thromboplastin and Glioblastoma

thromboplastin has been researched along with Glioblastoma* in 32 studies

Reviews

3 review(s) available for thromboplastin and Glioblastoma

ArticleYear
Biological basis of personalized anticoagulation in cancer: oncogene and oncomir networks as putative regulators of coagulopathy.
    Thrombosis research, 2016, Volume: 140 Suppl 1

    Activation of stromal response pathways in cancer is increasingly viewed as both a local and systemic extension of molecular alterations driving malignant transformation. Rather than reflecting passive and unspecific responses to anatomical abnormalities, the coagulation system is a target of oncogenic deregulation, impacting the role of clotting and fibrinolytic proteins, and integrating hemostasis, inflammation, angiogenesis and cellular growth effects in cancer. These processes signify, but do not depend on, the clinically manifest coagulopathy and thrombosis. In this regard, the role of driver mutations affecting oncoprotein coding genes such as RAS, EGFR or MET and tumour suppressors (PTEN, TP53) are well described as regulators of tissue factor (TF), protease activated receptors (PAR-1/2) and ectopic coagulation factors (FVII). Indeed, in both adult and pediatric brain tumours the expression patterns of coagulation and angiogenesis regulators (coagulome and angiome, respectively) reflect the molecular subtypes of the underlying diseases (glioblastoma or medulloblastoma) as defined by their oncogenic classifiers and clinical course. This emerging understanding is still poorly established in relation to the transforming effects of non-coding genes, including those responsible for the expression of microRNA (miR). Indeed, several miRs have been recently found to regulate TF and other effectors. We recently documented that in the context of the aggressive embryonal tumour with multilayered rosettes (ETMR) the oncogenic driver miR (miR-520g) suppresses the expression of TF and correlates with hypocoagulant tumour characteristics. Unlike in adult cancers, the growth of pediatric embryonal brain tumour cells as spheres (to maintain stem cell properties) results in upregulation of miR-520g and downregulation of TF expression and activity. We postulate that oncogenic protein and miR coding genes form alternative pathways of coagulation system regulation in different tumour settings, a property necessitating more personalised and biologically-based approaches to anticoagulation.

    Topics: Animals; Anticoagulants; Blood Coagulation; Blood Coagulation Disorders; Brain; Brain Neoplasms; Glioblastoma; Humans; Medulloblastoma; MicroRNAs; Oncogenes; Precision Medicine; Thromboplastin

2016
Brain neoplasms and coagulation.
    Seminars in thrombosis and hemostasis, 2013, Volume: 39, Issue:8

    Brain vasculature is uniquely programmed to protect central nervous system tissues and respond to their metabolic demands. These functions are subverted during the development of primary and metastatic brain tumors, resulting in vascular perturbations that are thought to contribute to progression and comorbidities of the underlying disease, including thrombosis and hemorrhage. Chronic activation of the coagulation system is particularly obvious in glioblastoma multiforme (GBM), where intratumoral vasoocclusive thrombosis may contribute to hypoxia, pseudopalisading necrosis, and angiogenesis. GBM is also associated with spontaneous or iatrogenic bleeding, and the emission of circulating procoagulants implicated in the unusually high risk of peripheral venous thromboembolism. Tissue factor (TF) expression is elevated in several types of brain tumors, including adult and pediatric GBM, as is the production of TF-containing microparticles (TF-MPs). Both TF expression and its vesicular emission are regulated by tumor microenvironment (e.g., hypoxia), in concert with activated oncogenic and growth factor pathways (RAS, EGFR, MET), as well as the loss of tumor suppressor gene activity (PTEN). Discovery of distinct oncogenic networks led to recognition of unique molecular subtypes within brain tumors, of which GBM (proneural, neural, classical, and mesenchymal), and medulloblastoma (SHH, WNT, group 3, and group 4) exhibit subtype-specific composition of the tumor coagulome. It remains to be established whether mechanisms of thrombosis and biological effects of coagulation in brain tumors are also subtype specific. In this regard, TF pathway represents a paradigm, and its impact on tumor dormancy, inflammation, angiogenesis, formation of cancer stem cell niches, and dissemination is a subject of considerable interest. However, establishing the extent to which TF and TF-MPs contribute to pathogenesis and thromboembolic disease in the context of primary and secondary brain tumors may require molecular stratification of patient populations. We suggest that a better understanding of these molecular linkages may pave the way to a more effective (targeted) therapy, prophylaxis, adjunctive use of anticoagulants, and other agents able to modulate interactions between brain tumors and the coagulation system.

    Topics: Adult; Blood Coagulation; Brain Neoplasms; Child; Glioblastoma; Humans; Models, Biological; Neovascularization, Pathologic; Signal Transduction; Thromboplastin

2013
Vaso-occlusive and prothrombotic mechanisms associated with tumor hypoxia, necrosis, and accelerated growth in glioblastoma.
    Laboratory investigation; a journal of technical methods and pathology, 2004, Volume: 84, Issue:4

    Glioblastoma (GBM) has explosive biologic properties with rapid clinical progression leading to death. Its distinguishing pathologic features, necrosis with surrounding pseudopalisades and microvascular hyperplasia, are believed to be instrumental to its accelerated growth. Microvascular hyperplasia arises in response to the secretion of proangiogenic factors by hypoxic pseudopalisades and allows for peripheral neoplastic expansion. Mechanisms underlying necrosis and hypoxia remain obscure, but vaso-occlusive and prothrombotic contributions could be substantial. Recent investigations on the origin of pseudopalisades suggest that this morphologic phenomenon is created by a tumor cell population actively migrating away from a central hypoxic region and that, in at least a significant subset, hypoxia-induced migration appears due to vaso-occlusion caused by intravascular thrombosis. Both vascular endothelial growth factor induced vascular permeability to plasma coagulation factors and the increased neoplastic expression of tissue factor likely contribute to a prothrombotic state favoring intravascular thrombosis. In addition to prothrombotic mechanisms, vaso-occlusion could also result from angiopoietin-2-mediated endothelial cell apoptosis and vascular regression, which follows neoplastic co-option of native vessels in animal models of gliomas. Vaso-occlusive and prothrombotic mechanisms in GBM could readily explain the presence of pseudopalisades and coagulative necrosis in tissue sections, the emergence of central contrast enhancement and its rapid peripheral expansion on neuroimaging, and the dramatic shift to an accelerated rate of clinical progression. Since the hypoxic induction of angiogenesis appears to support further neoplastic growth, therapeutic targeting of the underlying vascular pathology and thrombosis could provide a new means to prolong time to progression.

