Mutations of
APC are common in CRC, whereas mutations of
AXIN1, a gene encoding another component of the Wnt pathway, are infrequent in these cancers.
4AXIN1 and AXIN2 variants in gastrointestinal cancers.
The potential tumor-suppressor function of Axin1 in CRC remains unclear, but compounds stabilizing this protein have been reported to suppress the growth of Wnt-driven CRCs.
5- Huang S.M.
- Mishina Y.M.
- Liu S.
- et al.
Tankyrase inhibition stabilizes axin and antagonizes Wnt signalling.
,6- Lau T.
- Chan E.
- Callow M.
- et al.
A novel tankyrase small-molecule inhibitor suppresses APC mutation-driven colorectal tumor growth.
By contrast, a tumor-suppressor function for Axin1 has been shown clearly in hepatocellular carcinoma (HCC), for which
AXIN1 mutations are identified in almost 10% of cases.
7- Zucman-Rossi J.
- Villanueva A.
- et al.
Genetic landscape and biomarkers of hepatocellular carcinoma.
It has been suggested that these mutations are associated with the role of Axin1 in the down-regulation of Wnt/β-catenin signaling.
8- Satoh S.
- Daigo Y.
- Furukawa Y.
- et al.
AXIN1 mutations in hepatocellular carcinomas, and growth suppression in cancer cells by virus-mediated transfer of AXIN1.
However, most human HCCs with
AXIN1 mutations studied have not been found to be associated with higher levels of β-catenin activation, and the HCCs developing in mice with
Axin1 inactivation in hepatocytes do not display aberrant activation of the Wnt pathway.
9- Abitbol S.
- Dahmani R.
- Coulouarn C.
- et al.
AXIN deficiency in human and mouse hepatocytes induces hepatocellular carcinoma in the absence of beta-catenin activation.
Nevertheless, Axin1 initially was identified as a negative regulator of the canonical Wnt pathway.
10- Zeng L.
- Fagotto F.
- Zhang T.
- et al.
The mouse fused locus encodes Axin, an inhibitor of the Wnt signaling pathway that regulates embryonic axis formation.
,11- Kishida S.
- Yamamoto H.
- Ikeda S.
- et al.
Axin, a negative regulator of the Wnt signaling pathway, directly interacts with adenomatous polyposis coli and regulates the stabilization of beta-catenin.
Several lines of evidence indicate that Axin1 and its homolog Axin2 play a key role in down-regulating the stability of β-catenin, encoded by
Ctnnb1, which is crucial for the cellular response Wnt/β-catenin signaling.
12Wnt/beta-catenin signaling and disease.
In the absence of the Wnt signal, Axins act as scaffold proteins, together with the tumor-suppressor Adenomatous polyposis coli, in the destruction complex targeting β-catenin for degradation. However, Axin1 also is involved in transducing the Wnt signal, leading to an increase in cytoplasmic and nuclear β-catenin levels and the expression of β-catenin target genes.
12Wnt/beta-catenin signaling and disease.
Axin1 also acts as a scaffold protein for other non-Wnt pathways, including the c-Jun N-terminal kinase, transforming growth factor-β, p53, and AMP-activated protein kinase pathways.
13- Salahshor S.
- Woodgett J.R.
The links between axin and carcinogenesis.
,14AMPK: sensing glucose as well as cellular energy status.
Given the complex functions of Axin1, we decided to investigate its role in normal intestinal homeostasis and its tumor-suppressor function in CRC. Using mice with an epithelium-specific inactivation of Axin1, we showed that Axin1 was dispensable for normal intestinal homeostasis and redundant with Axin2 for down-regulation of the Wnt pathway. Mice that have combined deficiencies for Axin1 and Axin2 phenocopied Apc-deficient mice. We found that Axin1 deficiency in intestinal epithelial cells promoted the development of azoxymethane (AOM)-induced colon tumorigenesis, but induced an improvement of dextran sulfate sodium (DSS)-induced colitis. Axin1 therefore promotes an inflammatory program with tumor-suppressor function in intestinal epithelial cells that we identified as an interferon (IFN)γ/helper T cell 1 (Th1) immune program. We defined an Axin1-dependent gene expression signature (GES), which we then used to probe databases of human CRC transcriptomes. Patients with tumor profiles defined by the signature as Axin1-deficient had poor antitumor immune response and poor clinical outcome, whereas those with the Axin1-proficient profile had a better survival and an immune profile of potential vulnerability to immune checkpoint blockade therapies.
Discussion
We show that Axin1 is redundant with Axin2 for down-regulation of Wnt/β-catenin signaling in the intestinal epithelium. Axin1 is dispensable for intestinal homeostasis, deficiency of both Axin1 and Axin2 in the intestinal epithelium is required to phenocopy intestine-specific Apc knockout mice. This might explain why the Wnt/β-catenin pathway does not appear to be involved in the tumor-suppressor role of Axin1 in the intestine. We identify an unrecognized role for Axin1 in the control of the immune response in the intestine capable of accounting for its tumor-suppressor function. We found that Axin1 deficiency in intestinal epithelial cells rendered mice more susceptible to chemically induced colon carcinogenesis, but reduced DSS-induced colitis by attenuating the induction of a proinflammatory program. RNA-seq analyses identified an IFNγ/Th1 immune program controlled by Axin1 that enhances the inflammatory response and protects against CRC. These observations in mice were complemented by in silico analysis of 2239 human CRC tumors, which showed that patients with weak expression of the Axin1-dependent GES had a worse prognosis and suggested that those with strong expression of the Axin1-dependent GES might benefit from immune checkpoint blockade.
