WT1 Promotes Invasion of NSCLC via Suppression of CDH1

WT1 Promotes Invasion of NSCLC via Suppression of CDH1

ORIGINAL ARTICLE WT1 Promotes Invasion of NSCLC via Suppression of CDH1 Chen Wu, PhD,* Weiyou Zhu, MD, PhD,* Jing Qian, MS,* Shaohua He, MS,* Changpi...

2MB Sizes 0 Downloads 22 Views

ORIGINAL ARTICLE

WT1 Promotes Invasion of NSCLC via Suppression of CDH1 Chen Wu, PhD,* Weiyou Zhu, MD, PhD,* Jing Qian, MS,* Shaohua He, MS,* Changping Wu, MD, PhD,‡ Yijiang Chen, MD, PhD,† and Yongqian Shu, MD, PhD* Introduction: The Wilms’ tumor gene (WT1) has been identified as an oncogene in many malignant diseases, and aberrant WT1 expression has been linked to development, progression, and prognosis of non–small-cell lung cancer (NSCLC). We sought to investigate the underlying mechanism of WT1 and metastasis in NSCLC. Methods: Real-time polymerase chain reaction was applied to detect WT1 and CDH1 mRNA in 159 NSCLC samples and corresponding adjacent tissues. Stable clones with overexpression and knockdown of WT1 were generated with plasmid and shRNA via lentivirus technology in H1568 and H1650 NSCLC cell lines. Wound-healing assay, transwell assays, and polymerase chain reaction array were carried out for invasion evaluation. Dual luciferase reporter assay was performed to validate the effect of WT1 on CDH1. Results: The level of the WT1 mRNA was negatively correlated with that of E-cadherin (CDH1) and associated with pathological stage, metastasis, and survival rate of 159 NSCLC patients. A series of genes were regulated by WT1, and WT1 could suppress CDH1 transcription via direct binding to its promoter and may enhance the invasive ability of H1568 and H1650 NSCLC cell lines. Conclusions: WT1 expression was correlated with clinical stage, metastasis, and survival rate in 159 NSCLC patients. Via direct binding to the promoter, WT1 could suppress CDH1 and promote NSCLC invasion. Key Words: WT1, CDH1, Non–small-cell lung cancer, Invasion, Metastasis. (J Thorac Oncol. 2013;8: 1163-1169)

L

ung cancer (including non–small-cell lung cancer [NSCLC] and small-cell lung cancer) remains the leading cause of cancer-related deaths in the world.1 Although great efforts have been made toward improving early diagnosis and effective treatment, including findings of new biomarkers,2–4 establishment of modified operation, and development of specific drugs,5–9 the prognosis is still poor. Departments of *Oncology, †Thoracic and Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, People’s Republic of China; and ‡Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, People’s Republic of China. Disclosure: The authors declare no conflict of interest. Address for correspondence: Yongqian Shu, MD, PhD, Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, No. 300, Guangzhou Road, Nanjing, 210029, People’s Republic of China. E-mail: [email protected] Copyright © 2013 by the International Association for the Study of Lung Cancer ISSN: 1556-0864/13/0809-1163

The most common features of malignancy are invasion and metastasis, which are characterized by the ability of cancer cells to invade into adjacent area, intravasate into blood or lymphatic vessels, and extravasate into a distant environment. Metastasis, which is primarily responsible for the low 5-year survival rates (approximately 10%–15%),10,11 is especially required for aggressive NSCLC, and once cancer cells lose contact with the neighboring cells, they become motile, invade the surrounding area, migrate, enter the circulation, extravasate into the target organ, proliferate, and metastasis. Therefore, the investigation of this area is important for patients with advanced NSCLC, but far from satisfaction to date. The Wilms’ tumor gene 1 (WT1) encodes a 49 to 52 kDa protein with an N-terminal domain involved in RNA/protein interactions12 and is an important regulator of cell growth and development in the embryo kidney, adult urogenital system, and central nervous system.13 The C-terminal domain harbors four zinc fingers, which permit binding to target DNA sequences for its transcriptional regulatory function.14 Through two alternative splicing motifs of exon 5 (17 amino acid) and exon 9 (3 amino acid, KTS region), four main protein isoforms of WT1 are designated as A (-/-), B (+/-), C (-/+), and D (+/+).15 A large number of genes coding for growth factors (e.g., TGF-β, CSF-1), growth factor receptors (e.g., insulin R, IGF-IR, EGFR), transcription factors (e.g., EGR, WT1, cMyc, Pax2, Dax-1, and Sry), and other proteins (e.g., ODC, MDR1, Hsp70, p21, Bcl-2) have been identified as WT1 target genes.16 Although first recognized as a tumor suppressor in Wlims’ tumor (nephroblastoma),17 based on accumulating evidence, WT1 has been demonstrated to act as an oncogene in other sorts of malignancies including leukaemia,18–20 breast cancer,21,22 and lung cancer23,24 et al., which suggested it acts in a dichotomous manner. The reason why WT1, the chameleon gene,25 plays the very opposite role still remains unclear. Studies have identified Sox9, Snail, and CDH1 as being deregulated by WT1 in WT1 conditional knockout mice, although available evidence for WT1 in direct transcriptional regulation is only available for Snail and CDH1, which was only reported in cardiovascular progenitor cells.26 The present study was designed to investigate the relationship between WT1 and its downstream molecule E-cadherin in NSCLC. We demonstrated that the expression of WT1 and E-cadherin (CDH1) were correlated and also associated with metastasis in NSCLC samples. We further showed that in vitro, WT1-D enhanced the invasiveness of H1568 and H1650 NSCLC cells via negative modulation of E-cadherin by directly bind to its promoter.

