Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Platelets ; 33(7): 1031-1042, 2022 Oct 03.
Article in English | MEDLINE | ID: mdl-35132909

ABSTRACT

Extracellular vesicles (EVs) released from activated platelets contain microRNAs, the most abundant of which is hsa-miR-223-3p. Endogenous hsa-miR-223-3p suppresses the expression of tissue factor (TF), the initiator of the extrinsic coagulation pathway, in endothelial cells. Monocytes can be induced to express TF to enhance coagulation, but the role of hsa-miR-223-3p in regulating monocyte TF remains unknown. This study examined whether hsa-miR-223-3p from platelet-derived EVs (pdEVs) affects TF expression in monocytes. THP-1 cells, differentiated into a monocyte-like phenotype with 1α,25-dihydroxyvitaminD3, were transfected with hsa-miR-223-3p mimic or control microRNA. Alternatively, THP-1 cells were incubated with pdEVs from PAR1-agonist peptide activated-platelets, as platelet releasate, or pdEVs isolated by ultracentrifugation. Transfection with hsa-miR-223-3p mimic resulted in significant reductions in TF protein, determined by western blotting and flow cytometry and reduced procoagulant activity, measured by a TF-specific factor Xa generation assay, compared to cells transfected with control microRNA. This reduction was reversed by co-transfection with hsa-miR-223-3p inhibitor, AntagomiR-223. Incubation of THP-1 cells with pdEVs also decreased TF expression; however, this was not reversed by AntagomiR-223. Taken together, monocyte TF expression is downregulated by hsa-miR-223-3p, but when transferred via pdEVs the effect was not reversed with Antagomir-223, suggesting other pdEV components may contribute to TF regulation.Abbreviations: Tissue factor (TF), Factor VII (FVII), activated Factor VII (FVIIa), Factor X (FX), activated Factor X (FXa), extracellular vesicles (EVs), microvesicles (MVs), platelet-derived extracellular vesicles (pdEVs), protease-activated receptor 1 agonist peptide (PAR1-AP), lipopolysaccharide (LPS), P-selectin glycoprotein ligand-1 (PSGL-1), Tris-Buffered Saline Tween (TBST), room temperature (RT)[Figure: see text].


Subject(s)
Extracellular Vesicles , MicroRNAs , Endothelial Cells/metabolism , Extracellular Vesicles/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Receptor, PAR-1/metabolism , Thromboplastin/genetics , Thromboplastin/metabolism
2.
Arch Biochem Biophys ; 697: 108702, 2021 01 15.
Article in English | MEDLINE | ID: mdl-33275878

ABSTRACT

Kynurenine-3-monooxygenase (KMO) is an important therapeutic target for several brain disorders that has been extensively studied in recent years. Potent inhibitors towards KMO have been developed and tested within different disease models, showing great therapeutic potential, especially in models of neurodegenerative disease. The inhibition of KMO reduces the production of downstream toxic kynurenine pathway metabolites and shifts the flux to the formation of the neuroprotectant kynurenic acid. However, the efficacy of KMO inhibitors in neurodegenerative disease has been limited by their poor brain permeability. Combined with virtual screening and prodrug strategies, a novel brain penetrating KMO inhibitor has been developed which dramatically decreases neurotoxic metabolites. This review highlights the importance of KMO as a drug target in neurological disease and the benefits of brain permeable inhibitors in modulating kynurenine pathway metabolites in the central nervous system.


Subject(s)
Brain/metabolism , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Kynurenine 3-Monooxygenase/antagonists & inhibitors , Neurodegenerative Diseases/drug therapy , Animals , Brain/drug effects , Drug Discovery , Enzyme Inhibitors/therapeutic use , Humans , Neurodegenerative Diseases/enzymology , Neurodegenerative Diseases/metabolism
3.
Biochim Biophys Acta Mol Cell Res ; 1865(1): 12-24, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28962834

ABSTRACT

The exposure and release of TF is regulated by post-translational modifications of its cytoplasmic domain. Here, the potential of Pin1 to interact with the cytoplasmic domain of TF, and the outcome on TF function was examined. MDA-MB-231 and transfected-primary endothelial cells were incubated with either Pin1 deactivator Juglone, or its control Plumbagin, as well as transfected with Pin1-specific or control siRNA. TF release into microvesicles following activation, and also phosphorylation and ubiquitination states of cellular-TF were then assessed. Furthermore, the ability of Pin1 to bind wild-type and mutant forms of overexpressed TF-tGFP was investigated by co-immunoprecipitation. Additionally, the ability of recombinant or cellular Pin1 to bind to peptides of the C-terminus of TF, synthesised in different phosphorylation states was examined by binding assays and spectroscopically. Finally, the influence of recombinant Pin1 on the ubiquitination and dephosphorylation of the TF-peptides was examined. Pre-incubation of Pin1 with Juglone but not Plumbagin, reduced TF release as microvesicles and was also achievable following transfection with Pin1-siRNA. This was concurrent with early ubiquitination and dephosphorylation of cellular TF at Ser253. Pin1 co-immunoprecipitated with overexpressed wild-type TF-tGFP but not Ser258→Ala or Pro259→Ala substituted mutants. Pin1 did interact with Ser258-phosphorylated and double-phosphorylated TF-peptides, with the former having higher affinity. Finally, recombinant Pin1 was capable of interfering with the ubiquitination and dephosphorylation of TF-derived peptides. In conclusion, Pin1 is a fast-acting enzyme which may be utilised by cells to protect the phosphorylation state of TF in activated cells prolonging TF activity and release, and therefore ensuring adequate haemostasis.