We investigated the role of Axin1 in the intestine because of the crucial role of the Wnt pathway in the control of homeostasis and tumorigenesis of the gastrointestinal tract.
35Wnt/beta-catenin signaling, disease, and emerging therapeutic modalities.
We found that
Axin1 was dispensable for normal intestinal homeostasis, its loss being compensated by
Axin2. Mice with double
Axin1,
Axin2 deficiencies in the intestinal epithelium phenocopied Apc
ΔIEC mice and died rapidly. The reason for the lethality likely is owing to the failed differentiation of the intestinal lineages induced by the aberrant Wnt activation that should alter the function of the intestine.
17- Andreu P.
- Colnot S.
- Godard C.
- et al.
Crypt-restricted proliferation and commitment to the Paneth cell lineage following Apc loss in the mouse intestine.
,36- Sansom O.J.
- Reed K.R.
- Hayes A.J.
- et al.
Loss of Apc in vivo immediately perturbs Wnt signaling, differentiation, and migration.
We expected the compensation of
Axin1 loss by
Axin2 for Wnt pathway down-regulation to be observed only in tissues in which Wnt signaling was activated because
Axin2 is a canonical β-catenin target gene.
37- Lustig B.
- Jerchow B.
- Sachs M.
- et al.
Negative feedback loop of Wnt signaling through upregulation of conductin/axin2 in colorectal and liver tumors.
We therefore analyzed the combined inactivation of
Axin1 and
Axin2 in the liver, in which the activation of Wnt signaling is restricted to a ring of a few hepatocytes around the central vein constituting the perivenous region of the liver lobule.
19- Benhamouche S.
- Decaens T.
- Godard C.
- et al.
Apc tumor suppressor gene is the "zonation-keeper" of mouse liver.
The Wnt pathway is not activated in most hepatocytes, and it is even inhibited in the periportal area owing to high levels of
Apc expression.
19- Benhamouche S.
- Decaens T.
- Godard C.
- et al.
Apc tumor suppressor gene is the "zonation-keeper" of mouse liver.
We previously described the specific deletion of
Axin1 from all hepatocytes.
9- Abitbol S.
- Dahmani R.
- Coulouarn C.
- et al.
AXIN deficiency in human and mouse hepatocytes induces hepatocellular carcinoma in the absence of beta-catenin activation.
As reported here for intestinal cells, no particular phenotype was detected in the corresponding mice. Surprisingly, the combined deletion of
Axin1 and
Axin2 in hepatocytes led to an aberrant activation of β-catenin signaling throughout the liver lobule, even in the periportal area in which
Axin2 expression was not detected.
19- Benhamouche S.
- Decaens T.
- Godard C.
- et al.
Apc tumor suppressor gene is the "zonation-keeper" of mouse liver.
The liver phenotype of these mice mimicked that observed after targeted
Apc loss in hepatocytes.
19- Benhamouche S.
- Decaens T.
- Godard C.
- et al.
Apc tumor suppressor gene is the "zonation-keeper" of mouse liver.
These results therefore suggest that the compensation of A
xin1 loss by A
xin2 also occurs in tissues in which Wnt/β-catenin signaling is not activated and in which
Axin2 expression levels are thought to be very low, such as periportal liver tissues. This surprising result supports the computational model of Lee et al,
38- Lee E.
- Salic A.
- Kruger R.
- et al.
The roles of APC and Axin derived from experimental and theoretical analysis of the Wnt pathway.
who considered Axin1 to be rate-limiting for the destruction complex owing to its low levels.
We identified a new role for epithelial Axin1 in controlling gut inflammation that might be associated with its tumor-suppressor function. RNA-seq analyses of the colon of DSS-treated Axin1
ΔIEC and WT mice identified an IFNγ/Th1 proinflammatory program that was down-regulated in Axin1
ΔIEC mice. Flow cytometry analyses confirmed the reduction of the Th1 and Th17 cells in the lamina propria of DSS-treated Axin1
ΔIEC mice that corroborated the resistance of these mice to DDS-induced colitis. Thus, epithelial Axin1 appears to promote a proinflammatory response mediated by activation of the IFNγ/Th1 pathway. Interestingly, the same program also was found to be down-regulated in tumors developing in Axin1
ΔIEC mice after the AOM/DSS protocol. The IFNγ/Th1 pathway is known to have tumor-suppressor functions responsible for tumor clearance in several cancers, including CRC,
27- Galon J.
- Costes A.
- Sanchez-Cabo F.
- et al.
Type, density, and location of immune cells within human colorectal tumors predict clinical outcome.
,39- Shankaran V.
- Ikeda H.
- Bruce A.T.
- et al.
IFNgamma and lymphocytes prevent primary tumour development and shape tumour immunogenicity.
