Published OnlineFirst July 12, 2018; DOI: 10.1158/1541-7786.MCR-18-0392

Oncogenes and Tumor Suppressors Molecular Cancer Research Tumor Suppressor Activity of Selenbp1, a Direct Nkx2-1 Target, in Lung Adenocarcinoma Deborah R. Caswell1, Chen-Hua Chuang2, Rosanna K. Ma2, Ian P. Winters2, Eric L. Snyder3, and Monte M. Winslow1,2,4,5

Abstract

The Nkx2-1 promotes differentiation heterologous system, suggesting that these function in of lung epithelial lineages and suppresses malignant pro- a positive feedback loop. Selenbp1 inhibits clonal growth gression of lung adenocarcinoma. However, targets of Nkx2- and migration and suppresses growth of metastases in an in 1 that limit tumor growth and progression remain incom- vivo transplant model. Genetic inactivation of Selenbp1, pletely understood. Here, direct Nkx2-1 targets are identified using CRISPR/Cas9, also enhanced primary tumor growth whose expression correlates with Nkx2-1 activity in in autochthonous lung adenocarcinoma mouse models. human lung adenocarcinoma. Selenium-binding Collectively, these data demonstrate that Selenbp1 is a 1 (Selenbp1), an Nkx2-1 effector that limits phenotypes direct target of Nkx2-1, which inhibits lung adenocarcinoma associated with lung cancer growth and metastasis, was growth in vivo. investigated further. Loss- and gain-of-function approaches demonstrate that Nkx2-1 is required and sufficient for Implications: Selenbp1 is an important suppressor of lung Selenbp1 expression in lung adenocarcinoma cells. Inter- tumor growth that functions in a positive feedback loop with estingly, Selenbp1 knockdown also reduced Nkx2-1 expres- Nkx2-1, and whose loss is associated with worse patient sion and Selenbp1 stabilized Nkx2-1 protein levels in a outcome. Mol Cancer Res; 16(11); 1737–49. 2018 AACR.

Introduction Despite the experimental and clinical data underscoring the importance of NKX2-1 in lung adenocarcinoma, few direct Lineage-specific transcription factors have emerged as key reg- NKX2-1 targets that control these phenotypes have been identi- ulators of cancer growth, progression, and metastasis (1–7). fied and verified through functional studies. Myosin-binding Transcription factors related to Drosophila NK2 are key regulators protein H and occludin are both direct NKX2-1 targets that reduce of organ development and homeostasis (8, 9). In lung adeno- motility and metastatic ability (15, 16). However, additional carcinoma, several transcription factors have been identified that work is required to understand the subsets of NKX2-1 targets that influence tumorigenesis and metastatic ability, including the are critical for inhibiting tumor growth and progression in vivo. NK2-related transcription factor Nkx2-1 (3, 6, 7, Through a global analysis of putative direct Nkx2-1 target 10–12). In human lung adenocarcinoma and in genetically engi- genes, we identified selenium-binding protein 1 (Selenbp1/Sbp1) neered mouse models, Nkx2-1 expression is generally high in as an effector of the tumor growth–suppressive effect of Nkx2-1 in well-differentiated tumors but is downregulated in highly prolif- lung adenocarcinoma. Selenbp1 is highly expressed in several erative poorly differentiated tumor areas (10, 12, 13). Gain- and adult tissues, including the lung (17–20), and reduced SELENBP1 loss-of-function experiments suggest that Nkx2-1 can limit tumor expression is associated with poor patient outcome in many growth and metastatic ability (10, 12–14). Targeted inactivation human cancer types (18, 19, 21–25). We demonstrate that of Nkx2-1 in normal lung epithelial cells and early neoplasias Selenbp1 inhibits lung adenocarcinoma growth and migration increases proliferation (13). Thus, Nkx2-1 is a key regulator of and provide evidence that Selenbp1 and Nkx2-1 may exist in a multiple cancer-associated phenotypes. positive feedback loop. These results suggest that Selenbp1 is an important suppressor of lung tumor growth and progression.

1Cancer Biology Program, Stanford University School of Medicine, Stanford, California. 2Department of Genetics, Stanford University School of Medicine, Stanford, California. 3Department of Pathology and Huntsman Cancer Institute, Materials and Methods 4 University of Utah, Salt Lake City, Utah. Department of Pathology, Stanford Human expression analysis 5 University School of Medicine, Stanford, California. Stanford Cancer Institute, Human lung adenocarcinoma TCGA mRNA expression data Stanford University School of Medicine, Stanford, California. (26) were downloaded from cBioPortal, and correlations were Note: Supplementary data for this article are available at Molecular Cancer plotted using cBioPortal (27, 28). Correlations from the Director's Research Online (http://mcr.aacrjournals.org/). Challenge human lung adenocarcinoma microarray expression Corresponding Author: Monte M. Winslow, Stanford University School of data were plotted using Microsoft Excel (29). Analysis of lung Medicine, 279 Campus Drive, Beckman Center B256, Stanford, CA 94305. Phone: adenocarcinoma survival in KMPlotter (30) indicated that high 650-725-8696; Fax: 650-725-1534; E-mail: [email protected] SELENBP1 (Probe: 214422_s_at) predicts outcomes most signif- doi: 10.1158/1541-7786.MCR-18-0392 icantly in the lung adenocarcinoma subset of patients (Fig. 1H). 2018 American Association for Cancer Research. Data were split on the median.

www.aacrjournals.org 1737

Downloaded from mcr.aacrjournals.org on September 27, 2021. © 2018 American Association for Cancer Research. Published OnlineFirst July 12, 2018; DOI: 10.1158/1541-7786.MCR-18-0392

Caswell et al.

ChIP-seq analysis generated by transducing the cell lines with murine stem The chromatic immunoprecipitation sequencing (ChIP-seq) cell virus (MSCV) retrovirus containing tdTomato or CFP datasets were generated previously (13). ChIP-seq peaks with a fluorescent . Lentiviral vectors were used to knock peak significance above 10 6 were associated with genes using down Nkx2-1 in mouse cell lines with a previously verified GREAT (31). Peaks were assigned to genes if they were located short hairpin RNA (shRNA; ref. 12). Two hairpins (Sigma- 20,000 base pairs upstream or 1,000 base pairs downstream of the Aldrich) were validated for knockdown of Selenbp1 in mouse transcriptional start site (TSS). cell lines (TRCN0000101186, TRCN0000101188) and human cell lines (TRCN0000005702, TRCN0000005703). The MSCV Cell lines, lentiviral gene knockdown, and retroviral gene retroviral expression system was used to reexpress Nkx2-1. expression Maximilian Diehn's laboratory kindly shared the human lung The 394T4, 368T1, and 389T2 cell lines was generated from cancer cell line H441. Transfection of MSCV retroviral plasmids LSL-G12D/þ lung tumors in a lung adenocarcinoma–bearing Kras ; was used to express GFP, Nkx2-1, and Selenbp1 in 293T cells. flox/flox mouse and have been described (12, 32, 33). No Mycoplasma testing was performed on the cell lines, and cells The 394T4.Tom, 389T2.Tom and 389T2.CFP cell lines were were used for experiments within 3 to 5 days of thawing.

A B Lung adenocarcinoma C Lung adenocarcinoma 3,500 Sftpb (TCGA; n = 230) (TCGA; n = 230)

3,000 )

) 22 2 14 2 2,500 Atp11a 12 20 Tmem163 18

2,000 Cish 10 16 Selenbp1 8 14 Nkx2-1 1,500 (RSEM, log (RSEM, log 12 6 1,000 10 significance 4 8 Npc2 (-10*log(p-value)) 500 Napsa 2

C16orf89 6 Spearman: 0.57 Spearman: 0.63 Nkx2-1 ChIP Peak Nkx2-1 ChIP SFTPB 4 0 NKX2-1 1 Pearson: 0.60 Pearson: 0.68 0.40 0.50 0.60 0.70 1.00 2 6 8 10 12 14 6 8 10 12 14 NKX2-1 Correlation (Spearman) SELENBP1 (RSEM, log ) SELENBP1 (RSEM, log2) 2

D E F Lung adenocarcinoma (Shedden et al., n = 353) pos NKX2-1 14

) Pos Low/neg 2 12 (log

NKX2-1 H+E NKX2-1 SELENBP1 10 Pos 18 2 8 neg

SFTPB 6 Spearman: 0.52 SELENBP1 44 23

Pearson: 0.50 Low/neg 4

141210864 NKX2-1

SELENBP1 (log2)

G H Chr3 HR = 0.46 (0.36–0.59) 94,720 kb 94,730 kb 94,740 kb 0-75 1.0 Log-rank P = 1.1e−10 Nkx2-1 Input control 0-75 0-75 Foxa1/Foxa2 0.8 0-75

H3K4me1 0-75 H3K4me3 0.6 0-75 H3K27ac 0-75 0.4 H3K27me3 Selenbp1 Nkx2-1 Peaks 0.2 Selenbp1 Expression Nkx2-1 Motifs - High Probability of survival - Low 0.0 150100500 Times (months)

Figure 1. SELENBP1 is an NKX2-1 target that is coordinately expressed with NKX2-1 in human lung cancer. A, Selenbp1 has multiple proximal Nkx2-1 ChIP-seq peaks and SELENBP1 expression correlates with NKX2-1 expression in human lung adenocarcinoma. Each dot represents a ChIP-seq peak associated with the indicated gene. B and C, SELENBP1 expression correlates with NKX2-1 expression (B), and the expression of the NKX2-1 target gene surfactant protein b (SFTPB; C) in human lung adenocarcinoma. Each dot represents a tumor. D, SELENBP1 expression correlates with SFTPB expression in human lung adenocarcinoma. Each dot represents a tumor. E. Representative IHC staining of an NKX2-1pos and an NKX2-1neg human lung adenocarcinoma. Scale bar, 100 micrometer. F, SELENBP1 and NKX2-1 protein levels scored as low/negative (low/neg) or positive (pos) from the human lung adenocarcinoma TMA. The relationship between SELENBP1 and NKX2-1 is statistically significant (calculated using a 2 2 contingency). G, Lung adenocarcinoma patient outcome stratified by SELENBP1 expression. Analysis of 720 patients from GEO (Affymetrix microarrays), EGA, and TCGA (Szasz et al., 2016). P value and HR are indicated. H, ChIP-seq data for Nkx2-1, Foxa1/Foxa2, and histone marks around Selenbp1 in murine lung tumors.

