Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Thromb Res ; 238: 206-207, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38733692
2.
Thromb Res ; 229: 90-98, 2023 09.
Article in English | MEDLINE | ID: mdl-37421683

ABSTRACT

BACKGROUND: Colorectal cancer (CRC) is the third most prevalent cancer type. CRC-patients are at increased risk of venous and arterial thromboembolism (TE), but the magnitude of the risks, their predictors and consequences are not exactly known. OBJECTIVES: We aimed to determine incidence, predictors and prognosis of TE after incident CRC in a large, unselected population. METHODS: Using data from Statistics Netherlands and the Netherlands Comprehensive Cancer Organization, all incident CRC-patients were identified between 2013 and 2018 plus a sample of 1:2 age- and sex-matched control subjects. Incidence rates and cumulative incidences for TE were estimated. Predictor variables for TE were explored by univariable Cox regression. The association between TE and all-cause mortality was evaluated by multivariable time-dependent Cox regression. RESULTS: 68,238 incident CRC-patients were matched to 136,476 controls. CRC-patients had a 1-year cumulative venous TE (VTE) incidence of 1.93 % (95%CI 1.83-2.04), versus 0.24 % (95%CI 0.21-0.27) in controls (HR 8.85; 95%CI 7.83-9.99). For arterial TE (ATE), this was 2.74 % (95%CI 2.62-2.87) in CRC versus 1.88 % (95%CI 1.81-1.95) in controls (HR 1.57; 95%CI 1.47-1.66). Cancer stage, surgery, chemotherapy and asthma were predictors for VTE, whereas age, prior ATE and Parkinson's disease were predictors for ATE. CRC patients with TE had an increased risk of all-cause mortality (VTE HR; 3.68 (95%CI 3.30-4.10, ATE HR; 3.05 (95%CI 2.75-3.39)) compared with CRC-patients without TE. CONCLUSIONS: This Dutch nationwide cohort study adds detailed knowledge on the risk of VTE and ATE, their predictors and prognosis in CRC-patients. These findings may drive TE prophylactic management decisions.


Subject(s)
Colorectal Neoplasms , Venous Thromboembolism , Humans , Venous Thromboembolism/epidemiology , Venous Thromboembolism/etiology , Incidence , Cohort Studies , Netherlands/epidemiology , Prognosis , Colorectal Neoplasms/complications , Colorectal Neoplasms/epidemiology , Risk Factors
3.
Thromb Res ; 221: 51-57, 2023 01.
Article in English | MEDLINE | ID: mdl-36470070

ABSTRACT

Local coagulation activation has been shown to impact both primary tumor growth and metastasis in mice. It is well known that components of the blood clotting cascade such as tissue factor and thrombin play a role in tumor progression by activating cellular receptors and local formation of fibrin. However, whether venous thromboembolism (VTE) or a hypercoagulable state has a direct impact on cancer progression is unknown. Here we have combined an orthotopic murine breast cancer model, using female Nod-SCID mice, with siRNA-mediated silencing of antithrombin (siAT) leading to the induction of a systemic hypercoagulable state. We show that, compared to control siRNA-treated (not experiencing a hypercoagulable state) tumor-bearing mice, siAT treated tumor-bearing mice do not show enhanced tumor growth nor enhanced metastasis. We conclude that, in this murine model for hypercoagulability, induction of a hypercoagulable state does not contribute to breast cancer progression.


Subject(s)
Breast Neoplasms , Thrombophilia , Humans , Female , Animals , Mice , Antithrombins , Disease Models, Animal , Heterografts , Mice, SCID , Thrombophilia/genetics , Anticoagulants , Breast Neoplasms/complications , Breast Neoplasms/genetics , Antithrombin III/genetics , RNA, Small Interfering
4.
Thromb Res ; 201: 147-150, 2021 05.
Article in English | MEDLINE | ID: mdl-33798826
5.
J Thromb Haemost ; 17(1): 169-182, 2019 01.
Article in English | MEDLINE | ID: mdl-30472780

