Translational implication of Kallmann syndrome-1 gene expression in hepatocellular carcinoma

  • Authors:
    • Yuri Tanaka
    • Mitsuro Kanda
    • Hiroyuki Sugimoto
    • Dai Shimizu
    • Satoshi Sueoka
    • Hideki Takami
    • Kazuhiro Ezaka
    • Ryoji Hashimoto
    • Yukiyasu Okamura
    • Naoki Iwata
    • Chie Tanaka
    • Suguru Yamada
    • Tsutomu Fujii
    • Goro Nakayama
    • Masahiko Koike
    • Shuji Nomoto
    • Michitaka Fujiwara
    • Yasuhiro Kodera
  • View Affiliations

  • Published online on: April 16, 2015     https://doi.org/10.3892/ijo.2015.2965
  • Pages: 2546-2554
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Accumulation of epigenetic alterations causes inactivation of tumor suppressors and contributes to the initiation and progression of hepatocellular carcinoma (HCC). Identification of methylated genes is necessary to improve our understanding of the pathogenesis of HCC and develop novel biomarkers and therapeutic targets. The Kallmann syndrome-1 (KAL1) gene encodes an extracellular matrix-related protein with diverse oncological functions. However, the function of KAL1 in HCC has not been examined. We investigated the methylation status of the KAL1 promoter region in HCC cell lines, and evaluated KAL1 mRNA levels and those of genes encoding potential interacting cell adhesion factors. KAL1 mRNA expression level was heterogeneous in nine HCC cell lines, and reactivation of KAL1 mRNA expression was observed in cells with promoter hypermethylation of KAL1 gene after demethylation. In addition, KAL1 mRNA levels inversely correlated with those of ezrin in all nine HCC cell lines. KAL1 expression levels in 144 pairs of surgically-resected tissues were determined and correlated to clinicopathological parameters. KAL1 mRNA level was independent of the background liver status, whereas HCC tissues showed significantly lower KAL1 mRNA levels than corresponding noncancerous liver tissues. Downregulation of KAL1 mRNA in HCC was significantly associated with malignant phenotype characteristics, including elevated tumor markers, larger tumor size, vascular invasion, and hypermethylation of KAL1. Patients with downregulation of KAL1 were more likely to have a shorter overall survival than other patients, and multivariate analysis identified downregulation of KAL1 as an independent prognostic factor (hazard ratio 2.04, 95% confidence interval 1.11-3.90, P=0.022). Our results indicated that KAL1 may act as a putative tumor suppressor in HCC and is inactivated by promoter hypermethylation. KAL1 may serve as a biomarker of malignant phenotype of HCC.

Introduction

Hepatocellular carcinoma (HCC) is the most common primary malignancy of the liver and the third most common cause of cancer-related death worldwide (1,2). Development of HCC is considered as a discriminative event because it occurs in chronically damaged tissue due to chronic hepatitis and liver cirrhosis, whereas other common malignancies develop on otherwise healthy tissue (35). Because of the accumulated genome instability and numerous epigenetic alterations induced by the microenvironment of the background liver, HCC is a more heterogeneous disease (3).

Aberrant DNA methylation is one of the most common epigenetic alterations in malignancies and is specific to individual organs and diseases (68). Furthermore, several studies have shown that aberrant DNA methylation contributes to the initiation and progression of malignant tumors through inactivation of tumor suppressors (9,10). Therefore, identification of novel methylated genes is important for the development of both diagnostic markers and therapeutic targets, such as demethylation agents.

Kallmann syndrome-1 gene (KAL1), also named anosmin-1, encodes an extracellular matrix (ECM) related protein with a role in cellular adhesion. KAL1 contains a WAP domain and three FnIII domains, and promotes the migration of gonadotropin-releasing hormone neurons from the olfactory placode to the hypothalamus during development (1113). KAL1 also induces neurite outgrowth and cell migration through fibroblast growth factor receptor 1 (FGFR1) pathways (14,15). Studies have demonstrated that ECM proteins play a vital role in proliferation and invasion of tumor cells (16). However, to date, conflicting results have been reported regarding the oncological role of KAL1. Decreased KAL1 expression is observed in colon, lung, and ovarian cancers compared with corresponding adjacent normal tissues (17). Conversely, KAL1 overexpression promotes brain tumor malignancy through integrin signal pathways and facilitates colon cancer cell migration and anti-apoptotic capacity (15). These studies indicate that KAL1 exhibits diverse functions in cancer initiation and progression. To the best of our knowledge, there have been no studies of expression analysis of KAL1 in HCC. Moreover, although loss-of-function mutations of the KAL1 gene have been known to underlie Kallmann syndrome (18,19), the significance of the methylation status of the KAL1 gene has yet to be determined.

In our previous microarray project exploring HCC-related tumor suppressors, we found that KAL1 was downregulated in HCC tissues (Log2 ratio: −2.1) (16,2022). Accordingly, we hypothesized that KAL1 might act as a putative tumor suppressor and mediate tumorigenesis of HCC. To systematically address this idea, we examined the expression and methylation status of KAL1 in HCC.