,40Tumor immunology and tumor evolution: intertwined histories.
which could explain the increased AOM–DSS–driven colon tumorigenesis in Axin1
ΔIEC mice. Consistent with a decrease in immune surveillance, we detected by immunohistochemistry a decrease in the infiltration of the cytotoxic lymphocytes CD8
+ in tumors that developed in Axin1
ΔIEC mice. This correlates with the significantly large numbers of small tumors in the Axin1
ΔIEC mice and suggests that epithelial Axin1 suppresses the initiation of tumorigenesis. The tumor-suppressor function of Axin1 in the intestinal epithelial cells also involved a negative control of cell proliferation as shown by the strong increase in Ki-67 staining in tumors of Axin1
ΔIEC mice. Interestingly, the increase in cell proliferation in the tumors of Axin1
ΔIEC mice was not associated with an increase in Wnt/β-catenin signaling, likely owing to compensation by its paralog
Axin2, induced in the tumors (
Figure 8I).
The precise mechanism by which intestinal epithelial Axin1 exerts its tumor-suppressor function needs further clarification. Our data showed that Axin1
ΔIEC mice were more susceptible to AOM–DSS–driven colon tumorigenesis, but were more resistant to DSS-induced colitis. These observations are inconsistent with the paradigm that linked inflammation and cancer.
41- Grivennikov S.I.
- Greten F.R.
- Karin M.
Immunity, inflammation, and cancer.
However, inflammation has the role of a double-edged sword in cancer and may either induce antitumor immunity or promote tumor development, specifically in CRC.
40Tumor immunology and tumor evolution: intertwined histories.
Indeed, in human beings, an IFNγ/Th1 response is characteristic of cancer immune surveillance, associated with Th1CD4
+ and CD8
+T cells, which directly regulate tumor cell cytotoxicity and induce tumor suppression.
25- Dunn G.P.
- Koebel C.M.
- Schreiber R.D.
Interferons, immunity and cancer immunoediting.
Our data strongly suggest that intestinal epithelial Axin1 exerts its tumor-suppressor function by inducing an antitumorigenic IFNγ/Th1 response. How epithelial Axin1 promotes a proinflammatory IFNγ/Th1 response remains to be investigated. We examined if it was via an epithelium-extrinsic and/or -intrinsic function. Analysis of the intestinal permeability showed no significant difference between the increase in the DSS-induced clearance of the TRITC-dextran in the colon of Axin1
ΔIEC and WT mice, suggesting no specific alteration in the epithelial barrier integrity between the KO and WT mice upon the DSS challenge. This is in agreement with data showing that the systemic response to bacterial antigens can occur independently of epithelial barrier disruption.
42- Sydora B.C.
- Tavernini M.M.
- Doyle J.
- Fedorak R.N.
A defect in epithelial barrier integrity is not required for a systemic response to bacterial antigens or intestinal injury in T cell receptor-alpha gene-deficient mice.
These results favor an epithelium-intrinsic mechanism by which Axin1 promotes a proinflammatory response in the colon. From the RNA-seq/in silico analyses of both the tumors (
Figure 10A, and Tables 1 and 2 in
Figure 10) and the DSS-treated colon (
Figure 11C, and Tables 3 and 4 in
Figure 11), we can speculate that Axin1 participates in the transduction pathways that allow, under the sensing of microbial stimuli, the synthesis of immunoregulatory signals in the intestinal epithelial cells. In agreement with this hypothesis, a role for epithelial Axin1 in host–pathogen interactions has been described.
Salmonella typhimurium decreases Axin1 protein expression in intestinal epithelial cell lines at the post-transcriptional level, whereas its overexpression inhibits
Salmonella invasion bacterial infections in vitro.
43- Zhang Y.G.
- Wu S.
- Xia Y.
- et al.
Axin1 prevents Salmonella invasiveness and inflammatory response in intestinal epithelial cells.
The RNA-seq data identified IFNγ/Stat1 and TNFα as the inflammatory pathways targeted by epithelial Axin1. This is consistent with the phenotype of mice with a genetic or a pharmacologic inhibition of
Stat1 that showed a decrease in the Th1 response and a reduction in chemically induced colitis.
44- Bandyopadhyay S.K.
- de la Motte C.A.
- Kessler S.P.
- et al.
Hyaluronan-mediated leukocyte adhesion and dextran sulfate sodium-induced colitis are attenuated in the absence of signal transducer and activator of transcription 1.
,45- Wu X.
- Guo W.
- Wu L.
- et al.
Selective sequestration of STAT1 in the cytoplasm via phosphorylated SHP-2 ameliorates murine experimental colitis.
Strikingly, the specific deletion of
Stat1 in intestinal epithelial cells phenocopied Axin1
ΔIEC mice and showed a tumor-suppressor function of Stat1 in CRC. Stat1
ΔIEC mice were more resistant to DSS-induced colitis and showed an increased tumor load upon the AOM/DSS protocol.
46- Crncec I.
- Modak M.
- Gordziel C.
- et al.
STAT1 is a sex-specific tumor suppressor in colitis-associated colorectal cancer.
The mechanism by which epithelial Axin1 is able to activate the inflammatory pathways IFNγ/Stat1 and TNFα is largely unknown at present. Axin1 is a scaffolding protein for which many interactors have been described (
https://www.ncbi.nlm.nih.gov/gene/8312) and has not yet been connected to any inflammatory pathway. Among the already identified interactors, Card9 and Traf2 have been implicated in intestinal inflammatory responses and cancer,
47- Wang T.
- Fan C.
- Yao A.
- et al.
