Article Text
Abstract
Background Tumourigenesis in right-sided and left-sided colons demonstrated distinct features.
Objective We aimed to characterise the differences between the left-sided and right-sided adenomas (ADs) representing the early stage of colonic tumourigenesis.
Design Single-cell and spatial transcriptomic datasets were analysed to reveal alterations between right-sided and left-sided colon ADs. Cells, animal experiments and clinical specimens were used to verify the results.
Results Single-cell analysis revealed that in right-sided ADs, there was a significant reduction of goblet cells, and these goblet cells were dysfunctional with attenuated mucin biosynthesis and defective antigen presentation. An impairment of the mucus barrier led to biofilm formation in crypts and subsequent bacteria invasion into right-sided ADs. The regions spatially surrounding the crypts with biofilm occupation underwent an inflammatory response by lipopolysaccharide (LPS) and an apoptosis process, as revealed by spatial transcriptomics. A distinct S100A11+ epithelial cell population in the right-sided ADs was identified, and its expression level was induced by bacterial LPS and peptidoglycan. S100A11 expression facilitated tumour growth in syngeneic immunocompetent mice with increased myeloid-derived suppressor cells (MDSC) but reduced cytotoxic CD8+ T cells. Targeting S100A11 with well-tolerated antagonists of its receptor for advanced glycation end product (RAGE) (Azeliragon) significantly impaired tumour growth and MDSC infiltration, thereby boosting the efficacy of anti-programmed cell death protein 1 therapy in colon cancer.
Conclusion Our findings unravelled that dysfunctional goblet cells and consequential bacterial translocation activated the S100A11-RAGE axis in right-sided colon ADs, which recruits MDSCs to promote immune evasion. Targeting this axis by Azeliragon improves the efficacy of immunotherapy in colon cancer.
- colonic adenomas
- bacterial translocation
- colonic microflora
- immunotherapy
- cancer
Data availability statement
Data are available on reasonable request.
This is an open access article distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited, appropriate credit is given, any changes made indicated, and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/.
Statistics from Altmetric.com
WHAT IS ALREADY KNOWN ON THIS TOPIC
Left-sided and right-sided colon cancer differs in genetic features, prognosis and treatment response.
Neoplasia development varies according to the sidedness in the colon.
WHAT THIS STUDY ADDS
Mature goblet cells of right-sided adenomas (ADs) are depleted and dysfunctional in mucin biosynthesis and antigen presentation. Impairment of mucus barrier as decreased mucin expression and defective glycosylation leads to biofilm formation in the crypts and subsequent bacteria invasion into right-sided ADs.
The regions spatially surrounding the crypts with biofilm occupation undergo inflammatory response by lipopolysaccharide (LPS) and apoptosis process in right-sided ADs. The infiltrated immune cells in right-sided ADs experience inflammatory response against bacteria.
A distinct S100A11+ epithelial cell population in the right-sided ADs is identified and its expression level is induced by bacterial LPS and peptidoglycan in intestinal AD organoid cells. S100A11 expression facilitates the tumour growth of AD organoids and CT26 (p<0.01) allografts in syngeneic immunocompetent mice with increased myeloid-derived suppressor cells (MDSCs) but reduced cytotoxic CD8+ T cells.
Mechanistically, S100A11 acts extracellularly as a chemotactic signal for MDSC recruitment, promoting the growth of colon ADs and carcinomas in hosts through binding to the receptor for advanced glycation end products (RAGE).
HOW THIS STUDY MIGHT AFFECT RESEARCH, PRACTICE OR POLICY
Our findings indicate that targeting S100A11 with well-tolerated antagonists of its receptor RAGE (Azeliragon) significantly impairs tumour growth and MDSC infiltration and boosts the efficacy of anti-programmed cell death protein 1 therapy in colon cancer.
Introduction
Human colon is a single organ comprising two embryological origins with distinct physiological functions. Numerous studies suggested that left-sided and right-sided colon cancer differ in genetic features, prognosis and treatment response.1–3 Along the normal-adenoma-carcinoma axis, colon polyps represent the earliest stage of tumourigenesis. Polyps are primarily histologically divided into conventional adenomas (ADs) and serrated polyps (SPs).4 ADs are associated with mutations of adenomatous polyposis coli (APC) gene and WNT pathway activation, originating from stem cell expansion.4 SPs feature epigenetic disruption like MLH1 gene hypermethylation, and is more prevalent in the right-sided colon.5 This uneven distribution of SPs between left-sided and right-sided colon may mask the distinct characteristics of colonic polyps as a consequence of their sidedness. Thus, more work is needed to delineate the unique features of left-sided versus right-sided colon ADs in the context of congruent histological subtypes.
Gut microbial dysbiosis is a causative factor for tumourigenesis in colon.6 In particular, the biofilm formation, massive bacterial invasion of colonic mucus layer embedded in a matrix of extracellular polymeric substances, is associated with tumourigenesis.7 8 Biofilm formation enables bacteria to directly interact with colonic epithelium, instigating inflammatory response and oncogenic change.7 Of note, bacterial biofilms are prevalently observed in right-sided colon cancer, while rarely demonstrated in left-sided part.8 Currently, it is still unclear whether the sidedness preference of biofilm also exists in colonic polyps, the cause of its formation, and how it modulates the microenvironment in these precancerous lesions.