Materials and methods

Sample collection

We purchased nine HCC cell lines from the American Type Culture Collection (Manassas, VA, USA) and cultured cells as previously described (23). Primary HCC and adjacent liver tissues were collected from 144 patients who underwent hepatectomy for HCC at Nagoya University Hospital between January 1998 and January 2012. The ages of the 144 patients ranged from 34 to 84 years (median, 65.5 years), and the male-to-female ratio was 121:23. The median duration of patient follow-up was 40.1 months (range, 2.3–145 months). Thirty-seven were infected with hepatitis B and 80 patients were infected with hepatitis C virus. Ten patients had normal liver, 82 patients had chronic hepatitis, and 52 patients showed cirrhosis. Ninety, 37, and 17 patients were in stages I, II, and III, respectively.

Tissue samples were frozen immediately after resection and stored at −80°C until use

Genomic DNA and total RNA was extracted from both HCC and adjacent noncancerous tissues approximately 5 mm2 in diameter, avoiding necrotic areas. Specimens were classified histologically according to the 7th edition of the Union for International Cancer Control (24). Written informed consent for the use of clinical samples and data was obtained from all enrolled patients as required by the Institutional Review Board of Nagoya University, Japan.

Analysis of the KAL1 promoter region

Nucleotide sequencing was used to determine the presence of CpG islands in the KAL1 promoter region, defined as follows: ≥200 bp region with GC content >50% and an observed CpG/expected CpG ratio ≥0.6 (25). CpG Island Searcher software (http://cpgis-lands.usc.edu/) was used to determine the locations of CpG islands (26).

Quantitative real-time reverse transcription-polymerase chain reaction (qRT-PCR)

KAL1 mRNA levels were determined using qRT-PCR. Total RNA (10 μg) was isolated from nine HCC cell lines, 144 primary HCCs, and adjacent non-cancerous tissues and used as a template for complementary DNA synthesis. Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) mRNA (TaqMan, GAPDH control reagents, Applied Biosystems, Foster City, CA, USA) was quantified in each sample for standardization. Specific primers and annealing temperatures are listed in Table I. qRT-PCR was performed using the SYBR Green PCR Core Reagents kit (Applied Biosystems) as follows: one cycle at 95°C for 10 min, 40 cycles at 95°C for 5 sec, and 60°C for 60 sec. Real-time detection of SYBR Green fluorescence was conducted using an ABI StepOnePlus Real-Time PCR System (Applied Biosystems). All samples were analyzed in triplicate. The expression level of each sample is shown as the value of the KAL1 amplicon divided by that of GAPDH (27).

Table I

Primers and annealing temperatures.

Table I

Primers and annealing temperatures.

GeneExperimentTypeSequence (5′-3′)Product size (bp)Annealing temperature (°C)
KAL1qRT-PCRForward AACAATGGTTCCCTGGTTTG11060
Reverse TCACAAAAGCTTTGGCACTG
MSPForward GTGCGAACGGGAGAGGC10968
Reverse GTCAACTACGAACCCGAACG
U-MSPForward AAAACCCATAAACCAATCTCA12658
Reverse TGAATGGGAGAGGTGTTTGT
Bisulfite sequencingForward TATTGGGAGGGAGTTTGGGA41166
ReverseTAC TCC CCA CCC TCA AAC TA
EZRqRT-PCRForward GATAGTCGTGTTTTCGGGGA9160
Reverse CTCTGCATCCATGGTGGTAA
FAKqRT-PCRForward GCCAAAAGGATTTCTAAACCAG11064
Reverse CCTGGTCCACTTGATCAGCTA
SRCqRT-PCRForward CTGACCGCATGGACCGT10758
Reverse AAGCCAACCTGTCACTTGGTA
DPYSL3qRT-PCRForward AGAAGAAGGAGGGAGGGAGC11060
Reverse CTCCCTTGATAAGGAGACGG
GAPDHqRT-PCRForward GAAGGTGAAGGTCGGAGTC22660
Probe CAAGCTTCCCGTTCTCAGCC
Reverse GAAGATGGTGATGGGATTTC

[i] KAL1, Kallmann syndrome 1 sequence; EZR, ezrin; FAK, focal adhesion kinase; SRC, cellular SRC; DPYSL3, Dihydropyrimidinase-like 3; GAPDH, glyceraldehyde-3-phosphate dehydrogenase; qRT-PCR, quantitative real-time reverse-transcription polymerase chain reaction; MSP, methylation specific PCR; U-MSP, un-methylation specific PCR.

Methylation-specific PCR (MSP) and bisulfite sequence analysis

Genomic DNA samples from nine HCC cell lines and 144 HCC tissues were subjected to bisulfite treatment. MSP was conducted to determine the presence or absence of promoter hypermethylation of KAL1 gene. Bisulfite DNA from HCC cell lines was sequenced to determine the reliability of MSP results. Primer sequences are shown in Table I.

