The p53-inducible miR-34a and miR-34b/c genes are frequently silenced in colorectal cancer. To address the in vivo relevance of miR-34a/b/c function for suppression of intestinal tumor formation, we generated ApcMin/+ mice with deletions of the miR-34a and/or miR-34b/c genes separately or in combination. Combined deletion of miR-34a/b/c increased the number of intestinal stem cells as well as Paneth and Goblet cells, resulting in enlarged intestinal crypts. miR-34a/b/c-deficient ApcMin/+ mice displayed an increased tumor burden and grade and decreased survival. miR-34a/b/c-deficient adenomas showed elevated proliferation and decreased apoptosis and displayed pronounced bacterial infiltration, which may be due to an observed decrease in infiltrating immune cells and downregulation of barrier proteins. mRNA induction in miR-34a/b/c-deficient tumors was enriched for miR-34a/b/c seed-matching sites and for mRNAs encoding proteins related to epithelial–mesenchymal transition, stemness, and Wnt signaling. Accordingly, cells explanted from miR-34a/b/c-deficient adenomas formed tumor organoids at an increased rate. Several upregulated miR-34 targets displayed elevated expression in primary human colorectal cancers that was associated with lymph-node metastases (INHBB, AXL, FGFR1, and PDFGRB) and upregulation of INHBB and AXL in primary colorectal cancer was associated with poor patient survival. In conclusion, our results show that miR-34a/b/c suppress tumor formation caused by loss of Apc and control intestinal stem cell and secretory cell homeostasis by downregulation of multiple target mRNAs. Cancer Res; 77(10); 2746–58. ©2017 AACR.

Colorectal cancer is a leading cause of cancer death. In the United States alone, 134,490 new cases and 49,190 deaths from colorectal cancer were expected in 2016 (1). Colorectal cancer is a multistep process driven by mutational activation of several oncogenes and genetic/epigenetic inactivation of tumor suppressors (2). The majority of colorectal cancers originate from benign adenomas, which are caused by inactivating mutations in the adenomatous polyposis coli (APC) gene. APC mutation leads to the constitutive activation of the Wnt/β-catenin pathway, which results in the increased expression of β-catenin, a critical regulator of intestinal epithelial cell homeostasis (3). During colorectal cancer progression, additional mutations in key oncogenes and tumor suppressor genes, such as p53, are acquired. ApcMin/+ mice inherit a mutant Apc allele that results in a truncation of the APC protein at amino acid 850 (4) and spontaneously lose the wild-type Apc allele, resulting in multiple benign adenomas mainly throughout the small intestine (5). Thus, ApcMin/+ mice allow to study the influence of putative tumor suppressor genes on the initiation of intestinal tumorigenesis in vivo (6).

The tumor suppressor gene p53 is inactivated in the majority of colorectal cancers and encodes a transcription factor, which is posttranscriptionally induced by DNA damage and a number of additional cellular stresses (7). Besides regulating the expression of mRNAs, p53 also controls the expression of noncoding RNAs, such as lncRNAs and microRNAs (miRNA; refs. 8, 9). Many tumor suppressive functions of p53 are thought to be mediated by p53-induced miRNAs (8, 10). Among the p53-induced microRNA-encoding genes, miR-34a and miR-34b/c display the most consistent and pronounced induction, which may explain why they were identified as one of the first p53-regulated miRNAs (11).

Via downregulation of key-regulators, miR-34a/b/c suppress numerous cancer-associated processes, such as cell proliferation and survival (12), epithelial–mesenchymal transition (EMT; ref. 13), and stemness (14). Moreover, miR-34a/b/c presumably function as cell-fate determinants in colonic cancer stem cells (CSC) by suppressing Notch activity (15). Furthermore, miR-34a/b/c regulate the homeostasis of normal stem cells (16) and suppress the formation of induced pluripotent stem cells (IPSCs) (17).

Consistent with a tumor suppressive role in tumorigenesis, downregulation of miR-34a/b/c expression has been observed in a variety of human cancers (8). miR-34a/b/c expression is also epigenetically silenced by CpG methylation in colorectal cancer cell lines (18) and in primary colorectal cancers (19). These findings suggest an important role of miR-34a/b/c as downstream effectors of p53 and potential tumor suppressors in colorectal cancer. In line with its tumor suppressive potential miR-34a mimetics are currently tested in the clinics for treatment of advanced cancer (20).

Here, we report the generation of ApcMin/+ mice carrying targeted deletions of the miR-34a and miR-34b/c genes and the characterization of the resulting phenotypes at the organismal and molecular level. Our findings provide genetic proof of a tumor suppressive function of the miR-34a and miR-34b/c genes during intestinal tumorigenesis.

Animals

The generation of miR-34a−/− mice with a C57BL6/SV129 background has been described previously (21). miR-34b/cfl/fl mice were kindly provided by Dr. Alexander Nikitin (Cornell University, Ithaca, NY; ref. 16). In brief, gene-specific deletions were generated by using homologous recombination with a vector containing miR-34a or miR-34b/c sequences flanked by loxP sites and an intronic Neomycin resistance (Neo) cassette flanked frt sites individually (21). The Neo cassette was removed by crossing with flp-mice and germline miR-34a or miR-34b/c knock-out mice were generated by crossing with CMV-Cre mice. miR-34a−/−; b/c−/− compound mice were generated by crossing miR-34a−/− and miR-34b/c−/−. The resulting genotypes were obtained in the expected Mendelian ratios and the offspring displayed no overt phenotype. miR-34a−/−, miR-34b/c−/−, miR-34a−/−; miR-34b/c−/− and miR-34a+/+; miR-34bc+/+ mice were crossed with ApcMin/+ mice to obtain mice with the following genotypes: miR-34a−/−; ApcMin/+, miR-34b/c−/−; ApcMin/+, miR-34a/b/c−/−; ApcMin/+, and miR-34a/b/c+/+ApcMin/+. Mice were housed in individually ventilated cages (IVC). Animal studies were approved by the Government of Upper Bavaria, Germany (AZ 55.2-1-54-2532-4-2014).

