Download The Role of Tissue Factor Pathway Inhibitor

Survey
yes no Was this document useful for you?
   Thank you for your participation!

* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project

Document related concepts
no text concepts found
Transcript
The Role of Tissue Factor Pathway Inhibitor-2
in Cancer Biology
Ewa Sierko, M.D.,1,2 Marek Z. Wojtukiewicz, M.D.,1,2 and Walter Kisiel, Ph.D.3
ABSTRACT
Tissue factor pathway inhibitor-2 (TFPI-2), a member of the Kunitz-type serine
proteinase inhibitor family, is a structural homologue of tissue factor pathway inhibitor
(TFPI). The expression of TFPI-2 in tumors is inversely related to an increasing degree of
malignancy, which may suggest a role for TFPI-2 in the maintenance of tumor stability and
inhibition of the growth of neoplasms. TFPI-2 inhibits the tissue factor/factor VIIa (TF/
VIIa) complex and a wide variety of serine proteinases including plasmin, plasma kallikrein,
factor XIa, trypsin, and chymotrypsin. Aberrant methylation of TFPI-2 promoter cytosinephosphorothioate-guanine (CpG) islands in human cancers and cancer cell lines was widely
documented to be responsible for diminished expression of mRNA encoding TFPI-2 and
decreased or inhibited synthesis of TFPI-2 protein during cancer progression. Furthermore,
an aberrantly spliced variant of TFPI-2 mRNA (designated asTFPI-2) was detected, which
represents an untranslated form of TFPI-2. The levels of asTFPI-2 were very low or
undetectable in normal cells but markedly upregulated in neoplastic tissue. TFPI-2
functions in the maintenance of the stability of the tumor environment and inhibits
invasiveness and growth of neoplasms, as well as metastases formation. TFPI-2 has also
been shown to induce apoptosis and inhibit angiogenesis, which may contribute
significantly to tumor growth inhibition. Restoration of TFPI-2 expression in tumor
tissue inhibits invasion, tumor growth, and metastasis, which creates a novel possibility of
cancer patient treatment. However, more information is still needed to define the precise
role of TFPI-2 in human tumor biology.
KEYWORDS: Coagulation inhibitors, TFPI-2, cancer
T
issue factor pathway inhibitor-2 (TFPI-2) is
a homologue of tissue factor inhibitor (TFPI)—the
principal inhibitor of tissue factor–dependent blood
coagulation.1,2
STRUCTURE AND FUNCTION OF TFPI-2
The TFPI-2 molecule consists of three tandemly
arranged Kunitz-type proteinase inhibitor domains, a
1
Department of Oncology, Medical University, Bialystok, Poland;
Comprehensive Cancer Center, Bialystok, Poland; 3Department of
Pathology, University of New Mexico Health Sciences Center, University of New Mexico, Albuquerque, New Mexico.
Address for correspondence and reprint requests: Dr. Marek Z.
Wojtukiewicz, Department of Oncology, Medical University, 12
Ogrodowa St., 15-027 Bialystok, Poland (e-mail: m.wojtukiewicz
2
negatively charged amino terminal region, and a positively charged carboxy terminal region.3 The first
Kunitz domain with arginine in its P1 reactive site is
responsible for TFPI-2 inhibitory activity.3 Recently,
other residues flanking the P1 residue, particularly at P6
and P50 , were documented to influence the inhibitory
activity and specificity of human TFPI-2.4 In 1994,
Kisiel and co-workers,5 on the basis of available data
on molecular cloning, structure, and function, revealed
@neostrada.pl).
Inhibitors of Hemostatic System in Cancer: Basic and Clinical
Aspects; Guest Editors, Marek Z. Wojtukiewicz, M.D., and Ewa
Sierko, M.D.
Semin Thromb Hemost 2007;33:653–659. Copyright # 2007 by
Thieme Medical Publishers, Inc., 333 Seventh Avenue, New York,
653
654
SEMINARS IN THROMBOSIS AND HEMOSTASIS/VOLUME 33, NUMBER 7
the identity of TFPI-2 with previously described placental protein 5 (PP5).6 Three types of TFPI-2 molecules (33, 31, and 27 kDa), resulting from differential
glycosylation, were identified in the extracellular matrix
of human dermal fibroblasts and human umbilical vein
endothelial cells (HUVECs).7–9
Despite the structural similarity of TFPI-2 and
TFPI, TFPI-2 serves as a weak inhibitor of tissue factor/
factor VIIa (TF/VIIa) complex, in contrast with TFPI.1–3
The dominant activity of TFPI-2 is directed at the
inhibition of a wide variety of serine proteinases
including plasmin,3,6,10,11 plasma kallikrein,3 trypsin
and chymotrypsin,3,6,12 as well as factor XIa.3 TFPI-2
failed to exhibit any inhibitory activity toward glandular kallikrein, urinary plasminogen activator, tissue
plasminogen activator, human activated protein C,
leukocyte elastase, and thrombin.3,11
The gene responsible for human TFPI-2 synthesis is localized on chromosome 7q22.13 The human
TFPI-2 gene spans 7 kb and consists of five exons and
four introns.14 The TFPI-2 gene promoter exhibits
features typical of a housekeeping gene.15
LOCALIZATION OF TFPI-2 UNDER NORMAL
CONDITIONS
TFPI-2 is synthesized in endothelial cells (ECs) of
different blood vessels (venous, arterial, and capillaries)
but predominately by the ECs of small blood vessels16
and is abundantly secreted (60 to 90%) into the extracellular matrix (ECM).16 Furthermore, expression of the
gene encoding TFPI-2 was detected in human ciliary
epithelium,17 mature placenta, heart, liver, kidneys, and
skeletal muscles.12 TFPI-2 mRNA was also demonstrated in syncytiotrophoblasts,18 whereas the product
