Suppression of TAFI by siRNA inhibits invasion and migration of breast cancer cells

  • Authors:
    • Chao Yu
    • Yun Luan
    • Zejun Wang
    • Jingjie Zhao
    • Chengwei Xu
  • View Affiliations

  • Published online on: July 18, 2017     https://doi.org/10.3892/mmr.2017.7031
  • Pages: 3469-3474
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Human thrombin activatable fibrinolysis inhibitor (TAFI), also known as carboxypeptidase B2 (CPB2), is a procarboxypeptidase enzyme. The purpose of the present study was to observe the expression of TAFI in breast cancer (BC) and breast cancer cell (BCC) lines and to investigate the effect of TAFI suppression by small interfering (si)RNA gene silencing on invasion and migration of BCC lines. A significant increase in TAFI level was identified by immunohistochemical analysis in BC tissues compared with normal breast tissues. TAFI suppression also inhibited cell viability, invasion and migration ability as demonstrated by MTT, Transwell chamber, and wound scratch assays, respectively (P<0.05). The data suggested that suppression of TAFI by siRNA inhibits invasion and migration of breast cancer cells and that TAFI may be a new target for breast cancer therapy.

Introduction

Thrombin-activatable fibrinolysis inhibitor (TAFI) is a plasma zymogen that functions as a molecular link between coagulation and fibrinolysis. Numerous single nucleotide polymorphisms have been identified in carboxypeptidase basic (CPB2), the gene encoding TAFI, and are located in the 5′-flanking region, in the coding sequences and in the 3′-untranslated region of the CPB2 mRNA transcript (1). It has been suggested that CBB2 serves an important role in the interactions among procoagulant, anticoagulant and fibrinolytic systems (24). Activated TAFI (TAFIa) attenuates fibrinolysis by removing the carboxyl-terminal lysine residues from partially degraded fibrin that mediate positive feedback in plasmin generation (5). Pancreatic carboxypeptidase B (CPB) is a stable protease exhibiting high homology with TAFI but not to TAFIa (6).

Activation of TAFI by thrombin is increased 1,250-fold in the presence of the endothelial cell membrane protein thrombomodulin (TM) (7), which is expressed in tumors and is a prognostic factor in human cancer (8,9). Several studies have demonstrated that higher TAFI levels are associated with various types of cancer (1013), and plasma levels of TAFI are significantly increased in breast and lung cancer, gastric carcinoma and multiple myeloma compared with healthy individuals, suggesting that TAFI may serve a role in the pathogenesis of thrombotic disorders in cancer patients (14,15). However, the role of TAFI in tumor development remains to be fully elucidated. In the present study, the TAFI gene was knocked down by small interfering (si)RNA transfection to suppress the expression of TAFI, and the effects of the TAFI signal pathway on invasion and migration were investigated in a breast cancer cell (BCC) line.

Materials and methods

Patients and samples

The present study was approved by the ethics committee of The Second Hospital of Shandong University (Jinan, China) and informed consent was obtained from the participants. All eligible specimens were collected from patients with pathologically and clinically confirmed breast cancer who underwent surgical resection prior to any therapy from June 2013 to December 2016. All samples were re-evaluated by pathologists to confirm the diagnosis and to estimate the tumor cell content. Patients were aged from 25 to 65.

Cell culture

The human BCC lines MDA-MB-231 and MCF-7 were purchased from the American Type Cell Culture Collection (Manassas, VA, USA). The cells were cultured in Dulbecco's modified Eagle's medium with 10% heat-inactivated newborn calf serum (Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA), 5 mmol/l 4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid (Sigma-Aldrich; Merck KGaA, Darmstadt, Germany), 100 U/ml penicillin, and 100 U/ml streptomycin in 5% atmospheric CO2 at 37°C.