1738 Mol Cancer Res; 16(11) November 2018 Molecular Cancer Research

Downloaded from mcr.aacrjournals.org on September 27, 2021. © 2018 American Association for Cancer Research. Published OnlineFirst July 12, 2018; DOI: 10.1158/1541-7786.MCR-18-0392

Selenbp1, Nkx2-1 Target, Suppresses Lung Cancer

Histologic preparation and IHC membrane. Membranes were immunoblotted using primary Tissues were fixed in 4% formalin overnight and transferred to antibodies. 70% ethanol until paraffin embedding. IHC was performed on The following antibodies were used to assess protein levels: 4-mm section with the ABC Vectastain Kit (Vector Laboratories) Selenbp1: Abcam: ab90135 using the Cadenza system followed by hematoxylin counterstain- Nkx2-1: Abcam: ab76013 (EP1584Y) ing. The following primary antibodies were used: Nkx2-1 (Abcam: Hsp90: BD Transduction Laboratories: 610419 ab76013), Selenbp1 (Abcam: ab90135), RFP (Rockland: GFP: Cell Signaling Technology: 2956 600401379). Sections were developed with DAB and counter- Primary antibody incubations were followed by secondary stained with hematoxylin. For in vivo BrdU labeling, mice were HRP-conjugated anti-mouse (Santa Cruz Biotechnology, injected intraperitoneally with 50 mg/kg of BrdU 24 hours before sc-2005) and anti-rabbit (Santa Cruz Biotechnology, sc-2004) analysis. The number of BrdU-positive cancer cells was quantified antibodies, and membranes were developed with ECL 2 Western by IHC and direct counting, taking into account the morphologic blotting substrate (P180196, Thermo Fisher Scientific, Pierce). feature of cancer cells and excluding areas directly adjacent to necrotic areas. The lung cancer tissue microarray (TMA) LC20815, BrdU cell culture assay containing 208 human patient cores (85 lung adenocarcinoma) To assess proliferation, 105 cells were plated in each well of a was obtained from US Biomax, Inc., and stained for NKX2-1 and 6-well plate. Twelve hours later, the subconfluent cells were SELENBP1 using IHC. labeled with 10 mmol/L BrdU for 2 hours, followed by anti-BrdU staining using the BD APC Flow Kit according to the manufac- Low-density plating turer's instructions. A total of 3 103 cells were seeded per 10-cm plate. Mouse cell lines were allowed to grow for one week before quantification. Migration assay For migrations assays, cells were plated in triplicate in 10-cm Low-density plating quantification plates, overnight such that confluency was reached the next day. A ImageJ was used to analyze colony number. Each image was 200 mL pipette tip was used to initiate the scratch. The gap was changed to 8-bit greyscale; then, the threshold was adjusted to 180 monitored over time and the gap measurement/analysis was pixels. Under the process menu, the image was changed to binary quantified using ImageJ. and then converted to mask. The area for counting was circled with Migration assays were also performed in the presence of mito- freehand selections. The particles were analyzed with a size limit mycin C by seeding 1 106 cells in each well of a 6-well plate of 150 pixels to infinity. overnight. Two hours prior to the scratch assay, 5 mg/mL of mitomycin C was added to the media. A 200 mL pipette tip was RNA-seq analysis used to initiate the scratch. The gap was monitored over time, and G12D RNA-seq data generated previously from a Kras ;p53- the gap measurement/analysis was quantified using ImageJ. deficient;tdTomato-expressing (KPT) tumors (33). These data were aligned with genes identified from ChIP-seq data using a python Presto blue assay script, and genes with a log2 fold change larger than 2 (genes that Proliferation was assessed using PrestoBlue Cell Viability are downregulated in metastasis) were identified. Reagent (A13261, Invitrogen), with 2 103 cells seeded per well of a 96-well plate. Quantitative PCR qRT-PCR was performed using SYBR green according to stan- Cell transplantation and analysis dard protocols. Samples were run in triplicate. Mouse samples Six-week-old male B6129SF1/J mice (Jax stock #101043) of were normalized to Gapdh. Human samples were normalized to similar weights were used for the transplantation experiments. For ACTIN. The following primers were used to assess RNA levels: intravenous transplantation, 1 105 cells were injected into the lateral tail vein (in 200 mL PBS) of B6129SF1/J recipient mice. Mouse Nkx2-1: forward 50 gatggtacggcgccaacccag 30 and reverse Mice were analyzed 3 weeks after transplantation. For analyses, 50 actcatattcatgccgctcgc 30 lungs were examined under a fluorescent dissecting microscope Mouse Sftpb: forward 50 aaccccacacctctgagaac 30 and reverse for tdTomatopos tumor number, and then fixed for IHC. Tumor 50gtgcaggctgaggcttgt 30 area relative to total lung area was quantified using ImageJ on Mouse Selenbp1: forward 50 tcttcaaagatggcttcaacc 30 and reverse hematoxylin and eosin (H&E)–stained sections. 50 ctgccagtcccacacaaata 30? Mouse Gapdh: forward 50 cagcctcgtcccgtagac 30 and reverse 50 Mouse strains and tumor induction LSL-G12D/þ LSL-tdTomato LSL-Cas9 flox cattgctgacaatcttgagtga 30 Kras , R26 , H11 , and p53 mice have Human NKX2-1: forward 50 aggacaccatgaggaacag 30 and reverse been described previously (12, 13, 37). The Lenti-U6sgRNA/PGK- 50 cccatgaagcgggagat 30 Cre vector was generated as described previously (37). Lenti- Human SELENBP1: forward 50 cagcgcttctacaagaacga and reverse U6sgSelenbp1#1/PGK-Cre was cotransfected with packaging vec- 50 tgatcaggcctggcattt 30 tors (delta8.2 and VSV-G) into 293T cells using TransIT-LT1 Human ACTIN: forward 5; ccttgcacatgccggag 30 and reverse 50 (Mirus Bio). The supernatant was collected at 48 and 72 hours, gcacagagcctcgcctt 30 ultracentrifuged at 25,000 rpm for 90 minutes, and resuspended in PBS. Tumors were initiated by intratracheal transduction Western blotting of mice with Lenti-U6sgSelenbp1#1/PGK-Cre, and tumors For Western blotting, denatured protein samples were run on were allowed to develop for 4 months. A total of 2 105 viral a 4% to 12% Bis-Tris (NuPage) and transferred onto PVDF particles were injected into KT and KT;Cas9 mice, and 2.5 104

www.aacrjournals.org Mol Cancer Res; 16(11) November 2018 1739

Downloaded from mcr.aacrjournals.org on September 27, 2021. © 2018 American Association for Cancer Research. Published OnlineFirst July 12, 2018; DOI: 10.1158/1541-7786.MCR-18-0392

Caswell et al.