of the gene was found both in syncytiotrophoblast and
cytotrophoblast.19 TFPI-2 protein was also immunohistochemically detected in seminal vesicles,20 colon, breast,
pancreas, stomach, larynx, kidney, endometrium, and
brain tissue.21 The plasma concentration of TFPI-2 in
men and nonpregnant women is very low (0.43 to 0.49
ng/mL).19 Moreover, TFPI-2 is present in seminal
plasma,22 preovulatory follicular fluid,23 and in the
mucus of the uterus.19
TFPI-2 PRESENCE IN NEOPLASTIC
TISSUES
Immunohistochemical studies have provided suggestive
evidence that the expression of TFPI-2 decreases with an
increasing degree of malignancy (in the case of breast,
gastric, colon, pancreatic, renal, laryngeal, and endometrial cancer and glial neoplasms).21 The highest levels of
TFPI-2 mRNA and protein were detected in normal
brain, with lesser amounts in low-grade gliomas and
anaplastic astrocytomas. In glioblastomas, TFPI-2 was
2007
undetected.24 An inverse correlation between TFPI-2
distribution and the degree of malignancy was also
observed in choriocarcinoma18 and fibrosarcoma cell
lines.25 In addition, in about one third of non–small
cell lung cancers (NSCLCs), TFPI-2 gene expression is
decreased 4- to 120-fold compared with normal lung
tissue.26,27 Neither pancreatic cancer cell lines nor primary pancreatic ductal neoplasms revealed expression of
TFPI-2 mRNA.28 Interestingly, high expression of
TFPI-2 was observed exclusively in lobular carcinoma
of the breast, which is known to be associated with a
much more favorable prognosis than ductal invasive
breast carcinoma.21 Moreover, as much as 90% of
hepatocellular carcinomas were characterized by underexpression of TFPI-2.29 Thus, there is ample evidence
that TFPI-2 production is reduced or absent during
tumor progression. However, little is known concerning
the molecular underpinnings of this phenomenon.
GENETIC AND EPIGENETIC BASIS OF
TFPI-2 EXPRESSION IN CANCER
As mentioned above, the gene for the TFPI-2 is localized
in chromosome 7,13 which is frequently associated with
genetic changes in different cancers (e.g., head and neck
carcinomas,30 gastric,18 colon,31 prostate,31 vesicular,32
ovarian18 and testicular cancer,31 malignant melanoma,
and glial neoplasms32).Several highly aggressive cancers
delete the locus for the TFPI-2 gene in chromosome 7q
region, which results in a complete lack of TFPI-2
protein expression in these cells.33–35 In human choriocarcinoma cell line, the minimal TFPI-2 promoter was
demonstrated to be located between –166 and –111 from
the translation start site, and nuclear factor-1 (NF-1),
nuclear factor-kappa B (NF-kB), and early growth response gene-1 (egr-1)/specificity protein 1 (Sp1) binding
sites were crucial for TFPI-2 inducible expression.15 The
promoter region –313 to þ 1 is critical for minimal and
inducible promoter activity of human TFPI-2 in glioma
cells.36 This region harbors sites for several transcription
factors, including SP1, activiting enhancer-binding protein-1 (AP-1), NF-kB and NF-kB-like site, and lymphoid transcription factor-1 (Lyf-1).37 Mutations at
either of two AP-1 sites (–310 to –300 and –163 to
154), as well as either of two SP1 sites (–192 to 183 and –
135 to 128), lead to reduced TFPI-2 activity.37 Promoter
polymorphism of various genes is frequently responsible
for regulation of functions of particular proteins.26 However, none of 90 NSCLC patients exhibited genetic
variations in the TFPI-2 promoter region.26
Of particular interest is epigenetic inactivation of
TFPI-2 synthesis. Aberrant methylation of TFPI-2
promoter cytosine-phosphorothioate-guanine (CpG)
islands in human cancers and cancer cell lines was widely
documented to be responsible for diminished mRNA
encoding TFPI-2 and decreased or abolished synthesis
TFPI-2 ROLE IN CANCER/SIERKO ET AL
of TFPI-2 protein during cancer progression.27–30,38–41
The finding was associated with increased cancer
growth, invasion, and dissemination.27–29,38–41 Such a
mechanism of TFPI-2 gene silencing was demonstrated
in melanoma cell lines (HMV-I, MeWo, and WM-115),
as well as in 30% of metastatic tumors, but not in primary
melanoma surgical specimens.38 One third of NSCLC
patients, particularly at stage III and IV of the disease,
exhibited methylation of TFPI-2 gene promoter in
cancer cells.27 Approximately one half of NSCLC cases
with silenced TFPI-2 gene were lymph nodes positive.27
TFPI-2 methylation was observed in all cases of esophageal adenocarcinomas examined.39 Aberrant methylation of TFPI-2 was also revealed in as many as 73% of
pancreatic cancer xenografts and primary pancreatic adenocarcinomas.28 The finding was more frequently detected in older patients.28 Interestingly, TFPI-2
methylation in endoscopically aspirated pure pancreatic
juice was observed in 60% of pancreatic cancer patients in
contrast with 20% in patients with intraductal neoplasms or 5% in individuals with chronic pancreatitis.41
The incidence of the aberrant methylation of TFPI-2 in
the pure pancreatic juice reached 100% when liver
metastases were present and was as high as 90% in
patients who presented at stage IV of the disease.41
Promoter methylation was also documented to be
responsible for TFPI-2 gene silencing in SNB19 glioblastoma cell line.40 The finding was observed in 80%