Subject(s)
Cell-Derived Microparticles/metabolism , NIMA-Interacting Peptidylprolyl Isomerase/physiology , Thromboplastin/metabolism , Cells, Cultured , Endothelial Cells/metabolism , Hemostasis/genetics , Humans , NIMA-Interacting Peptidylprolyl Isomerase/genetics , Phosphorylation , Protein Stability , Protein Transport , Secretory Pathway/genetics
4.
Biochim Biophys Acta ; 1863(11): 2846-2857, 2016 11.
Article in English | MEDLINE | ID: mdl-27599717

ABSTRACT

Restriction of tissue factor (TF) activity at the cell surface and TF release are critical for prevention of excessive coagulation. This study examined the regulation of TF dephosphorylation and its release through ubiquitination. A plasmid containing the sequence to express the tandem protein TF-tGFP was mutated to include an arginine-substitution at Lys255 within TF. MDA-MB-231 cell line, and HCAEC endothelial cells were transfected and subsequently activated with PAR2-agonist peptide. The wild-type and mutant TF-tGFP were immunoprecipitated from the cell lysates and the ubiquitination and phosphorylation state of TF examined. Analysis of the proteins showed that arginine-substitution of Lys255 within TF prevented its ubiquitination while the wild-type TF-tGFP was oligoubiquitinated. The TF-associated oligoubiquitin chain was estimated to contain up to 4 ubiquitin units, with the linkage formed between Lys63 of one ubiquitin unit, and the C-terminus of the next unit. The Lys255→Arg substitution of TF-tGFP prolonged the phosphorylation of Ser253 within TF, compared to the wild-type TF-tGFP, lengthened the presence of TF-tGFP at the cell surface and extended the duration of TF-tGFP release from cells following PAR2 activation. A biotinylated 19-mer peptide corresponding to the C-terminus of TF (TFc) was used as substrate to show that the ubiquitination of TF was mediated by the Ube2D family of E2-enzymes and involved Mdm2. Moreover, double-phosphorylation of TFc was prerequisite for ubiquitination, with subsequent dephosphorylation of Ser253 by phosphatase PP2A. In conclusion, oligoubiquitination of Lys255 within TF permits PP2A to bind and dephosphorylate Ser253 and occurs to terminate TF release and contain its activity.


Subject(s)
Blood Coagulation , Endothelial Cells/metabolism , Thromboplastin/metabolism , Blood Coagulation/drug effects , Cell Line, Tumor , Endothelial Cells/drug effects , Female , Humans , Lysine , Oligopeptides/pharmacology , Phosphorylation , Protein Binding , Protein Phosphatase 2/metabolism , Proto-Oncogene Proteins c-mdm2/metabolism , Receptor, PAR-2/agonists , Receptor, PAR-2/metabolism , Serine , Thromboplastin/chemistry , Thromboplastin/genetics , Time Factors , Transfection , Ubiquitination
5.
J Biol Chem ; 286(14): 11977-84, 2011 Apr 08.
Article in English | MEDLINE | ID: mdl-21310953

ABSTRACT

The mechanisms that regulate the incorporation and release of tissue factors (TFs) into cell-derived microparticles are as yet unidentified. In this study, we have explored the regulation of TF release into microparticles by the phosphorylation of serine residues within the cytoplasmic domain of TF. Wild-type and mutant forms of TF, containing alanine and aspartate substitutions at Ser253 and Ser258, were overexpressed in coronary artery and dermal microvascular endothelial cells and microparticle release stimulated with PAR2 agonist peptide (PAR2-AP). The release of TF antigen and activity was then monitored. In addition, the phosphorylation state of the two serine residues within the released microparticles and the cells was monitored for 150 min. The release of wild-type TF as procoagulant microparticles peaked at 90 min and declined thereafter in both cell types. The TF within these microparticles was phosphorylated at Ser253 but not at Ser258. Aspartate substitution of Ser253 resulted in rapid release of TF antigen but not activity, whereas TF release was reduced and delayed by alanine substitution of Ser253 or aspartate substitution of Ser258. Alanine substitution of Ser258 prolonged the release of TF following PAR2-AP activation. The release of TF was concurrent with phosphorylation of Ser253 and was followed by dephosphorylation at 120 min and phosphorylation of Ser258. We propose a sequential mechanism in which the phosphorylation of Ser253 through PAR2 activation results in the incorporation of TF into microparticles, simultaneously inducing Ser258 phosphorylation. Phosphorylation of Ser258 in turn promotes the dephosphorylation of Ser253 and suppresses the release of TF.