ABSTRACT

Essentials Tissue factor (TF) isoforms are expressed in pancreatic neuroendocrine tumors (pNET). TF knockdown inhibits proliferation of human pNET cells in vitro. mTOR kinase inhibitor sapanisertib/MLN0128 suppresses TF expression in human pNET cells. Sapanisertib suppresses TF expression and activity and reduces the growth of pNET tumors in vivo. SUMMARY: Background Full-length tissue factor (flTF) and alternatively spliced TF (asTF) contribute to growth and spread of pancreatic ductal adenocarcinoma. It is unknown, however, if flTF and/or asTF contribute to the pathobiology of pancreatic neuroendocrine tumors (pNETs). Objective To assess TF expression in pNETs and the effects of mTOR complex 1/2 (mTORC1/2) inhibition on pNET growth. Methods Human pNET specimens were immunostained for TF. Human pNET cell lines QGP1 and BON were evaluated for TF expression and responsiveness to mTOR inhibition. shRNA were used to knock down TF in BON. TF cofactor activity was assessed using a two-step FXa generation assay. TF promoter activity was assessed using transient transfection of human TF promoter-driven reporter constructs into cells. Mice bearing orthotopic BON tumors were treated with the mTORC1/2 ATP site competitive inhibitor sapanisertib/MLN0128 (3 mg kg-1 , oral gavage) for 34 days. Results Immunostaining of pNET tissue revealed flTF and asTF expression. BON and QGP1 expressed both TF isoforms, with BON exhibiting higher levels. shRNA directed against TF suppressed BON proliferation in vitro. Treatment of BON with sapanisertib inhibited mTOR signaling and suppressed TF levels. BON tumors grown in mice treated with sapanisertib had significantly less TF protein and cofactor activity, and were smaller compared with tumors grown in control mice. Conclusions TF isoforms are expressed in pNETs. Sapanisertib suppresses TF mRNA and protein expression as well as TF cofactor activity in vitro and in vivo. Thus, further studies are warranted to evaluate the clinical utility of TF-suppressing mTORC1/2 inhibitor sapanisertib in pNET management.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Neuroendocrine Tumors/drug therapy , Pancreatic Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Pyrazoles/pharmacology , Pyrimidines/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Thromboplastin/metabolism , Animals , Cell Line, Tumor , Humans , Mechanistic Target of Rapamycin Complex 1/metabolism , Mechanistic Target of Rapamycin Complex 2/metabolism , Mice, Nude , Neuroendocrine Tumors/enzymology , Neuroendocrine Tumors/genetics , Neuroendocrine Tumors/pathology , Pancreatic Neoplasms/enzymology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Promoter Regions, Genetic , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Thromboplastin/genetics , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
6.
J Thromb Haemost ; 16(2): 293-302, 2018 02.
Article in English | MEDLINE | ID: mdl-29247594

ABSTRACT

Essentials The underlying pathophysiological mechanisms behind cancer-associated thrombosis are unknown. We compared expression profiles in tumor cells from patients with and without thrombosis. Tumors from patients with thrombosis showed significant differential gene expression profiles. Patients with thrombosis had a proinflammatory status and increased fibrin levels in the tumor. SUMMARY: Background Venous thromboembolism (VTE) is a frequent complication in patients with cancer, and is associated with significant morbidity and mortality. However, the mechanisms behind cancer-associated thrombosis are still incompletely understood. Objectives To identify novel genes that are associated with VTE in patients with colorectal cancer (CRC). Methods Twelve CRC patients with VTE were age-matched and sex-matched to 12 CRC patients without VTE. Tumor cells were isolated from surgical samples with laser capture microdissection approaches, and mRNA profiles were measured with next-generation RNA sequencing. Results This approach led to the identification of new genes and pathways that might contribute to VTE in CRC patients. Application of ingenuity pathway analysis indicated significant links with inflammation, the methionine degradation pathway, and increased platelet function, which are all key processes in thrombus formation. Tumor samples of patients with VTE had a proinflammatory status and contained higher levels of fibrin and fibrin degradation products than samples of those without VTE. Conclusion This case-control study provides a proof-of-principle that tumor gene expression can discriminate between cancer patients with low and high risks of VTE. These findings may help to further unravel the pathogenesis of cancer-related VTE. The identified genes could potentially be used as candidate biomarkers to select high-risk CRC patients for thromboprophylaxis.