Journal of Thoracic Oncology  ®  •  Volume 8, Number 9, September 2013

1163

Journal of Thoracic Oncology  ®  •  Volume 8, Number 9, September 2013

Wu et al.

PATIENTS AND METHODS Patients and Tissue Samples The study was performed on a total of randomly selected 159 NSCLC patients, of whom 102 had adenocarcinomas and the remaining 57 had squamous cell carcinomas. The patients were treated at People’s Hospital of Jiangsu Province between September 2007 and August 2009, and all the specimens were collected under a protocol approved by the Human Ethics Committee of Nanjing Medical University. Each patient participated after providing informed consent. The median age of the patients was 64 years (range 49–85 years) and most of them were men (73%). A total of 318 tissue specimens were included in the study with one tumor sample and one corresponding adjacent sample for each patient. All specimens were histologically classified by a professional pathologist according to the national NCCN guidelines for NSCLC version 3.201127 using blind method. Survival rate was calculated using 36 months as cutoff point.

DNA and RNA Preparation Total RNA was extracted from tissue specimens using the TRIzol method (Invitrogen, Shanghai, China) and cDNA was synthesized using reverse transcriptase kit (TAKARA, Tokyo, Japan) according to the manufacturers’ protocol.

Real-Time Polymerase Chain Reaction for WT1 and CDH1 RNA Expression WT1 and E-cadherin (CDH1) mRNA levels were measured by real-time polymerase chain reaction (RT-PCR) using SYBR Premix Ex Taq (TAKARA). WT1 and CDH1 transcription values were normalized against the expression of β-actin. Amplification conditions, primers, and probes sequences for the WT1 and β-actin were from the work by Sitaram et al.28 and for CDH1 were the same as those in the work by Martínez-Estrada et al.26

Cell Culture H1568 (CRL-5876), H1650 (CRL-5883), and 293T (CRL-11268) cell lines from ATCC (Manassas, VA) were used for the present study. H1568 and H1650 were cultured in RPMI 1640 medium whereas 293T was cultured in Dulbecco’s Modified Eagle Medium (DMEM) high-glucose medium, both supplemented with 10% fetal bovine serum (Invitrogen, Carlsbad, CA). All cells were maintained in a humidified 37°C incubator with 5% CO2.

Lentivirus Production and Transduction WT1 gene was synthesized (purchased from GenScript, Nanjing, China) with restrictive digestion site of MluI in both ends, subcloned into pLV-GFP plasmid (gifted by Prof. Beicheng Sun, University of Nanjing Medical University, China), and named pLV-GFP-WT1. To generate plasmidexpressing WT1-shRNA, double-stranded oligonucleotides were cloned into pLL3.7 vector (gifted by D. Yun Chen, University of Nanjing Medical University, China) and named pLL3.7-WT1-shRNA. The sequences of WT1-shRNA

1164

used are aac TCAGGGTTACAGCACGGTC ttcaagaga GACCGTGCTGTAACCCTGA tttttt c. The uppercase letters represent WT1-specific sequence, and lowercase letters represent hairpin sequences. Recombinant lentivirus was generated from 293T cells using calcium phosphate precipitation. H1568 and H1650 were transfected with lentivirus using polybrene (8 µg/ml).

