Missense-type p53 mutations have shown to acquire novel oncogenic roles through a gain-of-function mechanism. However, there is intratumor heterogeneity in stabilization of mutant p53 protein, and it has not been well understood about the interaction between p53-stabilized and -destabilized cells in the same tumors. We established mouse intestinal tumor–derived organoids carrying ApcΔ716, KrasG12D, and Tgfbr2−/− mutations with Trp53R270H or Trp53Null mutation (AKTPR270H and AKTPNull, respectively). Using these organoids, we found that the activation level of Wnt/β-catenin signaling is significantly higher in AKTPR270H cells compared with AKTPNull cells. Notably, Wnt activation in the AKTPNull cells was significantly increased when co-cultured with AKTPR270H cells. Expression analysis revealed that COX-2 is significantly upregulated in AKTPR270H but not in AKTPNull cells, suggesting that mutant p53 induces the COX-2/prostaglandin E2 (PGE2) pathway. Importantly, Wnt activation in co-cultured AKTPNull cells with AKTPR270H was significantly suppressed when treated with the inhibitor of COX-2 or PGE2 receptors EP2/EP4. Furthermore, stimulation with PGE2 increased Wnt signaling activity in AKTPNull cells. These results indicate that the COX-2/PGE2 pathway is activated in the p53-stabilized cells in the missense-type p53-mutant cancer, and secreted PGE2 may transactivate Wnt/β-catenin signaling in neighboring p53-destabilized tumor cells in the intratumor microenvironment. Therefore, targeting stabilized mutant p53 or the COX-2/PGE2 pathway may suppress Wnt/β-catenin signaling of both mutant p53–stabilized and –destabilized cells; thus, this can be a possible preventive or therapeutic strategy.

Significance:

There is intratumor heterogeneity in the stabilization of missense mutant p53, and it has been thought that only cells with nuclear accumulation of mutant p53 have oncogenic function. However, using mouse intestinal tumor–derived organoids, we show that mutant p53–stabilized cells transactivate Wnt/β-catenin signaling in neighboring p53-destabilized cells through activating the COX-2/PGE2 pathway. These results suggest that both p53-stabilized cells and p53-destabilized cells contribute to malignant progression through interaction within the intratumor microenvironment.

TP53 is one of the most frequently mutated genes in a wide range of cancers, including colorectal cancer (1, 2), and the TP53 mutations were found in about 80% of colorectal cancer cases that are associated with metastasis (3). These results suggest that TP53 mutation plays an important role in the late stage of cancer progression. The target molecules of p53 regulate a variety of biological processes, including cell-cycle arrest, DNA repair, and senescence, which may prevent tumor progression as a tumor suppressor (4). On the other hand, approximately 75% of TP53 mutations in cancer are missense mutations in hot spots, resulting in the expression of mutant p53 with single amino acid substitutions, and such mutant p53 is thought to acquire a novel oncogenic function (5, 6).

Such gain of function of mutant p53 has been demonstrated in mouse genetic studies (7, 8). Trp53-null mutant mice spontaneously develop soft-tissue sarcomas and lymphomas, whereas mice expressing missense-type mutant p53 develop adenocarcinomas in the intestine and lung. It has also been reported that mutant p53 induces stem cell properties in breast and colorectal cancer cells (9, 10). In addition, we showed that the Trp53R270H missense mutation (corresponding to human TP53R273H) causes submucosal invasion of intestinal tumors in ApcΔ716-mutant mice through activation of the inflammatory and Wnt signaling pathways (11). We also constructed a malignant intestinal tumor model by introducing ApcΔ716, KrasG12D, Tgfbr2−/−, and Trp53R270H mutations to generate quadruple-mutant mouse model (12) and showed that missense-type mutant p53 promotes the survival and clonal expansion ability of single cell–dissociated cancer cells, which may contribute to colonization of disseminated cells in distant organs (13). It has also been reported that in vivo growth of Trp53R248Q mouse tumors depends on sustained expression of mutant p53 (14). Similar results also showed that ablation of mutant p53R248Q suppressed growth and invasion of mutant p53–driven tumors (15). These results suggest a possibility of gain-of-function mutant p53 as a therapeutic target molecule.

Although the genetic evidence for gain of function of mutant p53 has been accumulated, the underlying mechanisms of mutant p53–induced tumor promotion have not been fully understood yet. It has been shown that missense mutant p53 dramatically alters the transcriptome in tumor cells through modification of chromatin regulation by upregulation of chromatin regulatory genes or interaction with the SWI/SNF complex (16, 17). However, the precise molecular mechanisms of how mutant p53 promotes malignant phenotypes are still unclear. Moreover, there is intratumor heterogeneity in stabilization of mutant p53 (18), and it has not yet been understood whether there is interaction in oncogenic function between p53-stabilized and -destabilized cells.

In this study, we used mouse intestinal tumor–derived organoids that carried Apc, Kras, and Tgfbr2 mutations together with Trp53R270H or Trp53−/− mutation to identify a tumor-promoting mechanism of mutant p53 and interaction between mutant p53-stabilized and -destabilized cells. Through the organoid experiments, we found that mutant p53R270H–expressing cells transactivate Wnt/β-catenin signaling of co-cultured Trp53−/− cancer cells through induction of the COX-2/prostaglandin E2 (PGE2) pathway. Accordingly, these results suggest that the COX-2/PGE2 pathway induced by stabilized mutant p53 activates Wnt/β-catenin signaling in neighboring p53-destabilized cells, which may contribute to malignant progression of tumor tissues.

Organoid lines and cell culture experiments

An AKTPR270H organoid was established from intestinal tumors of quadruple mutant ApcΔ716 (A), KrasG12D (K) Tgfbr2−/− (T), and Trp53R270H (P) mice (12). AKTPNull organoids were generated by the disruption of wild-type (WT) Trp53 in AKT triple-mutant cells using the p53 CRISPR/Cas9 plasmid (13). Trp53-disrupted cells were selected with 10 µmol/L nutlin-3 (Cayman Chemical). These organoid cells were cultured on two-dimensional dishes with Advanced DMEM/F-12 medium (Gibco) supplemented with 10% FBS, 5 µmol/L anaplastic lymphoma kinase inhibitor (A83-01; Tocris Bioscience), 5 µmol/L GSK inhibitor (CHIR99021; Tocris Bioscience), and 10 µmol/L ROCK inhibitor (Y27632; Wako).

For suppression experiment of Trp53R270H in AKTPR270H cells, siRNA for Trp53 or control-siRNA (Silencer select predesigned siRNA, Ambion, Thermo Fisher Scientific) and siRNA for Trp53 (ON-TARGETplus Mouse Trp53 siRNA-SMART pool, Dharmacon, Horizon) were transfected by Lipofectamine RNAiMAX (Thermo Fisher Scientific). Knockdown efficiency was examined by immunoblotting. Wnt signaling activation was examined after 9 hours of siRNA transfection.

For co-culture experiments, 2 × 102 AKTPNull and AKTPR270H cells were mixed and co-cultured, or 4 × 102 AKTPNull or AKTPR270H cells were cultured (monoculture) on Ultra-Low Attachment culture plates (Corning). After 72 hours, the size of organoids was measured using ImageJ (NIH) on the photographs. All cell culture experiments were repeated three times unless otherwise noted.