    Topics: Blood Vessels; Brain Neoplasms; Cell Division; Cell Hypoxia; Glioblastoma; Humans; Necrosis; Neovascularization, Pathologic; Thromboplastin; Thrombosis

2004

Other Studies

29 other study(ies) available for thromboplastin and Glioblastoma

ArticleYear
Systematic study of tissue factor expression in solid tumors.
    Cancer reports (Hoboken, N.J.), 2023, Volume: 6, Issue:2

    Elevated tissue factor (TF) expression, although restricted in normal tissue, has been reported in multiple solid cancers, and expression has been associated with poor prognosis. This manuscript compares TF expression across various solid tumor types via immunohistochemistry in a single study, which has not been performed previously.. To increase insight in the prevalence and cellular localization of TF expression across solid cancer types, we performed a detailed and systematic analysis of TF expression in tumor tissue obtained from patients with ovarian, esophageal, bladder, cervical, endometrial, pancreatic, prostate, colon, breast, non-small cell lung cancer (NSCLC), head and neck squamous cell carcinoma (HNSCC), and glioblastoma. The spatial and temporal variation of TF expression was analyzed over time and upon disease progression in patient-matched biopsies taken at different timepoints. In addition, TF expression in patient-matched primary tumor and metastatic lesions was also analyzed.. TF expression was detected via immunohistochemistry (IHC) using a validated TF-specific antibody. TF was expressed in all cancer types tested, with highest prevalence in pancreatic cancer, cervical cancer, colon cancer, glioblastoma, HNSCC, and NSCLC, and lowest in breast cancer. Staining was predominantly membranous in pancreatic, cervical, and HNSCC, and cytoplasmic in glioblastoma and bladder cancer. In general, expression was consistent between biopsies obtained from the same patient over time, although variability was observed for individual patients. NSCLC biopsies of primary tumor and matched lymph node metastases showed no clear difference in TF expression overall, although individual patient changes were observed.. This study shows that TF is expressed across a broad range of solid cancer types, and expression is present upon tumor dissemination and over the course of treatment.

    Topics: Carcinoma, Non-Small-Cell Lung; Female; Glioblastoma; Head and Neck Neoplasms; Humans; Lung Neoplasms; Male; Squamous Cell Carcinoma of Head and Neck; Thromboplastin

2023
Tissue factor is a critical regulator of radiation therapy-induced glioblastoma remodeling.
    Cancer cell, 2023, 08-14, Volume: 41, Issue:8

    Radiation therapy (RT) provides therapeutic benefits for patients with glioblastoma (GBM), but inevitably induces poorly understood global changes in GBM and its microenvironment (TME) that promote radio-resistance and recurrence. Through a cell surface marker screen, we identified that CD142 (tissue factor or F3) is robustly induced in the senescence-associated β-galactosidase (SA-βGal)-positive GBM cells after irradiation. F3 promotes clonal expansion of irradiated SA-βGal

    Topics: Brain Neoplasms; Cell Line, Tumor; Glioblastoma; Humans; Neoplasm Recurrence, Local; Signal Transduction; Thromboplastin; Tumor Microenvironment

2023
Glioblastoma cell populations with distinct oncogenic programs release podoplanin as procoagulant extracellular vesicles.
    Blood advances, 2021, 03-23, Volume: 5, Issue:6

    Vascular anomalies, including local and peripheral thrombosis, are a hallmark of glioblastoma (GBM) and an aftermath of deregulation of the cancer cell genome and epigenome. Although the molecular effectors of these changes are poorly understood, the upregulation of podoplanin (PDPN) by cancer cells has recently been linked to an increased risk for venous thromboembolism (VTE) in GBM patients. Therefore, regulation of this platelet-activating protein by transforming events in cancer cells is of considerable interest. We used single-cell and bulk transcriptome data mining, as well as cellular and xenograft models in mice, to analyze the nature of cells expressing PDPN, as well as their impact on the activation of the coagulation system and platelets. We report that PDPN is expressed by distinct (mesenchymal) GBM cell subpopulations and downregulated by oncogenic mutations of EGFR and IDH1 genes, along with changes in chromatin modifications (enhancer of zeste homolog 2) and DNA methylation. Glioma cells exteriorize their PDPN and/or tissue factor (TF) as cargo of exosome-like extracellular vesicles (EVs) shed from cells in vitro and in vivo. Injection of glioma-derived podoplanin carrying extracelluar vesicles (PDPN-EVs) activates platelets, whereas tissue factor carrying extracellular vesicles (TF-EVs) activate the clotting cascade. Similarly, an increase in platelet activation (platelet factor 4) or coagulation (D-dimer) markers occurs in mice harboring the corresponding glioma xenografts expressing PDPN or TF, respectively. Coexpression of PDPN and TF by GBM cells cooperatively affects tumor microthrombosis. Thus, in GBM, distinct cellular subsets drive multiple facets of cancer-associated thrombosis and may represent targets for phenotype- and cell type-based diagnosis and antithrombotic intervention.

    Topics: Animals; Extracellular Vesicles; Glioblastoma; Glioma; Humans; Mice; Thromboplastin; Thrombosis

2021
Dynamic MR imaging for functional vascularization depends on tissue factor signaling in glioblastoma.
    Cancer biology & therapy, 2018, 05-04, Volume: 19, Issue:5

    Glomeruloid vascular proliferation (GVP) is a diagnostic hallmark and links to aggressive behavior, therapy resistance and poor prognosis in glioblastoma (GBM). It lacks clinical approaches to predict and monitor its formation and dynamic change. Yet the mechanism of GVPs also remains largely unknown. Using an in situ GBM xenograft mouse model, combined clinical MRI images of pre-surgery tumor and pathological investigation, we demonstrated that the inhibition of tissue factor (TF) decreased GVPs in Mouse GBM xenograft model. TF shRNA reduced microvascular area and diameter, other than bevacizumab. TF dominantly functions via PAR2/HB-EGF-dependent activation under hypoxia in endothelial cells (ECs), resulting in a reduction of GVPs and cancer cells invasion. TF expression strongly correlated to GVPs and microvascular area (MVA) in GBM specimens from 56 patients, which could be quantitatively evaluated in an advanced MRI images system in 33 GBM patients. This study presented an approach to assess GVPs that could be served as a MRI imaging biomarker in GBM and uncovered a molecular mechanism of GVPs.

    Topics: Adult; Aged; Animals; Biomarkers, Tumor; Brain Neoplasms; Cell Line, Tumor; Glioblastoma; Heterografts; Humans; Magnetic Resonance Imaging; Male; Mice; Middle Aged; Neovascularization, Pathologic; Signal Transduction; Thromboplastin; Young Adult

2018
(99m)Tc-ixolaris targets glioblastoma-associated tissue factor: in vitro and pre-clinical applications.
    Thrombosis research, 2015, Volume: 136, Issue:2

    The clotting initiator protein tissue factor (TF) has recently been described as a potential target that can be exploited to image aggressive tumors. Ixolaris is a specific TF inhibitor that blocks tumor cell procoagulant activity and tumor growth.. Herein we evaluated the ability of (99m)Tc-ixolaris to target tumor-derived TF using an orthotopic glioblastoma (GBM) model in mice.. The right forebrains of Swiss mice were stereotactically inoculated with U87-MG human GBM cells. Histological and immunohistochemical analyses were performed on the resulting tumors after 35-45 days. The biodistribution of (99m)Tc-ixolaris was evaluated by semi-quantitative whole-body scintigraphy and a quantitative analysis of radioactivity in isolated organs.. No (99m)Tc-ixolaris uptake was observed in brain of tumor-free mice, independently of the integrity of brain-blood barrier. In contrast, the presence of TF-expressing brain tumor masses determined a significant (99m)Tc-ixolaris uptake.. (99m)Tc-ixolaris recognized TF-expressing GBM cells in vivo. Given the proposed role of TF in tumor progression, (99m)Tc-ixolaris is a promising radiopharmaceutical agent for quantifying cancer-associated TF in aggressive tumors, including GBM.