Subject(s)
Biomarkers, Tumor/genetics , Blood Coagulation/genetics , Colorectal Neoplasms/genetics , Venous Thromboembolism/genetics , Aged , Aged, 80 and over , Case-Control Studies , Colorectal Neoplasms/blood , Colorectal Neoplasms/complications , Colorectal Neoplasms/diagnosis , Female , Gene Expression Profiling , Gene Regulatory Networks , Genetic Predisposition to Disease , Humans , Male , Middle Aged , Phenotype , Proof of Concept Study , Risk Assessment , Risk Factors , Transcriptome , Venous Thromboembolism/blood , Venous Thromboembolism/diagnosis
7.
Thromb Res ; 156: 1-7, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28570958

ABSTRACT

BACKGROUND: Full length Tissue factor (flTF) is a key player in hemostasis and also likely contributes to venous thromboembolism (VTE), the third most common cardiovascular disease. flTF and its minimally coagulant isoform, alternatively spliced TF (asTF), have been detected in thrombi, suggesting participation of both isoforms in thrombogenesis, but data on participation of asTF in hemostasis is lacking. Therefore, we assessed the role of asTF in flTF cofactor activity modulation, using a co-expression system. OBJECTIVE: To investigate the interplay between flTF and asTF in hemostasis on endothelial cell surface. METHODS: Immortalized endothelial (ECRF) cells were adenovirally transduced to express asTF and flTF, after which flTF cofactor activity was measured on cells and microvesicles (MVs). To study co-localization of flTF/asTF proteins, confocal microscopy was performed. Finally, intracellular distribution of flTF was studied in the presence or absence of heightened asTF levels. RESULTS: Levels of flTF antigen and cofactor activity were not affected by asTF co-expression. asTF and flTF were found to localize in distinct subcellular compartments. Only upon heightened overexpression of asTF, lower flTF protein levels and cofactor activity were observed. Heightened asTF levels also induced a shift of flTF from non-raft to lipid raft plasma membrane fractions, and triggered the expression of ER stress marker BiP. Proteasome inhibition resulted in increased asTF - but not flTF - protein expression. CONCLUSION: At moderate levels, asTF appears to have negligible impact on flTF cofactor activity on endothelial cells and MVs; however, at supra-physiological levels, asTF is able to reduce the levels of flTF protein and cofactor activity.


Subject(s)
Alternative Splicing/physiology , Blood Coagulation Factors/metabolism , Endothelial Cells/metabolism , Thromboplastin/metabolism , Venous Thromboembolism/blood , Hemostasis , Humans
8.
J Thromb Haemost ; 13(9): 1683-93, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26179105

ABSTRACT

BACKGROUND: Procoagulant full-length tissue factor (flTF) and its minimally coagulant alternatively spliced isoform (asTF), promote breast cancer (BrCa) progression via different mechanisms. We previously showed that flTF and asTF are expressed by BrCa cells, resulting in autoregulation in a cancer milieu. BrCa cells often express hormone receptors such as the estrogen receptor (ER), leading to the formation of hormone-regulated cell populations. OBJECTIVE: To investigate whether TF isoform-specific and ER-dependent pathways interact in BrCa. METHODS: Tissue factor isoform-regulated gene sets were assessed using ingenuity pathway analysis. Tissues from a cohort of BrCa patients were divided into ER-positive and ER-negative groups. Associations between TF isoform levels and tumor characteristics were analyzed in these groups. BrCa cells expressing TF isoforms were assessed for proliferation, migration and in vivo growth in the presence or absence of estradiol. RESULTS: Ingenuity pathway analysis pointed to similarities between ER- and TF-induced gene expression profiles. In BrCa tissue specimens, asTF expression was associated with grade and stage in ER-positive but not in ER-negative tumors. flTF was only associated with grade in ER-positive tumors. In MCF-7 cells, asTF accelerated proliferation in the presence of estradiol in a ß1 integrin-dependent manner. No synergy between asTF and the ER pathway was observed in a migration assay. Estradiol accelerated the growth of asTF-expressing tumors but not control tumors in vivo in an orthotopic setting. CONCLUSION: Tissue factor isoform and estrogen signaling share downstream targets in BrCa; the concomitant presence of asTF and estrogen signaling is required to promote BrCa cell proliferation.