Cell lines

HCT116 (#CCL-247, RRID: CVCL_0291), SW480 (#CCL-228, RRID: CVCL_0546), SW620 (#CCL-227, RRID: CVCL_0547), and Caco-2 (#HTB-37, RRID: CVCL_0025) were obtained from ATCC. LOVO (RCB1639, RRID: CVCL_0399) and COLO320 (RCB1193, RRID: CVCL_1989) were obtained from RIKEN BRC. Cells were maintained in high-glucose DMEM (Gibco) supplemented with 100 U/mL penicillin, 100 μg/mL streptomycin (Lonza), and 10% FBS (HyClone). Cells were maintained in the culture for maximum of 15 passages. Cells were monitored monthly for Mycoplasma contamination, and cell lines were authenticated using short tandem repeat analysis (Labcorp).

Reporter assays

For measurement of Wnt signaling activity, the cells were transfected with Super 8× TOPFlash (RRID: Addgene_12456) or Super 8× FOPFlash (RRID: Addgene_12457; Addgene) using Lipofectamine LTX (Thermo Fisher Scientific). After 24 hours of transfection, the luciferase activity was measured using a Dual-Luciferase Reporter Assay system (Promega). For measurement of NF-κB activation, the cells were transfected with a pNL3.2.NF-κB-RE vector (Promega) using Lipofectamine LTX (Thermo Fisher Scientific). The luciferase activity was measured using a Nano-Glo Dual-Luciferase Reporter Assay System (Promega).

For measurement of Wnt signaling activity in co-culture experiment, AKTPNull cells were transfected with Super 8× TOPFlash (RRID: Addgene_12456) or Super 8× FOPFlash (RRID: Addgene_12457). At 24 hours after transfection, the culture medium was changed, and then AKTPNull cells were co-cultured with AKTPR270H cells. After 24 hours of co-culture, the luciferase activity was measured.

Inhibitors

For COX-2 inhibition, cells were cultured with 5 µmol/L celecoxib (Selleck Chemicals). For EP2 and EP4 inhibition, cells were cultured with 100 nmol/L PF-04418948 (Selleck Chemicals) or 10 µmol/L RQ-15986/CJ-042794 (AskAt Inc.), respectively (19). For PGE2 stimulation, 1 µmol/L PGE2 (Cayman Chemical) was added at 9 hours after transfection of the reporter vector.

TOP-Venus reporter and EdU labeling experiments

Venus cDNA was replaced with luciferase cDNA of the TOPFlash vector (Upstate) to generate TOP-Venus and transfected to AKTPNull cells using Lipofectamine LTX (Thermo Fisher Scientific). Transfectant TOP-Venus AKTPNull cells were selected with 200 to 500 ng/mL puromycin. A total of 2 × 102 TOP-Venus AKTPNull and AKTPR270H cells were mixed and co-cultured, or 4 × 102 TOP-Venus AKTPNull cells were monocultured on Ultra-Low Attachment culture plates (Corning). After 72 hours of culture, spheroids were picked up and embedded in Matrigel (Corning), and after another 72 hours of culture, spheroids were immunostained.

For 5-ethynyl-2'-deoxyuridine (EdU) labeling, we used 2 × 102 AKTPNull cells and Venus-labeled AKTPR270H cells that were previously described (20). These cells were co-cultured on Ultra-Low Attachment culture plates (Corning). After 48 hours of culture, cells were treated with 10 mmol/L EdU for 90 minutes. After fixation with 4% paraformaldehyde, organoids were embedded in Matrigel, and EdU-labeled AKTPNull cells were detected using Click-iT EdU cell proliferation assays (Thermo Fisher Scientific) and a confocal microscope (TCS SP8, Leica Microsystems). The numbers of EdU-labeled cells were counted at three different horizontal levels of each organoid, and the mean EdU labeling index was calculated.

Ctnnb1 short hairpin RNA experiments

Short hairpin RNA (shRNA) lentiviral vectors for Ctnnb1 in pLKO.1-puro (MISSION shRNA, Sigma-Aldrich) were co-transfected to HEK293T cells with pCMV-VSV-G (Addgene, #8485, RRID: Addgene_8454) and pCMV-dR8.2 dvpr (Addgene, #8455, RRID: Addgene_8455) using PEI MAX (Polysciences) for packaging. Lentiviruses were infected to AKTP cells using polybrene (Sigma-Aldrich), and shRNA-expressing cells were selected with puromycin. Cell growth of shRNA lentivirus–infected cells was examined using CellTiter-Glo 2.0 Cell Viability Assay (Promega).

Animal experiments

C57BL/6 mice (male; 6 weeks of age) were purchased (The Jackson Laboratory). AKTPR270H or AKTPNull organoids were treated with trypsin, and 3 to 5 × 105 cells were injected into the mouse spleen. Liver metastatic tumors were examined at 4 weeks after the injection (n = 3) as follows. All animal experiments were carried out according to the protocol approved by the Committee on Animal Experimentation of Kanazawa University, Japan.

Histology and IHC

Whole-liver tissues were collected from C57BL/6 mice. Human colorectal cancer samples were obtained from patients who underwent surgical resection at Ishikawa Prefectural Central Hospital, Japan. The experiments using human tissues were approved by the Human Genome/Gene Analysis Research Ethics Committee of Kanazawa University at #2021-002-603, and written informed consent was obtained from the patients. The tissues were fixated in 4% paraformaldehyde, paraffin-embedded, and sectioned at 4 μm thickness. For IHC, antibodies against E-cadherin (R&D Systems, Cat. #AF748, RRID: AB_355568), mouse p53 (Leica Biosystems, CM5, Cat. #NCL-p53-CM5p, RRID: AB_563933; Cell Signaling Technology, 1C12, Cat. #2524, RRID: AB_331743), human p53 (Santa Cruz Biotechnology, DO7, Cat. #47698, RRID: AB_628083), β-catenin (Cell Signaling Technology, D10A8, #8480, RRID: AB_11127855), and COX-2 (Cayman Chemicals, polyclonal antibody, aa584-598, RRID: AB_10079419) were used as primary antibodies. Staining signals were visualized using the VECTASTAIN Elite Kit (Vector Laboratories). For fluorescence IHC, Alexa Fluor 594– or Alexa Fluor 488–conjugated antibodies (Molecular Probes, Fluor 594, #A21207, RRID: AB_141637; Fluor 488, #A21206, RRID: AB_2535792) were used as the second antibody. The immunostained specimens were examined using a confocal microscope (Leica TCS SP8, Leica Microsystems). The number of p53-stabilized and β-catenin nuclear-accumulated cells were counted in three to four independent microscopic fields, and the mean number per tumor was calculated.

Organoid immunocytochemistry

The organoids were immunostained using antibodies against mouse p53 (Leica Biosystems, CM5, Cat. #NCL-p53-CM5p, RRID: AB_563933; Cell Signaling Technology, 1C12, Cat. #2524, RRID: AB_331743) and COX-2 (Cayman Chemicals, polyclonal antibody, aa584-598, RRID: AB_10079419) and Alexa Fluor 594– or Alexa Fluor 488–conjugated antibodies (Molecular Probes, Fluor 594, RRID: AB_141637; Fluor 488, RRID: AB_2535792) for the secondary antibody. The immunostained organoids were examined using a confocal microscope (Leica TCS SP8, Leica Microsystems).