    Topics: Animals; Biomarkers, Tumor; Brain Neoplasms; Cell Line, Tumor; Glioblastoma; Humans; Male; Metabolic Clearance Rate; Mice; Molecular Imaging; Organ Specificity; Positron-Emission Tomography; Radiopharmaceuticals; Reproducibility of Results; Salivary Proteins and Peptides; Sensitivity and Specificity; Technetium; Thromboplastin; Tissue Distribution

2015
Expression of tissue factor signaling pathway elements correlates with the production of vascular endothelial growth factor and interleukin-8 in human astrocytoma patients.
    Oncology reports, 2014, Volume: 31, Issue:2

    The expression levels of tissue factor (TF), the clotting initiator protein, have been correlated with angiogenesis and the histological grade of malignancy in glioma patients. The pro-tumor function of TF is linked to a family of G protein-coupled receptors known as protease-activated receptors (PARs), which may be activated by blood coagulation proteases. Activation of PARs elicits a number of responses, including the expression of vascular endothelial growth factor (VEGF) and interleukin-8 (IL-8). In the present study, we analyzed the expression of TF signaling pathway elements (TF, PAR1 and PAR2) and evaluated their correlation with the expression of downstream products (VEGF and IL-8) in human astrocytoma patients. Quantitative PCR (qPCR) showed a significant increase in TF expression in grade IV (glioblastoma) tumors, which was inversely correlated with the expression of the tumor-suppressor PTEN. Immunohistochemistry and qPCR analyses demonstrated a highly significant elevation in the expression of PAR1, but not PAR2, in tumor samples from high-grade astrocytoma patients. The elevated VEGF expression levels detected in the high-grade astrocytoma samples were positively correlated with TF, PAR1 and PAR2 expression. In addition, IL-8 was significantly increased in glioblastoma patients and positively correlated with TF and PAR2 expression. Further in vitro assays employing the human glioma cell lines U87-MG and HOG demonstrated that a synthetic peptide PAR2 agonist stimulated VEGF and IL-8 production. Our findings suggest a role for TF signaling pathway elements in astrocytoma progression, particularly in glioblastoma. Therefore, TF/PAR signaling elements may be suitable targets for the development of new therapies for the treatment of aggressive glioma.

    Topics: Brain Neoplasms; Glioblastoma; Humans; Interleukin-8; Neovascularization, Pathologic; PTEN Phosphohydrolase; Receptor, PAR-1; Receptor, PAR-2; Signal Transduction; Thromboplastin; Vascular Endothelial Growth Factors

2014
P14ARF suppresses tumor-induced thrombosis by regulating the tissue factor pathway.
    Cancer research, 2014, Mar-01, Volume: 74, Issue:5

    How necrotic areas develop in tumors is incompletely understood but can impact progression. Recent findings suggest that the formation of vascular microthrombi contributes to tumor necrosis, prompting investigation of coagulation cascades. Here, we report that loss of tumor suppressor P14ARF can contribute to activating the clotting cascade in glioblastoma. P14ARF transcriptionally upregulated TFPI2, a Kunitz-type serine protease in the tissue factor pathway that inhibits the initiation of thrombosis reactions. P14ARF activation in tumor cells delayed their ability to activate plasma clotting. Mechanistically, P14ARF activated the TFPI2 promoter in a p53-independent manner that relied upon c-JUN, SP1, and JNK activity. Taken together, our results identify the critical signaling pathways activated by P14ARF to prevent vascular microthrombosis triggered by glioma cells. Stimulation of this pathway might be used as a therapeutic strategy to reduce aggressive phenotypes associated with necrotic tumors, including glioblastoma.

    Topics: Genes, Tumor Suppressor; Glioblastoma; Humans; Promoter Regions, Genetic; Signal Transduction; Thromboplastin; Thrombosis; Transcriptional Activation; Tumor Cells, Cultured; Tumor Suppressor Protein p14ARF; Up-Regulation

2014
The contribution of tumor and host tissue factor expression to oncogene-driven gliomagenesis.
    Biochemical and biophysical research communications, 2014, Nov-14, Volume: 454, Issue:2

    Glioblastoma multiforme (GBM) is an aggressive form of glial brain tumors, associated with angiogenesis, thrombosis, and upregulation of tissue factor (TF), the key cellular trigger of coagulation and signaling. Since TF is upregulated by oncogenic mutations occurring in different subsets of human brain tumors we investigated whether TF contributes to tumourigenesis driven by oncogenic activation of EGFR (EGFRvIII) and RAS pathways in the brain. Here we show that TF expression correlates with poor prognosis in glioma, but not in GBM. In situ, the TF protein expression is heterogeneously expressed in adult and pediatric gliomas. GBM cells harboring EGFRvIII (U373vIII) grow aggressively as xenografts in SCID mice and their progression is delayed by administration of monoclonal antibodies blocking coagulant (CNTO 859) and signaling (10H10) effects of TF in vivo. Mice in which TF gene is disrupted in the neuroectodermal lineage exhibit delayed progression of spontaneous brain tumors driven by oncogenic N-ras and SV40 large T antigen (SV40LT) expressed under the control of sleeping beauty transposase. Reduced host TF levels in low-TF/SCID hypomorphic mice mitigated growth of glioma subcutaneously but not in the brain. Thus, we suggest that tumor-associated TF may serve as therapeutic target in the context of oncogene-driven disease progression in a subset of glioma.

    Topics: Adolescent; Adult; Animals; Brain; Brain Neoplasms; Cell Line, Tumor; ErbB Receptors; Gene Deletion; Gene Expression Regulation, Neoplastic; Glioblastoma; Glioma; Humans; Mice; Mice, SCID; Oncogenes; Prognosis; Thromboplastin

2014
Circulating microparticles of glial origin and tissue factor bearing in high-grade glioma: a potential prothrombotic role.
    Thrombosis and haemostasis, 2013, Volume: 110, Issue:2

    Venous thromboembolism (VTE) may complicate the clinical course of glioblastoma multiforme (GBM). Circulating microparticles (MPs) have been associated with cancer-related VTE. Sixty-one consecutive patients with GBM undergoing gross-total (41) or subtotal (20) surgical resection followed by radio-chemotherapy were prospectively evaluated. MPs numbers according to cellular origin and the procoagulant activity of annexin V positive (AV+) MPs (MP-activity) were measured before surgery and then 1 week and 1, 4, and 7 months after surgery. Glial (GFAP+) and endothelial (CD62E+) derived MPs, AV+ and tissue factor-bearing (TF+) MPs were measured using flow cytometry. Baseline levels of GFAP+/TF-, TF+/GFAP-, and GFAP+/TF+ MPs were significantly higher in GBM patients than in healthy controls, and significantly increased at each time point after surgery; at 7 months, a further significant increase over the level found a week after surgery was only seen in the subtotally resected patients. The number AV+/CD62E- MPs increased in GBM patients and correlated with MP activity. TF+/GFAP- MPs numbers were significantly higher in 11 GBM patients who developed VTE than in those who did not (p 0.04). TF+/GFAP- MPs levels above the 90th percentile (calculated in GBM patients without VTE) were associated with a higher risk of VTE (RR 4.17, 95% CI 1.57-11.03). In conclusion, the numbers of glial-derived and/or TF-bearing MPs were high in GBM patients both before and even more after the neoplasm was treated, especially in patients with subtotal resection likely according to disease progression. A contribution of TF+/GFAP- MPs to the risk of VTE is suggested.

