Forkhead box J1 expression is upregulated and correlated with prognosis in patients with clear cell renal cell carcinoma

  • Authors:
    • Pingyu Zhu
    • Yongrui Piao
    • Xiuzhe Dong
    • Zhehu Jin
  • View Affiliations

  • Published online on: June 16, 2015     https://doi.org/10.3892/ol.2015.3376
  • Pages: 1487-1494
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The forkhead box (FOX) family of transcription factors are considered to have a role in tumorigenesis. FOXJ1 is a member of the FOX family; however, its function in human renal cell carcinoma (RCC) has remained to be elucidated. Therefore, the present study evaluated the expression of FOXJ1 in human clear cell RCC and the effect of FOXJ1 on the proliferative ability of RCC cells. The RCC specimens analyzed in the present study were obtained from 286 patients with RCC who underwent nephrectomy. FOXJ1 mRNA expression levels were determined using reverse transcription‑quantitative polymerase chain reaction, and FOXJ1 protein expression levels were determined using immunohistochemistry and western blot analysis. To determine the effect of FOXJ1 on the proliferative ability of RCC cells, the expression of FOXJ1 was decreased using small interfering (si)RNA, and a FOXJ1 vector was stably transfected into RCC cell lines. The proliferative ability of RCC cells was then examined using a WST‑1 assay and xenograft experiments with BALB/c nude mice, where the association between FOXJ1 expression and patient survival was determined using Kaplan‑Meier analysis. FOXJ1 expression was significantly higher in RCC tissues compared with that of healthy renal tissues. Furthermore, FOXJ1 expression was associated with tumor stage, histologic grade and size. In addition, FOXJ1 significantly enhanced the proliferation of RCC cells in vitro and in vivo. The present study identified that FOXJ1 expression was upregulated in RCC and enhanced the proliferative ability of RCC cells. Therefore, FOXJ1 may serve as an independent prognostic marker and a therapeutic target for the treatment of patients with RCC.

Introduction

Human renal cell carcinoma (RCC) is the most common type of malignant kidney tumor in adults worldwide, and ~85% of RCCs are clear cell RCC (CCRCC) (1). RCC is regarded as a localized disease in the early stages, however, 30% of patients with RCC that present with localized disease at the time of diagnosis develop metastatic disease within three years (2). Furthermore, the prognosis for metastatic RCC is poor (3) as RCC is resistant to traditional chemotherapy (4,5) and alternative therapeutic strategies for advanced RCC are limited. At present, novel strategies for the treatment of advanced RCC include molecular targeted therapy (6), monoclonal antibodies (7), immunotherapy (8) and the suppression of signaling pathways (9). Although specific markers predicting the prognosis of advanced RCC and its potential therapeutic response to treatment have been investigated, the molecular mechanisms underlying the progression and development of RCC have remained elusive.

The forkhead box (FOX) family comprises numerous proteins with a wide spectrum of biological processes, including differentiation, metabolism, apoptosis, proliferation, migration and invasion. FOX proteins contain conserved transcriptional factors defined by a common DNA-binding domain (10). Furthermore, the FOX family is divided into 19 subclasses and consists of 50 genes in the human genome (11). Previous studies have determined that FOX proteins are associated with carcinogenesis and the progression of malignancies. For example, the expression of FOXM1 was increased in a variety of types of tumor, including basal cell and hepatocellular carcinoma, as well as lung, breast, prostate and colorectal cancer (1217). FOXM1 may be associated with carcinogenesis due to its role as a key regulator in the G1/S and G2/M phases of the cell cycle (1821). In addition, FOXO has been reported to be dysregulated in various types of tumor, including prostate and breast cancer, leukemia, glioblastoma and endometrial carcinoma (2228); FOXA1 was overexpressed in thyroid, lung and esophageal cancer (29,30); and FOXC2 appears to be a key gene involved in tumor progression and angiogenesis (31).

FOXJ1 is a transcription factor that is significant in the central nervous and reproductive systems (3234). Previous studies have demonstrated that abnormal expression of FOXJ1 is associated with autoimmune diseases and certain inflammatory diseases (35,36). This association appears to be due to the ability of FOXJ1 to suppress T cell activity, resulting in spontaneous autoimmunity (37). In addition, FOXJ1 inhibits the humoral immune response in B cells, with FOXJ1 deficiency in B cells being associated with germinal center formation and the development of autoantibodies (38). A previous study proposed that FOXJ1 expression was decreased in breast cancer, thus, functioning as a tumor suppressor gene (39). However, FOXJ1 expression was increased in hepatocellular carcinoma and was associated with poor prognosis. Furthermore, overexpression of FOXJ1 appears to be involved in proliferation and cell-cycle progression. In brief, little is known regarding the potential roles of FOXJ1 in carcinogenesis (40).