of hepatocellular carcinoma cell lines and in 50% of
human hepatocellular carcinomas.29
Although methylation of the TFPI-2 gene promoter leads to its silencing, it does not seem to provide
the sole explanation for this phenomenon.42 Rao et al42
raised the hypothesis that one or more components of
pathways regulating TFPI-2 expression had also undergone methylation-associated silencing in cancer cells. In
this context, recent observations on the extracellular
signal-regulated kinase family (ERK)/mitogen-activated
protein kinase (MAPK) pathway mediation of the
TFPI-2 promoter activity is of particular interest.43 It
has also been reported that TFPI-2 expression is downregulated in cells harboring activated ras oncogenes.25
The Ras/Raf/mitogen-activity protein kinase (MAPK)/
extracellular signal-regulated kinase (ERK) signaling
cascade regulates cell proliferation and differentiation.44
Activation of components of the latter pathway, specifically oncogenic Ras and constitutively activated ERKs,
are frequent findings in various human malignancies.45–47
A novel mechanism, by which tumor cells circumvent the effects of TFPI-2, was recently discovered.48
Specifically, an aberrantly spliced variant of TFPI-2
mRNA derived from TFPI-2 pre-mRNA splicing at
exon/intron boundaries, as well as at new sites within
exons and introns (designated asTFPI-2), was detected.48 This form of TFPI-2 mRNA is untranslated.48 The levels of asTFPI-2 were very low or
negligible in normal cells but markedly upregulated in
neoplastic tissue and in several cancer cell lines.48 Based
on the latter observation, it is conceivable that quantification of asTFPI-2 mRNA levels could be a marker of
tumor progression.
Another facet of TFPI-2 regulation derives from
its cleavage by other tumor environment components.
Recently, an extracellular metalloproteinase, ADAMTS1,
was shown to modify the binding properties of TFPI-2
that alter the extracellular location of the protein and
disrupt its function.49 It is particularly noteworthy that
ADAMTS1 is expressed in human malignant tumors
and plays a significant role in structural remodeling
facilitating cancer invasion processes.50
TFPI-2 gene is becoming increasingly recognized
as a tumor suppressor gene. Marked downregulation of
TFPI-2 expression is associated with increasing malignancy.21,24 However, this observation raises an important question regarding its biological relevance in
aggressive tumors.
THE ROLE OF TFPI-2 IN MALIGNANCY
TFPI-2, an ECM-derived inhibitor, may function in the
maintenance of the stability of the tumor environment
and inhibit the growth of neoplasms. It has been shown
to suppress invasion of choriocarcinoma tumor cells both
in vitro and in vivo.51 Similarly, TFPI-2 decreases
invasion of human lung cancer A549cell line,52 glioblastoma SNB19 cell line,24,53 fibrosarcoma HT-1080 cell
line,54 prostate cancer LNCaP cell line,55 and amelanotic melanoma C-32 cell line.56 The invasive potential
of cancer derives from proteolytic activity driven by
tumor cells, which results in ECM and basement membrane degradation.57 In fact, TFPI-2 is thought to play a
pivotal role in the regulation of plasmin-mediated ECM
proteolysis. Downregulation of TFPI-2 protein concentration enhances cancer cell ability to degrade the ECM
because TFPI-2 is a potent inhibitor of plasmin regardless of whether the enzyme is associated with the tumor
cells or ECM.3,58 TFPI-2 exerts inhibitory effect on
plasmin- or trypsin-dependent activation of pro-matrix
metalloproteinase (MMP)-1 and pro-MMP-3, which
subsequently leads to diminished ECM degradation and
decreased invasion of HT-1080 fibrosarcoma cell
lines.54,59 In addition, it is conceivable that TFPI-2
may inhibit the plasmin-mediated release of proangiogenic molecules from the ECM components. Moreover,
TFPI-2 has been demonstrated to inhibit MMP-2
formation in HT-1080 fibrosarcoma cell line25 and to
directly inhibit MMP-1, MMP-13, MMP-2, and
MMP-9 in experimental models.60 However, recently
performed studies revealed that native TFPI-2 failed to
form complexes with MMP-2, MMP-9, and MMP-1.61
TFPI-2 also failed to inhibit the proteolytic activity of
MMP-1 toward triple-helical collagen.61 Experimental
655
656
SEMINARS IN THROMBOSIS AND HEMOSTASIS/VOLUME 33, NUMBER 7
studies focused on the effect of restoration of TFPI-2
revealed inhibition of invasion and tumor growth of
several cancer lines.28,62,63
A unique characteristics of TFPI-2 were observed
in human hepatocellular carcinoma cell line.64 The
serine proteinase inhibitor was demonstrated to promote
invasion, an effect that was suggested to be a result of
TFPI-2 binding to TF-VIIa complex on cancer cells.64
It is not obvious if this phenomenon represents the
mechanism by which TFPI-2 exerts its activity in vivo
because the results of another study revealed that hypermethylation of its gene promoter was detected in as
many as 50% of human hepatocellular carcinomas,
and ectopic overexpression of TFPI-2 markedly reduced
the proliferation and invasiveness of hepatocellular carcinoma cells.29 Significantly higher amounts of TFPI-2
gene promoter hypermethylation occurs in metastatic
disease, in relation to its expression in localized cancer,38,41 suggesting that silencing of the TFPI-2 gene
is involved in the process of metastases formation.