The adaptor protein CARD9 protects against colon cancer by restricting mycobiota-mediated expansion of myeloid-derived suppressor cells.
,48- Piao J.H.
- Hasegawa M.
- Heissig B.
- et al.
Tumor necrosis factor receptor-associated factor (TRAF) 2 controls homeostasis of the colon to prevent spontaneous development of murine inflammatory bowel disease.
and may represent candidates meriting investigation.
Interestingly, the immune landscape has prognostic value
49- Bruni D.
- Angell H.K.
- Galon J.
The immune contexture and Immunoscore in cancer prognosis and therapeutic efficacy.
; notably, the features of the tumor microenvironment such as a lack of T-cell infiltration, and low levels of Th1 cell activity resulting in low levels of immune cytotoxicity, are predictive of adverse outcomes in patients with CRC; conversely, the presence of cytotoxic immune infiltration has been identified as a favorable prognostic marker.
27- Galon J.
- Costes A.
- Sanchez-Cabo F.
- et al.
Type, density, and location of immune cells within human colorectal tumors predict clinical outcome.
,50- Mlecnik B.
- Bindea G.
- Kirilovsky A.
- et al.
The tumor microenvironment and Immunoscore are critical determinants of dissemination to distant metastasis.
RNA-seq and flow cytometry analyses of tumors from
Axin1-deficient mice predicted a tumor microenvironment with low levels of IFNγ/Th1 cell activity. We therefore investigated whether the
Axin1-dependent gene signature that we identified was of prognostic value in human CRC. In our 2239-sample series, the
Axin1 signature-based classification was found to be an independent prognostic feature: patients from the Axin1-proficient-like group had a longer DFS than those from the Axin1-deficient-like group. The difference in 5-year DFS was small (5%) between the 2 groups, but was significant in multivariate analysis. These Axin1-proficient-like patients also had molecular traits suggestive of a significantly stronger Th1/IFNγ/cytotoxic antitumor response that would make them more likely to respond to immunotherapy than Axin1-deficient-like patients and opens up possibilities for treatment. First, patients with the Axin1-proficient-like signature were enriched for CMS1 class, which is associated with a microsatellite instability -high status and predicted to respond best to immunotherapy, and thus might be expected to benefit from immune checkpoint blockade. Second, the use of tankyrase inhibitors could be optimized; these drugs stabilize Axin proteins
5- Huang S.M.
- Mishina Y.M.
- Liu S.
- et al.
Tankyrase inhibition stabilizes axin and antagonizes Wnt signalling.
and have emerged as promising treatments for CRC,
6- Lau T.
- Chan E.
- Callow M.
- et al.
A novel tankyrase small-molecule inhibitor suppresses APC mutation-driven colorectal tumor growth.
although their clinical use is limited by their toxicity. The combined use of an immunotherapy approach together with tankyrase inhibitors might be expected to lower the effective concentration range for the use of these inhibitors, at least in the group of patients bearing the Axin1-proficient-like signature.
In conclusion, our study showed a new mechanism by which Axin1 has tumor-suppressive function in CRC, which might open new therapeutic perspectives in colorectal cancer.
Materials and Methods
Ethical Compliance Statement
All animal procedures were performed according to French legal regulations (Ministère de la Recherche, de l’Enseignement Supérieur et de l’Innovation), with the approval of the Ethics Committee of Paris Descartes University (Project autorisation pour projets utilisant animaux à des fins scientifiques [APAFIS] 8722 and 8612).
The human in silico study is based on public data from published studies in which the informed patients' consent to participate and the ethics and institutional review board already were obtained by authors. The study was approved by our Institutional Review Board from Institut Paoli-Calmettes (Marseille, France; Comité d’Orientation Stratégique).
Mice
Mice with an inducible specific deletion of
Axin1 in intestinal epithelial cells (Axin1
ΔIEC) were obtained by crossing
Axin1fl/fl mice
9- Abitbol S.
- Dahmani R.
- Coulouarn C.
- et al.
AXIN deficiency in human and mouse hepatocytes induces hepatocellular carcinoma in the absence of beta-catenin activation.
with Villin Cre-ERT2 mice expressing an inducible Cre-recombinase under the control of the villin promoter.
51- el Marjou F.
- Janssen K.P.
- Chang B.H.
- et al.
Tissue-specific and inducible Cre-mediated recombination in the gut epithelium.
Axin2-null mice
16- Yu H.M.
- Jerchow B.
- Sheu T.J.
- et al.
The role of Axin2 in calvarial morphogenesis and craniosynostosis.
were obtained from Anne-Amandine Chassot (Nice, France). For all experiments, approximately equal numbers of male and female mice were analyzed.
Mice were housed in colony cages in specific pathogen-free conditions, under a 12-hour light/dark cycle, in a controlled-temperature environment (21°C). They were fed ad libitum with a standard laboratory chow diet (65% carbohydrate, 11% lipids, and 24% proteins; SAFE 03, FRANCE).
Unless otherwise specified, we injected 4 doses of tamoxifen, 1 per day for 4 days (1 mg/mouse of tamoxifen, T5648; Sigma-Aldrich) in corn oil IP into 8- to 12-week-old mice.
For the inactivation of
Axin1 in hepatocytes, Ad5-CMV-cre and Ad5-GFP (2 × 10
9 plaque forming unit in 150 mL) were injected into the retro-orbital vein of 8-week-old mice anesthetized by isoflurane inhalation as previously described.
