Hepatocyte growth factor activator inhibitor type‑1 in cancer: Advances and perspectives (Review)

  • Authors:
    • Qiaoli Zheng
    • Haijian Wu
    • Jiang Cao
    • Jingjia Ye
  • View Affiliations

  • Published online on: October 13, 2014     https://doi.org/10.3892/mmr.2014.2628
  • Pages: 2779-2785
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Cancer is one of the most common diseases, with high morbidity and mortality rates. Large‑scale efforts have been made to understand the pathogenesis of the disease, particularly in the advanced stages, in order to develop effective therapeutic approaches. Hepatocyte growth factor activator inhibitor type-1 (HAI-1), also known as serine protease inhibitor Kunitz type 1, inhibits the activity of several trypsin-like serine proteases. In particular, HAI-1 suppresses hepatocyte growth factor (HGF) activator and matriptase, resulting in subsequent inhibition of HGF/scatter factor and macrophage‑stimulating protein (MSP). HGF and MSP are involved in cancer development and progression, via the receptors Met receptor tyrosine kinase (RTK) and Ron RTK, respectively. Therefore, HAI-1-mediated downregulation of HGF and MSP signaling may suppress tumorigenesis and progression in certain types of cancers. Abnormal HAI-1 expression levels have been observed in various types of human cancer. The exact function of HAI-1 in cancer pathogenesis, however, has not been fully elucidated. In this review, the focus is on the potential impact of aberrant HAI-1 expression levels on tumorigenesis and progression, the underlying mechanisms, and areas that require further investigation to clarify the precise role of HAI-1 in cancer.

1. Introduction

Hepatocyte growth factor activator inhibitor type-1 (HAI-1), encoded by the serine protease inhibitor, Kunitz type 1 gene, is a membrane-bound Kunitz-type serine protease inhibitor (1). HAI-1 was firstly purified from the conditioned medium of the MKN45 human stomach carcinoma cell line and identified as an inhibitor of hepatocyte growth factor activator (HGFA) (2). HAI-1 has also been demonstrated to inhibit a number of type-II transmembrane serine proteases (TTSPs), including matriptase, hepsin, transmembrane protease serine 13 (TMPRSS13) and human airway trypsin-like protease (HAT) (36). As a protein predominantly expressed in epithelial cells, HAI-1 is vital for cell growth, survival and mobility (1).

Increasing evidence has demonstrated that HAI-1 suppresses tumorigenesis and progression via regulation of the activity of a range of serine proteases in the tumor microenvironment. HGFA, a target trypsin-like serine protease of HAI-1, is secreted as a single-chain zymogen precursor and is activated by thrombin during blood coagulation. The activated HGFA induces the activation of two known macromolecular substrates, namely hepatocyte growth factor (HGF) and macrophage-stimulating protein (MSP), which are critical proteins involved in cancer pathogenesis (7,8). Downregulation of the activity of these two substrates through HAI-1-mediated HGFA inhibition, therefore, suppresses tumorigenesis and progression. In addition, TTSPs, another subtype of target HAI-1 proteases, facilitate epithelial carcinogenesis and progression (9). Therefore, HAI-1 is an important and promising therapeutic target in tumor treatment. This review focuses on recent advances in the understanding of HAI-1 with regard to the development and progression of cancer, and future studies concerning HAI-1 are proposed.

2. HAI-1 functional domains and proteases inhibited by HAI-1

HAI-1 is composed of an N-terminal extracellular region with two Kunitz domains (KD1 and KD2) separated by a low-density lipoprotein receptor (LDLR)-like domain, a transmembrane region and a short cytoplasmic region (Fig. 1) (10). The primary transmembrane form (66 kDa) of HAI-1 is released as several soluble proteins (58, 48, 40 and 39 kDa) into the extracellular milieu by proteolytic cleavage (11). The transmembrane and soluble forms of HAI-1 exhibit inhibitory activity against serine proteases in the pericellular microenvironment (11,12). Among these HAI-1 molecules, the 58 kDa and 40 kDa HAI-1 proteins are the predominant soluble forms in the conditioned medium from cancer cell cultures. The 40 kDa HAI-1, which lacks KD2, exhibits higher inhibitory activity against HGFA than the 58 kDa band (11). However, studies have demonstrated that KD1 is also responsible for protease inhibition via interaction with target proteins (1215). Furthermore, KD1-protease complex formation is enhanced by the LDLR-like domain but attenuated by KD2 (12).

HAI-1 exerts marked inhibitory activity against a variety of serine proteases, including HGFA, matriptase, hepsin, plasmin, trypsin, prostasin, TMPRSS13 and HAT (1,36,16,17). Studies have demonstrated that the proteases inhibited by HAI-1 clearly promote carcinogenesis and progression. For instance, HGFA expression is upregulated in breast, colorectal and renal cell carcinomas accompanied by downregulation of HAI-1 (1820). Matriptase, another protease inhibited by HAI-1, is overexpressed in a variety of malignant tumors, and possesses the ability to promote oncogenesis and progression (1). Hepsin-encoding gene, Hpn, is among the most consistently and quantitatively overexpressed genes in human prostate cancer, as detected by cDNA microarray and tissue array assays, and hepsin is the most reliable single marker to distinguish prostatic neoplasia from benign prostate hyperplasia (2123). Prostasin, one glycosylphosphatidylinositol-anchored serine protease, has been reported to be upregulated in ovarian cancer but downregulated in high-grade prostate cancer (17,24,25). Therefore, HAI-1 may contribute to the prevention of cancer growth and progression via inhibition of these serine protease activities.