Immunoblotting

Organoid cells were lysed in lysis buffer, and protein samples were separated in 10% SDS-PAGE. Antibodies against mouse p53 (Cell Signaling Technology, 1C12, Cat. #2524, RRID: AB_331743) and COX-2 (Cayman Chemicals, polyclonal antibody, aa584-598, RRID: AB_10079419) were used. Antibodies for GAPDH (FUJIFILM Wako, Cat. #016-25523, RRID: AB_2814991) and β-actin (Wako, 6D1, #010-27841, RRID: AB_2858279) were used as the internal control. The enhanced chemiluminescence detection system (GE Healthcare) was used to detect the signals.

Trp53 copy-number analysis

Trp53 genotyping in AKTPR270H cell lines was performed using a TaqMan SNP Genotyping assay (Thermo Fisher Scientific). The custom TaqMan probes for WT Trp53 (VIC dye) and mutant Trp53R270H (FAM dye) were designed in exon 8 including the R270 codon of mouse Trp53. Sequences of TaqMan probes are as follows:

Forward primer: CCG​GAT​AGT​GGG​AAC​CTT​CTG

Reverse primer: TCT​TCT​GTA​CGG​CGG​TCT​CT

The TaqMan probe to detect WT Trp53 (VIC dye) is CTT​TGA​GGT​TCGTGT​TTG​T, and the TaqMan probe to detect mutant Trp53R270H (FAM dye) is TTT​GAG​GTT​CATGT​TTG​T. The PCR results were analyzed using Allele discrimination in Agilent AriaMx (Agilent Technologies).

Public database for RNA sequencing

For RNA sequencing (RNA-seq) data of AKTPR270H and AKTPNull organoids and AKTFPR270H/LOH and AKTFPR270H/+ organoids, we used public datasets of the DNA Data Bank of Japan (accessions #PRJDB5631 and #PRJDB15570).

Statistical analysis

The data were analyzed using two-sided unpaired t tests unless otherwise mentioned and presented as the means ± SD. Statistical analyses for Fig. 1A, B, and G were performed using a one-way ANOVA test. The Fisher exact test was used for Fig. 1G. A value of P < 0.05 was considered statistically significant. GraphPad Prism 9 (GraphPad) was used for statistical analyses. All data were reproduced with at least two independent experiments and at least three biological replicates.

Figure 1

Promotion of Wnt/β-catenin signaling in missense-type p53-mutant colon tumors. A, Luciferase activities of NF-κΒ reporter assay are shown as a bar graph (mean ± SD). B, Luciferase activities of Wnt/β-catenin reporter assay (TOPFlash) are shown as a bar graph (mean ± SD). Three independent AKTPNull and AKTPR270H organoid lines for each were used in the reporter assays (A and B). Statistical analysis was performed using the one-way ANOVA test (A and B). C, Luciferase activities of Wnt/β-catenin reporter assay for control-transfected and Trp53 siRNA–transfected AKTPR270H organoid cells are shown as a bar graph (mean ± SD). The immunoblotting result for p53 is shown. GAPDH was used as the internal control. Each dot in AC graphs indicates an independent experiment. P values are indicated. D, Representative confocal microscopy images of fluorescence immunocytochemistry for β-catenin (green) and E-cadherin (red) in AKTPNull and AKTPR270H organoids. Arrowheads indicate cells with β-catenin stabilization. Bars, 25 μm. E, Representative photographs of fluorescence IHC for β-catenin (green) and E-cadherin (red) of AKTPNull and AKTPR270H liver metastatic tumors. Insets show enlarged images. Bars, 100 μm. Note that β-catenin accumulation was predominantly found in AKTPR270H tumors. F, Immunoblotting results for p53 in colorectal cancer cell lines. β-Actin was used as the internal control. G, Luciferase activities of Wnt/β-catenin reporter assay (TOPFlash) for colorectal cancer cell lines are shown as a bar graph (mean ± SD). Each dot indicates an independent experiment. The P value is provided. H, Representative photographs of immunostaining for p53 (left) and β-catenin (right) of p53-positive and β-catenin nuclear-accumulated (top) and p53-negative and β-catenin not nuclear–accumulated (bottom) human colorectal cancer. Insets show enlarged images of the boxed area. Arrowheads indicate p53 stabilization (left) and β-catenin nuclear accumulation (right). Bars, 200 μm. I, Ratio of β-catenin nuclear accumulation in p53-positive (+) and p53-negative (−) colorectal cancer scored using the IHC results in H. The P value is indicated.

Figure 1

Promotion of Wnt/β-catenin signaling in missense-type p53-mutant colon tumors. A, Luciferase activities of NF-κΒ reporter assay are shown as a bar graph (mean ± SD). B, Luciferase activities of Wnt/β-catenin reporter assay (TOPFlash) are shown as a bar graph (mean ± SD). Three independent AKTPNull and AKTPR270H organoid lines for each were used in the reporter assays (A and B). Statistical analysis was performed using the one-way ANOVA test (A and B). C, Luciferase activities of Wnt/β-catenin reporter assay for control-transfected and Trp53 siRNA–transfected AKTPR270H organoid cells are shown as a bar graph (mean ± SD). The immunoblotting result for p53 is shown. GAPDH was used as the internal control. Each dot in AC graphs indicates an independent experiment. P values are indicated. D, Representative confocal microscopy images of fluorescence immunocytochemistry for β-catenin (green) and E-cadherin (red) in AKTPNull and AKTPR270H organoids. Arrowheads indicate cells with β-catenin stabilization. Bars, 25 μm. E, Representative photographs of fluorescence IHC for β-catenin (green) and E-cadherin (red) of AKTPNull and AKTPR270H liver metastatic tumors. Insets show enlarged images. Bars, 100 μm. Note that β-catenin accumulation was predominantly found in AKTPR270H tumors. F, Immunoblotting results for p53 in colorectal cancer cell lines. β-Actin was used as the internal control. G, Luciferase activities of Wnt/β-catenin reporter assay (TOPFlash) for colorectal cancer cell lines are shown as a bar graph (mean ± SD). Each dot indicates an independent experiment. The P value is provided. H, Representative photographs of immunostaining for p53 (left) and β-catenin (right) of p53-positive and β-catenin nuclear-accumulated (top) and p53-negative and β-catenin not nuclear–accumulated (bottom) human colorectal cancer. Insets show enlarged images of the boxed area. Arrowheads indicate p53 stabilization (left) and β-catenin nuclear accumulation (right). Bars, 200 μm. I, Ratio of β-catenin nuclear accumulation in p53-positive (+) and p53-negative (−) colorectal cancer scored using the IHC results in H. The P value is indicated.

Close modal

Data availability

The data generated in this study are available upon request from the corresponding authors.

Missense mutant p53 promotes Wnt/β-catenin signaling in intestinal tumors

We previously established an intestinal tumor–derived organoid, AKTP, in which ApcΔ716, KrasG12D, Tgfbr2−/−, and Trp53R270H mutations were introduced (12). To investigate the role of oncogenic function of mutant p53R270H, we further constructed Trp53−/− AKTP organoid by disrupting Trp53 using the CRISPR/Cas9 (13). Here, we named AKTP organoid lines with Trp53R270H or Trp53−/− mutations as AKTPR270H or AKTPNull, respectively. We used three independently established AKTPR270H lines in this study, and genomic PCR analysis revealed that all lines had two copies of the Trp53R270H mutant alleles, indicating the loss of WT Trp53 by copy-number neutral loss of heterozygosity (LOH; Supplementary Fig. S1).