Tumor growth depends on the balance between
the ratio of proliferation of cancer cells and their apoptosis. Recently, the role of TFPI-2 in the induction of
apoptosis was described.65,66 Highly invasive human
glioblastoma cells either lack or contain undetectable
amounts of TFPI-2, whereas normal brain tissue and
low-grade glial tumors express this protein abundantly.21,24 An increase in apoptosis was shown in
aTFPI-2–positive multiforme glioblastoma cell line
(SNB-19) and in low-grade glioma cell line (Hs683).65
Restoration of TFPI-2 in human glioblastoma cell line
(U-251) activates both intrinsic and extrinsic caspasemediated, proapoptotic signaling pathways and induces
apoptosis.66
It is thoroughly documented that activation of
blood coagulation contributes to local progression of
cancer and facilitates metastatic spread as well.67–84
Tissue factor plays a key role in the initiation of coagulation activation in malignancy.62,82 The presence of
TF was observed in loco in many tumors and resulted in
increased invasiveness and higher rate of metastases
formation.67–84 The essential role in TF/VIIa inactivation is played by TFPI.1,2 Another inhibitor of TFdependent pathway of blood coagulation is TFPI-2. It
may inhibit tumor growth via interfering with TF
activity. However, data on the influence of TFPI-2 on
cancer biology by such a mechanism are scanty and will
require additional studies. Nonetheless, it seems that
there exists a regulatory self-restricting mechanism that,
to some extent, counteracts TF influence on cancer
progression. Namely, TF activity leads to thrombin
generation, which upregulates TFPI-2 synthesis.85 In
this way, thrombin may contribute to suppression of the
extrinsic pathway of blood coagulation and subsequently
facilitate the maintenance of ECM and may attenuate
TF-mediated impact on cancer progression.
2007
The process of angiogenesis is a rate-limiting step
in cancer progression.85 Several studies highlight the role
of TFPI-2 in the regulation of new blood vessel formation. In this regard, TFPI-2 was shown to inhibit
angiogenesis in experimental models.86,87 The inhibitor
is synthesized by ECs, particularly small blood vessels,
and secreted abundantly into the ECM.16 It may facilitate the maintenance of integrity of ECM and basement
membrane of small blood vessels, thus preventing initiation of angiogenesis. Moreover, TFPI-2 plays a role in
EC adhesion as anti-TFPI-2 IgG treatment of ECs
leads to their detachment from the ECM.16 Of interest,
TFPI-2 in ECs is upregulated by the essential proangiogenic growth factor vascular endothelial growth factor
(VEGF), which suppress proliferation of ECs.88 This
may represent the mechanism for negative-feedback
regulation of VEGF activity.
Several highly aggressive neoplasms (melanoma
as well as breast, lung, prostate, and ovarian cancer)
develop a vessel-like network formed by cancer cells
themselves; the process known as vasculogenic mimicry.89–95 The structures play a role in sustained blood
supply of the nascent tumor, and their formation was
correlated with tumor progression.89–95 In experimental
melanoma, TFPI-2 was demonstrated to strongly contribute to vasculogenic mimicry plasticity.96
The presence of TFPI-2 was demonstrated in
tumor-infiltrating macrophages in gastric and renal carcinomas.21 Cancer cells are able to synthesize and release
several different cytokines that exert multidirectional
influences on normal host cells97 and inflammatory
mediators, such as tumor necrosis factor-a (TNF-a),
endotoxin, and phorbol esters, which stimulate synthesis
and secretion of TFPI-2 by HUVECs.9 The precise role
of TFPI-2 localized in tumor-associated macrophages in
cancer biology is still unknown.
On the basis of gene profiling and protein
detection, it is tempting to speculate that TFPI-2
expression is attenuated during cancer progression,
which facilitates invasion, tumor growth, angiogenesis,
as well as metastases formation. Restoration of TFPI-2
in the tumor tissue inhibits the above-mentioned
processes suggesting a novel therapeutic target for the
treatment of cancer patients. However, additional information on the precise role of TFPI-2 in the tumor
biology of particular types of human cancer is needed
to design therapeutic options based on TFPI-2 structure and function.
ACKNOWLEDGMENTS
This work was supported in part by research grant 6
P05A 096 21 from the Polish Committee of Scientific
Research (KBN) to M.Z.W. and by research grant
HL64119 from the National Institutes of Health
(to W.K.).
TFPI-2 ROLE IN CANCER/SIERKO ET AL
ABBREVIATIONS
ADAMTS1 a disintegrin and metalloproteinase with
thrombospondin motifs
AP-1
activating enhancer-binding protein 1
CpG
cytosine-phosphorothioate-guanine
egr-1/Sp1
early growth response gene-1/specificity
protein 1
ECs
endothelial cells
ECM
extracellular matrix
ERK
extracellular signal-regulated kinase
HUVECs
human umbilical vein endothelial cells
Lyf-1
lymphoid transcription factor-1
MAPK
mitogen-activated protein kinase
MEK
MAPK/ERK
MMP
matrix metalloproteinase
NF-1
nuclear factor-1
NF-kB
nuclear factor-kappa B
NSCLC
non–small cell lung cancer
PP5
placental protein 5
SP1
specificity protein 1
TF
tissue factor
TFPI
tissue factor pathway inhibitor
TNF-a
tumor necrosis factor-a
VEGF
vascular endothelial growth factor
10.