3. Aberrant HAI-1 expression levels in cancer correlate with malignant phenotypes and clinicopathological parameters

Aberrant HAI-1 expression levels have been demonstrated in various types of cancer and have diagnostic and prognostic implications. The expression profiles and functions of HAI-1 have been investigated extensively in pre-clinical and clinical studies (Table I).

Table I

Expressional and functional studies of HAI-1 in cancer.

Table I

Expressional and functional studies of HAI-1 in cancer.

Cancer typeModelHAI-1 expressionConsequence/cancer associationReference
BreastMCF-7 cell lineHigh expression levelsLow invasionParr and Jiang(26)
MDA MB-231 cell lineLow expression levelsHigh invasionParr and Jiang (26)
MDA-MB-231 cell lineKnockdownEnhanced migration, proliferation, invasionParr and Jiang (31)
Breast cancer specimensLower levels in grade 3Decreased in poorly differentiated tumorsParr et al (18)
ColorectalPrimary colorectal carcinoma specimensLower levels in carcinoma tissueAssociated with disease progressionKataoka et al (19)
PancreaticSUIT-2 cell lineKnockdownReduced cell growth, but enhance invasionCheng et al (34)
SUIT-2 cell line, nude miceKnockdownEnhanced pulmonary metastasisFukushima et al (35)
OvarianOvarian cancer specimenLower levels in stage III/IVLoss of expression associated with advanced stageOberst et al (38)
GastricGastric cancer specimensLow expression levelsAssociated with invasion and lymph node metastasisZeng et al (39)
CervicalSiHa and HeLa cell linesOverexpressionInhibited growth, invasion, lead to apoptosisNakamura et al (28)
Cervical cancer specimensLow expression levelsPoor prognosisNakamura et al (28)
EndometrialKLE and HEC-251 cell linesOverexpressionInhibited growth, invasion and migrationNakamura et al (30)
Endometrial cancer specimensLow expression levelsPoor prognosisNakamura et al (30)
UterineSK-LMS-1 and SKN cell linesOverexpressionInhibited growth, invasion and migrationNakamura et al (29)
Uterine leiomyosarcoma specimensLow expression levelsPoor prognosisNakamura et al (29)
ProstatePC-3 and DU-145 cell linesKnockdownInhibited growth, enhance invasion and migrationSanders et al (32)
Prostate cancer specimensLow expression levelsAssociated with increasing aggressivenessSaleem et al (40)
Prostate cancer samplesHigh mean serum levelDistant metastasis and hormone resistanceNagakawa et al (47)
KidneyRenal cell carcinoma specimenLow expression levelsInvolved in cancer progressionYamauchi et al (20)
Oral cavityHSC-3 and SAS cell linesKnockdownReduced growth, but enhanced migrationBaba et al (33)
SAS cell line, nude miceKnockdownEnhanced tumorigenicityBaba et al (33)
Oral squamous cell carcinoma specimensReduced expression levels at the invasion frontAssociated with invasion, lymph node metastasisBaba et al (33)
LiverHep3B cell lineKnockdownInhibited growthNagata et al (37)
Hepatocellular carcinoma specimensPositive in 35% cancer tissuesInvolved in cancer progressionNagata et al (37)
Hepatocellular carcinoma specimensPositive in 31% cancer tissuesAssociated with poor prognosisFunagayama et al (41)

[i] HAI-1, hepatocyte growth factor activator inhibitor type-1.

Abnormal HAI-1 gene expression levels have been detected in a wide variety of human cancer cell lines; certain cell lines with a highly invasive nature exhibited low HAI-1 mRNA expression levels (26). In addition, as determined by in vitro and in vivo models, HAI-1 exerts a potential inhibitory effect on cancer cell invasion and migration, important hallmarks of cancer (27). Forced expression of HAI-1 significantly inhibited the invasion and migration of cervical, endometrial and uterine cancer cells in vitro (2830). Furthermore, breast, pancreatic, prostate and oral carcinoma cells exhibited enhanced invasive properties in vitro in response to HAI-1 knockdown (3134), a result also validated in nude mice bearing xenografts (33,35,36). Such findings are important for the analysis of the pathogenic role of HAI-1 in cancer cells. However, whether HAI-1 suppresses or promotes proliferation of cancer cells remains elusive (2834,37).

Immunohistochemical (IHC) staining has detected reduced HAI-1 expression levels in endometrial, cervical and colorectal carcinomas and uterine leiomyosarcoma, compared with adjacent normal tissues (19,2830). In addition, reduced HAI-1 mRNA levels have been detected by polymerase chain reaction in breast, gastric, colorectal and renal cell carcinoma (RCC) tissues (1820,38). Further detailed analysis revealed that reduced HAI-1 expression levels were associated with worse clinicopathological parameters (advanced stage, lymph node metastasis and distant metastasis) and/or poor prognosis (reduced disease-free survival and overall survival times) in ovarian, gastric, cervical, endometrial, renal cell and oral squamous cell carcinomas, and uterine leiomyosarcoma (19,20,2830,33,3940), but not in hepatocellular carcinoma (HCC) (37,41). Notably, HAI-1 was only marginally detectable in normal hepatocytes (42), while >30% HCC tissues were identified as HAI-1-positive by IHC (41), thus increased HAI-1 expression levels appear to be associated with advanced tumor stage and poor prognosis in HCC (37,41).