We have previously shown that the inflammatory pathway and Wnt/β-catenin signaling are activated in the ApcΔ716Trp53R270H double-mutant intestinal tumor–derived organoids compared with that in the simple ApcΔ716-mutant tumor organoids (11). Therefore, we examined the activation levels of NF-κB in the inflammatory pathway and Wnt/β-catenin signaling in the AKTP cells using reporter assays. We found that NF-κB activation was at the similar levels between AKTPR270H and AKTPNull organoid cells (Fig. 1A). In contrast, the levels of Wnt/β-catenin signaling were significantly higher in all AKTPR270H compared with AKTPNull organoid lines (Fig. 1B). Moreover, transfection of siRNA against Trp53 mRNA into AKTPR270H cells significantly suppressed Wnt signaling activation with a decrease in p53 expression levels (Fig. 1C). These results indicate that mutant p53R270H promotes Wnt/β-catenin signaling activation.

We confirmed p53R270H-induced Wnt signaling activation by other methods. First, we performed an Ingenuity Pathway Analysis using RNA-seq data of AKTPR270H and AKTPNull organoids (13) and found that the Wnt signaling pathways (CTNNB1 and TCF7L2 pathways) were significantly activated in AKTPR270H (Supplementary Fig. S2). In addition, immunostaining of cultured AKTPR270H organoids and histologic sections of liver metastasis of AKTPR270H organoid cells showed β-catenin stabilization and nuclear accumulation, which were rarely found in AKTPNull organoid cultures and metastatic tissues (Fig. 1D and E).

We further examined the relationships between mutant p53 and Wnt/β-catenin signaling using TP53 WT and TP53-mutant human colorectal cancer cell lines (21). As expected, stabilized p53 was detected in the missense-type TP53-mutant SW480, SW620, and COLO320 cells but not in TP53 WT HCT116 and LOVO cells (Fig. 1F). Notably, Wnt/β-catenin signaling is significantly higher in TP53-mutant cells than in TP53 WT cells (Fig. 1G). However, the Wnt/β-catenin signaling is also high in the TP53-null mutant Caco-2 cells (22). We further examined the primary colorectal cancer tissues by IHC using tissue microarray. In the p53 staining–positive colorectal cancer group, β-catenin nuclear accumulation was found in 39% of tissues, whereas the ratio was significantly decreased to 17% in p53-negative colorectal cancer tissues (Fig. 1H and I). These results collectively suggest that the Wnt signaling activation in human colorectal cancer may involve, though not exclusively, the presence of missense-type mutant p53, similar to that found in mouse intestinal tumors.

Missense mutant p53 promotes Wnt/β-catenin signaling in a non–cell autonomous mechanism

It has been reported that there is heterogeneity in p53 stabilization among TP53-mutant cells (18). We also found the heterogeneity in p53 stabilization in both human colorectal cancer and mouse AKTPR270H liver metastasis by IHC (Fig. 2A). In p53-positive human colorectal cancer, p53 stabilization was found in approximately 80% and 72% of tumor cells in the primary and liver metastases, respectively (Fig. 2A and B). Similarly, in AKTPR270H metastatic tumors, p53 stabilization was found in approximately 50% to 66% of two different cell lines in average. Specifically, not all cancer cells with p53 mutations have stabilized p53 protein, and there is intratumor heterogeneity in p53 stabilization. As stabilization of mutant p53 is thought to be a prerequisite for the oncogenic function, these results suggest that there is intratumor heterogeneity in the oncogenic function of mutant p53 in the p53-mutant tumors.

Figure 2

Heterogeneity of p53 and β-catenin stabilization in tumor cells. A, Representative photographs of IHC for p53 of human colon cancer (left) and AKTPR270H liver metastatic tumor (right). Insets show enlarged images. Arrowheads indicate p53-negative (destabilized) tumor cells. Bars, 100 μm. B, Ratio of p53-positive (+) cells in primary and metastatic colorectal cancer (CRC; squares) and AKTPR270H liver metastatic tumors of two independent lines (circles). Each dot indicates the results of the independent microscopic fields. C, Representative photographs of fluorescence IHC for p53 (green, left), β-catenin (red, center), and merged images (right) of AKTPR270H liver metastasis (top) and human colon cancer (bottom) Insets show enlarged images. Arrowheads and arrows indicate double-positive and single-positive cells, respectively. Asterisks indicate double-negative cells. Bars, 100 μm. D, Relative proportions (%) of p53-positive, β-catenin–positive, and double-negative cells in the liver metastasis of AKTPR270H cells (left), AKTPNull cells (right), and human colorectal cancer (center) are shown as bar graphs. Three biologically independent data for each are shown.

Figure 2

Heterogeneity of p53 and β-catenin stabilization in tumor cells. A, Representative photographs of IHC for p53 of human colon cancer (left) and AKTPR270H liver metastatic tumor (right). Insets show enlarged images. Arrowheads indicate p53-negative (destabilized) tumor cells. Bars, 100 μm. B, Ratio of p53-positive (+) cells in primary and metastatic colorectal cancer (CRC; squares) and AKTPR270H liver metastatic tumors of two independent lines (circles). Each dot indicates the results of the independent microscopic fields. C, Representative photographs of fluorescence IHC for p53 (green, left), β-catenin (red, center), and merged images (right) of AKTPR270H liver metastasis (top) and human colon cancer (bottom) Insets show enlarged images. Arrowheads and arrows indicate double-positive and single-positive cells, respectively. Asterisks indicate double-negative cells. Bars, 100 μm. D, Relative proportions (%) of p53-positive, β-catenin–positive, and double-negative cells in the liver metastasis of AKTPR270H cells (left), AKTPNull cells (right), and human colorectal cancer (center) are shown as bar graphs. Three biologically independent data for each are shown.

Close modal

We therefore examined whether the stabilization of p53 and β-catenin occurs simultaneously in the same cells or not by double-fluorescence immunostaining. In the AKTPR270H metastatic tumors, we found double-positive cells for β-catenin and p53 as expected; however, we also found certain populations of single-positive cells for β-catenin or p53 [Fig. 2C (top) and D (left)]. Similar immunostaining patterns, namely, double-positive and single-positive cells for p53 and β-catenin were also found in human colorectal cancer tissues [Fig. 2C (bottom) and D (center)]. In contrast, nuclear accumulation of β-catenin was rarely detected in AKTPNull metastatic tumors [Fig. 2D (right)]. Taken together, these results suggest that mutant p53 activates Wnt/β-catenin signaling not only in the p53-stabilized cells but also in the p53-destabilized cells possibly through a non–cell autonomous mechanism.

Mutant p53–induced COX-2/PGE2 pathway transactivates Wnt/β-catenin signaling in p53-null/destabilized cells

To investigate a possible mechanism for Wnt signaling activation in cancer cells in which the mutant p53 protein is destabilized and undetectable, we used AKTPNull as a model for these p53-destabilized cells in the p53-mutant tumors. After co-culture with AKTPR270H cells, we examined the Wnt signaling activity in AKTPNull cells using TOPFlash reporter assay (Fig. 3A). AKTPNull cells (AKTPNull-bottom cells in Fig. 3B) carry an Apc mutation; thus, reporter assay showed basal activation of Wnt signaling (Fig. 3B). Importantly, the TOPFlash level in AKTPNull-bottom cells was significantly increased when co-cultured with AKTPR270H-top cells compared with that in the co-culture with AKTPNull-top cells or the monoculture control, suggesting a paracrine mechanism for Wnt activation in AKTPNull cells by AKTPR270H cells (Fig. 3B).