Immunohistochemistry

Immunohistochemistry was performed to analyse TAFI expression in breast cancer (BC) tissues and adjacent normal tissues. In brief, human breast tissues in healthy and cancer patients were fixed in 10% formaldehyde, and then were embedded in paraffin. The sections were then deparaffinized with xylene, rehydrated and then treated with 3% hydrogen peroxide to quench the endogenous peroxidase activity. Subsequent antigen retrieval was performed by heating in citrate buffer solution (0.01 M) using a microwave oven. Sections were cut at 5 µm and stained with a TAFI antibody at 4°C overnight (catalog no. sc-67300; 1:100; Santa Cruz Biotechnology, Inc., Dallas, TX, USA) following the manufacturer's protocol. and then sections were further incubated with 3,3-diaminobenzidine tetrahydrochloride for 5 min at room temperature. The sections were observed and captured with a Nikon Eclipse 90i microscope (Nikon Corporation, Tokyo, Japan).

siRNA transfection

According to the principles of siRNA design, 3×19 bp sequences in TAFI cDNA, (GenBank accession number NM-001872.3) were identified as target sites using a web-based online software system (Thermo Fisher Scientific, Inc.) for computing highly effective siRNA (https://www.thermofisher.com/order/genome-database/details/sirna/104006?CID=&ICID=uc-sirna-TAFI). The sequences were submitted to BLAST® (National Centre for Biotechnology Information, Bethesda, MD, USA) to ensure that only the selected gene was targeted. The target sequences for the TAFI gene were: siRNA-1: 5′AGU UAU AUG GCC UAU GAA CC-3′ (sense strand) and 5′-UGGUUCAUAGGCCAUAUAACU-3′ (antisense strand), siRNA-2: 5′-UGAUUGUUCGCAUAGAAAGAA-3′ (sense strand) and 5′-UUCUUUCUAUGCGAACAAUCA-3′ (antisense strand) and siRNA-3: 5′-UAGGUAUAAGGUUUCUGAGCC-3′ (sense strand) and 5′-GGCUCAGAAACCUUAUACCUA-3′ (antisense strand). Negative control siRNA: 5′-GCGUAACGCGGGAAUUUACUU-3′ (sense strand) and 5′-GUAAAUUCCCGCGUUACGCUU-3′ (antisense strand). siRNA 3 achieved the greatest knockdown of TAFI, and therefore this was selected for further experiments. Briefly, 2 µl siRNA was diluted with 100 µl OPTi-MEM (Invitrogen; Thermo Fisher Scientific, Inc.) and 10 µl Lipofectamine 2000 (Invitrogen; Thermo Fisher Scientific, Inc.) was diluted with 100 µl OPTi-MEM for 30 min at room temperature. Dilutions were mixed together and incubated at room temperature for 20 min, and 200 µl of the transfection mixture was added to each well. Experiments were repeated three times. Then, 6 h following transfection, the medium was replaced by the common complete medium again. Transiently transfected cells were harvested at 24 and 48 h following transfection for different experiments.

Semi quantitative reverse transcription-polymerase chain reaction (RT-PCR)

Total RNA was extracted from transfected cells after 24 h using TRIzol reagent (Invitrogen; Thermo Fisher Scientific, Inc.) according to the manufacturer's protocols. The purity and concentration were determined by measuring the absorbance at 260 and 280 nm (A260/A280). Reverse transcription was performed with 1 µg of total RNA and the RevertAid First Strand cDNA Synthesis kit (Thermo Fisher Scientific, Inc.). PCR primers used were as follows: CPB2, forward 5′-AACTGCTTCACTGGCTACTA-3′ and reverse 5′-TATGCTTACAAAAATCCACA-3′ GAPDH forward 5′-CCACCCATGGCAAATTCCATGGCA-3′ and reverse 5′-TCTAGACGGCAGGTCAGGTCCACC-3′. PCR was carried out in a 20 µl reaction volume containing 50 ng of genomic DNA, 10 µmol of each primer, 200 µmmol/l of each dNTP, 4 µl5 × PCR buffer and 2 u taq DNA polymerase. Amplification was carried out with an initial denaturation step at 94°C for 5 min, followed by 35 cycles of denaturation at 94°C for 40 sec, extension at 72°C for 50 sec with a final extension at 72°C for 10 min. A densitometric scanning instrument (model GS-800; Bio-Rad Laboratories, Inc., Hercules, CA, USA) was used to quantify the bands and the relative amount of CPB2 and TAFI gene mRNA expression was estimated relative to the GAPDH mRNA detected in the same sample.

