Skip to main content

Bifidobacterium adolescentis induces Decorin+ macrophages via TLR2 to suppress colorectal carcinogenesis

A Correction to this article was published on 19 August 2023

This article has been updated

Abstract

Background

The interplay between gut microbiota and tumor microenvironment (TME) in the pathogenesis of colorectal cancer (CRC) is largely unknown. Here, we elucidated the functional role of B. adolescentis and its possible mechanism on the manipulation of Decorin+ macrophages in colorectal cancer.

Methods

The relative abundance of B. adolescentis in tumor or para-tumor tissue of CRC patients was analyzed. The role of B. adolescentis was explored in the CRC animal models. The single cell-RNA sequencing (scRNA-seq) was used to investigate the myeloid cells subsets in TME. The expression level of TLR2/YAP axis and its downstream Decorin in macrophages were tested by Western blot and qRT-PCR. Knockdown of Decorin in Raw264.7 was performed to investigate the effect of Decorin+ macrophages on subcutaneous tumor formation. Multi-immunofluorescence assay examined the number of Decorin+ macrophages on the CRC tissue.

Results

We found that the abundance of B. adolescentis was significantly reduced in tumor tissue of CRC patients. Supplementation with B. adolescentis suppressed AOM/DSS-induced tumorigenesis in mice. ScRNA-seq and animal experiment revealed that B. adolescentis increased Decorin+ macrophages. Mechanically, Decorin was activated by TLR2/YAP axis in macrophages. The abundance of B. adolescentis was correlated with the number of Decorin+ macrophages and the expression level of TLR2 in tumor tissue of CRC patients.

Conclusions

These results highlight that B. adolescentis induced Decorin+ macrophages and provide a novel therapeutic target for probiotic-based modulation of immune microenvironment in CRC.

Introduction

Colorectal cancer (CRC) ranks third in incidence and mortality globally [1]. Gut microbes are closely bound up with CRC via immune cells [2]. Accumulating evidence suggested that several special bacteria connected with the initiation and progression of CRC through immune pathway [3,4,5]. For instance, Helicobacter hepaticus induces T follicular helper cells and tertiary lymphoid structures to against CRC [6]. Among all the immune cells, macrophages are regarded as a critical part interacting with the intestinal flora in CRC. The gut microbiota regulates monocyte-like macrophages accumulation in a chemokine dependent manner and mediates an inflammatory response to facilitate colitis-associated tumorigenesis [7]; Tryptophan metabolites derived from microbes activate the aryl hydrocarbon receptor of tumor-associated macrophages to suppress anti-tumor immunity [8]. Accordingly, utilizing the flora to regulate macrophages is a potential therapeutic strategy in CRC [9].

Bifidobacterium plays a critical role in immune maturity and regulation [10,11,12]. Studies suggested that Bifidobacterium was associated with enhanced efficacy of checkpoint blockade immunotherapy [13, 14]. For example, cocktail of Bifidobacterium (B. breve and B.longum) promotes anti-tumor immunity and improves anti-PD-L1 efficacy [15]. Intratumoral accumulation of Bifidobacterium cocktail (B. bifidum, B. longum, B. lactis and B.breve) facilitates CD47-based immunotherapy via STING signaling [16]. Bifidobacterium pseudolongum enhances the anti-CTLA-4 therapy response by producing the metabolite inosine [17]. Most of these studies on cancer focus on Bifidobacterium cocktail and checkpoint blockade immunotherapy. However, the potential role of B.adolescentis on initiation and progression of CRC remains poorly understand. Our previous work indicated that B.adolescentis ameliorated chronic colitis by regulating immune response and gut microbiota remodeling [18].

In this study, we observed that supplementation with B.adolescentis suppressed colorectal tumorigenesis. Mechanically, we uncovered that B. adolescentis induced Decorin+ macrophages infiltration in TME and Decorin in macrophages was a crucial molecule mediating the anti-tumor effect of B.adolescentis. In addition, B. adolescentis increased Decorin+ macrophages via a TLR2/YAP-dependent signaling. Our results highlighted the protective role of B.adolescentis on CRC, which provided a new therapeutic strategy based on probiotics modulation of tumor immune microenvironment.

