Hypoxia regulates the expression of tissue factor pathway signaling elements in a rat glioma model

  • Authors:
    • Robson Q. Monteiro
    • Luize G. Lima
    • Nathália P. Gonçalves
    • Mayara R. Arruda De Souza
    • Ana C. Leal
    • Marcos A. Almeida Demasi
    • Mari C. Sogayar
    • Tatiana C. Carneiro‑Lobo
  • View Affiliations

  • Published online on: May 17, 2016     https://doi.org/10.3892/ol.2016.4593
  • Pages: 315-322
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Hypoxia and necrosis are fundamental features of glioma, and their emergence is critical for the rapid biological progression of this fatal tumor. The presence of vaso‑occlusive thrombus is higher in grade IV tumors [glioblastoma multiforme (GBM)] compared with lower grade tumors, suggesting that the procoagulant properties of the tumor contribute to its aggressive behavior, as well as the establishment of tumor hypoxia and necrosis. Tissue factor (TF), the primary cellular initiator of coagulation, is overexpressed in GBMs and likely favors a thrombotic microenvironment. Phosphatase and tensin homolog (PTEN) loss and hypoxia are two common alterations observed in glioma that may be responsible for TF upregulation. In the present study, ST1 and P7 rat glioma lines, with different levels of aggressiveness, were comparatively analyzed with the aim of identifying differences in procoagulant mechanisms. The results indicated that P7 cells display potent procoagulant activity compared with ST1 cells. Flow cytometric analysis showed less pronounced levels of TF in ST1 cells compared with P7 cells. Notably, P7 cells supported factor X (FX) activation via factor VIIa, whereas no significant FXa generation was observed in ST1 cells. Furthermore, the exposure of phosphatidylserine on the surface of P7 and ST1 cells was investigated. The results supported the assembly of prothrombinase complexes, accounting for the production of thrombin. Furthermore, reverse transcription‑quantitative polymerase chain reaction showed that CoCl2 (known to induce a hypoxic‑like stress) led to an upregulation of TF levels in P7 and ST1 cells. Therefore, increased TF expression in P7 cells was accompanied by increased TF procoagulant activity. In addition, hypoxia increased the shedding of procoagulant TF‑bearing microvesicles in both cell lines. Finally, hypoxic stress induced by treatment with CoCl2 upregulated the expression of the PAR1 receptor in both P7 and ST1 cells. In addition to PAR1, P7, but not ST1 cells, expressed higher levels of PAR2 under hypoxic stress. Thus, modulating these molecular interactions may provide additional insights for the development of more efficient therapeutic strategies against aggressive glioma.

Introduction

Tissue factor (TF), the primary initiator of the coagulation cascade, is a 47-kDa cell-associated transmembrane protein that acts as a high-affinity receptor for factor VIIa (FVIIa). Formation of the TF/FVIIa complex (extrinsic tenase complex) on cellular surfaces triggers the generation of active coagulation proteases, followed by deposition of activated platelets and fibrin (1). TF is constitutively expressed in specific cell types and has been shown to be upregulated in a number of pathological processes (2,3).

A strong correlation between TF expression and malignancy grade has been reported for several tumor types, and may result in vascular occlusion and hypoxia, therefore influencing various aspects of tumor progression, including angiogenesis, metastasis and other cellular responses (48). Increased TF expression has been also associated with cancer-associated thrombosis, a highly relevant systemic response (8,9). In fact, several studies suggest that circulating extracellular vesicles containing TF may be important in a patient's prothrombotic state (10,11).

The majority of the pro-tumoral responses induced by TF are associated with the functions of activated clotting factors [FVIIa, factor Xa (FXa) and thrombin] as well as other proteases, such as matrix metalloproteinase-1 (MMP1), in cleaving and activating G protein-coupled protease-activated receptors (PARs) (7,9,12). Activation of PAR1 by thrombin or MMP1 triggers a number of tumor cell responses, while the TF/FVIIa binary complex predominantly functions through activation of PAR2. In fact, it is proposed that PAR1 and PAR2 induce several redundant responses in tumor cells (7,13).

Glioblastoma multiforme (GBM), the most common type of adult brain tumor, infiltrates the normal brain area, rendering complete surgical resection impossible. The disease has a poor median survival time of 15 months after diagnosis, despite the application of aggressive multimodality treatments following surgery, such as radiation and chemotherapy (14). The aggressiveness of GBM has been correlated with severe hypoxia, which generates large areas of necrosis and an extensive, hyperpermeable vasculature (15). Hypoxia activation is a hallmark of growing tumors that occurs as a result of inadequate oxygen supply. Increased angiogenesis serves to restore the influx of nutrients and oxygen to the tumor environment (16). It has been proposed that the TF signaling pathway has a relevant role in GBM progression (17,18). Therefore, oncogenic signaling pathways, such as the expression of epidermal growth factor receptor (EGFR) and its mutant, EGFRvIII, as well as inactivation of the tumor suppressor phosphatase and tensin homolog (PTEN), correlate with the elevated expression of TF in GBM cell lines and patient samples (1922).