5-Aza-2′-deoxycytidine (5-aza-dC) treatment

To assess the relation of promoter hypermethylation to KAL1 transcription, HCC cell lines were treated with the DNA methylation inhibitor 5-aza-dC (Sigma-Aldrich, St. Louis, MO, USA) as previously described (10,28).

Expression of genes that encode cell adhesion factors

To identify cell adhesion proteins that may interact with KAL1, expression levels of Ezrin (EZR), focal adhesion kinase (FAK), cellular SRC (SRC) and dihydropyrimidinase-like 3 (DPYSL3) genes were determined by qRT-PCR in HCC cell lines (29,30). Primers specific for EZR, FAK, SRC and DPYSL3 are listed in Table I.

Immunohistochemical (IHC) staining

KAL1 protein localization was determined by IHC using 64 representative formalin-fixed and paraffin-embedded sections of well-preserved HCC tissue using a rabbit polyclonal antibody against KAL1 (ABN486, Millipore, Darmstadt, Germany) diluted 1:150 in antibody diluent (Dako, Glostrup, Denmark) as previously described (7,31). Samples were then washed with phosphate-buffered saline, followed by 10 min incubation with a biotinylated secondary antibody (Histofine SAB-PO(R), Nichirei, Tokyo, Japan). Sections were subsequently developed for 3 min using 3,3′-diaminobenzidine as substrate (Nichirei) and analyzed. To avoid bias, specimens were randomized, coded, and then analyzed by two independent observers who were uninformed of the identities of the samples.

Statistical analysis

The qualitative χ2 test and quantitative Mann-Whitney test were used to compare two groups. Correlations between mRNA levels of KAL1 and those of EZR, FAK, SRC, or DPYSL3 as well as tumor size and preoperative serum protein induced by vitamin K antagonists II (PIVKA-II) level were analyzed using the Spearman rank correlation test. Overall and disease-free survival rates were calculated using the Kaplan-Meier method, and the difference in survival curves was evaluated using the log rank test. A P-value <0.05 was considered statistically significant. All statistical analysis was performed using JMP 10® software (SAS Institute Inc., Cary, NC, USA).

Results

KAL1 mRNA expression and methylation status in HCC cell lines

The KAL1 gene harbors a CpG island around the promoter region (Fig. 1A), suggesting that hypermethylation of the CpG island may regulate KAL1 transcription. KAL1 mRNA expression levels were heterogeneous among nine HCC cell lines, regardless of differentiation (Fig. 1B). MSP revealed methylation in HLF, HuH1, HuH2, HuH7 and PLC/PRF/5 cells. When comparing the levels of KAL1 mRNA in HCC cell lines before and after demethylation by 5-aza-dC treatment, reactivation of KAL1 mRNA expression was observed in cells with promoter hypermethylation of the KAL1 gene (Fig. 1B). Direct sequence analysis revealed that all CpG sites in HuH2 cells (complete methylation) were CG (cytosine and guanine), whereas the corresponding positions in Hep3B cells (absence of methylation) were TG (thymine and guanine) (Fig. 2). These results confirm the accuracy of the MSP results.

Expression analysis of KAL1 and genes encoding putative functional partners in HCC cell lines

We next evaluated the expression levels of genes encoding other cell adhesion factors that could potentially functionally interact with KAL1. The relative expression levels of EZR, FAK, SRC, DPYSL3, and KAL1 mRNAs in HCC cell lines are shown in Fig. 3A. The results showed that KAL1 mRNA levels inversely correlated with those of EZR (correlation coefficient −0.667, P=0.049; Fig. 3B).

KAL1 status in surgically-resected tissues

We next examined KAL1 mRNA levels in 144 HCC tissues compared with the corresponding noncancerous liver tissues. Results showed that KAL1 mRNA levels were lower in HCC tissues compared with the corresponding noncancerous liver tissues in 106 (74%) of 144 patients. We next evaluated the association between expression levels of KAL1 mRNA and protein. Results of IHC, qPCR and MSP in representative patients are shown in Fig. 4A and B. One patient with reduced KAL1 mRNA levels showed reduced expression of KAL1 protein in the cytoplasm of HCC cells accompanied with promoter hypermethylation (Fig. 4A). Equivalent expression of KAL1 in cancer and normal cells was detected in a patient without downregulation of KAL1 mRNA and methylation (Fig. 4B). The expression patterns of KAL1 in 64 patients correlated significantly with those of KAL1 mRNA (P=0.023, Fig. 4C).

There were no significant differences in KAL1 mRNA levels between normal liver, chronic hepatitis, and cirrhosis in noncancerous liver tissues. In contrast, HCC tissues showed significantly decreased KAL1 mRNA levels compared with the corresponding noncancerous liver tissues (Fig. 5A). The KAL1 mRNA levels in HCCs correlated inversely with tumor size and preoperative serum PIVKA-II level (Fig. 5B). In 62 patients, KAL1 mRNA expression level in HCC was less than half of that in the corresponding noncancerous liver tissue, and these patients were categorized into the ‘downregulation of KAL1’ group for the following analyses. Downregulation of KAL1 was significantly associated with α-fetoprotein >20 ng/ml, PIVKA-II >40 mAU/ml, tumor size ≥3.0 cm, moderate to poor differentiation, formation of a capsule, vascular invasion, and hypermethylation of KAL1 (Table II).

