Although the 14-3-3σ gene was initially identified as a p53 target gene in colorectal cancer cells, its potential role in intestinal tumorigenesis has remained unknown. Here we determined that 14-3-3σ expression is significantly downregulated in primary human colorectal cancer when compared with adjacent normal colonic tissue in patient samples. Downregulation of 14-3-3σ in primary colorectal cancers was significantly associated with p53 mutation, increasing tumor stage, distant metastasis, and poor patient survival. Poor survival was more significantly associated with decreased 14-3-3σ expression in p53 wild-type than in p53-mutant colorectal cancers. 14-3-3σ expression was detected in enterocytes of the transit amplifying zone and gradually increased towards the apical villi in the small intestinal epithelium. In small and large intestinal epithelia and adenomas, 14-3-3σ expression was upregulated in differentiated areas. Deletion of 14-3-3σ in ApcMin mice increased the number and size of adenomas in the small intestine and colon, shortening the median survival by 64 days. 14-3-3σ–deficient adenomas displayed increased proliferation and decreased apoptosis, as well as increased dysplasia. In adenomas, loss of 14-3-3σ promoted acquisition of a mesenchymal-like gene expression signature, which was also found in colorectal cancers from patients with poor relapse-free survival. The transcriptional programs controlled by the 14-3-3σ-interacting factors SNAIL, c-JUN, YAP1, and FOXO1 were activated by deletion of 14-3-3σ, potentially contributing to the enhanced tumor formation and growth. Taken together, these results provide genetic evidence of a tumor-suppressor function of 14-3-3σ in the intestine.

Significance:

Downregulation of 14-3-3σ in colorectal cancer is associated with metastasis and poor survival of patients, and its inactivation in a murine tumor model drives intestinal tumor formation and epithelial-mesenchymal transition.

Colorectal cancer accounts for the second-highest mortality among the 10 most common cancers worldwide. Over 1.8 million new colorectal cancer cases and 881,000 deaths were estimated to occur in 2018, accounting for about 1 in 10 cancer cases and deaths (1). Treatment of advanced colorectal cancer involves intestinal resection and combined chemotherapies. The current 5-year survival rate for local colorectal cancer is around 64%, and 12% for colorectal cancer with distant metastases (2). Thus, a better understanding of the mechanisms underlying colorectal cancer initiation and progression is necessary to improve diagnostics and therapy of colorectal cancer.

We identified 14-3-3σ as a p53-induced gene, which mediates a cell-cycle arrest in the G2 phase in colorectal cancer cells (3). 14-3-3σ, or stratifin (Sfn), belongs to a gene family, which encodes 7 different 14-3-3 isoforms in mammalian cells (for review see ref. 4). Unlike the other 14-3-3 family members, the 14-3-3σ protein exclusively forms homodimers (5, 6). 14-3-3σ binds to protein ligands via a consensus-binding motif phosphorylated on serine or threonine residues. The binding to 14-3-3 proteins regulate the function of associated proteins through cytoplasmatic sequestration, masking of interaction domains and export/import sequences, prevention of degradation, modulation of enzymatic activity and transactivation, or facilitation of protein modifications (4). The exclusive homodimerization critically contributes to the ability of 14-3-3σ to inhibit cell proliferation in the presence of other 14-3-3 family members, which are often highly expressed (6). More recently, we demonstrated that 14-3-3σ-deficiency in mice leads to an increased frequency and size of DMBA/12-O-tetradecanoylphorbol-13-acetate (TPA)–induced papilloma (7).

14-3-3σ is the only 14-3-3 gene, which is induced after DNA damage (8). The p53-mediated induction of 14-3-3σ was shown to be necessary for the maintenance of a stable G2–M arrest after DNA damage in colorectal cancer cell lines (9). 14-3-3σ associates with the cyclin-dependent kinase (CDK) CDC2/cyclin B1 complex and prevents its nuclear localization, which is required for progression through mitosis (9). Furthermore, 14-3-3σ was shown to bind to G1-specific CDKs to suppress cell proliferation (10). We identified ∼100 additional proteins interacting with 14-3-3σ in colorectal cancer cells using a proteomic approach (11). It is therefore likely that 14-3-3σ serves as a nodal regulator of numerous cellular processes.

Here we performed a comprehensive analysis of human colorectal cancer cohorts and found that downregulation of 14-3-3σ is associated with colorectal cancer progression and poor patient survival. Then we used a genetic approach to determine whether these associations also have a causal relationship. The results show that deletion of 14-3-3σ in a mouse model of intestinal cancer enhances tumor initiation and growth. In addition, the expression profiles of 14-3-3σ–negative adenomas revealed a switch towards a mesenchymal state, which is in line with the proposed function of 14-3-3σ as a suppressor of epithelial–mesenchymal transition (EMT) and metastasis.

Generation and handling of mice

The generation of 14-3-3σ mice with a C57BL/6J background has been described previously (7). ApcMin/+ mice (12) were obtained from Marlon Schneider, Ludwig-Maximilians-Universität, Munich, Germany. All animals were housed in the animal facility at the Pathology Institute of the Ludwig-Maximilians-University (Munich, Germany) in individually ventilated cages (IVC). Mice identifications, health controls (scoring) and cage changings were performed under a laminar flow hood. This animal study was approved by the Government of Upper Bavaria, Germany (AZ: 55.2–1-54–2532–4-2014).

Genotyping

Genomic DNA was obtained by overnight digest of ear punches in lysis buffer (50 mmol/L KCl, 1.5 mmol/L MgCl2, 10 mmol/L Tris pH8.5, 0.01% gelatin, 0.45% NP40, 0.45% Tween 20, 100 μg/mL proteinase K). PCR was performed for 25 cycles with an annealing temperature of 65°C. Primers for detection of 14-3-3σ wild-type and 14-3-3σ knockout (KO) alleles were added in the same ratio to the PCR reaction mix. PCR products (wild-type = 106 and KO = 183 bp) were resolved on a 2% agarose gel. Genotyping of ApcMin/+ mice was performed as previously described (13), using primers ApcMin/+ wild-type, ApcMin/+-common, and ApcMin/+-mutant (Supplementary Table S1).

Breeding mice for survival analysis

For the survival analysis, littermates with the genotypes ApcMin+/− (control group) and ApcMin/+/14-3-3σ−/− (experimental group) were compared. The littermates with the respective genotypes were obtained by crossing ApcMin/+/14-3-3σ+/− with 14-3-3σ+/− mice, both in pure C57BL/6J background. Control group and experimental group mice were housed in separated cages with up to 5 female or up to 4 male mice per cage.

Average survival probability

Thirty-six ApcMin/+ (C57BL/6J; 15 female, 21 male) and 39 ApcMin/+/14-3-3σ−/− (C57BL/6J; 19 female, 20 male) mice were maintained and monitored over a period of 282 days. Moribund mice were euthanized, reason of and age at death (days) was documented and adenomas from small intestine and colon were counted, measured (mm2) and analyzed by IHC, in situ hybridization, qPCR, and next generation sequencing (NGS). Average survival probabilities were calculated according to Kaplan–Meier using GraphPad Prism 8.3.0.

Tissue preparation and tumor count

Moribund or 120 days-old mice were euthanized, intestinal tracts were isolated, flushed with ice-cold PBS, and opened longitudinally. Small intestines were separated into four equal parts (duodenum, 2x jejunum, ileum) and colon was left as a whole. Single parts were photographed, fixed as Swiss rolls in 4% buffered, formaldehyde and embedded in paraffin. Tumor numbers and sizes were evaluated after photo documentation using ImageJ 1.52a.

Histology and immunohistology

Paraffin-embedded tissues were sectioned at 5 μm and used for hematoxylin and eosin (H&E) staining and periodic acid–Schiff (PAS) staining according to standard protocols. Tumors from ApcMin/+ and ApcMin/+/14-3-3σ−/− mice were classified as adenomas with either low- or high-grade dysplasia, based on nuclear-cytoplasmic ratio, nucleus location, prominence of nucleoli, gland architecture, amount of interglandular stroma, and the presence of mucus secretion. IHC was performed using antibodies and reagents listed in Supplementary Table S2. For each IHC detection ≥ 20 adenomas per mouse were examined. Positive staining was evaluated using ImageJ 1.52a.