Tissue preparation and tumor count

Mice were sacrificed at 18 weeks of age. Whole intestines were isolated, separated into four equal parts, flushed with PBS, opened longitudinally, photographed, and fixed as Swiss rolls in 4% buffered formaldehyde. Tumor numbers and size were evaluated using ImageJ software.

Histology and IHC

Three-micron paraffin sections were used for hematoxylin and eosin staining and periodic acid–Schiff (PAS) staining according to standard protocols. Tumors from ApcMin/+ 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. Immunohistochemistry was performed using antibodies and reagents listed in Supplementary Table S1. For each immunohistochemical detection ≥ 40 tumors per genotype were examined. Positive staining was evaluated by Image-Pro plus and ImageJ software.

In situ hybridization and fluororescence in situ hybridization

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). in situ hybridization (ISH) was performed as described (22). To visualize intestinal bacteria by fluorescence in situ hybridization, 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 3-μm paraffin sections.

Transcriptomic analysis

Total RNA from tumors was isolated using the RNeasy Plus Mini Kit (Qiagen) with an on-column DNase digestion (three RNA samples per genotype; each tumor RNA sample represented a pool of 3 tumors isolated from the same mouse). Random primed cDNA libraries were generated and sequenced using the HiSeq2500 (Illumina) platform by GATC (Konstanz). Each sample was covered by at least 35 million single reads of 50 bp length. Data were normalized in R with the RUVSeq (23) module and differential expression analysis was performed using Chipster (24). The overlap of DEseq2 and edgeR results were considered to represent the most significantly, differentially expressed genes. Gene Ontology (GO) and KEGG pathway analysis was performed with DAVID Bioinformatics Resources (25). Gene Set Enrichment Analysis (GSEA) was performed using the GSEA (26) software. miR-34a/b/c targets were predicted with TargetScan 6.2 (27). Expression data were deposited in the Gene Expression Omnibus website (accession no. GSE84138).

Cell lines and culture, generation of cell pools with conditional pri-miR-34a expression, and tumor organoid culture

H1299 lung cancer cells used for 3′-UTR reporter assays were from own stocks and their negativity for p53 was validated. SW620 colorectal cancer cells used for ectopic pri-miR-34a expression were from own stocks and authenticated by STR analysis in 2014 (Eurofins Medigenomix Forensik GmbH, Ebersberg, Germany). H1299 and SW620 cancer cell lines were kept in Dulbecco's modified Eagle's medium (DMEM) and 10% fetal calf serum (Invitrogen) at 5% CO2. SW620 cells were transfected with episomal pRTR-pri-miR-34a expression plasmids as described before (13). After 24 hours, cell pools were selected by addition of puromycin (2 μg/mL) for 10 days. GFP expression was evaluated by fluorescence microscopy 48 hours after addition of 100 ng/mL doxycycline (DOX) to the cell pools. Intestinal adenoma cells from ApcMin/+ mice were isolated and counted using a hemocytometer. Single cells (15,000) were embedded in 50 μL Matrigel per well in 24-well plates. The tumor organoid culture medium was formulated as described before (28).

3′-UTR reporter assays

The 3′-UTR of Wasf1 was PCR amplified from genomic DNA isolated from mouse tissue, inserted into pGL3-control-MCS vector downstream of a firefly luciferase ORF, and verified by sequencing. H1299 cells were seeded in a 12-well plate with 3 × 104 cells/well and transfected with 100 ng of pGL3-Wasf1 plasmid, 20 ng of Renilla reporter plasmid for normalization, and 25 nmol/L miR-34a or miR-34c pre-miRNAs or a negative control oligonucleotide (Ambion). Forty-eight hours later, luciferase assays were performed with the Dual-Luciferase Reporter 1000 Assay System (Promega). Fluorescence intensities were measured with an Orion II Microplate Luminometer (Titertek-Berthold). Primers used for cloning and sequencing are listed in Supplementary Table S2.

Quantitative real-time PCR and Western blot analysis

Total RNA was isolated from tumor samples or cultured cell lines using the RNeasy Plus Mini Kit (Qiagen). cDNA was generated by Verso cDNA kit (Thermo Scientific) and quantitative real-time PCR (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 (29). The individual mRNA levels were normalized to β-actin. Primers used for qPCR are listed in Supplementary Table S3.

For protein lysates tumor samples or cultured cells were lysed in RIPA buffer containing cOmplete Mini protease inhibitor cocktail tablets (Roche Diagnostics). Lysates were sonicated and centrifuged. Whole-lysate proteins (30–60 μg) were loaded per lane. Gel electrophoresis and transfer to PVDF membranes (Millipore) were carried out using standard protocols (Bio-Rad Laboratories). Primary antibodies (Supplementary Table S1) were used in combination with HRP-coupled secondary antibodies. ECL (Millipore) signals were recorded with a 440CF imaging system (Kodak).