The expression of FOXJ1 and its function in human RCC is unclear. Therefore, the current study aimed to determine the expression of FOXJ1 in human RCC and its effect on the proliferative ability of human RCC cells.

Materials and methods

Patients and samples

The current study included 286 patients with RCC that had undergone radical nephrectomy in the Department of Urology of the Affiliated Hospital of Yanbian University (Yanji, China) between April 2002 and March 2003. The histological cell type of all specimen slices was determined by experienced pathologists and all samples were diagnosed as conventional CCRCC. The clinical tumor stages and characteristics were classified according to the tumor node metastasis (TNM) classification system (41), and the nuclear grade was evaluated according to the Fuhrman grading system of malignant tumors (42). RCC tissue samples and corresponding healthy kidney tissues located at a maximal distance from the tumor were collected immediately following surgical resection. The samples were formalin-fixed (Sigma-Aldrich, St. Louis, MO, USA), dehydrated and paraffin-embedded (Sigma-Aldrich). All tissue samples were maintained in liquid nitrogen (Sigma-Aldrich) prior to protein and RNA extraction. The patients were followed up every three months for a period of 120 months. The present study was approved by the Ethics Committee of the Affiliated Hospital of Yanbian University and written consent was obtained from all patients.

Immunohistochemistry

All paraffin-embedded tissue sections (4 µm) were deparaffinized in xylene (Sigma-Aldrich) and rehydrated. Subsequently, endogenous peroxidase activity was blocked by treatment with 0.4% hydrogen peroxide (Sigma-Aldrich) for 20 min followed by blocking with rabbit serum (Sigma-Aldrich) for 30 min. The sections were then incubated with primary FOXJ1 monoclonal mouse anti-rat antibody (cat. no. sc-53139; 1;1,000; Santa Cruz Biotechnology, Inc., Dallas, TX, USA) at 37°C for 1 h. The sections were washed with Tris buffer prior to incubation with biotinylated polyclonal goat anti-mouse antibody (cat. no. E0433; 1:2,000; Dako, Glostrup, Denmark) at 37°C for 2 h. Detection of the antibody reactions was performed using the standard streptavidin-biotin complex technique (43). The tissue sections were immunohistochemically examined under a light microscope (ZX-117M; Shenzhen Zhongxun Optics Instrument Co., Ltd., Shenzhen, China), with FOXJ1 expression semi-quantitatively determined according to staining intensity (−, negative; +, weak; ++, moderate; and +++, strong).

Western blot analysis

Western blot analysis was performed according to the manufacturer's instructions. Briefly, total protein was isolated from the CCRCC and healthy tissue samples using lysis buffer (Sigma-Aldrich) as previously described (44), and the total protein concentration was determined using a Bradford dye-binding protein assay (Bio-Rad Laboratories, Inc., Hercules, CA, USA). Subsequently, 10% SDS-PAGE (Bio-Rad Laboratories, Inc.) was performed. FOXJ1 monoclonal mouse anti-rat antibody (cat. no. sc-53139; 1;1,000; Santa Cruz Biotechnology, Inc.) was applied as the experimental antibody and anti-β-actin monoclonal mouse anti-human antibody (cat. no. ab6276; 1:5,000; Abcam, Cambridge, UK) was applied as a loading control at 37°C for 2 h. The immune complexes were evaluated using an enhanced chemiluminescence system (GE Healthcare Life Sciences, Chalfont, UK).

Reverse transcription-quantitative polymerase chain reaction (RT-qPCR)