Methods

Cell culture

Human colorectal cancer cell line (HCT116), mouse colorectal cancer cell line (CT26), human macrophage cell line (THP-1) and mouse macrophage cell line (Raw264.7) were purchased from American Type Culture Collection (ATCC). HCT116 cultured in Maccoy 5A (Genom, China) supplemented with 10% FBS (Sijiqing, China) at 37 °C in a humidified 5% CO2 atmosphere. CT26, THP-1 and Raw264.7 cultured in RPMI 1640 (GIBCO, China) supplemented with 10% FBS (Sijiqing, China) at 37 °C in a humidified 5% CO2 atmosphere.

Bacterial strains and culture conditions

B. adolescentis was purchased from American Type Culture Collection (ATCC 15703). B. adolescentis grown in Reinforced Clostridium Medium (BD Difco, USA) was cultured under an atmosphere of 10% H2, 10% CO2, and 80% N2 in an AW500SG anaerobic workstation (ELECTROTEK) at 37 °C. The E. coli strain MG1655 (ATCC 700926) was cultured in Luria–Bertani medium overnight at 37 °C.

DNA extraction and bacteria DNA quantification

Genomic DNAs extraction and purification from clinical or mice tissue were executed by QIAGEN DNA Mini Kit (Qiagen, Germany) according to the instructions. Quantitative real-time PCR was performed to assess the relative abundance of B. adolescentis. The abundance of B. adolescentis was calculated by -ΔCt method and the universal Eubacteria 16s was used as an internal reference gene. Primers used are listed in the Supplementary Table S1.

Human specimens

A total of 65 paired fresh tumor and adjacent non-tumor tissue were obtained from patients with primary CRC who had not received preoperative anti-tumor and antibiotic treatment from Sir Run Run Shaw Hospital of Zhejiang University (Hangzhou, China). All samples were frozen in liquid nitrogen until use. Clinical Research Ethics Committee of the Sir Run Shaw Hospital, Zhejiang University School of Medicine approved the protocol. All aspects of the study were conducted in accordance with the principles of the Declaration of Helsinki.

Carcinogen-induced cancer model

6-week-old male C57BL/6 wildtype mice were obtained from Shanghai SLAC Laboratory Animal, China. Before bacterial intragastric administration, as previous study reported, 2 mg/mL streptomycin (Cat. #MB1275, Meilunbio) solubilized in water was given to mice for 7 days to ensure the conformance of regular microbiota and facilitate B. adolescentis colonization. C57BL/6 mice were given one single intraperitoneal injection of carcinogen azoxymethane (AOM, Sigma) (10 mg/kg per mouse), and then were given 5 successive days of 2.5% dextran sodium sulfate (DSS, MP Biomedicals) in the drinking water, following by regular drinking water for 2 weeks. This cycle was then repeated for twice [19]. Two weeks later, mice in B.adolescentis and E. coli groups were administrated with 1 × 109 CFU B.adolescentis or E. coli suspended in 200 μL sterile PBS every day, and the control group was administered with PBS. After about 3 months, mice were sacrificed and the colorectum was surgically excised for further analysis.

Animal use and care

All animal studies were approved by the Institutional Animal Care and Use Committee (IACUC) of Zhejiang University (ZJU). All animal experiments strictly adhered to protocols, policies, and ethical guidelines formulated by our IACUC. BALB/c nude mice, BALB/c mice and C57BL/6 mice were purchased from Shanghai SLAC Laboratory Animal, China. All mice were maintained in ventilated cages with 12 h light/dark cycles, constant temperature and humidity, enriched water and ad libitum feeding under specific pathogen-free (SPF) conditions.