    Topics: Adult; Aged; Brain Neoplasms; Case-Control Studies; Cell-Derived Microparticles; E-Selectin; Endothelial Cells; Female; Glial Fibrillary Acidic Protein; Glioblastoma; Humans; Male; Middle Aged; Neuroglia; Risk Factors; Thromboplastin; Venous Thromboembolism

2013
A combination of radiosurgery and soluble tissue factor enhances vascular targeting for experimental glioblastoma.
    BioMed research international, 2013, Volume: 2013

    Radiosurgery for glioblastoma is limited to the development of resistance, allowing tumor cells to survive and initiate tumor recurrence. Based on our previous work that coadministration of tissue factor and lipopolysaccharide following radiosurgery selectively induced thrombosis in cerebral arteriovenous malformations, achieving thrombosis of 69% of the capillaries and 39% of medium sized vessels, we hypothesized that a rapid and selective shutdown of the capillaries in glioblastoma vasculature would decrease the delivery of oxygen and nutrients, reducing tumor growth, preventing intracranial hypertension, and improving life expectancy. Glioblastoma was formed by implantation of GL261 cells into C57Bl/6 mouse brain. Mice were intravenously injected tissue factor, lipopolysaccharide, a combination of both, or placebo 24 hours after radiosurgery. Control mice received both agents after sham irradiation. Coadministration of tissue factor and lipopolysaccharide led to the formation of thrombi in up to 87 ± 8% of the capillaries and 46 ± 4% of medium sized vessels within glioblastoma. The survival rate of mice in this group was 80% versus no survivor in placebo controls 30 days after irradiation. Animal body weight increased with time in this group (r = 0.88, P = 0.0001). Thus, radiosurgery enhanced treatment with tissue factor, and lipopolysaccharide selectively induces thrombosis in glioblastoma vasculature, improving life expectancy.

    Topics: Animals; Combined Modality Therapy; Disease Models, Animal; Glioblastoma; Humans; Lipopolysaccharides; Mice; Neoplasm Recurrence, Local; Neoplasms, Experimental; Radiosurgery; Thromboplastin; Thrombosis

2013
Anti-tissue factor (TF9-10H10) treatment reduces tumor cell invasiveness in a novel migratory glioma model.
    Neuropathology : official journal of the Japanese Society of Neuropathology, 2013, Volume: 33, Issue:5

    In vitro and descriptive studies of human tissue samples revealed the pro-coagulant glycoprotein tissue factor (TF) as a potent player in glioma cell infiltration that is activated by hypoxia and has also been shown to be upregulated by mutations of TP53 or PTEN. Here we present the morphological and genetic characterization of a novel glioblastoma in vivo model and provide evidence that treatment with an antibody targeting TF leads to reduced glioma cell invasiveness. Therefore, we established a murine xenograft treatment model by transplanting the angiogenic and diffusely infiltrating human glioma cell line MZ-18 with endogenous TF expression into nude mice brains and treating these mice with an intracranial osmotic pump system continuously infusing a monoclonal antibody against TF (mAb TF9-10H10). The human MZ-18 cell line harbors two TP53 mutations resulting in a strong nuclear accumulation of p53, thereby facilitating the unambiguous identification of tumor cells in the xenograft model. Intracranial application of TF9-10H10 significantly reduced invasion of MZ-18 cells compared to mock-treated control animals. The extent of activated blood vessels was also reduced upon anti-TF treatment. Thus, targeting the TF pathway might be a promising treatment strategy for future glioblastoma therapies, by affecting both invading tumor cells and tumor vasculature.

    Topics: Animals; Antibodies, Monoclonal; Brain Neoplasms; Cell Line; Glioblastoma; Humans; Mice; Mice, Nude; Neoplasm Invasiveness; Receptor, PAR-2; Thromboplastin; Tumor Suppressor Protein p53; Xenograft Model Antitumor Assays

2013
Oncogenic epidermal growth factor receptor up-regulates multiple elements of the tissue factor signaling pathway in human glioma cells.
    Blood, 2010, Aug-05, Volume: 116, Issue:5

    Cancer cells frequently overexpress tissue factor (TF) and become procoagulant. This conversion may be driven by genetic transformation, including through the expression of the oncogenic epidermal growth factor receptor (EGFR) and its mutant, EGFRvIII, present in glioblastoma multiforme (GBM). Here we show that the EGFRvIII-dependent GBM cell transformation is associated with the onset of the simultaneous overexpression of TF, protease-activated receptors 1 and 2 (PAR1 and PAR2), and ectopic synthesis of factor VII (FVII). Efficient generation of factor Xa by these cells still requires exogenous FVIIa. However, as a result of EGFRvIII-dependent transformation, GBM cells become hypersensitive to TF/PAR-mediated signaling and produce ample angiogenic factors (vascular endothelial growth factor and interleukin-8) on exposure to FVIIa and PAR1- or PAR2-activating peptides. Thus, oncogenes may cause complex changes in the ability of GBM cancer cells to interact with the coagulation system, thereby exacerbating its influence on angiogenesis and disease progression.

    Topics: Brain Neoplasms; Cell Line, Tumor; ErbB Receptors; Gene Expression Regulation, Neoplastic; Genes, erbB-1; Glioblastoma; Humans; Mutation; Neoplasm Proteins; Oncogenes; Receptor, PAR-1; Receptor, PAR-2; Signal Transduction; Thromboplastin; Up-Regulation

2010
The effect of epidermal growth factor receptor variant III on glioma cell migration by stimulating ERK phosphorylation through the focal adhesion kinase signaling pathway.
    Archives of biochemistry and biophysics, 2010, Oct-15, Volume: 502, Issue:2

    Epidermal growth factor receptor variant III (EGFRvIII), the most common EGFR mutation, is associated with cell migration of glioblastoma multiforme (GBM) cases; however, the mechanism has not been elucidated. In this study, we found that the EGFRvIII-promoted glioma cell migration was closely linked to high levels of tyrosine phosphorylation in focal adhesion kinase (FAK) Y397. We also demonstrated that EGFRvIII formed a complex with FAK, resulting in enhanced tyrosine phosphorylation levels of FAK Y397 and EGFR Y1068. After knockdown of FAK expression via anti-FAK shRNA, the U87ΔEGFR cell migration was significantly inhibited, accompanying with the reduced phosphorylation levels of extracellular signal-regulated kinase (ERK1/2). Furthermore, the role of ERK1/2 in FAK-regulated cell migration was confirmed. Taken together, our results suggest that FAK and its downstream molecule ERK were involved in EGFRvIII-promoted glioma cell migration in U87ΔEGFR cells.