Figure 3

Activation of Wnt/β-catenin signaling in p53-negative cells through the COX-2/PGE2 pathway. A, Schematic drawing of Wnt/β-catenin reporter assays (TOPFlash) of co-cultured AKTPNull-bottom cells with AKTPR270H-top cells or AKTPNull-top cells. B, Luciferase activities of Wnt reporter assays (TOPFlash) of AKTPNull-bottom cells co-cultured with AKTPNull-top cells or AKTPR270H-top cells are shown as a bar graph (mean ± SD). Each dot represents an independent experiment. The P value is provided. C, RNA-seq data for Ptgs2 in three independent AKTFPR270H/+ and AKTFPR270H/LOH organoid lines. fragments per kilobase of exon per million reads mapped, the mean fold change, and P value are provided. D, Immunoblotting results for p53 and COX-2 in three independent AKTPNull and AKTPR270H lines. GAPDH was used as the internal control. E, Representative fluorescence immunocytochemistry images for p53 (green) and COX-2 (red) of AKTPNull (left) and AKTPR270H (right) organoids. F, Representative image of fluorescence IHC for p53 (green) and COX-2 (red) of AKTPR270H liver metastatic tumor. Insets in E and F show enlarged images. Bars in E and F, 100 μm. G, Representative images of fluorescence IHC for COX-2 (red) and F4/80 (macrophages, green) or αSMA (myofibroblasts, green). Merged images are shown (right). Bars, 50 μm. H, Luciferase activities of Wnt reporter assays (TOPFlash) of AKTPNull-bottom cells in co-cultured AKTPNull-top or AKTPR270H-top cells or control AKTPNull-bottom monoculture cells in the presence or absence of COX-2 inhibitor, celecoxib, are shown as a bar graph (mean ± SD). Each dot represents an independent experiment. The P value is indicated. ns, not significant. I and J, Luciferase activities of Wnt/β-catenin reporter assays in AKTPNull cells treated with PGE2 (I) or AKTPNull cells co-cultured with AKTPR270H treated with EP2/EP4 inhibitors (J) are shown as bar graphs (mean ± SD). Data from two independent AKTPNull lines (#1 and #2) are shown. Each dot indicates an independent experiment. P values are provided.

Figure 3

Activation of Wnt/β-catenin signaling in p53-negative cells through the COX-2/PGE2 pathway. A, Schematic drawing of Wnt/β-catenin reporter assays (TOPFlash) of co-cultured AKTPNull-bottom cells with AKTPR270H-top cells or AKTPNull-top cells. B, Luciferase activities of Wnt reporter assays (TOPFlash) of AKTPNull-bottom cells co-cultured with AKTPNull-top cells or AKTPR270H-top cells are shown as a bar graph (mean ± SD). Each dot represents an independent experiment. The P value is provided. C, RNA-seq data for Ptgs2 in three independent AKTFPR270H/+ and AKTFPR270H/LOH organoid lines. fragments per kilobase of exon per million reads mapped, the mean fold change, and P value are provided. D, Immunoblotting results for p53 and COX-2 in three independent AKTPNull and AKTPR270H lines. GAPDH was used as the internal control. E, Representative fluorescence immunocytochemistry images for p53 (green) and COX-2 (red) of AKTPNull (left) and AKTPR270H (right) organoids. F, Representative image of fluorescence IHC for p53 (green) and COX-2 (red) of AKTPR270H liver metastatic tumor. Insets in E and F show enlarged images. Bars in E and F, 100 μm. G, Representative images of fluorescence IHC for COX-2 (red) and F4/80 (macrophages, green) or αSMA (myofibroblasts, green). Merged images are shown (right). Bars, 50 μm. H, Luciferase activities of Wnt reporter assays (TOPFlash) of AKTPNull-bottom cells in co-cultured AKTPNull-top or AKTPR270H-top cells or control AKTPNull-bottom monoculture cells in the presence or absence of COX-2 inhibitor, celecoxib, are shown as a bar graph (mean ± SD). Each dot represents an independent experiment. The P value is indicated. ns, not significant. I and J, Luciferase activities of Wnt/β-catenin reporter assays in AKTPNull cells treated with PGE2 (I) or AKTPNull cells co-cultured with AKTPR270H treated with EP2/EP4 inhibitors (J) are shown as bar graphs (mean ± SD). Data from two independent AKTPNull lines (#1 and #2) are shown. Each dot indicates an independent experiment. P values are provided.

Close modal

We previously performed RNA-seq of AKTFPR270H/LOH (Trp53R270H with loss of WT Trp53 by LOH) and AKTFPR270H/+ (Trp53R270H/+ heterozygous) organoids and found that the expression level of Ptgs2 encoding COX-2 was significantly upregulated in AKTFPR270H/LOH cells (Fig. 3C; ref. 23). As the mutant p53 stabilization is significantly increased by loss of WT p53 by LOH (24, 25), the expression data suggest that COX-2 upregulation is caused by stabilized mutant p53 function. As expected, we confirmed increased levels of COX-2 expression in all AKTPR270H organoid lines by immunoblotting compared with those in AKTPNull cells (Fig. 3D). Furthermore, we confirmed COX-2 expression in AKTPR270H cells by immunostaining, although the levels of COX-2 expression seemed to vary (Fig. 3E). On the other hand, COX-2 was not detected in AKTPNull cells. Liver metastatic tumors of AKTPR270H cells also showed COX-2 expression in p53-stabilized tumor cells (Fig. 3F). We also found COX-2–expressing macrophages but not α-smooth muscle actin (α-SMA)-expressing myofibroblasts in the tumor stroma of the liver metastatic tumors of AKTPR270H cells (Fig. 3G). COX-2 is a rate-limiting enzyme for prostaglandin biosynthesis, and several studies have shown an interaction between the COX-2/PGE2 pathway and Wnt/β-catenin signaling activation (2628). Importantly, treatment with celecoxib, a selective COX-2 inhibitor, dramatically suppressed Wnt/β-catenin signaling activation in AKTPNull-bottom cells co-cultured with AKTPR270H-top cells (Fig. 3H). In contrast, direct stimulation of AKTPNull cells with PGE2 significantly increased Wnt signaling activity (Fig. 3I). Furthermore, suppression of PGE2 signaling by treating cells with the EP2/EP4 receptor inhibitor suppressed Wnt signaling activation in AKTPNull-bottom cells that were co-cultured with AKTPR270H-top cells (Fig. 3J). The similar results were obtained in the two independently established AKTPNull organoid lines (Fig. 3I and J). Accordingly, these results indicate that AKTPR270H cells transactivate Wnt/β-catenin signaling in AKTPNull cells through induction of the COX-2/PGE2 pathway, and PGE2 signaling through the EP2 and/or EP4 receptor is vital for Wnt activation in AKTPNull cells. These results suggest that in p53-mutant tumors, cancer cells with stabilized mutant p53 activate Wnt/β-catenin signaling in neighboring p53-destabilized cells through the COX-2/PGE2 pathway. It is also possible that macrophage-secreted PGE2 contributes to Wnt/β-catenin signaling activation in p53-destabilized cells in the liver metastatic tumors, although it remains to be further investigated.

