Published OnlineFirst March 15, 2016; DOI: 10.1158/0008-5472.CAN-15-3010

Cancer Tumor and Stem Cell Biology Research

Analysis of Liver Tumor-Prone Mouse Models of the Hippo Kinase Scaffold RASSF1A and SAV1 Xiaoying Zhang1, Cai Guo1, Xiwei Wu2, Arthur X. Li2, Limin Liu1, Walter Tsark3, Reinhard Dammann4, Hui Shen5, Steven L. Vonderfecht3, and Gerd P. Pfeifer1,5

Abstract

The tumor suppressor RASSF1A is epigenetically silenced the oncogenic IkB kinase TBK1 as the most enriched RASSF1A- in most human cancers. As a binding partner of the kinases MST1 interacting proteins. The transcriptome of Rassf1a / livers was þ and MST2, the mammalian orthologs of the Drosophila Hippo more deregulated than that of Sav1 / livers, and the transcrip- þ kinase, RASSF1A is a potential regulator of the Hippo tumor tome of Rassf1a / , Sav1 / livers was similar to that of suppressor pathway. RASSF1A shares these properties with the Rassf1a / mice. We found that the levels of TBK1 were scaffold protein SAV1. The role of this pathway in human cancer substantially upregulated in livers lacking Rassf1a. Furthermore, has remained enigmatic inasmuch as Hippo pathway compo- transcripts of several b-tubulin isoforms were increased in the nents are rarely mutated in tumors. Here we show that Rassf1a Rassf1a-deficient livers presumably reflecting a role of RASSF1A as homozygous knockout mice develop liver tumors. However, a microtubule-stabilizing protein. In human liver cancer, heterozygous deletion of Sav1 or codeletion of Rassf1a and Sav1 RASSF1A frequently undergoes methylation at the promoter but produced liver tumors with much higher efficiency than single this was not observed for MST1, MST2, or SAV1. Our results deletion of Rassf1a. Analysis of RASSF1A-binding partners by suggest a multifactorial role of RASSF1A in suppression of liver mass spectrometry identified the Hippo kinases MST1, MST2, and carcinogenesis. Cancer Res; 76(9); 2824–35. 2016 AACR.

Introduction stage of tumor progression (3). Mice with Rassf1a gene deletion are prone to carcinogen-induced and spontaneous tumorigenesis Liver cancer is one of the leading causes of cancer-related death (4–6). worldwide. The Ras association domain family 1 (RASSF1) gene is Although RASSF1A encodes a Ras association domain, this located at 3p21.3 in a region frequently deleted in protein is effectively recruited to activated Ras only through human solid tumors. The RASSF1 locus encodes two major heterodimerization with its closest homolog NORE1A/ transcripts referred to as RASSF1A and RASSF1C, which code for RASSF5 (7). RASSF1A is a potent microtubule binding and two proteins differing in their N-termini (1). The RASSF1A tran- stabilizing protein (8, 9). The noncatalytic protein RASSF1A script and protein are frequently absent from human tumors due is found in a complex with the kinases MST1 and MST2, the to methylation of the RASSF1A isoform–specific promoter (1, 2). mammalian orthologs of the tumor-suppressive Drosophila Methylation of the RASSF1A promoter often occurs at an early Hippo kinase and is associated with several other proteins of the Hippo pathway (10, 11). RASSF1A inhibits the dephos- 1Department of Biology, Beckman Research Institute, City of Hope, phorylation of MST1/MST2 by phosphatases thus promoting Duarte, California. 2Department of Molecular and Cellular Biology, an activated state of MST1/MST2 (12). The core components Beckman Research Institute, City of Hope, Duarte, California. 3Division of the Hippo pathway are highly conserved across species. In of Comparative Medicine, Beckman Research Institute, City of Hope, Duarte, California. 4Institute for Genetics, Justus-Liebig-University, mammals, the kinases MST1/MST2 can form a complex with Giessen, Germany. 5Center for Epigenetics, Van Andel Research Insti- another adaptor protein, SAV1, the mammalian counterpart tute, Grand Rapids, Michigan. of the Drosophila protein Salvador (13). MST1/MST2 kinases Note: Supplementary data for this article are available at Cancer Research activate two downstream kinases, LATS1 and LATS2 (14). Online (http://cancerres.aacrjournals.org/). The best-studied effector of the LATS kinases is YAP, an Current address for X. Zhang: Department of Pediatrics, University of Tennessee oncogenic transcriptional coactivator (15). Phosphorylated Health Science Center, Memphis, TN; current address for C. Guo, Department of YAP is retained in the cytoplasm, which prevents it from Biology, California Institute of Technology, Pasadena, CA; and current address activating (16). In this model, the Hippo for L. Liu, Department of Biochemistry and Molecular Genetics, University of kinases are negative regulators of YAP. Hippo signaling is Virginia, Charlottesville, VA. an important pathway for inhibition of cancer development Corresponding Author: Gerd P. Pfeifer, Center for Epigenetics, Van Andel in the mouse liver. Liver-specificdeletionofMst1 and Mst2 Research Institute, 333 Bostwick Ave. NE, Grand Rapids, MI 49503. Phone: combined, or deletion of Sav1 alone leads to liver enlarge- 616-234-5398; Fax: 616-234-5398; E-mail: [email protected] ment and effective induction of hepatocellular carcinoma or doi: 10.1158/0008-5472.CAN-15-3010 mixed liver tumors in mice (hepatocellular carcinoma/cho- 2016 American Association for Cancer Research. langiocarcinoma; refs. 17–20).

2824 Cancer Res; 76(9) May 1, 2016

Downloaded from cancerres.aacrjournals.org on October 3, 2021. © 2016 American Association for Cancer Research. Published OnlineFirst March 15, 2016; DOI: 10.1158/0008-5472.CAN-15-3010