    Topics: Cell Movement; Epidermis; ErbB Receptors; Extracellular Signal-Regulated MAP Kinases; Focal Adhesion Kinase 1; Focal Adhesion Protein-Tyrosine Kinases; Glioblastoma; Glioma; Humans; Phosphorylation; Signal Transduction; Thromboplastin

2010
Epidermal growth factor receptor and PTEN modulate tissue factor expression in glioblastoma through JunD/activator protein-1 transcriptional activity.
    Cancer research, 2009, Mar-15, Volume: 69, Issue:6

    Hypoxia and necrosis are fundamental features of glioblastoma (GBM) and their emergence is critical for the rapid biological progression of this fatal tumor; yet, underlying mechanisms are poorly understood. We have suggested that vaso-occlusion following intravascular thrombosis could initiate or propagate hypoxia and necrosis in GBM. Tissue factor (TF), the main cellular initiator of coagulation, is overexpressed in GBMs and likely favors a thrombotic microenvironment. Epidermal growth factor receptor (EGFR) amplification and PTEN loss are two common genetic alterations seen in GBM but not in lower-grade astrocytomas that could be responsible for TF up-regulation. The most frequent EGFR mutation in GBM involves deletion of exons 2 to 7, resulting in the expression of a constitutively active receptor, EGFRvIII. Here, we show that overexpression of EGFR or EGFRvIII in human glioma cells causes increased basal TF expression and that stimulation of EGFR by its ligand, EGF, leads to a marked dose-dependent up-regulation of TF. In all cases, increased TF expression led to accelerated plasma coagulation in vitro. EGFR-mediated TF expression depended most strongly on activator protein-1 (AP-1) transcriptional activity and was associated with c-Jun NH(2)-terminal kinase (JNK) and JunD activation. Restoration of PTEN expression in PTEN-deficient GBM cells diminished EGFR-induced TF expression by inhibiting JunD/AP-1 transcriptional activity. PTEN mediated this effect by antagonizing phosphatidylinositol 3-kinase activity, which in turn attenuated both Akt and JNK activities. These mechanisms are likely at work in vivo, as EGFR expression was highly correlated with TF expression in human high-grade astrocytoma specimens.

    Topics: Brain Neoplasms; Cell Line, Tumor; Epidermal Growth Factor; ErbB Receptors; Glioblastoma; Humans; JNK Mitogen-Activated Protein Kinases; MAP Kinase Signaling System; Mitogen-Activated Protein Kinase 1; Mitogen-Activated Protein Kinase 3; Oncogene Protein v-akt; Phosphatidylinositol 3-Kinases; Phosphoinositide-3 Kinase Inhibitors; Promoter Regions, Genetic; Proto-Oncogene Proteins c-jun; PTEN Phosphohydrolase; Signal Transduction; Thromboplastin; Transcription Factor AP-1; Transcription, Genetic; Up-Regulation

2009
Ixolaris, a tissue factor inhibitor, blocks primary tumor growth and angiogenesis in a glioblastoma model.
    Journal of thrombosis and haemostasis : JTH, 2009, Volume: 7, Issue:11

    The expression levels of the clotting initiator protein Tissue Factor (TF) correlate with vessel density and the histological malignancy grade of glioma patients. Increased procoagulant tonus in high grade tumors (glioblastomas) also indicates a potential role for TF in progression of this disease, and suggests that anticoagulants could be used as adjuvants for its treatment.. We hypothesized that blocking of TF activity with the tick anticoagulant Ixolaris might interfere with glioblastoma progression.. TF was identified in U87-MG cells by flow-cytometric and functional assays (extrinsic tenase). In addition, flow-cytometric analysis demonstrated the exposure of phosphatidylserine in the surface of U87-MG cells, which supported the assembly of intrinsic tenase (FIXa/FVIIIa/FX) and prothrombinase (FVa/FXa/prothrombin) complexes, accounting for the production of FXa and thrombin, respectively. Ixolaris effectively blocked the in vitro TF-dependent procoagulant activity of the U87-MG human glioblastoma cell line and attenuated multimolecular coagulation complexes assembly. Notably, Ixolaris inhibited the in vivo tumorigenic potential of U87-MG cells in nude mice, without observable bleeding. This inhibitory effect of Ixolaris on tumor growth was associated with downregulation of VEGF and reduced tumor vascularization.. Our results suggest that Ixolaris might be a promising agent for anti-tumor therapy in humans.

    Topics: Animals; Cell Line, Tumor; Cell Proliferation; Disease Progression; Down-Regulation; Glioblastoma; Humans; Mice; Mice, Nude; Neovascularization, Pathologic; Salivary Proteins and Peptides; Thromboplastin; Vascular Endothelial Growth Factor A

2009
Aberrant NF-kappaB activity is critical in focal necrosis formation of human glioblastoma by regulation of the expression of tissue factor.
    International journal of oncology, 2008, Volume: 33, Issue:1

    Focal necrosis is a key pathologic feature that distinguishes glioblastoma from lower grade glioma. The presence of necrosis in a glioblastoma could promote its rapid growth and clinical progression. Focal necrosis of glioblastoma seems to be associated with thrombosis that result from hyper-coagulability. In the present study, we found that glioblastoma cells had a high level of constitutive nuclear factor (NF)-kappaB activity, which was directly correlated with necrosis in glioblastomas. We also found a direct correlation between NF-kappaB activity and the expression of tissue factor (TF), a potent procoagulant factor in gliomas. Inhibition of TF by an inhibitory antibody prevented the procoagulant activity of glioblastoma cells, indicating a TF-dependent mechanism. Blockade of NF-kappaB activation significantly inhibited TF expression and the procoagulant activity of glioblastoma cells in vitro. Blockade of NF-kappaB activation also significantly inhibited in vivo expression of TF, which was directly correlated with decreased necrosis formation and tumor growth of glioblastoma cells in nude mice. Collectively, these results suggest that elevated NF-kappaB activity in glioblastomas cells plays a critical role in necrosis formation of glioblastoma and that inhibition of NF-kappaB activity in glioblastoma can suppress necrosis formation and progressive growth.

    Topics: Cell Line, Tumor; Glioblastoma; Humans; I-kappa B Proteins; Immunohistochemistry; Interleukin-8; Necrosis; NF-kappa B; NF-KappaB Inhibitor alpha; Thromboplastin; Transfection; Vascular Endothelial Growth Factor A

2008
Early growth response gene-1 regulates hypoxia-induced expression of tissue factor in glioblastoma multiforme through hypoxia-inducible factor-1-independent mechanisms.
    Cancer research, 2006, Jul-15, Volume: 66, Issue:14

