Cryptic Vs. Active Tissue Factor: Cystine186-Cystine 209 Disulfide Bond Switch Is Not the Answer.

Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 333-333 ◽  
Author(s):  
Hema Kothari ◽  
Ramesh Nayak ◽  
L. Vijaya Mohan Rao ◽  
Usha Pendurthi

Abstract Abstract 333 Tissue factor (TF) plays the key role in initiation of blood coagulation by allosterically activating coagulation factor VIIa (FVIIa). TF is essential for hemostasis but aberrant expression would lead to thrombotic disorders. TF on cell surfaces resides mostly in a cryptic state. At present it is not entirely clear how cryptic TF differs from procoagulant active TF and how de-encryption occurs. Recent studies have suggested that cryptic and active TF may have different conformations, i.e., cryptic TF contains unpaired cysteine thiols at Cys 186 and Cys 209 in the membrane proximal domain whereas procoagulant active TF contains the oxidized Cys186-Cys209 disulfide bond. It has been suggested that the catalytic switching of this disulfide bond between reduced and oxidized states by protein disulfide isomerase (PDI) might modulate the activity status of TF. Although the validity of this hypothesis has been questioned, more recent reports have supported the contention that PDI-catalyzed disulfide bond formation of Cys186-Cys209 in TF plays a role in regulation of TF activity both in vitro and in vivo. To reconcile the conflicting data in the recent literature, we re-examined in the present study the central dogma on which the hypothesis was originated, i.e., a TF mutant lacking Cys186-Cys 209 disulfide bond is functionally inactive (cryptic) and that TF activation requires formation of the Cys186-Cys209 disulfide bond. CHO cells were transfected with wild type TF or TF mutants that selectively preclude Cys186-Cys209 disulfide bond formation, i.e., TFC186S, TFC209S, or TFC186S/C209S, and TF procoagulant and antigen expression levels at the cell surface were quantified accurately by measuring TF procoagulant activity in a factor × activation assay and TF antigen levels in radioligand binding studies utilizing two different TF mAb (10H10 and 5G9) and FVIIa. CHO cells transfected with TFC186S, TFC209S or TFC186S/C209S expressed very little procoagulant activity (less than 1% of CHO cells transfected with wild-type TF under identical experimental conditions). However, TF mAb and FVIIa binding studies revealed that CHO cells transfected with TFC186S, TFC209S or TFC186S/C209S expressed very little TF protein on the cell surface (∼ 1 to 2% of CHO cells transfected with wild-type TF). Similar results were obtained whether we mutated Cys 186 or Cys 209 to Ser or Ala or whether we used transient or stable transfections. To confirm that the lower expression of TFC186S, TFC209S or TFC186S/C209S is neither an artifact nor limited to CHO cell model system, we extended these studies to endothelial cells. Human umbilical vein endothelial cells (HUVECs) were transduced with adenovirus particles (25 moi/cell) encoding wild-type TF, TFC186S, TFC209S or TFC186S/C209S and TF antigen levels at the cell surface and in cell lysates were measured using multiple methods - confocal microscopy, FACS, TF antigen assay, immuno blot analysis, TF mAb binding and FVIIa binding. HUVEC transduced with adenoviral particles encoding TFC186S, TFC209S or TFC186S/C209S expressed very low levels of TF antigen both at the cell surface ( 1 to 5%) as well as in total (10 to 15%) (TFC186S > TFC209S >TFC186S/C209S) compared to HUVEC transduced with the same number of adenovirus particles expressing wild-type TF. Cell surface TF activity measured in the presence of saturating concentrations of FVIIa revealed that the TF procoagulant activity of the mutants correlate well with the amount of TF antigen present on the cell surface, i.e., there were no significant differences in the specific activity of TF mutants and wild-type TF (TF procoagulant activity/amount of TF antigen, pM FXa formed/femto mole TF: wild-type TF, 68 ± 6; TFC186S, 55 ± 10; TFC209S, 57 ± 17; and TFC186S/C209S, 64 ± 0.2). More importantly, treatment of HUVEC with the thiol-oxidizing agent HgCl2 or with ionomycin increased the cell surface TF activity to the same extent (200 to 400% increase over HUVEC treated with control vehicle) in HUVEC expressing TFC186S/C209S or wild-type TF. In summary the present data provide clear evidence for that TF lacking Cys186-Cys209 bond is coagulantly active and a lower TF procoagulant activity in cells expressing the mutant TF that precludes Cys186-Cys209 disulfide bond formation is due to severe impairment in TF protein synthesis/processing and not because the mutant TF assumes the cryptic conformation. Our data also show that TF de-encryption does not require the formation of Cys186-Cys209 disulfide bond. Disclosures: No relevant conflicts of interest to declare.