Liver Tumors in Rassf1a and Sav1 Knockout Mice

However, recent genome sequencing of human hepatocellular bank on an institutionally approved protocol. Tumor-free carcinomas has failed to uncover mutations in components of the liver DNA from accident victims was obtained from Biochain. Hippo pathway (e.g., MST1/2, LATS1/2, SAV1; refs. 21–23). Genomic DNA was isolated from frozen tissue using the Although amplification and/or overexpression of YAP as well as DNeasy Blood & Tissue Kit (Qiagen). One microgram of diminished YAP phosphorylation has been seen in a fraction of genomic DNA was treated with sodium bisulfite and the human liver cancers (19, 24, 25), the overall impact of this samples were subjected to "combined bisulfite restriction pathway on human cancer is still unclear. analysis" (COBRA; ref. 29) using primers as described previ- Our current report addresses the role of Rassf1a deletion on liver ously (30). PCR products were digested with BstUI or TaqI physiology and malignant transformation in the mouse. For restriction enzymes (New England Biolabs). These enzymes comparison, we analyzed mice with single homozygous deletion only cut initially methylated DNA after bisulfite conversion of Rassf1a or heterozygous deletion of Sav1, and also created a and PCR. The digested PCR products were separated by þ Sav1 / Rassf1a / double knockout mouse model to study the electrophoresis on 2% agarose gels. function of these in liver tumorigenesis. Antibodies Materials and Methods Antibodies against phospho-IkBa (Ser32; catalog no. 2859), MST1 (#3682), YAP (#4912), phospho-YAP (Ser127; #4911), Generation of single and double Rassf1a and Sav1 gene TBK1 (#3013), NF-kB1 (#13586), and phospho-p65 (Ser536; knockout mice, tumor studies, and liver tissue collection #3033) were purchased from Cell Signaling Technology. Anti- Rassf1a / mice were derived previously (4). We created Sav1- bodies against YAP1 (sc-15407), TAZ (sc-48805), SAV1 (sc- mutant mice by using standard gene targeting methods to delete 135394), and NF-kB/p65 (sc-109x) were purchased from Santa exons 1 and 2 in 129S1 mouse embryonic stem cells. Homozy- Cruz Biotechnology. Lamin B1 (ab16048), TUBB2A gous Sav1 knockouts died in utero as reported previously (26). The (#ab170931), and IkBa (ab32518) antibodies were obtained Sav1 knockout colony was maintained in the heterozygous state. from Abcam. Anti-b-actin (A5441) and anti-FLAG (F7425) anti- The Rassf1a- and Sav1-mutant mice were crossed to Cre-Deleter/ bodies were from Sigma. Anti-MST2 (#1943-1) was from Epi- 129 mice (27) to remove the neomycin selection cassette. Typical tomics, and anti-GAPDH (627408) was from Genetex. Anti- genotyping results are shown in Supplementary Fig. S1. The Sav1 RASSF1A antibodies (M3-04 and M5-06) were raised against the and Rassf1a-mutant mice were maintained on the 129S1 back- þ þ þ þ N-terminal exon of RASSF1A (10). ground. Four mouse cohorts, Rassf1a / Sav1 / (wild-type), þ þ þ þ þ þ Rassf1a / Sav1 / , Rassf1a / Sav1 / , and Rassf1a / Sav1 / , were established and monitored for spontaneous tumor forma- RNA isolation, microarray analysis, and cDNA reverse tion, morbidity, or death. Mice were euthanized when visibly transcription moribund or whenever a growth/tumor became manifest, and a For total RNA isolation, we followed the instruction of the complete necroscopy was performed. Normal appearing liver or Ambion Purelink RNA mini kit. cDNA was synthesized from RNA tumor tissues were fixed in 10% neutral-buffered formalin, and template by using SuperScript III reverse transcriptase (Invitro- the remaining parts of liver tumor tissue and adjacent normal gen). Real-time PCR amplifications were performed in 96-well tissue were instantly frozen in liquid nitrogen and then stored at optical reaction plates with SYBR Green by using Taq DNA 80C. The formalin-fixed tissues were embedded in paraffin, polymerase (Qiagen). We used Affymetrix Gene 1.0-ST micro- and then stained with hematoxylin and eosin (H&E). arrays for analysis of mouse liver RNAs. Micro- array data were analyzed using the Bioconductor package "Array- Western blot and immunoprecipitation analyses Tools." Differentially expressed genes were selected by using a Frozen tissue was ground in liquid nitrogen by using a pestle, cutoff of P ( 0.005 and fold change 1.5. Heatmaps were created and the total proteins were extracted with extraction buffer [50 by using Cluster v3.0 and visualized by Java Treeview. Raw data mmol/L HEPES, pH 7.4, 137 mmol/L NaCl, 10% glycerol, 0.1% was deposited into the GEO database (accession number Triton-X 100, and 1 complete protease inhibitor cocktail GSE73911). (Roche)]. Alternatively, soluble cytosolic fractions were prepared following a published procedure (28). Protein samples were The Cancer Genome Atlas data analysis resolved on 12% SDS–polyacrylamide gels and transferred onto DNA methylation data packages (Version 13) for the The nitrocellulose membranes (Bio-Rad). Nitrocellulose membranes Cancer Genome Atlas (TCGA) Liver Hepatocellular Carcinoma were blocked with 5% nonfat milk (Bio-Rad) in PBST (PBS with (LIHC) project were downloaded from the TCGA data portal on 0.1% Tween 20) and sequentially incubated with primary anti- Dec 22, 2015. The R package Homo.sapiens was used to obtain the bodies and HRP-conjugated secondary antibodies (Jackson genomic locations of each gene, and GenomicRanges was used to ImmunoResearch Laboratories) in 5% nonfat milk in PBST. After get DNA methylation probes interrogating 1,500 bp of the incubation, blots were washed three times with PBST. We visu- genomic range of the target gene. For each heatmap, the probes alized the immunoreactive bands with Amersham ECL Prime (rows) are arranged by their genomic location, and samples are Western Blotting Detection Reagent following the manufacturer's clustered with hierarchical clustering to reveal patterns. Normal- instructions. Immunoprecipitation was performed as described ized isoform-level RNAseq data were downloaded from Firehose previously (12). (http://gdac.broadinstitute.org). We used uc003dae for RASSF1A and uc003dab for RASSF1C expression levels, and for other genes Combined bisulfite restriction analysis only expressed isoforms were used. Corresponding Log2-trans- Human liver cancer specimens and adjacent normal liver formed gene expression levels are plotted on top of each sample tissue were obtained from the City of Hope frozen tumor with a relative scale.

www.aacrjournals.org Cancer Res; 76(9) May 1, 2016 2825

Downloaded from cancerres.aacrjournals.org on October 3, 2021. © 2016 American Association for Cancer Research. Published OnlineFirst March 15, 2016; DOI: 10.1158/0008-5472.CAN-15-3010

Zhang et al.