Western blot analysis

Cytoplasmic proteins of different groups were extracted using cytoplasmic extraction reagents (Beyotime Institute of Biotechnology, Haimen, China). Protein concentration in the supernatants was determined by bicinchoninic acid assay. A total of 20 µl protein was separated by 10% SDS-PAGE and transferred onto polyvinylidene fluoride membranes. The membranes were blocked in 5% skimmed milk in TBST buffer at room temperature for 1 h with gentle agitation and then incubated overnight at 4°C with mouse anti-human TAFI immunoglobulin (Ig)G antibody (catalog no. sc-67867; 1:200; Santa Cruz Biotechnology, Inc.) or rabbit anti-human GAPDH IgG antibody (catalog no. sc-25778; 1:1,000; Santa Cruz Biotechnology, Inc.). Following washing with TBST, the membranes were incubated with horseradish peroxidase-conjugated anti-human IgG antibody (catalog no. ab109489; 1:1,000; Abcam). The bound antibodies were visualized using an enhanced chemiluminescence reagent (EMD Millipore, Billerica, MA, USA) and quantified by densitometry using ChemiDoc XRS+ image analyzer (Bio-Rad Laboratories, Inc.). The expression levels of TAFI and GAPDH protein were quantified by densitometry analysis (model GS-800, Bio-Rad Laboratories, Inc.). The signal strength of each TAFI signal was normalized against the corresponding GAPDH control.

Cell viability assay

Viable cells were quantified by a standard 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) reduction assay. The tetrazolium salt MTT (Sigma-Aldrich; Merck KGaA) was used to compare the cellular metabolic capacities. Each well of a 96-well plate was seeded with 1×104 cells and preincubated in 5% atmospheric CO2 at 37°C. Then, 48 h following transfection, the MTT assay was performed by adding 20 µl of MTT (5 mg/ml; Sigma-Aldrich; Merck KGaA) for 4 h. The supernatant was removed and 150 µl DMSO added to each well. Optical density (OD) of the solution was measured at 490 nm. Triplicate experiments with triplicate samples were performed.

Cell migration assay

Wound healing assays were performed to quantify the rate of cell migration. A density of 5×105 cells/well were plated onto 24 well plates and were grown for 24 h to >90% confluence. The medium was removed and a wound line, that is a cell-free area, was created on the cell monolayers by manually scraping the cells with a plastic pipette tip. Debris was removed from the culture by washing with PBS twice and then the cells were cultured in RPMI-1640 (Sigma-Aldrich; Merck KGaA) containing 1% FBS. Wound sizes were verified using ImageJ software version 1.43 (National Institutes of Health, Bethesda, MD, USA). Cell migration=0 h wound width (1 mm)-6 h wound width. Images (original magnification, ×40) were obtained using a Nikon Eclipse 90i microscope (Nikon Corporation; Tokyo, Japan). These experiments were repeated three times.

Cell invasion assay

The cell invasion ability was evaluated using a modified Boyden chamber (BD Biosciences, San Jose, CA, USA) with a polycarbonate filter with 8 µm pores placed between the upper and lower chambers coated with 70 µl Matrigel (1 mg/ml) as previously described (16). Briefly, 1×105 cells in 0.1 ml of serum-free DMEM medium were placed in the upper chamber and the lower chamber was filled with DMEM medium containing 15% newborn calf serum. Following incubation for 24 h at 37°C in a 5% CO2 incubator, the cells on the top surface of the insert were removed by wiping with a cotton swab. Cells that migrated to the bottom surface of the insert were fixed in 100% methanol for 2 min, stained in 0.1% Crystal Violet for 20 min, rinsed in PBS and then subjected to microscopic inspection (original magnification, ×100). The magnitude of cells migration was evaluated by counting the migrated cells in 10 random high-power microscope fields. Migration activity was expressed as a percentage of the control group.