particles were injected into KPT and KPT;Cas9 mice. Mice were regions. The Nkx2-1–bound region at the promoter of Selenbp1 analyzed by examining the lungs, pleural cavity, liver, kidneys, was associated with H3K4me3 (a marker of active genes; and spleen under a fluorescent microscope for tumors. Lung tissue ref. 44) and H3K27ac (a marker of activity at promoters and was fixed for IHC staining. The Stanford Institute of Medicine enhancers; ref. 45), whereas the putative enhancer regions were Animal Care and Use Committee approved all animal studies and associated with H3K4me1 (a marker of enhancers and pro- procedures. moters) and H3K27ac (a marker of enhancers; ref. 46). H3K27me3 [a marker of Polycomb-mediated gene repression (47, 48)] was absent from both the promoter and putative Tracking indels by decomposition (TIDE analysis) enhancer regions (Fig. 1H). Collectively, this suggests that Cutting efficiency was calculated using the free online webtool Nkx2-1 likely regulates the expression of Selenbp1 through as described in ref. 38. both its enhancer and promoter regions. Foxa1/Foxa2 also bound the promoter and enhancer regions, suggesting core- Results gulation of Selenbp1 by Nkx2-1 and a Foxa transcription factor Selenbp1 is an Nkx2-1 target in lung adenocarcinoma (Fig. 1H; refs. 34, 49–52). The pattern of chromatin marks To identify candidate Nkx2-1–regulated genes in lung adeno- around other known Nkx2-1 targets, including Sftpb and Nkx2- carcinoma, we integrated datasets from human lung adenocarci- 1, itself were consistent with the coordinate regulation of noma and a defined mouse lung cancer model. To uncover genes a large number of genes by these factors (Supplementary that are likely to be directly regulated by Nkx2-1, we analyzed an Fig. S2B and S2C; refs. 13, 53–55). Nkx2-1 ChIP-seq dataset generated from oncogenic KrasG12D- We recently characterized the global changes driven murine lung tumors (13). We next determined how well that occur in neoplastic cells during tumor progression and the expression of these putative direct Nkx2-1 target genes corre- metastasis in a KrasG12D-driven, p53-deficient lung adenocar- lates with NKX2-1 expression in human lung adenocarcinoma cinoma mouse model (33). As anticipated, the expression of (27, 28, 39). This analysis uncovered several canonical Nkx2-1 Nkx2-1 and canonical Nkx2-1 target genes correlated with the target genes including surfactant protein B (Sftpb) and Nkx2-1 expression Selenbp1. In fact, Selenbp1 was more than 7-fold itself, as well as several potentially novel Nkx2-1 target genes (Fig. lower in pleural disseminated tumor cells and metastases than 1A). Consistent with the Nkx2-1 ChIP-seq data from mouse primary nonmetastatic tumors (Supplementary Figs. S3 and tumors, NKX2-1 also bound regions near NAPSA, NPC2, and S4A). IHC for Nkx2-1 and Selenbp1 on primary tumors and LSL-G12D/þ flox/flox LSL-tdTomato SELENBP1 in a human lung adenocarcinoma cell line (H441; metastases from Kras ;p53 ;R26 (KPT) ChIP-seq data from ref. 40; Supplementary Fig. S1A). To initially mice confirmed that Nkx2-1low/neg areas were most often assess the function of Napsa, Npc2, and Selenbp1 in lung adeno- Selenbp1low/neg.Thisisconsistentwiththelowerexpression carcinoma cells, we used multiple shRNAs to stably knock down of SELENBP1 protein in poorly differentiated human lung each gene in an Nkx2-1pos lung adenocarcinoma cell line derived adenocarcinomas (Fig. 2A and B; ref. 18). The early-stage LSL-G12D flox/flox LSL-G12D/þ from the genetically engineered Kras ;p53 (KP) lung tumors that develop in p53-proficient Kras ; LSL-tdTomato mouse model (Supplementary Fig. S1B and S1D; refs. 12, 41). R26 (KT) mice were almost exclusively Nkx2-1pos Selenbp1 knockdown elicited the most dramatic increase in and were most often Selenbp1pos (Supplementary Fig. S4B clonal growth ability when the cells were plated in low-density and S4C). conditions (Supplementary Fig. S1E and S1F). These data, in The Nkx2-1–bound regions upstream of Selenbp1 are closest to combination with the correlation between SELENBP1 and the Selenbp1 TSS, but these enhancer regions could nonetheless NKX2-1 in human lung adenocarcinoma, focused our additional regulate the expression of other nearby genes (Supplementary Fig. validation and mechanistic experiments on this novel Nkx2-1– S3C). Unlike Selenbp1 (Fig. 2C), the expression of other genes in regulated gene. proximity of the NKX2-1–bound enhancer regions did not cor- Analysis of multiple human lung adenocarcinoma gene relate with the expression of Nkx2-1 or Sftpb in cancer cells from expression datasets confirmed that SELENBP1 expression cor- different stages of metastatic progression from KT and KPT mice relates with NKX2-1 expression, as well as with the expression (Supplementary Figs. S3A and S4D). The other genes within the of the NKX2-1 target SFTPB (Fig. 1B–D; Supplementary Fig. same region as SELENBP1 also did not correlate with NKX2-1 S2A). IHC on human lung adenocarcinomas uncovered a expression in human lung adenocarcinoma (Fig. 1B–D; Supple- general correlation between NKX2-1 and SELENBP1 protein mentary Fig. S3C). expression (Fig. 1E and F; Supplementary Fig. S2B). Low Collectively, these data document a clear correlation between SELENBP1 expression occurred in some Nkx2-1pos tumors; the expression of the transcription factor Nkx2-1 and Selenbp1 in however, NKX2-1low/neg tumors were almost always SELENB- lung cancer, which is consistent with our initial identification of P1low/neg (Fig. 1E and F; Supplementary Fig. S2B). Higher Nkx2-1–bound regions proximal to Selenbp1. expression of NKX2-1 in lung adenocarcinoma correlates with better patient outcome (42, 43), and we confirmed that higher Nkx2-1 is required for Selenbp1 expression in lung expression of SELENBP1 was also significantly associated with adenocarcinoma cells better patient outcome (Fig. 1G). In cell lines generated from the KP lung cancer mouse model, To further investigate the regulation of Selenbp1 by Nkx2-1, Selenbp1 expression correlated with Nkx2-1 and Sftpb expression we examined the Nkx2-1 ChIP-seq peaks upstream of Selenbp1 (Supplementary Fig. S4E and S4F). Selenbp1 expression was high (Fig. 1H; ref. 13). One small Nkx2-1–bound peak was located in Nkx2-1pos cell lines generated from nonmetastatic primary neg in the promoter region of Selenbp1, whereas three larger peaks tumors (TnonMet) and low in Nkx2-1 cell lines generated from were identified further upstream of Selenbp1 (Fig.1H).Wenext metastatic primary tumors (TMet) and metastases (Met; Fig. 2D; examined the histone modifications associated with these ref. 12). To determine whether Nkx2-1 is sufficient and/or

1740 Mol Cancer Res; 16(11) November 2018 Molecular Cancer Research

Downloaded from mcr.aacrjournals.org on September 27, 2021. © 2018 American Association for Cancer Research. Published OnlineFirst July 12, 2018; DOI: 10.1158/1541-7786.MCR-18-0392

Selenbp1, Nkx2-1 Target, Suppresses Lung Cancer

A B C 100 Nkx2-1

pos Pos Low/neg 10 (FPKM) 28 1 1 Pos NKX2-1

neg 0.1 10 13 Spearman: 0.83 Selenbp1

Selenbp1 Pearson: 0.66

Low/neg 0.01 0.01 0.1 1 10 1,000100 NKX2-1 H+E NKX2-1 SELENBP1 Nkx2-1 (FPKM) D E F G * 10 *** 394T4 ** 1.2 1.2 ** ) 2 8 1.0 1.0

(log 0.8

0.8 GFP 6 sh 0.6 shNkx2-1 Expression

Expression 0.6 Selenbp1 4 0.4 0.4 Hsp90 Selenpb1 0.2 0.2 2 Nkx2-1 0.0 Selenbp1 0.0 Nkx2-1 Met T Met nonMet Hsp90 GFP T sh GFP sh shNkx2-1 shNkx2-1 394T4 394T4

H I J K 25 **** 15 **** 368T1 389T2 20 10 15 Expression sh GFP shNkx2-1 Puro Nkx2-1 Expression 10 Selenbp1 5 Selenbp1 Nkx2-1 5

Nkx2-1 Nkx2-1 Selenbp1 Hsp90 0 0 Hsp90 Puro Puro Nkx2-1 Nkx2-1 389T2 389T2

Figure 2. Selenbp1 and Nkx2-1 expression correlates in murine lung tumors and Nkx2-1 is required and sufficient for Selenbp1 expression. A, IHC staining for Nkx2-1 and Selenbp1 on lung tumors from KrasLSL-G12D/þ;p53flox/flox;R26LSL-tdTomato (KPT) mice. H&E staining illustrates less differentiated areas with low Nkx2-1 and Selenbp1 expression. Scale bar, 50 micrometers. B, Correlation between Nkx2-1 and Selenbp1 expression scored as low/negative (low/neg) or positive (pos) in KPT lung tumors quantified by IHC. , P < 0.0001. C, Correlation between Nkx2-1 and Selenbp1 expression in tumors from KrasLSL-G12D/þ;R26LSL-tdTomato (KT)andKPT mice. D, Selenbp1 expression is significantly lower in cell lines derived from metastatic primary lung tumors and metastases from KP mice. , P < 0.05; , P < 0.01.

E and F, Stable knockdown of Nkx2-1 reduces Nkx2-1 (E)andSelenbp1 (F) expression in a TnonMet cell line (394T4). Normalized expression relative to shGFP and Gapdh. qRT-PCR mean SD of triplicate wells. , P < 0.001; , P < 0.01. G and H, Western blot analysis for Nkx2-1 and Selenbp1 in TnonMet cell lines (394T4 and 368T1) with stable knockdown of Nkx2-1. Hsp90 shows loading. I and J, Reexpression of Nkx2-1 in a TMet cell line (389T2) increases Nkx2-1 and Selenbp1 expression. Expression relative to Gapdh and normalized to control (Puro) vector. qRT-PCR mean SD of triplicate wells. , P < 0.0001. K, Western blot analysis for Nkx2-1 and Selenbp1 in 389T2 cells reexpressing Nkx2-1. Hsp90 shows loading. required for Selenbp1 expression, we performed gain- and loss-of- Nkx2-1 knockdown greatly reduced Selenbp1 mRNA and protein function experiments with these mouse lung adenocarcinoma cell (Fig. 2F–H; Supplementary Fig. S5D). To determine whether lines. We stably knocked down Nkx2-1 using previously validated Nkx2-1 is sufficient to drive Selenbp1 expression, we overexpressed shRNAs, in two cell lines derived from nonmetastatic primary Nkx2-1 in a cell line derived from a metastatic primary tumor tumors from the KP mouse model (TnonMet; 394T4 and 368T1; (TMet; 389T2; Fig. 2I–K; Supplementary Fig. S5E; ref. 12). Over- ref. 12). We confirmed knockdown of Nkx2-1 at the RNA and expression of Nkx2-1 in these cells induced expression of Selenbp1, protein levels (Fig. 2E, G, and H; Supplementary Fig. S5A). as well as the expression of the canonical Nkx2-1 target Sftpb (Fig. Downregulation of the canonical Nkx2-1 target gene Sftpb con- 2I–K; Supplementary Fig. S5E). firmed reduced Nkx2-1 activity in the shNkx2-1 TnonMet cell lines To investigate whether NKX2-1 is required for SELENBP1 (Supplementary Fig. S5B and S5C). In both TnonMet cell lines, expression in human cell lines, we examined SELENBP1

www.aacrjournals.org Mol Cancer Res; 16(11) November 2018 1741

Downloaded from mcr.aacrjournals.org on September 27, 2021. © 2018 American Association for Cancer Research. Published OnlineFirst July 12, 2018; DOI: 10.1158/1541-7786.MCR-18-0392

Caswell et al.