ISH and FISH

For detection of intestinal stem cells an Olfm4-specific, DIG-labeled RNA probe was generated using a murine Olfm4 vector (kindly provided by Dr. Hans Clevers, Hubrecht Institute, Utrecht, the Netherlands) in combination with a DIG Northern Starter Kit (Roche Diagnostics). ISH was performed as described (14). For 14-3-3σ–ISH, we developed a novel 14-3-3σ–specific in situ RNA probe. In brief, we amplified murine 14-3-3σ cDNA (NCBI Ref. Seq.: NM_018754.2) from base pairs 14–1605 and reverse inserted the amplified sequence into pBluescriptII-KS(+) (Stratagene) between the restriction sites NotI and XhoI (NotI-1605–14-XhoI). We named the resulting vector pBSII-KS(+)Sfn-UTR. To generate a DIG labeled 14-3-3σ RNA probe, we linearized pBSII-KS(+)Sfn-UTR with ClaI and transcribed the 14-3-3σ RNA probe by using T7 primers and the negative control probe by T3 primers. Transcription using T7 primers results in a 775 bp RNA probe, which includes 459 bp 3′-UTR and 316 bp 3′-CDS of 14-3-3σ.

To visualize intestinal bacteria by fluorescence ISH, the universal eubacteria probe (EUB338) and negative control probe (NON338) were employed. The 5′-FITC–labeled EUB338 DNA (5′-GCTGCCTCCCGTAGGAGT-3′) and 5′-Cy3–labeled NON338 DNA (5′-CGACGGAGGGCATCCTCA-3′) probes were synthesized by Metabion (Planegg) and hybridized to 5-μm paraffin sections as described previously (15).

qPCR

Total RNA was isolated from tumor samples using the RNeasy Plus Mini Kit (Qiagen). cDNA was generated by Verso cDNA kit (Thermo Scientific) and qPCR was performed by using Fast SYBR Green Master Mix (Applied Biosystems) and a LightCycler 480 II (Roche Diagnostics). Relative gene expression was determined using the 2–ΔΔCt method (16). The individual mRNA levels were normalized to cyclophilin. Primers used for qPCR are listed in Supplementary Table S3.

Analysis of expression and clinical data from public databases

Expression and clinical data of The Cancer Genome Atlas (TCGA) colon adenocarcinoma (COAD) and rectal adenocarcinoma (READ) cohorts was obtained from the MD Anderson standardized data browser (http://bioinformatics.mdanderson.org/TCGA/databrowser/). The RNA sequencing (RNA-seq) by Expectation-Maximization (RSEM) normalized expression values from the Illumina RNASeqV2 (genes) datasets were used. Expression and clinical data of other colorectal cancer patient datasets was downloaded from NCBI Gene Expression Omnibus (GEO; www.ncbi.nlm.nih.gov/geo). Differential expression between tumors and adjacent normal colon tissue was calculated by paired t test. Differential expression between primary tumors from patients with colorectal cancer with and without metastasis was calculated using the limma R-package (17). Differential expression between tumors of different stages was calculated using one-way ANOVA with a posttest for linear trend from stage 1 to stage 4. The statistics for Forest plots and survival curves was calculated by log-rank test. For binary classification of cases (high/low expression), the Survminer R-package (https://CRAN.R-project.org/package=survminer) was used to determine optimal cut-off values. The correlation between differentially expressed genes and 14-3-3σ in public colorectal cancer patient datasets was analyzed using the Pearson correlation coefficient analysis. SNAIL-, c-JUN-, YAP1-, and FOXO1-regulated genes were identified on the basis of datasets from the NCBI GEO. The regulation of the expression of these genes was assessed using gene expression profiling datasets of cell lines/tissues with SNAIL, c-JUN, YAP1, or FOXO1 ectopic expression or knockdown (KD)/KO. The binding of SNAIL, c-JUN, YAP1, or FOXO1 to the regulatory regions of these genes was assessed using GEO chromatin immunoprecipitation–sequencing (ChIP-seq) datasets. Raw ChIP-seq data were downloaded from the NCBI Sequence Read Archive (SRA). Reads were mapped to the human or mouse reference genomes and processed using the ChIP-seq analysis module of the CLC Genomics Workbench software (Qiagen Bioinformatics) with default settings. The identifiers of all analyzed GEO public GEO microarray, RNA-seq, and ChIP-seq datasets are provided in Supplementary Tables S4–S7.

mRNA expression profiling by RNA-seq and bioinformatics analyses

Total RNA from murine adenomas was isolated using the RNeasy Plus Total RNA Isolation Kit (Qiagen) according to manufacturer's instructions. RNA profiling was done in quadruplicates. Random primed complementary DNA libraries were constructed and sequenced using the HiSeq2500 (Illumina) platform by Eurofins/GATC. Each sample was covered by at least 30 million single reads of 50-bp length. The raw reads (FASTQ files) were cleaned by trimming of adaptor sequences and low-quality sequences with average quality scores <20. Trimmed reads were mapped to the GRCm38/mm10 mouse reference genome and processed using the RNA-seq module of the CLC Genomics Workbench v8.0 software (Qiagen Bioinformatics) with default settings. RNA-seq data were filtered to exclude weakly expressed transcripts with less than ten mapped reads in all samples from the analysis and subjected to normalization using the R-package RUVSeq (remove unwanted variation from RNA-seq data) package (18). Differential gene expression analysis was performed using limma (17), edgeR (19), and DESeq2 (20) methods. Gene-set enrichment analyses (GSEA) were performed using GSEA software (Broad Institute, Boston, MA) and data from the molecular signature database (MSigDB). The significance of enrichments is presented by normalized enrichment scores with false discovery rate–adjusted q values. Heatmaps were generated with Morpheus (Broad Institute, Boston, MA).

Statistical analysis

A two-tailed Student t test was used to compare continuous variables. Categorical variables were compared using the χ2 method. Kaplan–Meier calculations were used to display the overall survival time and the results were compared with a log-rank test. The Sidak method was used to adjust P values when multiple comparisons were performed. P values less than 0.05 were considered significant and indicated by asterisks (*, P < 0.05; **, P < 0.01; or ***, P < 0.0001). GraphPad 8.3.0 was used for calculations.

Data and material availability

The RNA-seq data generated in this study have been deposited at the NCBI GEO with the accession number GSE163257.

14-3-3σmRNA expression and clinical associations in human colorectal cancer

14-3-3σ expression is downregulated in several types of human epithelial cancers, among them prostate, lung, breast, skin, and colorectal cancer (4, 21–23). However, 14-3-3σ expression in colorectal cancer has been so far only described in a small number of human patients (n = 116; ref. 23). In this study, downregulation of 14-3-3σ in colorectal cancers was associated with tumor progression and poor prognosis of patients. Thus, we analyzed 14-3-3σ expression and clinical associations in colorectal cancers from multiple patient cohorts deposited in TCGA and GEO databases (24). The expression of 14-3-3σ mRNA was significantly lower in tumors compared with adjacent normal colonic mucosa in 8 from 15 analyzed cohorts of matched pairs of tumor and normal colon tissue (Fig. 1A; n = 644). The expression of 14-3-3σ was also significantly lower in primary colorectal cancers from patients with metastasis compared with patients without metastasis in a pooled cohort of 3,103 patients (Fig. 1B). In colorectal cancers, 14-3-3σ displayed a gradually decreasing expression with increasing tumor stage in 4 of 5 analyzed patient cohorts (Fig. 1C). In colorectal cancers with mutant p53 the expression of 14-3-3σ was significantly lower than in p53 wild-type colorectal cancers (Fig. 1D). Decreased 14-3-3σ expression showed a significant correlation with poor survival in the combined analysis of nine colorectal cancer patient cohorts (Fig. 1E and F). Further analysis of the colorectal cancers with a known p53 status revealed, that this association was more pronounced in p53 wild-type than in p53-mutant colorectal cancers (Fig. 1G and H). Therefore, p53 mutations are presumably not a hidden cause for the association of decreased 14-3-3σ expression with poor survival. Taken together, the clinical associations linked to decreased 14-3-3σ expression identified in the cohorts of patients with colorectal cancer indicate that 14-3-3σ may function as a tumor suppressor in colorectal cancer.

Figure 1.

Association of 14-3-3σ mRNA expression in primary colorectal cancers with clinical and pathological characteristics in patient cohorts. A, Forest plot showing fold changes in 14-3-3σ expression between colorectal cancers and matched adjacent normal colonic mucosa for indicated cohorts. Dots represent fold change and horizontal lines show 95% confidence intervals (CI). P values were calculated using paired t test. B, Forest plot showing fold change in 14-3-3σ expression between primary colorectal cancers from patients with (pM1) and without metastasis (pM0) for indicated cohorts. Dots represent fold change and horizontal lines show 95% CI. P values were calculated using the limma method. C, Associations of 14-3-3σ expression with tumor stage. Significance was determined using one-way ANOVA with a posttest for linear trend from stage 1 to stage 4. D,14-3-3σ expression in p53 wt and mutant primary colorectal cancers (data from TCGA COAD+READ; Student t test). E, Forest plot showing HRs for relapse-free survival by comparing patients with high versus low expression of 14-3-3σ in indicated cohorts. Dots represent HRs and horizontal lines show 95% CI. P values were calculated using the log-rank method. F, Kaplan–Meier analysis of relapse-free survival using the pooled dataset of TCGA and GEO patient cohorts (n = 2,229 patients). G and H, Kaplan-Meier analysis of relapse-free survival using the TCGA COAD+READ (G) and the GSE39582 (H) patient cohorts stratified by the p53 mutation status. The significance was calculated with the log-rank test and the x-axis represents relapse-free survival in months. CRC, colorectal cancer.