Total RNA was isolated from the CCRCC and healthy kidney tissues using an illustra™ QuickPrep mRNA purification kit (GE Healthcare, Life Sciences), according to the manufacturer's instructions, and RT was performed using a First-Strand complementary (c)DNA synthesis kit (GE Healthcare Life Sciences). The PCR conditions were determined according to the manufacturer's instructions as follows: Denaturation at 95°C for 5 min, annealing for 30 cycles of 95°C for 30 sec, 60°C for 30 sec and 72°C for 1 min and extension at 72°C for 10 min. RT-qPCR was performed using LightCycler® FastStart DNA Master SYBR Green I (Roche Diagnostics GmbH, Indianapolis, USA) and the PCR products were detected by agarose gel electrophoresis, followed by quantification of the products using LightCycler (Roche Diagnostics GmbH). The primer sequences were as follows: FOXJ1 forward, 5′-TCGAGATGGCGGAGAGCTGG-3′ and reverse, 5′-GATCCCAAGAAGGCCCCCAC-3′; GAPDH forward, 5′-ATCAAGAAGGTGGTGAAGCAG-3′ and reverse, 5′-TGGAGGAGTGGGTGTCGC-3′. All RT-qPCR experimental procedures were conducted in accordance with Minimum Information for Publication of Quantitative Real-Time PCR Experiments guidelines (45).

Cell culture

Four RCC cell lines (Caki-1, NC65, ACHN, and A498) were purchased from the American Type Culture Collection (Manassas, VA, USA). The RCC cell lines were cultured in complete medium consisting of RPMI-1640 medium (Gibco Bio-Cult Diagnostics Ltd., Glasgow, Scotland, UK) supplemented with 25 mM HEPES, 2 mM glutamine, 10% heat-inactivated fetal bovine serum, 100 µg/ml streptomycin, 100 U/ml penicillin and 5% non-essential amino acids (all obtained from Sigma-Aldrich). All RCC cell lines were maintained as monolayers in 10-cm petri dishes (Corning Inc., Corning, NY, USA) and cultured in an incubator with a humidified atmosphere of 5% CO2 at 37°C.

RNA interference (RNAi) and transfection
RNAi

RCC cells were incubated in culture dishes with complete medium at 37°C until cell confluence reached 30–50%. Subsequently, the RCC cells were transfected with 50 ng/ml small interfering (si)RNA oligonucleotides against FOXJ1 using Lipofectamine® 2000 (Invitrogen Life Technologies, Carlsbad, CA, USA). The siRNA oligonucleotide sequences were designed using siDirect software (http://sidirect2.rnai.jp). Following incubation for 48 h, FOXJ1 expression was evaluated by RT-PCR.

Transfection

The coding sequence of normal human FOXJ1 was synthesized by RT-PCR using HK-2 (healthy kidney cell line) cDNA (American Type Culture Collection) as the substrate. The PCR products of FOXJ1 were then subcloned into the pcDEF3 vector (Sigma-Aldrich) as described previously (46). The expression vector containing full-length FOXJ1 cDNA was stably transfected into the four RCC cell lines using Lipofectamine 2000. G418 (Sigma-Aldrich) was used to select RCC cells successfully transfected with FOXJ1, and FOXJ1 expression was evaluated by RT-PCR.

Proliferative ability analysis

The effect of FOXJ1 on the proliferative ability of RCC cells was analyzed using a WST-1 assay. In brief, exponentially-growing RCC cells were obtained and seeded into a 96-well microtiter plate. Following incubation for 24, 48 and 72 h, 10 µl WST-1 (Roche Diagnostics GmbH, Penzberg, Germany) was added to each well, and incubated for an additional 2 h. The absorbance, which represents the cell count in each well, was examined using a microculture plate reader (Immunoreader NJ-2000; Japan Intermed Co., Ltd., Tokyo, Japan) at a wavelength of 450 nm.

RCC xenograft mouse models

Thirty BALB/c nude mice (age, 3–4 weeks; Affiliated Hospital of North Sichuan Medical College, Nanchong, China) were randomly divided into two groups (control and FOXJ1 vector groups). The mice were kept in pathogen-free conditions, at temperatures of 26–28°C and 30–40% humidity and were exposed to 12 h light/dark cycles with free access to food and water. A total of 4×108 RCC cells were administered via subcutaenous injection into the lumbar region of each mouse. All mice were observed continuously for five weeks and the volume of each tumor was measured once a week. Following five weeks, all mice were sacrificed under deep anesthesia and the final volume of each tumor was recorded. Tumor volumes (v) were calculated using the following formula: v = ab2π / 6, where a is the longest diameter and b is the longest perpendicular diameter.

Statistical analysis

Statistical calculations were performed using SPSS software (version 19.0; IBM SPSS, Armonk, NY, USA). All experiments were performed in triplicate and the results are presented as the mean ± standard deviation. Statistical significance was determined using a Student's t-test, and the χ2 test was performed to analyze the association between FOXJ1 expression and clinicopathological characteristics. In addition, survival curves were plotted using Kaplan-Meier analysis. P≤0.05 was considered to indicate a statistically significant difference.