    Hypoxia strongly up-regulates tissue factor and promotes plasma clotting by glioblastoma multiforme, but transcriptional mechanisms remain undefined. Here, we investigated the potential roles of early growth response gene-1 (Egr-1), Sp1, nuclear factor-kappaB (NF-kappaB), activator protein-1 (AP-1), and hypoxia-inducible factor-1 (HIF-1) in the hypoxic regulation of tissue factor by glioblastoma multiforme cells in vitro. Hypoxia (1% O2) strongly induced Egr-1 mRNA within 1 hour and led to nuclear localization of Egr-1 protein. Using luciferase reporter plasmids in glioma cells, we found that hypoxia dramatically increased luciferase activity in cells with constructs containing Egr-1-binding sites but not in cells with constructs containing AP-1- or NF-kappaB-binding sites. Electrophoretic mobility shift assays revealed hypoxia-induced Egr-1, but not Sp1, binding to oligonucleotides containing the Egr-1/Sp1 motif of tissue factor gene promoter. Using an expression vector containing the minimal tissue factor promoter (-111 to +14 bp) and small interfering RNA (siRNA) directed at Egr-1 and Sp1 mRNAs, we found that Egr-1 was required for maximal hypoxic induction of promoter activity. Forced overexpression of Egr-1 but not Sp1 by cDNA transfection caused up-regulation of tissue factor in glioma cells under normoxia (21% O2), whereas siRNA directed at Egr-1 strongly attenuated hypoxia-induced tissue factor expression. To examine the effects of HIF-1alpha on tissue factor expression, we used glioma cells stably transfected with a HIF-1alpha siRNA expression vector and found that HIF-1alpha mRNA silencing did not affect tissue factor expression under hypoxia. We conclude that hypoxic up-regulation of tissue factor in glioblastoma multiforme cells depends largely on Egr-1 and is independent of HIF-1.

    Topics: Cell Hypoxia; Cell Line, Tumor; Early Growth Response Protein 1; Glioblastoma; Humans; Hypoxia-Inducible Factor 1; NF-kappa B; Promoter Regions, Genetic; Proto-Oncogene Proteins c-jun; Sp1 Transcription Factor; Thromboplastin; Transcription Factor AP-1; Transfection; Up-Regulation; Vascular Endothelial Growth Factor A

2006
PTEN and hypoxia regulate tissue factor expression and plasma coagulation by glioblastoma.
    Cancer research, 2005, Feb-15, Volume: 65, Issue:4

    We have previously proposed that intravascular thrombosis and subsequent vasoocclusion contribute to the development of pseudopalisading necrosis, a pathologic hallmark that distinguishes glioblastoma (WHO grade 4) from lower grade astrocytomas. To better understand the potential prothrombotic mechanisms underlying the formation of these structures that drive tumor angiogenesis, we investigated tissue factor (TF), a potent procoagulant protein known to be overexpressed in astrocytomas. We hypothesized that PTEN loss and tumor hypoxia, which characterize glioblastoma but not lower grade astrocytomas, could up-regulate TF expression and cause intravascular thrombotic occlusion. We examined the effect of PTEN restoration and hypoxia on TF expression and plasma coagulation using a human glioma cell line containing an inducible wt-PTEN cDNA. Cell exposure to hypoxia (1% O(2)) markedly increased TF expression, whereas restoration of wt-PTEN caused decreased cellular TF. The latter effect was at least partially dependent on PTEN's protein phosphatase activity. Hypoxic cells accelerated plasma clotting in tilt tube assays and this effect was prevented by both inhibitory antibodies to TF and plasma lacking factor VII, implicating TF-dependent mechanisms. To further examine the genetic events leading to TF up-regulation during progression of astrocytomas, we investigated its expression in a series of human astrocytes sequentially infected with E6/E7/human telomerase, Ras, and Akt. Cells transformed with Akt showed the greatest incremental increase in hypoxia-induced TF expression and secretion. Together, our results show that PTEN loss and hypoxia up-regulate TF expression and promote plasma clotting by glioma cells, suggesting that these mechanisms may underlie intravascular thrombosis and pseudopalisading necrosis in glioblastoma.

    Topics: Blood Coagulation; Cell Hypoxia; Cell Line, Tumor; Glioblastoma; Humans; Phosphatidate Phosphatase; Phosphoric Monoester Hydrolases; Protein Serine-Threonine Kinases; Proto-Oncogene Proteins; Proto-Oncogene Proteins c-akt; PTEN Phosphohydrolase; ras Proteins; Thromboplastin; Tumor Suppressor Proteins; Up-Regulation

2005
Coxsackievirus and adenovirus receptor (CAR) is modified and shed in membrane vesicles.
    Biochemistry, 2004, Jun-29, Volume: 43, Issue:25

    Vesicles shed by U87-MG cells contain coxsackievirus and adenovirus receptor (CAR) protein that has been posttranslationally modified. Relative to full-length CAR, migration of the vesicle-associated soluble CAR antigen (CARd6) on SDS-polyacrylamide gels indicated a loss of approximately 6 kDa. HeLa and END-HHV6 cells also shed a similar vesicle-associated CAR protein. Vesicles shed by U87-MG cells following stimulation with calcium and A23187 contained CARd6 similar to that present in vesicles shed constitutively. RD cells transfected to express full-length CAR produced CARd6, but cells that expressed CAR with a truncated cytoplasmic domain produced no equivalent to CARd6. In U87-MG cells, calpain activity was required for release of CARd6 with shed vesicles, and accumulation of CARd6 in cells that rounded up and released from the plastic substrate in response to A23187 treatment was blocked by N-ethylmaleimide. These experiments show that CAR, posttranslationally modified in the cytoplasmic domain, can be released with vesicles shed by cells. Posttranslational modification of the CAR cytoplasmic domain occurs during cell rounding and release from the culture substrate. This modified, vesicle-associated CAR was the principal form of soluble CAR released by the cells.

    Topics: Adenoviridae; Calcimycin; Cell Adhesion; Cell Line, Tumor; Culture Media; Cytoplasmic Vesicles; Dipeptides; Endothelial Cells; Enterovirus; Glioblastoma; HeLa Cells; Humans; Protein Processing, Post-Translational; Protein Structure, Tertiary; Receptors, Virus; Thromboplastin; Umbilical Veins

2004
Tissue factor, osteopontin, alphavbeta3 integrin expression in microvasculature of gliomas associated with vascular endothelial growth factor expression.
    British journal of cancer, 2000, Volume: 82, Issue:12

    Vascular endothelial growth factor (VEGF) is a potent angiogenic factor in human gliomas. VEGF-induced proteins in endothelial cells, tissue factor (TF), osteopontin (OPN) and alphavbeta3 integrin have been implicated as important molecules by which VEGF promotes angiogenesis in vivo. Sixty-eight gliomas were immunohistochemically stained with TF, VEGF, OPN and alphavbeta3 integrin antibody. Twenty-three tumours, six normal brains and nine glioma cell lines were evaluated for their mRNA expression of VEGF and TF by reverse transcription polymerase chain reaction analysis. The data indicated that TF as well as VEGF was a strong regulator of human glioma angiogenesis. First, TF expression in endothelial cells which was observed in 74% of glioblastomas, 54% of anaplastic astrocytomas and none of low-grade astrocytomas, correlated with the microvascular density of the tumours. Double staining for VEGF and TF demonstrated co-localization of these two proteins in the glioblastoma tissues. Second, there was a correlation between TF and VEGF mRNA expression in the glioma tissues. Third, glioma cell conditioned medium containing a large amount of VEGF up-regulated the TF mRNA expression in human umbilical vein endothelial cells. OPN and alphavbeta3 integrin, were also predominantly observed in the microvasculature of glioblastomas associated with VEGF expression. Microvascular expression of these molecules could be an effective antiangiogenesis target for human gliomas.