Figure 1.

Association of 14-3-3σ mRNA expression in primary colorectal cancers with clinical and pathological characteristics in patient cohorts. A, Forest plot showing fold changes in 14-3-3σ expression between colorectal cancers and matched adjacent normal colonic mucosa for indicated cohorts. Dots represent fold change and horizontal lines show 95% confidence intervals (CI). P values were calculated using paired t test. B, Forest plot showing fold change in 14-3-3σ expression between primary colorectal cancers from patients with (pM1) and without metastasis (pM0) for indicated cohorts. Dots represent fold change and horizontal lines show 95% CI. P values were calculated using the limma method. C, Associations of 14-3-3σ expression with tumor stage. Significance was determined using one-way ANOVA with a posttest for linear trend from stage 1 to stage 4. D,14-3-3σ expression in p53 wt and mutant primary colorectal cancers (data from TCGA COAD+READ; Student t test). E, Forest plot showing HRs for relapse-free survival by comparing patients with high versus low expression of 14-3-3σ in indicated cohorts. Dots represent HRs and horizontal lines show 95% CI. P values were calculated using the log-rank method. F, Kaplan–Meier analysis of relapse-free survival using the pooled dataset of TCGA and GEO patient cohorts (n = 2,229 patients). G and H, Kaplan-Meier analysis of relapse-free survival using the TCGA COAD+READ (G) and the GSE39582 (H) patient cohorts stratified by the p53 mutation status. The significance was calculated with the log-rank test and the x-axis represents relapse-free survival in months. CRC, colorectal cancer.

Close modal

14-3-3σ expression and deletion in intestinal epithelium of mice

To obtain genetic evidence for a tumor suppressor function of 14-3-3σ in the intestine we decided to analyze the effect of 14-3-3σ deletion in the ApcMin/+ mouse model of inherited intestinal cancer/familial adenomatous polyposis (FAP). In ApcMin/+ mice, which carry a mutant Apc allele, the additional spontaneous loss of the remaining wild-type Apc allele leads to the development of multiple intestinal adenomas (12, 25). To determine whether 14-3-3σ suppresses intestinal adenoma initiation and/or promotion, we generated 14-3-3σ–deficient ApcMin/+ mice. First, we analyzed the 14-3-3σ mRNA expression pattern in ApcMin/+ mice by ISH. In normal appearing epithelium of the small intestine, 14-3-3σ was moderately expressed above the crypts in progenitor cells of the transient amplifying cell compartment and strongly in differentiated cells towards the apical villus epithelium (Fig. 2A). In the colorectal epithelium, 14-3-3σ was also expressed above the crypts in progenitor cells of the transient amplifying cell compartment and in the differentiated cells towards the colon lumen (Fig. 2B). As expected, 14-3-3σ expression was not detectable in the intestinal epithelium of 14-3-3σ–deficient ApcMin/+ mice (Fig. 2A and B, right). 14-3-3σ–deficient ApcMin/+ mice were born at a normal Mendelian ratio and were phenotypically indistinguishable from ApcMin/+ mice with wild-type 14-3-3σ. Deletion of 14-3-3σ did not affect the lengths of the small intestines (Supplementary Fig. S1A) and the width of the crypts in ApcMin/+ mice (Supplementary Fig. S1B). However, the depth of the crypts was slightly increased. In addition, the number of intestinal stem cells was not influenced by 14-3-3σ deletion, as determined by ISH with a stem cell specific Olfm4 in situ probe (Supplementary Fig. S1C). In addition, the numbers of Paneth (Supplementary Fig. S1D) and Goblet cells (Supplementary Fig. S1E) were unchanged.

Figure 2.

14-3-3σ expression in the small intestine and colon of ApcMin/+ mice. A and B,14-3-3σ mRNA expression pattern in the small intestinal (A) and colon epithelium (B) of ApcMin/+ and 14-3-3σ-deficient ApcMin/+ mice. 14-3-3σ mRNA was detected by ISH (dark purple stain) and nuclei were counterstained with nuclear fast red (bright pink) to visualize tissue structure. Curved dashed lines indicate the position of the crypt base cells of the small intestine. Scale bars, 100 μm.

Figure 2.

14-3-3σ expression in the small intestine and colon of ApcMin/+ mice. A and B,14-3-3σ mRNA expression pattern in the small intestinal (A) and colon epithelium (B) of ApcMin/+ and 14-3-3σ-deficient ApcMin/+ mice. 14-3-3σ mRNA was detected by ISH (dark purple stain) and nuclei were counterstained with nuclear fast red (bright pink) to visualize tissue structure. Curved dashed lines indicate the position of the crypt base cells of the small intestine. Scale bars, 100 μm.

Close modal

14-3-3σ deficiency shortens survival and increases tumorigenesis in ApcMin mice

Next, we performed a Kaplan–Meier analysis to determine the average survival of ApcMin/+ and 14-3-3σ–deficient ApcMin/+ mice. For the survival analyses, littermates with the respective mutated alleles were compared. ApcMin/+ mice showed a median survival of 192 days (Fig. 3A), which is in accordance with previous publications (26, 27). Notably, the median survival of 14-3-3σ deficient ApcMin/+ mice was decreased by 64 days. This effect was independent of the gender of the mice (Fig. 3B and C). Therefore, loss of 14-3-3σ provided a selective disadvantage for the tumor prone ApcMin/+ mice, which is consistent with a tumor suppressive function of 14-3-3σ. Consistently, the comparison of adenomas from age-matched, 120 days old ApcMin mice showed, that 14-3-3σ-deficiency increased the number of adenomas by 2.3-fold, total tumor area by 3.6-fold, and the average tumor area by 1.4-fold (Fig. 3D and E). These effects of 14-3-3σ loss were independent of the gender (Supplementary Fig. S2A and B). When mice, that became moribund due to intestinal tumor burden were analyzed, 14-3-3σ–deficient ApcMin/+ mice displayed an increased number of adenomas and a larger total tumor area (Fig. 3F). However, the mean tumor area showed no difference. The lack of a difference in the tumor area was presumably due to the longer lifetime of 14-3-3σ–proficient ApcMin mice, which allowed more cell divisions to occur. The effects of 14-3-3σ–deficiency on tumor incidence and size observed in moribund ApcMin mice were independent of the gender (Supplementary Fig. S3A and S3B).

Figure 3.

14-3-3σ deficiency decreases survival and increases frequency and size of adenomas of of ApcMin/+ mice. A–C, Kaplan–Meier survival analysis of ApcMin/+ mice with the indicated number of cases, genders, and genotypes. ***, P < 0.001. B, Only female mice. C, Only male mice. D, Macroscopic analysis of representative polyps in jejunum and colon of moribund mice of indicated genotypes. Scale bar, 1 cm. E and F, Quantification of adenoma numbers per mouse (left), mean area of adenoma per mouse (middle), and sum of adenoma areas (right) in the whole intestinal tract of 120 days old (E) and moribund mice (F) with the indicated genotypes and number of cases. Scatter-dot-plots show each value of an individual mouse with the indicated genotypes. Top and bottom whiskers represent the SD separated by the mean. E and F, Results represent the mean ± SD. Results were subjected to an unpaired, two-tailed Student t test with P values. *, P < 0.05; **, P < 0.01; ***, P < 0.001; n.s., not significant.

Figure 3.

14-3-3σ deficiency decreases survival and increases frequency and size of adenomas of of ApcMin/+ mice. A–C, Kaplan–Meier survival analysis of ApcMin/+ mice with the indicated number of cases, genders, and genotypes. ***, P < 0.001. B, Only female mice. C, Only male mice. D, Macroscopic analysis of representative polyps in jejunum and colon of moribund mice of indicated genotypes. Scale bar, 1 cm. E and F, Quantification of adenoma numbers per mouse (left), mean area of adenoma per mouse (middle), and sum of adenoma areas (right) in the whole intestinal tract of 120 days old (E) and moribund mice (F) with the indicated genotypes and number of cases. Scatter-dot-plots show each value of an individual mouse with the indicated genotypes. Top and bottom whiskers represent the SD separated by the mean. E and F, Results represent the mean ± SD. Results were subjected to an unpaired, two-tailed Student t test with P values. *, P < 0.05; **, P < 0.01; ***, P < 0.001; n.s., not significant.