Table II

Association between expression levels of KAL1 mRNA and clinicopathological parameters in 144 patients with hepato-cellular carcinoma (HCC).

Table II

Association between expression levels of KAL1 mRNA and clinicopathological parameters in 144 patients with hepato-cellular carcinoma (HCC).

Clinicopathological parametersDownregulation of KAL1 mRNA in HCCs (n=62)Others (n=82)P-value
Age0.312
 <65 year2540
 ≥65 year3742
Gender0.067
 Male5665
 Female617
Background liver0.679
 Normal liver37
 Chronic hepatitis3646
 Cirrhosis2329
Pugh-Child’s classification0.075
 A5579
 B73
Hepatitis virus0.329
 Absent1512
 HBV1423
 HCV3347
AFP (ng/ml)0.004a
 ≤202553
 >203729
PIVKA II (mAU/ml)0.002a
 ≤401642
 >404640
Tumor multiplicity0.928
 Solitary4864
 Multiple1418
Tumor size0.004a
 <3.0 cm1234
 ≥3.0 cm5048
Differentiation0.015a
 Well926
 Moderate to poor5356
Growth type0.126
 Expansive growth5565
 Invasive growth717
Serosal infiltration0.450
 Absent4564
 Present1718
Formation of capsule0.003a
 Absent1235
 Present5047
Infiltration to capsule0.066
 Absent2343
 Present3939
Septum formation0.370
 Absent1931
 Present4351
Vascular invasion0.033a
 Absent4167
 Present2115
Hypermethylation of KAL1 in HCCs0.019a
 Absent3258
 Present3024
UICC pathological stage0.062
 I3357
 II2215
 III710

{ label (or @symbol) needed for fn[@id='tfn2-ijo-46-06-2546'] } HBV, hepatitis B virus; HCV, hepatitis C virus; AFP, α-fetoprotein; PIVKA, protein induced by vitamin K antagonists; UICC, Union for International Cancer Control.

a P<0.05, statistically significant difference.

Impact of KAL1 mRNA expression on patient outcome

Patients with downregulation of KAL1 were more likely to have a shorter overall survival than other patients (5-year survival rates 51% and 78%, respectively, P=0.002) (Fig. 6A). In multivariate analysis, downregulation of KAL1 was identified as an independent prognostic factor (hazard ratio 2.04, 95% confidence interval 1.11–3.90, P=0.022; Table III). Additionally, patients with downregulation of KAL1 tended to have a shorter disease-free survival compared with other patients, although it did not reach statistical significance (3-year survival rates 32% and 50%, respectively, P=0.014) (Fig. 6B).

Table III

Prognostic factors of 144 patients with hepatocellular carcinoma (HCC) for overall survival.

Table III

Prognostic factors of 144 patients with hepatocellular carcinoma (HCC) for overall survival.

UnivariateMultivariate


VariablenHazard ratio95% CIP-valueHazard ratio95% CIP-value
Age (≥65)791.750.96–3.300.068
Gender (male)1211.820.78–5.290.178
Background liver (cirrhosis)521.530.84–2.750.161
Pugh-Child’s classification (B)101.680.50–4.190.360
AFP (>20 ng/ml)661.961.09–3.580.024a1.490.81–2.780.196
PIVKA II (>40 mAU/ml)861.901.03–3.710.041a1.040.50–2.060.909
Tumor multiplicity (multiple)321.830.94–3.380.073
Tumor size (≥3.0 cm)982.841.38–6.640.004a1.950.88–4.860.103
Tumor differentiation (well)350.720.34–1.410.349
Growth type (invasive growth)241.710.84–3.260.136
Serosal infiltration352.231.16–4.110.017a1.700.87–3.180.115
Formation of capsule970.950.52–1.810.861
Infiltration to capsule781.240.69–2.290.478
Septum formation940.770.43–1.430.402
Vascular invasion363.752.05–6.78<0.001a2.481.30–4.710.006a
Hypermethylation of KAL1 in HCCs541.230.67–2.330.511
Downregulation of KAL1 mRNA622.531.40–4.730.002a2.041.11–3.900.022a

{ label (or @symbol) needed for fn[@id='tfn4-ijo-46-06-2546'] } CI, confidence interval; AFP, a-fetoprotein; PIVKA, protein induced by vitamin K antagonists. Univariate analysis was performed using the log-rank test. Multivariate analysis was performed using the Cox proportional hazards model.

a P<0.05, statistically significant.