Results

Patient characteristics

The present cohort included 192 male and 94 female patients (age range, 51–84 years; median age, 67 years), with a tumor diameter of 1–17 cm (median size, 4.6 cm). The TNM staging distribution was as follows: Stage I, 147 patients; stage II, 73 patients; stage III, 41 patients; and stage IV, 25 patients. In addition, the Fuhrman staging distribution was as follows: Grade I, 124 patients; grade II, 97 patients; and grade III, 65 patients (Table I). The presenting symptoms included hematuria (28 patients), flank pain (36 patients) and palpable masses (19 patients). RCC was an incidental finding during the routine examination of 108 patients. Furthermore, laboratory analysis indicated an elevated erythrocyte sedimentation rate in 64 patients at the time of diagnosis, while thrombocytopenia, erythrocytosis and anemia existed in four patients each. Forty-nine patients exhibited one or more concomitant diseases, including angina, urolithiasis, diabetes mellitus and valvular heart disease; 12 patients with CCRCC had previously been treated with radical nephrectomy on the contralateral side; and 29 patients exhibited metastatic CCRCC at the time of diagnosis.

Table I.

Association between characteristics of patients with CCRCC and FOXJ1 expression, detected using quantitative polymerase chain reaction and immunohistochemistry.

Table I.

Association between characteristics of patients with CCRCC and FOXJ1 expression, detected using quantitative polymerase chain reaction and immunohistochemistry.

FOXJ1 protein expression

CharacteristicnFOXJ1 mRNA expression, mean ± SDP-value++++++P-value
Kidney disease state <0.05 <0.05
  CCRCC2862.64±0.35   246111289
  Healthy2860.71±0.18 15697  33  0
Gender >0.05 >0.05
  Male1922.67±0.25   1642  7460
  Female  942.58±0.28     819  3829
Age, years >0.05 >0.05
  <601592.67±0.23   1334  6250
  ≥601272.61±0.24   1127  5039
Tumor size, cm <0.05 <0.05
  ≤71472.07±0.22   1853  5026
  >71393.25±0.33     6  8  6263
Histological gradea <0.05 <0.05
  I1241.85±0.18   2142  59  2
  II  972.76±0.27     317  3542
  III  653.98±0.36     0  2  1845
Tumor stage <0.05 <0.05
  I1472.07±0.22   1853  5026
  II  732.74±0.26     6  7  3822
III  413.47±0.32     0  1  1723
  IV  254.35±0.41     0  0     718

a Determined using the Fuhrman grading system. CCRCC, clear cell renal cell carcinoma; FOXJ1, forkhead box J1; SD, standard deviation.

FOXJ1 protein expression in RCC

FOXJ1 protein expression in human CCRCC and healthy kidney tissues was investigated by immunohistochemical analysis. FOXJ1 expression appeared to be increased in CCRCC tissues (Fig. 1A and B) compared with that of corresponding healthy kidney tissues (Fig. 1C and D). FOXJ1 staining was detected in the cytoplasm and nuclei of 262/286 CCRCC samples (91.6%), but in only 130/286 (45.4%) healthy kidney tissue samples. A significant association was detected between increased FOXJ1 protein expression levels and various clinicopathological characteristics using χ2 analysis, including advanced tumor stage, high histological grade and tumor size (P≤0.05). However, the other investigated characteristics, including gender and age, did not exhibit a significant association with FOXJ1 protein expression (P>0.05; Table I). These results indicate that FOXJ1 may be involved in the carcinogenesis and progression of human CCRCC.

Evaluation of FOXJ1 expression using RT-PCR, western blotting and RT-qPCR

To clarify the elevated FOXJ1 protein expression observed in CCRCC by immunohistochemistry, RT-PCR (Fig. 2A) and western blot analysis (Fig. 2B) were performed to determine FOXJ1 expression levels in human CCRCC and healthy kidney tissues. The relative level of FOXJ1 expression was analyzed by RT-qPCR with reference to an internal control (Fig. 2C). The results indicated that FOXJ1 expression was significantly increased in CCRCC tissue compared with that of corresponding healthy kidney tissues, and FOXJ1 was expressed at levels similar to those detected by immunohistochemistry. The results of four pairs of CCRCC and corresponding healthy kidney tissue samples are indicated in Fig. 2.

