Prognostic significance of sodium-potassium ATPase regulator, FXYD3, in human hepatocellular carcinoma
- Authors:
- Published online on: December 21, 2017 https://doi.org/10.3892/ol.2017.7688
- Pages: 3024-3030
-
Copyright: © Wang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.
Abstract
Introduction
Hepatocellular carcinoma (HCC) is the 6th most common type of primary malignant neoplasm and the third leading cause of cancer-associated mortality worldwide. The morbidity and mortality rates of the disease increase annually (1–3). HCC forms heterogeneous tumors and its development is driven by numerous factors. Chronic hepatitis B virus (HBV) infection and HBV-associated cirrhosis account for the majority of cases of HCC, which constitutes 70–90% of liver cancer cases according to histological examination (4). The efficacies of existing key HCC therapies, including hepatectomy, percutaneous ethanol injection, radio frequency ablation and liver transplantation, have improved in recent years. However, the disease often progresses or recurs despite appropriate treatment regimes, and the overall survival (OS) of HCC patients remains poor (5). Therefore, novel molecular markers that contribute to the development of effective treatment strategies and the improved prognosis of patients with HCC are urgently required.
FXYD domain-containing ion transport regulator 3 (FXYD3), also known as mammary tumor 8, belongs to the FXYD protein family and is localized in the cell membrane and cytoplasm. It has been reported that FXYD3 acts as a regulator of sodium-potassium (Na+-K+) ATPase (6,7). One study demonstrated that this integral cell membrane protein was differentially expressed between tumor cells and normal cells, regulated cell proliferation, cell apoptosis and tumor metastasis, influenced the cell cycle, and participated in tumor angiogenesis and progression (8). However, the association between the expression of FXYD3 and tumor occurrence and development remains poorly understood. FXYD3 is expressed in numerous healthy tissues, including the uterus, stomach, colon and skin (9,10). However, FXYD3 expression has also been identified in various types of cancer, including breast cancer (7), prostate cancer (11,12), colorectal cancer (CRC) (13), pancreatic cancer (14–16), esophageal squamous carcinoma (ESCC) (17), lung cancer (18,19) and glioma (20). Furthermore, FXYD3 was demonstrated to be associated with the clinical prognoses of these types of cancer. To the best of our knowledge, the association between FXYD3 expression and HCC prognosis has not been previously evaluated. In the present study, the expression of FXYD3 in HCC tissues was investigated using reverse transcription-quantitative polymerase chain reaction (RT-qPCR), western blotting and immunohistochemical (IHC) staining analyses to determine the prognostic value of FXYD3 for patients with HCC.
Materials and methods
Study population
The present study was approved by the Research Ethics Committee of the Sun Yat-sen University Cancer Center (Guangzhou, China). All patients provided informed consent for study participation prior to undergoing tumor resection. Tissue samples were obtained from 217 patients diagnosed with HCC who had undergone curative resection at the Sun Yat-Sen University Cancer Center between August 2002 and November 2011. Among them, 193 were males and 24 were females, with age ranging from 20–75 years (median, 49 years). A total of 137 paraffin-embedded primary HCC tumor samples were selected (collected between January 2002 and June 2008). Furthermore, 80 pairs of HCC tissues and adjacent non-cancerous liver tissues were also selected from the Sun Yat-sen University Cancer Center specimen repository (originally collected between January 2009 and November 2011). A total of 6 paraffin-embedded breast cancer tissue samples from the Sun Yat-sen University Cancer Center specimen repository were also used in the present study as a positive control.
The following inclusion criteria were applied to the present study: Histologically verified primary HCC, no previous adjuvant chemotherapy, no diagnosis of distant metastases prior to surgery and access to complete follow-up data. Patient tissues were excluded from the study when there was evidence of Child-Pugh class B or C liver disease. Postoperative follow-up included regular recurrence and metastasis surveillance testing by measuring patient serum α-fetoprotein levels, abdominal ultrasonography, chest radiography, computed tomography, hepatic angiography and biopsies at 2–4-month intervals. Patients with confirmed disease recurrence received further treatment, including a second surgical resection. The median follow-up time was 43.9 months (range, 1–131 months). A total of 99 mortalities occurred during the follow-up period (45.6%) and 118 patients (54.4%) survived. OS was defined as the time from the date of surgery to the date of mortality from any cause, including HCC. Disease-free survival was defined as the time from the date of surgery to the date of regional relapse or distant metastasis, and was determined among the remaining patients on the date of their last follow-up appointment.