Subject(s)
Alternative Splicing , Breast Neoplasms/pathology , Carcinoma/pathology , Estrogens , Neoplasm Proteins/physiology , Neoplasms, Hormone-Dependent/pathology , Receptors, Estrogen/physiology , Signal Transduction/physiology , Thromboplastin/physiology , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Carcinoma/genetics , Carcinoma/metabolism , Cell Division/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Disease Progression , Estradiol/pharmacology , Female , Gene Expression Profiling , Humans , Integrin beta1/physiology , Neoplasm Grading , Neoplasm Proteins/genetics , Neoplasm Staging , Neoplasms, Hormone-Dependent/genetics , Neoplasms, Hormone-Dependent/metabolism , Protein Isoforms/genetics , Protein Isoforms/physiology , Software , Thromboplastin/genetics , Tissue Array Analysis
10.
J Thromb Haemost ; 11 Suppl 1: 285-93, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23809132

ABSTRACT

Tissue factor (TF) is a 47 kDa membrane protein that initiates coagulation by binding to FVII(a) and FX(a) and is a risk factor for thrombosis in many disease states. In addition to its coagulant activity, TF also influences cancer progression by triggering signaling effects via a group of G-protein coupled receptors named protease-activated receptors (PARs). TF localizes to cytoskeletal structures in migrating cells, influences cytoskeleton reorganization and promotes migration. Recently, integrins, important mediators of cell motility, have emerged as important binding partners for TF and influence both TF coagulant and PAR-2-dependent signaling functions. Direct binding of TF to integrins also impacts processes such as cell migration and signaling independent of PAR-2. A recently discovered alternatively spliced, soluble TF isoform also ligates integrins to augment angiogenesis, thus fuelling cancer progression. To date, the literature describes a complex interplay between different integrin subunits and distinct TF isoforms, but our understanding of TF-integrin bidirectional regulation remains clouded. In this review, we aim to summarize the existing knowledge on integrin-TF interaction and speculate on its relevance to physiology and pathology.


Subject(s)
Integrins/metabolism , Neoplasms/metabolism , Thromboplastin/metabolism , Thrombosis/metabolism , Humans , Neoplasms/pathology , Thrombosis/pathology
11.
J Thromb Haemost ; 9(10): 2087-96, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21812913

ABSTRACT

BACKGROUND: TF is highly expressed in cancerous and atherosclerotic lesions. Monocyte recruitment is a hallmark of disease progression in these pathological states. OBJECTIVE: To examine the role of integrin signaling in TF-dependent recruitment of monocytes by endothelial cells. METHODS: The expression of flTF and asTF in cervical cancer and atherosclerotic lesions was examined. Biologic effects of the exposure of primary microvascular endothelial cells (MVEC) to truncated flTF ectodomain (LZ-TF) and recombinant asTF were assessed. RESULTS: flTF and asTF exhibited nearly identical expression patterns in cancer lesions and lipid-rich plaques. Tumor lesions, as well as stromal CD68(+) monocytes/macrophages, expressed both TF forms. Primary MVEC rapidly adhered to asTF and LZ-TF, and this was completely blocked by anti-ß1 integrin antibody. asTF- and LZ-TF-treatment of MVEC promoted adhesion of peripheral blood mononuclear cells (PBMCs) under orbital shear conditions and under laminar flow; asTF-elicited adhesion was more pronounced than that elicited by LZ-TF. Expression profiling and western blotting revealed a broad activation of cell adhesion molecules (CAMs) in MVEC following asTF treatment including E-selectin, ICAM-1 and VCAM-1. In transwell assays, asTF potentiated PMBC migration through MVEC monolayers by ∼3-fold under MCP-1 gradient. CONCLUSIONS: TF splice variants ligate ß1 integrins on MVEC, which induces the expression of CAMs in MVEC and leads to monocyte adhesion and transendothelial migration. asTF appears more potent than flTF in eliciting these effects. Our findings underscore the pathophysiologic significance of non-proteolytic, integrin-mediated signaling by the two naturally occurring TF variants in cancer and atherosclerosis.