Western Blotting Assay Proteins were extracted from cultured cells, quantitated using a protein assay (bicinchoninic acid [BCA] method; Beyotime, Shanghai, China). Proteins were fractionated by sodium dodecyl sulfate polyacrylamide gel electrophoresis, transferred to polyvinylidene fluoride (PVDF) membrane, blocked in 4% dry milk at room temperature for 1 hour, and immunostained with primary antibodies at 4°C overnight using anti-WT1 (1:1000, 6F-H2; Millipore, Billerica, MA), anti-E-cadherin (1:1000; Abcam, Cambridge, United Kingdom), and β-actin (1: 20,000; Dako, Glostrup, Denmark). The results were visualized via a chemiluminescent detection system (Pierce ECL Substrate Western blot detection system; Thermo, Rockford, IL) and exposed in Molecular Imager ChemiDoc XRS System (Bio-Rad, Hercules, CA).

Cell-Proliferation Assay (Cell-Counting Kit-8) Cells were seeded into 96-well plates (6.0 × 103 cells per well). Cell viability was assessed by cell-counting kit-8 assay (Beyotime Institute of Biotechnology, Shanghai, China). The absorbance of each well was read on a spectrophotometer (Thermo) at 450 nm (A450). Three independent experiments were performed in quintuplicate.

Wound-Healing Assay Cells were seeded in six-well plates and cultured to confluence. Wounds of 2-mm width were created with a plastic scriber and the floating cells were washed away thrice with phosphate buffered saline (PBS). After incubation in a serumfree medium for 48 hours, cultures were observed and photos were taken under a microscope. A minimum of five randomly chosen areas was measured.

Transwell Invasion Assay The invasive ability of the cells was investigated using Transwells (8-µm pore size; Corning Costar Corp, Bedford, MA) put into the 24-well plates. First, 50 µl Matrigel (50 µg/ml; BD Biosciences, San Jose, CA) was added onto each surface of the chamber, incubated for 2 hours for solidification, then the supernatant was washed away with warm PBS. H1568 and H1650 were suspended in RPMI 1640 containing 2% fetal bovine serum. A total of 100 µl of the cell suspension (5 × 104 cells) was added to the upper chamber coated with Matrigel, and 400 µl of RPMI 1640 containing 10% fetal bovine serum was added to the lower compartment. After incubation for 48 hours at 37°C in a 5% CO2 humidified incubator, the Matrigel and cells on the upper surface of the filter were removed with cotton swabs and the cells that invaded into the lower surface were fixed with 2% paraformaldehyde, stained with crystal violet. Then the filters were removed from

Copyright © 2013 by the International Association for the Study of Lung Cancer

Journal of Thoracic Oncology  ®  •  Volume 8, Number 9, September 2013

the chambers, air-dried on the precleaned slides and applied with cover-slides using resina. Images were taken under an inverted microscope (Olympus Corp, Tokyo, Japan) at ×100 magnification over three random fields in each well. ImageJ 1.45s software (National Insititutes of Health, Bethesda, MD) was used for integrated optical density analysis. Each experiment was performed in triplicate.

RT-PCR Array Total RNA was extracted from H1568, H1568-WT1, and H1568-shWT1 and reverse transcribed to cDNA. Subsequently, cDNA was amplified by PCR using 23 Super Array PCR master mix (SuperArray Bioscience, Frederick, MD) and then RT-PCR was carried out using the Human Tumor Metastasis RT2 Profiler PCR array (SuperArray Bioscience) in an ABI PRISM7900 system (Applied Biosystems, Foster City, CA), according to the manufacturer’s instructions.

Luciferase Reporter Assay A dual-luciferase reporter assay was performed to investigate the transcriptional regulation of CDH1 by WT1. The promoter of CDH1-containing WT1 binding site (cgcccccac) was synthesized artificially and named control. The binding site is italicized and underlined. The promoter with the mutated binding site (gcgaaccga) was also synthesized and named mutant, as shown in Figure 3C. Products were validated via agarose gel electrophoresis, and cloned into the luciferase reporter vector (Promega, Madison, WI). WT1 gene was cloned into pcDNA3.1 plasmid (gifted by D. Beicheng Sun) and named pcDNA3.1-WT1. H1568 cells were seeded into 24-well plates and cotransfected with WT1-siRNA

WT1 Promotes Invasion of NSCLC

(pooled siGENOME SMART pool WT1 siRNA [50 nM per well]; Dhamacon, Chicago, IL) or pcDNA3.1-WT1, control siRNA (Dhamacon), and empty pcDNA3.1 as control, respectively, and luciferase reporter vectors using Lipofectamine 2000 (Invitrogen), following the instructions. Zero, 6, 12, and 24 hours after incubation, cells were collected and firefly and Renilla luciferase activity was measured with the dual-luciferase reporter assay system (Promega). All results were gained through three independent experiments.