RNA extraction and RT-qPCR
Total RNA was extracted from 80 pairs of HCC tissues and matched non-cancerous liver tissues, using TRIzol solution (Invitrogen; Thermo Fisher Scientific, Inc., Waltham, MA, USA) according to the manufacturer's instructions. RNA concentrations were determined using a NanoDrop 2000 Spectrophotometer (Thermo Fisher Scientific, Inc.). RNA with an absorbance ratio from 1.8 to 2.0 at 260 and 280 nm was considered as pure. Each cDNA sequence was synthesized from 2 µg total RNA using a RevertAid First Strand cDNA Synthesis kit (Toyobo Life Science, Osaka, Japan). The resulting cDNA was processed by RT-qPCR to determine the relative FXYD3 mRNA expression levels. GAPDH served as an internal control. RT-qPCR was performed in triplicate at a final volume of 10 µl, consisting of 5 µl 2X SYBR Green master mix (Toyobo Life Science), 0.4 µl 20 mmol/l forward primer, 0.4 µl reverse primer, 0.75 µl sample cDNA and 3.45 µl RNase-free water. The reaction mixture was preheated to 95°C for 10 min, followed by 45 cycles of 95°C for 30 sec and 60°C for 60 sec. The following primers were used for RT-qPCR: FXYD3 forward, 5′-GGACGCCAATGACCTAGAAG-3′ and reverse, 5′-GGGTGATGAGAGGTGGAGTC-3′; and GAPDH forward, 5′-GGAGCGAGATCCCTCCAAAAT-3′ and reverse, 5′-GGCTGTTGTCATACTTCTCATGG-3′. The data were analyzed using the quantification cycle (2−ΔΔCq) method (21) and the results were averaged and expressed in relative expression units after normalization.
Protein extraction and western blotting
Western blotting was performed to analyze the FXYD3 protein expression levels in the clinical specimens from patients with HCC and the matched control tissues. Total protein was extracted from the fresh-frozen tissue samples using radioimmunoprecipitation assay lysis buffer (Beyotime Institute for Biotechnology, Haimen, China) according to the manufacturer's instructions. Protein concentrations were determined using a Bicinchoninic Acid medProtein Assay kit (Thermo Fisher Scientific, Inc.). A total of 30 µg protein per sample was separated by 12% SDS-PAGE and electro-transferred onto polyvinylidene fluoride membranes (Sigma-Aldrich; Merck KGaA, Darmstadt, Germany). The membranes were blocked in 5% skim milk in Tris-buffered saline-Tween for 60 min at room temperature. Subsequently, the membranes were incubated overnight at 4°C with a rabbit anti-FXYD3 monoclonal antibody (cat. no. ab205534; dilution 1:1,000; Abcam, Cambridge, UK) (22) and GAPDH (cat. no. 10494-1-AP; dilution 1:5,000; ProteinTech Group, Inc., Chicago, IL, USA). Subsequently, the membranes were incubated with appropriate horseradish peroxidase (HRP)-conjugated anti-rabbit IgG (cat. no. 7074; dilution 1:2,000; Cell Signaling Technology, Inc., Danvers, MA, USA) for 60 min at room temperature. The membranes were washed with TBS with 20% Tween-3 times and peroxidase activity was detected on X-ray films using an enhanced chemiluminescence detection system (Cell Signaling Technology, Inc.). The band intensities were measured by densitometry using Quantity One software v4.6.9 (Bio-Rad Laboratories, Inc., Hercules, CA, USA). The size of the FXYD3 band was consistent with the predicted molecular weight of the FXYD3 protein and was normalized to GAPDH protein levels.