Therefore, we integrated single-cell RNA sequencing (scRNA-seq) and spatial transcriptomics to determine cellular composition and organisation of left-sided versus right-sided colonic ADs. We revealed that bacteria-induced S100A11 in neoplastic cells that is specific to right-sided colonic ADs, and deciphered S100A11 as a secretory signal for myeloid-derived suppressor cell (MDSC) recruitment and as a potential target for immunotherapy.
Materials and methods
Statistical analysis
All statistical analyses were conducted using R V.4.1.3 (https://www.r-project.org/) and GraphPad (V.9.0). Comparisons between two groups were performed by the unpaired two-tailed Student’s t-test or unpaired two-sided Wilcoxon rank-sum test. Multiple group comparisons were conducted using one-way analysis of variance (ANOVA) followed by multiple comparisons. A two-way ANOVA was used to compare growth curves. A two-sided p<0.05 was regarded as statistically significant.
Patient and public involvement
Patients and/or the public were not involved in the design, or conduct, or reporting or dissemination plans of this research.
Additional methods and details are provided in online supplemental methods and online supplemental tables S1 and S2.
Supplemental material
Supplemental material
Supplemental material
Results
Integrated scRNA-seq profiles of left-sided and right-sided colonic adenomas
To determine the single-cell profiles of colonic polyps in connection to their sidedness, we first compiled two single-cell transcriptomic datasets with 78 polyp samples from 44 patients (figure 1A), of which 46 samples were from right-sided colon (caecum, ascending colon and hepatic flexure) and the rest from left-sided colon (descending colon, sigmoid colon) (figure 1B). After quality control, a total of 75 559 cells were included, in which 30 274 cells and 45 285 cells were originated from left-sided and right-sided polyps, respectively. Highly variably expressed genes within these cells were used for principal component analysis (PCA) (online supplemental figure S1A). To mitigate batch effects, the scRNA-seq data were integrated by Harmony algorithm.9 Principal components corrected by Harmony were employed to generate the unified Uniform Manifold Approximation and Projection (UMAP) embedding space and perform graph-based clustering (online supplemental figure S1B). Cells were classified into three major cell types, including epithelial cells (EPCAM+),10 immune cells (PTPRC+)11 and mesenchymal stromal cells (COL1A1+)12 (online supplemental figure S1C). All major cell types originated from two datasets merged together without obvious bias (online supplemental figure S1D). These results indicated that integrated scRNA-seq datasets were suitable for further analysis.
Supplemental material
Colonic polyps were histologically categorised into conventional ADs and SPs. The distribution of epithelial cells was almost even across left-sided and right-sided ADs (47% vs 53%), while epithelial cells of SPs exhibited the enrichment in the right side (left vs right, 21% vs 79%) in line with previous research (figure 1C and online supplemental figure S1E).5 To decipher the alteration in polyps caused by colonic location only, SPs were excluded from further investigation. The remaining 55 792 epithelial cells from ADs were reclustered (figure 1D). Identified clusters were annotated to colonic cell types based on canonical marker genes, including mature colonocytes (SLC26A2, CA1, GUCA2A), mature goblet cells (mGCs) (MUC2, TFF3, SYTL2), stem cells (OLFM4, LGR5, ASCL2), transit amplifying cells (MKI67, TOP2A, PCNA), BEST4+ colonocytes (BEST4, CA7, OTOP2), enteroendocrine cells (CHGA, SCGN, MS4A8), tuft cells (PLCG2, LRMP, SH2D6) (figure 1E and online supplemental figure S1F). The composition of cell types was relatively homogenously distributed within left-sided and right-sided ADs. Of note, the right-sided ADs exhibited decreased mGCs, BEST4+ colonocytes and enteroendocrine cells, but increased proportions of stem cells and tuft cells (figure 1F). Altered cellular composition between left-sided and right-sided ADs was further validated by Ro/e analysis and Milo (figure 1G and H).13 14
Mature goblet cells of right-sided ADs are depleted and dysfunctional in mucin biosynthesis and antigen presentation
As mGCs showed the greatest variation according to ADs sidedness, goblet cell-associated fractions were subjected to clustering and UMAP visualisation (figure 2A). mGCs in right-sided ADs expressed a lower level of MUC2 (p<0.001) and reduced proportionally within the goblet cells cluster compared with that in left-sided ADs (p=0.007) (figure 2B and C). Since a fundamental function of mGCs is to synthesise and secrete mucin, mucin gene expression was examined in goblet cells of ADs. In right-sided ADs, both gel-forming mucins (MUC2, MUC5AC and MUC6) and membrane-associated mucins (MUC4, MUC12, MUC13 and MUC17) expression decreased, suggesting the deficiency of mucin synthesis in right-sided ADs (figure 2D). Apart from gene transcription, mucin biosynthesis involves glycosylation in the Golgi apparatus, oligomerisation and dense packing for storage in large granules before secretion.15 mGCs from right-sided ADs were defective in disulfide isomerase, glycosylation and secretion pathways (figure 2E), as evidenced by decreased expression of genes involved in protein transportation (ARF1 and TMED9) and glycosylation (GALNT6 and B3GNT7) compared with left-sided ADs (all p<0.001) (figure 2F). Sialylation of glycans on mucus is essential for its stability and integrity by protecting against bacterial degradation.16 Sialyltransferases expression in mGCs of right-sided ADs was largely reduced (ST6GALNAC1, ST6GALNAC6 and ST3GAL4) (all p<0.001) (figure 2G). Together, these results suggested that mucus as the intestinal barrier might be defective in right-sides ADs.