Analysis of online expression data sets

The Cancer Genome Atlas (TCGA; ref. 30) gene expression data and follow-up information of colon adenocarcinomas (COAD) were downloaded from NCI's Genomic Data Commons website (GDC; https://gdc.cancer.gov/). Normalized RSEM counts were used to determine the expression of relevant mRNAs. Clinical outcome data were divided into high, intermediate, and low expression groups according to the expression value of individual genes.

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). Univariate, age/gender/tumor grade-adjusted hazard ratios, and 95% confidence intervals (CI) were estimated with a Cox's proportional hazard model. Prism 6 (GraphPad software) or SPSS (IBM) programs were used for calculations.

miR-34a/b/c deletion modulates the architecture of the small intestine in mice

We generated mice carrying deletions of the miR-34a or miR-34b/c loci and combinations of these. Deletion of miR-34a, miR-34b/c, or miR-34a/b/c in the germline did not significantly influence the lifespan of mice (Fig. 1A). However, we observed a gender-independent increase in the width and depth of crypts of the small intestine in miR-34a/b/c-deficient mice, which presumably caused the minor increase in the total length of the small intestine (Fig. 1B). Notably, the numbers of Paneth cells per crypt and Goblet cells per villus were significantly increased in miR-34a/b/c-deficient mice (Fig. 1C and D). In addition, the frequency of stem cells at the crypt base was significantly increased in miR-34a/b/c-deficient mice as determined by detection of the stem cell marker Olfm4 using in situ hybridization (Fig. 1E). These phenotypes were independent of the gender (Fig. 1B–E).

In order to determine, whether inactivation of miR-34 genes affects intestinal tumor formation, we generated ApcMin/+ mice with deletions of miR-34a, miR-34b/c, or miR-34a/b/c. Also in ApcMin/+ mice deficiency for miR-34a/b/c resulted in an increase of the number of Paneth, Goblet, and Olmf4-positive cells per crypt in untransformed tissue of the small intestine (Supplementary Fig. S1A–D). Similarly, the width and depth of the crypts were increased significantly in miR-34a/b/c-deficient ApcMin/+ mice, but the minor increase in the length of the small intestine was not statistically significant (Supplementary Fig. S1E). Again, these architectural changes were observed in both, male and female mice. Therefore, deletion of one APC allele did not influence the morphological changes in the intestine caused by deletion of miR-34a/b/c.

miR-34a/b/c loss enhances tumorigenesis in ApcMin/+ mice

Notably, miR-34a/b/c−/−; ApcMin/+ mice showed signs of morbidity sooner and exhibited a shorter life-span than wild-type ApcMin/+ mice (Fig. 2A). In contrast, ApcMin/+ mice deficient for either the miR-34a or the miR-34b/c allele did not show a statistically significant change in life-span (Fig. 2A). When mice were sacrificed at the age of 18 weeks and the entire small intestinal tract was examined, miR-34a/b/c−/−; ApcMin/+ and, to a lesser extent, miR-34a−/−; ApcMin/+ knockout mice showed a significantly increased number of tumors when compared with ApcMin/+ mice (Fig. 2B and C). Furthermore, the total tumor area in miR-34a/b/c−/−; ApcMin/+ mice was ∼4 times larger than in wild-type ApcMin/+ mice (Fig. 2D). The frequency of large tumors (>6 mm2) was significantly higher when miR-34a/b/c, but not when only miR-34a or miR-34b/c genes had been deleted in ApcMin/+ mice (Fig. 2E). We also noted a significantly increased number of adenomas with high-grade dysplasia in miR-34a/b/c-deficient ApcMin/+ mice, whereas the inactivation of either miR-34a or miR-34b/c alone did not have this effect (Fig. 2F). Taken together, our findings show that the inactivation of miR-34a/b/c promotes intestinal tumor formation by increasing tumor initiation and enhancing the growth of tumors in ApcMin/+ mice. As the combined inactivation of miR-34a and miR-34b/c genes was necessary for these effects, these miRNAs may have overlapping functions and compensate each other. In addition, these results are in accordance with the combined epigenetic inactivation of miR-34a and miR-34b/c by DNA methylation, which has been detected in more than 75% of analyzed colorectal cancer samples (31). In the subsequent analyses, we therefore focused on studying mice with simultaneous deletion of miR-34a and miR-34b/c.

miR-34a/b/c loss affects proliferation, apoptosis, and infiltration by bacteria and immune cells of adenomas

Notably, tumors in miR-34a/b/c knockout ApcMin/+ mice showed a significant increase in proliferation and a significantly reduced rate of apoptosis (Fig. 3A). The frequency of stromal cells within the tumors was not affected by miR-34a/b/c deletion as determined by detection of vimentin. Because it was recently shown that bacterial infiltration promotes tumorigenesis in ApcMin/+ mice (32), we asked whether miR-34a/b/c deletion affects the presence of bacteria in intestinal tumors. By detection of bacterial 16S rRNA, we found that adenomas from miR-34a/b/c−/−; ApcMin/+ mice display extensive bacterial infiltration regardless of tumor size (Fig. 3A) or tumor grade (Supplementary Fig. S2A). However, the adjacent normal intestinal epithelium of all mice analyzed here was devoid of bacteria. Therefore, miR-34a/b/c-deficient mice do not display a general intestinal barrier defect. Unexpectedly, the frequency of immune cells, such as macrophages, T- and B-cell, was decreased in miR-34a/b/c-deficient adenomas (Fig. 3B; Supplementary Fig. S2B). Therefore, the increased bacterial infiltration of adenomas may be due to a defect in the tumor-associated immune defense in miR-34a/b/c-deficient mice.