To determine whether hypoxia can modulate the hypercoagulative activity of glioma cells, as well as TF signaling-associated elements, such as PAR receptors, the present study examined two cell lines derived from C6 rat glioma cells that differently respond to glucocorticoid treatment. Non-tumorigenic ST1 cells undergo a dramatic tumoral-to-normal phenotypic reversion, characterized by growth inhibition in monolayer cultures (with a blockage at the G0/G1 cell cycle phase), loss of the ability to form colonies in semi-solid medium, loss of ability to induce tumor formation in nude mice, and dramatic morphological changes. By contrast, P7 cells are highly aggressive and unaffected by glucocorticoid treatment (23,24).

The results of the present study show that hypoxia upregulates TF expression, as well as PAR1 and PAR2 expression, in GBM cell lines. In addition, hypoxic stress increases the shedding of procoagulant TF-bearing microvesicles (MVs) by tumor cells. Together, the results provide a better understanding of the processes involved in the deregulation of the coagulation system in GBM, which appears to be critical for glioma progression. Thus, modulating these molecular interactions may offer additional insights for the development of more efficient therapeutic strategies against aggressive glioma.

Materials and methods

Cell culture

ST1 and P7 rat glioma cell lines were originally established by Armelin et al (23,24) from rat C6 glial cells (American Type Culture Collection, Rockville, MD, USA) by subjecting cultures to successive passages of serum-free medium. Both cell lines were grown at 37°C in a humidified, 5% CO2 atmosphere in culture flasks by subconfluent passages in Dulbecco's modified Eagle medium/F12 (DMEM/F12; Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA) supplemented with 2 g/l HEPES, 60 mg/l streptomycin and 1.2 g/l sodium bicarbonate. For hypoxia experiments, cells were cultured in fresh medium containing 250 or 500 µM CoCl2 (Sigma-Aldrich, St. Louis, MO, USA) for 4, 12 or 24 h.

MV purification from cell culture supernatants

Cell culture supernatants were consecutively centrifuged at 800 × g for 10 min and at 20,000 × g for 20 min, both at 4°C. The final pellet was then washed once in phosphate-buffered saline (PBS), resuspended in PBS and stored at −80°C until utilization. MVs were quantified by counting in a FACSCalibur Flow Cytometer (BD Biosciences).

In vitro activation of plasma coagulation

The procoagulant activity of cells and MVs was measured by performing a clotting assay employing platelet-poor plasma (PPP) from rats. Cells or MVs (50 µl) resuspended in PBS at different concentrations were added to 50 µM PPP containing 3.8% sodium citrate (1:9 v/v dilution). After 1 min incubation at 37°C, 100 µM of 6.25 mM CaCl2 was added and the clotting times were recorded using a KC-4 coagulometer (Sigma Amelung, Lemgo, Germany).

Reverse transcription-quantitative polymerase chain reaction (RT-qPCR)

RNA was isolated from P7 or ST1 cells (2.5×105) using the TRIzol reagent (Invitrogen; Thermo Fisher Scientific, Inc.) and reverse transcribed into cDNA using SuperScript III Reverse Transcriptase (Invitrogen; Thermo Fisher Scientific, Inc.), according to the manufacturer's instructions. mRNA expression levels were quantified by qPCR on a 7300 Real-Time PCR System (Applied Biosystems; Thermo Fisher Scientific, Inc.) using SYBR Green Master Mix. Sequence-specific primers were designed using Primer Express software (version 3; Applied Biosystems; Thermo Fisher Scientific, Inc.). The qPCR primers were as follows: Forward, 5′-CAGAGCAGGACAGAAAAGGAAGAA-3 and reverse, 5′-GCGTCAGCCTCCTCGTCTAT-3′ for rat TF; forward, 5′-AACTGCTAGCCTCTGGATTTGATG and reverse, 5′-AAAGACAAGGCAACCGATACTTC-3′ for rat PTEN; forward 5′-TGTGCGGGCTGCTGCAATGAT-3′ and reverse 5′-TGTGCTGGCTTTGGTGAGGTTTGA-3′ for rat vascular endothelial growth factor (VEGF); forward, 5′-CCTGTGCGGTCCTTTGCT-3′ and reverse, 5′-CATCCTCTCAGATTCTGGCTGTCT-3′ for rat PAR1; forward, 5′-AGAGGTATTGGGTCATGTG-3′ and reverse, 5′-GCAGGAATGAACATGGTCTG-3′ for rat PAR2; forward, 5′-GCTGAAGATTTGGAAAGGTGT-3′ and reverse, 5′-GCTGAAGATTTGGAAAGGTGT-3′ for the control, rat HPRT. Gene expression levels were analyzed using the software provided by the PCR system's manufacturer. The ΔΔCq method (25) was used to quantify the amplification-fold difference between P7 and ST1 cells with the Cq value of the target genes being adjusted to the housekeeping gene (HPRT). Assays were performed in triplicate with variability <0.5 Ct.