Aside from mucin biosynthesis, goblet cells could form goblet cell-associated antigen passages and transmit luminal substances to antigen-presenting cells to induce adaptive immune responses.17 To evaluate the antigen presentation capacity of goblet cell in ADs, genes relative to major histocompatibility complex (MHC) were determined. Intriguingly, there was a marked decrease in MHC-associated gene expression (B2M, HLA.A, HLA.B and HLA.C) in mGCs of right-sided ADs (figure 2H), together with attenuated antigen folding, assembly and presentation pathways (figure 2I).
Next, we determined the possible involvement of regulons in goblet cells in ADs using single-cell regulatory network inference and clustering analysis. Transcription factors ATOH1 and SPDEF governing goblet cell development showed higher regulator activity in mGCs of both left-sided and right-sided ADs without discernible difference (figure 2J). Notably, the regulator activity of KLF4 involved in terminal differentiation of goblet cells were reduced in mGCs of right-sided ADs (figure 2J).18 Instead, HNF4A and HNF4G activities were enhanced in mGCs with dysfunctional status in right-sided ADs (figure 2J). To some extent, this result was in line with previous findings that loss of HNF4A/4G with SMAD4 disruption promotes goblet cell identities.19 Considering the altered function status and regulon activity, we pooled the goblet cells with stem cells from left-sided and right-sided ADs together to explore the developmental process via pseudotime analysis. This analysis showed that the stem cells were at the beginning of the trajectory path, whereas mGCs were at a terminal state along the differentiation trajectory from progenitor to mature cell fates (figure 2K, left). In particular, mGCs from left-sided and right-sided ADs positioned at the distinct ends of the developmental trajectory, implying dissimilar differentiation process (figure 2K, right). Furthermore, we validated our findings in clinical ADs samples via immunohistochemical and Alcian blue (AB)/Periodic acid-Schiff (PAS) staining. Immunohistochemistry (IHC) confirmed diminished expression of goblet cell markers, namely MUC2 and TFF3 (p=0.008 and p=0.004), in right-sided ADs as compared with their left-sided counterparts (figure 2L). The sections of right-sided ADs showed smaller mucus vacuoles (p=0.008) and thinner inner mucus layer (p<0.001), as assessed by AB/PAS staining on the Carnoy’s fixed sample (figure 2M). Taken together, our results indicated the loss of goblet cells along with defective mucin biosynthesis and antigen presentation in right-sided ADs.
Biofilm formation and bacterial invasion in right-sided ADs lead to epithelial inflammation response
Given that mucus production by goblet cells hampering bacterial translocation and avoiding inflammatory pathologies, we hypothesised that defective mGCs might instigate inflammation in right-sided ADs. Supporting this notion, goblet cells proportion negatively correlated with inflammatory response caused by lipopolysaccharide (LPS) in colonic ADs (R=−0.3, p=0.04) (figure 3A). In line with this, most cell types were enriched in inflammatory response caused by LPS pathway in right-sided ADs (p<0.001) (figure 3B). These cells also underwent epithelial cell apoptotic process (p<0.001) (online supplemental figure S2A). Epithelial cell apoptotic process was positively associated with inflammatory response caused by LPS among ADs samples (R=0.59, p<0.001) and across different cell types (R=0.88, p=0.002) (online supplemental figure S2B and S2C). This implied that apoptosis in epithelial cells of ADs might be a consequence of inflammatory response mediated by bacterial translocation. To spatially visualise the bacteria-induced inflammation within ADs tissues, we performed spatial transcriptomics using the tissue section from five ADs samples, including two left-sided and three right-sided ADs samples. Single-sample Gene Set Enrichment Analysis (ssGSEA) analysis was employed to assess the enrichment of LPS-induced inflammatory response pathway within spots. Coloured spots based on enrichment score revealed increased LPS-induced inflammation predominantly in the interior of the right-sided ADs section with overall significantly higher enrichment (p<0.001) (figure 3C), suggesting that bacteria and their toxins that cross the intestinal barrier may instigate inflammation in right-sided ADs. Thus, we speculated that there was a higher bacterial burden within these tissues. Quantitative PCR (qPCR) targeting universal 16s ribosomal DNA (rDNA) indicated an increased abundance of bacteria in right-sided ADs (p=0.025) (figure 3D). Intriguingly, IHC staining of bacteria component LPS showed the bacterial aggregation as penetrating and occupying into the crypts of ADs (figure 3E). It appeared as dense communities of bacteria encased in a matrix in contact with epithelial cells, representing typical biofilm morphology. Biofilm presence was more prevalent among right-sided ADs compared with the left-sided counterpart (p=0.039) (figure 3E, top). The existence of biofilm in crypts correlated with increased bacterial tissue invasion in right-sided ADs (p=0.002) (figure 3E, bottom). Consistently, the spots around the crypts with biofilm exhibited high enrichment of LPS-induced inflammation and apoptosis pathways (p=0.035 and p=0.032). The decreased trend of the pathway enrichment was observed in the spots aways from the crypts (figure 3F). These results indicated that the cells spatially surrounding the crypts with biofilm occupation underwent the bacteria-induced inflammation in right-sided ADs. To further determine the alteration in the composition and relative abundance of gut microbiota, we sequenced the 16s rDNA V3–V4 regions from the tissues of left-sided and right-sided AD samples. PCA revealed that the bacterial composition in right-sided ADs was separated from left-sided counterpart (p=0.003) (figure 3G). At phylum levels, Proteobacteria displayed relative high abundance exclusively in right-sided AD samples, while Fusobacteria were rare (figure 3H). This was in line with previous research, suggesting the predominant presence of Proteobacteria and non-detectable Fusobacteria in biofilms of Familial adenomatous polyposis samples.7 Together, biofilm occupation in the crypts facilitated the bacterial translocation, leading to inflammatory response in right-sided ADs.