Blood ◽  
2010 ◽  
Vol 115 (21) ◽  
pp. 4273-4283 ◽  
Author(s):  
Hema Kothari ◽  
Ramesh C. Nayak ◽  
L. Vijaya Mohan Rao ◽  
Usha R. Pendurthi

Tissue factor (TF) on cell surfaces resides mostly in a cryptic state. It is not entirely clear how cryptic TF differs from procoagulantly active TF and how deencryption occurs. Here, we critically evaluated the importance of cystine 186–cystine 209 (Cys186-Cys209) bond formation for TF procoagulant activity and its de-encryption. Chinese hamster ovary cells transfected with TFC186S, TFC209S, or TFC186S/C209S expressed little procoagulant activity at the cell surface. TF monoclonal antibody and activated factor VII (FVIIa) binding studies showed that little TF protein was present at the cell surface in cells expressing mutant TF. Similar data were obtained in human umbilical vein endothelial cells (HUVECs) transduced to express TFC186S, TFC209S, or TFC186S/C209S. Analysis of TF activity in HUVECs expressing similar levels of wild-type TF and TFC186S/C209S showed that TF mutant in the presence of saturating concentrations of FVIIa exhibited similar coagulant activity as that of wild-type TF. More importantly, treatment of HUVECs expressing TFC186S/C209S with HgCl2 or ionomycin increased the cell-surface TF activity to the same extent as that of the wild-type TF. Our data provide clear evidence that TF lacking the Cys186-Cys209 bond is coagulantly active once it is complexed with FVIIa, and TF de-encryption does not require Cys186-Cys209 disulfide bond formation.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 1694-1694
Author(s):  
Jian-Kie Tie ◽  
Mei-Yan Zheng ◽  
Darrel W. Stafford ◽  
David L. Straight

Abstract The vitamin K-dependent carboxylase is an integral membrane protein with five transmembrane domains (TMDs). It catalyzes the post-translational modification of specific glutamic acid residues of vitamin K-dependent proteins to gamma-carboxyglutamic acid residues. This posttranslational modification is critical for the biological functions of blood coagulation. The native enzyme is a single chain molecule with one disulfide bond. In this study, we have expressed carboxylase as two chains: residues 1–345 and 346–758 in the same insect cells. Our results show that these two fragments are assembled into a fully active enzyme and are joined by a disulfide. Affinity purification of the carboxylase C-terminal fragment (346–758) results in co-purification of the N-terminal fragment (1–345) even under reducing condition. This indicates that, in addition to the disulfide linkage between these two fragments, they are also linked by non-covalent interactions. One possibility is that the hydrophobic interactions between the TMDs play a role. According to carboxylase membrane topology, there are four TMDs (1–4) in the N-terminal fragment and one TMD (fifth) in the C-terminal fragment. The C-terminal fragment contains all glycosylation sites. When we introduced two prolines to disrupt the transmembrane helix in the wild type carboxylase’s fifth TMD, glycosylation was eliminated. This indicates that the domain is not inserted into the lumen of the ER, but remains in the cytoplasm. Therefore, as our results demonstrate, in the two chain carboxylase with its fifth TMD disrupted, the two chains do not form a disulfide bound nor do they associate through essential non-covalent TMD interactions. While proline residues can disrupt membrane helices as described above, they often occur at the interface between the membrane and the lumenal surface of ER; these prolines appear to affect the chain orientation as it exits the membrane. There is a proline at residue 378 near the lumenal surface of the fifth TMD helix of carboxylase. To examine P378’s effect on disulfide bond formation, we mutated it to leucine. Results show that less disulfide bond formed in the two chain mutant carboxylase and the protein was significantly degraded when compared to the unmutated two chain molecule. Based on our results, we conclude the following: 1) the two chain carboxylase is assembled into a single molecule in vivo and the two chains are joined by a disulfide bond, the enzyme carboxylates gla-containing substrates and binds propeptide with affinity similar to that of wild type enzyme. Therefore, this molecule is a good model for structural studies of TMD interactions and disulfide bond formation; 2) TMD association in the membrane is important for the orientation of the N- and C-terminal portions of carboxylase to be assembled into the active enzyme; 3) and finally proline residue 378 at the lumenal interface of the fifth TMD plays a key role in the conformation which promotes disulfide formation.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 1029-1029
Author(s):  
Prosenjit Sen ◽  
Samit Ghosh ◽  
Pernille K Holm ◽  
Mirella Ezban ◽  
Usha Pendurthi ◽  
...  