Discussion

Impaired expression of genes encoding ECM proteins plays an important role in the initiation and progression of HCC (16,32). KAL1, one of the ECM-related proteins, has been reported to have diverse oncological functions (15,17). In the present study, the clinical significance of the expression and methylation status of KAL1 was evaluated in HCC.

Consistent with earlier studies in colon, lung and breast cancer (17), our results showed that expression levels of KAL1 were reduced in HCC tissues compared to adjacent noncancerous liver tissues. Furthermore, KAL1 expression was independent of chronic inflammation or fibrosis of the background liver, suggesting that downregulation of KAL1 is a specific event in hepatocarcinogenesis or at later stages. Loss-of-function mutations in the KAL1 gene are responsible for Kallmann syndrome, a developmental disorder characterized by the association of hypogonadotropic hypogonadism and anosmia (14,18,19). However, no studies have investigated the regulatory mechanisms of KAL1 expression in malignancies. Since a CpG island was found in the promoter region of the KAL1 gene, we focused on aberrant DNA methylation, which is an important mechanism for inactivation of tumor suppressors (33,34). Our results showed that HCC cell lines with profoundly suppressed KAL1 expression also harbored promoter hypermethylation of the KAL1 gene, and expression levels of KAL1 were restored by demethylation. Additionally, there was a significant association between downregulation of KAL1 mRNA and hypermethylation of KAL1 gene in surgically resected HCC tissues. These findings implicated that aberrant methylation is a pivotal regulatory mechanism for KAL1 expression in HCC. Promoter hypermethylation of the KAL1 gene has the potential for becoming a novel biomarker of HCC as well as a therapeutic target for specific demethylation agents (34,35).

We also investigated the levels of other important ECM-related proteins, and found that the expression level of KAL1 had a significant inverse association with EZR expression. EZR is a cytoplasmic peripheral membrane protein that functions as a substrate of protein tyrosine kinases, regulates cellular survival, adhesion, migration, and invasion. Importantly, EZR is also one of the key factors involved in tumor progression and metastasis in HCC (3639). Our finding supports the notion that KAL1 may function through tumor suppressor mechanisms and led us to speculate that KAL1 may interact with EZR and mediate tumorigenesis of HCC.

The significant correlation between the IHC and qRT-PCR data allowed us to evaluate the prognostic significance of KAL1 mRNA levels in a quantitative manner. Downregulation of KAL1 mRNA in HCC was significantly associated with factors reflecting the malignant potential of HCC and consequently deteriorated patient outcomes after curative hepatectomy. In contrast to the previous study showing that KAL1 overexpression promotes brain tumor malignancy (15), our results instead support a tumor suppressive role for KAL1 in HCC.

KAL1 was first identified through its function in the development of gonadotropin-releasing hormone neurons. Previous studies demonstrated that the expression of KAL1 is modulated by FGFR1 and hypoxia inducible factor-1α (HIF-1α) (11,14). FGFR-1 expression was reported as low in normal liver epithelium and high in human liver cancer epithelium (40,41). FGFR-1 protein may be important in regulating cytoskeletal dynamics and function in cancer cell invasion and metastatic behavior (42). HIF-1α is an important transcription factor in essential adaptive responses to hypoxia, and plays a major role in the development of characteristic tumor phenotypes, including growth rate, angiogenesis, invasiveness, and metastasis, via activation of target genes by binding to hypoxia-responsive elements in the gene regulatory sequences (4345). The interactions with these major oncogenic pathways might provide a mechanism(s) underlying the correlation between KAL1 expression and malignant phenotype of HCC. Future studies, including pathway analysis in hepatocarcinogenesis, hypoxic stress and functional analysis, are required to elucidate the molecular mechanisms that underlie the biological function of KAL1 in HCC.

Taken together, our results indicate that KAL1 acts as a putative tumor suppressor in HCC that is inactivated by promoter hypermethylation. Our findings suggest that KAL1 may serve as a promising biomarker of malignant phenotype of HCC.

References

1 

Siegel R, Ward E, Brawley O and Jemal A: Cancer statistics, 2011: The impact of eliminating socioeconomic and racial disparities on premature cancer deaths. CA Cancer J Clin. 61:212–236. 2011. View Article : Google Scholar : PubMed/NCBI

2 

Galuppo R, Ramaiah D, Ponte OM and Gedaly R: Molecular therapies in hepatocellular carcinoma: What can we target? Dig Dis Sci. 59:1688–1697. 2014. View Article : Google Scholar : PubMed/NCBI

3 

Giannelli G, Rani B, Dituri F, Cao Y and Palasciano G: Moving towards personalised therapy in patients with hepatocellular carcinoma: The role of the microenvironment. Gut. 63:1668–1676. 2014. View Article : Google Scholar : PubMed/NCBI

4 

Kanda M, Nomoto S, Nishikawa Y, Sugimoto H, Kanazumi N, Takeda S and Nakao A: Correlations of the expression of vascular endothelial growth factor B and its isoforms in hepatocellular carcinoma with clinicopathological parameters. J Surg Oncol. 98:190–196. 2008. View Article : Google Scholar : PubMed/NCBI

5 

El-Serag HB: Hepatocellular carcinoma. N Engl J Med. 365:1118–1127. 2011. View Article : Google Scholar : PubMed/NCBI

6 

Sawan C, Vaissière T, Murr R and Herceg Z: Epigenetic drivers and genetic passengers on the road to cancer. Mutat Res. 642:1–13. 2008. View Article : Google Scholar : PubMed/NCBI

7 

Kanda M, Sugimoto H, Nomoto S, Oya H, Hibino S, Shimizu D, Takami H, Hashimoto R, Okamura Y, Yamada S, et al: B-cell translocation gene 1 serves as a novel prognostic indicator of hepatocellular carcinoma. Int J Oncol. 46:641–648. 2015.