Supplemental material
Immune cell infiltration in ADs is modulated by bacteria in right-sided ADs
As immune cells are effectors of inflammation, we examined immune cell composition and functional status in ADs (figure 4A). Immune cells (PTPRC+) were increased in right-sided ADs (p=0.016) (figure 4B). We clustered immune cells into monocytes/macrophages (CD14, CSF1R, CSF3R and C1QB), T cells (CD3D, CD247, TRBC1 and TRAC), B cells (CD79A, CD19, PAX5 and IGHM) and mast cells (TPSB2, VWA5A, MS4A2 and CPA3) (figure 4C). Among them, T cells are predominant in ADs, demonstrating markedly increased in right-sided ADs compared with left-sided ADs (p=0.007) (figure 4D). In parallel, T cells in right-sided ADs were enriched in T cell extravasation signature, implying dependence on peripheral lymphocytes for replenishment (figure 4E). In right-sided ADs, T cells, mast cells and monocytes/macrophages all showed the enrichment in defence response to Gram-negative bacterium geneset, while B cells were enriched in antimicrobial humoral response and interleukin 1α production signature, in line with increased bacterial infiltration (figure 4E). Single-cell regulatory network inference and clustering analysis further revealed that inflammation-associated regulons were enriched in the immune cells of right-sided ADs (eg, RELA for T cells, STAT5A for mast cells, REL for B cells and IRF7 for monocytes/macrophages) (figure 4F). Collectively, these results indicated that immune cells were involved in inflammatory response against bacterial invasion in right-sided ADs.
As T cells were dominant component in ADs, these cells were reclustered and grouped into three subpopulations (T cell-1, T cell-2 and T cell-3) (figure 4G, upper). T cell-1 subset was elevated in right-sided ADs, accompanied by downregulated T cell-2 cluster (figure 4G, lower). We next explored the dynamic immune states and cell transitions in these cells by inferring the state trajectories using Monocle.20 T cell-2 and T cell-3 clusters were positioned at the beginning and the middle of the trajectory path, while T cell-1 cluster was located at terminal suggesting its differentiated status (figure 4H). Next, we compared differentially expressed genes (DEG) between T cell-1 cluster cells and remaining T cells. Intriguingly, LGALS3, LGALS4 and DEF5A gene expression were increased in T cells from T cell-1 cluster in right-sided ADs (figure 4I). LGALS3 and LGALS4 encode galectin-3 and galectin-4, respectively, which has been shown to interact with and agglutinate commensal and pathogenic bacteria.21 22 To validate the role of bacterial components on LGALS3 and LGALS4 expression in T cells, we performed bacterial LPS and peptidoglycan (PGN) treatment in Jurkat T cells. As shown in figure 4J, both LPS and PGN induced LGALS3 and LGALS4 expression in human Jurkat T cells in a dose-dependent manner (p<0.05) (figure 4J). These results suggested that bacterial existence in right-sided ADs might induce LGALS3 and LGALS4 expression in T cells exercising antibacterial activities.
S100A11 expression is induced by bacteria in right-sided ADs
Next, we focused on two clusters of epithelial cells lacking expression of canonical colonic epithelial cell markers in ADs (figure 5A and online supplemental figure S3A). These clusters are enriched in colon cancer signature compared with colonocytes and goblet cells (p<0.001), inferring they are neoplastic cells in ADs with malignant potential (online supplemental figure S3B). Thus, the cells in these two clusters were reclustered and visualised in UMAP plot. They were divided into five clusters that are uniformly distributed across the datasets and samples (figure 5B and online supplemental figure S3C). Intriguingly, subclusters 0 and 4 were mainly positioned in left-sided ADs and then annotated as Epi-L cluster. Subclusters 1, 2 and 3 were predominantly located in right-sided ADs and named as Epi-R cluster (figure 5B). Consistently, cells of Epi-L cluster and Epi-R cluster were enriched in left-sided and right-sided ADs, respectively, and confirmed by Milo analysis (figure 5C). To delineate the dissimilarity in Epi-L and Epi-R clusters, we performed DEG and pathway analysis (online supplemental figure S3D). Epi-R clusters are enriched in inflammation-related pathways and response to bacterium (figure 5D). Hallmark pathway analysis further showed the glycolysis and epithelial mesenchymal transition (EMT) pathway were enriched in Epi-R cluster (online supplemental figure S3E). Of note, the score of glycolysis and EMT pathway in Epi-R cluster were highest among other cell types in epithelial component of ADs (all p<0.001) (online supplemental figure S3F and S3G), further suggesting its increased malignant potential. Hence, the Epi-R cluster, predominantly distributed in right-sided ADs, was enriched in inflammation response and EMT process.