11.
12.
13.
14.
15.
16.
17.
REFERENCES
1. Sprecher CA, Kisiel W, Mathews S, Foster DC. Molecular
cloning, expression, and partial characterization of a second
tissue-factor-pathway-inhibitor. Proc Natl Acad Sci USA
1994;91:3353–3357
2. Broze GJ Jr. Tissue factor pathway inhibitor. Thromb
Haemost 1995;74:90–93
3. Peterson LC, Sprecher CA, Foster DC, Blumberg H,
Hamamoto T, Kisiel W. Inhibitory properties of a novel
human Kunitz-type protease inhibitor homologous to tissue
factor pathway inhibitor. Biochemistry 1996;35:266–272
4. Chand HS, Schmidt AE, Bajaj SP, Kisiel W. Structurefunction analysis of the reactive site in the first Kunitz-type
domain of human factor pathway inhibitor-2. J Biol Chem
2004;279:17500–17507
5. Kisiel W, Sprecher CA, Foster DC. Evidence that a second
tissue factor pathway inhibitor (TFPI-2) and placental
protein 5 are equivalent. Blood 1994;84:4384–4385
6. Bohn H, Winckler W. Isolierung und Charakterisierung
des Plazenta-Proteins PP5. Arch Gynaek 1977;223:179–
186
7. Rao CN, Liu YY, Peavey CL, Woodley DT. Novel
extracellular matrix-associated serine proteinase inhibitors
from human skin fibroblasts. Arch Biochem Biophys 1995;
317:311–314
8. Rao CN, Peavey CL, Liu YY, Lapiere JC, Woodley DT.
Partial characterization of matrix-associated serine protease
inhibitors from human skin cells. J Invest Dermatol 1995;
104:379–383
9. Rao CN, Reddy P, Liu Y, et al. Extracellular matrixassociated serine protease inhibitors (Mr 33,000, 31,000,
27,000) are single-gene products with differential glycosylation: cDNA cloning of the 33-kDa inhibitor reveals its
18.
19.
20.
21.
22.
23.
24.
25.
identity to tissue factor pathway inhibitor-2. Arch Biochem
Biophys 1996;335:82–92
Siiteri JE, Koistinen HAT, Salem HAT, Bohn H, Seppaelae
M. Placental protein 5 is related to blood coagulation and
fibrinolytic systems. Life Sci 1982;30:1885–1891
Meisser A, Bischof P, Bohn H. Placental protein 5 (PP5)
inhibits thrombin-induced coagulation of fibrinogen. Arch
Gynecol 1985;236:197–201
Miyagi Y, Koshikawa N, Yasumitsu H, et al. 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. J Biochem (Tokyo)
1994;116:939–942
Miyagi Y, Yasumitsu H, Eki T, et al. Assignment of the
human PP5/TFPI-2 gene to 7q22 by FISH and PCR-based
human/rodent cell hybrid mapping panel analysis. Genomics
1996;35:267–268
Kamei S, Kazama Y, Foster DC, Kisiel W. Genomic
structure and promoter activity of the human tissue factor
pathway inhibitor-2 gene. Biochim Biophys Acta 2001;1517:
430–435
Hubé F, Reverdiau P, Iochmann S, Cherpi-Antar C, Gruel
Y. Characterization and functional analysis of TFPI-2 gene
promoter in a human choriocarcinoma cell line. Thromb Res
2003;109:207–215
Iino M, Foster DC, Kisiel W. Quantification and characterization of human endothelial cell-derived tissue factor pathway
inhibitor-2. Arterioscler Thromb Vasc Biol 1998; 18:40–46
Ortego J, Escribano J, Coca-Prados M. Gene expression of
proteases and proteases inhibitors in the human ciliary
epithelium and ODM-2 cells. Exp Eye Res 1997;65:289–299
Udagawa K, Miyagi Y, Hirahara F, et al. Specific expression
of PP5/TFPI-2 mRNA by syncytiotrophoblasts in human
placenta as revealed by in situ hybridization. Placenta 1998;
19:217–223
Buetzow R, Virtanen I, Seppaelae M, et al. Monoclonal
antibodies reacting with placental protein 5: use in radioimmunoassay, Western blot analysis, and immunohistochemistry. J Lab Clin Med 1988;111:249–256
Wahlstroem T, Bohn H, Seppaelae M. Immunohistochemical demonstration of placental protein 5 (PP5) – like
material in the seminal vesicles and the ampullar part of vas
deferens. Life Sci 1982;31:2723–2725
Wojtukiewicz MZ, Sierko E, Zimnoch L, Kozłowski L,
Sulkowski S, Kisiel W. Immunohistochemical localization of
tissue factor pathway inhibitor-2 in human tumor tissue.
Thromb Haemost 2003;90:140–146
Ranta T, Siiteri JE, Koistinen R, et al. Human seminal plasma
contains a protein that shares physiochemical and immunochemical properties with placental protein 5 from the human
placenta. J Clin Endocrinol Metab 1981;53:1087–1089
Seppaelae M, Tenhunen A, Koskimies AI, Wahlstroem T,
Koistinen R, Stenmam UH. Hyperstimulated human preovulatory follicular fluid contains placental protein 5 (PP5).