Expression profiling of miR-34a/b/c-deficient adenomas

To further illuminate the mechanisms by which miR-34a/b/c loss promotes intestinal tumorigenesis in ApcMin/+ mice, we obtained mRNA expression profiles of adenomas from 18-week-old miR-34a/b/c-deficient ApcMin/+ mice and wild-type ApcMin/+ mice. For each mouse (n = 3 per genotype), RNAs derived from three different tumors were pooled, libraries were generated and subjected to RNA-Seq to obtain more than 35 million reads per library. Thereby, we detected the upregulation of 1773 mRNAs, when mRNAs with a log2-fold change ≥ 0.6 and RPKM ≥ 0.25 in miR-34a/b/c−/−; ApcMin/+ mice versus wild-type ApcMin/+ mice were included (Fig. 4A). As expected, the percentage of mRNAs upregulated in the miR-34a/b/c-deficient tumors was higher for mRNAs harboring miR-34a/b/c seed-matching sequences than for mRNAs lacking these (Fig. 4B). Three hundred and twenty-four mRNAs were identified as differentially regulated due to miR-34a/b/c loss when the raw-count methods DESeq2 (33) and edgeR (34) were applied (Fig. 4C). After unsupervised clustering of these differentially expressed genes, one of the miR-34a/b/c-deficient tumor samples showed a divergent expression profile, which was presumably due to biological variation (Fig. 4D). Next, we used Gene Ontology (GO) and KEGG pathway analyses to group the differentially expressed transcripts into functional classes. The GO analysis indicated that the upregulated mRNAs are involved in cell adhesion, extracellular matrix, and growth factor binding functions (Supplementary Fig. S3A). The KEGG analysis showed that several pathways were significantly enriched, such as focal adhesion, ECM-receptor interaction, and pathways related to cancer (Supplementary Fig. S3B). On the contrary, both GO and KEGG analyses indicated that the downregulated genes are mainly involved in immune response functions (Supplementary Fig. S4A and S4B). A subsequent Gene Set Enrichment Analysis (GSEA) revealed that miR-34a/b/c deletion results in gene expression signatures related to EMT, hypoxia, angiogenesis, inflammatory response, Kras signaling, and apical junction processes (Fig. 4E). Interestingly, the expression signatures of miR-34a/b/c-deficient adenomas also showed a significant overlap with signatures of intestinal stem cells (ISC) (35), Lgr5+ positive cells (36) and the GO Wnt signaling signature. In addition, several regulators of barrier function in IEC were also detected as downregulated by NGS analysis in miR-34a/b/c-deficient adenomas (e.g., Muc1, Tff3, and Retnlb; fold change: −4.74, −1.44, and −3.34, respectively). The downregulated mRNA expression of these barrier components was validated by qPCR (Supplementary Fig. S5A). Furthermore, we confirmed the decreased protein expression of MUC1 in miR-34a/b/c-deficient adenomas by immunohistochemistry (Supplementary Fig. S5B). The repression of these barrier components may also contribute to the observed infiltration of bacteria into miR-34a/b/c-deficient adenomas.

miR-34a/b/c loss enhances the formation of intestinal tumor organoids

To obtain functional evidence for an enhanced stemness of adenoma cells with deletion of miR-34a/b/c, we performed a tumor organoid formation assay. Indeed, an increased tumor organoid formation rate was observed for miR-34a/b/c-deficient adenoma derived cells when compared with those derived from miR-34a/b/c-proficient adenomas (Fig. 5A).

miR-34a/b/c deletion enhances intestinal Wnt signaling in ApcMin/+ mice

Wnt/β-catenin signaling is critically involved in regulating the homeostasis and neoplastic transformation of the intestine. Because 34a/b/c-deficient adenomas showed a Wnt signaling signature (Fig. 4E) and the Wnt/β-catenin pathway is directly regulated by miR-34a/b/c (37) and by APC, we determined whether the expression of β-catenin is also affected in the intestinal epithelial cells of miR-34a/b/c knockout mice. Interestingly, we detected an increased nuclear accumulation of β-catenin/CTNNB1 protein in normal crypts after miR-34a/b/c deletion in ApcMin/+ mice, but not in miR-34a/b/c-deficient wild-type mice (Fig. 5B), indicating that miR-34a/b/c deficiency in the presence of hemizygous Apc allows the nuclear accumulation of β-catenin in untransformed cells, including the stem cells located at the crypt base, which represent the preferred cells of origin for intestinal adenomas (38). Therefore, this effect of miR-34a/b/c loss may explain or at least contribute to the increased rate of tumor initiation in miR-34a/b/c-deficient ApcMin/+ mice.

Validation of miR-34a/b/c target expression

By bioinformatics analyses of the mRNA profiles obtained here, we identified a set of 11 upregulated mRNAs with miR-34 seed-matching sites in their 3′-UTR, which may, at least in part, mediate the tumor suppressive function of miR-34a/b/c (Supplementary Table S4; Fig. 6A). These mRNAs and a selected set of mRNAs with known pro-tumorigenic functions were analyzed by qPCR to exemplarily validate the NGS results (Fig. 6B). Several of these factors have already been characterized as direct miR-34 targets by others and us (Pdgfra (39), Pdgfrb (39), and Axl (40)). In addition, components of the Wnt pathway and EMT regulators were analyzed. Notably, all of the tested mRNAs were expressed at significantly higher levels in the tumors of miR-34a/b/c knockout ApcMin/+ mice. Therefore, our screen identified numerous additional miR-34a/b/c targets, which may be upregulated in colorectal cancer and relevant for tumor progression.