Liver tumors developed in 19 of 45 mice (42%) of the þ þ Rassf1a / Sav1 / genotype (Table 1). These varied from small, solitary nodules to large, multinodular masses that occupied entire lobes of the liver. The tumors were designated as hepa- tocellular adenomas, hepatocellular carcinoma, mixed hepato- cellular adenomas (mHA), or mixed hepatocellular carcinoma (mHCC; Table 1). Microscopic features of hepatocellular ade- nomas and hepatocellular carcinoma were typical for these types of tumors (e.g., Supplementary Fig. S2) and diagnoses were based upon standard histopathologic criteria (31). Tumors designated as mixed hepatocellular adenomas or mHCC were morphologically distinct and did not clearly fit into any recognized classification of hepatocellular tumors. þ They are likely similar to tumors described in WW45 / (same þ as Sav1 / ) mice and designated as "mixed-type" liver tumors thought to arise from hepatic progenitor (oval) cells (17). The mHA and mHCC had a component with obvious hepatocel- Figure 1. lular morphology and a significant second component of cells Survival of Rassf1a and Sav1 knockout mice. Overall survival is plotted with small, oval to occasionally round or elongated nuclei and þ þ þ þ þ þ þ þ þ for Rassf1a / Sav1 / , Rassf1a / Sav1 / , Rassf1a / Sav1 / ,and indistinct cytoplasm. These "small cells" formed solid sheets or / þ/ Rassf1a Sav1 mice. irregular bands that penetrated into and sometimes surrounded nodules of neoplastic hepatocytes. The nuclei of the cells were occasionally arranged in rows or formed small, duct-like struc- Results tures (Fig. 2A–D). Because this second cellular component Tumor susceptibility of Rassf1a and Sav1 knockout mice was uniformly small, highly homogeneous, and lacked mitotic Previous reports suggested that both RASSF1A and SAV1 form a figures, the designation of mHA or mHCC was based solely complex through their C-terminal SARAH domains with the upon the morphology of the hepatocellular component of the þ þ Hippo kinases MST1 and MST2 (10, 11). To study the contribu- tumor. In comparison with the Rassf1a / Sav1 / mice, only 6 tion of these proteins to tumorigenesis, we generated mouse of 30 (20%) wild-type mice had hepatocellular tumors (includ- strains with germ line (whole body) deletion of Rassf1a (Rassf1a ing one microscopic tumor) and these were all hepatocellular isoform-specific knockout by deletion of exon 1) and/or Sav1. adenomas (Table 1), indicating increased tumor susceptibility Rassf1a homozygous gene knockout mice are viable and fertile and tumor progression in Rassf1a-deleted mice (P ¼ 0.046; c2 þ þ (4, 5), but, for the Sav1 gene knockout, only heterozygous test). For the Sav1-targeted mice, 47 of 49 (96%) Rassf1a / þ mice could be obtained. This is due to embryonic lethality of Sav1 / mice were found with liver tumors at ages greater than þ complete Sav1 deletion in mice as also reported previously (26). 1 year. Similarly, 35 of 39 Rassf1a / Sav1 / mice (90%) devel- þ Rassf1a / Sav1 / mice were viable and fertile. Mouse colonies of oped liver tumors, suggesting that loss of one copy of Sav1 is þ þ þ þ þ þ four different genotypes, Rassf1a / Sav1 / , Rassf1a / Sav1 / , a much more potent driver of liver tumor formation than loss þ þ þ þ Rassf1a / Sav1 / , and Rassf1a / Sav1 / , were established on of both copies of Rassf1a, making it likely that loss of Sav1 is the 129S1 genetic background and were observed for signs the dominant event in driving tumorigenesis in the double þ of tumor formation over extended periods of time. Overall knockout mice (Table 1). Most of the livers from Sav1 / survival was significantly different between wild-type and mice showed signs of oval cell hyperplasia (Fig. 2E and Sup- þ Rassf1a / Sav1 / mice (P ¼ 0.0063; log-rank test) and between plementary Fig. S3). This change was only occasionally observ- þ þ þ þ þ Rassf1a / Sav1 / and Rassf1a / Sav1 /- mice (P < 0.001) but ed in Rassf1a / Sav1 / mice and was not seen in wild-type þ þ not between wild-type and Rassf1a / Sav1 / (P ¼ 0.1016), mice. Oval cells are considered bipotential progenitors cap- þ þ þ wild-type and Rassf1a / Sav1 / (P ¼ 0.1144), or between able of differentiating into hepatocytes or cholangiocytes. þ þ þ þ Rassf1a / Sav1 / and Rassf1a / Sav1 / (P ¼ 0.287; Fig. 1). However, distinct features of CC, for example, clear gland þ þ Curiously, some Rassf1a / Sav1 / mice survived to very old age. formation, mucin production, and/or desmoplastic or fibrotic

Table 1. Liver tumor formation in Rassf1a- and Sav1-deficient mice Mean Median No. of mice with Hepatocellular Hepatocellular No. age age hepatocellular Hepatocellular CA, Hepatocellular adenoma, Rassf1a Sav1 of mice (days) (days) tumors CA mixed Hepatoblastoma adenoma mixeda wt þ/ 49 591 607 47 35 14 32 17 95.90% 71.40% 28.60% 0% 65.30% 34.70% /þ/ 39 583 626 35 30 9 1 31 15 89.70% 77% 23.10% 2.60% 79.50% 38.50% / wt 45 643 642 19 4 1 0 14 2 42% 8.90% 2.20% 0% 31.10% 4.40% wt wt 30 613 659 6 0 0 0 6 0 20% 0% 0% 0% 20% 0% aThe designation mixed tumor refers to tumors, which in addition to a hepatocellular component also had a "small cell/oval cell" component. Mean age and median age refer to overall survival of all mice of the respective genotype.

2826 Cancer Res; 76(9) May 1, 2016 Cancer Research

Downloaded from cancerres.aacrjournals.org on October 3, 2021. © 2016 American Association for Cancer Research. Published OnlineFirst March 15, 2016; DOI: 10.1158/0008-5472.CAN-15-3010

Liver Tumors in Rassf1a and Sav1 Knockout Mice

Figure 2. Characterization of liver tumors. A–D, liver tumor from a Sav1þ/þ Rassf1a/ mouse. Low magnification photomicrograph demonstrating multinodular "mixed" type tumor (A). Areas with obvious hepatocellular morphology (h) are intermixed with areas having "small cell" morphology (). This is shown at slightly higher magnification in B. At still higher magnification (C and D), the "small cell" component () of the mixed tumors is found to be composed of cells that have oval to occasionally round or elongated nuclei (C; arrows) and vary from having no organization (C) to well-defined (D) orientation into rows or duct-like structures (D; arrowheads). Areas of obvious hepatocellular morphology are labeled (h) in C and D. H&E staining. E, percentage of mice with oval cell hyperplasia of the liver. F, percentage of mice with bile duct hyperplasia. www.aacrjournals.org Cancer Res; 76(9) May 1, 2016 2827

Downloaded from cancerres.aacrjournals.org on October 3, 2021. © 2016 American Association for Cancer Research. Published OnlineFirst March 15, 2016; DOI: 10.1158/0008-5472.CAN-15-3010

Zhang et al.

Figure 3. Analysis of Hippo pathway components. A, Western blot analysis of Hippo pathway components in liver lysates of 6-month-old mice of the different genotypes. Each lane represents a sample from an individual mouse. B, reduction of SAV1 protein levels in liver þ þ þ tumors developing in Rassf1a / Sav1 / mice. C, expression differences of the YAP target gene Ctgf between liver tumors developing in Rassf1aþ/þSav1þ/ mice and adjacent normal liver in the same mice. The expression data were normalized relative to Gapdh expression. All tumors were hepatocellular carcinoma except tumors 3852, 3872, 3939, and 3979, which were adenomas.