However, the exact role of HAI-1 in several types of cancer, including breast, colorectal and prostate cancer, remains controversial. Although reduced HAI-1 expression levels were associated with poorly differentiated breast cancer (18), high-level expression of HAI-I was found to be associated with poor patient outcome in a breast cancer tissue microarray analysis (43). HAI-1 downregulation in colorectal cancer has been observed in a number of studies, but enhanced immunoreactivity of HAI-1 was detected in colorectal cancer cells at the invasion front, which may be involved in distant metastasis, although this trend was not statistically significant (44). In human prostate cancer tissues, the HAI-1 protein levels were elevated compared with those of benign prostate tissues (45,46). The mean serum levels of HAI-1 in 118 patients with prostate cancer were reported to be significantly higher than those in 27 patients with benign prostatic hyperplasia. Furthermore, increased HAI-1 levels in serum were associated with distant metastasis and the development of hormone-resistance in prostate cancer (47). However, another study observed using immunohistochemistry indicated that HAI-1 expression levels were reduced in all grades of prostate cancer specimens (40).

According to current research, HAI-1 may exhibit different functions in different types of cancer or even at different stages/sites in the same type of cancer (26,32,37,40,47). However, the differences in measuring HAI-1 expression levels, the lack of standardized methods (including antibody) among studies create difficulties in reaching a conclusion regarding HAI-1 expression in cancer and its association with clinicopathological parameters. Further studies with large samples and standardized criteria are warranted to elucidate the role of HAI-1 in tumor pathology, and to determine the diagnostic and prognostic value of HAI-1 expression.

4. Molecular mechanisms of HAI-1 in cancer

As described above, HAI-1 exerts a suppressive effect on cancer invasion and metastasis, processes which result in a poor prognosis for cancer patients (48); however the molecular basis of HAI-1-mediated cancer inhibition remains poorly understood. In the present review, advances in the understanding of the diverse molecular mechanisms regulating HAI-1-mediated effects via target serine proteases, particularly HGFA and matriptase, are summarized. Studies have shown that increased expression levels of HGFA and/or matriptase were accompanied by significantly downregulated HAI-1 expression. Thus, the net balance between HGFA/matriptase and HAI-1 was shifted in favor of HGFA/matriptase in various types of carcinoma, including breast, ovarian, renal, prostate and colorectal carcinoma (1820,38,40,43,49,50). In addition, in vitro studies have validated the finding that HAI-1 knockdown-induced enhanced migration is partially reversed by silencing of matriptase or other serine protease expression (33,34,36).

The two best-characterized HAI-1-inhibited proteases (HGFA and matriptase) activate pro-HGF and pro-MSP, and are responsible for the subsequent activation of Met receptor tyrosine kinase (RTK) and Ron RTK, respectively (1,7,51). Dysregulation of the HGF-Met signaling pathway has been implicated in the development and metastasis of human cancer (52,53). Tumor xenografts with overexpressed HGF or Met exhibit high metastatic ability in mouse models (5458). In addition, angiogenesis and lymphangiogenesis are promoted in tumors due to the induction of endothelial cell growth by HGF-Met cascade, as revealed by in vitro and in vivo studies (5961). The downstream effectors of Met RTK activate several distinct signaling cascades, among which the RAS-mitogen activated protein kinase (MAPK) and phosphatidylinositol 3 kinase (PI3K)-AKT signaling pathways are predominant. The RAS-MAPK cascade eventually activates the extracellular signal-regulated kinases (ERKs), which transmit signals downstream, and results in the transcription of genes controlling cell proliferation, differentiation, adhesion, migration and apoptosis (62). Activation of the PI3K-AKT-mammalian target of rapamycin signaling pathway results in cancer cell proliferation and invasion (63). Met activation may also enhance the function of Rap1 and modulate the adhesion molecules cadherin and integrin, and therefore promote cell migration (64,65). As with the signaling activation pattern activated by HGF-Met, MSP-Ron signaling is also mediated by the RAS-MAPK and PI3K-AKT signaling pathways (Fig. 2) (7). Therefore, HAI-1 inhibits tumor development and progression via suppression of protease-mediated downstream signaling pathways.