Flow cytometric analysis

For surface phosphatidylserine (PS) detection, tumor cells (100 µl at 1×106 cells/ml) were incubated for 15 min at room temperature with fluorescein isothiocyanate-conjugated Annexin V (Santa Cruz Biotechnology Inc., Santa Cruz, CA, USA) diluted to 1:100 in binding buffer (10 mM HEPES, 150 mM NaCl, 2.5 mM CaCl2). Tumor cells (100 µl at 1×106 cells/ml) were then resuspended in 100 µl PBS containing 1% BSA and incubated for 30 min at 4°C with a goat polyclonal antibody against rat TF (200 µg/ml; cat no. 4501; Sekisui Diagnostics, LLC, Lexington, MA, USA). After washing with 1% BSA in PBS to remove unbound antibody, cells were incubated with a fluorescein-conjugated anti-goat IgG (1:200, sc-2777, Santa Cruz Biotechnology Inc.). Cells were then washed again with 1% BSA in PBS and analyzed using a FACSCalibur Flow Cytometer (BD Biosciences, San Jose, CA, USA). In all cases, data were analyzed using CellQuest Pro software version 5.1.1 (BD Biosciences). Data were expressed as global geometric mean fluorescence intensity (ΔMFI), which represents the difference between the MFI of stained and the MFI of unstained samples compared with the P7 cell line

FX activation via hydrolysis of chromogenic substrates

Activation of FX by the extrinsic tenase complex (TF/FVII)awas performed in HEPES-BSA buffer [50 mM HEPES, 100 mM NaCl, 5 mM CaCl2, 1 mg/ml BSA (pH 7.5)], as follows: FVIIa (final concentration, 1 nM) was incubated with cells (5×105/ml) or MVs (5×103/ml) for 10 min at 37°C in HEPES-BSA buffer. The reaction was initiated by the addition of FX (final concentration, 100 nM) and aliquots of 25 µl were removed at different times into microplate wells containing 25 µl Tris-EDTA buffer [50 mM Tris-HCl, 150 mM NaCl, 20 mM EDTA, 1 mg/ml polyethyleneglycol-6000 (pH 7.5)]. Following the addition of 50 µl of 200 µM S-2765 (chromogenic substrate; Diapharma, West Chester, OH, USA) prepared in Tris-EDTA buffer at 37°C for 30 min, absorbance was recorded at 405 nm in 6-sec intervals using a ThermoMax Microplate Reader (Molecular Devices, LLC, Menlo Park CA, USA). Velocities [mean optical density (mOD)/min] obtained in the first minute of the reaction were used to calculate the amount of FXa formed, as compared with a standard curve using known enzyme concentrations. In order to ensure that FX activation was strictly dependent on extrinsic tenase complex assembly, negative controls were performed in the absence of FVIIa and showed no significant formation of FXa.

Prothrombin activation via hydrolysis of chromogenic substrates

Activation of prothrombin by the prothrombinase complex [FXa/factor Va (FVa)] was performed in HEPES-BSA buffer using a discontinuous assay. FXa (final concentration, 10 pM) was incubated with FVa (final concentration, 1 nM; Haematological Technologies Inc., Essex Junction, VT, USA) in the presence of cells (5×105/ml) for 2 min at 37°C. The reaction was initiated by addition of prothrombin (final concentration, 500 nM; Haematological Technologies Inc.) and aliquots of 10 µl were removed every 1 min into microplate wells containing 40 µl Tris-EDTA buffer. Following the addition of 50 µl of 200 µM S-2238 (Diapharma) prepared in Tris-EDTA buffer at 37°C for 20 min, absorbance was recorded at 405 nm in 6-sec intervals using a ThermoMax Microplate Reader (Molecular Devices, LLC). Velocities (mOD/min) obtained in the first minute of the reaction were used to calculate the amount of thrombin formed, as compared with a standard curve using known enzyme concentrations. In order to ensure that prothrombin activation was strictly dependent on prothrombinase complex assembly, negative controls were performed in the absence of FXa/FVa and showed no significant formation of thrombin.

Statistical analysis

One-way analysis of variance followed by Bonferroni's multiple comparison test or unpaired t-test were performed using GraphPad Prism software (version 5; GraphPad Software, Inc., San Diego, CA, USA). Data are presented as the mean ± standard deviation of three independent experiments. The differences were considered to be statistically significant at P<0.05.

Results

Differential expression of TF in rat GBM cell lines

Previous studies have demonstrated that TF expression is correlated with the histological grade of malignancies in glioma (26,27). Using a one-stage clotting assay, the present study observed that P7 cells display potent procoagulant activity when compared with ST1 cells (Fig. 1A). As TF significantly contributes to the activation of plasma coagulation, the levels of TF expression were also determined in both GBM cell lines. Analysis by RT-qPCR demonstrated that P7 cells express significantly higher levels of TF mRNA than ST1 cells (Fig. 1B). In vitro studies previously suggested that upregulation of TF in high-grade glioma is associated with loss of tumor suppressor PTEN (19,21,22). Consistent with this hypothesis, Fig. 1B shows that the levels of PTEN mRNA are inversely correlated with TF expression in both P7 and ST1 cells. In addition, flow cytometric analysis showed less pronounced levels of TF antigen on the surface of ST1 cells compared with P7 cells. The ST1 cell line presented an ~3.5-fold increase in ΔMFI (Fig. 1C). Finally, TF activity was analyzed in the P7 and ST1 cells. Notably, P7 cells supported the assembly of extrinsic tenase complex with significant FX activation by FVIIa compared with the control cells, whereas no significant FXa generation was observed for ST1 cells under the same conditions (Fig. 1D).