Next, we performed an exemplary characterization of Wasf1 as a direct miR-34a/b/c target, because Wasf1 is known to regulate cancer-relevant processes (41). A luciferase-based reporter containing the full-length 3′-UTR of the murine Wasf1 mRNA was significantly repressed by cotransfection of pre-miR-34a or pre-miR-34c (Fig. 6C). In addition, we detected a significant reduction of Wasf1 mRNA expression after ectopic expression of pri-miR-34a in SW620 colorectal cancer cells (Fig. 6D). The WASF1 protein showed a delayed repression 72 hours after activation of the pri-miR-34a allele, which may be due to an unusually stable WASF1 protein (Fig. 6E). Consistent with being a miR-34a/b/c target, the WASF1 protein showed increased expression in three out of four tumors isolated from miR-34a/b/c knockout ApcMin/+ mice, when compared with miR-34a/b/c-proficient tumors (Fig. 6F). Taken together, these results show that Wasf1 is a direct target of miR-34a/b/c.

Increased expression of miR-34a/b/c targets in human colorectal cancers is associated with poor survival

In order to determine whether the miR-34 targets described above are also clinically relevant, we analyzed their expression within the expression profiles of 460 human colorectal adenocarcinomas deposited in TCGA database (30). Interestingly, INHBB, PDGFRB, STC1, and COL4A2 showed a significantly elevated expression in primary colorectal cancers (Fig. 7A). Furthermore, a significantly increased expression of INHBB, AXL, FGFR1, and PDGFRB was detected in primary colorectal cancers from patients with nodal status N2 when compared with samples from patients with lower nodal status (Fig. 7B). Except for AXL, a significantly increased expression of these mRNAs was associated with an increased tumor stage (Supplementary Fig. S6A). In addition, increased expression of INHBB and AXL in primary colorectal cancers was significantly associated with decreased patient survival (Fig. 7C; Supplementary Table S5). Elevated expression of COL4A2, WASF1, STC1, and PDGFRB was also associated with poor survival, although not significantly (Supplementary Fig. S6B). Because poor survival is mostly due to metastatic spread, these results suggest that the elevated expression of these mRNAs, which may be due to loss of miR-34a/b/c, promotes metastasis.

Finally, we compiled the regulations identified here and combined them with previously published results on miR-34a/b/c-regulated pathways in a summarizing model shown in Fig. 7D. Notably, several of the miR-34 targets displaying elevated expression after miR-34a/b/c deletion are involved in transmembrane signal transductions, which also activate the Wnt signaling pathway by inhibition of GSK-3β. Taken together, miR-34a/b/c presumably suppresses intestinal tumorigenesis caused by loss of Apc by downregulating the expression of a large number of pro-tumorigenic factors. In case miR-34a/b/c expression is lost or silenced during tumor progression, these pathways may be further activated and thereby contribute to colorectal cancer progression.

The increase in the pool of intestinal stem cells (ISC) in the miR-34a/b/c-deficient mice observed here may underlie the increased rate of tumor formation in ApcMin/+ mice, because ISC were shown to serve as efficient tumor initiating cells during intestinal tumorigenesis (38). Paneth cells provide a niche for ISCs and communicate with these via multiple signaling pathways, among them the Delta/NOTCH and Wnt/APC/β-catenin pathways, which are under negative control by miR-34a/b/c (15, 37): that is, several key components of the Wnt signaling pathway are miR-34a/b/c targets (WNT1, WNT3, LRP6, LEF1, and β-catenin (37, 40). Moreover, GSK-3β, which phosphorylates β-catenin and thereby leads to its poly-ubiquitination and proteasomal degradation, is inhibited by the PKB/AKT and PI3K pathways, which are also under control of miR-34a/b/c. Therefore, the concerted deregulation of the NOTCH and WNT pathways caused by the loss of miR-34a/b/c is a likely cause for the increased number of Paneth and stem cells. In support of this scenario, we also noticed an increased tumor organoid formation rate in miR-34a/b/c−/−; ApcMin/+ adenomas as well as an increased accumulation of nuclear β-catenin in the untransformed, epithelial cells of intestinal crypts in miR-34a/b/c-deficient ApcMin/+ mice. In addition, a recent study suggests that activation of the Wnt/β-catenin pathway correlates with T-cell exclusion (42). Therefore, miR-34 may regulate immune responses through Wnt signaling. In addition, the expression of barrier proteins was decreased in miR-34a/b/c−/−; ApcMin/+ mice (e.g., Muc1, Tff3, and Retnlb). In combination with the decreased presence of immune cells, these effects of miR-34a/b/c loss may contribute, at least in part, to the increased bacterial infiltration observed in miR-34a/b/c−/−; ApcMin/+ adenomas.