þ reaction, were not obvious in mice with a Sav1 / or Rassf1a / age of 6 months (Fig. 3A). If the Hippo pathway is dysfunctional þ genotype although most of the livers from Sav1 / mice in gene-targeted mice, YAP protein should be less phosphorylated showed signs of oval cell expansion or bile duct hyperplasia and total levels of YAP should increase owing to diminished (Fig. 2E and F). degradation of YAP. First, we found that protein levels of both MST1 and MST2 were substantially reduced in the livers of þ þ þ þ þ Analysis of the Hippo pathway Rassf1a / Sav1 mice. In Rassf1a / Sav1 / mice, MST1 was Conditional biallelic deletion of the Sav1 gene in the mouse decreased in only one of three mice and MST2 levels were similar liver induces liver enlargement and liver tumor formation (17, to those in wild-type mice. In the double knockout mice, levels of þ þ þ 18). Although in our work close to 100% of Rassf1a / Sav1 / MST1 were strongly decreased, but MST2 appeared at wild-type þ mice or Rassf1a / Sav1 / mice developed liver tumors after 12 levels, suggesting complex regulation of MST levels upon loss of months of age, we did not find any liver enlargement before their scaffold proteins (Fig. 3A). tumors developed (Supplementary Fig. S4). This finding suggests However, levels of YAP and phosphorylated YAP, their ratio, or that one copy of Sav1 is sufficient to prevent liver enlargement but levels of the YAP homolog TAZ did not differ noticeably in livers is insufficient to prevent liver tumor formation. Complete loss of of all the genotypes (Fig. 3A). This suggested that although levels Rassf1a does not lead to liver enlargement either (Supplementary of MST1/2 were affected by deletion of SAV1, the standard read- Fig. S4). outs of the Hippo pathway, YAP and p-YAP, appeared largely As SAV1 and RASSF1A are components and potential regulators unaltered in nontumorous liver. Comparing liver tumors devel- þ of the Hippo pathway, we analyzed levels of MST1 and MST2, YAP oping in Sav1 / mice with adjacent normal liver tissue, we found protein levels and phosphorylation on serine 127 of YAP in much reduced levels of SAV1 protein in the majority of the tumors normal-appearing livers of mice of the different genotypes at the (Fig. 3B). Increased transcript level of the YAP target gene Ctgf is a

2828 Cancer Res; 76(9) May 1, 2016 Cancer Research

Downloaded from cancerres.aacrjournals.org on October 3, 2021. © 2016 American Association for Cancer Research. Published OnlineFirst March 15, 2016; DOI: 10.1158/0008-5472.CAN-15-3010

Liver Tumors in Rassf1a and Sav1 Knockout Mice

þ þ þ marker of a dysfunctional Hippo pathway (32). We determined to each other than to Rassf1a / Sav1 / mice (Fig. 5B; Supple- the gene expression levels of Ctgf in liver tumors developing in mentary Table S2). þ þ þ Rassf1a / Sav1 / mice. Thirteen of 14 liver tumor tissues analysis did not provide any enrichment for þ þ þ expressed increased levels of Ctgf compared with the adjacent specific gene or functional categories in the Rassf1a / Sav1 / normal tissues (Fig. 3C). The high level of Ctgf expression in mice. In contrast, the same analysis for the Rassf1a-deleted geno- two tumors that retained SAV1 protein may be due to Hippo/ types uncovered enrichment of two major functional categories: YAP-independent mechanisms. We did not retain sufficient the circadian clock and microtubule polymerization (Supplemen- þ þ numbers of frozen tumors from Rassf1a / Sav1 / mice to tary Fig. S5). This analysis did not reveal a gene category related to compare Ctgf levels. The data suggests that the remaining Sav1 the NF-kB pathway or to inflammatory responses in Rassf1a / copy or perhaps other components of the pathway in the livers genotype livers that could perhaps be traced back to increased þ of Sav1 / mice may eventually become disabled in founder levels of TBK1. Interestingly, however, we found that several cells giving rise to a dysfunctional Hippo pathway and pro- b-tubulin genes, including Tubb1, Tubb2a, Tubb2b, and Tubb5 were ducing liver tumors. However, the overall protein expression among the top 60 upregulated genes in Rassf1a knockout livers levels of YAP and phosphorylated YAP are not noticeably (Supplementary Table S1). These b-tubulin genes were signifi- þ þ altered in bulk liver tissue of Sav1 heterozygous or Rassf1a cantly upregulated (up to 4-fold) in Rassf1a / Sav1 / and þ þ þ þ homozygous mutant mice (Fig. 3A). Rassf1a / Sav1 / mice but not in Rassf1a / Sav1 / livers (Fig. 5C; Supplementary Table S1). In fact, most b-tubulin genes were TBK1, a new RASSF1A-interacting protein upregulated except for Tubb3 and Tubb4a, which are b-tubulin To identify potentially novel RASSF1A-interacting proteins, isoforms preferentially expressed in the nervous system. Genes we performed immunoprecipitation of endogenous RASSF1A coding for a- and g-tubulin proteins were not significantly dif- from HeLa cells. Coprecipitating proteins were identified by ferent in the knockouts. At the protein level, TUBB2A was mass spectrometry (Fig. 4A). The most significantly enriched increased only slightly in livers of Rassf1a knockout mice (Fig. 5C). proteins were three kinases, MST1, MST2, and TBK1 (Tank The enrichment of circadian factors in Rassf1a-deleted livers binding kinase 1). Figure 4B shows coimmunoprecipitation (Supplementary Fig. S5) was unexpected. One differentially of endogenous RASSF1A and TBK1. TBK1 is an oncogenic expressed gene was Usp2. The ubiquitin-specific protease USP2 noncanonical IkB kinase, which phosphorylates IkB to release is a key driver of the circadian clock (35). Interestingly, USP2 is NF-kB, which can induce the transcription of various proin- also a binding partner and negative regulator of the Rassf1a- flammatory and antiapoptotic genes (33, 34). We then ana- interacting kinase, TBK1 (36). Although Tbk1 itself was not þ þ lyzed protein expression levels of TBK1 in Rassf1a / Sav1 / transcriptionally misregulated in Rassf1a knockout mice, we mouse liver tissues and found that TBK1 protein expression was observed that at the transcript level, factors regulating consistently and substantially increased in all Rassf1a knockout TBK1 protein stability, including Usp2 and Dtx4, were dysregu- livers compared with the wild-type livers at the same age (18 lated. Usp2 was the second most significantly altered gene in þ þ months; Fig. 4C). A similar increase of TBK1 levels was con- Rassf1a / Sav1 / livers (downregulated 16-fold; Supplemen- sistently found at earlier and later ages of the mice and also was tary Table S1). Dtx4, the sixth most significantly altered (upregu- þ seen in Rassf1a /-Sav1 / double knockout mice but not in lated) gene (Supplementary Table S1) in this genotype is an E3 þ þ þ Rassf1a / Sav1 / mice (data not shown). ubiquitin ligase and key suppressor of TBK1 targeting this kinase The best-studied functions of TBK1 are through regulating the for degradation (37). The data suggest that a regulatory feedback classical NF-kB pathway via p65 phosphorylation. Therefore, we loop may exist between RASSF1A, TBK1, and its modifiers DTX4 determined the total protein expression of p65, its phosphory- and USP2, which is perturbed upon loss of RASSF1A. lated form p-p65 and its binding partner, NF-kB1/p50, which complexes with p65 and IkBa in the cytoplasm. p65 or p-p65 did Human liver cancers show frequent methylation of the þ þ not show a notable increase in Rassf1a / Sav1 / mice. Also, RASSF1A promoter, but minimal methylation of other Hippo p105 and its truncated form p50 did not consistently show higher pathway genes protein levels upon Rassf1a deletion (Fig. 4C). Promoter hypermethylation of tumor suppressor genes is a potential contributor to tumor formation. As mutations in The liver transcriptomes of Sav1 and Rassf1a knockout mice genes of the Hippo pathway are uncommon in human cancers We isolated RNA from livers of the four mouse genotypes at 1 (16), including liver cancer, we analyzed several genes of the year of age processing four biologic replicates for each genotype. pathway for methylation at their promoters in 22 pairs of The RNA was analyzed on Affymetrix 1.0-ST microarrays. The four human normal tissue and tumor tissue of the liver (Supple- genotypes could be well separated by heatmaps (Fig. 5A). Using a mentary Figs. S6 and S7; Supplementary Table S3). Liver cancer P value cutoff of P < 0.005, we found a relatively small number of patients often have predisposing conditions including cirrho- þ þ þ upregulated or downregulated genes in Rassf1a / Sav1 / mice sis, HBV, or HCV (Supplementary Table S3). We analyzed relative to wild-type mice (Fig. 5B; Supplementary Tables S1 and promoter methylation of the target genes by using COBRA on S2). As expected, levels of Sav1 transcripts were reduced to about normal tissues and tumor tissues (Supplementary Figs. S6 and 50%. However, at the same significance level, the transcriptomes S7). Seventeen of 22 (77%) of the liver cancer patients had þ þ þ of Rassf1a / Sav1 / and Rassf1a / Sav1 / mice were more higher RASSF1A methylation in the tumor tissues compared þ substantially altered in comparison with those of Rassf1a / with the adjacent normal tissues (Supplementary Fig. S6A). þ þ Sav1 / mice (Fig. 5B; Supplementary Tables S1 and S2). There Partial methylation of RASSF1A was seen in some tumor- was some overlap between the genotypes. Most notably, expres- adjacent livers and in liver DNA from an 83-year-old tumor- þ þ sion differences in both directions of Rassf1a / Sav1 / and free liver. Our RASSF1A promoter methylation data are þ Rassf1a / Sav1 / mice relative to wild-type were more similar consistent with other groups' earlier findings showing extensive