Statistical Analysis Statistical analysis was performed using GraphPad Prism (version 5.01; GraphPad Software, Inc, La Jolla, CA) statistical software. The Student’s t test and paired t test were used to analyze significance between independent groups and paired materials, respectively. The correlation test was used to analyze the correlation between WT1 and CDH1. The χ2 test was used to test the significance of observed differences in proportions except when the cells size was less than 5 (Fisher’s exact tests). The significance was accepted as p value was less than 0.05.

RESULTS WT1 and CDH1 RNA levels were analyzed in NSCLC specimens and corresponding adjacent samples cut off from all 159 patients. Significantly higher WT1 (p < 0.0001; ­Fig. 1A) and lower CDH1 (p < 0.0001; Fig. 1B) RNA levels were detected in tumor specimens than in adjacent areas. Moreover, a negative correlation was observed between WT1 and CDH1 (R = −0.697; p < 0.0001; Fig. 1C) in tumor samples. The results suggested up-regulated expression of WT1 and down-regulation of CDH1 in NSCLC.

FIGURE 1.  Expression of WT1 and CDH1 transcripts and correlation between expressions of these two genes in non–small-cell lung cancer patients. A, Significantly higher RNA expression of WT1 in tumor specimens than in adjacent tissue samples (p < 0.0001). B, Significantly decreased RNA expression of CDH1 in tumor specimens in comparison with adjacent tumor-free tissues (p < 0.0001). C, A negative correlation was found between RNA expression of WT1 and CDH1 in tumor samples (R = −0.697; p < 0.0001). Copyright © 2013 by the International Association for the Study of Lung Cancer

1165

Wu et al.

Journal of Thoracic Oncology  ®  •  Volume 8, Number 9, September 2013

FIGURE 2.  WT1 promotes migration and invasion of NSCLC cells. The result of Transwell assay showed that the invasive ability of H1568 was significantly enhanced after WT1 overexpression (B versus A) and suppressed after WT1 silencing (C versus A) at a 48-hour time point, which was confirmed by integrated optical density (IOD) value evaluation (D). Similar effect of WT1 was observed in H1650 NSCLC cells (E–H). The result of wound-healing assay showed that migration ability of H1568 (I) and H1650 (J) cells were promoted by WT1 overexpression and restrained by WT1 silencing at 48-hour time point. The cell-proliferation assay test showed no distinct differences on proliferation after manipulation of WT1 in both H1568 (K) or H1650 cells (L) at 12-hour, 24-hour, and 48-hour time points. *p < 0.05. NSCLC, non–small-cell lung cancer; WT1, Wilms’ tumor gene.

A total of 159 NSCLC were divided into two groups using median RNA level of WT1 (median = 3.0013) and CDH1 (median = 0.4031) as threshold, respectively. The relationship of WT1 (CDH1) expression levels and clinical (pathological) characteristics were shown in Table1. Significant higher pathological stage (p < 0.0001), metastasis rate (p < 0.0001), and lower survival rate (p < 0.0001) were observed in both highWT1 and low-CDH1 expression groups, which supported the result of correlation analysis, indicating a potential functional link of WT1 and CDH1. Age, sex, and histologic diagnosis were not associated with WT1 or CDH1, as shown in Table1. In vitro, both H1568 and H1650 cells exhibited an invasive phenotype (Fig. 2A, E) that could be significantly enhanced when WT1 was overexpressed (Fig. 2B, F) and evidently suppressed when WT1 was down-regulated by shRNA (Fig. 2C, G), which was confirmed by integrated optical density value testing (Fig. 2D, H; p < 0.005). The results of wound-healing assay were consistent with those of the transwell assay, which proved that WT1 overexpression could enhance the migration and that down-regulation could suppress the process in both H1568 (Fig. 2I) and H1650 (Fig. 2J). WT1 overexpression or silencing could not affect cell proliferation (Fig. 2K, L), which, taken together, proved that the differences between the penetrated cells after WT1 plasmid or shRNA transfection were caused by variety of invasive rather than proliferative abilities.