In addition, we found that the deletion of miR-34a/b/c in adenomas leads to the upregulation of previously described miR-34a/b/c target mRNAs, such as Pdgfra, Pdgfrb, and Axl, which are known to enhance colorectal cancer formation (43). Furthermore, a set of novel, putative miR-34a/b/c target mRNAs (Wasf1, Fgfr1, Igf1, Stc1, Cacna2d2, Col6a2, Col4a2, and Inhbb), which were reported to be involved in tumorigenesis (41, 44–49), was upregulated in miR-34a/b/c-deficient adenomas. The factors encoded by these genes form complex signaling and functional networks (see also Fig. 7D): PDGFRA, PDGFRB, FGFR1, AXL, and IGF1, which are either ligands or receptors of the tyrosine kinase family, activate the PI3K and MAPK signaling pathway, and thereby promote cell growth, survival, EMT, and metastasis. Notably, CACNA2D2, a voltage-dependent calcium channel, deregulates calcium homeostasis when ectopically expressed (47). The resulting calcium release may induce the activation of PKB/AKT and RAS to promote cell proliferation and angiogenesis (47). INHBB assembles into activins, which are critical modulators of growth and survival (49). COL4A2 is a major structural protein of the basement membrane and COL6A2 has an anchoring function. Both collagens stimulate integrin signaling and are involved in cell growth, angiogenesis, and tumor metastasis (48). WASF1, a new miR-34a/b/c target characterized here, mediates actin polymerization, lamellipodia formation, and plays a critical role in cancer cell migration and invasion (41). STC1 is an endocrine regulator of calcium and phosphate homeostasis, which promotes cell proliferation and inhibits apoptosis through CCND1 and CDK2/4 (50). Taken together, miR-34a/b/c presumably act as global regulators to fine tune multiple cellular functions, which are necessary for the homeostasis of intestinal epithelia. Because miR-34a has been shown to form bimodal switches with at least some of its targets (15), the loss of miR-34a/b/c during colorectal cancer progression may have significant effects on the regulation of these signaling and expression networks, which ultimately promote intestinal tumorigenesis. miR-34a/b/c have a multitude of targets in intestinal epithelial and colon cancer cells as shown here and in previous studies (10). Therefore, it seems plausible that the effects of miR-34a/b/c loss are mediated by the combined activity of multiple upregulated targets and not by one or a few targets.

A subset of the direct targets of miR-34a/b/c may significantly contribute to human colorectal tumorigenesis because their elevated expression was associated with poor clinical outcomes of colorectal cancer patients. However, further analysis will be necessary to corroborate the role of miR-34a/b/c silencing for their increased expression in colorectal tumors. Our results suggest that miR-34 mimetics may be used to target multiple key pathways simultaneously and could thereby potentially prevent the emergence of resistance caused by mutations of single pathways. Therefore, miR-34a/b/c replacement therapy may represent a potential option for the treatment of colorectal cancer.

No potential conflicts of interest were disclosed.

Conception and design: H. Hermeking

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): L. Jiang, H. Hermeking

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): L. Jiang, H. Hermeking

Writing, review, and/or revision of the manuscript: L. Jiang, H. Hermeking

Study supervision: H. Hermeking

Other (experimental design): L. Jiang

We are grateful to Alexander Nikitin for providing miR-34b/cfl/fl mice, Marlon Schneider for ApcMin/+ mice, Hans Clevers for the Olfm4 plasmid, Matjaz Rokavec for initial help with setting up the ApcMin/+ mouse cohorts, and Markus Kaller for advice on bioinformatics analyses. We would also like to thank Peter Jung for discussions and Ursula Götz for technical assistance.

H. Hermeking received support from a Deutsche Krebshilfe Grant (No. 109531) and the Rudolf Bartling Stiftung (V/124). L. Jiang received a fellowship from the China Scholarship Council.