Abstract Frequent spontaneous joint bleedings in severe hemophilia leads to chronic arthopathy, which severely reduces the quality of life of severe hemophiliacs. Prophylactic administration of FVIII/FIX concentrates was introduced in the early 1960’s to convert severe hemophilia into a moderate form that cause less frequent joint bleedings. Recent data in hemophilia patients with inhibitors have provided evidence for that daily dose of rFVIIa can act in secondary prophylaxis in patients with frequent bleeds. The plasma half-life of rFVIIa is approx 2–3 hours which thus may limit its efficacy and convenience in prevention of bleeds. Therefore, development of rFVIIa molecules that could remain in the circulation for a prolonged period may potentially improve prophylactic options of rFVIIa. PEGylation is an established and clinically proven strategy for prolonging the circulatory life-time of bio-therapeutic proteins. Recently, GlycoPEGylation technology was used to generate PEGylated rFVIIa derivatives (Stennicke et al., 2008 Thromb. Haemost, in press) and they appeared to activate factor X at a similar rate as of rFVIIa (Stennicke et al., 2008 Thromb. Haemost, in press; and Ghosh et al., 2008, J. Thromb. Haemost, in press). In the present study, we further characterized the glycoPEGylated rFVIIa, rFVIIa-10K PEG and rFVIIa-40K PEG, particularly in reference to their interaction with tissue factor (TF) and endothelial cell protein C receptor (EPCR) on cell surfaces and their catabolism. rFVIIa and glycoPEGylated rFVIIa were labeled with 125I and the radio-iodinated proteins were used to monitor rFVIIa binding and uptake in endothelial cells, CHO cells stably transfected with EPCR, and fibroblasts. PEG modification of rFVIIa resulted in a marked decrease in the rate of rFVIIa uptake in endothelial cells. The reduction in the uptake of rFVIIa following attachment of PEG was primarily due to the reduced association of rFVIIa to the cell surface. The level of glycoPEGylated rFVIIa binding to endothelial cells was about 20 to 30% of that was obtained with the rFVIIa. No significant differences were found between rFVIIa-10K PEG and rFVIIa-40K PEG in their association with endothelial cells. rFVIIa-40K PEG uptake was slightly lower compared to rFVIIa-10K PEG. The reduction in the uptake of glycoPEGylated rFVIIa by endothelial cells appeared to be the result of a decreased rate of glycoPEGylated rFVIIa binding to EPCR. Consistent with this glycoPEGylated rFVIIa binding, relative to rFVIIa, was markedly lower to CHO cells expressing EPCR whereas no significant differences were found in the basal binding of rFVIIa and glycoPEGylated rFVIIa to wild-type CHO cells. The radioligand binding studies also revealed marked differences between rFVIIa and glycoPEGylated rFVIIa binding to TF on fibroblasts. Although the total amounts of radioligands associated with cell surface TF at saturating concentrations were similar, the affinity of rFVIIa-10K PEG and rFVIIa-40K PEG to TF, relative to rFVIIa affinity to TF, was lowered by about 6 to 20-fold, respectively. Surprisingly, if the binding affinities were evaluated in factor X activation studies, the differences between rFVIIa and glycoPEGylated rFVIIa were minimal. The differences between rFVIIa and glycoPEGylated rFVIIa binding to TF in these two different assay systems suggest that rFVIIa and glycoPEGylated rFVIIa may interact with active TF with equal affinity whereas they interact differently with cryptic TF. Alternatively, glycoPEGylated rFVIIa binds initially to cell surface TF with a similar affinity as of rFVIIa, but may dissociate readily from TF upon the washing that is required to measure the binding of radioligands to cells. Additional studies yielded data that favor the later possibility. Overall the present data suggest that the reduced affinity of rFVIIa associated with glycoPEGylation on binding to EPCR and TF is likely attributable to either steric hindrance or changes in electrostatic binding properties rather than modification of binding sites itself. Further, the reduced uptake of glycoPEGylated rFVIIa by cells stems primarily from the decreased association of the modified rFVIIa to cell surfaces rather than specific impairment in their internalization.