Interestingly, Wnt signaling activity in AKTPR270H cells was not affected by treatment with the COX-2 inhibitor or EP2/EP4 inhibitor (Supplementary Fig. S3). We previously showed that missense p53 mutations resulted in Wnt/β-catenin pathway activation with increased promoter accessibility (11). It is possible that Wnt/β-catenin signaling is activated in the p53-stabilized AKTPR270H cells by the COX-2/PGE2 pathway–independent mechanism.

Mutant p53 promotes proliferation of p53-null/destabilized cells

We further examined the Wnt signaling activity by imaging analysis using TOP-Venus reporter–transfected AKTPNull cells that express Venus in response to Wnt/β-catenin signaling. Notably, the number of Venus-positive AKTPNull cells increased significantly when co-cultured with AKTPR270H cells compared with that of control AKTPNull cell monocultures (Fig. 4A and B). Similar results were obtained in the two independent AKTPNull organoid lines. These results confirmed that AKTPR270H cells transactivate the Wnt signaling activity of AKTPNull cells.

Figure 4

Promotion of cell proliferation of p53-negative tumor cells co-cultured with p53-mutant cells. A, Representative fluorescent immunocytochemistry images for p53 (red) and TOP-Venus–positive (Wnt/β-catenin–activated) cells (green) of monocultured AKTPNull (left) and co-cultured AKTPNull with AKTPR270H (right) organoids. Insets show enlarged images. Bars, 50 μm. B, Ratios of TOP-Venus–positive (Wnt/β-catenin–activated) cells per microscopic field. Each dot indicates the result in the independent field. Data from two independent AKTPNull lines (#1 and #2) are shown. P values are provided. C, Representative images of fluorescent immunocytochemistry for EdU-labeled cells (red) of monocultured AKTPNull (left) and co-cultured AKTPNull with Venus-labeled AKTPR270H organoids (green; right). Bars, 25 μm. D, Ratios of EdU-labeled cells to total monocultured AKTPNull cells or co-cultured AKTPNull cells with AKTPR270H. Each dot indicates the results in the independent microscopic field. Data from two independent AKTPNull lines (#1 and #2) are shown. P values are indicated. E, Representative photographs of monocultured AKTPNull (left) and AKTPR270H organoids (right) and co-cultured AKTPNull and AKTPR270H organoids (center). Results from two independent AKTPNull lines (#1 and #2) are shown. Bars, 200 μm. F, Relative organoid sizes of monocultured AKTPNull and AKTPR270H cells and co-cultured AKTPNull and AKTPR270H cells. Each dot indicates the independent organoid size. Results of two independent AKTPNull lines (#1 and #2) are shown. P values are shown. ns, not significant. G, Immunoblotting results for total β-catenin in two lines of sh-β-catenin–transfected AKTPNull cells and sh-control–transfected cells. GAPDH was used as the internal control. H, Relative cell growth rate of sh-β-catenin–transfected AKTPNull cells (#1 and #2) with sh-control–transfected cells. The results of two independent AKTPR270H lines are shown (G and H). Asterisks and daggers, P < 0.05 vs. sh-control.

Figure 4

Promotion of cell proliferation of p53-negative tumor cells co-cultured with p53-mutant cells. A, Representative fluorescent immunocytochemistry images for p53 (red) and TOP-Venus–positive (Wnt/β-catenin–activated) cells (green) of monocultured AKTPNull (left) and co-cultured AKTPNull with AKTPR270H (right) organoids. Insets show enlarged images. Bars, 50 μm. B, Ratios of TOP-Venus–positive (Wnt/β-catenin–activated) cells per microscopic field. Each dot indicates the result in the independent field. Data from two independent AKTPNull lines (#1 and #2) are shown. P values are provided. C, Representative images of fluorescent immunocytochemistry for EdU-labeled cells (red) of monocultured AKTPNull (left) and co-cultured AKTPNull with Venus-labeled AKTPR270H organoids (green; right). Bars, 25 μm. D, Ratios of EdU-labeled cells to total monocultured AKTPNull cells or co-cultured AKTPNull cells with AKTPR270H. Each dot indicates the results in the independent microscopic field. Data from two independent AKTPNull lines (#1 and #2) are shown. P values are indicated. E, Representative photographs of monocultured AKTPNull (left) and AKTPR270H organoids (right) and co-cultured AKTPNull and AKTPR270H organoids (center). Results from two independent AKTPNull lines (#1 and #2) are shown. Bars, 200 μm. F, Relative organoid sizes of monocultured AKTPNull and AKTPR270H cells and co-cultured AKTPNull and AKTPR270H cells. Each dot indicates the independent organoid size. Results of two independent AKTPNull lines (#1 and #2) are shown. P values are shown. ns, not significant. G, Immunoblotting results for total β-catenin in two lines of sh-β-catenin–transfected AKTPNull cells and sh-control–transfected cells. GAPDH was used as the internal control. H, Relative cell growth rate of sh-β-catenin–transfected AKTPNull cells (#1 and #2) with sh-control–transfected cells. The results of two independent AKTPR270H lines are shown (G and H). Asterisks and daggers, P < 0.05 vs. sh-control.

Close modal

We next analyzed the cell proliferation rate of monocultured AKTPNull cells and co-cultured AKTPNull cells with AKTPR270H-Venus cells by EdU labeling assay. Fluorescent immunostaining of the organoids revealed that the number of EdU-labeled AKTPNull cells increased significantly when co-cultured with AKTPR270H cells compared with that of monocultured cells (Fig. 4C and D). The similar results were obtained in the two independent AKTPNull organoid lines.

The size of organoids developed in the Matrigel was significantly larger in AKTPR270H compared with AKTPNull cells, indicating a higher proliferation rate of AKTPR270H cells (Fig. 4E and F). Interestingly, the size of mixed organoids of AKTPNull and AKTPR270H cells was intermediate; however, it tended to be of similar size to AKTPR270H cells at least in one AKTPNull organoid line [Fig. 4F (left)]. These results indicate that AKTPR270H cells promote the proliferation of AKTPNull cells.

Finally, we examined a role of Wnt/β-catenin signaling in AKTPR270H cell proliferation. Notably, partial suppression of β-catenin expression by shRNA transfection significantly suppressed proliferation in the independent AKTPR270H organoid lines (Fig. 4G and H). Accordingly, it is possible that transactivation of Wnt/β-catenin signaling in AKTPNull cells may contribute to increased proliferation.

Recently, it has been reported that removal of mutant p53 genes in human cancer cells did not affect the proliferation and survival of cancer cells including colorectal cancer (29). On the other hand, we previously showed that mutant p53 enhances the clonal expansion and colony formation ability of the single cell–dissociated tumor cells, suggesting increased stemness of the p53-mutant cancer cells (13). As is generally thought, Wnt signaling is essential for the maintenance of tissue and tumor stem cells (30). Therefore, it is possible that mutant p53 plays an important role in colonization of the disseminated tumor cells in the liver through Wnt/β-catenin activation rather than growth of established tumors. Consistently, it has been reported that p53 mutations induce stem cell properties in cancer cells (9, 10).

Although the precise molecular mechanisms underlying the gain-of-function activity by mutant p53 remain unclear, previous studies have indicated the potential involvement of epigenetic modification by mutant p53, which resulted in significant transcriptome changes, including VEGFR2 upregulation (16, 17). Additionally, our recent findings suggest that the upregulation of the transcription factor Hmga2 by mutant p53 contributes to partial epithelial-to-mesenchymal transition and cluster migration of cancer cells (23). However, these mutant p53–induced oncogenic functions are considered cell intrinsic mechanisms. Namely, it was thought that only mutant p53–stabilized cells carry malignant phenotypes. However, in the present study, we demonstrate for the first time that mutant p53–stabilized cells may activate Wnt/β-catenin signaling of p53-destabilized cells by a paracrine mechanism. Therefore, it seems that mutant p53 enhances Wnt/β-catenin signaling of whole-tumor tissues through cell-autonomous and non–cell autonomous mechanisms.