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.
Siegel
RL
,
Miller
KD
,
Jemal
A
. 
Cancer statistics, 2016
.
CA Cancer J Clin
2016
;
66
:
7
30
.
2.
Kinzler
KW
,
Vogelstein
B
. 
Lessons from hereditary colorectal cancer
.
Cell
1996
;
87
:
159
70
.
3.
Bienz
M
,
Clevers
H
. 
Linking colorectal cancer to Wnt signaling
.
Cell
2000
;
103
:
311
20
.
4.
Moser
AR
,
Mattes
EM
,
Dove
WF
,
Lindstrom
MJ
,
Haag
JD
,
Gould
MN
. 
ApcMin, a mutation in the murine Apc gene, predisposes to mammary carcinomas and focal alveolar hyperplasias
.
Proc Natl Acad Sci U S A
1993
;
90
:
8977
81
.
5.
Luongo
C
,
Moser
AR
,
Gledhill
S
,
Dove
WF
. 
Loss of Apc+ in intestinal adenomas from Min mice
.
Cancer Res
1994
;
54
:
5947
52
.
6.
Moser
AR
,
Luongo
C
,
Gould
KA
,
McNeley
MK
,
Shoemaker
AR
,
Dove
WF
. 
ApcMin: a mouse model for intestinal and mammary tumorigenesis
.
Eur J Cancer
1995
;
31A
:
1061
4
.
7.
Vogelstein
B
,
Lane
D
,
Levine
AJ
. 
Surfing the p53 network
.
Nature
2000
;
408
:
307
10
.
8.
Hermeking
H
. 
MicroRNAs in the p53 network: micromanagement of tumour suppression
.
Nat Rev Cancer
2012
;
12
:
613
26
.
9.
Hunten
S
,
Kaller
M
,
Drepper
F
,
Oeljeklaus
S
,
Bonfert
T
,
Erhard
F
, et al
p53-Regulated networks of protein, mRNA, miRNA, and lncRNA expression revealed by integrated pulsed stable isotope labeling with amino acids in cell culture (pSILAC) and next generation sequencing (NGS) analyses
.
Mol Cell Proteomics
2015
;
14
:
2609
29
.
10.
Rokavec
M
,
Li
H
,
Jiang
L
,
Hermeking
H
. 
The p53/miR-34 axis in development and disease
.
J Mol Cell Biol
2014
;
6
:
214
30
.
11.
Hermeking
H
. 
p53 enters the microRNA world
.
Cancer Cell
2007
;
12
:
414
8
.
12.
Tarasov
V
,
Jung
P
,
Verdoodt
B
,
Lodygin
D
,
Epanchintsev
A
,
Menssen
A
, et al
Differential regulation of microRNAs by p53 revealed by massively parallel sequencing: miR-34a is a p53 target that induces apoptosis and G1-arrest
.
Cell Cycle
2007
;
6
:
1586
93
.
13.
Siemens
H
,
Jackstadt
R
,
Hunten
S
,
Kaller
M
,
Menssen
A
,
Gotz
U
, et al
miR-34 and SNAIL form a double-negative feedback loop to regulate epithelial-mesenchymal transitions
.
Cell Cycle
2011
;
10
:
4256
71
.
14.
Liu
C
,
Kelnar
K
,
Liu
B
,
Chen
X
,
Calhoun-Davis
T
,
Li
H
, et al
The microRNA miR-34a inhibits prostate cancer stem cells and metastasis by directly repressing CD44
.
Nat Med
2011
;
17
:
211
5
.
15.
Bu
P
,
Wang
L
,
Chen
KY
,
Srinivasan
T
,
Murthy
PK
,
Tung
KL
, et al
A miR-34a-numb feedforward loop triggered by inflammation regulates asymmetric stem cell division in intestine and colon cancer
.
Cell Stem Cell
2016
;
18
:
189
202
.
16.
Cheng
CY
,
Hwang
CI
,
Corney
DC
,
Flesken-Nikitin
A
,
Jiang
L
,
Oner
GM
, et al
miR-34 cooperates with p53 in suppression of prostate cancer by joint regulation of stem cell compartment
.
Cell Rep
2014
;
6
:
1000
7
.
17.
Choi
YJ
,
Lin
CP
,
Ho
JJ
,
He
X
,
Okada
N
,
Bu
P
, et al
miR-34 miRNAs provide a barrier for somatic cell reprogramming
.
Nat Cell Biol
2011
;
13
:
1353
60
.
18.
Lodygin
D
,
Tarasov
V
,
Epanchintsev
A
,
Berking
C
,
Knyazeva
T
,
Korner
H
, et al
Inactivation of miR-34a by aberrant CpG methylation in multiple types of cancer
.
Cell cycle
2008
;
7
:
2591
600
.
19.
Siemens
H
,
Neumann
J
,
Jackstadt
R
,
Mansmann
U
,
Horst
D
,
Kirchner
T
, et al
Detection of miR-34a promoter methylation in combination with elevated expression of c-Met and β-catenin predicts distant metastasis of colon cancer
.
Clin Cancer Res
2013
;
19
:
710
20
.
20.
Adams
BD
,
Parsons
C
,
Slack
FJ
. 
The tumor-suppressive and potential therapeutic functions of miR-34a in epithelial carcinomas
.
Expert Opin Ther Targets
2016
;
20
:
737
53
.
21.
Rokavec
M
,
Oner
MG
,
Li
H
,
Jackstadt
R
,
Jiang
L
,
Lodygin
D
, et al
IL-6R/STAT3/miR-34a feedback loop promotes EMT-mediated colorectal cancer invasion and metastasis
.
J Clin Invest
2014
;
124
:
1853
67
.
22.
Gregorieff
A
,
Clevers
H
. 
In situ hybridization to identify gut stem cells
.
Curr Protoc Stem Cell Biol
2010
;
Chapter 2
:
Unit 2F 1
.
23.
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
.
24.
Kallio
MA
,
Tuimala
JT
,
Hupponen
T
,
Klemela
P
,
Gentile
M
,
Scheinin
I
, et al
Chipster: user-friendly analysis software for microarray and other high-throughput data
.
BMC Genom
2011
;
12
:
507
.
25.
Huang da
W
,
Sherman
BT
,
Lempicki
RA
. 
Bioinformatics enrichment tools: paths toward the comprehensive functional analysis of large gene lists
.
Nucleic Acids Res
2009
;
37
:
1
13
.
26.
Subramanian
A
,
Tamayo
P
,
Mootha
VK
,
Mukherjee
S
,
Ebert
BL
,
Gillette
MA
, et al
Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles
.
Proc Natl Acad Sci U S A
2005
;
102
:
15545
50