Fertil Steril 1984;41:62–65
Rao CN, Lakka SS, Kin Y, et al. Expression of tissue factor
pathway inhibitor 2 inversely correlates during the progression of human gliomas. Clin Cancer Res 2001;7:570–576
Izumi H, Takahashi C, Oh J, Noda M. Tissue factor
pathway inhibitor-2 suppresses the production of active
matrix metalloproteinase-2 and is down-regulated in cells
harboring activated ras oncogene. FEBS Lett 2000;481:
31–36
657
658
SEMINARS IN THROMBOSIS AND HEMOSTASIS/VOLUME 33, NUMBER 7
26. Rollin J, Régina S, Vourc’h P, et al. Influence of MMP-2 and
MMP-9 promoter polymorphisms on gene expression and
clinical outcome of non-small cell lung cancer. Lung Cancer
2007;56:273–280
27. Rollin J, Iochmann S, Bléchet C, et al. Expression and
methylation status of tissue factor pathway inhibitor-2 gene
in non-small-cell lung cancer. Br J Cancer 2005;92:775–
783
28. Sato N, Parker AR, Fukushima N, et al. Epigenetic
inactivation of TFPI-2 as a mechanism associated with
growth and invasion of pancreatic ductal adenocarcinoma.
Oncogene 2005;24:850–858
29. Wong CM, Ng YL, Lee JM, et al. Tissue factor pathway
inhibitor-2 as a frequently silenced tumor suppressor gene in
hepatocellular carcinoma. Hepatology 2007;45:1129–1138
30. Osella P, Carlson A, Wyandt H, Milunsky A. Cytogenetic
studies of eight squamous cell carcinomas of the head and
neck. Deletion of 7q, a possible primary chromosomal event.
Cancer Genet Cytogenet 1992;59:73–78
31. Atkin NB, Baker MC. Chromosome 7q deletions: observations on 13 malignant tumors. Cancer Genet Cytogenet
1993;67:123–125
32. Rowley JD, Mitelman F. Principles of molecular cell biology
of cancer: chromosome abnormalities in human cancer and
leukemia. In: DeVita VT, Hellman S, Rosenberg SA, eds.
Cancer. Principles and Practice of Oncology. 4th ed.
Philadelphia, PA: JB Lippincott; 1993:67–91
33. Saito E, Okamoto A, Saito M, et al. Genes associated with
the genesis of leiomyoma of the uterus in a commonly deleted
chromosomal region at 7q22. Oncol Rep 2005;13:2386–2397
34. Dong JT. Chromosomal deletions and tumor suppressor
genes in prostate cancer. Cancer Metastasis Rev 2001;20:
171–193
35. Sell SM, Tullis C, Stracner D, Song CY, Gewin J. Minimal
interwal defined on 7q in uterine leiomyoma. Cancer Genet
Cytogenet 2005;157:67–69
36. Konduri SD, Osman FA, Rao CN, et al. Minimal and
inducible regulation of tissue factor pathway inhibitor-2 in
human gliomas. Oncogene 2002;21:921–928
37. Konduri SD, Yanamandra N, Dinh DH, et al. Physiological
and chemical inducers of tissue factor pathway inhibitor-2 in
human glioma cells. Int J Oncol 2003;22:1277–1283
38. Nobeyama Y, Okochi-Takada E, Furuta J, et al. Silencing of
tissue factor pathway inhibitor-2 gene in malignant melanomas. Int J Cancer 2007;121:301–307
39. Steiner FA, Hong JA, Fischette MR, et al. Sequential 5-Aza
2’-deoxycytidine/depsipeptide FK228 treatment induces tissue factor pathway inhibitor 2 (TFPI-2) expression in cancer
cells. Oncogene 2005;23:2386–2397
40. Konduri SD, Srivenugopal KS, Yanamandra N, et al. Promoter
methylation and silencind of the tissue factor pathway
inhibitor-2 (TFPI-2), a gene encoding an inhibitor of matrix
metalloproteinases in human gliomas cells. Oncogene 2003;22:
4509–4516
41. Jiang P, Watanabe H, Okada G, et al. Diagnostic utility of
aberrant methylation of tissue factor pathway inhibitor 2 in
pure pancreatic carcinoma. Cancer Sci 2006;97:1267–1272
42. Rao CN, Segawa T, Navari JR, et al. Methylation of TFPI-2
gene is not the sole cause of its silencing. Int J Oncol 2003;