1166

A variety of genes were screened out after WT1 overexpression and down-regulation via the PCR array of human metastasis pathway (Supplementary Table 1, Supplemental Digital Content 1, http://links.lww.com/JTO/A459) and nine of the genes whose variation larger than twofolds were picked and displayed in Figure 3A. CDH1 increased (4.09-fold) the most in H1568-shWT1 cells and NR4A3 decreased the most (−3.57fold) among these genes in H1568-WT1 cells. The Western blot assay confirmed the negative regulatory effect of WT1 on CDH1 (E-cadherin) in both H1568 and H1650 cell lines (Fig. 3B). To demonstrate the functional link between WT1 and CDH1, we performed luciferase reporter assay in H1568 cell lines with pcDNA3.1-WT1 plasmid and WT1 siRNA for transient transfection experiments. The luciferase activity representing CDH1 transcription was weakened after WT1 overexpression and, in contrast, enhanced after WT1 silencing, both with time increases (Fig. 3D, E). Moreover, no differences of luciferase activity were observed with mutated CDH1 promoter (Fig. 3F), suggesting a regulation function of WT1 on CDH1 through direct binding to its promoter.

Discussion Lung cancer is an aggressive malignancy, causing the most cancer-related deaths in the world.1 The best treatment for early-stage patients who are defined by the tumor, node,

Copyright © 2013 by the International Association for the Study of Lung Cancer

Journal of Thoracic Oncology  ®  •  Volume 8, Number 9, September 2013

WT1 Promotes Invasion of NSCLC

FIGURE 3.  CDH1 was a direct target of WT1. Via PCR array, nine metastasis-related genes varied larger than twofolds (including Cdh1) in H1568 cells after WT1 overexpression and silencing (A); Western blot assay validated the result of PCR array of negative trend of WT1 and E-cadherin in both H1568 and H1650 non–small-cell lung cancer cells (B); A schematic diagram showed the CDH1 promoter containing WT1 binding motif and its relationship with transcriptional start site (C); Luciferase reporter assay demonstrated that at 0-hour, 6-hour, 12-hour, and 24-hour time points, pcDNA3.1-WT1 down-regulated (D) and WT1-siRNA enhanced (E) Cdh1 transcription (versus empty plasmid) both in a time-dependent manner. In control group (wild-type CDH1 promoter), pcDNA-3.1-WT1 could inhibit and WT1-siRNA could enhance CDH1 transcription, but no regulatory function of WT1 in mutant group (mutant CDH1 promoter) were detected in H1568 (F). *p < 0.05. PCR, polymerase chain reaction; WT1, Wilms’ tumor gene.

metastasis (TNM) classification continues to be surgical resection, however, a considerable number of these patients will develop metastasis or relapse even when the surgeries are successful.29,30 The elucidation of the mechanisms of metastatis is crucial for improving surgery and treatment outcome, especially for selecting and customizing chemotherapy. Recently, numerous regulatory functions of WT1 have been reported and, most notable among these was that WT1 was found to be involved in inducing and mediating cardiovascular progenitor cell formation.26 In our study, we discovered a relationship of WT1 RNA level and pathological stage, metastasis and survival rate, found the association between WT1 and CDH1, and demonstrated that WT1 could promote the metastasis of NSCLC cells by direct regulation of CDH1. Furthermore, as reported, CDH1 may play a role in inducing

epithelial–mesenchymal transition (EMT) and mesenchymal–epithelial transition (MET), which are considered to be involved in migration, tumor invasion, and dissemination.31 Metastasis is a very complicated, polygene-involved, and multistage cascade. A variety of studies have revealed that EMT and MET processes were essential for metastasis.32–35 The balance of EMT and MET may be the decisive factor for original tumor to possess metastatic ability.36,37 Very recently, Martínez-Estrada et al.26 reported that WT1 was required for cardiovascular progenitor cell formation in which process EMT played important role. Miller-Hodges et al.38 summarized the role of WT1 in proper control of the mesenchymal–epithelial balance of cells. The role of Cdh1 in regulating metastasis and invasion of cancer cells have been demonstrated in many studies. Geiger et al.39 systemically concluded the structure,

Copyright © 2013 by the International Association for the Study of Lung Cancer

1167

Journal of Thoracic Oncology  ®  •  Volume 8, Number 9, September 2013

Wu et al.