Subject(s)
Cell-Derived Microparticles/metabolism , Serine/metabolism , Thromboplastin/metabolism , Alanine/genetics , Alanine/metabolism , Apoptosis/drug effects , Aspartic Acid/genetics , Aspartic Acid/metabolism , Blotting, Western , Cell-Derived Microparticles/drug effects , Cells, Cultured , Humans , Oligopeptides/pharmacology , Phosphorylation/drug effects , Serine/genetics , Signal Transduction/drug effects , Thromboplastin/genetics
6.
Biomedicines ; 10(9)2022 Sep 15.
Article in English | MEDLINE | ID: mdl-36140394

ABSTRACT

The flavoprotein kynurenine 3-monooxygenase (KMO) is localised to the outer mitochondrial membrane and catalyses the synthesis of 3-hydroxykynurenine from L-kynurenine, a key step in the kynurenine pathway (KP) of tryptophan degradation. Perturbation of KP metabolism due to inflammation has long been associated with the pathogenesis of several neurodegenerative disorders, including Huntington's disease (HD)-which is caused by the expansion of a polyglutamine stretch in the huntingtin (HTT) protein. While HTT is primarily localised to the cytoplasm, it also associates with mitochondria, where it may physically interact with KMO. In order to test this hypothesis, we employed bimolecular fluorescence complementation (BiFC) and found that KMO physically interacts with soluble HTT exon 1 protein fragment in living cells. Notably, expansion of the disease-causing polyglutamine tract in HTT leads to the formation of proteinaceous intracellular inclusions that disrupt this interaction with KMO, markedly decreasing BiFC efficiency. Using confocal microscopy and ultrastructural analysis, we determined KMO and HTT localisation within the cell and found that the KMO-HTT interaction is localized to the outer mitochondrial membrane. These data suggest that KMO may interact with a pool of HTT at the mitochondrial membrane, highlighting a possible physiological role for mitochondrial HTT. The KMO-HTT interaction is abrogated upon polyglutamine expansion, which may indicate a heretofore unrecognized relevance in the pathogenesis of this disorder.

7.
Arterioscler Thromb Vasc Biol ; 30(9): 1810-7, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20616308

ABSTRACT

OBJECTIVE: Increased levels of circulating tissue factor (TF) in the form of microparticles increase the risk of thrombosis. However, any direct influence of microparticle-associated TF on vascular endothelial cell proliferation is not known. In this study, the influence of recombinant and microparticle-associated TF on endothelial cell proliferation and mitogen-activated protein kinase signaling mechanisms was examined. METHODS AND RESULTS: Incubation of human coronary artery endothelial cells with lipidated recombinant full-length TF, or TF-containing microparticles (50 to 200 pmol/L TF), increased the rate of cell proliferation and induced phosphorylation of extracellular signal regulated kinase 1 in a TF-dependent manner. Inhibition of extracellular signal regulated kinase 1/2 using PD98059 or extracellular signal regulated kinase 1/2 antisense oligonucleotides or inhibition of c-Jun N-terminal kinase reduced recombinant TF-mediated cell proliferation. PD98059 also reduced cell proliferation in response to TF-containing microparticles. Inclusion of FVIIa (5 nmol/L) and FXa (10 nmol/L) or preincubation of cells with an inhibitory anti-FVIIa antibody had no additional influence on TF-mediated cell proliferation. However, preincubation of exogenous TF with a beta1-integrin peptide (amino acids 579 to 799) reduced TF-mediated proliferation. CONCLUSIONS: High concentrations of recombinant or microparticle-associated TF stimulate endothelial cell proliferation through activation of the extracellular signal regulated kinase 1/2 pathway, mediated through a novel mechanism requiring the interaction of exogenous TF with cell surface beta1-integrin and independent of FVIIa.


Subject(s)
Cell Proliferation , Cell-Derived Microparticles/metabolism , Endothelial Cells/enzymology , Integrin beta1/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Signal Transduction , Thromboplastin/metabolism , Apoptosis , Cell Proliferation/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Endothelial Cells/drug effects , Endothelial Cells/pathology , Enzyme Activation , Factor VIIa/metabolism , Factor Xa/metabolism , Humans , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/genetics , Oligonucleotides, Antisense/metabolism , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Recombinant Proteins/metabolism , Signal Transduction/drug effects , Time Factors
8.
Mol Cell Biochem ; 345(1-2): 119-30, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20730477

ABSTRACT

Recent evidence has shown that prolonged exposure to exogenous tissue factor (TF) can alter the cellular functions of cardiomyocytes resulting in cardiac dysfunction. The effect of TF may arise from local inflammation within or in the vicinity of the heart. The aim of this study was to investigate the effect of TF on cardiomyocyte proliferation and growth. H9c2 rat cardiomyocytes were exposed to a range of concentrations of recombinant TF (rTF) (1.3-52 ng/ml) for up to 10 days and the outcome on cell proliferation and induction of apoptosis measured. At lower concentrations examined (1.3 ng/ml), rTF had a proliferative influence on the H9c2 cells. In contrast, elevated concentrations of rTF (52 ng/ml) induced cellular apoptosis as indicated by increased caspase-3 activity and nuclear localisation of p53. Moreover, incubation with intermediate concentrations of rTF (13 ng/ml) resulted in an initial increase in proliferation but subsequently, led to cellular apoptosis by day 7 of the incubation. In order to determine if these effects induced hypertrophic cell growth, expression of mechano-growth factor (MGF) was analysed. Incubation of cells with rTF resulted in enhanced expression of MGF particularly at the intermediate concentrations of rTF (13 ng/ml) as well as mean cellular transverse diameter. In addition, there was a rapid increase in the expression of atrial natriuretic factor (ANF) in the cells, on incubation with rTF but diminished rapidly when exposed to higher concentrations of rTF. These data indicate that exposure to increasing concentrations of rTF can accelerate the rate of cardiomyocyte turnover which may ultimately lead to depletion of viable cells within the heart. Moreover, at lower concentrations of rTF, the induction of cell proliferation together with hypertrophic markers indicates that rTF may contribute to the induction and progression of cardiac hypertrophy.