Subject(s)
Alternative Splicing , Cell Adhesion Molecules/metabolism , Endothelium, Vascular/cytology , Integrins/metabolism , Monocytes/cytology , Signal Transduction , Thromboplastin/genetics , Blotting, Western , Cells, Cultured , Female , Humans , Reverse Transcriptase Polymerase Chain Reaction , Uterine Cervical Neoplasms/metabolism , Uterine Cervical Neoplasms/pathology
12.
Hamostaseologie ; 30(3): 144-9, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20680231

ABSTRACT

Full-length tissue factor (flTF) initiates coagulation, but also exerts non-hemostatic functions such as inflammation and angiogenesis through protease activated receptors (PARs). In 2003 a soluble variant of flTF was described which results from alternative splicing. Since its discovery the role of alternatively spliced tissue factor (asTF) in coagulation has been debated. asTF may have pro-coagulant properties but due to structural differences when compared to flTF, asTF coagulant function may be relatively low. Nevertheless, similar to flTF, asTF appears to have non-hemostatic properties; asTF expression in tumors correlates with increased tumor size, vessel number and poor survival in some cancer types, and drives tumor growth in animal models. Interestingly, unlike flTF, asTF does not promote angiogenesis through activating PARs but rather via integrin ligation. flTF is a critical determinant in cardiovascular disease but little is known about asTF in cardiovascular disease. asTF is produced by monocytes and macrophages, thus macrophage-derived asTF may contribute to atherosclerotic disease. In conclusion, unraveling asTF's non-hemostatic properties may generate new insights in the pathophysiology and diagnostics of cancer and cardiovascular disease.


Subject(s)
Alternative Splicing , Blood Coagulation/genetics , Hemostasis/physiology , Thromboplastin/genetics , Cardiovascular Diseases/blood , Cardiovascular Diseases/genetics , Humans , Neoplasms/blood , Neoplasms/genetics , RNA Precursors/genetics , Thromboplastin/physiology
13.
Proc Natl Acad Sci U S A ; 106(46): 19497-502, 2009 Nov 17.
Article in English | MEDLINE | ID: mdl-19875693

ABSTRACT

The initiator of coagulation, full-length tissue factor (flTF), in complex with factor VIIa, influences angiogenesis through PAR-2. Recently, an alternatively spliced variant of TF (asTF) was discovered, in which part of the TF extracellular domain, the transmembrane, and cytoplasmic domains are replaced by a unique C terminus. Subcutaneous tumors produced by asTF-secreting cells revealed increased angiogenesis, but it remained unclear if and how angiogenesis is regulated by asTF. Here, we show that asTF enhances angiogenesis in matrigel plugs in mice, whereas a soluble form of flTF only modestly enhances angiogenesis. asTF dose-dependently upregulates angiogenesis ex vivo independent of either PAR-2 or VIIa. Rather, asTF was found to ligate integrins, resulting in downstream signaling. asTF-alphaVbeta3 integrin interaction induces endothelial cell migration, whereas asTF-dependent formation of capillaries in vitro is dependent on alpha6beta1 integrin. Finally, asTF-dependent aortic sprouting is sensitive to beta1 and beta3 integrin blockade and a TF-antibody that disrupts asTF-integrin interaction. We conclude that asTF, unlike flTF, does not affect angiogenesis via PAR-dependent pathways but relies on integrin ligation. These findings indicate that asTF may serve as a target to prevent pathological angiogenesis.


Subject(s)
Alternative Splicing , Integrin alpha6beta1/metabolism , Integrin alphaVbeta3/metabolism , Neovascularization, Pathologic/genetics , Thromboplastin/genetics , Animals , Aorta/growth & development , Aorta/metabolism , Capillaries/growth & development , Capillaries/metabolism , Cell Movement , Endothelium, Vascular/metabolism , Factor V/metabolism , Mice , Mice, Inbred C57BL , Receptor, PAR-2/metabolism
15.
J Thromb Haemost ; 6(9): 1550-7, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18647225