TABLE 1.  Correlation between WT1 (or Cdh1) Status and Clinical (pathological) Characteristics in Patients with NSCLC

Characteristics n Age (yr)  Median (range) Sex  Male  Female Histology  Adenocarcinoma  Suqamous cell carcinoma Pathological stagec  IA  IB  IIA  IIB  IIIA  IIIB Metastasis  Yes  No Survival (rate)  Alive  Dead

WT1 High Expression (≥Mediana)

WT1 Low Expression (
79

80

64 (49–85)

64 (49–85)

63.5 (50–85)

116 43

59 (37.10%) 21 (13.20%)

57 (35.90%) 22 (13.80%)

102 57

52 (32.70%) 28 (17.60%)

50 (31.40%) 29 (18.20%)

34 44 30 30 10 11

6 (3.80%) 11 (6.90%) 24 (15.10%) 19 (11.90%) 9 (5.70%) 11 (6.90%)

28 (17.60%) 33 (20.80%) 6 (3.80%) 11 (6.92%) 1 (0.63%) 0 (0.00%)

46 113

38 (23.90%) 42 (26.42%)

8 (5.03%) 71 (44.65%)

79 80

17 (10.69%) 63 (39.62%)

62 (38.99%) 17 (10.69%)

All Patients 159

p

Cdh1 High Expression (≥Medianb)

Cdh1 Low Expression (
79

80

66 (50–85)

64 (49–84)

58 (36.48%) 23 (14.47%)

58 (36.48%) 20 (12.58%)

54 (33.96%) 27 (16.98%)

48 (30.19%) 30 (18.87%)

25 (15.72%) 31 (19.50%) 9 (5.70%) 11 (6.90%) 1 (0.63%) 1 (0.63%)

9 (5.66%) 13 (8.18%) 21 (13.20%) 19 (11.90%) 9 (5.90%) 10 (6.29%)

9 (5.90%) 69 (43.40%)

37 (23.27%) 44 (27.67%)

59 (37.11%) 19 (11.95%)

20 (12.58%) 61 (38.36%)

0.8427

p 0.8390

0.821

0.696

0.822

0.500

<0.0001

<0.0001

<0.0001

<0.0001

<0.0001

<0.0001

Median = 3.0013. Median = 0.4031. Pathological stage was based on NCCN Guidelines Version 3.2011 NSCLC. NCCN, National Comprehensive Cancer Network; NSCLC, non–small-cell lung cancer. a b c

function, and signal transduction of Cadherin and explained that it played a very important role in cell adhesion. Takeichi et al.40 proved that the perturbation of cadherin function causes temporary or permanent disaggregation of tumor cells and may thus promote their invasion and metastasis. Pinheiro et al.41 expounded that Cdh1 may initiate a variety of transcriptional events to regulate invasion and angiogenesis. According to our study, WT1’s functions on EMT, metastasis and invasion, are probably through Cdh1, the very classical invasion suppressor. However, although WT1 has a dichotomous function in either tissue development or tumor formation, the current studies could only illustrate the importance of WT1 in a tissuedependent manner. Similarly, although we tried to elucidate the mechanism of WT1 on metastasis and invasion within limits of NSCLC, the biological and pathological functions of WT1 in general, however, remain an unsolved mystery. A plethora of its target genes have been found since Knudson et al.42 first discovered, located, and named WT1 in 1972. As a transcriptional factor, via direct and indirect regulation, WT1 participates in nearly all types of biological processes. As summarized in Supplementary Table 1 (Supplemental Digital Content 1, http://links.lww.com/JTO/ A459) and illustrated in Figure 3A, genes that up-regulated in H1568-shWT1 cells were almost tumor metastasis inhibitors (e.g., CDH1) or suppressors (e.g., CD82 and MCAM) but

1168

those that were down-regulated were promoters of metastasis (e.g., FLT4 and MTA1) or proliferation (e.g., IGF1); accordingly, very contradictory results of these gene-expression levels were received in WT1 overexpressed H1568 cells, suggesting that changes in metastasis should not be attributed to alternations of one or a small quantity of genes. In summary, we identified the negative correlation of WT1 and CDH1 in our 159 NSCLC cases, both of which were associated with pathological stage, metastasis, and survival rate and proved the regulatory function of WT1 on CDH1 through direct binding of its promoter. As the limit on the number of NSCLC samples and cell types, more elaborate studies will be necessary for further exploration of the link between WT1 and CDH1 and its potential role in tumorigenesis and metastasis.