Our previous studies have demonstrated that viable tumor cells may expose the anionic phospholipid PS on the outer leaflet of the cell membrane (28,29). This exposure is required for the assembly of membrane-dependent procoagulant complexes. Therefore, the present study investigated whether differential PS exposure could contribute to the variable procoagulant properties of P7 and ST1 cells. Flow cytometric analysis employing Annexin V indicated that P7 cells presented an ~1.7-fold increase in global geometric ΔMFI compared with ST1 cells (Fig. 2A). Furthermore, the assembly of the prothrombinase complex (FVa/FXa/prothrombin), a process that is critically dependent on the presence of PS-rich anionic membranes, was investigated. Consistently, P7 cells supported prothrombin activation more efficiently than ST1 cells (Fig. 2B). Controls performed in the absence of FXa and FVa showed negligible thrombin formation.

The results indicate that the higher procoagulant activity of P7 cells relative to ST1 appears to be particularly associated with their expression of the primary clotting initiator, TF. However, distinct patterns of PS exposure may also augment these discrepancies.

Hypoxic stress upregulates TF expression and TF-FVIIa activity

In GBM, intratumoral activation of the coagulation system is directly associated with vascular occlusions, triggering interlinked processes, such as hypoxia, necrosis and angiogenesis (17,30). It is well established that hypoxia induces the expression of angiogenic regulators, such as VEGF, in GBM cells, promoting the formation of new vessels and driving rapid tumor growth. Fig. 3A reveals upregulation of VEGF mRNA expression in P7 and ST1 cells upon exposure to CoCl2 (250 or 500 µM CoCl2 for 4 h), which is known to induce hypoxic-like stress. Next, the present study investigated whether treatment with CoCl2 could upregulate TF expression in cancer cells. P7 and ST1 cells were exposed to CoCl2, and TF mRNA and protein levels were determined by RT-qPCR and flow cytometry, respectively. In comparison to normoxic conditions, the cultivation of P7 cells in 500 µM CoCl2 for 4 h significantly increased TF mRNA levels; however, this change was not observed in ST1 cells (Fig. 3B). Accordingly, flow cytometry showed a time-dependent upregulation of TF levels in P7 cells upon treatment with CoCl2 (250 or 500 μM), with a peak at 12 h (data not shown). To evaluate whether hypoxia-induced upregulation of TF expression is accompanied by an increase in TF procoagulant activity, an FXa generation assay by the extrinsic tenase complex was performed. P7 and ST1 cells were cultured under normoxic and hypoxic conditions (250 or 500 µM CoCl2) for 12 h. The FX activation supported by P7 cells was enhanced at both CoCl2 concentrations (Fig. 3C). However, no increase in TF activity was observed for stimulated ST1 cells. The results of the present study indicate that hypoxia upregulates functionally active TF antigen on the surface of P7 cells, but not on ST1 cells.

Shedding of procoagulant MVs by P7 and ST1 cells

TF has been shown to circulate in the blood as a component of cell-derived MVs. Increased secretion of TF-positive MVs has been observed in several types of cancer and has been associated with the activation of the hemostatic system (7,31). Therefore, the present study analyzed the spontaneous shedding of MVs by P7 and ST1 cell lines in vitro. Differential centrifugation was performed to isolate MVs from the supernatants of P7 and ST1 cell cultures. Flow cytometric analysis revealed a similar rate of MV production for both cell lines (Fig. 4A). Using a one-stage clotting assay, it was observed that MVs derived from P7 and ST1 cells shortened the coagulation time of rat plasma at similar concentration ranges (Fig. 4B). TF activity was also investigated in these MVs. Comparably to their parental cells, P7 MVs supported FX activation by the extrinsic tenase complex (TF/FVIIa), while ST1 MVs showed no TF activity (Fig. 4C). No significant FXa generation was observed in the absence of FVIIa.

Hypoxia has been shown to markedly increase TF-containing extracellular vesicle secretion in glioma cells (32). Therefore, the present study investigated whether hypoxia increases the secretion of MVs into the conditioned media of P7 and ST1 cell cultures. The incubation of both cell lines with 250 or 500 µM CoCl2 for 24 h promoted a significant enhancement in MV secretion when compared with normoxic conditions (Fig. 4D).

Taken together, these results demonstrate that P7 cells and TF-bearing MVs exhibit higher TF activity than ST1 cells, a process that is significantly enhanced upon exposure to hypoxia.