In this study, we elucidated that mutant p53 induces COX-2 expression, and the downstream product PGE2 activates EP2/EP4 receptor signaling in surrounding p53-destabilized cells, leading to the activation of Wnt/β-catenin signaling (Fig. 5). Previously, we showed that the missense-type p53 mutation in the intestinal tumor cells leads to a dramatic transcriptomic shift through an increase in promoter accessibility (11), and inflammatory pathways, including TNFα/NF-κB, are significantly activated in p53-mutant AKTPR270H cells (13). TNFα has been shown to induce COX-2 expression via NF-κB activation (31). Accordingly, it is possible that the TNFα/NF-κB pathway may be a potential driver of COX-2 induction in AKTPR270H cells.

Figure 5

Schematic drawing of mutant p53–induced transactivation of Wnt/β-catenin signaling in p53-deficient cells. Double-fluorescent IHC for p53 and β-catenin of liver metastatic tumors of AKTPR270H cells (left) and sketch of tumor cells from the photograph with the Wnt activation mechanism are shown (right).

Figure 5

Schematic drawing of mutant p53–induced transactivation of Wnt/β-catenin signaling in p53-deficient cells. Double-fluorescent IHC for p53 and β-catenin of liver metastatic tumors of AKTPR270H cells (left) and sketch of tumor cells from the photograph with the Wnt activation mechanism are shown (right).

Close modal

The COX-2/PGE2 pathway has been reported to activate Wnt signaling in colorectal cancer cells (26, 27), and the interaction between PGE2 and Wnt signaling has been shown to be important in the regulation of hematopoietic stem cells and organ regeneration processes (28). These results suggest that COX-2/PGE2-induced Wnt activation is thought to play an important role in the proliferation of colorectal cancers harboring p53 mutations. However, we could not elucidate the mechanism of PGE2-induced Wnt/β-catenin pathway activation in the present study. Both AKTPR270H and AKTPNull cells lack the WT Apc gene, resulting in constitutive stabilization of β-catenin. Accordingly, it is possible that PGE2 signaling may be involved in the regulation of nuclear localization or the degradation process of β-catenin, although it remains to be examined. In this study, we found that infiltrating macrophages in the metastatic tumor stroma also express COX-2. It is thus possible that macrophage-derived PGE2 may contribute to Wnt signaling activation in the p53-destabilized cancer cells, although this requires further investigation.

Accumulating evidence from genetic studies has revealed that the COX-2/PGE2 pathway plays a critical role in colorectal cancer development by generating an inflammatory microenvironment (32, 33). In addition, activation of the COX-2/PGE2 pathway in fibroblasts surrounding intestinal crypts has recently been reported to activate YAP via EP4 receptors on intestinal epithelial cells, thereby promoting tumorigenesis (34). These results suggest that activation of the COX-2/PGE2 pathway in mutant p53–stabilized cells may have significant effects on tumorigenesis possibly beyond activation of the Wnt/β-catenin pathway in the neighboring p53-destabilized cells. Accordingly, it is possible that the COX-2/PGE2 pathway is an effective therapeutic target against both p53-stabilized and -destabilized cells in TP53-mutant cancer.

M. Nakayama reports grants from the Ministry of Education, Culture, Sports, Science and Technology of Japan during the conduct of the study. M. Oshima reports grants from the Japan Agency for Medical Research and Development and Ministry of Education, Culture, Sports, Science and Technology of Japan during the conduct of the study. No disclosures were reported by the other authors.

M. Nakayama: Funding acquisition, investigation, writing–review and editing. H. Saito: Investigation. K. Murakami: Methodology. H. Oshima: Investigation. M. Oshima: Conceptualization, supervision, writing–original draft, writing–review and editing.

We thank Manami Watanabe and Ayako Tsuda for their technical assistance. This work was supported by Japan Agency for Medical Research and Development (22ck0106541h0003; 22gm4010012h0002; M. Oshima), and Grants-in-Aid for Scientific Research (A; 22H00454; M. Oshima and C; 20K07585 and 23K06631; M. Nakayama) from the Ministry of Education, Culture, Sports, Science and Technology of Japan.

Note: Supplementary data for this article are available at Cancer Research Communications Online (https://aacrjournals.org/cancerrescommun/).