ACKNOWLEDGMENT

The authors thank Prof. Beicheng Sun and D. Yun Chen from University of Nanjing Medical University for providing plv-GFP, pcDNA 3.1, and pLL-3.7 plasmids. REFERENCES 1. Siegel R, Naishadham D, Jemal A. Cancer statistics, 2012. CA Cancer J Clin 2012;62:10–29.

Copyright © 2013 by the International Association for the Study of Lung Cancer

Journal of Thoracic Oncology  ®  •  Volume 8, Number 9, September 2013

2. Kuang P, Zhou C, Li X, et al. Proteomics-based identification of secreted protein dihydrodiol dehydrogenase 2 as a potential biomarker for predicting cisplatin efficacy in advanced NSCLC patients. Lung Cancer 2012;77:427–432. 3. Rosser CJ, Goodison S. CD24, a promising biomarker in NSCLC. Biomark Med 2010;4:495. 4. Tsao AS, Liu S, Lee JJ, et al. Clinical outcomes and biomarker profiles of elderly pretreated NSCLC patients from the BATTLE trial. J Thorac Oncol 2012;7:1645–1652. 5. Bradbury PA, Shepherd FA. Chemotherapy and surgery for operable NSCLC. Lancet 2007;369:1903–1904. 6. Tsao AS, Roth JA, Herbst RS. Surgery: future directions in multimodality therapy for NSCLC. Nat Rev Clin Oncol 2010;7:10–12. 7. van Meerbeeck JP. The controversial role of surgery in stage III NSCLC. Lancet Oncol 2008;9:607–608. 8. Dubey S, Schiller JH. Three emerging new drugs for NSCLC: pemetrexed, bortezomib, and cetuximab. Oncologist 2005;10:282–291. 9. Maione P, Gridelli C, Troiani T, Ciardiello F. Combining targeted therapies and drugs with multiple targets in the treatment of NSCLC. Oncologist 2006;11:274–284. 10. Sanchez de Cos J, Sojo Gonzalez MA, Montero MV, et al. Non-small cell lung cancer and silent brain metastasis. Survival and prognostic factors. Lung Cancer 2009;63:140–145. 11. Furák J, Troján I, Szöke T, et al. Lung cancer and its operable brain metastasis: survival rate and staging problems. Ann Thorac Surg 2005;79:241– 7; discussion 241. 12. Call KM, Glaser T, Ito CY, et al. Isolation and characterization of a zinc finger polypeptide gene at the human chromosome 11 Wilms’ tumor locus. Cell 1990;60:509–520. 13. Scharnhorst V, van der Eb AJ, Jochemsen AG. WT1 proteins: functions in growth and differentiation. Gene 2001;273:141–161. 14. Bruening W, Moffett P, Chia S, Heinrich G, Pelletier J. Identification of nuclear localization signals within the zinc fingers of the WT1 tumor suppressor gene product. FEBS Lett 1996;393:41–47. 15. Haber DA, Sohn RL, Buckler AJ, Pelletier J, Call KM, Housman DE. Alternative splicing and genomic structure of the Wilms tumor gene WT1. Proc Natl Acad Sci U S A 1991;88:9618–9622. 16. Ariyaratana S, Loeb DM. The role of the Wilms tumour gene (WT1) in normal and malignant haematopoiesis. Expert Rev Mol Med 2007;9:1–17. 17. Haber DA, Buckler AJ, Glaser T, et al. An internal deletion within an 11p13 zinc finger gene contributes to the development of Wilms’ tumor. Cell 1990;61:1257–1269. 18. Ogawa H, Tamaki H, Ikegame K, et al. The usefulness of monitoring WT1 gene transcripts for the prediction and management of relapse following allogeneic stem cell transplantation in acute type leukemia. Blood 2003;101:1698–1704. 19. Scheibenbogen C, Letsch A, Thiel E, et al. CD8 T-cell responses to Wilms tumor gene product WT1 and proteinase 3 in patients with acute myeloid leukemia. Blood 2002;100:2132–2137. 20. Cilloni D, Gottardi E, De Micheli D, et al. Quantitative assessment of WT1 expression by real time quantitative PCR may be a useful tool for monitoring minimal residual disease in acute leukemia patients. Leukemia 2002;16:2115–2121. 21. Tuna M, Chavez-Reyes A, Tari AM. HER2/neu increases the expression of Wilms’ Tumor 1 (WT1) protein to stimulate S-phase proliferation and inhibit apoptosis in breast cancer cells. Oncogene 2005;24:1648–1652.