Hypoxic induction of PAR expression in P7 and ST1 cells

In addition to TF, hypoxic stress has been shown to modulate PAR1 expression in tumor cell lines (32,33). Furthermore, our recent study observed increased PAR1 and PAR2 staining around necrotic areas in samples from patients with GBM (22). In this context, the present study analyzed the effect of CoCl2 treatment on PAR expression in rat glioma cells. RT-qPCR showed that PAR1 and PAR2 mRNA are expressed in P7 and ST1 cells, and that P7 cells express higher mRNA levels of both receptors when compared with ST1 cells (Fig. 5A). Following treatment with 250 or 500 µM CoCl2 for 4 h, mRNA expression of PAR1 was significantly upregulated in both cell lines, whereas an increase in PAR2 expression was observed only in P7 cells (Fig. 5B). Conversely, ST1 cells exhibited significantly reduced levels of PAR2 mRNA when treated with 500 µM CoCl2.

Discussion

A dramatic shift in biological behavior occurs following the transition from astrocytoma to GBM. These changes include accelerated growth and rapid progression to mortality. The mechanisms responsible for this abrupt onset of rapid growth are still being defined but are likely associated with the development of necrosis and intense angiogenesis, which are two defining features of GBM histology and powerful predictors of poor prognosis (15,34). Almost all GBM specimens show microscopic intravascular thrombosis within tissue specimens and this event has been documented as an additional distinguishing pathological feature of GBM compared with lower grade astrocytoma (35). Furthermore, necrosis and subsequent hypoxia-induced angiogenesis may be initiated or propagated by intravascular thrombosis.

In the present study, it was demonstrated that hypoxia, a key event in GBM, induces increased expression of the potent procoagulant TF in GBM cells and promotes TF/FVIIa activity. This phenomenon markedly differs between the GBM cell lines P7, a highly aggressive, glucocorticoid-resistant cell line, and ST1, a non-tumorigenic, glucocorticoid-sensitive cell line. Under normoxia, P7 exhibits ~3.5-fold higher expression of TF as compared with ST1, with strong significant inverse correlation of PTEN expression. This reflects the ability of these cell lines to promote plasma clotting and FX activation. Notably, hypoxic stress promoted by treatment with CoCl2 significantly enhanced TF expression and activity in the P7 cell line.

In addition to TF, the present study also demonstrated the presence of the procoagulant phospholipid PS on the outer leaflet of P7 and ST1 cells, which supports FX activation through the assembly of the prothrombinase complex, leading to rapid thrombin generation. Both cell lines exhibit PS exposure, with the lipid exposure more prominent in the aggressive cell line, P7. Notably, Blanco et al recently demonstrated that increased PS exposure occurs in vivo in a xenograft mice model (36). Therefore, PS is proposed as a target for the delivery of nanovesicles in the treatment and imaging of GBM.

Previous studies have demonstrated that hypoxia elevates the release of MVs by tumor cells (31,37). Herein, it was demonstrated that hypoxic stress significantly increases the production of MVs from P7 or ST1 cells. The increased incidence of venous thrombosis throughout the course of GBM has been well documented (38). In fact, the elevated levels of circulating MVs in cancer patients has been correlated with thrombosis in a number of cancer types (9,11,30). However, it has previously been demonstrated that elevated levels of TF-positive MVs do not correlate with thrombosis occurrence in patients with GBM (39). Additionally, intratumoral expression of TF has not been correlated with thrombosis in patients with GBM (40). However, increased MV production upon the induction of hypoxic stress has been associated with TF transfer from tumor to endothelial cells in a GBM model. This process triggers a proangiogenic cascade dependent on PAR2 signaling in endothelial cells that in turn results in an increase in PAR2 expression upon hypoxia (32). In addition, TF-containing MVs may promote TF transfer between tumor cell lines with different aggressive behaviors, thus, modulating their procoagulant properties (41).

Several lines of evidence suggest that PAR1 has a significant role in GBM progression. For example, Zhang et al observed elevated PAR1 expression in GBM and identified a correlation with patient survival (42). In addition, our previous study demonstrated that PAR1 expression was significantly enhanced in patients with GBM compared with those exhibiting lower grade gliomas, and PAR1 expression was positively correlated with VEGF expression (22). PAR1 expression has also been documented in a rat glioma model (43). Herein, it was demonstrated that hypoxic stress upregulates PAR1 expression in P7 and ST1 cell lines. Notably, the aggressive cell line, P7, exhibited a significantly higher degree of PAR1 expression under normoxia than the ST1 cell line. Therefore, PAR1 may contribute to GBM progression and may have an important role in hypoxia-driven events.

In addition to PAR1, PAR2 has been associated with GBM progression. In vitro assays demonstrate that PAR2 activation mediates several pro-tumoral responses in GBM cell lines (2024,2646). Therefore, blocking the TF/PAR2 signaling axis has been identified as a feasible target for the treatment of GBM (45,47,48). As observed with PAR1, the P7 cell line expressed higher levels of PAR2 than ST1 under normoxia in the present study. Notably, hypoxic stress significantly enhanced PAR2 expression in P7 but not ST1 cells. Thus, PAR2 signaling may be involved in hypoxia-driven progression of patients with GBM, resulting in a more aggressive pattern.

In conclusion, the present study demonstrates that hypoxia enhances TF expression and MVs generation in rat GBM cell lines. In addition, hypoxic stress upregulates PAR1 and PAR2 expression, a process that was highly evident in a glucocorticoid resistant cell line. The results indicate that therapies targeted against TF-PAR signaling axis may be relevant in GBM, particularly those presenting with more aggressive behavior.