IHC
Paraffin-embedded tissue sections, 4-µm thick, were cut and mounted on glass slides. The samples were subsequently deparaffinized in dimethylbenzene and rehydrated using 100, 95, 90, 80 and 70% ethanol solutions. Endogenous peroxidase activity was blocked using 0.3% hydrogen peroxide at room temperature for 10 min. For antigen retrieval, the slides were boiled in citrate-hydrochloric acid (pH 6.0) (cat. no. ZLI-9065; OriGene Technologies, Inc.) for 20 min in a pressure cooker. Subsequent to washing in PBS, the slides were treated with 5% sheep serum albumin (cat. no. ab7481; Abcam) at room temperature for 15 min to prevent non-specific binding. The tissue sections were then incubated with rabbit anti-FXYD3 (cat. no. ab205534; dilution 1:400; Abcam) overnight at 4°C. Following the primary antibody incubation, the tissue sections were incubated with an HRP-conjugated secondary antibody (Dako; Agilent Technologies, Inc., Santa Clara, CA, USA) for 30 min at room temperature. The slides were then incubated with 3,3′-diaminobenzidine tetrahydrochloride for 10 min at room temperature, all sections were counterstained with hematoxylin for ~2 min at room temperature. For the negative control, the primary antibody was replaced with rabbit IgG (cat. no. 3900; dilution 1:400; Cell Signaling Technology, Inc.) (data not shown). A positive control was provided by breast cancer tissue samples. The semi-quantitative FXYD3 expression score was calculated as the sum of the score for the proportion of positively stained tumor cells and the score for staining intensity. These scores were determined by two pathologists blinded to the clinical characteristics of the patients. The proportions of positively stained tumor cells were scored as follows: 0, <5% (negative); 1, 5–25% (sporadic); 2, 25–50% (focal); and 3, >50% (diffuse). Staining intensity was graded according to the following scale: 0, no staining; 1, weak staining (light yellow); 2, moderate staining (yellow-brown); and 3, strong staining (brown). The total immunostaining score was calculated by multiplying the proportion of positively stained cells by the staining intensity score, yielding a value ranging from 0–9.
Statistical analysis
Statistical analysis was performed using SPSS 20.0 software (IBM Corp., Armonk, NY, USA). Scatterplots and bar graphs were created using GraphPad Prism version 6.0 (GraphPad Software, Inc., La Jolla, CA, USA). The distributions of the baseline characteristics of the two groups, which were separated according to a cutoff FXYD3 level of 1.285, were compared using χ2 or Fisher's exact tests. The Kaplan-Meier method and log-rank test were used to plot survival curves and to calculate the difference in survival between the groups. Parameters found to be significant by univariate analysis were further evaluated by multivariate Cox regression analysis. P<0.05 was considered to indicate a statistically significant difference.
Results
Patients and clinicopathological data
The expression of FXYD3 was analyzed in each tissue sample at the mRNA and protein levels. FXYD3 expression in the 80 pairs of HCC tissues and matched non-cancerous liver tissues was analyzed by RT-qPCR, and 24 pairs were also evaluated by western blotting. Furthermore, 137 paraffin-embedded primary HCC tumor samples were analyzed by IHC analysis. The associations between patient clinicopathological data and FXYD3 expression levels in patients with HCC are summarized in Table I. χ2 tests demonstrated that FXYD3 expression levels were significantly different between patients with one vs. multiple tumors (P=0.011).
FXYD3 overexpression in HCC
RT-qPCR and western blotting were performed to analyze the expression of FXYD3 in HCC and non-cancerous liver tissue samples. This revealed that FXYD3 expression was significantly elevated in the HCC tissues compared with non-tumor tissues at the mRNA and protein levels (Fig. 1). Fig. 1B is a representative image of 3 replicates. The western blots of 5 paired samples (20.8%) showed significant protein expression and the remaining samples (79.2%) did not exhibit visible expression. IHC analysis of 137 HCC tissues was used to assess the function and prognostic significance of FXYD3 in HCC. Positive staining for FXYD3 was indicated by the presence of brown granules in the cell membrane and cytoplasm. The specimens were divided into a high FXYD3 expression group (n=60; 43.8%) and a low FXYD3 expression group (n=77; 56.2%), which exhibited staining ranging from strongly and moderately positive to weakly positive and negative, respectively (Fig. 2).