Close modal

The increased size of adenomas in 120 days old 14-3-3σ–deficient ApcMin/+ mice may be caused by an increase in tumor cell proliferation and/or decrease in apoptosis. Indeed, the number of Ki-67–positive cells in adenomas from 120 days–old 14-3-3σ-deficient ApcMin/+ mice was significantly larger than in ApcMin/+ mice (Fig. 4A). Consistent with an increased proliferation in 14-3-3σ–deficient intestinal tumors/adenomas we detected a decreased expression of the mRNAs encoding the cell-cycle inhibitors p16INK4a and p19ARF, as well as increased expression of the mitosis promoting cyclin B1 in 14-3-3σ–deficient ApcMin/+ adenomas (Fig. 4B). Furthermore, the number of cells positive for cleaved-caspase-3 was significantly decreased in 14-3-3σ–deficient ApcMin/+ mice (Fig. 4C). Therefore, a decrease in apoptosis presumably also contributes to the increase in tumor size observed after deletion of 14-3-3σ.

Figure 4.

Effects of 14-3-3σ-deficiency on tumor characteristics in ApcMin/+ mice. A, Adenomas from moribund ApcMin/+ mice with the indicated genotypes were subjected to IHC detection of Ki-67 (3 adenomas per mouse and n = 3 mice per genotype). B, mRNA expression of selected cell-cycle regulatory genes in intestinal adenomas of similar size from moribund mice with the indicated genotypes was determined by qPCR (three adenomas per mouse and n = 6 mice per genotype). Results represent the mean ± SD and were subjected to an unpaired, two-tailed Student t test with P values. *, P < 0.03; n.s., not significant. C, IHC of cleaved caspase-3 (≥20 adenomas per mouse and n = 3 mice per genotype). D, FISH of universal eubacteria probe EUB338 (≥20 adenomas per genotype and n = 3 mice per genotype). E, H&E staining for tumor staging (low grade adenoma versus high-grade adenoma) of ≥ 36 intestinal adenomas per genotype (n = 6 mice per genotype). Scale bars, 100 μm. A, C, and D, Scatter-dot-plots show each value of an individual mouse with the indicated genotypes. Top and bottom whiskers represent the SD separated by the mean. Results represent the mean ± SD. Results were subjected to an unpaired, two-tailed Student t test with P values. *, P < 0.05; **, P < 0.01; ***, P < 0.001. C, D, and E, genotypes as indicated in A.

Figure 4.

Effects of 14-3-3σ-deficiency on tumor characteristics in ApcMin/+ mice. A, Adenomas from moribund ApcMin/+ mice with the indicated genotypes were subjected to IHC detection of Ki-67 (3 adenomas per mouse and n = 3 mice per genotype). B, mRNA expression of selected cell-cycle regulatory genes in intestinal adenomas of similar size from moribund mice with the indicated genotypes was determined by qPCR (three adenomas per mouse and n = 6 mice per genotype). Results represent the mean ± SD and were subjected to an unpaired, two-tailed Student t test with P values. *, P < 0.03; n.s., not significant. C, IHC of cleaved caspase-3 (≥20 adenomas per mouse and n = 3 mice per genotype). D, FISH of universal eubacteria probe EUB338 (≥20 adenomas per genotype and n = 3 mice per genotype). E, H&E staining for tumor staging (low grade adenoma versus high-grade adenoma) of ≥ 36 intestinal adenomas per genotype (n = 6 mice per genotype). Scale bars, 100 μm. A, C, and D, Scatter-dot-plots show each value of an individual mouse with the indicated genotypes. Top and bottom whiskers represent the SD separated by the mean. Results represent the mean ± SD. Results were subjected to an unpaired, two-tailed Student t test with P values. *, P < 0.05; **, P < 0.01; ***, P < 0.001. C, D, and E, genotypes as indicated in A.

Close modal

Next, we analyzed the potential effect of 14-3-3σ deletion on the epithelial barrier function by evaluating bacterial infiltration. ISH with the pan-bacteria probe EUB338 revealed that bacterial infiltration in 14-3-3σ–deficient ApcMin/+ adenomas is lower than in ApcMin/+ adenomas (Fig. 4D). Bacterial infiltration was not detected in villi of the small intestinal epithelium of ApcMin/+ and 14-3-3σ-deficient ApcMin/+ (Supplementary Fig. S4A). Furthermore, the frequency of infiltrating immune cells, such as T cells, macrophages, B cells, and neutrophils/granulocytes, was not changed significantly in 14-3-3σ–deficient ApcMin/+ adenomas (Supplementary Fig. S4B). Subsequently, we evaluated whether deletion of 14-3-3σ influences the degree of dysplasia present in adenomas obtained from moribund ApcMin/+ mice. As described before (28), low-grade dysplasia in adenomas was associated with enlarged, pencil-like, dark nuclei and partial loss of cell polarity, whereas high-grade dysplasia was characterized by enlarged, round, transparent nuclei with visible nucleoli and loss of cell polarity and dense growth. The evaluation of H&E-stained adenoma sections showed that moribund 14-3-3σ–deficient ApcMin/+ mice displayed significantly more adenomas with high-grade dysplasia (Fig. 4E). The high-grade dysplastic morphology of cells within 14-3-3σ–deficient adenomas implies that loss of 14-3-3σ promotes the dedifferentiation of tumor cells.

mRNA expression profiling of 14-3-3σ–deficient adenomas

To further illuminate the mechanism by which the loss of 14-3-3σ promotes intestinal tumorigenesis in ApcMin/+ mice, we compared the mRNA expression profiles of intestinal adenomas from ApcMin/+ and ApcMin/+/14-3-3σ−/− mice. For each genotype, 12 adenomas derived from four mice were analyzed (3 per mouse). Principal component analysis revealed that the expression profiles of adenomas from ApcMin/+/14-3-3σ−/− mice showed high similarities and were substantially different from profiles of adenomas from ApcMin/+ mice (Fig. 5A). Differential gene expression analyses using the Limma, edgeR, and Deseq2 algorithms revealed that 2,953 mRNAs were commonly, differentially expressed (P < 0.05) between adenomas from ApcMin/+ and ApcMin/+/14-3-3σ−/− mice (Fig. 5B). Out of these, 618 mRNAs were downregulated and 816 mRNAs were upregulated in 14-3-3σ-deficient adenomas with a fold change in expression > 1.5 (Fig. 5C and D). The mRNAs upregulated in 14-3-3σ-deficient adenomas generally showed a negative correlation with the expression of 14-3-3σ in colorectal cancer samples from 13 patient cohorts, whereas the downregulated mRNAs showed a positive correlation with the expression of 14-3-3σ in colorectal cancers (Supplementary Fig. S5).

Figure 5.

RNA-seq analysis of intestinal adenomas from ApcMin/+ and 14-3-3σ-deficient ApcMin/+ mice. A, Principal component analysis of mRNA expression in ApcMin/+ and ApcMin/+/14-3-3σ−/− mice-derived intestinal adenomas. B, Venn diagram displaying differentially expressed mRNAs (P < 0.05) between ApcMin/+ and ApcMin/+/14-3-3σ−/− mice-derived intestinal adenomas as determined by edgeR, limma, and DESeq2. C, Volcano plot showing expression fold change (FC) and significance of differentially expressed mRNAs between ApcMin/+ and ApcMin/+/14-3-3σ−/− mice-derived intestinal adenomas. Downregulated (fold change ≤ 0.667) and upregulated (fold change ≥ 1.5) mRNAs are indicated in blue and red, respectively. mRNAs with fold changes between 0.667 and 1.5 and/or statistically nonsignificant (P < 0.05) changes in expression are indicated in black. Dashed blue and red vertical lines indicate 1.5-fold change cutoff. Dashed horizontal line indicates the cutoff for P values < 0.05. D, Heatmap showing the expression of differentially expressed mRNAs (fold change ≥ 1.5, P < 0.05) between ApcMin/+ and ApcMin/+/14-3-3σ−/− mice-derived intestinal adenomas. Colors indicate relative expression values from minimum (blue) to maximum (red). E, GSEA analyses of mRNAs upregulated in 14-3-3σ-deficient intestinal adenomas. The most significant gene sets are shown for the indicated categories. F, Forest plot showing HRs for relapse-free survival by comparing patients with colorectal cancer with high versus low expression of the 14-3-3σ KO signature in indicated cohorts (details about the generation of the 14-3-3σ KO signature are in Materials and Methods). Dots represent HRs and horizontal lines show 95% CI. P values were calculated using the log-rank method. G, Kaplan–Meier curve showing association of the 14-3-3σ knockout signature with relapse-free survival in the pooled cohort. The statistics were calculated using the log-rank test. H, Expression of the 14-3-3σ KO signature in human colorectal tumors belonging to indicated CMS subtypes. CRC, colorectal cancer.