2005 ◽  
Vol 187 (12) ◽  
pp. 3941-3949 ◽  
Author(s):  
Robert E. Feissner ◽  
Caroline S. Beckett ◽  
Jennifer A. Loughman ◽  
Robert G. Kranz

ABSTRACT Transposon mutagenesis of Bordetella pertussis was used to discover mutations in the cytochrome c biogenesis pathway called system II. Using a tetramethyl-p-phenylenediamine cytochrome c oxidase screen, 27 oxidase-negative mutants were isolated and characterized. Nine mutants were still able to synthesize c-type cytochromes and possessed insertions in the genes for cytochrome c oxidase subunits (ctaC, -D, and -E), heme a biosynthesis (ctaB), assembly of cytochrome c oxidase (sco2), or ferrochelatase (hemZ). Eighteen mutants were unable to synthesize all c-type cytochromes. Seven of these had transposons in dipZ (dsbD), encoding the transmembrane thioreduction protein, and all seven mutants were corrected for cytochrome c assembly by exogenous dithiothreitol, which was consistent with the cytochrome c cysteinyl residues of the CXXCH motif requiring periplasmic reduction. The remaining 11 insertions were located in the ccsBA operon, suggesting that with the appropriate thiol-reducing environment, the CcsB and CcsA proteins comprise the entire system II biosynthetic pathway. Antiserum to CcsB was used to show that CcsB is absent in ccsA mutants, providing evidence for a stable CcsA-CcsB complex. No mutations were found in the genes necessary for disulfide bond formation (dsbA or dsbB). To examine whether the periplasmic disulfide bond pathway is required for cytochrome c biogenesis in B. pertussis, a targeted knockout was made in dsbB. The DsbB− mutant makes holocytochromes c like the wild type does and secretes and assembles the active periplasmic alkaline phosphatase. A dipZ mutant is not corrected by a dsbB mutation. Alternative mechanisms to oxidize disulfides in B. pertussis are analyzed and discussed.