Prognostic significance of FXYD3 protein expression in HCC patients
Receiver operating characteristic (ROC) curve analysis was performed to determine the cutoff score for FXYD3 protein expression. The score closest to the point at which maximum sensitivity and specificity were displayed was selected as the cutoff score. The area under the ROC curve was calculated to estimate the power of FXYD3 to predict the OS rate over the entire range of scores. According to the ROC curve analysis results (Fig. 3A), the cohort was classified into a high expression group and a low expression group to evaluate the prognostic value of the FXYD3 in HCC patients.
The IHC results were used to evaluate the association between HCC clinicopathological parameters and FXYD3 expression. The cutoff score (1.285) generated by ROC curve analysis, was used for classification by high and low FXYD3 expression. Kaplan-Meier analysis and the log-rank test were performed to analyze the association between FXYD3 protein expression and OS. OS time was significantly higher in the group with low FXYD3 expression levels than in the group with high FXYD3 expression levels (P<0.001; Fig. 3B).
Univariate Cox regression analysis demonstrated that FXYD3 expression (P=0.001), tumor size (P=0.007), tumor number (P<0.001) and tumor stage (TNM) (P<0.001) were significant risk factors for OS (Table II). Multivariate Cox regression analysis indicated that FXYD3 expression (P=0.008), tumor size (P=0.027), and tumor number (P<0.001) were independent prognostic factors in patients with HCC (Table II).
Discussion
The predominant clinical staging system for liver cancer is the Barcelona Clinic Liver Cancer system, which is useful for predicting prognoses and stratifying patients for treatment (23). A series of predictive biomarkers, including meprin A subunit α, vascular endothelial growth factor and insulin-like growth factor-1, have facilitated improvements in the OS and prognosis for patients with HCC in the early and advanced stages of the disease when used in combination with the aforementioned staging system (24–26). The identification of additional novel predictive markers may significantly improve patient clinical outcomes.
A series of studies have reported that FXYD3 is expressed in numerous types of cancer. FXYD3 expression was initially described in breast cancer (7), and subsequently detected in prostate cancer, CRC, pancreatic cancer and ESCC, in which FXYD3 was reported to be significantly upregulated in tumor tissues compared with normal adjacent mucosal tissues (11–17). However, in lung cancer and glioma, FXYD3 was demonstrated to be expressed at low levels compared with healthy paracancerous tissues (18–20). FXYD3 was originally identified in murine mammary tumors. Morrison and Leder (6) determined that FXYD3 expression was induced by the Neu and Ras genes rather than the c-Myc and Int-2 genes. In human mammary epithelial cells, transforming growth factor-β has been demonstrated to cause downregulation of FXYD3 expression through the zinc finger E-box binding homeobox 1 pathway (27,28). In CRC, FXYD3 expression is hypothesized to be associated with p53 expression, as the FXYD3 promoter contains the p53 binding site. Increased CRC cell apoptosis, despite p53 mutations, has been demonstrated to be associated with small interfering FXYD3 (29,30). However, there is little knowledge regarding the association between FXYD3 expression and the clinical prognosis of HCC patients.
The Na+-K+ ATPase is composed of tetramers of α and β subunits and is located in the plasma membrane. The channel transports three Na+ ions out of the cell and two K+ ions into the cell to maintain cellular homeostasis (31). The Na+-K+ ATPase has been demonstrated to be associated with cancer initiation, growth, development and metastasis (32). It has also been demonstrated to function in the p38 mitogen-activated protein kinase/extracellular signal-regulated kinase signaling cascade, Src kinase activity, phosphoinositide 3-kinase/Akt/mechanistic target of rapamycin signaling and the epithelial-mesenchymal transition, all of which are important pathways in tumorigenesis and tumor progression (32). Furthermore, it has been reported that cardiac steroids (cardiac glycosides), which are potent Na+-K+ pump inhibitors, can postpone tumor recurrence and metastasis, prolong survival times and increase survival rates of post-surgical patients with HCC (33–36). FXYD3 can function as a β-subunit of the Na+-K+ ATPase and modulate certain cell functions. However, FXYD3 can also affect glycosylation of the β-subunit of X, K-ATPase when co-expressed in Xenopus oocytes (33–35). Glycosylation is closely associated with tumorigenesis (36,37); however, whether FXYD3-mediated glycosylation of Na+-K+ ATPase contributes to HCC growth, invasion and/or metastasis requires investigation in future studies.