Subject(s)
Apoptosis/drug effects , Cardiomegaly/chemically induced , Cell Proliferation/drug effects , Myocytes, Cardiac/pathology , Thromboplastin/pharmacology , Animals , Atrial Natriuretic Factor , Cell Line , Dose-Response Relationship, Drug , Insulin-Like Growth Factor I , Rats , Recombinant Proteins
9.
Mol Cancer Res ; 6(12): 1807-18, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19074826

ABSTRACT

Increased expression of tissue factor (TF) has been associated with invasive forms of breast cancer. Conversely, the loss of estrogen receptor alpha (ERalpha) is associated with increased cell invasiveness. We have examined the influence of exogenous truncated recombinant TF (rTF) on ERalpha expression and cell invasiveness and investigated the mechanism of rTF signaling. The influence of rTF on ERalpha expression in MCF-7 and T47D cell lines was investigated using reverse transcription-PCR and ELISA. Cell invasion was measured using Boyden chamber-based invasion assays. Additionally, the interaction of fluorescein-labeled rTF with the surface of MCF-7 cells and particularly with beta(1)-integrin was examined. Treatment of cells with rTF resulted in the down-regulation of ERalpha mRNA and protein over 24 h, which required beta(1)-integrin and involved the mitogen-activated protein kinase pathway but did not require PAR2 activation. The addition of rTF reduced estradiol-mediated cell proliferation as well as increased cell invasiveness requiring both PAR2 and beta(1)-integrin activation. Fluorescein-labeled rTF was shown to bind to the surface of MCF-7 cells within 5 min and peaked at 15 min. The bound rTF colocalized with cellular beta(1)-integrin and was disrupted in the presence of excess unlabeled rTF and an anti-beta(1) polyclonal antibody. Finally, affinity purification of beta(1)-integrin using rTF-conjugated agarose showed a requirement for the presence of divalent cations but not factor VIIa. The results indicate that rTF is capable of down-regulating ERalpha expression in breast cancer cells, resulting in decreases in estrogen-mediated cell proliferation and increased invasiveness. Furthermore, the mechanisms by which rTF induces these changes involve both PAR2 and beta(1)-integrin.


Subject(s)
Breast Neoplasms , Estrogen Receptor alpha/genetics , Integrin beta1/metabolism , MAP Kinase Signaling System/physiology , Receptor, PAR-2/metabolism , Thromboplastin/pharmacology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Breast Neoplasms/physiopathology , Cell Division/physiology , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/drug effects , Hemostatics/metabolism , Hemostatics/pharmacology , Humans , Neoplasm Invasiveness/physiopathology , Receptors, Cell Surface/metabolism , Thromboplastin/metabolism
10.
Thromb Res ; 175: 13-20, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30677622

ABSTRACT

Alterations in the endothelial permeability occur in response to the activation of coagulation mechanisms in order to control clot formation. The activation of the protease activated receptors (PAR) can induce signals that regulate such cellular responses. PAR2 is a target for the coagulation factor Xa (fXa) and tissue factor-factor VIIa (TF-fVIIa) complex. By measuring the permeability of dextran blue across endothelial monolayer, we examined the mechanisms linking coagulation and endothelial permeability. Activation of PAR2 using the agonist peptide (PAR2-AP) resulted in increased permeability across the monolayer and was comparable to that obtained with VEGF at 60 min. Incubation of cells with activated factor Xa (fXa) resulted in an initial decrease in permeability by 30 min, but then significantly increased at 60 min. These responses required fXa activity, and were abrogated by incubation of the cells with a PAR2-blocking antibody (SAM11). Activation of PAR2 alone, or inhibition of PAR1, abrogated the initial reduction in permeability. Additionally, inclusion of Rivaroxaban (0.6 µg/ml) significantly inhibited the response to fXa. Finally, incubation of the endothelial monolayers up to 2 h with TF-containing microvesicles derived from MDA-MB-231 cells, in the presence or absence of fVIIa, did not influence the permeability across the monolayers. In conclusion, fXa but not TF-fVIIa is a noteworthy mediator of endothelial permeability. The rapid initial decrease in permeability requires PAR2 and PAR1 which may act to constrain bleeding. The longer-term response is mediated by PAR2 with increased permeability, presumably to enhance clot formation at the site of damage.