.
27.
Lewis
BP
,
Burge
CB
,
Bartel
DP
. 
Conserved seed pairing, often flanked by adenosines, indicates that thousands of human genes are microRNA targets
.
Cell
2005
;
120
:
15
20
.
28.
Sato
T
,
Stange
DE
,
Ferrante
M
,
Vries
RG
,
Van Es
JH
,
Van den Brink
S
, et al
Long-term expansion of epithelial organoids from human colon, adenoma, adenocarcinoma, and Barrett's epithelium
.
Gastroenterology
2011
;
141
:
1762
72
.
29.
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
.
30.
Cancer Genome Atlas
N
. 
Comprehensive molecular characterization of human colon and rectal cancer
.
Nature
2012
;
487
:
330
7
.
31.
Vogt
M
,
Munding
J
,
Gruner
M
,
Liffers
ST
,
Verdoodt
B
,
Hauk
J
, et al
Frequent concomitant inactivation of miR-34a and miR-34b/c by CpG methylation in colorectal, pancreatic, mammary, ovarian, urothelial, and renal cell carcinomas and soft tissue sarcomas
.
Virchows Arch
2011
;
458
:
313
22
.
32.
Lee
SH
,
Hu
LL
,
Gonzalez-Navajas
J
,
Seo
GS
,
Shen
C
,
Brick
J
, et al
ERK activation drives intestinal tumorigenesis in Apc(min/+) mice
.
Nat Med
2010
;
16
:
665
70
.
33.
Love
MI
,
Huber
W
,
Anders
S
. 
Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2
.
Genome Biol
2014
;
15
:
550
.
34.
Robinson
MD
,
McCarthy
DJ
,
Smyth
GK
. 
edgeR: a Bioconductor package for differential expression analysis of digital gene expression data
.
Bioinformatics
2010
;
26
:
139
40
.
35.
Munoz
J
,
Stange
DE
,
Schepers
AG
,
van de Wetering
M
,
Koo
BK
,
Itzkovitz
S
, et al
The Lgr5 intestinal stem cell signature: robust expression of proposed quiescent ‘+4′ cell markers
.
EMBO J
2012
;
31
:
3079
91
.
36.
Merlos-Suarez
A
,
Barriga
FM
,
Jung
P
,
Iglesias
M
,
Cespedes
MV
,
Rossell
D
, et al
The intestinal stem cell signature identifies colorectal cancer stem cells and predicts disease relapse
.
Cell Stem Cell
2011
;
8
:
511
24
.
37.
Kim
NH
,
Kim
HS
,
Kim
N-G
,
Lee
I
,
Choi
H-S
,
Li
X-Y
, et al
p53 and miRNA-34 are Suppressors of Canonical Wnt Signaling
.
Sci Signal
2011
;
4
:
ra71
.
38.
Barker
N
,
Ridgway
RA
,
van Es
JH
,
van de Wetering
M
,
Begthel
H
,
van den Born
M
, et al
Crypt stem cells as the cells-of-origin of intestinal cancer
.
Nature
2009
;
457
:
608
11
.
39.
Garofalo
M
,
Jeon
YJ
,
Nuovo
GJ
,
Middleton
J
,
Secchiero
P
,
Joshi
P
, et al
MiR-34a/c-Dependent PDGFR-alpha/beta downregulation inhibits tumorigenesis and enhances TRAIL-Induced apoptosis in lung cancer
.
PloS One
2013
;
8
:
e67581
.
40.
Kaller
M
,
Liffers
S-T
,
Oeljeklaus
S
,
Kuhlmann
K
,
Röh
S
,
Hoffmann
R
, et al
Genome-wide characterization of miR-34a induced changes in protein and mRNA expression by a combined pulsed SILAC and microarray analysis
.
Mol Cell Proteo
2011
;
10
:
M111. 010462
.
41.
Zhang
J
,
Zhou
S
,
Tang
L
,
Shen
L
,
Xiao
L
,
Duan
Z
, et al
WAVE1 gene silencing via RNA interference reduces ovarian cancer cell invasion, migration and proliferation
.
Gynecol Oncol
2013
;
130
:
354
61
.
42.
Corrales
L
,
Matson
V
,
Flood
B
,
Spranger
S
,
Gajewski
TF
. 
Innate immune signaling and regulation in cancer immunotherapy
.
Cell Res
2016
;
27
:
96
108
.
43.
Craven
RJ
,
Xu
LH
,
Weiner
TM
,
Fridell
YW
,
Dent
GA
,
Srivastava
S
, et al
Receptor tyrosine kinases expressed in metastatic colon cancer
.
Int J Cancer
1995
;
60
:
791
7
.
44.
Grose
R
,
Dickson
C
. 
Fibroblast growth factor signaling in tumorigenesis
.
Cytokine Growth Factor Rev
2005
;
16
:
179
86
.
45.
Olivo‐Marston
SE
,
Hursting
SD
,
Lavigne
J
,
Perkins
SN
,
Maarouf
RS
,
Yakar
S
, et al
Genetic reduction of circulating insulin‐like growth factor‐1 inhibits azoxymethane‐induced colon tumorigenesis in mice
.
Mol Carcinogen
2009
;
48
:
1071
6
.
46.
Tamura
S
,
Oshima
T
,
Yoshihara
K
,
Kanazawa
A
,
Yamada
T
,
Inagaki
D
, et al
Clinical significance of STC1 gene expression in patients with colorectal cancer
.
Anticancer Res
2011
;
31
:
325
9
.
47.
Warnier
M
,
Roudbaraki
M
,
Derouiche
S
,
Delcourt
P
,
Bokhobza
A
,
Prevarskaya
N
, et al
CACNA2D2 promotes tumorigenesis by stimulating cell proliferation and angiogenesis
.
Oncogene
2015
;
34
:
5383
94
.
48.
Worthley
DL
,
Giraud
AS
,
Wang
TC
. 
The extracellular matrix in digestive cancer
.
Cancer Microenviron
2010
;
3
:
177
85
.
49.
Togashi
Y
,
Kogita
A
,
Sakamoto
H
,
Hayashi
H
,
Terashima
M
,
de Velasco
MA
, et al
Activin signal promotes cancer progression and is involved in cachexia in a subset of pancreatic cancer
.
Cancer Lett
2015
;
356
:
819
27
.
50.
Du
Y-Z
,
Gu
X-H
,
Cheng
S-F
,
Li
L
,
Liu
H
,
Hu
L-P
, et al
The oncogenetic role of stanniocalcin 1 in lung adenocarcinoma: a promising serum candidate biomarker for tracking lung adenocarcinoma progression
.
Tumor Biol
2015
:
1
12
.