FXYD3 expression was examined at the transcriptional and translational levels by RT-qPCR, western blotting and IHC. Using RT-qPCR, it was demonstrated that FXYD3 expression was significantly elevated in HCC tumor tissues compared with that in non-cancerous liver tissues. These results were supported by those achieved through western blotting. It is important to note that microvascular invasion is a key determinant of prognosis in patients with HCC. However, the present study indicated no significant association between FXYD3 expression and microvascular invasion using χ2 analysis. ROC curve analysis was performed to identify a cutoff value with which to classify patients into high or low FXYD3 expression groups. Using Kaplan-Meier analysis and the log-rank test, it was determined that FXYD3 expression is associated with OS at the protein level (P<0.001) and that high FXYD3 expression was predictive of a poor prognosis in patients with HCC. Univariate and multivariate Cox regression analyses indicated that FXYD3 protein expression level was an independent prognostic factor (P=0.008) in patients with HCC. However, it is possible that bias may have resulted in group-group differences in survival.
In conclusion, elevated FXYD3 mRNA and protein expression levels are predictive of poor survival. The present study partially clarifies the role of FXYD3 in HCC, however, the precise mechanism underlying the association between the expression of FXYD3 and HCC remains to be elucidated. Further large multicenter studies are required to achieve this.
Acknowledgements
This study was supported by the National Natural Science Foundation of China (grant nos. 81625017 and 81572385) and the Fundamental Research Funds for the Central Universities of China (grant no. 16ykjc36).
References
Forner A, Llovet JM and Bruix J: Hepatocellular carcinoma. Lancet. 379:1245–1255. 2012. View Article : Google Scholar : PubMed/NCBI | |
He G, Dhar D, Nakagawa H, Font-Burgada J, Ogata H, Jiang Y, Shalapour S, Seki E, Yost SE, Jepsen K, et al: Identification of liver cancer progenitors whose malignant progression depends on autocrine IL-6 signaling. Cell. 155:384–396. 2013. View Article : Google Scholar : PubMed/NCBI | |
Li Z, Tuteja G, Schug J and Kaestner KH: Foxa1 and Foxa2 are essential for sexual dimorphism in liver cancer. Cell. 148:72–83. 2012. View Article : Google Scholar : PubMed/NCBI | |
Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J and Jemal A: Global cancer statistics, 2012. CA Cancer J Clin. 65:87–108. 2015. View Article : Google Scholar : PubMed/NCBI | |
Maluccio M and Covey A: Recent progress in understanding, diagnosing, and treating hepatocellular carcinoma. CA Cancer J Clin. 62:394–399. 2012. View Article : Google Scholar : PubMed/NCBI | |
Morrison BW and Leder P: Neu and ras initiate murine mammary tumors that share genetic markers generally absent in c-myc and int-2-initiated tumors. Oncogene. 9:3417–3426. 1994.PubMed/NCBI | |
Morrison BW, Moorman JR, Kowdley GC, Kobayashi YM, Jones LR and Leder P: Mat-8, a novel phospholemman-like protein expressed in human breast tumors, induces a chloride conductance in Xenopus oocytes. J Biol Chem. 270:2176–2182. 1995. View Article : Google Scholar : PubMed/NCBI | |
Zhuang L, Xu L, Wang P, Jiang Y, Yong P, Zhang C, Zhang H, Meng Z and Yang P: Na+/K+-ATPase α1 subunit, a novel therapeutic target for hepatocellular carcinoma. Oncotarget. 6:28183–28193. 2015. View Article : Google Scholar : PubMed/NCBI | |