22:843–848
43. Kast C, Wang M, Whiteway M. The ERK/MAPK pathway
regulates the activity of the human tissue factor pathway
inhibitor-2 promoter. J Biol Chem 2003;278:6787–6794
2007
44. Khosravi-Far R, Campbell S, Rossman KL, Der CJ.
Increasing complexity of Ras signal transduction: involvement of Rho family proteins. Adv Cancer Res 1998;72:57–
107
45. Shapiro P. Ras-MAP kinase signaling pathways and control
of cell proliferation: relevance to cancer therapy. Crit Rev
Clin Lab Sci 2002;39:285–330
46. Sivaraman VS, Wang H, Nuovo GJ, Malbon CC. Hyperexpression of mitogen-activated protein kinase in human
breast cancer. J Clin Invest 1997;99:1478–1483
47. Oka H, Chatani Y, Hoshino R, et al. Constitutive activation
of mitogen-activated protein (MAP) kinases in human renal
cell carcinoma. Cancer Res 1995;55:4182–4187
48. Kempaiah P, Chand HS, Kisiel W. Identification of a human
TFPI-2 slice variant that is upregulated in human tumor
tissue. Mol Cancer 2007;6:20–31
49. Torres-Collado AX, Kisiel W, Iruela-Arispe ML, Rodriguez-Manzaneque JC. ADAMTS1 interacts with, cleaves,
and modifies the extracellular location of the matrix inhibitor
tissue factor pathway inhibitor-2. J Biol Chem 2006;28:
17827–17837
50. Mochizuki S, Okada Y. ADAMs in cancer cell proliferation
and progression. Cancer Sci 2007;98:621–628
51. Jin M, Udagawa K, Miyagi E, et al. Expression of serine
proteinase inhibitor PP5/TFPI-2/MSPI decreases the invasive potential of human choriocarcinoma cells in vitro and in
vivo. Gynecol Oncol 2001;83:325–333
52. Lakka SS, Konduri SD, Mohanam S, Nicolson GL, Rao JS.
In vitro modulation of human lung cancer cell line
invasiveness by antisense cDNA of tissue factor pathway
inhibitor-2. Clin Exp Metastasis 2000;18:239–244
53. Konduri SD, Rao CN, Chandrasekar N, et al. A novel
function of tissue factor pathway inhibitor-2 (TFPI-2) in
human glioma invasion. Oncogene 2001;20:6938–6945
54. Rao CN, Cook B, Liu Y, et al. HT-1080 fibrosarcoma cell
matrix degradation and invasion are inhibited by the matrixassociated serine protease inhibitor TFPI-2/33 kDa MSPI.
Int J Cancer 1998;76:749–756
55. Konduri SD, Tasiou A, Chandrasekar N, Rao JS. Overexpression of tissue factor pathway inhibitor-2 (TFPI-2)
decreases the invasiveness of prostate cancer cells in vitro. Int
J Cancer 2001;18:127–131
56. Konduri SD, Tasiou A, Chandrasekar N, Nicolson GL, Rao
JS. Role of tissue factor pathway inhibitor-2 (TFPI-2) in
amelanotic melanoma (C-32) invasion. Clin Exp Metastasis
2000;18:303–308
57. Carroll V, Binder B. The role of the plasminogen activation
system in cancer. Semin Thromb Hemost 1999;25:183–
197
58. Siiteri JE, Koistinen HAT, Salem HAT, Bohn H, Seppaelae
M. Placental protein 5 is related to blood coagulation and
fibrinolytic systems. Life Sci 1982;30:1885–1891
59. Rao CN, Mohanam S, Puppala A, Rao JS. Regulation of
pro-MMP-1 and pro-MMP-3 activation by tissue factor
pathway inhibitor-2/matrix associated serine protease inhibitor. Biochem Biophys Res Commun 1999;255:94–98
60. Herman MP, Sukhova GK, Kisiel W, et al. Tissue factor
pathway inhibitor-2 is a novel inhibitor of matrix metalloproteinases with implications for atherosclerosis. J Clin
Invest 2001;107:1117–1126
61. Du X, Chand HS, Kisiel W. Human tissue factor pathway
inhibitor-2 does not bind or inhibit activated matrix metalloproteinase-1. Biochim Biophys Acta 2003;162:242–245
TFPI-2 ROLE IN CANCER/SIERKO ET AL
62. Kondraganti S, Gondi CS, Gujrati M, et al. Restoration of
tissue factor patway inhibitor inhibits invasion and trumor
growth in vitro and in vivo a malignant meningioma cell line.
Int J Oncol 2006;29:25–32
63. Sun Y, Xie M, Liu M, Jin D, Li P. Growth suppression of
human laryngeal squamous cell carcinoma by adenovirusmediated tissue factor pathway inhibitor gene 2. Laryngoscope 2006;116:596–601
64. Neaud V, Hisaka T, Monvoisin A, et al. Paradoxical proinvasive effect of the serine proteinase inhibitor tissue factor
pathway inhibitor-2 on human hepatocellular carcinoma
cells. J Biol Chem 2000;275:35565–35569
65. Tasiou A, Konduri SD, Yanamandra N, et al. A novel role of
tissue factor pathway inhibitor-2 in apoptosis of malignant
human gliomas. Int J Cancer 2001;19:591–597
66. George J, Gondi CS, Dinh DH, Gujrati M, Rao JS.
Restoration of tissue factor patway inhibitor-2 in a human
glioblastoma cell line triggers caspase-mediated pathway and
apoptosis. Clin Cancer Res 2007;13:3507–3517
67. Zacharski LR, Wojtukiewicz MZ, Costantini V, Ornstein
DL, Memoli VA. Pathways of coagulation/fibrinolysis
activation in malignancy. Semin Thromb Hemost 1992;18:
104–116
68. Hillen HFP. Thrombosis in cancer patients. Ann Oncol
2000;11(Suppl. 3):273–276
69. Wojtukiewicz MZ, Rucinska M, Zacharski RR, et al.
Localization of blood coagulation factors in situ in pancreatic
carcinoma. Thromb Haemost 2001;86:1416–1420
70. Wojtukiewicz MZ, Rucińska M, Zimnoch L, et al.
Expression of prothrombin fragment 1 þ 2 in cancer tissue
as an indicator of local activation of blood coagulation.
Thromb Res 2000;97:335–342
71. Shoji M, Hancock WW, Abe K, et al. Activation of blood
coagulation and angiogenesis in cancer. Am J Pathol 1998;
152:399–411
72. Wojtukiewicz MZ, Zacharski LR, Memoli VA, et al.
Indirect activation of blood coagulation in colon cancer.