References

1 

Williams JC and Mackman N: Tissue factor in health and disease. Front Biosci. 1:358–372. 2012. View Article : Google Scholar

2 

Østerud B and Bjørklid E: Sources of tissue factor. Semin Thromb Hemost. 32:11–23. 2006. View Article : Google Scholar : PubMed/NCBI

3 

Francischetti IM, Seydel KB and Monteiro RQ: Blood coagulation, inflammation, and malaria. Microcirculation. 15:81–107. 2008. View Article : Google Scholar : PubMed/NCBI

4 

Brat DJ and Van Meir EG: Vaso-occlusive and prothrombotic mechanisms associated with tumor hypoxia, necrosis, and accelerated growth in glioblastoma. Lab Invest. 84:397–405. 2004. View Article : Google Scholar : PubMed/NCBI

5 

Rak J, Milsom C, Magnus N and Yu J: Tissue factor in tumour progression. Best Pract Res Clin Haematol. 22:71–83. 2009. View Article : Google Scholar : PubMed/NCBI

6 

Kasthuri RS, Taubman MB and Mackman N: Role of tissue factor in cancer. J Clin Oncol. 27:4834–4838. 2009. View Article : Google Scholar : PubMed/NCBI

7 

Ruf W, Disse J, Carneiro-Lobo TC, Yokota N and Schaffner F: Tissue factor and cell signalling in cancer progression and thrombosis. J Thromb Haemost. 9(Suppl 1): S306–S315. 2011. View Article : Google Scholar

8 

van den Berg YW, Osanto S, Reitsma PH and Versteeg HH: The relationship between tissue factor and cancer progression: Insights from bench and bedside. Blood. 119:924–932. 2012. View Article : Google Scholar : PubMed/NCBI

9 

Lima LG and Monteiro RQ: Activation of blood coagulation in cancer: Implications for tumour progression. Biosci Rep. 33:e000642013. View Article : Google Scholar : PubMed/NCBI

10 

Lima LG, Oliveira AS, Campos LC, Bonamino M, Chammas R, Werneck C, Vicente CP, Barcinski MA, Petersen LC and Monteiro RQ: Malignant transformation in melanocytes is associated with increased production of procoagulant microvesicles. Thromb Haemost. 106:712–723. 2011. View Article : Google Scholar : PubMed/NCBI

11 

Zhou L, Qi XL, Xu MX, Mao Y, Liu ML and Song HM: Microparticles: New light shed on the understanding of venous thromboembolism. Acta Pharmacol Sin. 35:1103–1110. 2014. View Article : Google Scholar : PubMed/NCBI

12 

Boire A, Covic L, Agarwal A, Jacques S, Sherifi S and Kuliopulos A: PAR1 is a matrix metalloprotease-1 receptor that promotes invasion and tumorigenesis of breast cancer cells. Cell. 120:303–313. 2005. View Article : Google Scholar : PubMed/NCBI

13 

Albrektsen T, Sørensen BB, Hjortø GM, Fleckner J, Rao LV and Petersen LC: Transcriptional program induced by factor VIIa-tissue factor, PAR1 and PAR2 in MDA-MB-231 cells. J Thromb Haemost. 5:1588–1597. 2007. View Article : Google Scholar : PubMed/NCBI

14 

Wen PY and Kerasi S: Malignant gliomas in adults. N Engl J Med. 359:492–507. 2008. View Article : Google Scholar : PubMed/NCBI

15 

Evans SM, Judy KD, Dunphy I, Jenkins WT, Nelson PT, Lustig RA, Jenkins K, Magarelli DP, Hahn SM, Collins RA, et al: Hypoxia is important in the biology and aggression of human glial brain tumors. Clin Cancer Res. 10:8177–8184. 2004. View Article : Google Scholar : PubMed/NCBI

16 

Brown JM and Wilson WR: Exploiting tumour hypoxia in cancer treatment. Nat Rev Cancer. 4:437–447. 2004. View Article : Google Scholar : PubMed/NCBI

17 

Anand M and Brat DJ: Oncogenic regulation of tissue factor and thrombosis in cancer. Thromb Res. 129(Suppl 1): S46–S49. 2012. View Article : Google Scholar : PubMed/NCBI

18 

Magnus N, D'Asti E, Meehan B, Garnier D and Rak J: Oncogenes and the coagulation system-forces that modulate dormant and aggressive states in cancer. Thromb Res. 133(Suppl 2): S1–S9. 2014. View Article : Google Scholar : PubMed/NCBI

19 

Rong Y, Post DE, Pieper RO, Durden DL, Van Meir EG and Brat DJ: PTEN and hypoxia regulate tissue factor expression and plasma coagulation by glioblastoma. Cancer Res. 65:1406–1413. 2005. View Article : Google Scholar : PubMed/NCBI

20 

Magnus N, Garnier D and Rak J: Oncogenic epidermal growth factor receptor up-regulates multiple elements of the tissue factor signaling pathway in human glioma cells. Blood. 116:815–818. 2010. View Article : Google Scholar : PubMed/NCBI