In addition, the activation of the RAS-MAPK and PI3K-AKT signaling pathways is crucial for RTK-mediated epithelial-to-mesenchymal transition (EMT) in cancer cells (7). EMT is recognized as a potential mechanism for carcinoma metastasis and the loss of E-cadherin is a hallmark of EMT (66). The predominant transcriptional repressors of E-cadherin are zinc finger transcription factors, including Snail (Snai1), Slug, smad-interacting protein 1 (SIP1) and a basic helix-loop-helix transcription factor, Twist (67). An increasing amount of evidence has demonstrated that the interactions among HAI-1 and target serine proteases contribute to EMT in certain carcinoma cells. Support for this concept includes the finding that human pancreatic cancer cells with stable knockdown of HAI-1 exhibited an elongated spindle-like morphology and an enhanced migratory ability. Vimentin, SIP1 and matrix metalloproteinase (MMP)-9 expression was upregulated in these cells but E-cadherin expression was downregulated. The subsequent silencing of matriptase in these HAI-1 knockdown cells resulted in reversal in the expression levels of MMP-9 accompanied by a recovery of E-cadherin expression levels (34). In another study, HAI-1 overexpression resulted in a significant increase in E-cadherin expression levels but a reduction in Vimentin, SIP1, Snail and Twist expression levels in human endometrial cancer cell lines (30). The involvement of HAI-1 in EMT was further confirmed by other studies: Reduced E-cadherin expression levels in HAI-1-knockdown pancreatic cancer cells was reversed by recombinant KD1 (35) and HAI-1 knockdown oral squamous cell carcinoma cell lines exhibited more elongated morphology and reduced E-cadherin expression levels (33). All evidence reveals that HAI-1 inhibits tumor metastasis, partly by inhibiting EMT.

HAI-1 may also suppress the invasion and metastasis of tumor cells by inhibiting the activity of certain cognate serine proteases that activate fibrinolytic enzymes, MMPs and single-chain urokinase-type plasminogen activator (4,6870). These enzymes are responsible for the degradation of extracellular matrix components and further potentiate local tumor invasion and metastasis (71).

Recently, a transgenic mouse model revealed that HAI-1 suppressed intestinal tumorigenesis. Enhanced tumor formation was observed in mice with deficient intestinal HAI-1 expression. Notably, a total of 22 genes (including those encoding ligands, receptors, transcription factors and downstream genes) associated with the Wnt signaling pathway were identified by microarray analysis to be augmented in the tumors. Furthermore, the expression of several other genes involved in mucosal permeability and angiogenesis, including cldn2, lt1, cdh13, cdh5 and tnfrsf12a, was also upregulated (72). As barrier dysfunction may contribute to epithelial malignancy (73,74), HAI-1 may suppress tumorigenesis resulting from inhibition of the expression of these associated genes.

HAI-1 may exhibit different functions depending on the presence of cognate serine proteases in the intra- or extra-cellular milieu. The membrane-form HAI-1 acts not only as an inhibitor to HGFA, but also as an acceptor molecule, generating a reservoir of active HGFA on the cell surface; the HAI-1-HGFA complex on cell membrane may be dissociated and release the active HGFA into the surrounding microenvironment (75). Another study demonstrated that HAI-1 acted as an essential cofactor in the activation of pro-matriptase (76). Therefore, abnormal HAI-1 function may potentially contribute to tumor development and progression under specific conditions.

In conclusion, these findings established that HAI-1 is key in the development and progression of cancer; however, identification of the acute mechanism remains incomplete and requires further investigation.

5. Conclusion and future perspectives

HAI-1 is a vital protein involved in a number of biological and pathological processes due to its ability to inhibit cognate serine proteases in the extracellular milieu. The majority of these serine proteases are involved in the development and progression of cancer; therefore, HAI-1 exerts a suppressive function in cancer through regulation of these proteases.

Thus far, considerable achievements have been gained in the understanding of the pathological role of HAI-1 in tumors, particularly in the impact of aberrant HAI-1 expression levels on tumor growth, invasion, angiogenesis and metastasis. Existing studies have identified several of the molecular mechanisms mediated by HAI-1 and the target serine proteases. As determined by these findings, the prognostic and pharmaceutical properties of HAI-1 render the molecule a promising factor in cancer diagnosis and treatment.

However, paradoxical results have been obtained regarding HAI-1 expression patterns in certain types of cancer. The regulatory mechanisms that result in aberrant HAI-1 expression levels under different circumstances remain elusive. Further investigation into HAI-1 is important not only for providing greater insight into the molecular aspects of HAI-1 in cancer, but also for the possible development of novel diagnostic and therapeutic approaches. Even at the early stages of HAI-1 clinical investigation, understanding the acute roles of HAI-1 in cancer no doubt contributes, at least partly, to the eventual control of human cancer.

Acknowledgements

This study was supported by the National Natural Science Foundation of China (nos. 30271450 and 30672365) and the Natural Science Foundation of Zhejiang Province (no. 300466).