8 

Hernandez-Gea V, Toffanin S, Friedman SL and Llovet JM: Role of the microenvironment in the pathogenesis and treatment of hepatocellular carcinoma. Gastroenterology. 144:512–527. 2013. View Article : Google Scholar : PubMed/NCBI

9 

Arzumanyan A, Reis HM and Feitelson MA: Pathogenic mechanisms in HBV- and HCV-associated hepatocellular carcinoma. Nat Rev Cancer. 13:123–135. 2013. View Article : Google Scholar : PubMed/NCBI

10 

Kanda M, Sugimoto H, Nomoto S, Oya H, Shimizu D, Takami H, Hashimoto R, Sonohara F, Okamura Y, Yamada S, et al: Clinical utility of PDSS2 expression to stratify patients at risk for recurrence of hepatocellular carcinoma. Int J Oncol. 45:2005–2012. 2014.PubMed/NCBI

11 

Soussi-Yanicostas N, de Castro F, Julliard AK, Perfettini I, Chédotal A and Petit C: Anosmin-1, defective in the X-linked form of Kallmann syndrome, promotes axonal branch formation from olfactory bulb output neurons. Cell. 109:217–228. 2002. View Article : Google Scholar : PubMed/NCBI

12 

Di Schiavi E and Andrenacci D: Invertebrate models of kallmann syndrome: Molecular pathogenesis and new disease genes. Curr Genomics. 14:2–10. 2013.PubMed/NCBI

13 

Liu J, Cao W, Chen W, Xu L and Zhang C: Decreased expression of Kallmann syndrome 1 sequence gene (KAL1) contributes to oral squamous cell carcinoma progression and significantly correlates with poorly differentiated grade. J Oral Pathol Med. 44:109–114. 2015. View Article : Google Scholar

14 

González-Martínez D, Kim SH, Hu Y, Guimond S, Schofield J, Winyard P, Vannelli GB, Turnbull J and Bouloux PM: Anosmin-1 modulates fibroblast growth factor receptor 1 signaling in human gonadotropin-releasing hormone olfactory neuroblasts through a heparan sulfate-dependent mechanism. J Neurosci. 24:10384–10392. 2004. View Article : Google Scholar : PubMed/NCBI

15 

Choy CT, Kim H, Lee JY, Williams DM, Palethorpe D, Fellows G, Wright AJ, Laing K, Bridges LR, Howe FA, et al: Anosmin-1 contributes to brain tumor malignancy through integrin signal pathways. Endocr Relat Cancer. 21:85–99. 2014. View Article : Google Scholar :

16 

Kanda M, Nomoto S, Okamura Y, Hayashi M, Hishida M, Fujii T, Nishikawa Y, Sugimoto H, Takeda S and Nakao A: Promoter hypermethylation of fibulin 1 gene is associated with tumor progression in hepatocellular carcinoma. Mol Carcinog. 50:571–579. 2011. View Article : Google Scholar : PubMed/NCBI

17 

Jian B, Nagineni CN, Meleth S, Grizzle W, Bland K, Chaudry I and Raju R: Anosmin-1 involved in neuronal cell migration is hypoxia inducible and cancer regulated. Cell Cycle. 8:3770–3776. 2009. View Article : Google Scholar : PubMed/NCBI

18 

Guioli S, Incerti B, Zanaria E, Bardoni B, Franco B, Taylor K, Ballabio A and Camerino G: Kallmann syndrome due to a translocation resulting in an X/Y fusion gene. Nat Genet. 1:337–340. 1992. View Article : Google Scholar : PubMed/NCBI

19 

Hardelin JP, Levilliers J, del Castillo I, Cohen-Salmon M, Legouis R, Blanchard S, Compain S, Bouloux P, Kirk J, Moraine C, et al: X chromosome-linked Kallmann syndrome: Stop mutations validate the candidate gene. Proc Natl Acad Sci USA. 89:8190–8194. 1992. View Article : Google Scholar : PubMed/NCBI

20 

Nomoto S, Kanda M, Okamura Y, Nishikawa Y, Qiyong L, Fujii T, Sugimoto H, Takeda S and Nakao A: Epidermal growth factor-containing fibulin-like extracellular matrix protein 1, EFEMP1, a novel tumor-suppressor gene detected in hepato-cellular carcinoma using double combination array analysis. Ann Surg Oncol. 17:923–932. 2010. View Article : Google Scholar

21 

Kanda M, Nomoto S, Oya H, Takami H, Hibino S, Hishida M, Suenaga M, Yamada S, Inokawa Y, Nishikawa Y, et al: Downregulation of DENND2D by promoter hypermethylation is associated with early recurrence of hepatocellular carcinoma. Int J Oncol. 44:44–52. 2014.