Capendeguy O and Horisberger JD: Functional effects of Na+, K+-ATPase gene mutations linked to familial hemiplegic migraine. Neuromolecular Med. 6:105–116. 2004. View Article : Google Scholar : PubMed/NCBI | |
Capurro C, Coutry N, Bonvalet JP, Escoubet B, Garty H and Farman N: Cellular localization and regulation of CHIF in kidney and colon. Am J Physiol. 271:C753–C762. 1996. View Article : Google Scholar : PubMed/NCBI | |
Vaarala MH, Porvari K, Kyllönen A and Vihko P: Differentially expressed genes in two LNCaP prostate cancer cell lines reflecting changes during prostate cancer progression. Lab Invest. 80:1259–1268. 2000. View Article : Google Scholar : PubMed/NCBI | |
Grzmil M, Voigt S, Thelen P, Hemmerlein B, Helmke K and Burfeind P: Up-regulated expression of the MAT-8 gene in prostate cancer and its siRNA-mediated inhibition of expression induces a decrease in proliferation of human prostate carcinoma cells. Int J Oncol. 24:97–105. 2004.PubMed/NCBI | |
Widegren E, Onnesjö S, Arbman G, Kayed H, Zentgraf H, Kleeff J, Zhang H and Sun XF: Expression of FXYD3 protein in relation to biological and clinicopathological variables in colorectal cancers. Chemotherapy. 55:407–413. 2009. View Article : Google Scholar : PubMed/NCBI | |
Iacobuzio-Donahue CA, Maitra A, Olsen M, Lowe AW, van Heek NT, Rosty C, Walter K, Sato N, Parker A, Ashfaq R, et al: Exploration of global gene expression patterns in pancreatic adenocarcinoma using cDNA microarrays. Am J Pathol. 162:1151–1162. 2003. View Article : Google Scholar : PubMed/NCBI | |
Friess H, Ding J, Kleeff J, Fenkell L, Rosinski JA, Guweidhi A, Reidhaar-Olson JF, Korc M, Hammer J and Büchler MW: Microarray-based identification of differentially expressed growth- and metastasis-associated genes in pancreatic cancer. Cell Mol Life Sci. 60:1180–1199. 2003. View Article : Google Scholar : PubMed/NCBI | |
Logsdon CD, Simeone DM, Binkley C, Arumugam T, Greenson JK, Giordano TJ, Misek DE, Kuick R and Hanash S: Molecular profiling of pancreatic adenocarcinoma and chronic pancreatitis identifies multiple genes differentially regulated in pancreatic cancer. Cancer Res. 63:2649–2657. 2003.PubMed/NCBI | |
Zhu ZL, Yan BY, Zhang Y, Yang YH, Wang MW, Zentgraf H, Zhang XH and Sun XF: Overexpression of FXYD-3 is involved in the tumorigenesis and development of esophageal squamous cell carcinoma. Dis Markers. 35:195–202. 2013. View Article : Google Scholar : PubMed/NCBI | |
Gordon GJ, Richards WG, Sugarbaker DJ, Jaklitsch MT and Bueno R: A prognostic test for adenocarcinoma of the lung from gene expression profiling data. Cancer Epidemiol Biomarkers Prev. 12:905–910. 2003.PubMed/NCBI | |
Okudela K, Yazawa T, Ishii J, Woo T, Mitsui H, Bunai T, Sakaeda M, Shimoyamada H, Sato H, Tajiri M, et al: Down-regulation of FXYD3 expression in human lung cancers: Its mechanism and potential role in carcinogenesis. Am J Pathol. 175:2646–2656. 2009. View Article : Google Scholar : PubMed/NCBI | |
Wang MW, Gu P, Zhang ZY, Zhu ZL, Geng Y, Kayed H, Zentgraf H and Sun XF: FXYD3 expression in gliomas and its clinicopathological significance. Oncol Res. 18:133–139. 2009. View Article : Google Scholar : PubMed/NCBI | |
Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI | |
Instructions of the Anti-FXYD3 antibody: Anti-FXYD3 antibody [EPR17160] ab205534. http://www.abcam.com/fxyd3-antibody-epr17160-ab205534.html | |
Llovet JM, Brú C and Bruix J: Prognosis of hepatocellular carcinoma: The BCLC staging classification. Semin Liver Dis. 19:329–338. 1999. View Article : Google Scholar : PubMed/NCBI | |