Figure 5.

RNA-seq analysis of intestinal adenomas from ApcMin/+ and 14-3-3σ-deficient ApcMin/+ mice. A, Principal component analysis of mRNA expression in ApcMin/+ and ApcMin/+/14-3-3σ−/− mice-derived intestinal adenomas. B, Venn diagram displaying differentially expressed mRNAs (P < 0.05) between ApcMin/+ and ApcMin/+/14-3-3σ−/− mice-derived intestinal adenomas as determined by edgeR, limma, and DESeq2. C, Volcano plot showing expression fold change (FC) and significance of differentially expressed mRNAs between ApcMin/+ and ApcMin/+/14-3-3σ−/− mice-derived intestinal adenomas. Downregulated (fold change ≤ 0.667) and upregulated (fold change ≥ 1.5) mRNAs are indicated in blue and red, respectively. mRNAs with fold changes between 0.667 and 1.5 and/or statistically nonsignificant (P < 0.05) changes in expression are indicated in black. Dashed blue and red vertical lines indicate 1.5-fold change cutoff. Dashed horizontal line indicates the cutoff for P values < 0.05. D, Heatmap showing the expression of differentially expressed mRNAs (fold change ≥ 1.5, P < 0.05) between ApcMin/+ and ApcMin/+/14-3-3σ−/− mice-derived intestinal adenomas. Colors indicate relative expression values from minimum (blue) to maximum (red). E, GSEA analyses of mRNAs upregulated in 14-3-3σ-deficient intestinal adenomas. The most significant gene sets are shown for the indicated categories. F, Forest plot showing HRs for relapse-free survival by comparing patients with colorectal cancer with high versus low expression of the 14-3-3σ KO signature in indicated cohorts (details about the generation of the 14-3-3σ KO signature are in Materials and Methods). Dots represent HRs and horizontal lines show 95% CI. P values were calculated using the log-rank method. G, Kaplan–Meier curve showing association of the 14-3-3σ knockout signature with relapse-free survival in the pooled cohort. The statistics were calculated using the log-rank test. H, Expression of the 14-3-3σ KO signature in human colorectal tumors belonging to indicated CMS subtypes. CRC, colorectal cancer.

Close modal

GSEA of upregulated mRNAs showed the most pronounced enrichment within EMT and inflammatory cytokine signaling, as well as cell motility and extracellular matrix MSigDB gene sets (Fig. 5E). Because it has been suggested that 14-3-3σ is involved in the regulation of p53 activity (29), we evaluated the expression of p53 target genes. Although, p53 target gene signatures showed a tendency for enrichment in wild-type compared with 14-3-3σ–deficient tumors, the effects were not statistically significant (Supplementary Fig. S6). Therefore, loss of 14-3-3σ did not affect p53-driven gene expression significantly, which is in line with the preferential binding of other 14-3-3 isoforms to p53 (30).

Next, we defined a 14-3-3σ KO associated signature by subtracting the sum of normalized expressions of 618 downregulated mRNAs from the sum of normalized expressions of 816 upregulated mRNAs. High prevalence of the 14-3-3σ KO signature was significantly associated with poor relapse-free survival in six of nine colorectal cancer patient cohorts (Fig. 5F). Moreover, combined analysis of the pooled data of all nine patient cohorts showed that high expression of the 14-3-3σ KO signature is strongly associated with poor relapse-free survival (Fig. 5G). Finally, the expression of the 14-3-3σ KO signature was highest in the CMS4 (mesenchymal) molecular subtype of colorectal cancer (Fig. 5H), which is associated with metastasis and poor prognosis (31).

As the most pronounced changes in expression associated with the loss of 14-3-3σ were within the EMT and inflammatory cytokine signaling signatures, we inspected these signatures in more detail. The MSigDB Hallmark EMT signature contains 200 mesenchymal state–associated genes, of which, 62 were significantly elevated in 14-3-3σ-deficient adenomas (Fig. 6A). Moreover, the expression of core EMT-TFs SNAI1, TWIST1, VIM, ZEB1, and ZEB2, was also elevated in adenomas derived from ApcMin/+/14-3-3σ−/− mice (Fig. 6A). Interestingly, the expression of epithelial state associated mRNAs, such as CDH1, EPCAM, and various keratins was downregulated in 14-3-3σ-deficient adenomas (Fig. 6A), indicating that the loss of 14-3-3σ leads to an EMT. Interestingly, the keratins Krt14 and Krt17 are known to directly interact with 14-3-3σ protein to regulate epithelial cell functions (32, 33). The expression of mesenchymal state associated genes generally showed a negative correlation with the expression of 14-3-3σ in tumor samples from 13 public colorectal cancer patient cohorts, whereas the expression of epithelial state associated genes showed a positive correlation with the expression of 14-3-3σ in tumors from a patient with colorectal cancer (Fig. 6A). qPCR analysis of differentially expressed EMT-TFs Twist1 and Snai1, as well epithelial state–associated mRNAs Krt7, Krt18, and Krt19 validated the differential expressions detected by RNA-seq analysis (Fig. 6B). Finally, 14-3-3σ-deficient adenomas showed elevated expression of several inflammatory cytokines and their receptors, such as the protumorigenic IL6 (34) and hepatocyte growth factor (HGF), which is the ligand of the c-MET proto-oncogene that also stimulates the expression of EMT-transcription factors (TF; Fig. 6C; refs. 35, 36). The expression of inflammatory cytokines and their receptors generally showed a negative correlation with the expression of 14-3-3σ in tumors from patients with colorectal cancer (Fig. 6C). Altogether, our expression profiling results suggest that EMT may be involved in the increased dedifferentiation and dysplasia of 14-3-3σ-deficient adenomas. Moreover, the altered cytokine–receptor signaling suggests a role of 14-3-3σ downregulation in enhancing tumor/stroma interactions.

Figure 6.

14-3-3σ loss induces EMT and inflammatory cytokine–related expression in adenomas. A, Left, heatmap showing the expression of EMT-related genes in ApcMin/+ and ApcMin/+/14-3-3σ−/− adenomas. Right, heatmap showing correlations between the expression of each mRNA and the expression of 14-3-3σ in tumors from the indicated colorectal cancer patient cohorts. Bold indicates EMT-associated mRNAs added to the Hallmark EMT gene signature. B, qPCR validation of differential regulation of EMT-associated mRNAs in ApcMin/+ adenomas. RNA was isolated from intestinal adenomas of similar size from moribund mice with the indicated genotypes (three adenomas per mouse and n = 4 mice per genotype). Results represent the mean ± SD and were subjected to an unpaired, two-tailed Student t test with P values. *, P < 0.05; **, P < 0.01; ***, P < 0.001. C, Left, heatmap showing the expression of cytokine signaling-related genes in ApcMin/+ and ApcMin/+/14-3-3σ−/− adenomas. Right, heatmap showing correlations between differentially expressed mRNAs and 14-3-3σ in colorectal cancers from the indicated patient cohorts. CRC, colorectal cancer.

Figure 6.

14-3-3σ loss induces EMT and inflammatory cytokine–related expression in adenomas. A, Left, heatmap showing the expression of EMT-related genes in ApcMin/+ and ApcMin/+/14-3-3σ−/− adenomas. Right, heatmap showing correlations between the expression of each mRNA and the expression of 14-3-3σ in tumors from the indicated colorectal cancer patient cohorts. Bold indicates EMT-associated mRNAs added to the Hallmark EMT gene signature. B, qPCR validation of differential regulation of EMT-associated mRNAs in ApcMin/+ adenomas. RNA was isolated from intestinal adenomas of similar size from moribund mice with the indicated genotypes (three adenomas per mouse and n = 4 mice per genotype). Results represent the mean ± SD and were subjected to an unpaired, two-tailed Student t test with P values. *, P < 0.05; **, P < 0.01; ***, P < 0.001. C, Left, heatmap showing the expression of cytokine signaling-related genes in ApcMin/+ and ApcMin/+/14-3-3σ−/− adenomas. Right, heatmap showing correlations between differentially expressed mRNAs and 14-3-3σ in colorectal cancers from the indicated patient cohorts. CRC, colorectal cancer.