Statistical analysis

Data are presented as the mean ± standard deviation. Comparisons between treatments were analyzed by one-way analysis of variation followed by S-N-K post hoc test using SPSS software version 18.0 (SPSS, Inc., Chicago, IL, USA). P<0.05 was considered to indicate a statistically significant difference.

Results

TAFI is expressed in BC tissues

BC samples, together with their adjacent normal tissues, from 10 cases breast cancer patients were analyzed. Immunohistochemistry demonstrated that in tumor-adjacent normal tissues the expression of TAFI was relevantly low (Fig. 1A), while in BC tissues TAFIs were visibly elevated (Fig. 1B). The expression of TAFI mRNA was assessed using RT-PCR. TAFI was expressed at higher levels in BC tissues than in matched normal tissues (Fig. 1C).

siRNA-mediates CPB2 knockdown

Specific siRNA targeting CPB2 was transfected in MDA-MB-231 and MCF-7 cells for 48 h, then TAFI protein expression levels were detected. In the present study, RT-PCR detection demonstrated that siRNA blocked the expression of CPB2 gene expression, and western blot analysis confirmed that CPB2 gene silencing successfully suppressed expression of the corresponding TAFI protein compared with non-transfected group (control; P<0.05; Fig. 2A and B).

TAFI depletion reduces BCC viability

The effect of TAFI depletion on cell viability was evaluated using the MTT assay. Following treatment with siRNA for 48 h, the cell viability exhibited a significant decrease in MDA-MB-231 and MCF-7 cells (P<0.05; Fig. 3). This indicated that TAFI has an effect on the regulation of BCC viability.

Effects of RNA interference TAFI on cell migration and invasion

Next, the role of TAFI in migration and invasion of BCCs was determined using wound scratch and Transwell chamber assays, respectively. As demonstrated in Fig. 4A and B, transfection with TAFI siRNA significantly inhibited migration and invasion in MDA-MB-231 cells compared with non-transfected group (control; P<0.05). The present study revealed similar effects of TAFI inhibition on cell migration and invasion in the less metastatic MCF-7 cells compared with the more metastatic MDA-MB-231 cells (data not shown). Together these results demonstrated that knockdown of TAFI markedly inhibited BCC migration (at 6 h) and invasion (at 24 h).

Discussion

BC is one of the most frequent malignancies worldwide. There is an upward trend in incidence; the prognosis of patients with advanced BC is closely associated with metastasis with few effective treatments (17). BC initiation and progression is a complicated process which is associated with the loss of the normal regulatory pathways between cell proliferation, differentiation and apoptosis (18,19). In recent years, the incidence of BC in China has been increasing, according to the national cancer centre statistics (20), BC is the predominant female malignant tumor (21). Tumor metastasis is the basic characteristic of malignant tumors, and also the main difficulty in curing them. Previous studies have demonstrated that an abnormal increase in circulating coagulation protein conducive to thrombophilia is associated with different types of cancer (12,13). Coagulation-associated abnormality in cancer patients is an increased plasma fibrinogen level. Thrombotic and hemorrhagic complications are among the common causes of mortality in cancer patients. Impairment of coagulation may be an important early symptom of tumor progression (22). Studies have identified that cancer patients with tumor growth, angiogenesis and end-organ damage, may also experience coagulation activation (15,23).