21 

D'Asti E, Fang Y and Rak J: Brain neoplasms and coagulation-lessons from heterogeneity. Rambam Maimonides Med J. 5:e00302014. View Article : Google Scholar : PubMed/NCBI

22 

Carneiro-Lobo TC, Lima MT, Mariano-Oliveira A, Dutra-Oliveira A, Oba-Shinjo SM, Marie SK, Sogayar MC and Monteiro RQ: Expression of tissue factor signaling pathway elements correlates with the production of vascular endothelial growyh factor and interleukin-8 in human astrocytoma patients. Oncol Rep. 31:679–686. 2014.PubMed/NCBI

23 

Armelin MC and Armelin HÀ: Glucocorticoid hormone modulation of both cell surface and cytoskeleton related to growth control of rat glioma cells. J Cell Biol. 97:459–465. 1983. View Article : Google Scholar : PubMed/NCBI

24 

Armelin MC, Stocco RC and Armelin HA: Control of rat C6 glioma cell proliferation: Uncoupling of the inhibitory effects of hydrocortisone hormone in suspension and monolayer cultures. J Cell Biol. 97:455–458. 1983. View Article : Google Scholar : PubMed/NCBI

25 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2-ΔΔCt. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

26 

Hamada K, Kuratsu J, Saitoh Y, Takeshima H, Nishi T and Ushio Y: Expression of tissue factor correlates with grade of malignancy in human glioma. Cancer. 77:1877–1883. 1996. View Article : Google Scholar : PubMed/NCBI

27 

Guan M, Jin J, Su B, Liu WW and Lu Y: Tissue factor expression and angiogenesis in human glioma. Clin Biochem. 35:321–325. 2002. View Article : Google Scholar : PubMed/NCBI

28 

Fernandes RS, Kirszberg C, Rumjanek VM and Monteiro RQ: On the molecular mechanisms for the highly procoagulant pattern of C6 glioma cells. J Thromb Haemost. 4:1546–1552. 2006. View Article : Google Scholar : PubMed/NCBI

29 

Kirszberg C, Lima LG, Da Silva de Oliveira A, Pickering W, Gray E, Barrowcliffe TW, Rumjanek VM and Monteiro RQ: Simultaneous tissue factor expression and phosphatidylserine exposure account for the highly procoagulant pattern of melanoma cell lines. Melanoma Res. 19:301–308. 2009. View Article : Google Scholar : PubMed/NCBI

30 

Brat DJ, Castellano-Sanchez AA, Hunter SB, Pecot M, Cohen C, Hammond EH, Devi SN, Kaur B and Van Meir EG: Pseudopalisades in glioblastoma are hypoxic, express extracellular matrix proteases and are formed by an actively migrating cell population. Cancer Res. 64:920–927. 2004. View Article : Google Scholar : PubMed/NCBI

31 

Rautou PE and Mackman N: Microvesicles as risk markers for venous thrombosis. Expert Rev Hematol. 6:91–101. 2013. View Article : Google Scholar : PubMed/NCBI

32 

Svensson KJ, Kucharzewska P, Christianson HC, Sköld S, Löfstedt T, Johansson MC, Mörgelin M, Bengzon J, Ruf W and Belting M: Hypoxia triggers a proangiogenic pathway involving cancer cell microvesicles and PAR-2-mediated heparin-binding EGF signaling in endothelial cells. Proc Natl Acad Sci USA. 108:13147–13152. 2011. View Article : Google Scholar : PubMed/NCBI

33 

Naldini A, Filippi I, Ardinghi C, Silini A, Giavazzi R and Carraro F: Identification of a functional role for the protease-activated receptor-1 in hypoxic breast cancer cells. Eur J Cancer. 45:454–460. 2009. View Article : Google Scholar : PubMed/NCBI

34 

Hammoud MA, Sawaya R, Shi W, Thall PF and Leeds NE: Prognostic significance of preoperative MRI scans in glioblastoma multiforme. J Neurooncol. 27:65–73. 1996. View Article : Google Scholar : PubMed/NCBI

35 

Tehrani M, Friedman TM, Olson JJ and Brat DJ: Intravascular thrombosis in central nervous system malignancies: A potential role in astrocytoma progression to glioblastoma. Brain Pathol. 18:164–171. 2008. View Article : Google Scholar : PubMed/NCBI

36 

Blanco VM, Chu Z, Vallabhapurapu SD, Sulaiman MK, Kendler A, Rixe O, Warnick RE, Franco RS and Qi X: Phosphatidylserine-selective targeting and anticancer effects of SapC-DOPS nanovesicles on brain tumors. Oncotarget. 5:7105–7118. 2014. View Article : Google Scholar : PubMed/NCBI

37 

Wang T, Gilkes DM, Takano N, Xiang L, Luo W, Bishop CJ, Chaturvedi P, Green JJ and Semenza GL: Hypoxia-inducible factors and RAB22A mediate formation of microvesicles that stimulate breast cancer invasion and metastasis. Proc Natl Acad Sci USA. 111:E3234–E3242. 2014. View Article : Google Scholar : PubMed/NCBI