Close modal

14-3-3σ is known to associate with the TFs SNAIL, c-JUN, YAP1, and FOXO1, thereby promoting nuclear export and/or degradation, and ultimately suppressing activity of these TFs (37–40). Here we analyzed whether gene regulations mediated by these TFs are involved in the tumor-suppressing functions of 14-3-3σ in the ApcMin/+ mouse model. First, we established signatures of SNAIL, c-JUN, YAP1, and FOXO1 target genes by retrieving expression data from multiple public GEO datasets representing ectopic expression/KD/KO studies to identify genes that are consistently regulated by these TFs. Moreover, we analyzed multiple GEO ChIP-seq datasets to identify genes directly bound by these TFs. Notably, SNAIL, c-JUN, YAP1, and FOXO1 target gene signatures were significantly enriched in 14-3-3σ-deficient adenomas, implying that these TFs are activated by the deletion of 14-3-3σ in adenomas of ApcMin/+ mice (Fig. 7A, C, E, and G). Figure 7B, D, F, and H show examples of SNAIL, c-JUN, YAP1, and FOXO1 target genes that are differentially expressed between 14-3-3σ-proficient and -deficient ApcMin/+ adenomas and thus might be involved in the tumor-suppressing functions of 14-3-3σ (see also Supplementary Table S4–S7 for details of the analyzed public GEO microarray, RNA-seq, and ChIP-seq datasets). For example, connective tissue growth factor (CTGF) was upregulated in 14-3-3σ-deficient adenomas and represents a well-established YAP1 target gene (41), that has been associated with the induction of EMT and tumor progression (42, 43). Analysis of colorectal cancer cohorts revealed that elevated expression of CTGF in colorectal cancers is consistently associated with poor survival of patients, CMS4 subtype, and showed a gradually increased expression with tumor progression (Fig. 7I–K). Furthermore, fibroblast growth factor receptor 1 (FGFR1), which is known to be directly induced by c-JUN (44), was upregulated in 14-3-3σ-deficient adenomas and was also found to be associated with poor survival, CMS4 subtype, and tumor stage (Fig. 7L–N). Taken together, the effects of 14-3-3σ loss on adenoma-specific gene expression detected here indicate that the inactivation of 14-3-3σ during tumor progression may have profound effects on multiple cancer relevant processes, such as EMT and tumor–stroma signaling. Furthermore, our results indicate that tumor suppression by 14-3-3σ is, at least in part, mediated by the inhibition of 14-3-3σ–associated TFs which is lost upon 14-3-3σ inactivation.

Figure 7.

Activation of SNAIL-, c-JUN-, YAP1-, or FOXO1-mediated transcriptional programs in 14-3-3σ–deficient adenomas. A, C, E, and G, GSEA analyses of SNAIL (A), c-JUN (C), YAP1 (E), and FOXO1 (G) target gene signatures in ApcMin/+ and ApcMin/+/14-3-3σ−/− intestinal adenomas. B, D, F, and H, Left, heatmaps showing the expression of indicated genes in 8 adenomas from ApcMin/+ and ApcMin/+/14-3-3σ−/− mice. Middle, heatmaps showing the fold change in expression of indicated mRNAs in GEO datasets after SNAIL, c-JUN, YAP1, or FOXO1 ectopic expression or KD/KO. Right, black boxes indicate promoter occupancy by SNAIL, c-JUN, YAP1, or FOXO1 according to GEO ChIP-seq datasets. Results are shown from left to right in the order of the underlying datasets listed in Supplementary Tables 4–7. EMT-associated genes are indicated in bold. I and L, Forest plots showing HRs for relapse-free survival by comparing patients with high versus low expression of CTGF (I) and FGFR1 (L) in colorectal cancers. J and M, Expression of CTGF (J) and FGFR1 (M) in human colorectal cancers belonging to the indicated CMS subtypes. K and N, Association of CTGF (K) and FGFR1 (N) expression with tumor stage.

Figure 7.

Activation of SNAIL-, c-JUN-, YAP1-, or FOXO1-mediated transcriptional programs in 14-3-3σ–deficient adenomas. A, C, E, and G, GSEA analyses of SNAIL (A), c-JUN (C), YAP1 (E), and FOXO1 (G) target gene signatures in ApcMin/+ and ApcMin/+/14-3-3σ−/− intestinal adenomas. B, D, F, and H, Left, heatmaps showing the expression of indicated genes in 8 adenomas from ApcMin/+ and ApcMin/+/14-3-3σ−/− mice. Middle, heatmaps showing the fold change in expression of indicated mRNAs in GEO datasets after SNAIL, c-JUN, YAP1, or FOXO1 ectopic expression or KD/KO. Right, black boxes indicate promoter occupancy by SNAIL, c-JUN, YAP1, or FOXO1 according to GEO ChIP-seq datasets. Results are shown from left to right in the order of the underlying datasets listed in Supplementary Tables 4–7. EMT-associated genes are indicated in bold. I and L, Forest plots showing HRs for relapse-free survival by comparing patients with high versus low expression of CTGF (I) and FGFR1 (L) in colorectal cancers. J and M, Expression of CTGF (J) and FGFR1 (M) in human colorectal cancers belonging to the indicated CMS subtypes. K and N, Association of CTGF (K) and FGFR1 (N) expression with tumor stage.

Close modal

14-3-3σ expression is epigenetically silenced in several types of human cancers (21), suggesting that 14-3-3σ may also represent a tumor-suppressor in colorectal cancer. However, the in vivo role of 14-3-3σ in tumor suppression has so far only been analyzed genetically in breast and skin cancers (7, 45, 46) and not in colorectal cancer, where its tumor-suppressive function has initially been suggested (3). Here, we describe a protumorigenic effect of 14-3-3σ deletion in the ApcMin/+ mouse model of colorectal cancer and thereby demonstrate that 14-3-3σ represents a suppressor of intestinal tumorigenesis.

In the normal epithelium of the small intestine of ApcMin mice we detected 14-3-3σ expression in transient amplifying (TA) crypt cells and differentiated enterocytes of the villi. 14-3-3σ deficiency caused an increase in the frequency and size of small intestinal and colonic tumors in ApcMin/+ mice. We suggest that the function of 14-3-3σ in the TA cell compartment is different from its function in the differentiated, apical villi cells. In the highly proliferative TA cell compartment 14-3-3σ might be involved in the negative regulation of TA cell proliferation as part of genome surveillance by p53. However, in differentiated, apical villi cells, 14-3-3σ is presumably involved in the regulation of terminal differentiation. This hypothesis is supported by our finding that 14-3-3σ–deficient adenomas are larger and show an increase in proliferating and a decrease in apoptotic cells. Interestingly, the expression of mRNAs encoding the CDK inhibitor (CDKI) p16INK4a, which inhibits G1–S-transition, and p19ARF was decreased in 14-3-3σ–deficient adenomas. Because p19ARF and p16INK4a harbor antiproliferative activity, their decreased expression may contribute to the increased proliferation and size of 14-3-3σ–deficient adenomas.

Tumor formation in ApcMin/+ mice is invariably associated with loss of the remaining wild-type allele (12), which occurs early in colorectal cancer tumorigenesis. The frequency is presumably influenced by the degree of chromosomal instability (CIN; ref. 47). Interestingly, 14-3-3σ–deficient colorectal cancer cells show an increased rate of CIN as evidenced by more frequent chromosomal aberrations, such as telomere-telomere fusions, unbalanced translocations, and chromosomal breaks, upon ionizing radiation (48). Because 14-3-3σ has been implicated as a mediator of a G2–M cell-cycle arrest after DNA damage (3, 9), loss of the second Apc allele might be more frequent in 14-3-3σ-deficient ApcMin/+ mice, due to their inability to maintain a stable G2 arrest, which would prevent the accumulation of unrepaired chromosomal aberrations. Interestingly, Apc loss of heterozygosity (LOH) leads to microadenomas, of which, not all develop into macroadenomas (49). Therefore, a tumor-suppressive arrest exists at the early stage of adenoma formation, which may involve the activation of 14-3-3σ expression. Inactivation/downregulation of 14-3-3σ by CpG methylation or other mechanisms may allow, or at least contribute to, the formation of macroadenomas.

Our NGS analysis of ApcMin/+ adenomas showed that 14-3-3σ-deficiency in adenomas leads to the upregulation of the EMT-TF Snail, which is known to promote the cellular transition from an epithelial to a mesenchymal state (50). It has been suggested, that 14-3-3σ inhibits the transcriptional activity of Snail by sequestering phosphorylated Snail in the cytoplasm and thereby keeps cells in an epithelial-like state (37, 51). Therefore, upregulation of 14-3-3σ may inhibit Snail-induced EMT, whereas downregulation/inactivation of 14-3-3σ might accelerate Snail-induced EMT. Thus, decreased levels of 14-3-3σ might be necessary to maintain a mesenchymal colorectal cancer phenotype, which is found in those colorectal cancers that belong to the CMS4 subtype. This hypothesis is supported by the findings of Raychaudhuri and colleagues (38): Human colorectal cancer cells (HCT116) deficient for 14-3-3σ display upregulation of EMT-markers, increased migration and invasion. Therefore, 14-3-3σ-deficient HCT116 cells presumably passed through an EMT. Thus, 14-3-3σ is presumably involved in the inhibition of EMT (i.e., MET), whereas downregulation of 14-3-3σ may promote EMT during colorectal cancer progression. In line with this scenario, the expression signature associated with deletion of 14-3-3σ characterized here was associated with poor relapse-free survival of patients with colorectal cancer, which is mainly determined by the formation of distant metastasis.