www.aacrjournals.org Cancer Res; 76(9) May 1, 2016 2829

Downloaded from cancerres.aacrjournals.org on October 3, 2021. © 2016 American Association for Cancer Research. Published OnlineFirst March 15, 2016; DOI: 10.1158/0008-5472.CAN-15-3010

Zhang et al.

Figure 4. RASSF1A binds to and regulates the levels of the oncogenic kinase TBK1. A, immunoprecipitation of RASSF1A-interacting proteins from HeLa cells. Endogenous RASSF1A was immunoprecipitated with two anti-RASSF1A antibodies, M3-04 and M5-06. Prominent bands on Coomassie blue–stained gels were excised and proteins indicated by arrows were identified by mass spectrometry. B, proteins from HeLa cells were immunoprecipitated with anti-RASSF1A, anti-TBK1 antibodies, or control IgG (IP). After separation on SDS gels, Western blot analyses (WB) were conducted with the indicated antibodies. , IgG bands. C, soluble cytosolic liver protein extracts from mice of different genotypes at 18 months of age were analyzed by Western blot analysis for levels of TBK1 and proteins of the NF-kB pathway. Each lane represents a sample from an individual mouse.

methylation of RASSF1A in hepatocellular carcinoma (38–40). methylation of the RASSF1A but not the RASSF1C or SAV1 However, we detected no methylation at the SAV1 promoter promoters in hepatocellular carcinoma in the TCGA dataset. The (Supplementary Fig. S6B). Furthermore, we found no methyl- more highly methylated tumors (indicated by a dotted line in Fig. ation in human liver tumors at the promoters of the genes 6A) had the lowest expression of RASSF1A. Although the TCGA MST1 and MST2 (Supplementary Fig. S7). sample number for normal liver is still limited, there seems to be a To support this analysis, we analyzed hepatocellular carcinoma trend for lower expression of SAV1 in some hepatocellular car- DNA methylation data from TCGA (Fig. 6). This analysis shows cinomas. However, about half of the hepatocellular carcinomas

2830 Cancer Res; 76(9) May 1, 2016 Cancer Research

Downloaded from cancerres.aacrjournals.org on October 3, 2021. © 2016 American Association for Cancer Research. Published OnlineFirst March 15, 2016; DOI: 10.1158/0008-5472.CAN-15-3010

Liver Tumors in Rassf1a and Sav1 Knockout Mice

Figure 5. Gene expression differences in livers from mice with Rassf1a and/or Sav1 deletion. A, heatmap showing differentially expressed genes in livers of 12-month-old mice with genotypes Rassf1aþ/þSav1þ/þ (WT), Rassf1aþ/þSav1þ/ (SA), Rassf1a/-Sav1þ/þ (RA), and Rassf1a/Sav1þ/ (RA_SA). Samples with the same genotype cluster together. B, Venn diagram showing differentially expressed genes (P ( 0.005), upregulated or downregulated, in the livers of the three knockout genotypes versus wild-type liver. C, differences in expression levels for the b-tubulin genes Tubb1, Tubb2a, Tubb2b,andTubb5 as determined from the þ þ microarray data. Levels of TUBB2A protein in wild-type and Rassf1a / Sav1 / mouse livers were assessed by Western blot analysis using b-actin as a control. Each lane represents a sample from an individual mouse.

www.aacrjournals.org Cancer Res; 76(9) May 1, 2016 2831

Downloaded from cancerres.aacrjournals.org on October 3, 2021. © 2016 American Association for Cancer Research. Published OnlineFirst March 15, 2016; DOI: 10.1158/0008-5472.CAN-15-3010

Zhang et al.

Figure 6. Methylation and expression of RASSF1A and SAV1 in human hepatocellular carcinoma. A, data for the RASSF1 locus were extracted from TCGA data portal (http://cancergenome.nih.gov/). Expression (RNA-seq) and DNA methylation (Illumina 450 k array) data were displayed for individual specimens (each column). The methylation data are plotted for individual CpG sites along the locus (each row). The dotted bar above the methylation data indicates groups of hepatocellular carcinoma specimens with higher level of DNA methylation at the RASSF1A CpG island that also show lack of gene expression (green). White bars show samples lacking expression data. B, TCGA data for the SAV1 locus.

2832 Cancer Res; 76(9) May 1, 2016 Cancer Research

Downloaded from cancerres.aacrjournals.org on October 3, 2021. © 2016 American Association for Cancer Research. Published OnlineFirst March 15, 2016; DOI: 10.1158/0008-5472.CAN-15-3010