    Topics: Brain Neoplasms; Endothelial Growth Factors; Glioblastoma; Glioma; Humans; Lymphokines; Microcirculation; Neovascularization, Pathologic; Osteopontin; Receptors, Vitronectin; Sialoglycoproteins; Thromboplastin; Tumor Cells, Cultured; Vascular Endothelial Growth Factor A; Vascular Endothelial Growth Factors

2000
Suppression of the tissue factor-dependent coagulation cascade: a contributing factor for the development of intratumoral hemorrhage in glioblastoma.
    International journal of molecular medicine, 2000, Volume: 6, Issue:3

    To clarify factors that may contribute to the development of intratumoral hemorrhage, we analyzed the expression of tissue factor (TF), an initiator of the extrinsic coagulation pathway, and of tissue factor pathway inhibitor (TFPI) in glioblastomas with or without massive intratumoral hematoma. Among 196 glioma cases reviewed, there were 13 with macroscopic intratumoral hemorrhage. We focused on the glioblastomas and used immunoblot- and immunohistochemical methods to compare the expression of TF and TFPI in 9 glioblastomas with macroscopic hematoma and 30 glioblastomas without macroscopic hemorrhage. Although TF was expressed in most glioblastomas irrespective of the presence or absence of macroscopic hemorrhage, the staining patterns differed significantly: TF-positive glioma cells were diffusely present in the non-hemorrhage group; in the group with hemorrhage, positive cells, primarily macrophages, were scattered throughout the tissue examined. The expression of TFPI was significantly higher in the group with than in the group without hemorrhage. Our results suggest that local suppression of the TF-dependent coagulation cascade is a contributing factor that permits the occurrence of intratumoral hemorrhage.

    Topics: Adult; Aged; Brain Neoplasms; Child, Preschool; Female; Glioblastoma; Glioma; Hematoma; Hemorrhage; Humans; Immunoblotting; Immunohistochemistry; Lipoproteins; Macrophages; Magnetic Resonance Imaging; Male; Middle Aged; Thromboplastin; Tomography, X-Ray Computed

2000
Sp1 decoy transfected to carcinoma cells suppresses the expression of vascular endothelial growth factor, transforming growth factor beta1, and tissue factor and also cell growth and invasion activities.
    Cancer research, 2000, Nov-15, Volume: 60, Issue:22

    Vasculature development is thought to be an important aspect in the growth and metastasis of solid tumors. Among the angiogenic factors produced by tumor cells, vascular endothelial growth factor is considered to be the most potent and pathologically important. The synthesis of this growth factor has been shown to be modulated through Sp1 function following stimulation by tumor necrosis factor alpha (TNF-alpha). Oligodeoxynucleotides (ODNs) were synthesized with either the consensus sequence for Sp1 binding (Sp1 decoy ODNs) or a mutated form of this sequence (mt-Sp1 decoy ODNs). Using the hemagglutinating virus of Japan (HVJ)-liposome method, we transferred these ODNs into cultured cancer cells (A549 and U251 cells). The TNF-alpha-mediated expression of both VEGF and transforming growth factor beta1 and tissue factor (TF) by the cancer cells could be simultaneously suppressed to less than 30% by transfection of Sp1 decoy ODNs but not by mt-Sp1 decoy ODNs. In addition, in vitro invasiveness, synthesis of mRNA for urokinase-type plasminogen activator, and cell proliferation of both cell lines were also inhibited to 40% by the transfection of only Sp1 decoy ODNs. These results suggested that the Sp1 decoy strategy would be effective for regulating tumor growth by simultaneously reducing cancer cell (a) angiogenic growth factor expression, (b) proliferation, and (c) invasiveness.

    Topics: Adenocarcinoma; Binding Sites; Cell Division; Cell Movement; Endothelial Growth Factors; Fluorescein-5-isothiocyanate; Fluorescent Dyes; Glioblastoma; Humans; Liposomes; Lung Neoplasms; Lymphokines; Neoplasm Invasiveness; Oligonucleotides; Respirovirus; RNA, Messenger; Sp1 Transcription Factor; Thromboplastin; Transcriptional Activation; Transfection; Transforming Growth Factor beta; Transforming Growth Factor beta1; Tumor Cells, Cultured; Tumor Necrosis Factor-alpha; Vascular Endothelial Growth Factor A; Vascular Endothelial Growth Factors

2000
Tissue factor and cell morphology variations in cell lines subcloned from U87-MG.
    Blood coagulation & fibrinolysis : an international journal in haemostasis and thrombosis, 1998, Volume: 9, Issue:6

    Tissue factor is heterogeneously distributed within and among cells in cultures of U87-MG, a glioblastoma-derived line. The heterogeneity among cells may reflect the presence of distinct populations within the U87-MG cultures. This hypothesis has been confirmed by cloning of five distinct sublines from the parent population. These subpopulations have remained distinct through 4 months of growth in culture and one cycle of cryogenic preservation and thawing. The cultures differ in growth rates, amounts of tissue factor activity expressed, tissue factor antigen measured by flow cytometry, and patterns of tissue factor distribution studied by immunofluorescence microscopy. Characterization of these sublines allowed us to recognize that the tissue factor distribution on polarized cells (e.g. spindle-shaped) differed from that on cells with less polar morphologies. Finely speckled tissue factor staining tended to be localized to polarized aspects of the cell body where actin stress fibers are commonly present, whereas larger distinct foci of tissue factor were present in regions of membrane spreading. These results show that tissue factor is distributed differently in distinct regions of plasma membrane differentiation. Furthermore, the isolation of distinct stable subpopulations by dilutional cloning of U87-MG cultures serves as a reminder that cell culture heterogeneity can complicate experiments using molecular genetic manipulation of cultured cells which require clonal isolation of genetically altered lines.

    Topics: Cell Division; Cell Membrane; Cell Size; Clone Cells; Cryopreservation; Flow Cytometry; Fluorescent Antibody Technique; Glioblastoma; Thromboplastin; Tumor Cells, Cultured

1998
Expression of tissue factor correlates with grade of malignancy in human glioma.
    Cancer, 1996, May-01, Volume: 77, Issue:9

    Tissue factor (TF), a cell surface receptor of factor VII/VIIa, was initially recognized as an initiator of the extrinsic coagulation pathway. TF has recently been found to be expressed highly in certain types of malignant tumors. In addition, TF belongs to the interferon receptor family and is one of the immediate early genes, suggesting that TF may participate in the regulation of cell growth. However, the correlation between the expression of TF and cell growth is still unclear.. Expression of TF in 6 glioma cell lines and 44 glioma surgical specimens was studied by Northern blot analysis, Western blot analysis, immunohistochemistry, and in situ hybridization.. Northern blot analysis showed that glioma cells expressed minor novel transcripts of 3.3 kb and 1.6 kb, in addition to the transcripts of 2.2 kb and 3.1 kb that were previously reported. Western blot analysis revealed that the level of TF protein did not correlate with that of TF transcripts. Although immunohistochemical analysis of surgical specimens showed that all gliomas were positive for TF, it was interesting that 1 of 10 benign gliomas (10%) was positive for TF (malignancy Grade I-II), 13 of 14 anaplastic astrocytomas (86%) (malignancy grade III) and 19 of 20 glioblastomas (95%) (malignancy grade IV) were moderately or strongly positive for TF. In situ hybridization showed the expression of TF mRNA in glioma cells.. TF is expressed in glioma and the level of expression correlates with the histologic grade of malignancy.