References

1 

Parr C, Sanders AJ and Jiang WG: Hepatocyte growth factor activation inhibitors - therapeutic potential in cancer. Anticancer Agents Med Chem. 10:47–57. 2010. View Article : Google Scholar : PubMed/NCBI

2 

Shimomura T, Denda K, Kitamura A, et al: Hepatocyte growth factor activator inhibitor, a novel Kunitz-type serine protease inhibitor. J Biol Chem. 272:6370–6376. 1997. View Article : Google Scholar : PubMed/NCBI

3 

Lin CY, Anders J, Johnson M and Dickson RB: Purification and characterization of a complex containing matriptase and a Kunitz-type serine protease inhibitor from human milk. J Biol Chem. 274:18237–18242. 1999. View Article : Google Scholar : PubMed/NCBI

4 

Kirchhofer D, Peek M, Lipari MT, et al: Hepsin activates pro-hepatocyte growth factor and is inhibited by hepatocyte growth factor activator inhibitor-1B (HAI-1B) and HAI-2. FEBS Lett. 579:1945–1950. 2005. View Article : Google Scholar

5 

Hashimoto T, Kato M, Shimomura T and Kitamura N: TMPRSS13, a type II transmembrane serine protease, is inhibited by hepatocyte growth factor activator inhibitor type 1 and activates pro-hepatocyte growth factor. FEBS J. 277:4888–4900. 2010. View Article : Google Scholar : PubMed/NCBI

6 

Kato M, Hashimoto T, Shimomura T, et al: Hepatocyte growth factor activator inhibitor type 1 inhibits protease activity and proteolytic activation of human airway trypsin-like protease. J Biochem. 151:179–187. 2012. View Article : Google Scholar : PubMed/NCBI

7 

Yao HP, Zhou YQ, Zhang R and Wang MH: MSP-RON signalling in cancer: pathogenesis and therapeutic potential. Nat Rev Cancer. 13:466–481. 2013. View Article : Google Scholar : PubMed/NCBI

8 

Eigenbrot C, Ganesan R and Kirchhofer D: Hepatocyte growth factor activator (HGFA): molecular structure and interactions with HGFA inhibitor-1 (HAI-1). FEBS J. 277:2215–2222. 2010. View Article : Google Scholar : PubMed/NCBI

9 

Bugge TH, Antalis TM and Wu Q: Type II transmembrane serine proteases. J Biol Chem. 284:23177–23181. 2009. View Article : Google Scholar : PubMed/NCBI

10 

Kataoka H, Miyata S, Uchinokura S and Itoh H: Roles of hepatocyte growth factor (HGF) activator and HGF activator inhibitor in the pericellular activation of HGF/scatter factor. Cancer Metastasis Rev. 22:223–236. 2003. View Article : Google Scholar : PubMed/NCBI

11 

Shimomura T, Denda K, Kawaguchi T, et al: Multiple sites of proteolytic cleavage to release soluble forms of hepatocyte growth factor activator inhibitor type 1 from a transmembrane form. J Biochem. 126:821–828. 1999. View Article : Google Scholar

12 

Kojima K, Tsuzuki S, Fushiki T and Inouye K: Roles of functional and structural domains of hepatocyte growth factor activator inhibitor type 1 in the inhibition of matriptase. J Biol Chem. 283:2478–2487. 2008. View Article : Google Scholar : PubMed/NCBI

13 

Denda K, Shimomura T, Kawaguchi T, Miyazawa K and Kitamura N: Functional characterization of Kunitz domains in hepatocyte growth factor activator inhibitor type 1. J Biol Chem. 277:14053–14059. 2002. View Article : Google Scholar : PubMed/NCBI

14 

Kirchhofer D, Peek M, Li W, et al: Tissue expression, protease specificity, and Kunitz domain functions of hepatocyte growth factor activator inhibitor-1B (HAI-1B), a new splice variant of HAI-1. J Biol Chem. 278:36341–36349. 2003. View Article : Google Scholar : PubMed/NCBI

15 

Miyata S, Fukushima T, Kohama K, et al: Roles of Kunitz domains in the anti-invasive effect of hepatocyte growth factor activator inhibitor type 1 in human glioblastoma cells. Hum Cell. 20:100–106. 2007. View Article : Google Scholar : PubMed/NCBI

16 

Shia S, Stamos J, Kirchhofer D, et al: Conformational lability in serine protease active sites: structures of hepatocyte growth factor activator (HGFA) alone and with the inhibitory domain from HGFA inhibitor-1B. J Mol Bio. 346:1335–1349. 2005. View Article : Google Scholar : PubMed/NCBI

17 

Fan B, Wu TD, Li W and Kirchhofer D: Identification of hepatocyte growth factor activator inhibitor-1B as a potential physiological inhibitor of prostasin. J Biol Chem. 280:34513–34520. 2005. View Article : Google Scholar : PubMed/NCBI

18 

Parr C, Watkins G, Mansel RE and Jiang WG: The hepatocyte growth factor regulatory factors in human breast cancer. Clin Cancer Res. 10:202–211. 2004. View Article : Google Scholar : PubMed/NCBI

19 

Kataoka H, Hamasuna R, Itoh H, Kitamura N and Koono M: Activation of hepatocyte growth factor/scatter factor in colorectal carcinoma. Cancer Res. 60:6148–6159. 2000.PubMed/NCBI

20 

Yamauchi M, Kataoka H, Itoh H, et al: Hepatocyte growth factor activator inhibitor types 1 and 2 are expressed by tubular epithelium in kidney and down-regulated in renal cell carcinoma. J Urol. 171:890–896. 2004. View Article : Google Scholar : PubMed/NCBI

21 

Dhanasekaran SM, Barrette TR, Ghosh D, et al: Delineation of prognostic biomarkers in prostate cancer. Nature. 412:822–826. 2001. View Article : Google Scholar : PubMed/NCBI