Supplemental material
To further characterise the Epi-L and Epi-R clusters, we identified the distinct markers of these cells. The cells in Epi-R cluster specially expressed DUOX2, SORCS2, ANPEP, TMEM132C, S100A11 and ANXA2 (figure 5E). To determine the effect of bacteria on these genes, we constructed organoid models derived from intestinal ADs of ApcMin/+ mice (figure 5F and online supplemental figure S4A and S4B). Treatment of these organoids with bacterial LPS and PGN upregulated DUOX2 (encoding dual oxidase 2) expression (online supplemental figure S4C), consistent with previous reports (p<0.01).23 Notably, S100A11 mRNA was also elevated with LPS and PGN treatment (p<0.05) (figure 5G). Induction of S100A11 by LPS and PGN was validated by flow cytometry analysis (p<0.05) (figure 5H). For further verification, biofilm-forming enteroaggregative Escherichia coli (EAEC) was administered to ApcMin/+ mice with or without antibiotic cocktail treatment (figure 5I). EAEC-induced biofilm was visualised by fluorescence in situ hybridisation (FISH) using a bacterial 16s rRNA probe (EUB338). Co-staining with S100A11 revealed that its expression was upregulated in biofilm-positive AD crypts from EAEC-treated ApcMin/+ mice, an observation abrogated in antibiotics-treated groups (all p<0.01) (figure 5J). To further verify that biofilm-induced S100A11 expression is sidedness-specific, we used ApcMin/+ mice treated with 2% dextran sulfate sodium (DSS), which developed tumours along the proximal-distal axis of the colon (figure 5K). FISH detection using a 16s rRNA probe revealed that biofilm formation was more prevalent in tumours from proximal colon in mice (p=0.03) compared with those from distal colon (figure 5L and M), mirroring widespread occurrence of bacterial biofilm in human right-sided ADs. In line with this, S100A11 level was increased in the proximal colon tumours as compared with distal counterparts (p=0.009) (figure 5N). Of note, S100A11 expression was exclusively elevated in biofilm-residing tumours from the proximal colons (p=0.014) (figure 5O), suggesting that bacterial biofilm might mediate S100A11 expression in proximal colon tumours. Furthermore, to validate the increased S100A11 expression specifically in the human right-sided ADs, we integrated the Epi-L and Epi-R cluster cells with the epithelial cells from the colon tissue samples in HS and HV datasets for further analysis (online supplemental figure S5A). Cells in colon tissue exhibited the higher expression of canonical markers, while demonstrating markedly lower S100A11 expression compared with the Epi-R and Epi-L cluster cells (online supplemental figure S5B and S5C). In particular, no significant alteration was observed in S100A11 expression in cells from left-colon and right-colon tissues (online supplemental figure S5C). Consistently, tumour-specific elevation of S100A11 was demonstrated in the right-sided ADs in our cohort, as determined by reverse transcription-qPCR, ELISA and spatial transcriptomics (all p<0.01) (figure 5P, online supplemental figure S6A and S5D). Intriguingly, there was a significant correlation between bacterial 16s and S100A11 expression in right-sided ADs (r=0.671, p=0.016) but not in left-sided ADs (r=−0.083, p=0.778) (figure 5Q). Taken together, these results implied that the existence of bacteria and biofilm might induce tumour-specific S100A11 expression in right-sided ADs.
Supplemental material
Supplemental material
Supplemental material
S100A11 suppresses antitumour immunity through MDSC recruitment
To probe the potential role of S100A11 in tumourigenesis, we overexpressed S100A11 in murine AD organoids (lower endogenous S100A11 expression), and accordingly performed S100A11 knockdown in CT26 colon carcinoma cell line (higher endogenous S100A11 expression) (online supplemental figure S6B, figure 6A, B and E). S100A11 overexpression had no significant effect on cell proliferation (figure 6C). Nevertheless, S100A11 overexpression promote the tumour growth of AD organoids in immunocompetent C57BL/6 hosts (p=0.002), implying that S100A11 might suppress antitumour immune responses to promote tumourigenesis (figure 6D). Intriguingly, ELISA results revealed that S100A11 levels in the overexpression allograft tumours could mimic the physiologically higher S100A11 levels in the human right-sided ADs (online supplemental figure S6A and S6C). Furthermore, immunosuppressive role of S100A11 was validated in CT26 allografts. We observed significantly slower tumour progression of S100A11-KD CT26 cells in wild-type BALB/c hosts (p=0.001), but not in immunodeficient hosts (figure 6F and G). Taken together, these results suggested that S100A11 promote tumour growth of colon AD and carcinoma through impairing antitumour immunity.