Subcutaneous tumor models

Male BALB/c nude mice (4 weeks old) were reared in specific-pathogen-free (SPF) facilities. HCT116 cells were mixed with B.adolescentis, E.coli or PBS (MOI of 10:1) and macrophages (THP-1 or Raw264.7; cancer cells: macrophages = 1:1) with 50 μL Matrigel matrix (Cat. #354234,Corning Biocoat), and hypodermically injected into BALB/c nude mice (100 μL, 2 × 106 cells/per mouse). For BALB/c mice (6-8 weeks old), CT26 cells were mixed with B.adolescentis, E.coli or PBS (MOI of 10:1) and Raw264.7 (CT26: Raw264.7 = 10:1) with 50 μL Matrigel matrix (Cat. #354234,Corning Biocoat), and hypodermically injected into BALB/c mice (100 μL, 2 × 106 cells/per mouse). After 6-7 days inoculation, the tumor volume of the mice was examined regularly and calculated as follows: Volume = 0.54 × L × W2, where L is the longest diameter and W is the shortest diameter. For TLR2 inhibition, 3 mg/kg Cu-CPT22(Selleck Cat. #S8677) was injected intraperitoneally to mice every two days until sacrifice. For YAP inhibition, 50 mg/kg Verteporfin (MCE Cat. #CL 318952) was injected intraperitoneally to mice every day until sacrifice.

Isolation of primary macrophages

For primary mouse macrophages BMDMs, bone marrow cells were harvested from 8-10 weeks old C57BL/6 mice as previously done [20]. 6 days later, BMDMs were confirmed by flow cytometry for CD11b and F4/80 and further used. For primary human macrophages, after isolating the CD14+ monocytes from PBMC using the MojoSort Human CD14 Selection Kit (Biolegend Cat No:480025) according to the manufacturer’s protocol, we cultured them in RPMI 1640 supplemented with 10% (v/v) FBS, penicillin/streptomycin, and 50 ng/mL M-CSF for 5 days to induce macrophages differentiation [21]. For polarization of M1 macrophages, BMDMs and THP-1 cells were treated by 100 ng/mL IFN-γ (Cat. #C746, Novoprotein) for 24 h as previous study reported [22], then the expression of aimed genes were examined by qRT-PCR.

Cytotoxicity experiments

For cytotoxicity experiments, BMDMs were pretreated with B.adolescentis, E. coli or vehicle (PBS) for 24 h, and then co-cultured with HCT116 or CT26 (BMDMs: tumor cells = 1:1) for 24 h. The level of LDH was evaluated according to the manufacturer’s instructions [23].

Isolation of colorectal lamina propria cells

Isolation of colorectal lamina propria cells was performed as previously done [20]. In brief, the posterior 1/3 of the colorectum without lymph and adipose tissue was cut into small pieces and washed in RPMI 1640 medium (GIBCO, China). The tissue was incubated in D-Hanks (Cat. #MA0039, Meilunbio) buffer supplemented with 1 mmol/L DTT (Cat. #MB30471, Meilunbio) and 5 mmol/L EDTA (Cat. #MB2514, Meilunbio) on a shaker (200 rpm) for 30 min at 37℃. And then the remaining colorectum was cut into 1 mm pieces and further digested in Hanks buffer (Cat. #MA0041, Meilunbio) supplemented with 1 mg/mL Type IV collagenase (Cat. #A005318, Sangon Biotech) for 30 min at 37℃ with 200 rpm shaking. After complete digestion, the cell suspension was passed through a 300-mesh filter and then centrifuged at 500 g for 5 min. The isolated colorectal lamina propria cells were further analyzed.

Flow cytometry analysis

Cells were counted (5 × 106 cells/per sample) and stained for 20 min at 4℃ Fixable viability stain 510 (Cat. #564406, 2 mg/mL, BD Biosciences) for live cell staining, then the surface molecules of the cells were stained with antibodies for 30 min at 4℃. The panel of antibodies designed to analyze of myeloid cells: Alexa Fluor 700-CD45 (Cat. #560510, Clone 30-F11, 1 mg/mL, BD Biosciences), BV421-F4/80 (Cat. #565411, Clone T45-2342, 1 mg/mL, BD Biosciences), BV605-CD11b (Cat. #557672, Clone M1/70, 1 mg/mL, BD Biosciences), PE-MHC-II (Cat. #557000, Clone M5/114, 1 mg/mL, BD Biosciences), PE-CY7-CD11c (Cat. #117317, Clone N418, 1 mg/mL, BioLegend) and APC-CD206 (Cat. #17-2061-82, Clone MR6F3, 2 mg/mL, eBioscience). Samples were analyzed using Flow Cytometer (BD Biosciences). Subsequent analysis was performed with FlowJo software (Tree Star Inc.).