2006 ◽  
Vol 188 (12) ◽  
pp. 4190-4197 ◽  
Author(s):  
Yohei Hizukuri ◽  
Toshiharu Yakushi ◽  
Ikuro Kawagishi ◽  
Michio Homma

ABSTRACT The P ring of the bacterial flagellar motor consists of multiple copies of FlgI, a periplasmic protein. The intramolecular disulfide bond in FlgI has previously been reported to be essential for P-ring assembly in Escherichia coli, because the P ring was not assembled in a dsbB strain that was defective for disulfide bond formation in periplasmic proteins. We, however, found that the two Cys residues of FlgI are not conserved in other bacterial species. We then assessed the role of this intramolecular disulfide bond in FlgI. A Cys-eliminated FlgI derivative formed a P ring that complemented the flagellation defect of our ΔflgI strain when it was overproduced, suggesting that disulfide bond formation in FlgI is not absolutely required for P-ring assembly. The levels of the mature forms of the FlgI derivatives were significantly lower than that of wild-type FlgI, although the precursor protein levels were unchanged. Moreover, the FlgI derivatives were more susceptible to degradation than wild-type FlgI. Overproduction of FlgI suppressed the motility defect of ΔdsbB cells. Additionally, the low level of FlgI observed in the ΔdsbB strain increased in the presence of l-cystine, an oxidative agent. We propose that intramolecular disulfide bond formation facilitates the rapid folding of the FlgI monomer to protect against degradation in the periplasmic space, thereby allowing its efficient self-assembly into the P ring.


2008 ◽  
Vol 76 (7) ◽  
pp. 3086-3092 ◽  
Author(s):  
Aiping Qin ◽  
David W. Scott ◽  
Barbara J. Mann

ABSTRACT Francisella tularensis subsp. tularensis is a highly virulent bacterium that is a CDC select agent. Despite advancements in the understanding of its biology, details pertaining to virulence are poorly understood. In previous work, we identified a transposon insertion mutant in the FTT0107c locus that was defective in intracellular survival in HepG2 and J77A.1 cells. Here, we report that this mutant was also highly attenuated in vivo. The FTT0107c locus is predicted to encode an ortholog of the disulfide bond formation B protein (DsbB). This designation was confirmed by complementation of an Escherichia coli dsbB mutant. This dsbB mutant of Schu S4 was highly attenuated in mice, but unlike what has been reported for Francisella novicida, intranasal immunization with a sublethal dose did not induce protection against wild-type challenge. dsbB was found to be transcribed in an operon with acrA and acrB, which encode an RND-type efflux pump. However, this pump did not make a significant contribution to virulence because strains with nonpolar deletions in acrA and acrB behaved like wild-type strain Schu S4 with respect to intracellular growth and in vivo virulence. This result is in contrast to a report that an acrB mutant of a live vaccine strain of F. tularensis has decreased virulence in mice. Overall, these results demonstrate key differences between the virulence requirements of Schu S4 and less virulent subspecies of Francisella. We have shown that DsbB is a key participant in intracellular growth and virulence, and our results suggest that there are critical virulence factors that contain disulfide bonds.


1984 ◽  
Vol 99 (6) ◽  
pp. 2123-2130 ◽  
Author(s):  
D D Wagner ◽  
V J Marder

Biosynthesis of von Willebrand protein by human umbilical vein endothelial cells involved distinct processing steps marked by the presence of several intermediate molecular species. Examination of endoglycosidase H sensitivity of these intracellular intermediates indicated that the processing steps occurred in at least two separate cellular compartments. In the pre-Golgi apparatus (most probably the endoplasmic reticulum), the high mannose carbohydrates were added onto the precursor monomer chains and the 260,000-mol-wt monomers dimerized by interchain disulfide bond formation. The other processing steps have been localized to the Golgi apparatus and later compartments (e.g., Weibel-Palade bodies). High mannose carbohydrate was converted to the complex type, leading to the appearance of a larger precursor subunit of 275,000 mol wt. The 275,000-mol-wt species was not formed if carbohydrate processing was inhibited by the ionophore monensin. From the large pool of dimers of precursor subunits, the high molecular weight multimers were built. These dimer molecules appeared to have free sulfhydryls which might have been involved in the interdimer disulfide bond formation. Simultaneously with multimerization, the precursor subunits were cleaved to the 220,000-mol-wt form. The cleavage of the pro-sequence was not likely to be an absolute requirement for von Willebrand protein multimerization or secretion, as the 275,000-mol-wt precursor subunit was present in secreted high molecular weight multimers of the protein.