It has been shown that 14-3-3σ binds to the transcription factors SNAIL, c-JUN, YAP1, and FOXO1, and thereby suppresses their activity (37–40). The target gene signatures of these TFs were significantly enriched in 14-3-3σ-deficient adenomas, suggesting that the transcriptional programs mediated these TFs are activated after the loss of 14-3-3σ. For example, the known YAP1 target gene CTGF (41) and the c-JUN target gene FGFR1 (44) were upregulated in 14-3-3σ-deficient adenomas and presumably contribute to the enhanced tumor formation and growth in these mice. These genes have established oncogenic properties in colorectal cancer and other cancer types (38, 40, 51). Moreover, our analyses showed that elevated expression of CTGF and FGFR1 is consistently associated with poor survival in patients with colorectal cancer, CMS4 colorectal cancer subtype, and showed a gradual increased expression with increasing tumor stage in multiple colorectal cancer patient cohorts.

Furthermore, our results implicate a role of 14-3-3σ in the regulation of cytokine receptor signaling, cell motility, and extracellular matrix organization. Via influencing the tumor–stroma interactions, the deregulation of these processes caused by loss/downregulation of 14-3-3σ may contribute to colorectal cancer progression.

Tumor progression in the ApcMin mouse model is known to be limited to the development of noninvasive adenomas (52). Additional genetic lesions or treatments may result in further progression of these adenomas to invasive adenomas, although with a low frequency (e.g., loss of p53, ref. 53, or treatment with DSS, ref. 54). However, formation of local or distant metastasis has not been observed in ApcMin mice even after introduction of additional lesions (52). To determine the effect of 14-3-3σ loss on invasion and metastases formation in the context of additional lesions that occur during colorectal cancer progression it is therefore necessary to inactivate 14-3-3σ in mouse models of metastatic colorectal cancer. The results presented here warrant such efforts.

H. Hermeking reports grants from Deutsche Krebshilfe during the conduct of the study; in addition, H. Hermeking has a patent for “14-3-3sigma arrests the cell cycle” United States Patent 6.740.523 issued. No disclosures were reported by the other authors.

M. Winter: Investigation, writing–original draft. M. Rokavec: Investigation, bioinformatics analyses. H. Hermeking: Conceptualization, resources, supervision, funding acquisition, writing–original draft, writing–review and editing.

We thank Ursula Götz for technical assistance, Marlon Schneider for the ApcMin/+ mice, and Hans Clevers for the Olfm4 cDNA plasmid. The project was funded by grants of the Deutsche Krebshilfe (#70112441 and #70113975 to H. Hermeking).

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.