Cell culture in presence of B.adolescentis

BMDMs, THP-1 cells or Raw264.7 cells were seeded in 6-well plate at a density of 5 × 105 cells per well and cultured in DMEM or RPMI  1640 medium with 10% (vol/vol) FBS overnight. BMDMs, THP-1 cells or Raw264.7 cells were co-cultured with B.adolescentis at a MOI = 100:1 for 24 h. Finally, the protein and RNA of BMDMs, THP-1 cells or Raw264.7 cells were extracted for analysis.

RNA extraction and quantitative qRT-PCR

RNAs were extracted from macrophages or tissue samples using Trizol reagent (Invitrogen, USA), and total RNA were reversed by Evo M-MLV RT Kit (Accurate Biology, China) according to the manufacturer’s instructions. Quantitative RT-PCR analysis was performed in triplicate in ROCHE LightCycler480 System (Rotor gene 6000 Software, Sydney, Australia) with SYBR Premix Ex Taq (Takara, Japan). Relative abundance was demonstrated by -ΔCt method. Primers used are listed in the Supplementary Table S1.

Genomics library and single cell RNA sequencing

Primary Sequencing data were demultiplexed and converted to FASTQ format by Illumina bcl2fastq software. Cell Ranger pipeline (version 3.1.0) was used to perform sample demultiplexing, barcode processing and single-cell 3, gene counting. Then scRNA-seq data were aligned to Ensemble genome GRCm38 reference genome. After filtering and UMI (universal molecular identifier) counting, a total of 18,661 single cell captured from 2 samples were captured using 10 × Genomics Chromium Single Cell 3’ Solution. The mean reads per cell was 49,151 with median UMI Counts per Cell of 5,790 after aggregation.

All bioinformatics analysis was performed using R version 3.4.0 (R Foundation, https://www.rproject.org) and RStudio version 1.3.1093 (https://www.rstudio.com).

To further identify sub-clusters within myeloid cell type, we re-analyzed cell type annotated as myeloid cells. Specifically, subset of Seurat object was extracted from the original expression matrix by default Seurat function, then we re-performed dimensionality reduction and PCA analysis and generate clusters as described above. Sub-cell types identified from sub-clustering of the myeloid cells were performed by “SingleR” package with “ImmGen” as the reference database, which identified Neutrophils, Macrophages, Monocytes, DC, Basophils cells. Finally, differentially expressed genes between PBS and B.a group in each sub-cell subsets were determined by Seurat package and visualized by “pheatmap” R packages.

RNA sequencing

For RNA sequencing, BMDMs were co-cultured with B.adolescentis or vehicle (PBS) (MOI of 100:1) for 24 h. Total RNA was extracted from cells by Trizol reagent (Invitrogen, USA), and mRNA was purified by poly-T oligo-attached magnetic beads. Sequencing libraries were generated using NEBNext® Ultra™ RNA Library Prep Kit for Illumina® (NEB, USA) following manufacturer’s instruction. 150 bp paired-end libraries were sequenced by Illumina PE150 platform. Paired-end reads were aligned to the human genome version hg19 using Hisat2 v2.0.5. Differential expression analysis of two groups was performed using the DESeq2. P value < 0.05 and | log2 (fold change) |> 0.5 were considered as significant.

Western blot

The Western blot was performed as previously described. The PVDF membranes were blocked with 5% skim milk for 1 h and then reacted with Decorin-specific antibody (Cat. #ab137508, diluted 1:1,000, Abcam), TLR2-specific antibody (Cat. #13744, diluted 1:1,000, CST), YAP-specific antibody (Cat. #13584-1-AP, diluted 1:1,000, Proteintech) at 4℃ overnight. Membranes were then incubated with Goat anti-Rabbit IgG-HRP (Cat. #HA1001, diluted 1:10,000, HUABIO) or Goat anti-Mouse IgG-HRP (Cat. #HA1006, diluted 1:10,000, HUABIO) at room temperature for 1 h. GAPDH (Cat. #60004-1-Ig, diluted 1:3,000, Proteintech) was used as a loading control.