OuYang HY, Xu J, Luo J, Zou RH, Chen K, Le Y, Zhang YF, Wei W, Guo RP and Shi M: MEP1A contributes to tumor progression and predicts poor clinical outcome in human hepatocellular carcinoma. Hepatology. 63:1227–1239. 2016. View Article : Google Scholar : PubMed/NCBI | |
Kaseb AO, Hassan MM, Lin E, Xiao L, Kumar V, Pathak P, Lozano R, Rashid A, Abbruzzese JL and Morris JS: V-CLIP: Integrating plasma vascular endothelial growth factor into a new scoring system to stratify patients with advanced hepatocellular carcinoma for clinical trials. Cancer. 117:2478–2488. 2011. View Article : Google Scholar : PubMed/NCBI | |
Kaseb AO, Abbruzzese JL, Vauthey JN, Aloia TA, Abdalla EK, Hassan MM, Lin E, Xiao L, El-Deeb AS, Rashid A and Morris JS: I-CLIP: Improved stratification of advanced hepatocellular carcinoma patients by integrating plasma IGF-1 into CLIP score. Oncology. 80:373–381. 2011. View Article : Google Scholar : PubMed/NCBI | |
Yamamoto H, Mukaisho K, Sugihara H, Hattori T and Asano S: Down-regulation of FXYD3 is induced by transforming growth factor-β signaling via ZEB1/δEF1 in human mammary epithelial cells. Biol Pharm Bull. 34:324–329. 2011. View Article : Google Scholar : PubMed/NCBI | |
Herrmann P and Aronica SM: Estrogen and tamoxifen up-regulate FXYD3 on breast cancer cells: Assessing the differential roles of ER α and ZEB1. SpringerPlus. 4:2452015. View Article : Google Scholar : PubMed/NCBI | |
Bibert S, Aebischer D, Desgranges F, Roy S, Schaer D, Kharoubi-Hess S, Horisberger JD and Geering K: A link between FXYD3 (Mat-8)-mediated Na,K-ATPase regulation and differentiation of Caco-2 intestinal epithelial cells. Mol Biol Cell. 20:1132–1140. 2009. View Article : Google Scholar : PubMed/NCBI | |
Maxwell PJ, Longley DB, Latif T, Boyer J, Allen W, Lynch M, McDermott U, Harkin DP, Allegra CJ and Johnston PG: Identification of 5-fluorouracil-inducible target genes using cDNA microarray profiling. Cancer Res. 63:4602–4606. 2003.PubMed/NCBI | |
Kanai R, Ogawa H, Vilsen B, Cornelius F and Toyoshima C: Crystal structure of a Na+-bound Na+,K+-ATPase preceding the E1P state. Nature. 502:201–206. 2013. View Article : Google Scholar : PubMed/NCBI | |
Durlacher CT, Chow K, Chen XW, He ZX, Zhang X, Yang T and Zhou SF: Targeting Na+/K+-translocating adenosine triphosphatase in cancer treatment. Clin Exp Pharmacol Physiol. 42:427–443. 2015. View Article : Google Scholar : PubMed/NCBI | |
Crambert G, Li C, Claeys D and Geering K: FXYD3 (Mat-8), a new regulator of Na, K-ATPase. Mol Biol Cell. 16:2363–2371. 2005. View Article : Google Scholar : PubMed/NCBI | |
Yue Q, Zhen H, Huang M, Zheng X, Feng L, Jiang B, Yang M, Wu W, Liu X and Guo D: Proteasome inhibition contributed to the cytotoxicity of arenobufagin after its binding with Na, K-ATPase in human cervical carcinoma HeLa cells. PLoS One. 11:e01590342016. View Article : Google Scholar : PubMed/NCBI | |
Chen Z, Chen HY, Lang QB, Li B, Zhai XF, Guo YY, Yue XQ and Ling CQ: Preventive effects of jiedu granules combined with cinobufacini injection versus transcatheter arterial chemoembolization in post-surgical patients with hepatocellular carcinoma: A case-control trial. Chin J Integr Med. 18:339–344. 2012. View Article : Google Scholar : PubMed/NCBI | |
Hart GW and Copeland RJ: Glycomics hits the big time. Cell. 143:672–676. 2010. View Article : Google Scholar : PubMed/NCBI | |
Dalziel M, Crispin M, Scanlan CN, Zitzmann N and Dwek RA: Emerging principles for the therapeutic exploitation of glycosylation. Science. 343:12356812014. View Article : Google Scholar : PubMed/NCBI |