Liver Tumors in Rassf1a and Sav1 Knockout Mice

still express high levels of SAV1 (Fig. 6B). Analysis of TCGA data Rassf1a homozygous deletion is milder than that of Sav1 single for MST1 and MST2 also show a lack of methylation at their copy deletion but is still pronounced. It can be hypothesized that promoter-associated CpG islands (Supplementary Fig. S8). MST1 loss of Rassf1a cooperates with other tumor-driving events in liver is expressed at rather high levels and is downregulated in very few cancer formation as previously demonstrated for hematologic hepatocellular carcinomas. Unexpectedly, MST2 is upregulated in malignancies (6). about half the hepatocellular carcinomas relative to normal liver Besides its involvement in the Hippo pathway, RASSF1A has tissues (Supplementary Fig. S8B). These data suggest that the additional functions that when impaired may contribute to RASSF1A gene is frequently and extensively methylated and malignant transformation. As an established microtubule bind- silenced in human liver tumors but that SAV1 or MST1/2 are less ing and stabilizing protein (8), loss of RASSF1A may interfere with commonly affected. events such as cell migration (44) and mitosis (8, 10, 45, 46). Defects in cell migration or mitosis are likely contributors to Discussion oncogenesis. Interestingly, we observed that loss of Rassf1a leads to increased expression of a number of b-tubulin isoform-encod- In this study, we created three mouse models in which scaffold ing genes including Tubb1, Tubb2a, Tubb2b, and Tubb5. This protein binding partners of the mammalian Hippo kinases MST1 finding suggests that loss of microtubule stabilization by RASSF1A and MST2 were deleted. Liver-specificdeletionofMst1 and Mst2 may invoke compensatory events by increasing transcription of together (18–20), as well as liver-specific homozygous deletion of b-tubulin genes. However, this compensation may be insufficient Sav1 (17, 18), produced hepatomegaly and effectively induced contributing to the tumor-promoting phenotype of Rassf1a loss. hepatocellular carcinomas in mice. These deletions increase the We identified the kinase TBK1 as a strong RASSF1A-interacting pool of hepatic progenitor cells (oval cells), a phenomenon we also protein (Fig. 4). This interaction was further proven to be relevant þ observed in our Sav1 / mice and to a lesser extent in Rassf1a / in vivo by the clearly enhanced levels of TBK1 protein in every liver þ mice. Sav1 / mice also developed liver cancer in a previous study sample obtained from mice with Rassf1a deletion. The absence of (17). It remains unknown why constitutive deletion of Sav1 in the a protein commonly leads to diminished levels of its binding mouse has such a prominent effect on liver tumor formation and partner(s) owing to reduced protein stability in absence of the rarely affects other tissues even though YAP, its best-studied down- partner. However, this is not the case with TBK1, which showed stream effector, plays a general role in many cell types. increased levels upon deletion of Rassf1a. One possible explana- In our comparative mouse studies, we determined that homo- tion is that RASSF1A promotes degradation of TBK1 upon bind- zygous deletion of Rassf1a causes liver tumor susceptibility. In ing, a scenario, which requires additional investigation. In any previous studies, liver tumors were not observed as a common event, the data suggest that RASSF1A is a negative regulator of the type of tumor in Rassf1a / mice (4, 5). These mice were on a oncogenic kinase TBK1 because in the absence of RASSF1A, the different genetic background (129SvJ C57BL/6). The hetero- levels of this kinase are increased. TBK1 is best known as an zygous deletion of Sav1 was much more potent in promoting liver activator of the NF-kB pathway and has oncogenic properties (33, cancer (Table 1) even though both RASSF1A and SAV1 are binding 34). Deletion of Rassf1a did not lead to a noticeable activation of partners and activators of the MST kinases (10, 14, 41). Although the pathway and of NF-kB target genes in the liver (Fig. 4; we are unable to offer a definitive explanation for these differences Supplementary Table S1). We also did not observe an increase in tumor susceptibility between the Sav1 and Rassf1a deleted of inflammatory cells in sections of livers from Rassf1a / mice. genotypes at this time, one likely possibility is the redundancy For these reasons, NF-kB pathway–independent functions of of the RASSF protein family. While there is only one SAV1-like TBK1 may be important for tumor promotion in the liver and protein in mammalian genomes, there are 10 RASSF proteins these may be increased upon loss of Rassf1a. Mouse models have defined by the presence of a RAS association domain (42). Six of shown that TBK1 is critical in protecting the liver from apoptosis these proteins (RASSF1-6) have a C-terminal RAS association (47). TBK1 can directly activate AKT by phosphorylating the domain. The proteins most closely related to RASSF1 are RASSF3 activation loop (48). Furthermore, it has been shown that TBK1 and RASSF5 (also known as NORE1). We found that the Rassf5 is a mitotic kinase that phosphorylates and activates Polo-like gene was upregulated 1.24-fold in the liver of Rassf1a knockout kinase 1 (PLK1; ref. 49). Interference with mitotic progression is mice (Supplementary Table S1) but other Rassf genes were not generally a potential tumor-promoting event. Importantly, a role changed. Deletion of multiple members of the C-terminal Rassf of RASSF1A and the Hippo pathway in mitotic exit and prevention family may be necessary to elicit a more dramatic phenotype in of polyploidy has been proposed previously (10), suggesting that animal models. loss of RASSF1A may have a multipronged effect on correct Mice with heterozygous deletion of Sav1 or homozygous dele- execution of mitosis, which may explain aspects of the tumor- tion of Rassf1a did not show any signs of liver enlargement prior to predisposing phenotype of Rassf1a loss. development of tumors in contrast to mice with conditional Finally, our studies add information to the possible role of the ablation of Sav1 or Mst1 and Mst2 in the liver (17–20) or liver- Hippo pathway in human liver cancer. Several mouse models specific overexpression of Yap (43). These data suggest that the with deletions of core components of the pathway clearly suggest core Hippo pathway restraining tissue overgrowth by inhibiting that failure of Hippo signaling promotes liver cancer (50). How- YAP activity in the liver is functional in the absence of Rassf1a or in ever, mutations of SAV1, RASSF1A, MST1, MST2,orLATS1, LATS2 presence of only one copy of Sav1. However, these deletions led to are extremely rare in human cancer (16). Although upstream the formation of liver tumors. It is likely that the remaining Hippo signaling regulators such as FAT1, FAT4,orNF2 may be functional copy of Sav1 is lost or disabled in tumor-initiating mutated in certain types of malignancies, they are not commonly þ cells of Sav1 / livers. The downstream consequences are activa- mutated in human liver cancer either. Therefore, one may con- tion of YAP-dependent expression of progrowth genes as exem- sider epigenetic inactivation of Hippo pathway tumor suppressor plified by increased expression of Ctgf (Fig. 3C). The phenotype of genes as a potential tumor-driving event. However, our own

www.aacrjournals.org Cancer Res; 76(9) May 1, 2016 2833

Downloaded from cancerres.aacrjournals.org on October 3, 2021. © 2016 American Association for Cancer Research. Published OnlineFirst March 15, 2016; DOI: 10.1158/0008-5472.CAN-15-3010

Zhang et al.

studies (Supplementary Figs. S6 and S7) along with previous data Authors' Contributions (38–40) and data obtained from TCGA (Fig. 6 and Supplemen- Conception and design: C. Guo, G.P. Pfeifer tary Fig. S8) suggest that the only Hippo pathway gene commonly Development of methodology: L. Liu, R. Dammann, S.L. Vonderfecht silenced by epigenetic processes in hepatocellular carcinoma is Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): X. Zhang, C. Guo, X. Wu, L. Liu, W. Tsark, RASSF1A. This is particularly puzzling inasmuch as epigenetic S.L. Vonderfecht inactivation of SAV1 would be expected to be a much more Analysis and interpretation of data (e.g., statistical analysis, biostatistics, efficient tumor-promoting event than silencing of RASSF1A, given computational analysis): X. Zhang, X. Wu, A.X. Li, L. Liu, H. Shen, the data from our mouse models. Although SAV1 seems to be S.L. Vonderfecht, G.P. Pfeifer downregulated in a subset of hepatocellular carcinoma, MST2 is Writing, review, and/or revision of the manuscript: X. Zhang, X. Wu, A.X. Li, overexpressed rather than downregulated in many of the TCGA L. Liu, R. Dammann, S.L. Vonderfecht, G.P. Pfeifer Administrative, technical, or material support (i.e., reporting or organizing samples. Furthermore, although Sav1-mutant livers are charac- data, constructing databases): S.L. Vonderfecht terized by an expansion of the oval cell pool, recent data has cast Study supervision: G.P. Pfeifer doubt onto whether these progenitor cells can be considered general precursors of hepatocellular carcinoma in various liver Grant Support cancer models, as opposed to mature hepatocytes, which may This work was supported by a grant of the University of California Tobacco- undergo dedifferentiation (51). Future studies are needed to Related Disease Research Program (17RT-0116) and by NIH grant CA084469 to resolve the complex roles of the Hippo pathway in human G.P. Pfeifer. hepatocellular carcinoma. The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact. Disclosure of Potential Conflicts of Interest S.L. Vonderfecht has provided expert testimony for UCLA. No potential Received November 3, 2015; revised January 28, 2016; accepted February 25, conflicts of interest were disclosed by the other authors. 2016; published OnlineFirst March 15, 2016.