22 

Shimizu D, Kanda M, Nomoto S, Oya H, Takami H, Hibino S, Suenaga M, Inokawa Y, Hishida M, Takano N, et al: Identification of intragenic methylation in the TUSC1 gene as a novel prognostic marker of hepatocellular carcinoma. Oncol Rep. 31:1305–1313. 2014.

23 

Takami H, Kanda M, Oya H, Hibino S, Sugimoto H, Suenaga M, Yamada S, Nishikawa Y, Asai M, Fujii T, et al: Evaluation of MAGE-D4 expression in hepatocellular carcinoma in Japanese patients. J Surg Oncol. 108:557–562. 2013. View Article : Google Scholar : PubMed/NCBI

24 

Sobin LH, Gospodarowicz MK and Wittekind C: International Union Against Cancer, TNM Classification of Malignant Tumors. 7th edition. Wiley-Blackwell; New York: 2009

25 

Kanda M, Nomoto S, Oya H, Hashimoto R, Takami H, Shimizu D, Sonohara F, Kobayashi D, Tanaka C, Yamada S, et al: Decreased expression of prenyl diphosphate synthase subunit 2 correlates with reduced survival of patients with gastric cancer. J Exp Clin Cancer Res. 33:882014. View Article : Google Scholar : PubMed/NCBI

26 

Takai D and Jones PA: The CpG island searcher: A new WWW resource. In Silico Biol. 3:235–240. 2003.PubMed/NCBI

27 

Kanda M, Nomoto S, Oya H, Shimizu D, Takami H, Hibino S, Hashimoto R, Kobayashi D, Tanaka C, Yamada S, et al: Dihydropyrimidinase-like 3 facilitates malignant behavior of gastric cancer. J Exp Clin Cancer Res. 33:662014. View Article : Google Scholar : PubMed/NCBI

28 

Kanda M, Shimizu D, Nomoto S, Hibino S, Oya H, Takami H, Kobayashi D, Yamada S, Inokawa Y, Tanaka C, et al: Clinical significance of expression and epigenetic profiling of TUSC1 in gastric cancer. J Surg Oncol. 110:136–144. 2014. View Article : Google Scholar : PubMed/NCBI

29 

Kawahara T, Hotta N, Ozawa Y, Kato S, Kano K, Yokoyama Y, Nagino M, Takahashi T and Yanagisawa K: Quantitative proteomic profiling identifies DPYSL3 as pancreatic ductal adenocarcinoma-associated molecule that regulates cell adhesion and migration by stabilization of focal adhesion complex. PLoS One. 8:e796542013. View Article : Google Scholar : PubMed/NCBI

30 

Oya H, Kanda M, Sugimoto H, Shimizu D, Takami H, Hibino S, Hashimoto R, Okamura Y, Yamada S, Fujii T, et al: Dihydropyrimidinase-like 3 is a putative hepatocellular carcinoma tumor suppressor. J Gastroenterol. Aug 31–2014.(Epub ahead of print). PubMed/NCBI

31 

Kanda M, Shimizu D, Nomoto S, Takami H, Hibino S, Oya H, Hashimoto R, Suenaga M, Inokawa Y, Kobayashi D, et al: Prognostic impact of expression and methylation status of DENN/MADD domain-containing protein 2D in gastric cancer. Gastric Cancer. 18:288–296. 2015. View Article : Google Scholar

32 

Yam JW, Tse EY and Ng IO: Role and significance of focal adhesion proteins in hepatocellular carcinoma. J Gastroenterol Hepatol. 24:520–530. 2009. View Article : Google Scholar : PubMed/NCBI

33 

Maunakea AK, Nagarajan RP, Bilenky M, Ballinger TJ, D’Souza C, Fouse SD, Johnson BE, Hong C, Nielsen C, Zhao Y, et al: Conserved role of intragenic DNA methylation in regulating alternative promoters. Nature. 466:253–257. 2010. View Article : Google Scholar : PubMed/NCBI

34 

Khare S, Zhang Q and Ibdah JA: Epigenetics of hepatocellular carcinoma: Role of microRNA. World J Gastroenterol. 19:5439–5445. 2013. View Article : Google Scholar : PubMed/NCBI

35 

Miki D, Ochi H, Hayes CN, Aikata H and Chayama K: Hepato-cellular carcinoma: Towards personalized medicine. Cancer Sci. 103:846–850. 2012. View Article : Google Scholar : PubMed/NCBI