expression in a published gene expression dataset from a human a series of assays to investigate the function of Selenbp1 on lung adenocarcinoma cell line (H441) with and without NKX2-1 proliferation and cell migration. Selenbp1 knockdown had little knockdown (40). NKX2-1 knockdown resulted in a 4-fold if any effect on cell growth under standard culture conditions (Fig. decrease in NKX2-1 expression and an approximately 2-fold 3C; Supplementary Fig. S5K–S5O). In contrast, when plated at decrease in SELENBP1 expression (Supplementary Fig. S5F and low density, Selenbp1 knockdown cells formed significantly larger S5G). SFTPB expression was also approximately 2-fold lower in and considerably more numerous colonies than shGFP control NKX2-1 knockdown H441 cells (Supplementary Fig. S5H; ref. 40). cells (Fig. 3D–F). These data suggest that Nkx2-1 regulates the expression of Several studies have suggested that Nkx2-1 can inhibit migra- Selenbp1 in lung adenocarcinoma. tory ability (7, 15, 56). We used a 2D migration assay to determine whether Nkx2-1 controls the migratory ability of lung adenocar- Selenbp1 inhibits clonal growth and migration cinoma cells in culture. Nkx2-1 knockdown increased the migra- To assess the molecular and cellular phenotypes controlled by tory ability of TnonMet cells, whereas doxycycline-regulatable Selenbp1, we knocked down Selenbp1 with two independent induction of Nkx2-1 in a TMet line decreased migratory ability – shRNAs in the TnonMet cell lines and confirmed Selenbp1 knock- (Fig. 3G and H; Supplementary Fig. S6A S6D). Both cell-intrinsic down at the RNA and protein levels (Fig. 3A and B; Supplementary and secreted factors can influence migration; therefore, we deter- Fig. S5I and S5J). Using these knockdown cell lines, we performed mined whether the effect of Nkx2-1 knockdown on migration was

A **** B C D 394T4 1.2 **** 394T4 1.6 NS

#4 NS shGFP shNkx2-1 1.0 1.4 NS 0.8 1.2 1.0 0.6 0.8 shSelenbp1 shGFP shSelenbp1 #1 0.6 0.4 shSelenbp1#1 shSelenbp1#4 Selenbp1 0.4 0.2 Selenbp1 Expression 0.2 0.0 Hsp90 0.0 Normalized % BrdU incorporation shGFP shGFP shNkx2-1 shSelenbp1#1 shSelenbp1#1 shSelenbp1#4 shSelenbp1#4 394T4 394T4

E F G H *** * 10.0 *** 5.0 *** 0.20 * 394T4.Tom 8.0 4.0 *** shGFP shNkx2-1 shSelenbp1#1 shSelenbp1#4 0.15 6.0 *** 3.0 *** ** 0.10 4.0 2.0 0 hours 0.05 2.0 1.0 Normalized colony size % Gap remaining (12 h) Normalized colony number 0.0 0.0 0.00 12 hours shGFP shGFP shGFP shNkx2-1 shNkx2-1 shNkx2-1 shSelenbp1#1 shSelenbp1#4 shSelenbp1#1 shSelenbp1#4 shSelenbp1#1 shSelenbp1#4 394T4 394T4 394T4

Figure 3. Nkx2-1 and its target Selenbp1 inhibit clonal growth and migratory ability. A, shRNA-mediated knockdown of Selenbp1 using two hairpins significantly reduces Selenbp1 expression in a TnonMet cell line (394T4). Normalized expression relative to shGFP and Gapdh. qRT-PCR mean SD of triplicate wells. , P < 0.0001. B, Western blot analysis demonstrating stable knockdown of Selenbp1 394T4 cells. Hsp90 shows loading. C, BrdU incorporation is not significantly increased by stable knockdown of Nkx2-1 or Selenbp1 in 394T4 cells. Data are representative of four independent experiments. D, Representative images of low density colony formation of 394T4 cells with stable knockdown of Nkx2-1 or Selenbp1. Scale bar, 1 cm. E and F, Colony number (E) and size (F) formed by 394T4 cells is significantly increased with stable knockdown of Nkx2-1 or Selenbp1. , P < 0.01; , P < 0.001. Data are representative of three independent experiments. G, Representative images of scratch assays using the 394T4 cells with knockdown of Nkx2-1 or Selenbp1 at 0 and 12 hours after scratching. Scale bar, 5 mm. H, The TnonMet cell line (394T4) migrates significantly faster with knockdown of Nkx2-1 or Selenbp1. Mean SD of triplicate wells. , P < 0.05; , P < 0.001.

1742 Mol Cancer Res; 16(11) November 2018 Molecular Cancer Research

Downloaded from mcr.aacrjournals.org on September 27, 2021. © 2018 American Association for Cancer Research. Published OnlineFirst July 12, 2018; DOI: 10.1158/1541-7786.MCR-18-0392

Selenbp1, Nkx2-1 Target, Suppresses Lung Cancer

cell autonomous. By performing migration assays with differen- the total tumor burden as assessed by lung weight and percent tially labeled cocultured control TMet and TMet.Nkx2-1 expressing tumor area (Fig. 4A–E). Although Selenbp1 knockdown did cells, we found that reexpression of Nkx2-1 only reduced the not significantly alter tumor number, the tumors that formed migration of the Nkx2-1 expressing cells, consistent with its effect were larger suggesting that Selenbp1 inhibits tumor growth being cell autonomous (Supplementary Fig. S6A–S6D). (Fig. 4C–E). Given the effect of Nkx2-1 on migration, we examined whether Selenbp1 also regulates migration in vitro. Selenbp1 knockdown significantly increased migratory ability of T cells (Fig. 3G Selenbp1 increases Nkx2-1 protein level nonMet Selenbp1 and H). To rule out that the increased migration of these cells was In the course of our experiments on the knockdown Selenbp1 due to the slight increase in proliferation, we performed addi- cells, we unexpectedly found that knockdown of resulted tional migration assays in the presence of proliferation inhibitor in reduced Nkx2-1 expression at both the RNA and protein levels – mitomycin C (Supplementary Fig. S6E and S6F). The migratory (Fig. 5A D). Given that Nkx2-1 regulates its own mRNA expres- fl ability of Selenbp1 knockdown cells was still significantly higher sion, these results could re ect an effect of Selenbp1 on either Nkx2-1 than control cells, confirming that the effect of Selenbp1 on gene expression or Nkx2-1 protein level. To determine fl migration is independent of cellular proliferation (Supplemen- whether Selenbp1 can in uence Nkx2-1 protein levels in a het- tary Fig. S6E and S6F). erologous system, we expressed Nkx2-1 with and without exog- enous Selenbp1 in 293T cells. Nkx2-1 and Selenbp1 were expressed from plasmids in which constitutive expression was Selenbp1 suppresses tumor growth in vivo driven by the MSCV long terminal repeat. Nkx2-1 protein was To investigate the importance of Selenbp1 in tumor growth consistently higher in the presence of coexpressed Selenbp1, and metastasis in vivo, we intravenously transplanted TnonMet suggesting that Selenbp1 may stabilize Nkx2-1 protein (Fig. 5E; Selenbp1 knockdown and control cells into syngeneic recipient Supplementary Fig. S7A and S7B). We also examined Nkx2-1 mice (Fig. 4A–E). Selenbp1 knockdown significantly increased protein expression in the tumors that formed after intravenous

A B 394T4.Tom C NS **** 90 0.4 shGFP shSelenbp1#4 *** 75 0.3 60 Figure 4. Selenbp1 suppresses tumor growth 0.2 Tomato 45 in vivo. A, Knockdown of Selenbp1 Tumor # in an intravenously injected TnonMet cell 30 line (394T4), results in significantly 0.1 increased lung weight 3 weeks (g) Lung weight 15 postinjection. Each dot represents H+E a mouse, and the bar is the mean. 0.0 0 , P < 0.001; , P < 0.0001. B, Increased total tumor mass of a Lung shGFP Normal TnonMet cell line (394T4.Tom), with shGFP Selenbp1 knockdown is shown by tdTomato fluorescent and H&E shSelenbp1#4 images. Fluorescent images, scale bar, shSelenbp1#4 5 mm, H&E images, scale bar ¼ 1 mm. 394T4.Tom 394T4.Tom C, Knockdown of Selenbp1 in a TnonMet cell line (394T4), intravenously injected, results in a slight but not D **** fi E signi cant increase in tumor number. 25 30 ** Each dot represents a mouse, and the bar is the mean. NS, not significant. 25 20 )

D, Knockdown of Selenbp1 in a T 2 nonMet 20 cell line (394T4), intravenously injected, induces significantly larger 15 15 tumors than control cells. Each dot 10 represents a tumor, and the bar is the 10 mean. , P < 0.0001. E, Tumor area Tumor area (%) 5

fi area (mm Tumor is signi cantly higher after 5 0 intravenous injection of a TnonMet cell line (394T4.Tom) with Selenbp1 0 knocked down compared with control cells. Each dot represents a mouse, shGFP and the bar is the mean. , P < 0.01. shGFP shSelenbp1#4 394T4.Tom shSelenbp1#4 394T4.Tom

www.aacrjournals.org Mol Cancer Res; 16(11) November 2018 1743

Downloaded from mcr.aacrjournals.org on September 27, 2021. © 2018 American Association for Cancer Research. Published OnlineFirst July 12, 2018; DOI: 10.1158/1541-7786.MCR-18-0392

Caswell et al.

injection of control TnonMet and Selenbp1 knockdown cells. Com- To determine whether this feedback loop exists in human lung pared with tumors from control cells, many more of the tumors adenocarcinoma, we stably knocked down SELENBP1 with two that formed from shSelenbp1 cells were Nkx2-1neg (Fig. 5F), This separate shRNAs in a human lung adenocarcinoma cell line further supports the importance of Selenbp1 in maintaining (H441). SELENBP1 knockdown resulted in a significant decrease Nkx2-1 expression. in NKX2-1 mRNA and protein expression (Fig. 5G–I). This