19- Benhamouche S.
- Decaens T.
- Godard C.
- et al.
Apc tumor suppressor gene is the "zonation-keeper" of mouse liver.
DSS-Induced Colitis and Histologic Score
This experiment was performed as previously described.
52- Chassaing B.
- Aitken J.D.
- Malleshappa M.
- Vijay-Kumar M.
Dextran sulfate sodium (DSS)-induced colitis in mice.
Six- to 8-week-old WT (Axin1
fl/fl) and KO (Axin1
ΔIEC) mice received 3% DSS (weight/volume) in drinking water for 7 days and then were given regular drinking water until they were killed. Mice from each strain and gender were kept in separate cages. Mice were monitored daily for weight loss and clinical disease (soft feces).
A histologic score was obtained according to the criteria described by Erben et al
53- Erben U.
- Loddenkemper C.
- Doerfel K.
- et al.
A guide to histomorphological evaluation of intestinal inflammation in mouse models.
based on inflammation, hyperplasia, epithelial damage, and mucosal architecture.
Induction of Colorectal Carcinogenesis
Colorectal tumorigenesis was induced in mice with the AOM-plus-DSS model, as previously described.
22- Neufert C.
- Becker C.
- Neurath M.F.
An inducible mouse model of colon carcinogenesis for the analysis of sporadic and inflammation-driven tumor progression.
Briefly, 6- to 8-week-old WT (Axin1
fl/fl) and KO (Axin1
ΔIEC) mice received a single IP injection of AOM (10 mg/kg body weight; Sigma). Mice from different strains and sexes were kept in separate cages. Over the next few weeks, mice were exposed to 3 cycles of treatment with 2.5% DSS (weight/volume) in drinking water for 7 days, with a 14-day interval in which the mice received regular water between cycles, for recovery. Mice were killed on day 100 after AOM injection.
Immunoblot Analysis
Total protein extracts were obtained from 100 mg frozen mouse liver or from mouse primary hepatocytes homogenized in lysis buffer (50 mmol/L Tris-HCl, pH 7.5, 150 mmol/L NaCl, 5 mmol/L EDTA, 30 mmol/L Na4P2O7, 50 mmol/L Sodium fluoride [NaF], 1% Triton X-100 [Sigma-Aldrich], 1 mmol/L dithiothreitol) protease inhibitor cocktail (32953; Pierce, Thermo Fisher Scientific) supplemented with phosphatase inhibitor cocktail (8867; Pierce, Thermo Fisher Scientific) in a bead mill, with the Tissue Lyser disruption system (Qiagen, Hilden, Germany). Proteins were resolved by sodium dodecyl sulfate–polyacrylamide gel electrophoresis. The resulting bands were transferred to nitrocellulose membranes, which were blocked by incubation with 5% bovine serum albumin or 5% milk. Blots were incubated overnight at 4°C with specific primary antibodies, washed, incubated with the corresponding horseradish-peroxidase–conjugated secondary antibodies (Cell Signaling), and developed by an enhanced chemiluminescence technique (Thermo Fisher Scientific). Primary antibodies were obtained from Cell Signaling Technologies (Axin1, ref: C76H11, 1:500) and Santa Cruz Biotechnology (glyceraldehyde-3-phosphate dehydrogenase [Gapdh], ref: sc-25778, 1:1000).
Histology and Immunohistochemistry
Immediately after death, the entire gastrointestinal tract of the mouse was removed, splayed open along its length, and rolled up from the proximal to the distal end to form a Swiss roll. Tissues were fixed by incubation in 4% paraformaldehyde overnight at 4°C and embedded in paraffin wax. Sections (3-μm thick) were cut and stained with H&E. Immunohistochemistry was performed as previously described.
54- Trentesaux C.
- Fraudeau M.
- Pitasi C.L.
- et al.
Essential role for autophagy protein ATG7 in the maintenance of intestinal stem cell integrity.
We treated 5-μm sections with 3% hydrogen peroxide for 15 minutes at room temperature. Antigens then were retrieved by boiling for 15 minutes in citrate buffer (10 mmol/L, pH 6) or for 40 minutes in Tris-EDTA buffer (100 mmol/L Tris, 12.6 mmol/L, pH 9) in a microwave pressure cooker (EZ Retriever; Biogenex). The sections were incubated in blocking solution (2% goat serum, 1% bovine serum albumin diluted in Tris Buffer Saline Tween) for 20 minutes at room temperature. They then were incubated overnight at 4°C with the primary antibodies against the following proteins, diluted in blocking solution: cleaved caspase-3 (9661S, 1:200; Cell Signaling), Ki-67 (12202S, 1:1500; Cell Signaling), lysozyme (EC.3.2.1.17, 1:200; Dako), Olfm4 (CS 39141S, 1:400; Cell Signaling), p53 (NCL-p53-CM5p, 1:200; Leica), γH2AX (05-636, 1:300; Millipore), chromogranin B (HPA012602, 1:1000; Atlas Antibodies), villin (ab130751, 1:500; Abcam), glutamine synthetase (610518, 1:400; BD Biosciences), and CD8 (14-0808-80, 1:100; eBiosciences). Specific binding was detected with a biotinylated secondary antibody and ABC reagent (Vector) for immunohistochemistry, and the signal was revealed with 3,3′-diaminobenzidine tetra hydrochloride solution (Vector). Hematoxylin was used as a nuclear counterstain. Apoptosis was assessed with a TUNEL assay kit, in accordance with the manufacturer's instructions (QIA33; Calbiochem). In the crypt, the 5–6 cells located above the +4 cells were considered transit-amplifying cells.