Effect of FOXJ1 on the proliferation of RCC cells

A pcDEF3 vector containing full-length FOXJ1 cDNA was stably transfected into Caki-1, NC65, ACHN and A498 cell lines. Additionally, FOXJ1 expression was suppressed using siRNA. Successful transfections were confirmed using RT-PCR, where FOXJ1 expression was markedly increased by the FOXJ1 vector insert and markedly decreased by siRNA (Fig. 3A). The effect of FOXJ1 on the proliferation of RCC cells was determined by performing a WST-1 assay. RCC cells expressing high levels of FOXJ1 exhibited a significantly increased proliferative ability compared with that of the control cells. By contrast, RCC cells expressing low levels of FOXJ1 exhibited lower proliferative ability compared with that of the control cells (Fig. 3B). The observed increase in proliferation associated with increased FOXJ1 expression was supported by identical results obtained from the in vivo xenograft investigations of BALB/c nude mice (Fig. 3C).

Prognostic significance of FOXJ1 expression

Due to the significant association identified between FOXJ1 expression, and clinical stage and pathologic grade in CCRCC, the present study aimed to determine whether FOXJ1 was able to be regarded as a prognostic marker in human CCRCC. In the current cohort, 15 patients succumbed to myocardial infarction and 11 patients succumbed to advanced malignant disease. Kaplan-Meier analysis was performed to calculate the association between FOXJ1 expression and survival in CCRCC. It was demonstrated that the survival time of patients with CCRCC significantly differed between the low and high FOXJ1 expression groups (P<0.05; Fig. 4). Furthermore, following 10 years of follow-up, it was determined that patients expressing immunohistochemically low levels of FOXJ1 (− and +) lived significantly longer compared with patients in whom immunohistochemical staining demonstrated high FOXJ1 expression (++ and +++). These results indicated that FOXJ1 expression may serve as an independent marker for predicting the prognosis of patients with CCRCC.

Discussion

Various members of the FOX family, including FOXM1, FOXO, FOXA1 and FOXC2, have been studied, with the results indicating that FOX sub-families may be important in the tumorigenesis and progression of certain carcinomas (10). However, the function of FOXJ1 in carcinogenesis has remained unclear. To date, the role of FOXJ1 has generated considerable attention in certain types of tumor, with a number of studies analyzing its expression in human tumors. For example, a recent study indicated that FOXJ1 expression was increased and associated with aggressive characteristics in hepatocellular carcinoma. Thus, FOXJ1 was proposed as a prognostic marker in patients with hepatocellular carcinoma (40). By contrast, a previous study proposed that FOXJ1 was decreased and may function as a tumor suppressor gene in breast cancer (39). The expression of FOXJ1 and its role in RCC has remained to be determined.

To the best of our knowledge, the current study was the first to investigate FOXJ1 expression in human RCC. FOXJ1 expression levels were determined in human CCRCC samples using RT-PCR, western blot analysis and RT-qPCR. These methods identified that FOXJ1 expression levels were similar to those detected by immunohistochemistry. Additionally, the current study revealed that FOXJ1 expression was significantly increased in CCRCC compared with that of healthy kidney tissues. Furthermore, the expression of FOXJ1 was significantly associated with tumor stage, histological grade and tumor size. These findings indicated that FOXJ1 may function as a significant gene that is key in the tumorigenesis and progression of CCRCC. In vitro and in vivo analysis of the effect FOXJ1 expression on RCC cell proliferation indicated that FOXJ1 significantly enhanced the proliferation of RCC cells. Similar results were detected in xenograft investigations using BALB/c nude mice. The present study also used Kaplain-Meier analysis to investigate the association between FOXJ1 expression and the survival of patients with CCRCC. The results indicated that high expression of FOXJ1 was associated with poor prognosis in patients with CCRCC. Thus, it was proposed that FOXJ1 may be considered as an oncogene and an independent marker for predicting prognosis in patients with RCC. In addition, the FOXJ1 gene may be important in the tumorigenesis of renal cancer in adults and high expression levels of FOXJ1 may accelerate the progression of human RCC. The effects of FOXJ1 observed in both CCRCC tissues and RCC cells indicate that the conclusions drawn from these results are likely to apply to RCC in general. Thus, future studies should analyze the detailed molecular mechanisms regulated by FOXJ1 in human RCC.

In conclusion, the current results indicate that FOXJ1 expression was increased in human RCC and that FOXJ1 enhanced the proliferation of RCC cells. These findings indicate that FOXJ1 is a significant gene that may be crucial in the tumorigenesis and progression of human RCC. Thus, silencing of FOXJ1 expression may present a novel treatment strategy for patients with RCC.