1.
Bray
F
,
Ferlay
J
,
Soerjomataram
I
,
Siegel
RL
,
Torre
LA
,
Jemal
A
. 
Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries
.
CA Cancer J Clin
2018
;
68
:
394
424
.
2.
Siegel
RL
,
Miller
KD
,
Jemal
A
. 
Cancer statistics, 2019
.
CA Cancer J Clin
2019
;
69
:
7
34
.
3.
Hermeking
H
,
Lengauer
C
,
Polyak
K
,
He
TC
,
Zhang
L
,
Thiagalingam
S
, et al
14–3-3 sigma is a p53-regulated inhibitor of G2/M progression
.
Mol Cell
1997
;
1
:
3
11
.
4.
Hermeking
H
. 
The 14–3-3 cancer connection
.
Nat Rev Cancer
2003
;
3
:
931
43
.
5.
Benzinger
A
,
Popowicz
GM
,
Joy
JK
,
Majumdar
S
,
Holak
TA
,
Hermeking
H
. 
The crystal structure of the non-liganded 14–3-3sigma protein: insights into determinants of isoform specific ligand binding and dimerization
.
Cell Res
2005
;
15
:
219
27
.
6.
Verdoodt
B
,
Benzinger
A
,
Popowicz
GM
,
Holak
TA
,
Hermeking
H
. 
Characterization of 14–3-3sigma dimerization determinants: requirement of homodimerization for inhibition of cell proliferation
.
Cell Cycle
2006
;
5
:
2920
6
.
7.
Winter
M
,
Lodygin
D
,
Verdoodt
B
,
Hermeking
H
. 
Deletion of 14–3-3sigma sensitizes mice to DMBA/TPA-induced papillomatosis
.
Oncotarget
2016
;
7
:
46862
70
.
8.
Samuel
T
,
Weber
HO
,
Rauch
P
,
Verdoodt
B
,
Eppel
JT
,
McShea
A
, et al
The G2–M regulator 14–3-3sigma prevents apoptosis through sequestration of Bax
.
J Biol Chem
2001
;
276
:
45201
6
.
9.
Chan
TA
,
Hermeking
H
,
Lengauer
C
,
Kinzler
KW
,
Vogelstein
B
. 
14–3-3Sigma is required to prevent mitotic catastrophe after DNA damage
.
Nature
1999
;
401
:
616
20
.
10.
Laronga
C
,
Yang
HY
,
Neal
C
,
Lee
MH
. 
Association of the cyclin-dependent kinases and 14–3-3 sigma negatively regulates cell cycle progression
.
J Biol Chem
2000
;
275
:
23106
12
.
11.
Benzinger
A
,
Muster
N
,
Koch
HB
,
Yates
JR
 3rd
,
Hermeking
H
. 
Targeted proteomic analysis of 14–3-3 sigma, a p53 effector commonly silenced in cancer
.
Mol Cell Proteomics
2005
;
4
:
785
95
.
12.
Su
LK
,
Kinzler
KW
,
Vogelstein
B
,
Preisinger
AC
,
Moser
AR
,
Luongo
C
, et al
Multiple intestinal neoplasia caused by a mutation in the murine homolog of the APC gene
.
Science
1992
;
256
:
668
70
.
13.
Jaeckel
S
,
Kaller
M
,
Jackstadt
R
,
Gotz
U
,
Muller
S
,
Boos
S
, et al
Ap4 is rate limiting for intestinal tumor formation by controlling the homeostasis of intestinal stem cells
.
Nat Commun
2018
;
9
:
3573
.
14.
Gregorieff
A
,
Clevers
H
. 
In situ hybridization to identify gut stem cells
.
Curr Protoc Stem Cell Biol
2010
;
Chapter 2:Unit 2F 1
.
15.
Oner
MG
,
Rokavec
M
,
Kaller
M
,
Bouznad
N
,
Horst
D
,
Kirchner
T
, et al
Combined inactivation of TP53 and MIR34A promotes colorectal cancer development and progression in mice via increasing levels of IL6R and PAI1
.
Gastroenterology
2018
;
155
:
1868
82
.
16.
Livak
KJ
,
Schmittgen
TD
. 
Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method
.
Methods
2001
;
25
:
402
8
.
17.
Ritchie
ME
,
Phipson
B
,
Wu
D
,
Hu
Y
,
Law
CW
,
Shi
W
, et al
limma powers differential expression analyses for RNA-sequencing and microarray studies
.
Nucleic Acids Res
2015
;
43
:
e47
.
18.
Risso
D
,
Ngai
J
,
Speed
TP
,
Dudoit
S
. 
Normalization of RNA-seq data using factor analysis of control genes or samples
.
Nat Biotechnol
2014
;
32
:
896
902
.
19.
Robinson
MD
,
McCarthy
DJ
,
Smyth
GK
. 
edgeR: a Bioconductor package for differential expression analysis of digital gene expression data
.
Bioinformatics
2010
;
26
:
139
40
.
20.
Love
MI
,
Huber
W
,
Anders
S
. 
Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2
.
Genome Biol
2014
;
15
:
550
.
21.
Lodygin
D
,
Hermeking
H
. 
Epigenetic silencing of 14–3-3sigma in cancer
.
Semin Cancer Biol
2006
;
16
:
214
24
.
22.
Ferguson
AT
,
Evron
E
,
Umbricht
CB
,
Pandita
TK
,
Chan
TA
,
Hermeking
H
, et al
High frequency of hypermethylation at the 14–3-3 sigma locus leads to gene silencing in breast cancer
.
Proc Natl Acad Sci U S A
2000
;
97
:
6049
54
.
23.
Shao
Z
,
Cai
Y
,
Xu
L
,
Yao
X
,
Shi
J
,
Zhang
F
, et al
Loss of the 14–3-3sigma is essential for LASP1-mediated colorectal cancer progression via activating PI3K/AKT signaling pathway
.
Sci Rep
2016
;
6
:
25631
.
24.
Clough
E
,
Barrett
T
. 
The gene expression omnibus database
.
Methods Mol Biol
2016
;
1418
:
93
110
.
25.
Moser
AR
,
Pitot
HC
,
Dove
WF
. 
A dominant mutation that predisposes to multiple intestinal neoplasia in the mouse
.
Science
1990
;
247
:
322
4
.
26.
Hoshiko
S
,
Kawaguchi
M
,
Fukushima
T
,
Haruyama
Y
,
Yorita
K
,
Tanaka
H
, et al
Hepatocyte growth factor activator inhibitor type 1 is a suppressor of intestinal tumorigenesis
.
Cancer Res
2013
;
73
:
2659
70
.
27.
Guillen-Ahlers
H
,
Suckow
MA
,
Castellino
FJ
,
Ploplis
VA
. 
Fas/CD95 deficiency in ApcMin/+ mice increases intestinal tumor burden
.
PLoS One
2010
;
5
:
e9070
.
28.
Jiang
L
,
Hermeking
H
. 
miR-34a and miR-34b/c suppress intestinal tumorigenesis
.
Cancer Res
2017
;
77
:
2746
58
.
29.
Waterman
MJ
,
Stavridi
ES
,
Waterman
JL
,
Halazonetis
TD
. 
ATM-dependent activation of p53 involves dephosphorylation and association with 14–3-3 proteins
.
Nat Genet
1998
;
19
:
175
8
.
30.
Stavridi
ES
,
Chehab
NH
,
Malikzay
A
,
Halazonetis
TD
. 
Substitutions that compromise the ionizing radiation-induced association of p53 with 14–3-3 proteins also compromise the ability of p53 to induce cell cycle arrest
.
Cancer Res
2001
;
61
:
7030
3
.
31.
Guinney
J
,
Dienstmann
R
,
Wang
X
,
de Reynies
A
,
Schlicker
A
,
Soneson
C
, et al
The consensus molecular subtypes of colorectal cancer
.
Nat Med
2015
;
21
:
1350
6
.
32.
Guo
Y
,
Redmond
CJ
,
Leacock
KA
,
Brovkina
MV
.,
Ji
S
,
Jaskula-Ranga
V
, et al
Keratin 14-dependent disulfides regulate epidermal homeostasis and barrier function via 14–3-3sigma and YAP1
.
Elife
2020
;
9
:
e53165
.
33.
Kim
S
,
Wong
P
,
Coulombe
PA
. 
A keratin cytoskeletal protein regulates protein synthesis and epithelial cell growth
.
Nature
2006
;
441
:
362
5
.
34.
Taniguchi
K
,
Karin
M
. 
IL-6 and related cytokines as the critical lynchpins between inflammation and cancer
.
Semin Immunol
2014
;
26
:
54
74
.
35.
Parizadeh
SM
,
Jafarzadeh-Esfehani
R
,
Fazilat-Panah
D
,
Hassanian
SM
,
Shahidsales
S
,
Khazaei
M
, et al
The potential therapeutic and prognostic impacts of the c-MET/HGF signaling pathway in colorectal cancer
.
IUBMB Life
2019
;
71
:
802
11
.
36.
Thiery
JP
,
Acloque
H
,
Huang
RY
,
Nieto
MA
. 
Epithelial-mesenchymal transitions in development and disease
.
Cell
2009
;
139
:
871
90
.
37.
Du
C
,
Zhang
C
,
Hassan
S
,
Biswas
MH
,
Balaji
KC
. 
Protein kinase D1 suppresses epithelial-to-mesenchymal transition through phosphorylation of snail
.
Cancer Res
2010
;
70
:
7810
9
.
38.
Raychaudhuri
K
,
Chaudhary
N
,
Gurjar
M
,
D'Souza
R
,
Limzerwala
J
,
Maddika
S
, et al
14–3-3sigma gene loss leads to activation of the epithelial to mesenchymal transition due to the stabilization of c-Jun protein
.
J Biol Chem
2016
;
291
:
16068
81
.
39.
Sambandam
SAT
,
Kasetti
RB
,
Xue
L
,
Dean
DC
,
Lu
Q
,
Li
Q
. 
14–3-3sigma regulates keratinocyte proliferation and differentiation by modulating Yap1 cellular localization
.
J Invest Dermatol
2015
;
135
:
1621
8
.
40.
Su
YW
,
Hao
Z
,
Hirao
A
,
Yamamoto
K
,
Lin
WJ
,
Young
A
, et al
14–3-3sigma regulates B-cell homeostasis through stabilization of FOXO1
.
Proc Natl Acad Sci U S A
2011
;
108
:
1555
60
.
41.
Zhao
B
,
Ye
X
,
Yu
J
,
Li
L
,
Li
W
,
Li
S
, et al
TEAD mediates YAP-dependent gene induction and growth control
.
Genes Dev
2008
;
22
:
1962
71
.
42.
Shen
YW
,
Zhou
YD
,
Chen
HZ
,
Luan
X
,
Zhang
WD
. 
Targeting CTGF in cancer: an emerging therapeutic opportunity
.
Trends Cancer
2021
;
7
:
511
24
.
43.
Wells
JE
,
Howlett
M
,
Cole
CH
,
Kees
UR
. 
Deregulated expression of connective tissue growth factor (CTGF/CCN2) is linked to poor outcome in human cancer
.
Int J Cancer
2015
;
137
:
504
11
.
44.
Weekes
D
,
Kashima
TG
,
Zandueta
C
,
Perurena
N
,
Thomas
DP
,
Sunters
A
, et al
Regulation of osteosarcoma cell lung metastasis by the c-Fos/AP-1 target FGFR1
.
Oncogene
2016
;
35
:
2852
61
.
45.
Ling
C
,
Su
VM
,
Zuo
D
,
Muller
WJ
. 
Loss of the 14–3-3sigma tumor suppressor is a critical event in ErbB2-mediated tumor progression
.
Cancer Discov
2012
;
2
:
68
81
.
46.
Ingles-Esteve
J
,
Morales
M
,
Dalmases
A
,
Garcia-Carbonell
R
.,
Jene-Sanz
A
,
Lopez-Bigas
N
, et al
Inhibition of specific NF-kappa B activity contributes to the tumor suppressor function of 14–3-3 sigma in breast cancer
.
PLoS One
2012
;
7
:
e38347
.
47.
McIlhatton
MA
,
Boivin
GP
,
Groden
J
. 
Manipulation of DNA repair proficiency in mouse models of colorectal cancer
.
Biomed Res Int
2016
;
2016
:
1414383
.
48.
Dhar
S
,
Squire
JA
,
Hande
MP
,
Wellinger
RJ
,
Pandita
TK
. 
Inactivation of 14–3-3sigma influences telomere behavior and ionizing radiation-induced chromosomal instability
.
Mol Cell Biol
2000
;
20
:
7764
72
.
49.
Yamada
Y
,
Hata
K
,
Hirose
Y
,
Hara
A
,
Sugie
S
,
Kuno
T
, et al
Microadenomatous lesions involving loss of Apc heterozygosity in the colon of adult Apc(Min/+) mice
.
Cancer Res
2002
;
62
:
6367
70
.
50.
Lamouille
S
,
Xu
J
,
Derynck
R
. 
Molecular mechanisms of epithelial-mesenchymal transition
.
Nat Rev Mol Cell Biol
2014
;
15
:
178
96
.
51.
Hou
Z
,
Peng
H
,
White
DE
,
Wang
P
,
Lieberman
PM
,
Halazonetis
T
, et al
14–3-3 binding sites in the snail protein are essential for snail-mediated transcriptional repression and epithelial-mesenchymal differentiation
.
Cancer Res
2010
;
70
:
4385
93
.
52.
Jackstadt
R
,
Sansom
OJ
. 
Mouse models of intestinal cancer
.
J Pathol
2016
;
238
:
141
51
.
53.
Halberg
RB
,
Katzung
DS
,
Hoff
PD
,
Moser
AR
,
Cole
CE
,
Lubet
RA
, et al
Tumorigenesis in the multiple intestinal neoplasia mouse: redundancy of negative regulators and specificity of modifiers
.
Proc Natl Acad Sci U S A
2000
;
97
:
3461
6
.
54.
Tanaka
T
,
Kohno
H
,
Suzuki
R
,
Hata
K
,
Sugie
S
,
Niho
N
, et al
Dextran sodium sulfate strongly promotes colorectal carcinogenesis in Apc(Min/+) mice: inflammatory stimuli by dextran sodium sulfate results in development of multiple colonic neoplasms
.
Int J Cancer
2006
;
118
:
25
34
.