Some S100 family members, such as S100A8 and S100A9, are secreted extracellularly and promote the chemotaxis of immune cells, such as MDSCs.24 We thus hypothesised that S100A11 might drive MDSC infiltration. Flow cytometry showed that MDSC infiltration were induced in AD organoid allografts with S100A11 overexpression (p=0.001) (figure 6H and online supplemental figure S 7). IHC staining results indicated that elevated MDSC level was observed in the colonic tumours from biofilm-forming EAEC-treated ApcMin/+ mice in line with the increased S100A11 levels (p=0.008) (online supplemental figure S6D). Accordingly, knockdown of intrinsic S100A11 level led to decreased MDSC infiltration in AD organoid tumours and CT26 tumours (all p<0.01) (figure 6H and online supplemental figure S6F and S6E). Particularly, granulocytic MDSCs were reduced in CT26 S100A11-KD allograft tumours (p=0.002), while no significant change was found for monocytic MDSCs (online supplemental figure S6G). MDSC dampens antitumour immune response by suppressing T cell-mediated immune responses.25 In coincidence with this, activated and cytotoxic T cells were decreased in S100A11-OE organoid allografts (all p<0.05), while they were significantly elevated in CT26 allografts with S100A11-KD (all p<0.05) (figure 6I). To examine whether secreted S100A11 was chemotactic signal for MDSC recruitment, we performed in vitro transwell assays. Conditional medium from CT26 cells enhanced MDSC migration compared with that of organoid cells (p<0.001), which correlates with higher S100A11 expression and MDSC infiltration in CT26 allograft tumours (all p<0.05) (online supplemental figure S6J, S6C and S6H). Furthermore, conditional medium from AD organoid cells with S100A11 OE promoted MDSC migration (p< 0.05), an effect was blunted by S100A11 neutralising antibody (p=0.005) (figure 6J and online supplemental figure S6I). S100A11 neutralising antibody also abrogated MDSC migration in conditioned medium from CT26-shNC (p =0.007), but not in CT26-shS100A11 cells (figure 6J). Thus, S100A11 functions as chemotactic signal for MDSC recruitment. We next investigated the functional role of S100A11-mediated MDSC recruitment in vivo. Administration of S100A11 neutralising antibody inhibited growth of CT26-shNC allografts, as evidenced by reduced tumour volume (p<0.001) and weight (p=0.008), but had no effect on S100A11-KD tumours (figure 6K). Flow cytometry showed that S100A11 neutralisation antibody suppressed MDSC infiltration in controls but not in S100A11 knockdown tumours (figure 6L). This further supports the role of S100A11 in MDSC recruitment and facilitating tumour growth.
Supplemental material
S100A11-RAGE axis mediates MDSC infiltration in colonic adenoma and carcinoma as therapeutic target in combination with anti-PD1 therapy
S100 family proteins are small calcium binding proteins with high structural homology.26 Several S100 members including S100A11 are ligands of the receptor for advanced glycation end products (RAGE).27 S100A8/A9 can bind to RAGE expressed on MDSC to promote intratumoral accumulation.24 We thus examined whether S100A11-RAGE axis mediated recruitment of MDSC in colonic AD and carcinoma. The RAGE antagonist FPS-ZM1 (5 mg/kg) impaired the growth of AD organoid allografts with S100A11 overexpression, as evidenced by reduced tumour volume (p=0.003) and weight (p=0.003) (figure 7A). Flow cytometry revealed FPS-ZM1 suppressed MDSC infiltration induced by S100A11 overexpression in AD organoid allografts (p=0.019) (figure 7B). In CT26 allografts, FPS-ZM1 inhibited the tumour growth of shNC allografts (p=0.037) without significant effects on S100A11-KD tumours (figure 7C). Consistently, reduced MDSC caused by FPS-ZM1 treatment was observed in shNC allografts (p=0.002) but not in S100A11-KD group (figure 7D). These results suggested that RAGE mediated the S100A11-induced MDSC recruitment in colonic AD and carcinoma. To further verify this, FPS-ZM1 were applied to in vitro transwell assays. FPS-ZM1 suppressed the enhanced migration of MDSC in conditioned medium from S100A11-OE AD organoids (p<0.001) (figure 7E). Similarly, the MDSC migration was abrogated in conditioned medium of CT26-shNC (p=0.003), but not in S100A11-KD group (figure 7E). These were consistent in vivo results, further supporting that S100A11-induced MDSC recruitment depends on RAGE.