Subject(s)
Endothelium/metabolism , Factor VIIa/metabolism , Factor Xa/metabolism , Receptor, PAR-2/metabolism , Cell Line, Tumor , Humans , Permeability
11.
Microvasc Res ; 76(3): 152-60, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18725234

ABSTRACT

Hyperglycaemia and the associated formation of advanced glycation end-products (AGE) have been implicated in the pathogenesis of diabetic vasculopathy. In addition to its role in coagulation, tissue factor (TF) is known to regulate vascular proliferation and angiogenesis. In this study, the influence of AGE and glucose on the expression of TF in human renal mesangial cells (HRMC) and the subsequent induction of capillary formation by human dermal microvascular endothelial cells (HDMEC) were measured. Furthermore, the activity of TF, incorporated into microparticles was investigated. Both AGE and elevated glucose were capable of upregulating the expression of TF expression in a concentration-dependent manner in HRMC but not in HDMEC. This TF antigen and activity in the conditioned media from HRMC was associated with microparticles. Moreover, the formation of capillaries was readily induced on supplementation of HDMEC with conditioned media, from AGE-treated or high glucose-treated HRMC but not on incubation of HDMEC with either AGE or hyperphysiological concentrations of glucose. Furthermore, the rate of capillary formation was suppressed on incubation of the conditioned media with a polyclonal antibody against TF but not against VEGF. This study indicates that TF-containing microparticles are an important pro-inflammatory mediator acting as a mediator between elevated glucose and the development of diabetic vasculopathy by altering the angiogenic properties of endothelial cells and offers one explanation for the correlation between diabetes and microvascular disease.


Subject(s)
Endothelial Cells/drug effects , Endothelial Cells/physiology , Glucose/pharmacology , Glycation End Products, Advanced/pharmacology , Mesangial Cells/drug effects , Mesangial Cells/physiology , Serum Albumin/pharmacology , Thromboplastin/physiology , Base Sequence , Cell-Derived Microparticles/physiology , Cells, Cultured , Culture Media, Conditioned , DNA Primers/genetics , Diabetic Angiopathies/etiology , Diabetic Angiopathies/pathology , Diabetic Angiopathies/physiopathology , Endothelial Cells/cytology , Humans , Inflammation Mediators/physiology , Mesangial Cells/ultrastructure , Microvessels/cytology , Microvessels/drug effects , Microvessels/growth & development , Neovascularization, Pathologic/etiology , Neovascularization, Pathologic/physiopathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Serum Albumin, Human , Skin/blood supply , Thromboplastin/antagonists & inhibitors , Thromboplastin/genetics
12.
Atherosclerosis ; 190(2): 343-51, 2007 Feb.
Article in English | MEDLINE | ID: mdl-16697385

ABSTRACT

The association between Chlamydia pneumoniae (C. pneumoniae) infection and the onset and progression of atherosclerosis has become apparent recently. Moreover, increased expression of tissue factor (TF) as a result of C. pneumoniae infection has been previously demonstrated. We have examined the expression of TF on the surface of endothelial cells and the release of TF-containing cell-derived microparticles, over seven days. Additionally, using cells expressing a procoagulantly active EGFP-TF hybrid protein, we examined the kinetics of TF trafficking on the cells and incorporation into shed microparticles. Finally, in an attempt to associate this with the activation of NFkappaB, we used a luciferase reporter to measure the duration of the activation of this transcription factor. TF-containing microparticles were released within 24h of infection and continued for up to 7 days. Moreover, the initial release of TF containing microparticles was associated with NFkappaB activation and was suppressed on inclusion of an NFkappaB inhibitor, pyrrolidinedithiocarbamate ammonium. Moreover, persistent dissemination of TF-containing microparticles at later stages of infection was associated with the release of the infective C. pneumoniae elementary bodies. The released procoagulant, cellular microparticles are known to be strongly atherogenic and therefore we suggest a mechanism for the involvement of C. pneumoniae in the onset and progression of vascular disease.


Subject(s)
Chlamydophila Infections/physiopathology , Chlamydophila pneumoniae , Endothelium, Vascular/microbiology , Endothelium, Vascular/physiology , Thromboplastin/genetics , Base Sequence , Cell Line , DNA Primers , Humans , Reverse Transcriptase Polymerase Chain Reaction , Thromboplastin/metabolism
13.
Blood Coagul Fibrinolysis ; 28(6): 452-459, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28151805

ABSTRACT

: Tissue factor (TF) is not usually expressed by endothelial cells but can be induced in these cells by inflammatory cytokines. Many studies have used human umbilical vein endothelial cells (HUVEC) as a model to examine the regulation of TF expression. However, there is a question as to whether this reflects TF expression in adult endothelial cells. This study compared TF expression and the release of TF-positive microvesicles in HUVEC and adult human dermal blood endothelial cells (HDBEC) in response to tumour necrosis factor α (TNFα) and interleukin-1 ß (IL-1ß). Cells were treated with the inflammatory cytokines and TF mRNA and total protein expression was examined by real-time RT-PCR and TF ELISA. Cell surface TF activity was measured in the calibrated automated thrombogram assay, as were microvesicle concentrations and microvesicle-associated TF activity. The TF antigen content of the microvesicles was determined by TF ELISA. Both HUVEC and HDBEC expressed increased levels of TF mRNA in response to TNFα and IL-1ß within 2 h. TF antigen expression increased in both cell types, reaching a maximum at 6 h, with HUVEC expressing significantly higher levels compared with HDBEC in response to TNFα. However, increases in TF-specific thrombin generation were similar on both HUVEC and HDBEC and both cell types also released comparable levels of TF-positive microvesicles. HUVEC and HDBEC respond similarly to TNFα and IL-1ß in terms of TF expression, and both are suitable models to examine cell surface TF activity and TF-positive microvesicle release in endothelial cells.