Acknowledgements

The present study was supported by grants from the China State Scholarship Fund (grant no. 201408220020) and the Yanbian University Science and Technology Development Item (grant no. 201259).

References

1 

Deng FM and Melamed J: Histologic variants of renal cell carcinoma: does tumor type influence outcome? Urol Clin North Am. 39:119–132. 2012. View Article : Google Scholar : PubMed/NCBI

2 

Athar U and Gentile TC: Treatment options for metastatic renal cell carcinoma: a review. Can J Urol. 15:3954–3966. 2008.PubMed/NCBI

3 

Jemal A, Siegel R, Ward E, Murray T, Xu J, Smigal C and Thun MJ: Cancer statistics, 2006. CA Cancer J Clin. 56:106–130. 2006. View Article : Google Scholar : PubMed/NCBI

4 

Yu DS, Chang SY and Ma CP: The expression of mdr-1-related gp-170 and its correlation with anthracycline resistance in renal cell carcinoma cell lines and multidrug-resistant sublines. Br J Urol. 82:544–547. 1998. View Article : Google Scholar : PubMed/NCBI

5 

Hartmann JT and Bokemeyer C: Chemotherapy for renal cell carcinoma. Anticancer Res. 19:1541–1543. 1999.PubMed/NCBI

6 

Sciarra A, Gentile V, Salciccia S, Alfarone A and Di Silverio F: New anti-angiogenic targeted therapy in advanced renal cell carcinoma (RCC): Current status and future prospects. Rev Recent Clin Trials. 3:97–103. 2008. View Article : Google Scholar : PubMed/NCBI

7 

Dalle S, Thieblemont C, Thomas L and Dumontet C: Monoclonal antibodies in clinical oncology. Anticancer Agents Med Chem. 8:523–532. 2008. View Article : Google Scholar : PubMed/NCBI

8 

Coppin C: Immunotherapy for renal cell cancer in the era of targeted therapy. Expert Rev Anticancer Ther. 8:907–919. 2008. View Article : Google Scholar : PubMed/NCBI

9 

Simpson D and Curran MP: Temsirolimus: In advanced renal cell carcinoma. Drugs. 68:631–638. 2008. View Article : Google Scholar : PubMed/NCBI

10 

Myatt SS and Lam EW: The emerging roles of forkhead box (Fox) proteins in cancer. Nat Rev Cancer. 7:847–859. 2007. View Article : Google Scholar : PubMed/NCBI

11 

Jackson BC, Carpenter C, Nebert DW and Vasiliou V: Update of human and mouse forkhead box (FOX) gene families. Hum Genomics. 4:345–352. 2010.PubMed/NCBI

12 

Wonsey DR and Follettie MT: Loss of the forkhead transcription factor FoxM1 causes centrosome amplification and mitotic catastrophe. Cancer Res. 65:5181–5189. 2005. View Article : Google Scholar : PubMed/NCBI

13 

Kim IM, Ackerson T, Ramakrishna S, et al: The Forkhead Box m1 transcription factor stimulates the proliferation of tumor cells during development of lung cancer. Cancer Res. 66:2153–2161. 2006. View Article : Google Scholar : PubMed/NCBI

14 

Lee JS, Chu IS, Heo J, et al: Classification and prediction of survival in hepatocellular carcinoma by gene expression profiling. Hepatology. 40:667–676. 2004. View Article : Google Scholar : PubMed/NCBI

15 

Teh MT, Wong ST, Neill GW, Ghali LR, Philpott MP and Quinn AG: FOXM1 is a downstream target of Gli1 in basal cell carcinomas. Cancer Res. 62:4773–4780. 2002.PubMed/NCBI

16 

Kalin TV, Wang IC, Ackerson TJ, et al: Increased levels of the FoxM1 transcription factor accelerate development and progression of prostate carcinomas in both TRAMP and LADY transgenic mice. Cancer Res. 66:1712–1720. 2006. View Article : Google Scholar : PubMed/NCBI

17 

Yoshida Y, Wang IC, Yoder HM, Davidson NO and Costa RH: The forkhead box M1 transcription factor contributes to the development and growth of mouse colorectal cancer. Gastroenterology. 132:1420–1431. 2007. View Article : Google Scholar : PubMed/NCBI

18 

Laoukili J, Kooistra MR, Brás A, et al: FoxM1 is required for execution of the mitotic programme and chromosome stability. Nat Cell Biol. 7:126–136. 2005. View Article : Google Scholar : PubMed/NCBI