22 

Wu QY and Parry G: Hepsin and prostate cancer. Front Biosci. 12:5052–5059. 2007. View Article : Google Scholar

23 

Klezovitch O, Chevillet J, Mirosevich J, et al: Hepsin promotes prostate cancer progression and metastasis. Cancer Cell. 6:185–195. 2004. View Article : Google Scholar : PubMed/NCBI

24 

Mok SC, Chao J, Skates S, et al: Prostasin, a potential serum marker for ovarian cancer: identification through microarray technology. J Natl Cancer Inst. 93:1458–1464. 2001. View Article : Google Scholar : PubMed/NCBI

25 

Takahashi S, Suzuki S, Inaguma S, et al: Down-regulated expression of prostasin in high-grade or hormone-refractory human prostate cancers. Prostate. 54:187–193. 2003. View Article : Google Scholar : PubMed/NCBI

26 

Parr C and Jiang WG: Expression of hepatocyte growth factor/scatter factor, its activator, inhibitors and the c-Met receptor in human cancer cells. Int J Oncol. 19:857–863. 2001.PubMed/NCBI

27 

Hanahan D and Weinberg RA: Hallmarks of cancer: the next generation. Cell. 144:646–674. 2011. View Article : Google Scholar : PubMed/NCBI

28 

Nakamura K, Abarzua F, Hongo A, et al: The role of hepatocyte growth factor activator inhibitor-1 (HAI-1) as a prognostic indicator in cervical cancer. Int J Oncol. 35:239–248. 2009.PubMed/NCBI

29 

Nakamura K, Abarzua F, Hongo A, et al: Hepatocyte growth factor activator inhibitors (HAI-1 and HAI-2) are potential targets in uterine leiomyosarcoma. Int J Oncol. 37:605–614. 2010. View Article : Google Scholar

30 

Nakamura K, Hongo A, Kodama J and Hiramatsu Y: The role of hepatocyte growth factor activator inhibitor (HAI)-1 and HAI-2 in endometrial cancer. Int J Cancer. 128:2613–2624. 2011. View Article : Google Scholar : PubMed/NCBI

31 

Parr C and Jiang WG: Hepatocyte growth factor activation inhibitors (HAI-1 and HAI-2) regulate HGF-induced invasion of human breast cancer cells. Int J Cancer. 119:1176–1183. 2006. View Article : Google Scholar

32 

Sanders AJ, Parr C, Mason MD and Jiang WG: Suppression of hepatocyte growth factor activator inhibitor-1 leads to a more aggressive phenotype of prostate cancer cells in vitro. Int J Mol Med. 20:613–619. 2007.PubMed/NCBI

33 

Baba T, Kawaguchi M, Fukushima T, et al: Loss of membrane-bound serine protease inhibitor HAI-1 induces oral squamous cell carcinoma cells’ invasiveness. J Pathol. 228:181–192. 2012.PubMed/NCBI

34 

Cheng H, Fukushima T, Takahashi N, Tanaka H and Kataoka H: Hepatocyte growth factor activator inhibitor type 1 regulates epithelial to mesenchymal transition through membrane-bound serine proteinases. Cancer Res. 69:1828–1835. 2009. View Article : Google Scholar

35 

Fukushima T, Kawaguchi M, Yamasaki M, et al: Hepatocyte growth factor activator inhibitor type 1 suppresses metastatic pulmonary colonization of pancreatic carcinoma cells. Cancer Sci. 102:407–413. 2011. View Article : Google Scholar : PubMed/NCBI

36 

Ye J, Kawaguchi M, Haruyama Y, et al: Loss of hepatocyte growth factor activator inhibitor type 1 participates in metastatic spreading of human pancreatic cancer cells in a mouse orthotopic transplantation model. Cancer Sci. 105:44–51. 2014. View Article : Google Scholar

37 

Nagata K, Hirono S, Ido A, et al: Expression of hepatocyte growth factor activator and hepatocyte growth factor activator inhibitor type 1 in human hepatocellular carcinoma. Biochem Biophys Res Commun. 289:205–211. 2001. View Article : Google Scholar : PubMed/NCBI

38 

Zeng L, Cao J and Zhang X: Expression of serine protease SNC19/matriptase and its inhibitor hepatocyte growth factor activator inhibitor type 1 in normal and malignant tissues of gastrointestinal tract. World J Gastroenterol. 11:6202–6207. 2005.PubMed/NCBI

39 

Oberst MD, Johnson MD, Dickson RB, et al: Expression of the serine protease matriptase and its inhibitor HAI-1 in epithelial ovarian cancer: correlation with clinical outcome and tumor clinicopathological parameters. Clin Cancer Res. 8:1101–1107. 2002.

