cDNA Cloning and mRNA Expression of a Serine Proteinase Inhibitor Secreted by Cancer Cells: Identification as Placental Protein 5 and Tissue Factor Pathway Inhibitor-2

1994 ◽  
Vol 116 (5) ◽  
pp. 939-942 ◽  
Author(s):  
Yohei Miyagi ◽  
Naohiko Koshikawa ◽  
Hidetaro Yasumitsu ◽  
Etsuko Miyagi ◽  
Fumiki Hirahara ◽  
...  
1998 ◽  
Vol 79 (01) ◽  
pp. 217-221 ◽  
Author(s):  
Koichi Kokame ◽  
Toshiyuki Miyata ◽  
Naoaki Sato ◽  
Hisao Kato

SummaryThrombotic complications are frequently associated with atherosclerosis. Lysophosphatidylcholine (LPC), a component accumulated in oxidatively modified LDL (ox-LDL), is known to play a crucial role in the initiation and progression of atherosclerotic vascular lesions. Since a vascular anticoagulant, tissue factor pathway inhibitor (TFPI), has the function of regulating the initial reaction of tissue factor (TF)-induced coagulation, we investigated the effect of LPC on TFPI synthesis in cultured human umbilical vein endothelial cells (HUVEC). The treatment of HUVEC with LPC for 24 h decreased TFPI antigen levels in both the culture medium and the cell lysate in a dose-dependent manner. Northern blot analysis revealed that LPC caused a time-dependent decrease in the TFPI mRNA levels. The levels of TFPI antigen and mRNA were decreased to 72% and 38%, respectively, by the incubation with 50 μM LPC for 24 h. The down-regulation by LPC of TFPI mRNA expression was not observed in the presence of cycloheximide, suggesting that protein synthesis was involved in the suppression of TFPI mRNA expression. The TFPI mRNA levels in actinomycin D-treated cells were relatively stable, indicating that the down-regulation of TFPI mRNA by LPC would be partly explained by the enhanced mRNA destabilization. In contrast to the significant down-regulatory effects of LPC on TFPI expression, LPC did not induce TF mRNA expression in HUVEC. These results indicate that LPC accumulated in the atherosclerotic vascular wall would suppress endothelial TFPI synthesis, reducing the antithrombotic property of endothelial cells.


1992 ◽  
Vol 111 (5) ◽  
pp. 681-687 ◽  
Author(s):  
Kei-ichi Enjyoji ◽  
Mitsuru Emi ◽  
Tsunehiro Mukai ◽  
Hisao Kato

2000 ◽  
Vol 83 (01) ◽  
pp. 141-147 ◽  
Author(s):  
Yoshiaki Kazama ◽  
Shintaro Kamei ◽  
Joseph Kuijper ◽  
Donald Foster ◽  
Walter Kisiel

SummaryTissue factor pathway inhibitor-2 (TFPI-2), also known as placental protein 5, is a 32 kDa extracellular matrix-associated serine proteinase inhibitor consisting of three tandemly-arranged Kunitz-type domains. Two overlapping genomic clones containing sequences encoding murine TFPI-2 were isolated from a A FIXII 129 SVJ mouse genomic library, and the complete nucleotide sequence of the gene was determined. The murine TFPI-2 gene spans approximately 9.3 kilobases and consists of five exons and four introns. The nucleotide sequences surrounding all the exon-intron boundaries are highly conserved and obey the GT-AG rule. Each Kunitz-type domain is encoded by a single exon, similar to that observed for other Kunitz-type proteinase inhibitors. A total of 1,577 bp of the 3’-flanking region contains a probable polyadenylation site (ATTAAA) at +5,759 and an apparent cleavage or termination site (CATTG) at +6,170. The 5’-flanking region of the murine TFPI-2 gene contains a prototypical TATA box, a GC box and two CAAT boxes. In addition, several candidate transcription factor binding sites responsible for placenta-, endothelial cell-, and smooth muscle cell-specific expression of the TFPI-2 gene were also identified.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 4165-4165
Author(s):  
Prakasha Kempaiah ◽  
Walter Kisiel

Abstract Human tissue factor pathway inhibitor-2 (TFPI-2) is a 32 kDa extracellular matrix-associated Kunitz-type serine proteinase inhibitor that regulates the plasmin and trypsin- mediated activation of zymogen matrix metalloproteinases and growth factors essential for tumor growth, invasion, and metastasis. We previously demonstrated that HT-1080 human fibrosarcoma cells do not express TFPI-2, but restoration of TFPI-2 expression in these cells by stable transfection with the human TFPI-2 cDNA markedly inhibited their growth and metastasis in-vivo. In this study, 1 μM concentrations of either recombinant human TFPI-2 or its mutated first Kunitz-type domain (R24K KD1), were offered to HT-1080 cells, incubated for 48 h, and the degree of apoptosis assessed by nuclear fragmentation, ethidium bromide/acridine orange (EB/AO) staining, fluorescence-activated cell sorting (FACS) and immunoblotting. Agarose gel electrophoresis of DNA from HT-1080 cells treated with either TFPI-2 or R24K KD1 for 48h indicated DNA fragmentation with a ladder pattern typically associated with apoptosis. EB/AO staining of TFPI-2- and R24K KD1- treated cells revealed that 40–70% of the cells were apoptotic in relation to vehicle-treated cells as judged by fluoresence microscopy. Co-administration of TFPI-2 with polymyxin B produced similar results, suggesting that apoptosis was not endotoxin-dependent. Consistent with our earlier studies showing its enhanced inhibitory activity relative to TFPI-2, R24K KD1 was able to induce apoptosis in 68% of HT-1080 cells after 48h of treatment compared to 39% for the parent TFPI-2 at an equivalent concentration. Moreover, HT-1080 cells treated with a KD1 preparation lacking the reactive site arginine residue (R24Q KD1) produced only an 18% apoptosis rate, thereby linking the observed apoptosis with serine proteinase inhibition. FACS analysis, using propidium iodide and annexin-V labeling, revealed similar apoptotic rates to that seen by EB/AO staining assays. In addition, immunoblotting experiments of vehicle and TFPI-2-treated cells indicated increased caspase-3 activation in TFPI-2-treated cells, thus providing evidence that apoptosis is caspase-mediated. We also observed up-regulation of the proapoptotic Bax protein by immunoblotting following treatment of HT-1080 cells with either TFPI-2 or R24K KD1. Taken together, our results demonstrate that treatment of HT-1080 cells with exogenous TFPI-2 or R24K KD1 activates caspase-mediated, proapoptotic signaling pathways and induces apoptosis. In addition, our data provides suggestive evidence that peritumor application of either TFPI-2 or R24K KD1 may facilitate tumor apoptosis in-vivo.


Oncogene ◽  
2005 ◽  
Vol 24 (14) ◽  
pp. 2386-2397 ◽  
Author(s):  
Federico A Steiner ◽  
Julie A Hong ◽  
Maria R Fischette ◽  
David G Beer ◽  
Zong-Sheng Guo ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document