A B C D **** 394T4 ** 1.2 **** 1.2 * 368T1

1.0 1.0

0.8 0.8 shSelenbp1#4 0.6 shGFP shSelenbp1#1 0.6 shSelenbp1#4 shSelenbp1#1 shGFP

0.4 Nkx2-1 0.4 Nkx2-1 Nkx2-1 Expression Nkx2-1 Expression Hsp90 Hsp90 0.2 0.2

0.0 0.0 shGFP shGFP shSelenbp1#1 shSelenbp1#4 shSelenbp1#1 shSelenbp1#4 394T4 368T1

EGF H pos **** * Nkx2-1 1.2 *** 1.4 ** Nkx2-1neg 1.0 1.2 100 100 1.0 0.8 Nkx2-1+Selenbp1 Mock Nkx2-1+GFP GFP+Selenbp1 0.8 0.6 Nkx2-1 Expression 0.6 0.4 50 50 0.4 Selenbp1 Tumor (%) Tumor Tumor (%) Tumor 0.2 NKX2-1 0.2 SELENBP1 Expression GFP 0.0 0.0 0 0 HSP90 1 2 3 1 2 3 shGFP shGFP shSelenbp1 shGFP I H441 shSELENBP1#2 shSELENBP1#2 shSELENBP1#1 shSELENBP1#1 H441 H441 shGFP shSELENBP1#1 shSELENBP1#2 NKX2-1 SELENBP1 HSP90

Figure 5.

Selenbp1 functions in a positive feedback loop with Nkx2-1. A and B, Knockdown of Selenbp1 in the TnonMet cell line (394T4) significantly reduces Nkx2-1 mRNA (A)and protein (B) expression. Normalized gene expression relative to Gapdh. qRT-PCR mean SD of triplicate wells. Hsp90 shows loading. , P < 0.0001.

C and D, Knockdown of Selenbp1 in the TnonMet cell line (368T1) significantly reduces Nkx2-1 mRNA (C) and protein (D) expression. Normalized gene expression relative to Gapdh. qRT-PCR mean SD of triplicate wells. Hsp90 shows loading. , P < 0.05; , P < 0.01. E, Transient transfection of Selenbp1 increases the level of exogenous Nkx2-1 protein in 293 cells. HSP90 shows loading. F, Quantification of IHC for Nkx2-1 expression in shGFP and shSelenbp1 lung tumors (from Fig. 4). A higher percentage of Selenbp1 knockdown tumors are Nkx2-1neg compared with control shGFP tumors. Data from 3 mice from each group is shown. G and H, shRNA knockdown of SELENBP1 using two hairpins results in reduced SELENBP1 (G)andNKX2-1 (H) expression in the human lung adenocarcinoma cell line H441. Normalized expression relative to ACTIN. qRT-PCR mean SD of triplicate wells. , P < 0.05; , P < 0.01; , P < 0.001; , P < 0.0001. I, In the human lung adenocarcinoma cell line H441, SELENBP1 knockdown using two hairpins decreases SELENBP1 and NKX2-1 protein levels. HSP90 shows loading.

1744 Mol Cancer Res; 16(11) November 2018 Molecular Cancer Research

Downloaded from mcr.aacrjournals.org on September 27, 2021. © 2018 American Association for Cancer Research. Published OnlineFirst July 12, 2018; DOI: 10.1158/1541-7786.MCR-18-0392

Selenbp1, Nkx2-1 Target, Suppresses Lung Cancer

supports the possibility of a positive feedback loop between To investigate whether Selenbp1 deficiency could also increase SELENBP1 and NKX2-1 in lung adenocarcinoma, which could the growth of KrasG12D-driven p53-deficient tumors in vivo,we LSL-G12D/þ contribute to the downregulation of NKX2-1 during tumor initiated tumors with Lenti-sgSelenbp1/Cre in Kras ; flox/flox LSL-tdTomato LSL-Cas9 progression. p53 ;R26 ;H11 (KPT;Cas9) and control KPT mice. Inactivation of Selenbp1 in KPT;Cas9 mice resulted in significantly larger tumors as measured by lung weight and Selenbp1 suppresses primary tumor growth percent tumor area as compared with control KPT mice (Fig. in vivo To determine whether Selenbp1 affects tumor growth , 6D–F). Lenti-sgSelenbp1/Cre-initiated tumors in KPT;Cas9 mice – we used somatic CRISPR/Cas9 mediated genome editing to had indels at the expected site in Selenbp1 and a higher proportion Selenbp1 KrasG12D inactivate in -driven autochthonous lung of lung tumors in KPT;Cas9 mice were Selenbp1neg (Supplemen- tumors (34, 57). We designed and validated an sgRNA-target- tary Fig. S8A and S8d). The expression of Nkx2-1 and Selenbp1, Selenbp1 ing exon 2 of . We generated a lentiviral vector encod- quantified by IHC, significantly correlated in KPT;Cas9 lung Selenbp1/Cre ing this sgRNA and Cre-recombinase (Lenti-sg ). tumors with a higher number of tumors being either Selenbp1/Cre KrasLSL- We initiated tumors with Lenti-sg in Nkx2-1pos;Selenbp1pos or Nkx2-1neg;Selenbp1neg (Supplementa- G12D/þ;R26LSL-tdTomato;H11LSL-Cas9 KT;Cas9 KT ( ) and control ry Fig. S8E). Inactivation of Selenbp1 also increased the number of KT;Cas9 mice. Four months after tumor initiation, mice had Hmga2pos;Nkx2-1neg;Selenbp1neg areas in KPT;Cas9 mice versus KT greater total tumor burden than control mice, as measured KPT mice (Supplementary Fig. S8F). High expression of Hmga2 is – by lung weight and percent tumor area (Fig. 6A C). a marker of poorly differentiated metastatic lung adenocarcinoma Selenbp1 Cre KT;Cas9 Lenti-sg / -initiated tumors in mice had (32), and the increased number of Hmga2pos regions in KPT;Cas9 Selenbp1 indels at the expected site in (Supplementary mice suggests that Selenbp1 inactivation may lead not only to KT;Cas9 Fig. S8A). A higher proportion of lung tumors in mice increased tumor growth but also to increased tumor progression. were Selenbp1neg,comparedwithKT lung tumors (Supple- KT;Cas9 mentary Fig. S8B). mice had a higher proportion of Discussion Selenbp1neg;Nkx2-1neg lung tumors, and a lower number of Selenbp1pos;Nkx2-1pos lung tumors compared with KT mice These studies were initiated to gain additional insight into the (Supplementary Fig. S8C). function of the Nkx2-1 transcription factor in lung cancer. Our

A B * C ** Figure 6. Lenti-sgSelenbp1/Cre 1.2 50 Selenbp1 suppresses lung tumor KT KT;Cas9 growth. A and B, CRISPR/Cas9– 1.0 40 mediated targeting of Selenbp1 in KT;Cas9 0.8 mice increases tumor burden. 30 Representative fluorescence 0.6 dissecting scope and H&E images (A) 20 and lung weight (B)ofKT and KT;Cas9 Tomato 0.4 with Lenti-sgSelenbp1/Cre initiated % Tumor area

Lung weight (g) 10 tumors 4 months after initiation. Each 0.2 dot represents a mouse, and the bar is the mean. Normal lung weight is 0.0 0 indicated with a dashed line. , P < 0.05. Fluorescent images scale H+E KT

bars, 5 mm, H&E images scale bars, 1 KT KT;Cas9 mm. C, KT;Cas9 mice with Lenti- KT;Cas9 sgSelenbp1/Cre initiated tumors have sgSelenbp1 sgSelenbp1 greater tumor burden than KT mice. Each dot represents a mouse and the bar is the mean. , P < 0.01. D and E, D E F CRISPR/Cas9–mediated targeting of ** * Selenbp1 in KPT;Cas9 mice increases Lenti-sgSelenbp1/Cre 2.0 50 tumor burden. Representative fluorescence dissecting scope and KPT KPT;Cas9 1.6 40 H&E images (D) and lung weight (E)of KPT and KPT;Cas9 with Lenti- 1.2 30 sgSelenbp1/Cre initiated tumors 4 months after initiation. Each dot 0.8 20 represents a mouse, and the bar is the Tomato % Tumor area mean. Normal lung weight is indicated Lung weight (g) 0.4 10 with a dashed line. , P < 0.01. Fluorescent images scale bars, 5 mm, 0.0 0 H&E images scale bars, 1 mm. F, KPT; Cas9 mice with Lenti-sgSelenbp1/Cre initiated tumors have greater tumor KPT H+E burden than mice. Each dot KPT KPT KPT;Cas9 represents a mouse and the bar is the KPT;Cas9 mean. , P < 0.05. sgSelenbp1 sgSelenbp1

www.aacrjournals.org Mol Cancer Res; 16(11) November 2018 1745

Downloaded from mcr.aacrjournals.org on September 27, 2021. © 2018 American Association for Cancer Research. Published OnlineFirst July 12, 2018; DOI: 10.1158/1541-7786.MCR-18-0392

Caswell et al.