Histopathologic analysis

Colorectal tumors were fixed with 4% paraformaldehyde overnight at room temperature and embedded in paraffin. Hematoxylin and eosin (H&E) stain was performed as previously described.

For IHC, sections of paraffin-embedded tissue were stained by Ki67-specific antibody (Cat. #GB111141, diluted 1:1000, Servicebio), CD31-specific antibody (Cat. #GB11063-2, diluted 1:1000, Servicebio) and visualized by DAB staining (Cat. #G1212, Servicebio) according to the manufacturer’s instructions.

Immunofluorescence staining was performed as previously described. Tissue was incubated with DCN-specific antibody (Cat. #GB11300, diluted 1:1000, Servicebio) CD68-specific antibody (Cat. #GB14043, diluted 1:500, Servicebio) and F4/80-specific antibody (Cat. #GB14043, diluted 1:500, Servicebio). Respectively, sections were visualized by AF488-TSA (G1222, diluted 1:1000, Servicebio) and AF594-TSA (G1223, diluted 1:2000, Servicebio). Finally, sections were counterstained with 40,6-diamidino-2-phenylindole (DAPI; G1012, Servicebio) and imaged with NIKON digital sight DS-FI2 (NIKON Eclipse ci). For each section, the number of positive (F4/80+, CD68+, CD68+DCN+ and F4/80+DCN+) cells were analyzed in at least 5 randomly selected high-power views.

Statistical analysis

Statistical analysis was performed using the GraphPad Prism 9.0 software. Data were analyzed with paired or unpaired Student t test, Mann–Whitney test, ANOVA test or linear regression as showed in figure legends. P value less than 0.05 was considered statistically significant.

Results

Supplementation of B. adolescentis suppressed colorectal tumorigenesis in mice

We observed that the abundance of B.adolescentis was significantly lower in tumor tissue from patients with CRC than in para-tumor (Supplementary Fig. 1A). To investigate the role of B.adolescentis in CRC, we utilized the AOM/DSS-induced CRC model in C57BL/6 mice, which were pretreated with streptomycin (2 mg/mL) for 7 days and administrated with B.adolescentis, Escherichia coli (E. coli) or vehicle (PBS) control (Supplementary Fig. 1B, C), and found that supplementation with B.adolescentis resulted in considerably fewer and smaller tumors than E. coli or vehicle group (Fig. 1A-C). The expression of cell proliferation marker Ki67 and tumor angiogenesis marker CD31 were lower in colorectal tumor tissue from B.adolescentis-treated mice (Fig. 1D-F). Collectively, these data indicated that B.adolescentis could suppress colorectal tumorigenesis in vivo.

Fig. 1
figure 1

Supplementation of B. adolescentis suppressed colorectal tumorigenesis in mice. A Representative colorectum images of AOM/DSS mice treated with B.adolescentis (B.a; n = 5), E. coli (n = 5), or vehicle (PBS) (n = 5). The red arrows indicate the tumor locations. BC Tumor number and load in the colorectum were measured. D-F Representative images and percentages of Ki67 and CD31 in colorectal cancer tissue by immunostaining; white scale bars, 200 μm; black scale bars, 50 μm. Data are shown as mean ± SD. * P < 0.05, ** P < 0.01, **** P < 0.0001; ANOVA test (B, C, E, F)

B.adolescentis recruited macrophages to suppress colorectal tumorigenesis

To understand the potential mechanism by which B.adolescentis suppresses CRC, we profiled the tumor from above AOM/DSS model using single-cell RNA sequencing (scRNA-seq) (Fig. 2A) and focused on the myeloid-derived immune cells (Supplementary Fig. 1D), as previous studies have shown that intratumoral Bifidobacterium facilitates innate immune response and scRNA-seq analyses informed the key role of myeloid-targeted therapies in CRC [16, 24]. We then divided myeloid-derived immune cells into five groups based on their top expression markers (Fig. 2B, Supplementary Fig. 1E) and found that B.adolescentis group exhibited an increase in the proportion of intratumoral macrophages and basophiles, and a decrease in dendritic cells (DCs) and monocytes (Fig. 2C, Supplementary Fig. 1F).