19 

Costa RH: FoxM1 dances with mitosis. Nat Cell Biol. 7:108–110. 2005. View Article : Google Scholar : PubMed/NCBI

20 

Wang X, Kiyokawa H, Dennewitz MB and Costa RH: The Forkhead Box m1b transcription factor is essential for hepatocyte DNA replication and mitosis during mouse liver regeneration. Proc Natl Acad Sci USA. 99:16881–16886. 2002. View Article : Google Scholar : PubMed/NCBI

21 

Wang IC, Chen YJ, Hughes D, et al: Forkhead box M1 regulates the transcriptional network of genes essential for mitotic progression and genes encoding the SCF (Skp2-Cks1) ubiquitin ligase. Mol Cell Biol. 25:10875–10894. 2005. View Article : Google Scholar : PubMed/NCBI

22 

Accili D and Arden KC: FoxOs at the crossroads of cellular metabolism, differentiation and transformation. Cell. 117:421–426. 2004. View Article : Google Scholar : PubMed/NCBI

23 

Hu MC, Lee DF, Xia W, et al: IkappaB kinase promotes tumorigenesis through inhibition of forkhead FOXO3a. Cell. 117:225–237. 2004. View Article : Google Scholar : PubMed/NCBI

24 

Seoane J, Le HV, Shen L, Anderson SA and Massagué J: Integration of Smad and forkhead pathways in the control of neuroepithelial and glioblastoma cell proliferation. Cell. 117:211–223. 2004. View Article : Google Scholar : PubMed/NCBI

25 

Modur V, Nagarajan R, Evers BM and Milbrandt J: FOXO proteins regulate tumor necrosis factor-related apoptosis inducing ligand expression. Implications for PTEN mutation in prostate cancer. J Biol Chem. 277:47928–47937. 2002. View Article : Google Scholar : PubMed/NCBI

26 

Parry P, Wei Y and Evans G: Cloning and characterization of the t(X;11) breakpoint from a leukemic cell line identify a new member of the forkhead gene family. Genes Chromosomes Cancer. 11:79–84. 1994. View Article : Google Scholar : PubMed/NCBI

27 

Ward EC, Hoekstra AV, Blok LJ, et al: The regulation and function of the forkhead transcription factor, Forkhead box O1, is dependent on the progesterone receptor in endometrial carcinoma. Endocrinology. 149:1942–1950. 2008. View Article : Google Scholar : PubMed/NCBI

28 

Goto T, Takano M, Albergaria A, et al: Mechanism and functional consequences of loss of FOXO1 expression in endometrioid endometrial cancer cells. Oncogene. 27:9–19. 2008. View Article : Google Scholar : PubMed/NCBI

29 

Nucera C, Eeckhoute J, Finn S, et al: FOXA1 is a potential oncogene in anaplastic thyroid carcinoma. Clin Cancer Res. 15:3680–3689. 2009. View Article : Google Scholar : PubMed/NCBI

30 

Lin L, Miller CT, Contreras JI, et al: The hepatocyte nuclear factor 3 alpha gene, HNF3alpha (FOXA1), on chromosome band 14q13 is amplified and overexpressed in esophageal and lung adenocarcinomas. Cancer Res. 62:5273–5279. 2002.PubMed/NCBI

31 

Sano H, Leboeuf JP, Novitskiy SV, et al: The Foxc2 transcription factor regulates tumor angiogenesis. Biochem Biophys Res Commun. 392:201–206. 2010. View Article : Google Scholar : PubMed/NCBI

32 

Brody SL, Yan XH, Wuerffel MK, Song SK and Shapiro SD: Ciliogenesis and left-right axis defects in forkhead factor HFH-4-null mice. Am J Respir Cell Mol Biol. 23:45–51. 2000. View Article : Google Scholar : PubMed/NCBI

33 

Hackett BP, Brody SL, Liang M, Zeitz ID, Bruns LA and Gitlin JD: Primary structure of hepatocyte nuclear factor/forkhead homologue 4 and characterization of gene expression in the developing respiratory and reproductive epithelium. Proc Natl Acad Sci USA. 92:4249–4253. 1995. View Article : Google Scholar : PubMed/NCBI

34 

Clevidence DE, Overdier DG, Tao W, et al: Identification of nine tissue-specific transcription factors of the hepatocyte nuclear factor 3/forkhead DNA-binding-domain family. Proc Natl Acad Sci USA. 90:3948–3952. 1993. View Article : Google Scholar : PubMed/NCBI