TAFI represents the important molecular link between the coagulation and fibrinolytic pathways (24). TAFI activity is generated in the process of coagulation through binding to thrombin, thrombin-TM complex and orplasmin, which in turn cleave TAFI protein at Arg114 (residue Arg92 following removal of the signal peptide) into N-terminal activation peptide and catalytic domains, leading to exposure of the active site cleft of activated TAFIa (2527). The plasma concentration of TAFI is controlled by genetic and non-genetic factors. It is suggested that a variety of diseases are positively associated with plasma TAFI levels (28). Reports showed that (12,13) databases indicate a high expression of TAFI levels in breast, ovarian and hepatic cancer cell lines. Plasma levels of TAFI are also increased in a number of types of cancers, including BC (10,29). In addition, higher levels of TAFI have been associated with more advanced stages of cancer (30). The fibrinolytic system, more appropriately referred to as the plasminogen activator system, controls not only the intravascular fibrin deposition but is also involved in a wide variety of physiologic and pathologic processes. These components are involved in tumor growth, invasion and metastasis in cancer cells (31). The pathogenesis of the hypercoagulable state of the patients is extremely complex. However, the enhanced coagulation of tumor cells serves an important function in it. Malignant tumors are involved in the hemostasis system by several pathways. Studies have demonstrated that the cytokines which tumor cells excrete contribute to the adhesiveness of the epithelium, thus aiding metastasis (32,33). Anticoagulant drugs like acetaminophen (34) may reduce the migration of tumor cells by inhibiting coagulation. Cancer cell invasion and metastasis require the degradation of the extracellular matrix during local invasion, angiogenesis, intravasation and extravasation. Invasive tumor cells express a plasminogen activation system composed of a urokinase- and/or tissue-type plasminogen activator, plasminogen and protease receptors on their membrane surface. There is a negative correlation between the TM expression level and cell malignancy; patients with low TM expression in BC tissues have a significantly lower rate of disease-free survival compared with patients with high TM expression (35). However, soluble type TM reduced invasion and metastasis of cancer cells, indicating that TM is not only an anticoagulant protein but also a regulator of tumor invasion and metastasis, (36,37). However, the mechanisms are not completely understood and the importance of TAFI in homeostasis and fibrinolysis remains to be elucidated.

The present study inhibited TAFI expression using siRNA in BCC lines and the results demonstrated that decreasing TAFI expression may reduce cell growth, migration and invasion in two types of BCC cells. Although the mechanism of TAFI synthesis in the BCC cells remains to be elucidated, the present study may offer a novel therapeutic approach for the treatment of BC metastasis.

Acknowledgements

We are grateful to Central Research Laboratory, the Second Hospital of Shandong University for technical assistance and the generous support. This study was supported by a grant from the Youth Foundation of the Second Hospital of Shandong University (grant no. Y2014010044) and the Natural Science Foundation of Shandong Province (grant no. ZR2014HM023)..

References

1 

Boffa MB, Maret D, Hamill JD, Bastajian N, Crainich P, Jenny NS, Tang Z, Macy EM, Tracy RP, Franco RF, et al: Effect of single nucleotide polymorphisms on expression of the gene encoding thrombin-activatable fibrinolysis inhibitor: A functional analysis. Blood. 111:183–189. 2008. View Article : Google Scholar : PubMed/NCBI

2 

Bajzar L: Thrombin activatable fibrinolysis inhibitor and an antifibrinolytic pathway. Arterioscler Thromb Vasc Biol. 20:2511–2518. 2000. View Article : Google Scholar : PubMed/NCBI

3 

Redlitz A, Tan AK, Eaton DL and Plow EF: Plasma carboxypeptidases as regulators of theplasminogen system. J Clin Invest. 96:2534–2538. 1995. View Article : Google Scholar : PubMed/NCBI

4 

Wang W, Boffa MB, Bajzar L, Walker JB and Nesheim ME: A study of the mechanism of inhibition of fibrinolysis by activated thrombin-activable fibrinolysis inhibitor. J Biol Chem. 273:27176–27181. 1998. View Article : Google Scholar : PubMed/NCBI