Cancers ◽  
2021 ◽  
Vol 13 (15) ◽  
pp. 3718
Author(s):  
Yahya Madkhali ◽  
Araci M. R. Rondon ◽  
Sophie Featherby ◽  
Anthony Maraveyas ◽  
John Greenman ◽  
...  

Procoagulant activity of tissue factor (TF) in response to injury or inflammation is accompanied with cellular signals which determine the fate of cells. However, to prevent excessive signalling, TF is rapidly dissipated through release into microvesicles, and/or endocytosis. To elucidate the mechanism by which TF signalling may become moderated on the surface of cells, the associations of TF, fVII/fVIIa, PAR2 and caveolin-1 on MDA-MB-231, BxPC-3 and 786-O cells were examined and compared to that in cells lacking either fVII/fVIIa or TF. Furthermore, the localisation of labelled-recombinant TF with cholesterol-rich lipid rafts was explored on the surface of primary human blood dermal endothelial cells (HDBEC). Finally, by disrupting the caveolae on the surface of HDBEC, the outcome on TF-mediated signalling was examined. The association between TF and PAR2 was found to be dependent on the presence of fVIIa. Interestingly, the presence of TF was not pre-requisite for the association between fVII/fVIIa and PAR2 but was significantly enhanced by TF, which was also essential for the proliferative signal. Supplementation of HDBEC with exogenous TF resulted in early release of fVII/fVIIa from caveolae, followed by re-sequestration of TF-fVIIa. Addition of labelled-TF resulted in the accumulation within caveolin-1-containing cholesterol-rich regions and was also accompanied with the increased assimilation of cell-surface fVIIa. Disruption of the caveolae/rafts in HDBEC using MβCD enhanced the TF-mediated cellular signalling. Our data supports a hypothesis that cells respond to the exposure to TF by moderating the signalling activities as well as the procoagulant activity of TF, through incorporation into the caveolae/lipid rafts.


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.


1997 ◽  
Vol 77 (05) ◽  
pp. 0975-0980 ◽  
Author(s):  
Angel Gálvez ◽  
Goretti Gómez-Ortiz ◽  
Maribel Díaz-Ricart ◽  
Ginés Escolar ◽  
Rogelio González-Sarmiento ◽  
...  

SummaryThe effect of desmopressin (DDAVP) on thrombogenicity, expression of tissue factor and procoagulant activity (PCA) of extracellular matrix (ECM) generated by human umbilical vein endothelial cells cultures (HUVEC), was studied under different experimental conditions. HUVEC were incubated with DDAVP (1, 5 and 30 ng/ml) and then detached from their ECM. The reactivity towards platelets of this ECM was tested in a perfusion system. Coverslips covered with DD A VP-treated ECMs were inserted in a parallel-plate chamber and exposed to normal blood anticoagulated with low molecular weight heparin (Fragmin®, 20 U/ml). Perfusions were run for 5 min at a shear rate of 800 s1. Deposition of platelets on ECMs was significantly increased with respect to control ECMs when DDAVP was used at 5 and 30 ng/ml (p <0.05 and p <0.01 respectively). The increase in platelet deposition was prevented by incubation of ECMs with an antibody against human tissue factor prior to perfusion. Immunofluorescence studies positively detected tissue factor antigen on DDAVP derived ECMs. A chromogenic assay performed under standardized conditions revealed a statistically significant increase in the procoagulant activity of the ECMs produced by ECs incubated with 30 ng/ml DDAVP (p <0.01 vs. control samples). Northern blot analysis revealed increased levels of tissue factor mRNA in extracts from ECs exposed to DDAVP. Our data indicate that DDAVP in vitro enhances platelet adhesion to the ECMs through increased expression of tissue factor. A similar increase in the expression of tissue factor might contribute to the in vivo hemostatic effect of DDAVP.


Sign in / Sign up

Export Citation Format

Share Document