1.
Kandoth
C
,
McLellan
MD
,
Vandin
F
,
Ye
K
,
Niu
B
,
Lu
C
, et al
.
Mutational landscape and significance across 12 major cancer types
.
Nature
2013
;
502
:
333
9
.
2.
Cancer Genome Atlas Network
.
Comprehensive molecular characterization of human colon and rectal cancer
.
Nature
2012
;
487
:
330
7
.
3.
Brannon
AR
,
Vakiani
E
,
Sylvester
BE
,
Scott
SN
,
McDermott
G
,
Shah
RH
, et al
.
Comparative sequencing analysis reveals high genomic concordance between matched primary and metastatic colorectal cancer lesions
.
Genome Biol
2014
;
15
:
454
.
4.
Vousden
KH
,
Lu
X
.
Live or let die: the cell’s response to p53
.
Nat Rev Cancer
2002
;
2
:
594
604
.
5.
Brosh
R
,
Rotter
V
.
When mutants gain new powers: news from the mutant p53 field
.
Nat Rev Cancer
2009
;
9
:
701
13
.
6.
Muller
PAJ
,
Vousden
KH
.
Mutant p53 in cancer: new functions and therapeutic opportunities
.
Cancer Cell
2014
;
25
:
304
17
.
7.
Olive
KP
,
Tuveson
DA
,
Ruhe
ZC
,
Yin
B
,
Willis
NA
,
Bronson
RT
, et al
.
Mutant p53 gain of function in two mouse models of Li-Fraumeni syndrome
.
Cell
2004
;
119
:
847
60
.
8.
Lang
GA
,
Iwakuma
T
,
Suh
Y-A
,
Liu
G
,
Rao
VA
,
Parant
JM
, et al
.
Gain of function of a p53 hot spot mutation in a mouse model of Li-Fraumeni syndrome
.
Cell
2004
;
119
:
861
72
.
9.
Lu
X
,
Liu
DP
,
Xu
Y
.
The gain of function of p53 cancer mutant in promoting mammary tumorigenesis
.
Oncogene
2013
;
32
:
2900
6
.
10.
Solomon
H
,
Dinowitz
N
,
Pateras
IS
,
Cooks
T
,
Shetzer
Y
,
Molchadsky
A
, et al
.
Mutant p53 gain of function underlies high expression levels of colorectal cancer stem cells markers
.
Oncogene
2018
;
37
:
1669
84
.
11.
Nakayama
M
,
Sakai
E
,
Echizen
K
,
Yamada
Y
,
Oshima
H
,
Han
T-S
, et al
.
Intestinal cancer progression by mutant p53 through the acquisition of invasiveness associated with complex glandular formation
.
Oncogene
2017
;
36
:
5885
96
.
12.
Sakai
E
,
Nakayama
M
,
Oshima
H
,
Kouyama
Y
,
Niida
A
,
Fujii
S
, et al
.
Combined mutation of Apc, Kras, and Tgfbr2 effectively drives metastasis of intestinal cancer
.
Cancer Res
2018
;
78
:
1334
46
.
13.
Nakayama
M
,
Hong
CP
,
Oshima
H
,
Sakai
E
,
Kim
S-J
,
Oshima
M
.
Loss of wild-type p53 promotes mutant p53-driven metastasis through acquisition of survival and tumor-initiating properties
.
Nat Commun
2020
;
11
:
2333
.
14.
Alexandrova
EM
,
Yallowitz
AR
,
Li
D
,
Xu
S
,
Schulz
R
,
Proia
DA
, et al
.
Improving survival by exploiting tumour dependence on stabilized mutant p53 for treatment
.
Nature
2015
;
523
:
352
6
.
15.
Schulz-Heddergott
R
,
Stark
N
,
Edmunds
SJ
,
Li
J
,
Conradi
L-C
,
Bohnenberger
H
, et al
.
Therapeutic ablation of gain-of-function mutant p53 in colorectal cancer inhibits stat3-mediated tumor growth and invasion
.
Cancer Cell
2018
;
34
:
298
314.e7
.
16.
Zhu
J
,
Sammons
MA
,
Donahue
G
,
Dou
Z
,
Vedadi
M
,
Getlik
M
, et al
.
Gain-of-function p53 mutants co-opt chromatin pathways to drive cancer growth
.
Nature
2015
;
525
:
206
11
.
17.
Pfister
NT
,
Fomin
V
,
Regunath
K
,
Zhou
JY
,
Zhou
W
,
Silwal-Pandit
L
, et al
.
Mutant p53 cooperates with the SWI/SNF chromatin remodeling complex to regulate VEGFR2 in breast cancer cells
.
Genes Dev
2015
;
29
:
1298
315
.
18.
Xue
Y
,
San Luis
B
,
Lane
DP
.
Intratumour heterogeneity of p53 expression; causes and consequences
.
J Pathol
2019
;
249
:
274
85
.
19.
Takeuchi
K
,
Tanaka
A
,
Kato
S
,
Aihara
E
,
Amagase
K
.
Effect of (S)-4-(1-(5-Chloro-2-(4-fluorophenyoxy)benzamido)ethyl) benzoic acid (CJ-42794), a selective antagonist of prostaglandin E receptor subtype 4, on ulcerogenic and healing responses in rat gastrointestinal mucosa
.
J Pharmacol Exp Ther
2007
;
322
:
903
12
.
20.
Kok
SY
,
Oshima
H
,
Takahashi
K
,
Nakayama
M
,
Murakami
K
,
Ueda
HR
, et al
.
Malignant subclone drives metastasis of genetically and phenotypically heterogenous cell clusters through fibrotic niche generation
.
Nat Commun
2021
;
12
:
863
.
21.
Ahmed
D
,
Eide
PW
,
Eilertsen
IA
,
Danielsen
SA
,
Eknæs
M
,
Hektoen
M
, et al
.
Epigenetic and genetic features of 24 colon cancer cell lines
.
Oncogenesis
2013
;
2
:
e71
.
22.
Djelloul
S
,
Forgue-Lafitte
ME
,
Hermelin
B
,
Mareel
M
,
Bruyneel
E
,
Baldi
A
, et al
.
Enterocyte differentiation is compatible with SV40 large T expression and loss of p53 function in human colonic Caco-2 cells. Status of the pRb1 and pRb2 tumor suppressor gene products
.
FEBS Lett
1997
;
406
:
234
42
.
23.
Wang
D
,
Nakayama
M
,
Hong
CP
,
Oshima
H
,
Oshima
M
.
Gain-of-function p53 mutation acts as a genetic switch for TGFβ signaling-induced epithelial-to-mesenchymal transition in intestinal tumors
.
Cancer Res
2024
;
84
:
56
68
.
24.
Alexandrova
EM
,
Mirza
SA
,
Xu
S
,
Schulz-Heddergott
R
,
Marchenko
ND
,
Moll
UM
.
p53 loss-of-heterozygosity is a necessary prerequisite for mutant p53 stabilization and gain-of-function in vivo
.
Cell Death Dis
2017
;
8
:
e2661
.
25.
Nakayama
M
,
Oshima
M
.
Mutant p53 in colon cancer
.
J Mol Cell Biol
2019
;
11
:
267
76
.
26.
Castellone
MD
,
Teramoto
H
,
Williams
BO
,
Druey
KM
,
Gutkind
JS
.
Prostaglandin E2 promotes colon cancer cell growth through a Gs-axin-β-catenin signaling axis
.
Science
2005
;
310
:
1504
10
.
27.
Shao
J
,
Jung
C
,
Liu
C
,
Sheng
H
.
Prostaglandin E2 stimulates the β-catenin/T cell factor-dependent transcription in colon cancer
.
J Biol Chem
2005
;
280
:
26565
72
.
28.
Goessling
W
,
North
TE
,
Loewer
S
,
Lord
AM
,
Lee
S
,
Stoick-Cooper
CL
, et al
.
Genetic interaction of PGE2 and Wnt signaling regulates developmental specification of stem cells and regeneration
.
Cell
2009
;
136
:
1136
47
.
29.
Wang
Z
,
Burigotto
M
,
Ghetti
S
,
Vaillant
F
,
Tan
T
,
Capaldo
BD
, et al
.
Loss-of-function but not gain-of-function properties of mutant TP53 are critical for the proliferation, survival, and metastasis of a broad range of cancer cells
.
Cancer Discov
2024
;
14
:
362
79
.
30.
Hageman
JH
,
Heinz
MC
,
Kretzschmar
K
,
van der Vaart
J
,
Clevers
H
,
Snippert
HJG
.
Intestinal regeneration: regulation by the microenvironment
.
Dev Cell
2020
;
54
:
435
46
.
31.
Nakao
S
,
Ogtata
Y
,
Shimizu
E
,
Yamazaki
M
,
Furuyama
S
,
Sugiya
H
.
Tumor necrosis factor alpha (TNF-alpha)-induced prostaglandin E2 release is mediated by the activation of cyclooxygenase-2 (COX-2) transcription via NFkappaB in human gingival fibroblasts
.
Mol Cell Biochem
2002
;
238
:
11
8
.
32.
Oshima
M
,
Dinchuk
JE
,
Kargman
SL
,
Oshima
H
,
Hancock
B
,
Kwong
E
, et al
.
Suppression of intestinal polyposis in Apc delta716 knockout mice by inhibition of cyclooxygenase 2 (COX-2)
.
Cell
1996
;
87
:
803
9
.
33.
Sonoshita
M
,
Takaku
K
,
Sasaki
N
,
Sugimoto
Y
,
Ushikubi
F
,
Narumiya
S
, et al
.
Acceleration of intestinal polyposis through prostaglandin receptor EP2 in Apc (Delta 716) knockout mice
.
Nat Med
2001
;
7
:
1048
51
.
34.
Roulis
M
,
Kaklamanos
A
,
Schernthanner
M
,
Bielecki
P
,
Zhao
J
,
Kaffe
E
, et al
.
Paracrine orchestration of intestinal tumorigenesis by a mesenchymal niche
.
Nature
2020
;
580
:
524
9
.
This open access article is distributed under the Creative Commons Attribution 4.0 International (CC BY 4.0) license.