WT1 Promotes Invasion of NSCLC

22. Oji Y, Miyoshi Y, Kiyotoh E, et al. Absence of mutations in the Wilms’ tumor gene WT1 in primary breast cancer. Jpn J Clin Oncol 2004;34:74–77. 23. Menssen HD, Bertelmann E, Bartelt S, et al. Wilms’ tumor gene (WT1) expression in lung cancer, colon cancer and glioblastoma cell lines compared to freshly isolated tumor specimens. J Cancer Res Clin Oncol 2000;126:226–232. 24. Oji Y, Miyoshi S, Takahashi E, et al. Absence of mutations in the Wilms’ tumor gene wt1 in de novo non-small cell lung cancers. Neoplasma 2004;51:17–20. 25. Huff V. Wilms’ tumours: about tumour suppressor genes, an oncogene and a chameleon gene. Nat Rev Cancer 2011;11:111–121. 26. Martínez-Estrada OM, Lettice LA, Essafi A, et al. Wt1 is required for cardiovascular progenitor cell formation through transcriptional control of Snail and E-cadherin. Nat Genet 2010;42:89–93. 27. Ward JH. NCCN Guidelines and the International Community. J Natl Compr Canc Netw 2011;9:133–134. 28. Sitaram RT, Degerman S, Ljungberg B, et al. Wilms’ tumour 1 can suppress hTERT gene expression and telomerase activity in clear cell renal cell carcinoma via multiple pathways. Br J Cancer 2010;103:1255–1262. 29. Johnston MR. The limits of surgical resection alone for non-small cell lung cancer. Lung Cancer 1997;17(Suppl 1):S99–102. 30. Van Schil PE. Surgery for non-small cell lung cancer. Lung Cancer 2001;34(Suppl 2):S127–S132. 31. Ye J, Wu D, Shen J, et al. Enrichment of colorectal cancer stem cells through epithelial-mesenchymal transition via CDH1 knockdown. Mol Med Rep 2012;6:507–512. 32. Tsai JH, Donaher JL, Murphy DA, Chau S, Yang J. Spatiotemporal regulation of epithelial-mesenchymal transition is essential for squamous cell carcinoma metastasis. Cancer Cell 2012;22:725–736. 33. Yan W, Cao QJ, Arenas RB, Bentley B, Shao R. GATA3 inhibits breast cancer metastasis through the reversal of epithelial-mesenchymal transition. J Biol Chem 2010;285:14042–14051. 34. Yang MH, Chang SY, Chiou SH, et al. Overexpression of NBS1 induces epithelial-mesenchymal transition and co-expression of NBS1 and Snail predicts metastasis of head and neck cancer. Oncogene 2007;26:1459–1467. 35. Zhang H, Meng F, Liu G, et al. Forkhead transcription factor foxq1 promotes epithelial-mesenchymal transition and breast cancer metastasis. Cancer Res 2011;71:1292–1301. 36. Thompson EW, Williams ED. EMT and MET in carcinoma—clinical observations, regulatory pathways and new models. Clin Exp Metastasis 2008;25:591–592. 37. Chen J, Han Q, Pei D. EMT and MET as paradigms for cell fate switching. J Mol Cell Biol 2012;4:66–69. 38. Miller-Hodges E, Hohenstein P. WT1 in disease: shifting the epithelialmesenchymal balance. J Pathol 2012;226:229–240. 39. Geiger B, Ayalon O. Cadherins. Annu Rev Cell Biol 1992;8:307–332. 40. Takeichi M. Cadherins in cancer: implications for invasion and metastasis. Curr Opin Cell Biol 1993;5:806–811. 41. Pinheiro H, Carvalho J, Oliveira P, et al. Transcription initiation arising from E-cadherin/CDH1 intron2: a novel protein isoform that increases gastric cancer cell invasion and angiogenesis. Hum Mol Genet 2012;21:4253–4269. 42. Knudson AG Jr, Strong LC. Mutation and cancer: a model for Wilms’ tumor of the kidney. J Natl Cancer Inst 1972;48:313–324.

Copyright © 2013 by the International Association for the Study of Lung Cancer

1169