5 

Boffa MB, Hamill JD, Bastajian N, Dillon R, Nesheim ME and Koschinsky ML: A role for CCAAT/enhancer-binding protein in hepatic expression of thrombin-activable fibrinolysis inhibitor. J Biol Chem. 277:25329–25336. 2002. View Article : Google Scholar : PubMed/NCBI

6 

Pereira PJ Barbosa, Segura-Martin S, Oliva B, Ferrer-Orta C, Avilés FX, Coll M, Gomis-Rüth FX and Vendrell J: Human procarboxypeptidase B: Three-dimensional structure and implications for thrombin-activatable fibrinolysis inhibitor (TAFI). J Mol Biol. 321:537–547. 2002. View Article : Google Scholar : PubMed/NCBI

7 

Hosaka Y, Takahashi Y and Ishii H: Thrombomodulin in human plasma contributes to inhibit fibrinolysis through acceleration of thrombin-dependent activation of plasma procarboxypeptidase B. Thromb Haemost. 79:371–377. 1998.PubMed/NCBI

8 

Tamura A, Hebisawa A, Hayashi K, Sagara Y, Fukushima K, Kurashima A, Yotsumoto H, Mori M and Komatsu H: Prognostic significance of thrombomodulin expression and vascular invasion in stage I squamous cell carcinoma of the lung. Lung Cancer. 34:375–382. 2001. View Article : Google Scholar : PubMed/NCBI

9 

Reijerkerk A, Meijers JC, Havik SR, Bouma BN, Voest EE and Gebbink MF: Tumor growth and metastasis are not affected in thrombin-activatable fibrinolysis inhibitor-deficient mice. J Thromb Haemost. 2:769–779. 2004. View Article : Google Scholar : PubMed/NCBI

10 

Fawzy MS, Mohammed EA, Ahmed AS and Fakhr-Eldeen A: Thrombin-activatable fibrinolysis inhibitor Thr325Ile polymorphism and plasma level in breast cancer: A pilot study. Meta Gene. 4:73–84. 2015. View Article : Google Scholar : PubMed/NCBI

11 

Fawzy MS and Toraih EA: Data supporting the structural and functional characterization of Thrombin-Activatable Fibrinolysis Inhibitor in breast cancer. Data Brief. 5:981–989. 2015. View Article : Google Scholar : PubMed/NCBI

12 

Yeung TL, Leung CS, Wong KK, Samimi G, Thompson MS, Liu J, Zaid TM, Ghosh S, Birrer MJ and Mok SC: TGF-β modulates ovarian cancer invasion by upregulating CAF-derived versican in the tumor microenvironment. Cancer Res. 73:5016–5028. 2013. View Article : Google Scholar : PubMed/NCBI

13 

Miller TW, Balko JM, Fox EM, Ghazoui Z, Dunbier A, Anderson H, Dowsett M, Jiang A, Smith RA, Maira SM, et al: ERα-dependent E2F transcription can mediate resistance to estrogen deprivation in human breast cancer. Cancer Discov. 1:338–351. 2011. View Article : Google Scholar : PubMed/NCBI

14 

Hataji O, Taguchi O, Gabazza EC, Yuda H, D'Alessandro-Gabazza CN, Fujimoto H, Nishii Y, Hayashi T, Suzuki K and Adachi Y: Increased circulating levels of thrombin-activatable fibrinolysis inhibitor in lung cancer patients. Am J Hematol. 76:214–219. 2004. View Article : Google Scholar : PubMed/NCBI

15 

Fidan E, Kavgaci H, Orem A, Yilmaz M, Yildiz B, Fidan S, Akcan B, Ozdemir F and Aydin F: Thrombin activatable fibrinolysis inhibitor and thrombin-antithrombin-III-complex levels in patients with gastric cancer. Tumour Biol. 33:1519–1525. 2012. View Article : Google Scholar : PubMed/NCBI