analysis uncovered Selenbp1 as a tumor-suppressive Nkx2-1 or too little can be antitumorigenic. In lung cancer, several studies target gene. Cell culture assays, transplant experiments, and the have examined the role of selenium as a chemopreventative agent analysis of autochthonous mouse models of lung cancer collec- (67, 72). A recent clinical trial and meta-analysis concluded that tively indicate that Selenbp1 suppresses tumor growth in vivo. selenium supplementation does not decrease lung cancer inci- SELENBP1 has been identified as a potentially important factor dence in an unselected population, but significantly decreases in several different cancer types (17–19, 21, 58). Our data suggest incidence among patients with low baseline selenium (73, 74). that Selenbp1 inhibits migration of lung adenocarcinoma cells in Understanding the regulation of Nkx2-1 remains an important culture, which is consistent with its effect on colorectal cancer and question as Nkx2-1 serves dual functions as both an oncogene in hepatocellular carcinoma cell lines (Fig. 3G and H; Supplemen- some human lung adenocarcinomas and a suppressor of cancer tary Fig. S6E and S6F; refs. 17, 60). In lung cancer, Selenbp1 also progression in others (3, 5, 10, 12, 75). Thus, within these reduced clonal growth, whereas in prostate cancer, SELENBP1 different contexts, a better understanding of the regulators of inhibits anchorage-independent growth (59). Finally, using trans- Nkx2-1 expression could uncover strategies to downregulate plant models and CRISPR/Cas9–mediated inactivation in autoch- oncogenic NKX2-1 or increase NKX2-1 expression to limit tumor thonous mouse models of lung cancer, we show that Selenbp1 growth and progression. Despite its importance in lung cancer suppresses cancer growth in vivo. Functional evidence suggests that and known ability to regulate its own expression, other pathways SELENBP1 inhibits growth in colorectal cancer and hepatocellular that control Nkx2-1 expression in lung cancer are not well under- carcinoma and that overexpression of SELENBP1 enhances cis- stood (54, 55, 76). Several other transcription factors bind to the platin sensitivity of esophageal adenocarcinoma (17, 58, 60, 61). promoter of Nkx2-1 in lung epithelial cells; however, whether The capacity of Selenbp1 to change multiple phenotypes across these factors also contribute to the regulation of Nkx2-1 in lung numerous cancer types suggests that Selenbp1 plays a broad role cancer has not been well characterized (6, 55, 76–78). Our in cancer growth and progression. Although Selenbp1 expression knockdown data suggest that Selenbp1 may positively regulate is mechanistically linked to Nkx2-1 in lung cancer, different Nkx2-1. Other factors also act as regulators of Nkx2-1 expression, factors must control Selenbp1 expression in other cancer types. emphasizing the importance of Nkx2-1 function in development The molecular functions of Selenbp1 in cancer are relatively and cancer (35, 79). However, Selenbp1 inactivation in vivo poorly understood. Selenbp1 is expressed widely across many appears insufficient to lead to the loss of Nkx2-1 in lung adeno- adult tissues and is an alpha-beta protein with several loop regions mas and early-stage lung adenocarcinomas in vivo, suggesting that and one cysteine residue (Cys57; ref. 19). In human liver, breast, Selenbp1 downregulation likely contributes to Nkx2-1 down- and colorectal cancer cell lines, Selenbp1 inhibits glutathione regulation only within certain cellular contexts. Rescue experi- peroxidase 1 (GPX1) enzymatic activity and decreased GPX1 ments examining how Selenbp1 reexpression in Selenbp1 knock- activity correlates with decreased invasiveness (60, 62). In prostate down cell lines would be beneficial in a deeper understanding of cancer cells, Selenbp1 binds to deubiquitinating factors, poten- how Selenbp1 impacts the tumor-suppressive phenotypes uncov- tially playing a role in the regulation of ubiquitin-mediated ered in this article. In addition, defined mutant forms of Selenbp1, protein degradation. However, whether ubiquitination is expressed in knockdown cell lines, would provide additional decreased or increased with Selenbp1 binding remains unclear insight into the function of Selenbp1 and into its mechanism of (59, 63). Most recently, Selenbp1 was identified as a novel human tumor suppression. methanethiol oxidase that, when mutated, causes extraoral hal- KrasG12D-driven Nkx2-1 heterozygous lung tumors in mice can itosis (64). Our findings suggest that Selenbp1 may play an progress to become Nkx2-1neg mucinous adenocarcinomas, con- important role in stabilizing Nkx2-1 protein. Whether the effect sistent with the development of mucinous lung adenocarcinoma LSL-G12D/þ flox/flox of Selenbp1 on Nkx2-1 protein levels represents a direct bio- in Kras ;Nkx2-1 mice (13, 14, 36). Inactivation of chemical link between these proteins remains unknown. Both Selenbp1 by somatic CRISPR/Cas9 genome editing in KrasG12D- Nkx2-1 and Selenbp1 knock-down inhibits clonal growth and driven mouse models did not induce mucinous adenocarcinoma migration (Fig. 3). These data suggest that either the knockdown over the timeframe of our studies. In KT;Cas9 mice with Lenti- of Nkx2-1 inhibits these phenotypes through a signaling cascade sgSelenbp1/Cre–initiated tumors, more than half of Selenbp1neg mediated by the feedback loop between Selenbp1 and Nkx2-1, or tumors were also Nkx2-1neg. However, the presence of Selenbp1neg that other factors downstream of Selenbp1 induce the phenotypes Nkx2-1pos tumors suggests that loss of Selenbp1 alone is not observed. In human lung adenocarcinomas some of Nkx2-1pos sufficient to lead to complete loss of Nkx2-1 in vivo. tumors have low Selenbp1 expression, indicating that Nkx2-1 Our data suggest that Nkx2-1–driven expression of Selenbp1 may be necessary but not sufficient for endogenous expression of inhibits tumor growth and progression of lung tumors. Selenbp1 Selenbp1. Understanding the connection between Nkx2-1 and may play an important role across cancer types, and future work Selenbp1 will be an important area for future study. on the molecular mechanism by which Selenbp1 alters cellular Selenium itself has been investigated for nearly half a century as phenotypes associated with tumor growth and metastases will a cancer therapeutic (65–69). Selenium containing proteins lead to an improved understanding of the mediators of tumor include proteins with nonspecifically incorporated selenium, progression. specific selenium-binding proteins (like Selenbp1), and seleno- cysteine-containing proteins (70). Interestingly, in a mouse mod- fl el of familial adenomatous polyposis (Apcmin), treatment with an Disclosure of Potential Con icts of Interest fl organoselenium compound significantly decreased the develop- No potential con icts of interest were disclosed. ment of small intestine and colon tumors (71). In a transgenic mouse model of liver cancer, both selenium deficiency and Authors' Contributions selenium supplementation suppressed carcinogenesis, suggesting Conception and design: D.R. Caswell, C.-H. Chuang, M.M. Winslow that there may be an ideal level of selenium, and either too much Development of methodology: D.R. Caswell, C.-H. Chuang

1746 Mol Cancer Res; 16(11) November 2018 Molecular Cancer Research

Downloaded from mcr.aacrjournals.org on September 27, 2021. © 2018 American Association for Cancer Research. Published OnlineFirst July 12, 2018; DOI: 10.1158/1541-7786.MCR-18-0392

Selenbp1, Nkx2-1 Target, Suppresses Lung Cancer

Acquisition of data (provided animals, acquired and managed patients, Foundation Graduate Research Fellowship Program (GRFP). I.P. Winters was provided facilities, etc.): D.R. Caswell, C.-H. Chuang, R.K. Ma, I.P. Winters, additionally supported by NIH F31-CA210627 and NIHT32-HG000044. C.-H. E.L. Snyder Chuang was supported by an American Lung Association Fellowship. E.L. Analysis and interpretation of data (e.g., statistical analysis, biostatistics, Snyder was supported by a Career Award for Medical Scientists from the computational analysis): D.R. Caswell, C.-H. Chuang, R.K. Ma Burroughs Wellcome Fund, a V Foundation Scholar award, and NIH R01- Writing, review, and/or revision of the manuscript: D.R. Caswell, CA212415. This work was supported by NIH R01-CA175336 (to M.M. C.-H. Chuang, R.K. Ma, I.P. Winters, E.L. Snyder, M.M. Winslow Winslow), a V Foundation Scholar award (to M.M. Winslow), and in part by Administrative, technical, or material support (i.e., reporting or organizing the Stanford Cancer Institute support grant (NIH P30-CA124435). data, constructing databases): D.R. Caswell, R.K. Ma, I.P. Winters Study supervision: M.M. Winslow The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked Acknowledgments advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate The authors thank Pauline Chu for technical assistance, the Stanford Shared this fact. FACS facility for technical support, Sean Dolan and Alexandra Orantes for administrative support, Sebastian Hoersch for help with the Nkx2-1 ChIP-seq analysis, and members of the Winslow laboratory for helpful comments. Received April 23, 2018; revised June 7, 2018; accepted June 29, 2018; D.R. Caswell and I.P. Winters were supported by the National Science published first July 12, 2018.