Quantification of the labeled positive cells was performed in almost 40 consecutive crypts in the small intestine and in 20 consecutive crypts in the colon. This provides an average of positive cells in the crypts per animal. Quantification was performed on 3 or 4 different mice in each group and statistics were performed on these combined averaged numbers.
Mouse livers were cut into 3-mm–thick sections and fixed by incubation in 4% paraformaldehyde for 12 hours. They then were embedded in paraffin. Immunohistochemistry was performed as previously described.
19- Benhamouche S.
- Decaens T.
- Godard C.
- et al.
Apc tumor suppressor gene is the "zonation-keeper" of mouse liver.
An antibody against glutamine synthetase was used (610518, 1:400; BD Biosciences).
Intestinal Permeability
Four hours (average time of intestinal transit in mice) before killing, overnight fasted mice were orally gavaged with a solution of TRITC dextran 4 kilodaltons at 50 mg/mL in phosphate-buffered saline (PBS) (TdB Labs, Sweden). Blood then was collected at the tail tip. Plasma was analyzed for the TRITC dextran 4 kilodalton concentration using an automatic fluorescence microplate reader at 544-nm excitation and 580-nm emission wavelengths to determine the intestinal epithelium permeability. TRITC concentrations were determined using standard curves prepared by diluting various amounts of TRITC-dextran in PBS.
RNA Extraction and Quantitative Real-Time PCR Analysis
Total RNA was extracted from mouse tissues with TRIzol Reagent (Life Technologies) in accordance with the manufacturer’s protocol. Reverse-transcription was performed with 1 μg total RNA and the Transcriptor First-Strand Complementary DNA Synthesis Kit (Roche Diagnostics), with random hexamer primers. Quantitative PCR was performed with the Light Cycler 480 Sybr Green I Master kit (Roche) and specific primers (Eurogentec) on a Light Cycler 480 thermocycler (Roche). RNA levels were calculated by the 2(-Delta Ct) method, with 18S as the internal control, relative to RNA levels in control (Axin1flfl) mice.
RNA-Seq Analysis
RNA extracted from colon tissue (distal colon) was treated with DNAse1 according to the manufacturer’s protocol (RNA Clean and Concentrator-5, R1013; Zymo Research). Three different mice per genotype were used for RNA-seq analysis. Library preparation and sequencing were performed at the GenomIC’ sequencing facility using 3 RNA samples per genotype (
https://www.institutcochin.fr/core_facilities/genomesequencing-studies). The libraries were prepared following the TruSeq Stranded messenger RNA protocol from Illumina, starting from 800 ng high-quality total RNA (RNA Integrity Number, >8). Paired-end (2 × 75 bp) sequencing was performed on an Illumina Nextseq 500 platform, 20–40 millions paired end reads were used for the analysis. After sequencing, a primary analysis was performed with AOZAN software (ENS, Paris) for demultiplexing and quality control analysis on the raw data (based on FastQC modules, version 0.11.5).
The STAR algorithm (version 2.7.6a; Genomic'IC) then was used to align the Fastq files with the Ensembl Mus musculus GRCm38 reference, release 101 (Genomic'IC). Reads then were counted with RSEM (v1.3.1; Genomic'IC) and statistical analyses were performed on the read counts with R (version 3.6.3; Genomic'IC) and the DESeq2 package (DESeq2_1.26.0; Genomic'IC) to determine the proportion of genes differentially expressed between 2 conditions.
We used the standard DESeq2 normalization method (DESeq2 median of ratios with the DESeq function), with a prefilter of reads and genes (reads uniquely mapped on the genome, or up to 10 different loci with a count adjustment, and genes with at least 10 reads in at least 3 different samples).
In accordance with the recommendations for this package, we used the Wald test with the contrast function and the Benjamini–Hochberg false discovery rates control procedure to identify DEGs.
Ontology Analyses
The list of genes showing significant differential expression was explored by both over-representation and GSEAs.
Significantly over-represented terms were identified with a right-tailed Fisher exact test, which calculated an overlap P value determining the probability of each term being associated with our lists of differential transcripts by chance alone.
The IPA z-score is a statistical measurement of correlation between the direction of the relationship and experimental levels of protein. It was calculated to assess the activation (positive z-score) or repression (negative score) of each term. The z-score was considered significant if its absolute value was greater than 2. GSEA was performed with Broad Institute GSEA software v4.1.0 (
http://software.broadinstitute.org/gesa/index.jsp).
55- Subramanian A.
- Tamayo P.
- Mootha V.K.
- et al.
Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles.
GSEA determined whether gene sets from the Molecular Signatures Database Hallmark collection (v7.4) were distributed randomly throughout our ranked transcript lists or whether they were located toward the top or bottom of these lists.
Heatmaps were generated with Genesis software (
https://genome.tugraz.at).
56- Sturn A.
- Quackenbush J.
- Trajanoski Z.
Genesis: cluster analysis of microarray data.