16 

Huang Z, Miao X, Luan Y, Zhu L, Kong F, Lu Q, Pernow J, Nilsson G and Li N: PAR1-stimulated platelet releasate promotes angiogenic activities of endothelial progenitor cells more potently than PAR4-stimulated platelet releasate. J Thromb Haemost. 13:465–476. 2015. View Article : Google Scholar : PubMed/NCBI

17 

Cui J, Hollmén M, Li L, Chen Y, Proulx ST, Reker D, Schneider G and Detmar M: New use of an old drug: Inhibition of breast cancer stem cells by benztropine mesylate. Oncotarget. 8:1007–1022. 2017.PubMed/NCBI

18 

Chung SS and Vadgama JV: Curcumin and epigallocatechin gallate inhibit the cancer stem cell phenotype via down-regulation of STAT3-NFκB signaling. Anticancer Res. 35:39–46. 2015.PubMed/NCBI

19 

Ramachandran C and You W: Differential sensitivity of human mammary epithelial and breast carcinoma cell lines to curcumin. Breast Cancer Res Treat. 54:269–278. 1999. View Article : Google Scholar : PubMed/NCBI

20 

Hu X, Jiang L, Li Q and Gu Y: Quantitative assessment of background parenchymal enhancement in breast magnetic resonance images predicts the risk of breast cancer. Oncotarget. 8:10620–10627. 2017.PubMed/NCBI

21 

Zhang F, Zhang Y, Deng Z, Xu P, Zhang X, Jin T and Liu Q: Genetic variants in the acylphosphatase 2 gene and the risk of breast cancer in a Han Chinese population. Oncotarget. 7:86704–86712. 2016.PubMed/NCBI

22 

Kvolik S, Jukic M, Matijevic M, Marjanovic K and Glavas-Obrovac L: An overview of coagulation disorders in cancer patients. Surg Oncol. 19:e33–e46. 2010. View Article : Google Scholar : PubMed/NCBI

23 

De Cicco M: The prothrombotic state in cancer: Pathogenic mechanisms. Crit Rev Oncol Hematol. 50:187–196. 2004. View Article : Google Scholar : PubMed/NCBI

24 

Mosnier LO and Bouma BN: Regulation of fibrinolysis by thrombin activatable fibrinolysis inhibitor, an unstable carboxypeptidase B that unites the pathways of coagulation and fibrinolysis. Arterioscler Thromb Vasc Biol. 26:2445–2453. 2006. View Article : Google Scholar : PubMed/NCBI

25 

Antovic JP and Blombäck M: Thrombin-activatable fibrinolysis inhibitor antigen and TAFI activity in patients with APC resistance caused by factor V Leiden mutation. Thromb Res. 106:59–62. 2002. View Article : Google Scholar : PubMed/NCBI

26 

Miah MF and Boffa MB: Functional analysis of mutant variants of thrombin-activatable fibrinolysis inhibitor resistant to activation by thrombin or plasmin. J Thromb Haemost. 7:665–672. 2009. View Article : Google Scholar : PubMed/NCBI

27 

Nesheim M: Thrombin and fibrinolysis. Chest. 124:(Suppl 3). 33S–39S. 2003. View Article : Google Scholar : PubMed/NCBI

28 

Boffa MB and Koschinsky ML: Curiouser and curiouser: Recent advances in measurement of thrombin-activatable fibrinolysis inhibitor (TAFI) and in understanding its molecular genetics, gene regulation, and biological roles. Clin Biochem. 40:431–442. 2007. View Article : Google Scholar : PubMed/NCBI

29 

Chengwei X, Xiaoli M, Yuan Z, Li P, Shengjiang W, Chao Y and Yunshan W: Plasma thrombin-activatable fibrinolysis inhibitor levels and its Thr325Ile polymorphism in breast cancer. Blood Coagul Fibrinolysis. 24:698–703. 2013. View Article : Google Scholar : PubMed/NCBI