As MDSCs are the major barrier for tumour immunotherapy, targeting S100A11-RAGE axis might be potential approach to inhibit MDSC infiltration and boost immunotherapy efficacy. Indeed, S100A11 knockdown plus anti-PD1 exerted synergistic effects in suppressing tumour growth as compared with single treatment, leading to tumour regression (p<0.01) and reduction in tumour weight (p<0.05) (figure 7F). Flow cytometry revealed S100A11 knockdown plus anti-PD1 synergised to reduce MDSC and promote activated and cytotoxic T-cell infiltration (figure 7G). We further examined whether targeting S100A11-RAGE axis using RAGE antagonist have synergistic effect in combination with anti-PD1. The results showed that FPS-ZM1 plus anti-PD1 synergistic suppressed tumour progression in mice (p<0.001) (figure 7H). Another RAGE antagonist Azeliragon is orally active and well-tolerated as a granted orphan drug for patient with glioblastoma. Oral administration of Azeliragon (4 mg/kg) suppressed tumour growth by ~50% in CT26 allografts without body weight reduction in hosts (figure 7I and online supplemental figure S6K), proving it as effective and safe medication for CRC. Importantly, Azeliragon plus anti-PD1 exerted synergistic effects as compared with single treatment, leading to tumour regression (p<0.05) and reduced tumour weight (p<0.05) (figure 7I). A combination of Azeliragon and anti-PD1 synergised to reduce MDSC while promoting cytotoxic T-cell infiltration (figure 7J). These results implied that targeting S100A11-RAGE axis with Azeliragon was promising in combination with anti-PD1 therapy.
Discussion
This study focused on the implication of sidedness in cellular alternation and development of colonic ADs. Right-sided ADs showed goblet cell depletion and dysfunction in mucin biosynthesis and antigen presentation, implying the impaired barrier function. Consequently, biofilm formation and bacteiral invasion were observed predominantly in the crypts and its surrounding tissues of right-sided ADs. Specific cell cluster Epi-R featuring S100A11 expression was identified in right-sided ADs. Bacterial components LPS and PGN induced S100A11 expression in tumour cells, functioning as secretory signal for MDSC recruitment.
As one of the earliest signs of colon tumourigenesis, the disruption of mucosal homeostasis is characterised by expansion of proliferative crypt compartment, accompanied by a delay or inhibition in normal cellular differentiation.28 The loss of MUC2 expression, goblet cell depletion and increased tight junction permeability associated with the transformation of colonic epithelium all compromise barrier function and promote bacterial penetration.29–32 Numerous studies have implicated individual bacteria in CRC tumourigenesis,33 while few researches linked the organised bacterial community namely biofilm to tumour progression. It was reported that human colon mucosal biofilms, either from healthy or colon cancer hosts, were carcinogenic.34 In human CRC, bacterial biofilms were prevalent in right-sided tumours.8 It seems that the extents of biofilm-epithelium interactions varied across sample types and disease status. Biofilm invasion into mucus layer was usually confined on the surface of colonic epithelium in human CRC samples,8 while it was observed in the colonic crypts of murine colitis model.35 In this study, bacterial biofilms adhered tightly to the bottom of crypts of the right-sided ADs, identified as previously unremarked pattern (figure 3E). Furthermore, biofilm formation allows bacteria to grow in close interaction with intestinal epithelial cells, providing the essential condition for bacterial invasion and instigating inflammatory responses.8 Accordingly, we observed the bacterial invasion in the tissue regions surrounding the crypts with biofilm formation (figure 3E). Multiple cell types in right-sided ADs exhibited inflammatory response against bacteria (figure 3B). Intriguingly, the neoplastic cells of Epi-R clusters, primarily found in right-sided ADs, displayed enrichment of epithelial-mesenchymal transition pathways (online supplemental figure S3E). The epithelial-mesenchymal process, recognised as the hallmarks of cancer, reveals that its transcriptional features could be induced by bacteria.36 This implied that bacterial existence may promote the malignant transformation in right-sided ADs. Taken together, biofilm formation may be associate with right-sided tumour progression.
Colonic mucus is mainly composed of MUC2 mucin, a glycoprotein with glycans.37 Mucins are mainly biosynthesised, glycosylated and secreted by goblet cells. Our study revealed that mucin biosynthesis and glycosylation were impaired in goblet cells of the right-sided ADs, evidenced by the decreased expression of mucins and enzymes involved in glycosylation (figure 2D, E and F). Particularly, sialyltransferase (ST6GALNAC1, ST6GALNAC6 and ST3GAL4) expression involved in mucin sialylation was decreased in goblet cells of the right-sided ADs (figure 2G). Sialylation is the terminal addition of sialic acid (SA) to glycans, as a form of glycosylation occurring on mature mucin proteins.16 Intriguingly, ST6GALNAC1 mutant mice demonstrated a marked thinning of the inner mucus layer in the colon,16 implying the presence of sialylated mucin in the inner layer of mucus. Furthermore, recent studies demonstrated that mucin glycosylation restrain the bacterial biofilm formation.38 The colonic mucus of mice lacking ST6GALNAC1/6 was less sialylated, thinner and more permeable to microbiota.16 In line with this, our study showed that bacterial invasion and biofilm formation were prevalent in the tumour crypts of right-sided ADs, corresponding to their reduced sialyltransferase levels (figure 2G). More studies are needed to demonstrate the distribution of glycosylated mucin in the inner mucus layer and its role in disrupting biofilm. Since the bottom of the colonic crypt harbours stem/progenitor cells in undifferentiated state, it is crucial to prevent the bacterial biofilm formation and subsequent inflammation response in crypts of right-sided ADs due to their potential for malignant transformation.