Thromb Haemost 1989;62:1062–1066
73. Wojtukiewicz MZ, Zacharski LR, Rucińska M, et al.
Expression of tissue factor and tissue factor pathway inhibitor
in situ in laryngeal carcinoma. Thromb Haemost 1999;82:
1659–1662
74. Wojtukiewicz MZ, Zacharski LR, Memoli VA, et al.
Fibrinogen-fibrin transformation in situ in renal cell
carcinoma. Anticancer Res 1990;10:579–582
75. Wojtukiewicz MZ, Zacharski LR, Memoli VA, et al.
Abnormal regulation of coagulation/fibrinolysis in small cell
carcinoma of the lung. Cancer 1990;65:481–485
76. Wojtukiewicz MZ, Zacharski LR, Memoli VA, et al.
Malignant melanoma. Interaction with coagulation and
fibrinolysis pathways in situ. Am J Clin Pathol 1990;93:
516–521
77. Wojtukiewicz MZ, Tang TG, Ben-Josef E, Renaud C,
Walz DA, Honn KV. Solid tumor cells express functional
‘‘tethered ligand’’ thrombin receptor. Cancer Res 1995;55:
698–704
78. Wojtukiewicz MZ, Tang TG, Ciarelli JJ, et al. Thrombin
increases the metastatic potential of tumor cell. Int J Cancer
1993;54:793–806
79. Wojtukiewicz MZ, Tang TG, Nelson KK, Walz DA, Diglio
CA, Honn KV. Thrombin enhances tumor cell adhesive and
80.
81.
82.
83.
84.
85.
86.
87.
88.
89.
90.
91.
92.
93.
94.
95.
96.
97.
metastatic properties via increased aIIbß3 expression on the
cell surface. Thromb Res 1992;68:233–245
Even-Ram S, Uziely B. Cohen P met al. Thrombin receptor
overexpression in malignant and physiological invasion
processes. Nat Med 1998;4:909–914
Folkman J. Angiogenesis in cancer, vascular, rheumatoid and
other diseases. Nat Med 1995;1:27–31
Rao LVM. Tissue factor as a tumor procoagulant. Cancer
Metastasis Rev 1992;11:249–266
Tsopanoglou NE, Maragoudakis ME. On the mechanism of
thrombin-induced angiogenesis. J Biol Chem 1999;274:
23969–23976
Zhang Y, Deng Y, Luther T, et al. Tissue factor controls the
balance of angiogenic and antiangiogenic properties of tumor
cells in mice. J Clin Invest 1994;94:1320–1327
Neaud V, Duplantier JG, Mazzocco C, Kisiel W. Thrombin
up-regulates tissue factor pathway inhibitor-2 synthesis
through a cyclogenase-2-dependent, epidermal growth factor
receptor-independent mechanism. J Biol Chem 2004;279:
5200–5206
Yanamandra N, Kondraganti S, Gondi CS, et al. Recombinant adeno-associated virus (rAAV) expressing TFPI-2
inhibits invasion, angiogenesis and tumor growth in a human
glioblastoma cell line. Int J Cancer 2005;115:998–1005
Ivanciu L, Gerard RD, Tang H, Lupu F, Lupu C.
Adenovirus-mediated expression of tissue factor pathway
inhibitor-2 inhibits endothelial cell migration and angiogenesis. Arterioscler Thromb Vasc Biol 2007;27:310–316
Xu Z, Maiti D, Kisiel W, Duh EL. Tissue factor pathway
inhibitor-2 is up-regulated by vascular endothelial growth
factor and suppresses growth factor-induced proliferation of
endothelial cells. Arterioscler Thromb Vasc Biol 2006;26:
2819–2825
Ruf W, Seftor E, Petrovan RJ, et al. Differential role of tissue
factor pathway inhibitor 1 and 2 in melanoma vasculogenic
mimicry. Cancer Res 2003;63:5381–5389
Liu C, Huang H, Donate F, et al. Prostate-specific
membrane antigen directed selective thrombotic infarction
of tumors. Cancer Res 2002;62:560–566
Shirakawa K, Tsuda H, Heike Y, et al. Absence of
endothelial cells, central necrosis, and fibrosis are associated
with aggressive inflammatory breast cancer. Cancer Res 2001;
61:445–451
Pezzella F. Evidence for novel non-angiogenic pathway in
breast-cancer metastasis. Lancet 2000;355:1787–1800
Sood AK, Seftor EA, Fletcher MS, et al. Molecular
determinants of ovarian cancer plasticity. Am J Pathol 2001;
158:1279–1288
Sharma N, Seftor REB, Seftor EA, et al. Prostatic tumor cell
plasticity involves cooperative interactions of distinct phenotypic subpopulations: role in vasculogenic mimicry. Prostate
2002;50:189–201
Passalidou E, Trivella M, Singh N, et al. Vascular phenotype
in angiogenic and non-angiogenic lung non-small cell
carcinomas. Br J Cancer 2002;86:244–249
Ruf W, Seftor EA, Petrovan RJ, et al. Differential role of
tissue factor pathway inhibitors 1 and 2 in melanoma
vasculogenic mimicry. Cancer Res 2003;63:5381–5389
Falanga A, Rickles FR. Pathophysiology of the thrombophilic state in the cancer patient. Semin Thromb Hemost
1999;25:173–182
659