    Topics: Astrocytoma; Blotting, Northern; Blotting, Western; Cell Division; Gene Expression Regulation, Neoplastic; Genes, Immediate-Early; Glioblastoma; Glioma; Humans; Immunohistochemistry; In Situ Hybridization; Receptors, Cell Surface; Receptors, Interferon; RNA, Messenger; Thromboplastin; Transcription, Genetic; Tumor Cells, Cultured

1996
Expression of tissue factor in glioma.
    Noshuyo byori = Brain tumor pathology, 1996, Volume: 13, Issue:2

    Tissue factor (TF) is a cell surface glycoprotein that initiates the extrinsic coagulation protease cascade and it is expressed in some tumor cells. TF belongs to the interferon receptor family, and it is one of the early immediate genes, suggesting that TF has a biological function other than hemostasis. We investigated the expression of TF in gliomas. Immunocytochemistry showed the expression of TF in 3 glioma cell lines. Immunohistochemical analysis of 44 surgical specimens revealed that all gliomas were positive for TF, and 19 (95%) of 20 glioblastomas, 12 (86%) of 14 anaplastic astrocytomas and 1 (10%) of 10 benign gliomas were moderately or strongly positive for TF. Our study showed that TF is expressed in gliomas, and that the level of TF expression is correlated with the grade of malignancy of the glioma, suggesting that TF may participate in cell growth.

    Topics: Brain Neoplasms; Cell Division; Glioblastoma; Glioma; Humans; Immunohistochemistry; Neoplasm Staging; Thromboplastin; Tumor Cells, Cultured

1996
Hyperfibrinolysis during intracranial surgery: effect of high dose aprotinin.
    Journal of neurology, neurosurgery, and psychiatry, 1995, Volume: 58, Issue:1

    A patient undergoing intracranial surgery developed disseminated intravascular coagulation with life threatening peroperative bleeding. Thromboelastography established the diagnosis of hyperfibrinolysis, usually a fatal complication of a neurosurgical operation. With the administration of a high dose regimen of aprotinin (Trasylol) the haemorrhage was controlled and the hyperfibrinolytic state reversed. Evaluation of blood samples from the jugular bulb suggested that there was a pronounced local release of tissue plasminogen activator into the circulation.

    Topics: Aged; Aprotinin; Brain; Brain Neoplasms; Craniotomy; Dose-Response Relationship, Drug; Fatal Outcome; Fibrinolysis; Glioblastoma; Hemorrhage; Humans; Injections, Intravenous; Male; Postoperative Complications; Prothrombin; Thrombelastography; Thromboplastin

1995
A high-sensitivity thromboplastin reagent prepared from cultured human cells.
    Blood coagulation & fibrinolysis : an international journal in haemostasis and thrombosis, 1994, Volume: 5, Issue:4

    High-sensitivity thromboplastin reagents suitable for use in the prothrombin time (PT) assay are typically prepared from human brain and placenta, tissues that are in limited supply and subject to viral contamination. Cloning and expression of recombinant human tissue factor (TF) has enabled production of a new generation of thromboplastin reagents whose performance and utility are under active investigation. The purpose of this study was to determine the feasibility of producing a sensitive human thromboplastin reagent from a non-recombinant source: cultured human cells. Several human cell lines with apparently high constitutive TF synthesis were identified, and a viable thromboplastin reagent (Humaplastin) was produced from a human lung cell line via a non-conventional process that did not require reconstitution or rehydration of TF in cell membranes. When calibrated against BCT/253, a human brain international reference thromboplastin, Humaplastin exhibited a mean normal prothrombin time of 12.6 +/- 0.7 s (mean +/- SD: n = 20) and an International Sensitivity Index of 1.09 +/- 0.019. The performance of this reagent was well correlated (r = 0.983) with Thromborel S, a commercially available human placental thromboplastin reagent. Orthogonal least squares regression of the log PT values from the placental thromboplastin reagent versus Humaplastin and two recombinant TF-based thromboplastin reagents suggested that the latter three reagents are somewhat more sensitive than the placental thromboplastin reagent, although such differences should not be expected to have a significant impact on clinical utility. It is concluded that cultured human lung cells represent a suitable source of tissue thromboplastin for production of a high-sensitivity non-recombinant thromboplastin reagent.

    Topics: Adenocarcinoma; Anticoagulants; Astrocytoma; Blood Coagulation Factors; Brain; Brain Neoplasms; Calibration; Carcinoma, Squamous Cell; Cells, Cultured; Choriocarcinoma; Feasibility Studies; Female; Glioblastoma; Histiocytosis, Langerhans-Cell; Humans; Indicators and Reagents; Lung; Lung Neoplasms; Neoplasm Proteins; Placenta; Prothrombin Time; Recombinant Proteins; Reference Standards; Sensitivity and Specificity; Thromboplastin; Tumor Cells, Cultured; Uterine Neoplasms

1994
Immunofluorescent studies of tissue factor on U87MG cells: evidence for non-uniform distribution.
    Blood coagulation & fibrinolysis : an international journal in haemostasis and thrombosis, 1993, Volume: 4, Issue:6

    U87MG cells (human glioblastoma) express tissue factor and shed membrane-derived vesicles enriched in procoagulant activity. Tissue factor antigen has been studied by flow cytometry, immunofluorescent microscopy and Western blotting. Flow cytometric analysis utilized monoclonal antibodies recognizing the tissue factor extracellular domain and the carboxyl-terminal nine amino acids. Studies with intact and permeabilized cells support the location of the carboxyl-terminal domain in the cytoplasm, as previously predicted from the protein sequence. Immunofluorescent microscopy revealed a heterogeneous staining pattern, indicating that tissue factor antigen may be clustered on the cell surface. Intense staining was occasionally observed in cytoplasmic extensions and membrane regions that appeared to be extruding from the cells. Western blot analysis of vesicles shed into the culture medium revealed a principal tissue factor band with mobility marginally slower than that of placental tissue factor. Both extracellular and cytoplasmic epitopes were present in this vesicular tissue factor.

    Topics: Flow Cytometry; Glioblastoma; Humans; Microscopy, Fluorescence; Thromboplastin; Tumor Cells, Cultured

1993
Blood clotting changes in guinea pigs with a heterologous intracranial neoplasm.
    Laboratory investigation; a journal of technical methods and pathology, 1970, Volume: 23, Issue:2

    Topics: Animals; Blood Coagulation Disorders; Blood Coagulation Tests; Blood Platelets; Brain Neoplasms; Factor V; Factor VIII; Fibrinogen; Glioblastoma; Guinea Pigs; Humans; Neoplasm Transplantation; Neoplasms, Experimental; Prothrombin; Prothrombin Time; Thromboplastin; Transplantation, Heterologous

1970