Venn diagrams were produced with the FunRich functional enrichment analysis tool v3.1.3 (
http://funrich.org).
57- Fonseka P.
- Pathan M.
- Chitti S.V.
- et al.
FunRich enables enrichment analysis of OMICs datasets.
Flow Cytometric Analyses
Lymphocytes were extracted from lamina propria as previously described.
58- Kim E.
- Tran M.
- Sun Y.
- Huh J.R.
Isolation and analyses of lamina propria lymphocytes from mouse intestines.
Briefly, small intestines were rinsed in cold PBS, minced into small pieces, incubated with EDTA (2 mmol/L), and digested with collagenase (1.5 mg/mL) for 30 minutes at 37°C. A discontinuous Percoll separation method (40% and 80%) was used to purify immune cells. Cell suspensions were centrifuged and the pellet was resuspended in 40% Percoll layered by 80% Percoll. The cells concentrated at the interface were collected and washed in cold PBS. Cells were suspended in RPMI1640 containing 5% fetal bovine serum, counted, incubated with human FcgammaR-binding inhibitor Receptor Binding Inhibitor Polyclonal Antibody (14-9161-73; eBioscience) and stained with specific antibodies.
Cells were subjected to fluorescence-activated cell sorting (FACS) analysis with the following antibodies: anti-CD4 (RM4-5 clone, 558107; BD Biosciences), anti-CD19 (1D3 clone, 550992; BD Biosciences) anti-IFNγ (XMG1.2 clone, 554412; BD Biosciences), or anti-transcription factor forkhead box P3 (FOXP3) (FJK-16s clone, 12-5773; eBioscience), anti-IL10 (JES5-16E3 clone, 554467; BD Biosciences), anti-IL17A (TC11-18H10 clone, 560220; BD Biosciences), and anti-RAR-related orphan receptor gamma (RORγ) (t) (REA278 clone, 130-123-248; Miltenyi Biotec). Dead cells were removed using the LIVE/DEAD Fixable Aqua Dead Cell Stain Kit (L34957; Invitrogen).
For intracellular staining, cells were pretreated with phorbol myristate acetate/ionomycin for 4 hours at 37°C 5% CO2 and brefeldin A was added for the last 3 hours. Cells then were stained with specific extracellular antibodies, permeabilized with BD cytoFix/cytoPerm solution (555028; BD Biosciences) or with transcription factor staining buffer set (00-5523-00; eBiosicence), and, finally, stained intracellularly for IFNγ, IL10, RORγ(t), IL17, or Foxp3. FACS analysis was performed with the BD LSR Fortessa flow cytometer (BD Biosciences) with FlowJo software (BD Bioscience).
Gene Expression Analysis on Colon Cancer Clinical Samples
We assessed the biological and clinical relevance of our 327-gene signature (
Axin1-deficient mice vs
Axin1-proficient mice) in human CRC samples by analyzing our database including clinical, pathologic, and gene expression data for primary CRC samples from 10 publicly available data sets and our own data set generated at the Institut Paoli-Calmettes (Marseille, France). Data were collected from the National Center for Biotechnology Information/GenBank Gene Expression Ommnibus, ArrayExpress, and The Cancer Genome Atlas (TCGA) databases (
Table 1). Samples were profiled with DNA microarrays (Affymetrix) or by RNA sequencing (Illumina). The data set analyzed contained data for 2239 primary CRC samples. The characteristics of these samples are summarized in
Table 2. Briefly, 88% of patients were older than age 50 years, and 53% were male. The proximal colon was the site of the CRC in 51% of cases. There were more stage 2 cancers (41%) than stage 3 cancers (34%). MMR status was classified as pMMR in 81% of cases. CMS determination identified 20% of samples as CMS1 (microsatellite instability immune), 32% as CMS2 (canonical), 17% as CMS3 (metabolic), and 31% as CMS4 (mesenchymal). DFS data were available for 1843 nonmetastatic patients with CMS subtyping data who had undergone surgery.
Data analysis required a preanalytical processing step, as previously described.
59- Guo Y.
- Gabola M.
- Lattanzio R.
- et al.
Cyclin A2 maintains colon homeostasis and is a prognostic factor in colorectal cancer.
We then applied several multigene molecular signatures separately to each tumor in each data set. We first used the 327 DEGs between the tumors of
Axin1-deficient mice and tumors from WT mice to define an
Axin1-deficient vs
Axin1-proficient expression profile. We then calculated the Pearson correlation distance between each sample and this profile: the samples with negative correlations were classified as
Axin1-proficient-like (
N = 1053) and those with positive correlations were classified as
Axin1-deficient-like (
N = 1186). The other gene expression signatures applied included the CMS classification,
60- Guinney J.
- Dienstmann R.
- Wang X.
- et al.
The consensus molecular subtypes of colorectal cancer.
the signatures of 24 different innate and adaptive immune cell subpopulations from Bindea et al,
28- Bindea G.
- Mlecnik B.
- Tosolini M.
- et al.
Spatiotemporal dynamics of intratumoral immune cells reveal the immune landscape in human cancer.
the immune constant of rejection classifier,
30- Bertucci F.
- Finetti P.
- Simeone I.
- et al.
The immunologic constant of rejection classification refines the prognostic value of conventional prognostic signatures in breast cancer.
metagenes associated with the T-cell–inflamed signature,
29- Ayers M.