30 

Balcik OS, Albayrak M, Uyar ME, Dagdas S, Yokus O, Ceran F, Cipil H, Kosar A and Ozet G: Serum thrombin activatable fibrinolysis inhibitor levels in patients with newly diagnosed multiple myeloma. Blood Coagul Fibrinolysis. 22:260–263. 2011. View Article : Google Scholar : PubMed/NCBI

31 

Higuchi T, Nakamura T, Kakutani H and Ishii H: Thrombomodulin suppresses invasiveness of HT1080 tumor cells by reducing plasminogen activation on the cell surface through activation of thrombin-activatable fibrinolysis inhibitor. Biol Pharm Bull. 32:179–185. 2009. View Article : Google Scholar : PubMed/NCBI

32 

Luo J, Lee S Ok, Liang L, Huang CK, Li L, Wen S and Chang C: Infiltrating bone marrow mesenchymal stem cells increase prostate cancer stem cell population and metastatic ability via secreting cytokines to suppress androgen receptor signaling. Oncogene. 33:2768–2778. 2014. View Article : Google Scholar : PubMed/NCBI

33 

Eichbaum C, Meyer AS, Wang N, Bischofs E, Steinborn A, Bruckner T, Brodt P, Sohn C and Eichbaum MH: Breast cancer cell-derived cytokines, macrophages and cell adhesion: Implications for metastasis. Anticancer Res. 31:3219–3227. 2011.PubMed/NCBI

34 

Hasanein P and Sharifi M: Effects of rosmarinic acid on acetaminophen-induced hepatotoxicity in male Wistar rats. Pharm Biol. 55:1809–1816. 2017. View Article : Google Scholar : PubMed/NCBI

35 

Bazzi ZA, Lanoue D, El-Youssef M, Romagnuolo R, Tubman J, Cavallo-Medved D, Porter LA and Boffa MB: Activated thrombin-activatable fibrinolysis inhibitor (TAFIa) attenuates breast cancer cell metastatic behaviors through inhibition of plasminogen activation and extracellular proteolysis. BMC Cancer. 16:3282016. View Article : Google Scholar : PubMed/NCBI

36 

McMahon B and Kwaan HC: The plasminogen activator system and cancer. Pathophysiol Haemost Thromb. 36:184–194. 2008. View Article : Google Scholar : PubMed/NCBI

37 

Vairaktaris E, Yapijakis C, Nkenke E, Vassiliou S, Vylliotis A, Nixon AM, Derka S, Ragos V, Spyridonidou S, Tsigris C, et al: The 1040C/T polymorphism influencing thermal stability and activity of thrombin activatable fibrinolysis inhibitor is associated with risk for oral cancer. Am J Hematol. 82:1010–1012. 2007. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

September-2017
Volume 16 Issue 3

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Yu C, Luan Y, Wang Z, Zhao J and Xu C: Suppression of TAFI by siRNA inhibits invasion and migration of breast cancer cells. Mol Med Rep 16: 3469-3474, 2017
APA
Yu, C., Luan, Y., Wang, Z., Zhao, J., & Xu, C. (2017). Suppression of TAFI by siRNA inhibits invasion and migration of breast cancer cells. Molecular Medicine Reports, 16, 3469-3474. https://doi.org/10.3892/mmr.2017.7031
MLA
Yu, C., Luan, Y., Wang, Z., Zhao, J., Xu, C."Suppression of TAFI by siRNA inhibits invasion and migration of breast cancer cells". Molecular Medicine Reports 16.3 (2017): 3469-3474.
Chicago
Yu, C., Luan, Y., Wang, Z., Zhao, J., Xu, C."Suppression of TAFI by siRNA inhibits invasion and migration of breast cancer cells". Molecular Medicine Reports 16, no. 3 (2017): 3469-3474. https://doi.org/10.3892/mmr.2017.7031