36 

Kang YK, Hong SW, Lee H and Kim WH: Prognostic implications of ezrin expression in human hepatocellular carcinoma. Mol Carcinog. 49:798–804. 2010.PubMed/NCBI

37 

Chen Y, Wang D, Guo Z, Zhao J, Wu B, Deng H, Zhou T, Xiang H, Gao F, Yu X, et al: Rho kinase phosphorylation promotes ezrin-mediated metastasis in hepatocellular carcinoma. Cancer Res. 71:1721–1729. 2011. View Article : Google Scholar : PubMed/NCBI

38 

Ghaffari A, Hoskin V, Szeto A, Hum M, Liaghati N, Nakatsu K, Madarnas Y, Sengupta S and Elliott BE: A novel role for ezrin in breast cancer angio/lymphangiogenesis. Breast Cancer Res. 16:4382014. View Article : Google Scholar : PubMed/NCBI

39 

Piao J and Liu S, Xu Y, Wang C, Lin Z, Qin Y and Liu S: Ezrin protein overexpression predicts the poor prognosis of pancreatic ductal adenocarcinomas. Exp Mol Pathol. 98:1–6. 2014. View Article : Google Scholar : PubMed/NCBI

40 

Ogasawara S, Yano H, Iemura A, Hisaka T and Kojiro M: Expressions of basic fibroblast growth factor and its receptors and their relationship to proliferation of human hepatocellular carcinoma cell lines. Hepatology. 24:198–205. 1996. View Article : Google Scholar : PubMed/NCBI

41 

Huang X, Yu C, Jin C, Kobayashi M, Bowles CA, Wang F and McKeehan WL: Ectopic activity of fibroblast growth factor receptor 1 in hepatocytes accelerates hepatocarcinogenesis by driving proliferation and vascular endothelial growth factor-induced angiogenesis. Cancer Res. 66:1481–1490. 2006. View Article : Google Scholar : PubMed/NCBI

42 

Wang J, Li J, Wang X, Zheng C and Ma W: Downregulation of microRNA-214 and overexpression of FGFR-1 contribute to hepatocellular carcinoma metastasis. Biochem Biophys Res Commun. 439:47–53. 2013. View Article : Google Scholar : PubMed/NCBI

43 

Zheng SS, Chen XH, Yin X and Zhang BH: Prognostic significance of HIF-1α expression in hepatocellular carcinoma: A meta-analysis. PLoS One. 8:e657532013. View Article : Google Scholar

44 

Wu L, Fu Z, Zhou S, Gong J, Liu CA, Qiao Z and Li S: HIF-1α and HIF-2α: Siblings in promoting angiogenesis of residual hepatocellular carcinoma after high-intensity focused ultrasound ablation. PLoS One. 9:e889132014. View Article : Google Scholar

45 

Yamada S, Utsunomiya T, Morine Y, Imura S, Ikemoto T, Arakawa Y, Kanamoto M, Iwahashi S, Saito Y, Takasu C, et al: Expressions of hypoxia-inducible factor-1 and epithelial cell adhesion molecule are linked with aggressive local recurrence of hepatocellular carcinoma after radiofrequency ablation therapy. Ann Surg Oncol. 21(Suppl 3): S436–S442. 2014. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

June-2015
Volume 46 Issue 6

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Tanaka Y, Kanda M, Sugimoto H, Shimizu D, Sueoka S, Takami H, Ezaka K, Hashimoto R, Okamura Y, Iwata N, Iwata N, et al: Translational implication of Kallmann syndrome-1 gene expression in hepatocellular carcinoma. Int J Oncol 46: 2546-2554, 2015
APA
Tanaka, Y., Kanda, M., Sugimoto, H., Shimizu, D., Sueoka, S., Takami, H. ... Kodera, Y. (2015). Translational implication of Kallmann syndrome-1 gene expression in hepatocellular carcinoma. International Journal of Oncology, 46, 2546-2554. https://doi.org/10.3892/ijo.2015.2965
MLA
Tanaka, Y., Kanda, M., Sugimoto, H., Shimizu, D., Sueoka, S., Takami, H., Ezaka, K., Hashimoto, R., Okamura, Y., Iwata, N., Tanaka, C., Yamada, S., Fujii, T., Nakayama, G., Koike, M., Nomoto, S., Fujiwara, M., Kodera, Y."Translational implication of Kallmann syndrome-1 gene expression in hepatocellular carcinoma". International Journal of Oncology 46.6 (2015): 2546-2554.
Chicago
Tanaka, Y., Kanda, M., Sugimoto, H., Shimizu, D., Sueoka, S., Takami, H., Ezaka, K., Hashimoto, R., Okamura, Y., Iwata, N., Tanaka, C., Yamada, S., Fujii, T., Nakayama, G., Koike, M., Nomoto, S., Fujiwara, M., Kodera, Y."Translational implication of Kallmann syndrome-1 gene expression in hepatocellular carcinoma". International Journal of Oncology 46, no. 6 (2015): 2546-2554. https://doi.org/10.3892/ijo.2015.2965