References 1. Osada H, Tatematsu Y, Yatabe Y, Horio Y, Takahashi T. ASH1 gene is a in vitro and in nude mouse xenografts. Navarro A, editor. PLoS One specific therapeutic target for lung cancers with neuroendocrine features. 2009;4:e7774. Cancer Res 2005;65:10680–5. 18. Chen G, Wang H, Miller CT, Thomas DG, Gharib TG, Misek DE, 2. Garraway LA, Sellers WR. Lineage dependency and lineage-survival onco- et al. Reduced selenium-binding protein 1 expression is associated genes in human cancer. Nat Rev Cancer 2006;6:593–602. with poor outcome in lung adenocarcinomas. J Pathol 2004;202: 3. Kwei KA, Kim YH, Girard L, Kao J, Pacyna-Gengelbach M, Salari K, et al. 321–9. Genomic profiling identifies TITF1 as a lineage-specific oncogene amplified 19. Raucci R, Colonna G, Guerriero E, Capone F, Accardo M, Castello G, et al. in lung cancer. Oncogene 2008;27:3635–40. Structural and functional studies of the human selenium binding protein-1 4. Nishikawa E, Osada H, Okazaki Y, Arima C, Tomida S, Tatematsu Y, et al. and its involvement in hepatocellular carcinoma. Biochim Biophys Acta miR-375 is activated by ASH1 and inhibits YAP1 in a lineage-dependent 2011;1814:513–22. manner in lung cancer. Cancer Res 2011;71:6165–73. 20. Chang PW, Tsui SK, Liew C, Lee CC, Waye MM, Fung KP. Isolation, 5. Yamaguchi T, Hosono Y, Yanagisawa K, Takahashi T. NKX2-1/TTF-1: an characterization, and chromosomal mapping of a novel cDNA clone enigmatic oncogene that functions as a double-edged sword for cancer cell encoding human selenium binding protein. J Cell Biochem 1997;64: survival and progression. Cancer Cell 2013;23:718–23. 217–24. 6. Cheung WK, Zhao M, Liu Z, Stevens LE, Cao PD, Fang JE, et al. Control of 21. Kim H, Kang HJ, You KT, Kim SH, Lee KY, Kim TI. Suppression of human alveolar differentiation by the lineage transcription factors GATA6 and seleniumbinding protein 1 is a late event in colorectal carcinogenesis and is HOPX inhibits lung adenocarcinoma metastasis. Cancer Cell 2013;23: associated with poor survival. Proteomics 2006;6:3466–76. 725–38. 22. Zhang S, Li F, Younes M, Liu H, Chen C, Yao Q. Reduced selenium-binding 7. Li CM, Gocheva V, Oudin MJ, Bhutkar A, Wang SY, Date SR, et al. Foxa2 and protein 1 in breast cancer correlates with poor survival and resistance to the Cdx2 cooperate with Nkx2-1 to inhibit lung adenocarcinoma metastasis. anti-proliferative effects of selenium. PLoS One 2013;8:e63702. Genes Develop 2015;29:1850–62. 23. Zeng GQ, Yi H, Zhang PF, Li XH, Hu R, Li MY, et al. The function and 8. Gehring WJ. Homeo boxes in the study of development. Science significance of SELENBP1 downregulation in human bronchial epithelial 1987;236:1245–52. carcinogenic process. PLoS One 2013;8:e71865. 9. Kim Y, Nirenberg M. Drosophila NK-homeobox genes. Proc Nat Acad Sci 24.HaYS,LeeGT,KimYH,KwonSY,ChoiSH,KimTH,etal.Decreased USA 1989;86:7716–20. selenium-binding protein 1 mRNA expression is associated with poor 10. Caswell DR, Chuang CH, Yang D, Chiou SH, Cheemalavagu S, Kim-Kiselak prognosis in renal cell carcinoma. World J Surg Oncol 2014;12: C, et al. Obligate progression precedes lung adenocarcinoma dissemina- 288. tion. Cancer Discov 2014;4:781–9. 25. Zhang C, Wang YE, Zhang P, Liu F, Sung CJ, Steinhoff MM, et al. 11. Nguyen DX, Chiang AC, Zhang XH, Kim JY, Kris MG, Ladanyi M, et al. Progressive loss of selenium-binding protein 1 expression correlates WNT/TCF signaling through LEF1 and HOXB9 mediates lung adenocar- with increasing epithelial proliferation and papillary complexity in cinoma metastasis. Cell 2009;138:51–62. ovarian serous borderline tumor and low-grade serous carcinoma. Hum 12. Winslow MM, Dayton TL, Verhaak RG, Kim-Kiselak C, Snyder EL, Feldser Pathol 2010;41:255–61. DM, et al. Suppression of lung adenocarcinoma progression by Nkx2-1. 26. The Cancer Genome Atlas Research Network, Weinstein JN, Collisson EA, Nature 2011;473:101–4. Mills GB, Shaw KRM, Ozenberger BA, et al. The Cancer Genome Atlas Pan- 13. Snyder EL, Watanabe H, Magendantz M, Hoersch S, Chen TA, Wang DG, Cancer analysis project. Nat Genet 2013;45:1113–20. et al. Nkx2-1 represses a latent gastric differentiation program in lung 27. Cerami E, Gao J, Dogrusoz U, Gross BE, Sumer SO, Aksoy BA, et al. The cBio adenocarcinoma. Molecular Cell 2013;50:185–99. cancer genomics portal: an open platform for exploring multidimensional 14. Maeda Y, Tsuchiya T, Hao H, Tompkins DH, Xu Y, Mucenski ML, et al. Kras cancer genomics data: figure 1. Cancer Discov 2012;2:401–4. (G12D) and Nkx2-1 haploinsufficiency induce mucinous adenocarcinoma 28. Gao J, Aksoy BA, Dogrusoz U, Dresdner G, Gross B, Sumer SO, et al. of the lung. J Clin Invest 2012;122:4388–400. Integrative analysis of complex cancer genomics and clinical profiles using 15. Hosono Y, Yamaguchi T, Mizutani E, Yanagisawa K, Arima C, the cBioPortal. Sci Signal 2013;6:pl1. Tomida S, et al. MYBPH, a transcriptional target of TTF-1, inhibits 29. Director's Challenge Consortium for the Molecular Classification of ROCK1, and reduces cell motility and metastasis. EMBO J 2012;31: Lung Adenocarcinoma, Shedden K, Taylor JM, Enkemann SA, Tsao MS, 481–93. Yeatman TJ, et al. Gene expression-based survival prediction in lung 16. Runkle EA, Rice SJ, Qi J, Masser D, Antonetti DA, Winslow MM, et al. adenocarcinoma: a multi-site, blinded validation study. Nat Med Occludin is a direct target of thyroid transcription factor-1 (TTF-1/ 2008;14:822–7. NKX2-1). J Biol Chem 2012;287:28790–801. 30. Gyorffy B, Surowiak P, Budczies J, Lanczky A. Online survival analysis 17. Pohl NM, Tong C, Fang W, Bi X, Li T, Yang W. Transcriptional Regulation software to assess the prognostic value of biomarkers using transcriptomic and biological functions of selenium-binding protein 1 in colorectal cancer data in non-small-cell lung cancer. PLoS One 2013;8:e82241.

www.aacrjournals.org Mol Cancer Res; 16(11) November 2018 1747

Downloaded from mcr.aacrjournals.org on September 27, 2021. © 2018 American Association for Cancer Research. Published OnlineFirst July 12, 2018; DOI: 10.1158/1541-7786.MCR-18-0392

Caswell et al.