38 

Marras LC, Geerts WH and Perry JR: The risk of venous thromboembolism is increased throughout the course of malignant glioma: An evidence-based review. Cancer. 89:640–664. 2000. View Article : Google Scholar : PubMed/NCBI

39 

Thaler J, Ay C, Mackman N, Bertina RM, Kaider A, Marosi C, Key NS, Barcel DA, Scheithauer W, Kornek G, et al: Microparticle-associated tissue factor activity, venous thromboembolism and mortality in pancreatic, gastric, colorectal and brain cancer patients. J Thromb Haemost. 10:1363–1370. 2012. View Article : Google Scholar : PubMed/NCBI

40 

Thaler J, Preusser M, Ay C, Kaider A, Marosi C, Zielinski C, Pabinger I and Hainfellner JA: Intratumoral tissue factor expression and risk of venous thromboembolism in brain tumor patients. Thromb Res. 131:162–165. 2013. View Article : Google Scholar : PubMed/NCBI

41 

Lima LG, Leal AC, Vargas G, Porto-Carreiro I and Monteiro RQ: Intercellular transfer of tissue factor via the uptake of tumor-derived microvesicles. Thromb Res. 132:450–456. 2013. View Article : Google Scholar : PubMed/NCBI

42 

Zhang Y, Zhan H, Xu W, Yuan Z, Lu P, Zhan L and Li Q: Upregulation of matrix metalloproteinase-1 and proteinase-activated receptor-1 promotes the progression of human gliomas. Pathol Res Pract. 207:24–29. 2011. View Article : Google Scholar : PubMed/NCBI

43 

Itsekson-Hayosh Z, Shavit-Stein E, Last D, Goez D, Daniels D, Bushi D, Gera O, Zibly Z, Mardor Y, Chapman J and Harnof S: Thrombin activity and thrombin receptor in rat glioblastoma model: Possible markers and targets for intervention? J Mol Neurosci. 56:644–651. 2015. View Article : Google Scholar : PubMed/NCBI

44 

Dutra-Oliveira A, Monteiro RQ and Mariano-Oliveira A: Protease-activated receptor-2 (PAR2) mediates VEGF production through the ERK1/2 pathway in human glioblastoma cell lines. Biochem Biophys Res Commun. 421:221–227. 2012. View Article : Google Scholar : PubMed/NCBI

45 

Harter PN, Dützmann S, Drott U, Zachskorn C, Hattingen E, Capper D, Gessler F, Senft C, Seifert V, Plate KH, et al: Anti-tissue factor (TF9-10H10) treatment reduces tumor cell invasiveness in a novel migratory glioma model. Neuropathology. 33:515–525. 2013.PubMed/NCBI

46 

Luo R, Wang X, Dong Y, Wang L and Tian C: Activation of protease-activated receptor 2 reduces glioblastoma cell apoptosis. J Biomed Sci. 21:252014. View Article : Google Scholar : PubMed/NCBI

47 

Carneiro-Lobo TC, Konig S, Machado DE, Nasciutti LE, Forni MF, Francischetti IM, Sogayar MC and Monteiro RQ: Ixolaris, a tissue factor inhibitor, blocks primary tumor growth and angiogenesis in a glioblastoma model. J Thromb Haemost. 7:1855–1864. 2009. View Article : Google Scholar : PubMed/NCBI

48 

Carneiro-Lobo TC, Schaffner F, Disse J, Ostergaard H, Francischetti IM, Monteiro RQ and Ruf W: The tick-derived inhibitor Ixolaris prevents tissue factor signaling on tumor cells. J Thromb Haemost. 10:1849–1858. 2012. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

July-2016
Volume 12 Issue 1

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Monteiro RQ, Lima LG, Gonçalves NP, De Souza MR, Leal AC, Demasi MA, Sogayar MC and Carneiro‑Lobo TC: Hypoxia regulates the expression of tissue factor pathway signaling elements in a rat glioma model. Oncol Lett 12: 315-322, 2016
APA
Monteiro, R.Q., Lima, L.G., Gonçalves, N.P., De Souza, M.R., Leal, A.C., Demasi, M.A. ... Carneiro‑Lobo, T.C. (2016). Hypoxia regulates the expression of tissue factor pathway signaling elements in a rat glioma model. Oncology Letters, 12, 315-322. https://doi.org/10.3892/ol.2016.4593
MLA
Monteiro, R. Q., Lima, L. G., Gonçalves, N. P., De Souza, M. R., Leal, A. C., Demasi, M. A., Sogayar, M. C., Carneiro‑Lobo, T. C."Hypoxia regulates the expression of tissue factor pathway signaling elements in a rat glioma model". Oncology Letters 12.1 (2016): 315-322.
Chicago
Monteiro, R. Q., Lima, L. G., Gonçalves, N. P., De Souza, M. R., Leal, A. C., Demasi, M. A., Sogayar, M. C., Carneiro‑Lobo, T. C."Hypoxia regulates the expression of tissue factor pathway signaling elements in a rat glioma model". Oncology Letters 12, no. 1 (2016): 315-322. https://doi.org/10.3892/ol.2016.4593