- Lunceford J.
- Nebozhyn M.
- et al.
IFN-gamma-related mRNA profile predicts clinical response to PD-1 blockade.
the tertiary lymphoid structures signature,
34- Coppola D.
- Nebozhyn M.
- Khalil F.
- et al.
Unique ectopic lymph node-like structures present in human primary colorectal carcinoma are identified by immune gene array profiling.
the cytolytic activity score,
31- Rooney M.S.
- Shukla S.A.
- Wu C.J.
- et al.
Molecular and genetic properties of tumors associated with local immune cytolytic activity.
the antigen processing/presentation machinery score,
33- Thompson J.C.
- Davis C.
- Deshpande C.
- et al.
Gene signature of antigen processing and presentation machinery predicts response to checkpoint blockade in non-small cell lung cancer (NSCLC) and melanoma.
and the pathway activation scores from Gatza et al.
32- Gatza M.L.
- Lucas J.E.
- Barry W.T.
- et al.
A pathway-based classification of human breast cancer.
In statistical analysis, the correlations between the Axin1-proficient-like and Axin1-deficient-like groups and the clinical and pathologic variables were calculated with the Fisher exact test. The DFS was calculated from the date of diagnosis until the date of metastatic relapse or death from CRC. Follow-up evaluation was measured from the date of diagnosis to the date of last contact for event-free patients. Survival was calculated by the Kaplan–Meier method, and curves were compared in log-rank tests. Univariate and multivariate prognostic analyses for DFS were performed by Cox regression analysis (Wald test). The variables tested in univariate analysis included Axin1-proficient-like vs Axin1-deficient-like status, patient age (>50 y vs ≤50 y), sex (male vs female), tumor site (proximal vs distal), pathologic stage (2 vs 1, 3 vs 1), MMR status (pMMR vs deficient MMR), and CMS classification (CMS2 vs CMS1, CMS3 vs CMS1, and CMS4 vs CMS1). Multivariate analysis incorporated all variables with a P value less than 5% in univariate analysis. The correlations of molecular variables with Axin1-proficient-like and Axin1-deficient-like status were assessed by logistic regression analysis with the lm function (R statistical package; significance estimated by specifying a binomial family for models with a logit link). All statistical tests were 2-tailed and were performed with an α risk of 5%. Statistical analysis was performed with the survival package (version 2.43) in R software (version 3.5.2)
Statistical Analysis
Statistical analyses were performed with GraphPad Prism (9.04). Graphic data are represented as means ± SD unless specified. Data were checked for normality and unpaired 2-tailed Student t tests were used to assess differences between groups. P values less than .05 were considered significant.
Acknowledgments
The authors thank S. Robine (Institut Curie, Paris, France), A. Berns (Netherlands Cancer Institute, Amsterdam, The Netherlands), and Anne-Amandine Chassot (University of Nice Sophia Antipolis, Nice, France) for supplying mutant mice. The authors also thank Trinath Jamma (BITS Pilani-Hyderabad Campus, Telangana, India) for help and support for the cell sort at the Flow Core Facility, the staff of the Cochin animal facility, Isabelle Lagoutte from the Imagerie du Vivant facility of Cochin Institute, and Lucie Adoux from the Genomic facility of Cochin Institute.
CRediT Authorship Contributions
Christine Perret, PhD (Conceptualization: Lead; Data curation: Lead; Formal analysis: Lead; Funding acquisition: Lead; Investigation: Lead; Methodology: Lead; Supervision: Lead; Writing – original draft: Lead)
Romain Sanson (Formal analysis: Lead; Investigation: Lead; Methodology: Lead)
Silvia Luna Lazzara (Investigation: Equal; Methodology: Equal)
Caterina Luana Pitasi (Investigation: Supporting; Methodology: Supporting)
David Cune (Investigation: Supporting; Methodology: Supporting)
Coralie Trentesaux (Investigation: Supporting; Methodology: Supporting)
Marie Fraudeau (Investigation: Supporting; Methodology: Supporting)
Franck Letourneur (Methodology: Equal; Software: Equal)
Benjamin Saintpierre (Resources: Lead; Software: Lead)
Morgane Le Gall ( Software: Lead)
Pascale Bossard (Methodology: Supporting)
Benoit Terris (Methodology: Equal)
Pascal Finetti (Data curation: Equal; Resources: Equal; Software: Equal)
François Bertucci (Resources: Lead; Software: Lead; Writing – original draft: Supporting)
Emilie Mamessier (Resources: Equal; Software: Lead; Writing – original draft: Supporting)
Béatrice Romagnolo (Conceptualization: Equal; Funding acquisition: Lead; Supervision: Lead)
Data Availability Statement
Article info
Publication history
Published online: November 07, 2022
Accepted:
October 27,
2022
Received:
March 17,
2022
Footnotes
Conflicts of interest The authors disclose no conflicts.
Funding Supported by the National Institute of Health and Medical Research (France), Ligue Contre le Cancer (Equipe Labellisée LNCC 2020), Labex Who Am I? (France; R.S. and C.L.P.), Cancer Research Personalized Medicine, and a Ministère de la Recherche et de l’Enseignement Supérieur fellowship (R.S. and D.C.).
Copyright
© 2022 The Authors. Published by Elsevier Inc. on behalf of the AGA Institute.