ABSTRACT

BACKGROUND: Tissue factor (TF) and factor (F) VIIa are the primary initiators of the coagulation cascade, but also promote non-hemostatic events, such as angiogenesis and tumor growth, via activation of protease activated receptor-2 (PAR2). Our previous findings indicated that the TF:FVIIa complex activates signal transducer and activator of transcription (STAT) signaling, leading to cell survival in TF-transfected baby hamster kidney (BHK) cells. METHODS: Using BHK TF, keratinocytes (HaCaT) and human umbilical vein endothelial cells (HUVEC), FVIIa-induced phosphorylation and activation of the transcription factor cyclic AMP-responsive binding protein (CREB) were tested and compared to that elicited by thrombin and FXa. In addition, the effect of these factors on cell survival and expression of apoptosis-associated proteins was monitored. RESULTS: Factor VIIa led to a TF-dependent, but TF cytoplasmic domain-independent phosphorylation and activation of CREB in BHK TF, HaCaT and HUVEC. CREB activation was sensitive to blockade of the extracellular-signal regulated kinase 1/2 pathway and PAR2. Surprisingly, FVIIa decreased cell survival in HaCaT cells but not other cell types and upregulated the pro-apoptotic proteins Bak and Puma in a CREB-dependent manner. Factor Xa, but not FIIa, induced phosphorylation of CREB, but did not have an effect on apoptosis. CONCLUSION: TF:FVIIa induces CREB phosphorylation and activation in several cell types, but TF:FVIIa induces pro-apoptotic proteins and apoptosis only in selected cell types.


Subject(s)
Apoptosis , Cyclic AMP Response Element-Binding Protein/metabolism , Factor VIIa/metabolism , Receptor, PAR-2/metabolism , Thromboplastin/metabolism , Up-Regulation , Animals , Blotting, Western , Cells, Cultured , Cricetinae , Factor Xa/metabolism , GTP-Binding Proteins/metabolism , Humans , Phosphorylation , Signal Transduction , Thrombin/metabolism
16.
J Thromb Haemost ; 2(12): 2185-93, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15613025

ABSTRACT

Intervention studies blocking tissue factor (TF) driven coagulation show beneficial effects on survival in endotoxemia models by reducing cytokine production. It is unknown, however, if moderately reduced TF levels influence endotoxemia. The objective was to investigate whether TF haploinsufficiency reduces endotoxin-induced cytokine production in murine cells or in mice. We analyzed the intrinsic capacity of heterozygous TF deficient (TF+/-) leukocytes to produce cytokines. In addition, we determined the consequences of TF haploinsufficiency on endotoxin-induced inflammation during murine endotoxemia. Endotoxin induced the production of tumor necrosis factor-alpha (TNF-alpha), interleukin (IL)-6 and keratinocyte-derived chemokine (KC) in both whole blood and macrophages. Heterozygous TF deficiency reduced endotoxin induced IL-6 and KC levels about two-fold, while TNF-alpha levels were indistinguishable between TF+/- and wild-type cells. In vivo, endotoxin induced a biphasic coagulant response and significant increases in cytokine levels. Surprisingly, both the inflammatory and the coagulant responses were indistinguishable between wild-type and TF+/- mice. At baseline tissues of TF+/- mice showed a 50% reduction in TF activity compared to wild type. Upon endotoxin administration, TF activity increased and the difference between TF+/- and wild-type mice disappeared after 4 h. After 12 h the baseline difference in TF activity was re-established. TF deficiency reduces cytokine production in vitro, but an enhanced induction of TF during murine endotoxemia eliminates this effect in vivo.


Subject(s)
Endotoxemia/blood , Endotoxins/metabolism , Inflammation/chemically induced , Thromboplastin/genetics , Thromboplastin/metabolism , Animals , Antithrombins/metabolism , Blood Coagulation , Cell Line , Chemokines/metabolism , Cytokines/metabolism , Endotoxins/pharmacology , Heterozygote , Immunohistochemistry , Interleukin-6/blood , Interleukin-6/metabolism , Keratinocytes/metabolism , Leukocytes/metabolism , Lipopolysaccharides/metabolism , Macrophages/metabolism , Mice , Mice, Transgenic , Thrombin/metabolism , Time Factors , Tumor Necrosis Factor-alpha/biosynthesis
18.
J Thromb Haemost ; 1(5): 1012-8, 2003 May.
Article in English | MEDLINE | ID: mdl-12871370