Subject(s)
Endothelial Cells/metabolism , Thromboplastin/metabolism , Adult , Cell-Derived Microparticles/metabolism , Fetus/cytology , Gene Expression/drug effects , Human Umbilical Vein Endothelial Cells , Humans , Interleukin-1beta/pharmacology , RNA, Messenger/analysis , Thromboplastin/analysis , Thromboplastin/genetics , Tumor Necrosis Factor-alpha/pharmacology
14.
Thromb Haemost ; 117(11): 2034-2044, 2017 11.
Article in English | MEDLINE | ID: mdl-29044292

ABSTRACT

We have previously shown that phosphorylation of tissue factor (TF) at Ser253 increases the incorporation of TF into microvesicles (MVs) following protease-activated receptor 2 (PAR2) activation through a process involving filamin A, whereas phosphorylation of TF at Ser258 suppresses this process. Here, we examined the contribution of the individual phosphorylation of these serine residues to the interaction between filamin A and TF, and further examined how filamin A regulates the incorporation of TF into MVs. In vitro binding assays using recombinant filamin A C-terminal repeats 22-24 with biotinylated phospho-TF cytoplasmic domain peptides as bait showed that filamin A had the highest binding affinities for phospho-Ser253 and double-phosphorylated TF peptides, while the phospho-Ser258 TF peptide had the lowest affinity. Analysis of MDA-MB-231 cells using an in situ proximity ligation assay revealed increased proximity between the C-terminus of filamin A and TF following PAR2 activation, which was concurrent with Ser253 phosphorylation and TF-positive MV release from these cells. Knock-down of filamin A expression suppressed PAR2-mediated increases in cell surface TF procoagulant activity without reducing cell surface TF antigen expression. Disrupting lipid rafts by pre-incubation with methyl-ß-cyclodextrin prior to PAR2 activation reduced TF-positive MV release and cell surface TF procoagulant activity to the same extent as filamin A knock-down. In conclusion, this study shows that the interaction between TF and filamin A is dependent on the differential phosphorylation of Ser253 and Ser258. Furthermore, the interaction of TF with filamin A may translocate cell surface TF to cholesterol-rich lipid rafts, increasing cell surface TF activity as well as TF incorporation and release into MVs.


Subject(s)
Cell-Derived Microparticles/metabolism , Filamins/metabolism , Thromboplastin/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Endothelial Cells/metabolism , Female , Filamins/genetics , Humans , Membrane Microdomains/metabolism , Phosphorylation , Protein Binding , Protein Interaction Domains and Motifs , Protein Transport , Receptor, PAR-2/metabolism , Serine , Thromboplastin/genetics , Time Factors
15.
Thromb Haemost ; 111(4): 647-55, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24258684

ABSTRACT

We previously reported that the incorporation of tissue factor (TF) into cell-derived microvesicles (MVs) is regulated by the phosphorylation of the cytoplasmic domain of TF. Since the cytoskeletal protein filamin-A is known to bind to the cytoplasmic domain of TF in a phosphorylation-dependent manner, the involvement of filamin-A in the incorporation of TF into MVs was examined. Endothelial cells were transfected to express TF, whereas MDA-MB-231 cells were used to examine endogenously expressed TF. MV release was induced by activating protease-activated receptor-2 (PAR2). Partial suppression of filamin-A expression using two different filamin-A siRNA sequences resulted in significant reductions in the incorporation of TF antigen into MVs as determined by TF-ELISA and western blot analysis, and was reflected in reduced thrombin-generation and FXa-generation capacities of these MVs. Deletion of the cytoplasmic domain of TF also resulted in reduced incorporation of TF into MVs, whereas the suppression of filamin-A expression had no additional effect on the incorporation of truncated TF into MVs. Partial suppression of filamin-A expression had no effect on the number and size distribution of the released MVs. However, >90% suppression of filamin-A expression resulted in increased MV release, possibly as a result of increased instability of the plasma membrane and underlying cytoskeleton. In conclusion, the presence of filamin-A appears to be essential for the incorporation of TF into MVs following PAR2 activation, but is not required for the process of MV formation and release following PAR2 activation.