In our study, we identified the specific epithelial subpopulation named Epi-R featuring S100A11 expression, predominantly found in right-sided ADs (figure 5C and E). S100A11 belongs to damage-associated molecular patterns, the proteins rapidly released by the host during infection.27 Consistently, we found that bacterial component LPS and PGN could induce S100A11 expression in the colonic AD organoids cells (figure 5G). DSS-treated ApcMin/+ mice showed exacerbated biofilm formation in proximal colon tumours, together with elevated S100A11 (figure 5L). DSS causes colitis, which promotes tumourigenesis in colon and has diverse effects on colonic mucosa, including impaired tight junction and mucin production,39 depleted tissue-adhesion bacteria40 and intestinal epithelial cell apoptosis.41 Biofilm formation might also be uneven due to differential exposure of colonic mucosa to DSS. Nevertheless, administration of biofilm-forming EAEC to ApcMin/+ validated that biofilm promotes S100A11 upregulation in colon even in the absence of DSS treatment (figure 5J). Furthermore, previous research highlights the intracellular function of S100A11 in plasma membrane repair.42 Our study demonstrated the extracellular role of S100A11 as the chemotactic signal for MDSC recruitment (figure 6H and J). The extracellular function of S100A11 was mediated by its receptor RAGE, evidenced by its antagonist FPS-ZM1 suppressed the S100A11-induced MDSC recruitment (figure 7A, C and E). Finally, we revealed that another RAGE antagonist Azeliragon attenuated the MDSC infiltration and boosted anti-PD1 efficacy in colon cancer (figure 7I and J). Azeliragon is orally active and well-tolerated, granted as the orphan drug for glioblastoma. Thus, Azeliragon combined with anti-PD1 therapy shows promising translational value to clinical practice. We suggest that orally available RAGE antagonist Azeliragon might have potential therapeutic effects on the right-sided ADs. The therapeutic value of orally available RAGE antagonist Azeliragon needs the clinical trials for further investigation.
In summary, our work suggests impaired mucus barrier caused by dysfunctional goblet cells leads to bacterial biofilm formation and sequential inflammatory response in right-sided colon ADs. S100A11 induced by bacterial components exerts immunosuppressive effects by MDSC recruitment through S100A11-RAGE axis. RAGE antagonist Azeliragon may have alternative application for immunotherapy (figure 8).
Data availability statement
Data are available on reasonable request.
Ethics statements
Patient consent for publication
Ethics approval
This study was approved by Medical Ethics Committee of NanFang Hospital of Southern Medical University (NFEC-2023-588). All the animal work is approved by the Institutional Animal Care and Use Committee (IACUC) of Nanfang Hospital (Guangzhou, China) (IACUC-LAC-20231007-011 and IACUC-LAC-20240322-004). For human samples, informed consent was obtained for all patients.
Acknowledgments
This work was supported by National Natural Science Foundation of China (no. 82372864, 82373240 and 82172960), Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Cancer (no. 2020B121201004), Key‐Area Research and Development Programme of Guangdong Province (no. 2021B0101420005), President Foundation of Nanfang Hospital, Southern Medical University (no. 2023Y001), Guangdong Basic and Applied Basic Research Foundation (no. 2023A1515110178) and China Postdoctoral Science Fund (no. 2024M751339 and 2024T170388). Key Clinical Technique of Guangzhou (2023P-ZD01). The authors thank Dr Jianming Zeng (University of Macau), and all the members of his bioinformatics team, biotrainee, for generously sharing their experience and codes. The authors also thank Dr Shi Yu for drawing the illustration in BioRender (https://www.biorender.com/). Images in figure 1A, 5F, 5I, 5K, 6A, 6E, 6J and figure 8 were created with BioRender.com.
References
Supplementary materials
Supplementary Data
This web only file has been produced by the BMJ Publishing Group from an electronic file supplied by the author(s) and has not been edited for content.
Footnotes
Contributors QZ, LL and JC (Cheng J) contributed equally to this work. QZ performed the bioinformatic analysis and drafted the manuscript. LL, JC (Cheng J) and JC (Chen J) conducted the organoid culture and animal experiments. JC (Chen J) and YD conducted the IF and IHC staining. BZ performed the 16s rRNA sequencing analysis. XZ, QL, CL and HD assisted in the experiments. CCW, BZ, GL and XB designed and supervised the project and revised the manuscript. XB acted as guarantor.
Funding Key Clinical Technique of Guangzhou (2023P-ZD01)
Competing interests None declared.
Patient and public involvement Patients and/or the public were not involved in the design, or conduct, or reporting, or dissemination plans of this research.
Provenance and peer review Not commissioned; externally peer reviewed.
Supplemental material This content has been supplied by the author(s). It has not been vetted by BMJ Publishing Group Limited (BMJ) and may not have been peer-reviewed. Any opinions or recommendations discussed are solely those of the author(s) and are not endorsed by BMJ. BMJ disclaims all liability and responsibility arising from any reliance placed on the content. Where the content includes any translated material, BMJ does not warrant the accuracy and reliability of the translations (including but not limited to local regulations, clinical guidelines, terminology, drug names and drug dosages), and is not responsible for any error and/or omissions arising from translation and adaptation or otherwise.