accumulation but not for fibrin generation and hemostasis in mice.


Sign in / Sign up

Export Citation Format

Share Document