Subject(s)
Cell-Derived Microparticles/metabolism , Endothelial Cells/physiology , Factor Xa/biosynthesis , Filamins/metabolism , Receptor, PAR-2/metabolism , Thrombin/biosynthesis , Thromboplastin/metabolism , Cell-Derived Microparticles/chemistry , Cells, Cultured , Factor Xa/genetics , Filamins/genetics , Humans , Particle Size , Protein Engineering , Protein Structure, Tertiary/genetics , RNA, Small Interfering/genetics , Secretory Pathway , Sequence Deletion/genetics , Thrombin/genetics , Thromboplastin/genetics
16.
Article in English | MEDLINE | ID: mdl-25206957

ABSTRACT

Microvesicles were isolated from the conditioned media of 3 cell lines (MDA-MB-231, AsPC-1 and A375) by ultracentrifugation at a range of relative centrifugal forces, and the tissue factor (TF) protein and activity, microvesicle number, size distribution and relative density compared. Also, by expressing TF-tGFP in cells and isolating the microvesicles, the relative density of TF-containing microvesicles was established. Nanoparticle tracking analysis (NTA) indicated that the larger-diameter microvesicles (>200 nm) were primarily sedimented at 100,000g and possessed TF-dependent thrombin and factor Xa generation potential, while in the absence of factor VII, all microvesicles possessed some thrombin generation capacity. Immuno-precipitation of TF-containing microvesicles followed by NTA also indicated the range of these microvesicles to be 200-400 nm. Analysis of the microvesicles by gradient density centrifugation showed that lower-density (<1.1 g/ml) microvesicles were mainly present in the samples recovered at 100,000g and were associated with TF antigen and activity. Analysis of these fractions by NTA confirmed that these fractions were principally composed of the larger-diameter microvesicles. Similar analysis of microvesicles from healthy or patient plasma supported those obtained from conditioned media indicating that TF activity was mainly associated with lower-density microvesicles. Furthermore, centrifugation of healthy plasma, supplemented with TF-tGFP-containing microvesicles, resulted in 67% retrieval of the fluorescent microvesicles at 100,000g, but only 26% could be recovered at 20,000g. Pre-centrifugation of conditioned media or plasma at 10,000g improved the speed and yield of recovered TF-containing microvesicles by subsequent centrifugation at either 20,000g or 100,000g. In conclusion, TF appears to be associated with low-density (1.03-1.08 g/ml), larger-diameter (200-350 nm) microvesicles.

17.
Thromb Haemost ; 109(1): 61-71, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23152142

ABSTRACT

The role of tissue factor (TF)-containing microparticles in clot propagation has been established, but the ability of circulating microparticles to initiate coagulation has been disputed. However, TF-bearing microparticles, particularly endothelial-microparticles generated during disease, may interact with extracellular matrices which in turn can localise circulating TF to sites of injury. In order to examine this hypothesis in vitro , microparticles were isolated from human coronary artery endothelial cells transfected to overexpress TF, tumour-necrosis factor (TNF) α-treated cells or non-transfected cells lacking TF. The ability of microparticles to bind collagen-IV, fibronectin and fibrin was examined under static conditions and arterial shear rates (650 s⁻¹), and also in the presence of inhibitory antibodies against ß1-, ß3-, α3- and αv-integrins or an anti-TF antibody. TF-microparticles showed increases of up to 43% and 24% in adherence to collagen-IV and fibronectin, respectively, compared to control microparticles under shear flow. Furthermore, TF-containing microparticles, but not the transfected parent cells had increased levels of ß1-integrin compared to TF-deficient microparticles. Pre-incubation of microparticles with a ß1-integrin-blocking antibody counteracted the additional adhesion of TF-microparticles compared to control microparticles. Finally, adherence of TF microparticles to collagen-IV or fibronectin resulted in increased TF activity by concentrating TF onto the surface. In conclusion, the presence of TF within microparticles enhances the interactions of endothelial cell-derived microparticles with extracellular matrices in an integrin-dependent manner. Accumulation and localisation of these microparticles in turn results in the enhancement of TF activity. This may be an innate mechanism by which TF-bearing microparticles induce coagulation upon vascular injury.


Subject(s)
Blood Coagulation , Cell-Derived Microparticles/metabolism , Collagen Type IV/metabolism , Endothelial Cells/metabolism , Fibronectins/metabolism , Thromboplastin/metabolism , Antibodies, Blocking/metabolism , Cells, Cultured , Fibrin/metabolism , Humans , Integrin alpha3/immunology , Integrin alpha3/metabolism , Integrin alphaV/immunology , Integrin alphaV/metabolism , Integrin beta1/immunology , Integrin beta1/metabolism , Integrin beta3/immunology , Integrin beta3/metabolism , Protein Binding , Thromboplastin/genetics , Thromboplastin/immunology , Time Factors , Transfection , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation
18.
Cancer Immunol Immunother ; 57(9): 1347-55, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18297283