35 

Li CS, Zhang Q, Lim MK, et al: Association of FOXJ1 polymorphisms with systemic lupus erythematosus and rheumatoid arthritis in Korean population. Exp Mol Med. 39:805–811. 2007. View Article : Google Scholar : PubMed/NCBI

36 

Li CS, Chae SC, Lee JH, Zhang Q and Chung HT: Identification of single nucleotide polymorphisms in FOXJ1 and their association with allergic rhinitis. J Hum Genet. 51:292–297. 2006. View Article : Google Scholar : PubMed/NCBI

37 

Srivatsan S and Peng SL: Cutting edge: Foxj1 protects against autoimmunity and inhibits thymocyte egress. J Immunol. 175:7805–7809. 2005. View Article : Google Scholar : PubMed/NCBI

38 

Lin L, Brody SL and Peng SL: Restraint of B cell activation by Foxj1-mediated antagonism of NF-kappa B and IL-6. J Immunol. 175:951–958. 2005. View Article : Google Scholar : PubMed/NCBI

39 

Demircan B, Dyer LM, Gerace M, Lobenhofer EK, Robertson KD and Brown KD: Comparative epigenomics of human and mouse mammary tumors. Genes Chromosomes Cancer. 48:83–97. 2009. View Article : Google Scholar : PubMed/NCBI

40 

Chen HW, Huang XD, Li HC, et al: Expression of FOXJ1 in hepatocellular carcinoma: correlation with patients' prognosis and tumor cell proliferation. Mol Carcinog. 52:647–659. 2013. View Article : Google Scholar : PubMed/NCBI

41 

Elmore JM, Kadesky KT, Koeneman KS and Sagalowsky AI: Reassessment of the 1997 TNM classification system for renal cell carcinoma. Cancer. 98:2329–2334. 2003. View Article : Google Scholar : PubMed/NCBI

42 

Hong SK, Jeong CW, Park JH, Kim HS, Kwak C, Choe G, Kim HH and Lee SE: Application of simplified Fuhrman grading system in clear-cell renal cell carcinoma. BJU Int. 107:409–415. 2011. View Article : Google Scholar : PubMed/NCBI

43 

Abourbih S, Sircar K, Tanguay S, Kassouf W, Aprikian A, Mansure J and Brimo F: Aldehyde dehydrogenase 1 expression in primary and metastatic renal cell carcinoma: an immunohistochemistry study. World J Surg Oncol. 11:2982013. View Article : Google Scholar : PubMed/NCBI

44 

Dutta KK, Nishinaka Y, Masutani H, et al: Two distinct mechanisms for loss of thioredoxin-binding protein-2 in oxidative stress-induced renal carcinogenesis. Lab Invest. 85:798–807. 2005. View Article : Google Scholar : PubMed/NCBI

45 

Bustin SA, Benes V, Garson JA, et al: The MIQE guidelines: Minimum information for publication of quantitative real-time PCR experiments. Clin Chem. 55:611–622. 2009. View Article : Google Scholar : PubMed/NCBI

46 

Goldman LA, Cutrone EC, Kotenko SV, Krause CD and Langer JA: Modifications of vectors pEF-BOS, pcDNA1 and pcDNA3 result in improved convenience and expression. Biotechniques. 21:1013–1015. 1996.PubMed/NCBI

Related Articles

Journal Cover

September-2015
Volume 10 Issue 3

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhu P, Piao Y, Dong X and Jin Z: Forkhead box J1 expression is upregulated and correlated with prognosis in patients with clear cell renal cell carcinoma. Oncol Lett 10: 1487-1494, 2015
APA
Zhu, P., Piao, Y., Dong, X., & Jin, Z. (2015). Forkhead box J1 expression is upregulated and correlated with prognosis in patients with clear cell renal cell carcinoma. Oncology Letters, 10, 1487-1494. https://doi.org/10.3892/ol.2015.3376
MLA
Zhu, P., Piao, Y., Dong, X., Jin, Z."Forkhead box J1 expression is upregulated and correlated with prognosis in patients with clear cell renal cell carcinoma". Oncology Letters 10.3 (2015): 1487-1494.
Chicago
Zhu, P., Piao, Y., Dong, X., Jin, Z."Forkhead box J1 expression is upregulated and correlated with prognosis in patients with clear cell renal cell carcinoma". Oncology Letters 10, no. 3 (2015): 1487-1494. https://doi.org/10.3892/ol.2015.3376