40 

Saleem M, Adhami VM, Zhong W, et al: A novel biomarker for staging human prostate adenocarcinoma: overexpression of matriptase with concomitant loss of its inhibitor, hepatocyte growth factor activator inhibitor-1. Cancer Epidemiol Biomarkers Prev. 15:217–227. 2006. View Article : Google Scholar

41 

Funagayama M, Kondo K, Chijiiwa K and Kataoka H: Expression of hepatocyte growth factor activator inhibitor type 1 in human hepatocellular carcinoma and postoperative outcomes. World J Surg. 34:1563–1571. 2010. View Article : Google Scholar : PubMed/NCBI

42 

Kataoka H, Suganuma T, Shimomura T, et al: Distribution of hepatocyte growth factor activator inhibitor type 1 (HAI-1) in human tissues. Cellular surface localization of HAI-1 in simple columnar epithelium and its modulated expression in injured and regenerative tissues. J Histochem Cytochem. 47:673–682. 1999. View Article : Google Scholar

43 

Kang JY, Dolled-Filhart M, Ocal IT, et al: Tissue microarray analysis of hepatocyte growth factor/Met pathway components reveals a role for Met, matriptase, and hepatocyte growth factor activator inhibitor 1 in the progression of node-negative breast cancer. Cancer Res. 63:1101–1105. 2003.

44 

Nagaike K, Kohama K, Uchiyama S, et al: Paradoxically enhanced immunoreactivity of hepatocyte growth factor activator inhibitor type 1 (HAI-1) in cancer cells at the invasion front. Cancer Sci. 95:728–735. 2004. View Article : Google Scholar : PubMed/NCBI

45 

Warren M, Twohig M, Pier T, et al: Protein expression of matriptase and its cognate inhibitor HAI-1 in human prostate cancer: a tissue microarray and automated quantitative analysis. Appl Immunohistochem Mol Morphol. 17:23–30. 2009. View Article : Google Scholar : PubMed/NCBI

46 

Yasuda K, Komiya A, Watanabe A, et al: Expression of Hepatocyte growth factor activator inhibitor type-1 (HAI-1) in prostate cancer. Anticancer Res. 33:575–581. 2013.PubMed/NCBI

47 

Nagakawa O, Yamagishi T, Akashi T, Nagaike K and Fuse H: Serum hepatocyte growth factor activator inhibitor type I (HAI-I) and type 2 (HAI-2) in prostate cancer. Prostate. 66:447–452. 2006. View Article : Google Scholar : PubMed/NCBI

48 

Hudson BD, Kulp KS and Loots GG: Prostate cancer invasion and metastasis: insights from mining genomic data. Brief Funct Genomics. 12:397–410. 2013. View Article : Google Scholar : PubMed/NCBI

49 

Vogel LK, Saebø M, Skjelbred CF, et al: The ratio of Matriptase/HAI-l mRNA is higher in colorectal cancer adenomas and carcinomas than corresponding tissue from control individuals. BMC Cancer. 6:1762006. View Article : Google Scholar

50 

Tsai WC, Sheu LF, Chao YC, et al: Decreased Matriptase/HAI-1 ratio in advanced colorectal adenocarcinoma of Chinese patients. Chin J Physiol. 50:225–231. 2007.PubMed/NCBI

51 

Naldini L, Weidner KM, Vigna E, et al: Scatter factor and hepatocyte growth factor are indistinguishable ligands for the MET receptor. EMBO J. 10:2867–2878. 1991.PubMed/NCBI

52 

Graveel C, Su Y, Koeman J, et al: Activating Met mutations produce unique tumor profiles in mice with selective duplication of the mutant allele. Proc Natl Acad Sci USA. 101:17198–17203. 2004. View Article : Google Scholar

53 

Ponzo MG, Lesurf R, Petkiewicz S, et al: Met induces mammary tumors with diverse histologies and is associated with poor outcome and human basal breast cancer. Proc Natl Acad Sci USA. 106:12903–12908. 2009. View Article : Google Scholar : PubMed/NCBI

54 

Rong S, Segal S, Anver M, Resau JH and Vande Woude GF: Invasiveness and metastasis of NIH 3T3 cells induced by Met-hepatocyte growth factor/scatter factor autocrine stimulation. Proc Natl Acad Sci USA. 91:4731–4735. 1994. View Article : Google Scholar : PubMed/NCBI

55 

Meiners S, Brinkmann V, Naundorf H and Birchmeier W: Role of morphogenetic factors in metastasis of mammary carcinoma cells. Oncogene. 16:9–20. 1998. View Article : Google Scholar

56 

Gallego MI, Bierie B and Hennighausen L: Targeted expression of HGF/SF in mouse mammary epithelium leads to metastatic adenosquamous carcinomas through the activation of multiple signal transduction pathways. Oncogene. 22:8498–8508. 2003. View Article : Google Scholar

57 

Jeffers M, Fiscella M, Webb CP, et al: The mutationally activated Met receptor mediates motility and metastasis. Proc Natl Acad Sci USA. 95:14417–14422. 1998. View Article : Google Scholar : PubMed/NCBI

58 

Moshitch-Moshkovitz S, Tsarfaty G, Kaufman DW, et al: In vivo direct molecular imaging of early tumorigenesis and malignant progression induced by transgenic expression of GFP-Met. Neoplasia. 8:353–363. 2006. View Article : Google Scholar

59 

Abounader R and Laterra J: Scatter factor/hepatocyte growth factor in brain tumor growth and angiogenesis. Neuro Oncol. 7:436–451. 2005. View Article : Google Scholar : PubMed/NCBI

60 

Bussolino F, Di Renzo MF, Ziche M, et al: Hepatocyte growth factor is a potent angiogenic factor which stimulates endothelial cell motility and growth. J Cell Biol. 119:629–641. 1992. View Article : Google Scholar