ABSTRACT

AIMS: We previously reported that overexpression of tissue factor (TF) protected HT29 tumour cells from cellular cytotoxicity through a mechanism requiring the presence of the cytoplasmic domain of TF. In this investigation the mechanism of TF-mediated immune evasion has been examined. METHODS: The influence of alanine-substitution at Ser253 and Ser258 of TF (TF(Ala253) and TF( Ala258)) on the induction of cytotoxic evasion, as well as expression of vascular cell adhesion molecule-1 and intra-cellular adhesion molecule-1 (VCAM-1 and ICAM-1) was investigated. Moreover, we examined the effect of transfection of four 20-mer peptides, corresponding to the C-terminal residues of TF, with different phosphorylation states, on promotion of evasion from cell cytotoxicity. RESULTS: Cells overexpressing TF(Ala258) and to a lesser extent overexpressing TF(Ala253,) exhibited a reduced ability to evade cellular cytotoxicity compared to cells overexpressing the wild-type TF. Furthermore, the increase in protection acquired was greatest on transfection of Ser258-phosphsorylated form of the cytoplasmic peptide, lower in double-phosphorylated and Ser253-phosphorylated peptides respectively, and lowest in the unphosphorylated form. Finally, the expression of VCAM-1 mRNA as well as surface antigen was reduced on overexpression of TF(wt) but was partially reverted in the cells transfected to overexpress TF(Ala253) or TF(Ala258). CONCLUSIONS: These data show that the phosphorylation of TF at Ser258 and to a lesser extent Ser253, plays an essential role in the protective influence of TF on immune evasion by tumour cells, and that the mechanism could involve the downregulation of key surface antigens, such as adhesion proteins, involved in cell:cell interaction.


Subject(s)
Gene Expression Regulation, Neoplastic , Thromboplastin/metabolism , Alanine/chemistry , Cell Line, Tumor , Humans , Immune System , Models, Biological , Mutagenesis, Site-Directed , Neoplasm Metastasis , Peptides/chemistry , Phosphorylation , Protein Structure, Tertiary , Serine/chemistry , Transfection , Vascular Cell Adhesion Molecule-1/metabolism
19.
Atherosclerosis ; 194(1): 88-101, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17137581

ABSTRACT

In this study we examined the ability of tissue factor (TF) alone, or in conjunction with factor VIIa, factor Xa and TFPI in activating a number of key signalling pathways associated with cellular growth, stress and differentiation responses in human endothelial cells. We used luciferase reporter systems to demonstrate the activation of p42/44 MAPK by the TF-FVIIa complex, mediated via the PAR1 receptor. TF alone was capable of interacting with the cell surface and was sufficient to activate the JNK-SAPK pathway and subsequently AP-1, but the level of activation was enhanced by the activity of FXa on PAR1 and 2. Furthermore, the phosphorylated form of the transmembrane-cytoplasmic domain of TF was directly responsible for activation of these pathways. CREB activation occurred in response to TF-FVIIa in a non-protease dependent manner but was lowered on addition of FXa. Finally, NFkappaB activation occurred in response to FVIIa or FXa, with the latter exhibiting higher levels of activation. In conclusion, we have shown that TF is capable of activating differing signalling pathways, via more than one mechanism. The differential influence of TF is modified depending on the presence of other coagulation factors and ultimately acts as a deciding factor in the determination of cellular fate.


Subject(s)
Endothelial Cells/metabolism , MAP Kinase Signaling System/physiology , Thromboplastin/metabolism , Cells, Cultured , Cyclic AMP Response Element-Binding Protein/metabolism , Drug Synergism , Endothelial Cells/cytology , Endothelium, Vascular/cytology , Factor VIIa/metabolism , Factor VIIa/pharmacology , Factor Xa/metabolism , Factor Xa/pharmacology , Flow Cytometry , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Lipoproteins/metabolism , Lipoproteins/pharmacology , Luciferases/genetics , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , NF-kappa B/metabolism , Thromboplastin/genetics , Thromboplastin/pharmacology , Transcription Factor AP-1/metabolism , Transfection , Umbilical Veins/cytology
20.
Cancer Immunol Immunother ; 55(11): 1301-8, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16453151

ABSTRACT

AIMS: The association between tissue factor (TF) expression and increased rate of tumour metastasis is well established. In this study, we have examined the hypothesis that the expression of TF by disseminated tumour cells confers protection against immune recognition and cytotoxicity. MATERIALS AND METHODS: A hybrid EGFP-TF protein was expressed in HT29 colon carcinoma and K562 lymphoblast cell lines. To assess the cytotoxic activity against tumour cells over-expressing TF, a novel method was used, based on the direct measurement of fluorescently labelled HT29 or K562 target cells. RESULTS: Upon challenge with peripheral blood mononuclear cells (PBMC), tumour cells expressing TF partially evaded cellular cytotoxicity (Delta=15-40% reduction in cytotoxicity). Moreover, the influence of TF was not primarily dependent on its procoagulant function, although the inclusion of 20% (v/v) plasma did lower the rate of cytotoxicity against untransfected cells. However, expression of a truncated form of TF, devoid of the cytoplasmic domain, did not mediate any degree of inhibition of cytotoxicity, suggesting that the protective function of TF is principally due to this domain. CONCLUSIONS: We conclude that TF can promote immune evasion in tumour cells expressing this protein leading to increased survival and therefore metastatic rate in such cells.


Subject(s)
Colorectal Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Lymphocytes/metabolism , Thromboplastin/physiology , Cell Line, Tumor , Colorectal Neoplasms/pathology , Cytoplasm/metabolism , Humans , K562 Cells , Leukocytes, Mononuclear/metabolism , Neoplasm Metastasis , Protein Structure, Tertiary , Prothrombin Time , Thromboplastin/biosynthesis , Thromboplastin/genetics
SELECTION OF CITATIONS
SEARCH DETAIL