31. McLean CY, Bristor D, Hiller M, Clarke SL, Schaar BT, Lowe CB, et al. 54. Nakazato M, Endo T, Saito T, Harii N, Onaya T. Transcription of the thyroid GREAT improves functional interpretation of cis-regulatory regions. Nat transcription factor-1 (TTF-1) Gene from a newly defined start site: positive Biotechnol 2010;28:495–501. regulation by TTF-1 in the thyroid. Biochem Biophys Res Commun 32. Brady JJ, Chuang CH, Greenside PG, Rogers ZN, Murray CW, Caswell DR, 1997;238:748–52. et al. An Arntl2-driven secretome enables lung adenocarcinoma metastatic 55. Boggaram V. Thyroid transcription factor-1 (TTF-1/Nkx2.1/TITF1) gene self-sufficiency. Cancer Cell 2016;29:697–710. regulation in the lung. Clin Sci 2009;116:27. 33. Chuang CH, Greenside PG, Rogers ZN, Brady JJ, Yang D, Ma RK, 56. Saito RA, Watabe T, Horiguchi K, Kohyama T, Saitoh M, Nagase T, et al. et al. Molecular definition of a metastatic lung cancer state reveals Thyroid transcription factor-1 inhibits transforming growth factor- -medi- a targetable CD109–Janus kinase–Stat axis. Nat Med 2017;23: ated epithelial-to-mesenchymal transition in lung adenocarcinoma cells. 291–300. Cancer Res 2009;69:2783–91. 34. Martis PC. C/EBP is required for lung maturation at birth. Development 57. Rogers ZN, McFarland CD, Winters IP, Naranjo S, Chuang CH, Petrov 2006;133:1155–64. D, et al. A quantitative and multiplexed approach to uncover the 35. Mu D. The complexity of thyroid transcription factor 1 with both pro- and fitness landscape of tumor suppression in vivo. Nat Methods 2017; anti-oncogenic activities. J Biol Chem 2013;288:24992–5000. 14:737–47. 36. Finberg KE, Sequist LV, Joshi VA, Muzikansky A, Miller JM, Han M, et al. 58. Ansong E, Ying Q, Ekoue DN, Deaton R, Hall AR, Kajdacsy-Balla A, et al. Mucinous differentiation correlates with absence of EGFR mutation and Evidence that selenium binding protein 1 is a tumor suppressor in prostate presence of KRAS mutation in lung adenocarcinomas with bronchioloal- cancer. PLoS One 2015;10:e0127295. veolar features. J Mol Diagn 2007;9:320–6. 59. Jeong JY, Zhou JR, Gao C, Feldman L, Sytkowski AJ. Human selenium 37. Chiou S-H, Winters IP, Wang J, Naranjo S, Dudgeon C, Tamburini FB, et al. binding protein-1 (hSP56) is a negative regulator of HIF-1a and suppresses Pancreatic cancer modeling using retrograde viral vector delivery and in the malignant characteristics of prostate cancer cells. BMB Rep 2014;47: vivo CRISPR/Cas9-mediated somatic genome editing. Genes Dev 2015;29: 411–6. 1576–85. 60. Huang C, Ding G, Gu C, Zhou J, Kuang M, Ji Y, et al. Decreased selenium- 38. Brinkman EK, Chen T, Amendola M, van Steensel B. Easy quantitative binding protein 1 enhances glutathione peroxidase 1 activity and down- assessment of genome editing by sequence trace decomposition. Nucleic regulates HIF-1a to promote hepatocellular carcinoma invasiveness. Clin Acids Res 2014;42:e168. Cancer Res 2012;18:3042–53. 39. The Cancer Genome Atlas Research Network. Comprehensive molecular 61. Silvers AL, Lin L, Bass AJ, Chen G, Wang Z, Thomas DG, et al. Decreased profiling of lung adenocarcinoma. Nature 2014;511:543–50. selenium-binding protein 1 in esophageal adenocarcinoma results from 40. Isogaya K, Koinuma D, Tsutsumi S, Saito R-A, Miyazawa K, Aburatani H, posttranscriptional and epigenetic regulation and affects chemosensitivity. et al. A Smad3 and TTF-1/NKX2-1 complex regulates Smad4-independent Clin Cancer Res 2010;16:2009–21. gene expression. Cell Res 2014;24:994–1008. 62. Fang W, Goldberg ML, Pohl NM, Bi X, Tong C, Xiong B, et al. Functional and 41. Jackson EL. The differential effects of mutant p53 alleles on advanced physical interaction between the selenium-binding protein 1 (SBP1) and murine lung cancer. Cancer Res 2005;65:10280–8. the glutathione peroxidase 1 selenoprotein. Carcinogenesis 2010;31: 42. Schilsky JB, Ni A, Ahn L, Datta S, Travis WD, Kris MG, et al. Prognostic 1360–6. impact of TTF-1 expression in patients with stage IV lung adenocarcinomas. 63. Jeong JY, Wang Y, Sytkowski AJ. Human selenium binding protein-1 Lung Cancer 2017;108:205–11. (hSP56) interacts with VDU1 in a selenium-dependent manner. Biochem 43. Berghmans T, Paesmans M, Mascaux C, Martin B, Meert AP, Haller A, et al. Biophys Res Commun 2009;379:583–8. Thyroid transcription factor 1–a new prognostic factor in lung cancer: a 64. Pol A, Renkema GH, Tangerman A, Winkel EG, Engelke UF, de meta-analysis. Ann Oncol 2006;17:1673–6. Brouwer APM, et al. Mutations in SELENBP1, encoding a novel 44. Ruthenburg AJ, Allis CD, Wysocka J. Methylation of lysine 4 on histone H3: human methanethiol oxidase, cause extraoral halitosis. Nat Genet intricacy of writing and reading a single epigenetic mark: molecular cell. 2018;50:120–9. Mol Cell 2007;25:15–30. 65. Shamberger RJ. Relationship of selenium to cancer. I. Inhibitory 45. Creyghton MP, Cheng AW, Welstead GG, Kooistra T, Carey BW, Steine effect of selenium on carcinogenesis. J Natl Cancer Inst 1970;44: EJ, et al. Histone H3K27ac separates active from poised enhancers and 931–6. predicts developmental state. Proc Nat Acad Sci U S A 2010;107: 66. Shamberger RJ, Frost DV. Possible protective effect of selenium against 21931–6. human cancer. Can Med Assoc J 1969;100:682. 46. Calo E, Wysocka J. Modification of enhancer chromatin: what, how, and 67. Clark LC, Combs GF, Turnbull BW, Slate EH, Chalker DK, Chow J, why? Molecular Cell 2013;49:825–37. et al. Effects of selenium supplementation for cancer prevention in 47. Morey L, Helin K. Polycomb group protein-mediated repression of tran- patients with carcinoma of the skin. A randomized controlled trial. scription. Trends Biochem Sci 2010;35:323–32. Nutritional Prevention of Cancer Study Group. JAMA 1996;276: 48. Ferrari KJ, Scelfo A, Jammula S, Cuomo A, Barozzi I, Stutzer€ 1957–63. A, et al. Polycomb-Dependent H3K27me1 and H3K27me2 68. Lippman SM, Klein EA, Goodman PJ, Lucia MS, Thompson IM, Ford LG, regulate active transcription and enhancer fidelity. Mol Cell 2014;53: et al. Effect of selenium and vitamin E on risk of prostate cancer and other 49–62. cancers: the Selenium and Vitamin E cancer prevention trial (SELECT). 49. Ikeda K, Shaw-White JR, Wert SE, Whitsett JA. Hepatocyte nuclear factor 3 JAMA 2009;301:39–51. activates transcription of thyroid transcription factor 1 in respiratory 69.KarpDD,LeeSJ,KellerSM,WrightGS,AisnerS,BelinskySA,etal. epithelial cells. Mol Cell Biol 1996;16:3626–36. Randomized, double-blind, placebo-controlled, phase III chemopre- 50. DeFelice M, Silberschmidt D, DiLauro R, Xu Y, Wert SE, Weaver TE, et al. vention trial of selenium supplementation in patients with resected TTF-1 phosphorylation is required for peripheral lung morphogenesis, stage I non-small-cell lung cancer: ECOG 5597. J Clin Oncol 2013; perinatal survival, and tissue-specific gene expression. J Biol Chem 31:4179–87. 2003;278:35574–83. 70. Behne D, Kyriakopoulos A. Mammalian selenium-containing proteins. 51. Maeda Y, Dave V, Whitsett JA. Transcriptional control of lung morpho- Annu Rev Nutr 2001;21:453–73. genesis. Physiol Rev 2007;87:219–44. 71. Rao CV, Cooma I, Rodriguez JG, Simi B, El-Bayoumy K, Reddy BS. 52. Watanabe H, Francis JM, Woo MS, Etemad B, Lin W, Fries DF, et al. Chemoprevention of familial adenomatous polyposis development in the Integrated cistromic and expression analysis of amplified NKX2-1 in lung APCmin mouse model by 1,4-phenylene bis(methylene)selenocyanate. adenocarcinoma identifies LMO3 as a functional transcriptional target. Carcinogenesis 2000;21:617–21. Genes Develop 2013;27:197–210. 72. Patterson BH, Levander OA. Naturally occurring selenium compounds in 53. Oguchi H, Kimura S. Multiple transcripts encoded by the thyroid-specific cancer chemoprevention trials: a workshop summary. Cancer Epidemiol enhancer-binding protein (T/EBP)/thyroid-specific transcription factor-1 Biomarkers Prev 1997;6:63–9. (TTF-1) gene: evidence of autoregulation 1. Endocrinology 1998;139: 73. Reid ME, Duffield-Lillico AJ, Garland L, Turnbull BW, Clark LC, Marshall 1999–2006. JR. Selenium supplementation and lung cancer incidence: an update of the

1748 Mol Cancer Res; 16(11) November 2018 Molecular Cancer Research

Downloaded from mcr.aacrjournals.org on September 27, 2021. © 2018 American Association for Cancer Research. Published OnlineFirst July 12, 2018; DOI: 10.1158/1541-7786.MCR-18-0392

Selenbp1, Nkx2-1 Target, Suppresses Lung Cancer

nutritional prevention of cancer trial. Cancer Epidemiol Biomarkers Prev 77. Li C, Ling X, Yuan B, Minoo P. A novel DNA element mediates transcription 2002;11:1285–91. of Nkx2.1 by Sp1 and Sp3 in pulmonary epithelial cells. Biochim Biophys 74. Fritz H, Kennedy D, Fergusson D, Fernandes R, Cooley K, Seely A, et al. Acta 2000;1490:213–24. Selenium and lung cancer: a systematic review and meta analysis. PLoS One 78. Kang Y, Hebron H, Ozbun L, Mariano J, Minoo P, Jakowlew SB. Nkx2.1 2011;6:e26259. transcription factor in lung cells and a transforming growth factor-?1 75. Clarke N, Biscocho J, Kwei KA, Davidson JM, Sridhar S, Gong X, et al. heterozygous mouse model of lung carcinogenesis. Mol Carcinog Integrative genomics implicates EGFR as a downstream mediator in NKX2- 2004;40:212–31. 1 amplified non-small cell lung cancer. PLoS One 2015;10:1–17. 79. Herriges MJ, Tischfield DJ, Cui Z, Morley MP, Han Y, Babu A, et al. The 76. Shaw-White JR, Bruno MD, Whitsett JA. GATA-6 activates transcription of NANCI-Nkx2.1 gene duplex buffers Nkx2.1 expression to maintain lung thyroid transcription factor-1. J Biol Chem 1999;274:2658–64. development and homeostasis. Genes Dev 2017;31:889–903.

www.aacrjournals.org Mol Cancer Res; 16(11) November 2018 1749

Downloaded from mcr.aacrjournals.org on September 27, 2021. © 2018 American Association for Cancer Research. Published OnlineFirst July 12, 2018; DOI: 10.1158/1541-7786.MCR-18-0392

Tumor Suppressor Activity of Selenbp1, a Direct Nkx2-1 Target, in Lung Adenocarcinoma

Deborah R. Caswell, Chen-Hua Chuang, Rosanna K. Ma, et al.

Mol Cancer Res 2018;16:1737-1749. Published OnlineFirst July 12, 2018.

Updated version Access the most recent version of this article at: doi:10.1158/1541-7786.MCR-18-0392

Supplementary Access the most recent supplemental material at: Material http://mcr.aacrjournals.org/content/suppl/2018/07/12/1541-7786.MCR-18-0392.DC1

Cited articles This article cites 79 articles, 26 of which you can access for free at: http://mcr.aacrjournals.org/content/16/11/1737.full#ref-list-1

Citing articles This article has been cited by 3 HighWire-hosted articles. Access the articles at: http://mcr.aacrjournals.org/content/16/11/1737.full#related-urls

E-mail alerts Sign up to receive free email-alerts related to this article or journal.

Reprints and To order reprints of this article or to subscribe to the journal, contact the AACR Publications Department at Subscriptions [email protected].

Permissions To request permission to re-use all or part of this article, use this link http://mcr.aacrjournals.org/content/16/11/1737. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC) Rightslink site.

Downloaded from mcr.aacrjournals.org on September 27, 2021. © 2018 American Association for Cancer Research.