References 1. Dammann R, Li C, Yoon JH, Chin PL, Bates S, Pfeifer GP. Epigenetic 16. Harvey KF, Zhang X, Thomas DM. The Hippo pathway and human cancer. inactivation of a RAS association domain family protein from the lung Nat Rev Cancer 2013;13:246–57. tumour suppressor locus 3p21.3. Nat Genet 2000;25:315–9. 17. Lee KP, Lee JH, Kim TS, Kim TH, Park HD, Byun JS, et al. The Hippo- 2. Agathanggelou A, Cooper WN, Latif F. Role of the Ras-association domain Salvador pathway restrains hepatic oval cell proliferation, liver size, and family 1 tumor suppressor gene in human cancers. Cancer Res 2005;65: liver tumorigenesis. Proc Natl Acad Sci U S A 2010;107:8248–53. 3497–508. 18. Lu L, Li Y, Kim SM, Bossuyt W, Liu P, Qiu Q, et al. Hippo signaling is a 3. Grawenda AM, O'Neill E. Clinical utility of RASSF1A methylation in potent in vivo growth and tumor suppressor pathway in the mammalian human malignancies. Br J Cancer 2015;113:372–81. liver. Proc Natl Acad Sci U S A 2010;107:1437–42. 4. Tommasi S, Dammann R, Zhang Z, Wang Y, Liu L, Tsark WM, et al. Tumor 19. Zhou D, Conrad C, Xia F, Park JS, Payer B, Yin Y, et al. Mst1 and Mst2 susceptibility of Rassf1a knockout mice. Cancer Res 2005;65:92–8. maintain hepatocyte quiescence and suppress hepatocellular carcinoma 5. van der Weyden L, Tachibana KK, Gonzalez MA, Adams DJ, Ng BL, Petty R, development through inactivation of the Yap1 oncogene. Cancer Cell et al. The RASSF1A isoform of RASSF1 promotes microtubule stability and 2009;16:425–38. suppresses tumorigenesis. Mol Cell Biol 2005;25:8356–67. 20. Song H, Mak KK, Topol L, Yun K, Hu J, Garrett L, et al. Mammalian Mst1 6. van der Weyden L, Papaspyropoulos A, Poulogiannis G, Rust AG, Rashid M, and Mst2 kinases play essential roles in organ size control and tumor Adams DJ, et al. Loss of RASSF1A synergizes with deregulated RUNX2 suppression. Proc Natl Acad Sci U S A 2010;107:1431–6. signaling in tumorigenesis. Cancer Res 2012;72:3817–27. 21. Fujimoto A, Furuta M, Shiraishi Y, Gotoh K, Kawakami Y, Arihiro K, et al. 7. Ortiz-Vega S, Khokhlatchev A, Nedwidek M, Zhang XF, Dammann R, Whole-genome mutational landscape of liver cancers displaying biliary Pfeifer GP, et al. The putative tumor suppressor RASSF1A homodimerizes phenotype reveals hepatitis impact and molecular diversity. Nat Commun and heterodimerizes with the Ras-GTP binding protein Nore1. Oncogene 2015;6:6120. 2002;21:1381–90. 22. Schulze K, Imbeaud S, Letouze E, Alexandrov LB, Calderaro J, Rebouissou S, 8. Liu L, Tommasi S, Lee DH, Dammann R, Pfeifer GP. Control of microtubule et al. Exome sequencing of hepatocellular carcinomas identifies new stability by the RASSF1A tumor suppressor. Oncogene 2003;22:8125–36. mutational signatures and potential therapeutic targets. Nat Genet 9. El-Kalla M, Onyskiw C, Baksh S. Functional importance of RASSF1A 2015;47:505–11. microtubule localization and polymorphisms. Oncogene 2010;29: 23. Huang J, Deng Q, Wang Q, Li KY, Dai JH, Li N, et al. Exome sequencing of 5729–40. hepatitis B virus-associated hepatocellular carcinoma. Nat Genet 2012; 10. Guo C, Tommasi S, Liu L, Yee JK, Dammann R, Pfeifer GP. RASSF1A is part 44:1117–21. of a complex similar to the Drosophila Hippo/Salvador/Lats tumor-sup- 24. Zhao B, Wei X, Li W, Udan RS, Yang Q, Kim J, et al. Inactivation of YAP pressor network. Curr Biol 2007;17:700–5. oncoprotein by the Hippo pathway is involved in cell contact inhibition 11. Khokhlatchev A, Rabizadeh S, Xavier R, Nedwidek M, Chen T, Zhang XF, and tissue growth control. Genes Dev 2007;21:2747–61. et al. Identification of a novel Ras-regulated proapoptotic pathway. Curr 25. Zender L, Spector MS, Xue W, Flemming P, Cordon-Cardo C, Silke J, et al. Biol 2002;12:253–65. Identification and validation of oncogenes in liver cancer using an inte- 12. Guo C, Zhang X, Pfeifer GP. The tumor suppressor RASSF1A prevents grative oncogenomic approach. Cell 2006;125:1253–67. dephosphorylation of the mammalian STE20-like kinases MST1 and 26. Lee JH, Kim TS, Yang TH, Koo BK, Oh SP, Lee KP, et al. A crucial role of MST2. J Biol Chem 2011;286:6253–61. WW45 in developing epithelial tissues in the mouse. EMBO J 2008;27: 13. Tapon N, Harvey KF, Bell DW, Wahrer DC, Schiripo TA, Haber D, et al. 1231–42. Salvador promotes both cell cycle exit and apoptosis in Drosophila and is 27. Tang SH, Silva FJ, Tsark WM, Mann JR. A Cre/loxP-deleter transgenic line in mutated in human cancer cell lines. Cell 2002;110:467–78. mouse strain 129S1/SvImJ. Genesis 2002;32:199–202. 14. Harvey K, Tapon N. The Salvador-Warts-Hippo pathway - an emerging 28. Dimauro I, Pearson T, Caporossi D, Jackson MJ. A simple protocol for the tumour-suppressor network. Nat Rev Cancer 2007;7:182–91. subcellular fractionation of skeletal muscle cells and tissue. BMC Res Notes 15. Huang J, Wu S, Barrera J, Matthews K, Pan D. The 2012;5:513. coordinately regulates cell proliferation and apoptosis by inactivating 29. Xiong Z, Laird PW. COBRA: a sensitive and quantitative DNA methylation Yorkie, the Drosophila Homolog of YAP. Cell 2005;122:421–34. assay. Nucleic Acids Res 1997;25:2532–4.