61 

Grant DS, Kleinman HK, Goldberg ID, et al: Scatter factor induces blood vessel formation in vivo. Proc Natl Acad Sci USA. 90:1937–1941. 1993. View Article : Google Scholar : PubMed/NCBI

62 

Santarpia L, Lippman SM and El-Naggar AK: Targeting the MAPK-RAS-RAF signaling pathway in cancer therapy. Expert Opin Ther Targets. 16:103–119. 2012. View Article : Google Scholar : PubMed/NCBI

63 

Sami A and Karsy M: Targeting the PI3K/AKT/mTOR signaling pathway in glioblastoma: novel therapeutic agents and advances in understanding. Tumour Biol. 34:1991–2002. 2013. View Article : Google Scholar : PubMed/NCBI

64 

Gherardi E, Birchmeier W, Birchmeier C and Vande Woude G: Targeting MET in cancer: rationale and progress. Nat Rev Cancer. 12:89–103. 2012. View Article : Google Scholar : PubMed/NCBI

65 

Boettner B and Van Aelst L: Control of cell adhesion dynamics by Rap1 signaling. Curr Opin Cell Biol. 21:684–693. 2009. View Article : Google Scholar : PubMed/NCBI

66 

Thiery JP: Epithelial-mesenchymal transitions in tumour progression. Nat Rev Cancer. 2:442–454. 2002. View Article : Google Scholar : PubMed/NCBI

67 

Berx G, Raspé E, Christofori G, Thiery JP and Sleeman JP: Pre-EMTing metastasis? Recapitulation of morphogenetic processes in cancer. Clin Exp Metastasis. 24:587–597. 2007. View Article : Google Scholar : PubMed/NCBI

68 

Herter S, Piper DE, Aaron W, et al: Hepatocyte growth factor is a preferred in vitro substrate for human hepsin, a membrane-anchored serine protease implicated in prostate and ovarian cancers. Biochem J. 390:125–136. 2005.PubMed/NCBI

69 

Lee SL, Dickson RB and Lin CY: Activation of hepatocyte growth factor and urokinase/plasminogen activator by matriptase, an epithelial membrane serine protease. J Biol Chem. 275:36720–36725. 2000. View Article : Google Scholar : PubMed/NCBI

70 

Takeuchi T, Harris JL, Huang W, et al: Cellular localization of membrane-type serine protease 1 and identification of protease-activated receptor-2 and single-chain urokinase-type plasminogen activator as substrates. J Biol Chem. 275:26333–26342. 2000. View Article : Google Scholar

71 

Liotta LA: Tumor invasion and metastases - role of the extracellular matrix: Rhoads Memorial Award lecture. Cancer Res. 46:1–7. 1986.PubMed/NCBI

72 

Hoshiko S, Kawaguchi M, Fukushima T, et al: Hepatocyte growth factor activator inhibitor type 1 is a suppressor of intestinal tumorigenesis. Cancer Res. 73:2659–2670. 2013. View Article : Google Scholar : PubMed/NCBI

73 

Turner JR: Intestinal mucosal barrier function in health and disease. Nat Rev Immunol. 9:799–809. 2009. View Article : Google Scholar : PubMed/NCBI

74 

Turksen K and Troy TC: Junctions gone bad: Claudins and loss of the barrier in cancer. Biochim Biophys Acta. 1816:73–79. 2011.PubMed/NCBI

75 

Kataoka H, Shimomura T, Kawaguchi T, et al: Hepatocyte growth factor activator inhibitor type 1 is a specific cell surface binding protein of hepatocyte growth factor activator (HGFA) and regulates HGFA activity in the pericellular microenvironment. J Biol Chem. 275:40453–40462. 2000. View Article : Google Scholar

76 

Oberst MD, Williams CA, Dickson RB, Johnson MD and Lin CY: The activation of matriptase requires its noncatalytic domains, serine protease domain, and its cognate inhibitor. J Biol Chem. 278:26773–26779. 2003. View Article : Google Scholar

Related Articles

Journal Cover

December-2014
Volume 10 Issue 6

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
Zheng Q, Wu H, Cao J and Ye J: Hepatocyte growth factor activator inhibitor type‑1 in cancer: Advances and perspectives (Review). Mol Med Rep 10: 2779-2785, 2014
APA
Zheng, Q., Wu, H., Cao, J., & Ye, J. (2014). Hepatocyte growth factor activator inhibitor type‑1 in cancer: Advances and perspectives (Review). Molecular Medicine Reports, 10, 2779-2785. https://doi.org/10.3892/mmr.2014.2628
MLA
Zheng, Q., Wu, H., Cao, J., Ye, J."Hepatocyte growth factor activator inhibitor type‑1 in cancer: Advances and perspectives (Review)". Molecular Medicine Reports 10.6 (2014): 2779-2785.
Chicago
Zheng, Q., Wu, H., Cao, J., Ye, J."Hepatocyte growth factor activator inhibitor type‑1 in cancer: Advances and perspectives (Review)". Molecular Medicine Reports 10, no. 6 (2014): 2779-2785. https://doi.org/10.3892/mmr.2014.2628