ABSTRACT

BACKGROUND: In recent years it has become clear that factor (F)VIIa is not a passive mediator involved in the linear transduction of the coagulation cascade, but actively engages target cells to induce signal transduction and that this signal transduction fulfills critical functions in angiogenesis, arteriosclerosis and inflammatory processes. OBJECTIVES: The details of coagulation factor-dependent signal transduction are among the least understood in biology and thus we set out to establish the molecular events responsible for MAP kinase activation induced by the interaction of FVIIa with its cellular binding partner tissue factor (TF). METHODS: Two different TF-expressing cell types, BHKTF and HaCaT cells, were assayed for p21Ras activation using a pull-down assay that is specific for activated Ras. This activation was visualized by means of Western blotting. In addition, the upstream pathways leading to FVIIa-induced Ras activation were characterized using phosphospecific antibodies and specific inhibitors. RESULTS: We observed that in both BHKTF and HaCaT cells FVIIa-induced MAP kinase activation correlates with p21Ras activation, and that this p21Ras activation is essential for FVIIa-induced MAP kinase activation. In BHKTF cells, early p21Ras activation was mediated by the activation of protein kinase C (PKC), whereas late p21Ras activation employed alternative mechanisms. In HaCaT cells, stimulation of the Src kinase family mediated FVIIa-dependent p21Ras activation. Finally, in both cell types, Raf activity was mandatory for MAP kinase activation. CONCLUSIONS: p21Ras activation is instrumental in FVIIa signal transduction and the FVIIa-dependent activation of p21Ras involves either PKC or Src-dependent mechanisms, depending on the cell type investigated.


Subject(s)
Factor VIIa/pharmacology , MAP Kinase Signaling System/drug effects , Oncogene Protein p21(ras)/metabolism , Animals , Blotting, Western , Cell Line , Factor VIIa/physiology , Humans , Mitogen-Activated Protein Kinases/drug effects , Mitogen-Activated Protein Kinases/metabolism , Oncogene Protein p21(ras)/drug effects , Phosphorylation/drug effects , Protein Kinase C/metabolism , src-Family Kinases/metabolism
19.
Article in English | MEDLINE | ID: mdl-12062209

ABSTRACT

The immunomodulatory effect of hyperbaric oxygen, involving altered cytokine release by macrophages, is well described. Importantly, however, it is not known what the relative contribution is of the hyperbaric environment of the cells vs. increased oxygen tension on these hyperbaric oxygen-dependent effects. We compared, therefore, cytokine release by murine macrophages under hyperbaric oxygen, hyperpressure of normal air and normobaric conditions. We observed that hyperbaric oxygen enhanced cytokine release of both unstimulated as well as lipopolysaccharide (LPS)-challenged macrophages. Hyperpressure of normal air, however, enhanced LPS-induced cytokine production but did not elicit cytokine release in unstimulated macrophages. To further investigate the molecular details underlying the effects of hyperbaric oxygen, we investigated the effect of the p42/p44 mitogen-activated protein (MAP) kinase inhibitor PD98059 and the p38 MAP kinase inhibitor SB203580. Neither inhibitor, however, had a significant effect on the modulatory effects of hyperbaric oxygen on cytokine release. We concluded that the immunomodulatory effect of hyperbaric oxygen contains a component for which hyperpressure is sufficient and a component that apart from hyperpressure also requires hyperoxygenation.


Subject(s)
Hyperbaric Oxygenation , Macrophages/drug effects , Macrophages/immunology , Oxygen/pharmacology , Animals , Cells, Cultured , Dimethyl Sulfoxide/pharmacology , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Imidazoles/pharmacology , Lipopolysaccharides/pharmacology , Macrophages/enzymology , Macrophages/metabolism , Mice , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , Pyridines/pharmacology , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism , p38 Mitogen-Activated Protein Kinases
20.
Trends Cardiovasc Med ; 11(8): 335-9, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11728883

ABSTRACT

Tissue factor (TF) is a 47-kDa transmembrane protein, involved in the onset of coagulation. However, it also influences pathophysiological processes such as inflammation and tumor angiogenesis. Although the molecular mechanisms responsible for these phenomena remain unclear, it is thought that they are brought about by the action of intracellular signaling, resulting in gene transcription and subsequent protein synthesis. In this review we focus on FVIIa/TF-induced intracellular signaling and its possible role in physiology.


Subject(s)
Angiogenesis Inducing Agents/physiology , Inflammation/physiopathology , Signal Transduction/physiology , Thromboplastin/physiology , Blood Coagulation/physiology , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...