2834 Cancer Res; 76(9) May 1, 2016 Cancer Research

Downloaded from cancerres.aacrjournals.org on October 3, 2021. © 2016 American Association for Cancer Research. Published OnlineFirst March 15, 2016; DOI: 10.1158/0008-5472.CAN-15-3010

Liver Tumors in Rassf1a and Sav1 Knockout Mice

30. Schagdarsurengin U, Richter AM, Hornung J, Lange C, Steinmann K, 1A in hepatitis B virus-associated hepatocellular carcinomas. Clin Cancer Dammann RH. Frequent epigenetic inactivation of RASSF2 in thyroid Res 2003;9:3376–82. cancer and functional consequences. Mol Cancer 2010;9:264. 41. Udan RS, Kango-Singh M, Nolo R, Tao C, Halder G. Hippo promotes 31. Thoolen B, Maronpot RR, Harada T, Nyska A, Rousseaux C, Nolte T, et al. proliferation arrest and apoptosis in the Salvador/Warts pathway. Nat Cell Proliferative and nonproliferative lesions of the rat and mouse hepato- Biol 2003;5:914–20. biliary system. Toxicol Pathol 2010;38(7 Suppl):5S–81S. 42. Pfeifer GP, Dammann R, Tommasi S. RASSF proteins. Curr Biol 2010;20: 32. Zhao B, Ye X, Yu J, Li L, Li W, Li S, et al. TEAD mediates YAP-dependent gene R344–5. induction and growth control. Genes Dev 2008;22:1962–71. 43. Dong J, Feldmann G, Huang J, Wu S, Zhang N, Comerford SA, et al. 33. Barbie DA, Tamayo P, Boehm JS, Kim SY, Moody SE, Dunn IF, et al. Elucidation of a universal size-control mechanism in Drosophila and Systematic RNA interference reveals that oncogenic KRAS-driven cancers mammals. Cell 2007;130:1120–33. require TBK1. Nature 2009;462:108–12. 44. Dallol A, Agathanggelou A, Tommasi S, Pfeifer GP, Maher ER, Latif F. 34. Chien Y, Kim S, Bumeister R, Loo YM, Kwon SW, Johnson CL, et al. Involvement of the RASSF1A tumor suppressor gene in controlling cell RalB GTPase-mediated activation of the IkappaB family kinase TBK1 migration. Cancer Res 2005;65:7653–9. couples innate immune signaling to tumor cell survival. Cell 2006; 45. Dallol A, Agathanggelou A, Fenton SL, Ahmed-Choudhury J, Hesson L, Vos 127:157–70. MD, et al. RASSF1A interacts with microtubule-associated proteins and 35. Scoma HD, Humby M, Yadav G, Zhang Q, Fogerty J, Besharse JC. The de- modulates microtubule dynamics. Cancer Res 2004;64:4112–6. ubiquitinylating enzyme, USP2, is associated with the circadian clockwork 46. Vos MD, Martinez A, Elam C, Dallol A, Taylor BJ, Latif F, et al. A role for the and regulates its sensitivity to light. PLoS One 2011;6:e25382. RASSF1A tumor suppressor in the regulation of tubulin polymerization 36. Zhang L, Zhao X, Zhang M, Zhao W, Gao C. Ubiquitin-specific protease 2b and genomic stability. Cancer Res 2004;64:4244–50. negatively regulates IFN-beta production and antiviral activity by targeting 47. Bonnard M, Mirtsos C, Suzuki S, Graham K, Huang J, Ng M, et al. Deficiency TANK-binding kinase 1. J Immunol 2014;193:2230–7. of T2K leads to apoptotic liver degeneration and impaired NF-kappaB- 37. Cui J, Li Y, Zhu L, Liu D, Songyang Z, Wang HY, et al. NLRP4 negatively dependent gene transcription. EMBO J 2000;19:4976–85. regulates type I interferon signaling by targeting the kinase TBK1 for 48. Ou YH, Torres M, Ram R, Formstecher E, Roland C, Cheng T, et al. TBK1 degradation via the ubiquitin ligase DTX4. Nat Immunol 2012;13: directly engages Akt/PKB survival signaling to support oncogenic trans- 387–95. formation. Mol Cell 2011;41:458–70. 38. Schagdarsurengin U, Wilkens L, Steinemann D, Flemming P, Kreipe HH, 49. Kim JY, Welsh EA, Oguz U, Fang B, Bai Y, Kinose F, et al. Dissection of TBK1 Pfeifer GP, et al. Frequent epigenetic inactivation of the RASSF1A gene in signaling via phosphoproteomics in lung cancer cells. Proc Natl Acad Sci hepatocellular carcinoma. Oncogene 2003;22:1866–71. U S A 2013;110:12414–9. 39. Zhang YJ, Ahsan H, Chen Y, Lunn RM, Wang LY, Chen SY, et al. High 50. Avruch J, Zhou D, Fitamant J, Bardeesy N. Mst1/2 signalling to Yap: frequency of promoter hypermethylation of RASSF1A and p16 and its gatekeeper for liver size and tumour development. Br J Cancer 2011;104: relationship to aflatoxin B1-DNA adduct levels in human hepatocellular 24–32. carcinoma. Mol Carcinog 2002;35:85–92. 51. Font-Burgada J, Shalapour S, Ramaswamy S, Hsueh B, Rossell D, Umemura 40. Zhong S, Yeo W, Tang MW, Wong N, Lai PB, Johnson PJ. Intensive A, et al. Hybrid periportal hepatocytes regenerate the injured liver without hypermethylation of the CpG island of Ras association domain family giving rise to cancer. Cell 2015;162:766–79.

www.aacrjournals.org Cancer Res; 76(9) May 1, 2016 2835

Downloaded from cancerres.aacrjournals.org on October 3, 2021. © 2016 American Association for Cancer Research. Published OnlineFirst March 15, 2016; DOI: 10.1158/0008-5472.CAN-15-3010

Analysis of Liver Tumor-Prone Mouse Models of the Hippo Kinase Scaffold Proteins RASSF1A and SAV1

Xiaoying Zhang, Cai Guo, Xiwei Wu, et al.

Cancer Res 2016;76:2824-2835. Published OnlineFirst March 15, 2016.

Updated version Access the most recent version of this article at: doi:10.1158/0008-5472.CAN-15-3010

Supplementary Access the most recent supplemental material at: Material http://cancerres.aacrjournals.org/content/suppl/2016/03/15/0008-5472.CAN-15-3010.DC1

Cited articles This article cites 51 articles, 18 of which you can access for free at: http://cancerres.aacrjournals.org/content/76/9/2824.full#ref-list-1

Citing articles This article has been cited by 2 HighWire-hosted articles. Access the articles at: http://cancerres.aacrjournals.org/content/76/9/2824.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://cancerres.aacrjournals.org/content/76/9/2824. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC) Rightslink site.

Downloaded from cancerres.aacrjournals.org on October 3, 2021. © 2016 American Association for Cancer Research.