Fig. 2
figure 2

B.adolescentis recruited macrophages to suppress colorectal tumorigenesis. A Schematic representation of single cell preparation and scRNA-seq. B Myeloid cells were divided into 5 groups according to the top expression markers. C The percentage of 5 groups in B.adolescentis and vehicle (PBS) groups. D-E Flow cytometry representation and percentage of macrophages in colorectal lamina propria. F-G The number of F4/80+ cells was examined by immunofluorescence in the colorectal tumor of AOM/DSS mice treated with B.adolescentis, E.coli and vehicle (PBS); scale bars, 20 μm. H-J HCT116 cells were injected into BALB/c nude mice combined with THP-1 cells pretreated with B.adolescentis, E. coli or vehicle (PBS) for 24 h (n = 5 per group). Tumor volume and weight were recorded after 6 days. The independent experiment was repeated three times. Data are shown as mean ± SD. ** P < 0.01, *** P < 0.001, **** P < 0.0001; ANOVA test (E, G, I, J)

An increased percentage of macrophages was observed in colorectum lamina propria (CLP) and blood of AOM/DSS model treated with B.adolescentis when compared to E.coli or vehicle groups by flow cytometry assays (Fig. 2D, E, Supplementary Fig. 3A), whereas the percentage of DCs had no significant difference (Supplementary Fig. 3B). Similar results were confirmed in mice tumor tissue by immunofluorescence staining (Fig. 2F, G). Meanwhile, we co-cultured BMDMs or THP-1 cells with B.adolescentis for 24 h in vitro and found that B.adolescentis-treated macrophages exhibited a dramatic cell migration compared to E.coli or vehicle groups (Supplementary Fig. 3C, D).

To further determine the effect of B.adolescentis-treated macrophages on CRC progression, we injected HCT116 combined with B.adolescentis, E. coli or vehicle treated THP-1 cells (HCT116: THP-1 = 10:1) into BALB/c nude mice, and found that B.adolescentis-treated macrophages suppressed the tumor growth compared with E. coli or vehicle groups (Fig. 2H-J). In consideration of the extensive crosstalk among immune cells, the subcutaneous tumor experiment was performed in BALB/c immune-competent mice using CT26 and Raw264.7. B.adolescentis-treated macrophages consistently suppressed the tumor growth compared with E. coli or vehicle groups (Supplementary Fig. 3E-G). These data suggested that B.adolescentis promoted macrophages infiltration and had a suppressive effect on CRC.

B.adolescentis facilitated the infiltration of Decorin+ macrophages to suppress CRC

For exploration of the underlying mechanism by which B.adolescentis-treated macrophages exert anti-tumor function, we performed RNA-seq in BMDMs with or without B.adolescentis treatment (Fig. 3A), and its differentially expressed genes were analyzed and then overlapped with the differentially expressed genes of macrophages in vivo from the scRNA-seq mentioned above, and finally obtained 11 genes (Fig. 3B, C, Supplement Fig. 4A). We verified that Decorin (Dcn) had the highest fold change of expression in BMDMs treated with B.adolescentis in vitro (Fig. 3D). Furthermore, immunofluorescence staining showed that B.adolescentis increased the DCN+ macrophages (F4/80+DCN+) in tumor tissue of AOM/DSS model (Fig. 3E, F). Meanwhile, we analyzed the expression of Dcn in different cells subtypes in scRNA-seq, and found that macrophages exhibited higher Dcn expression compared to other cell subsets (Supplementary Fig. 4B). We found a significant increase in Dcn+ macrophages in B.adolescentis group through scRNA-seq data. (Supplementary Fig. 4C). Western blot analysis also confirmed that B.adolescentis increased the expression level of DCN in macrophages (Fig. 3G). Studies have reported that Dcn as a multivalent therapeutic agent against cancer by engaging multiple receptor tyrosine kinases like EGFR, Met and VEGFR2 [25], and Dcn deficiency promoted epithelial-mesenchymal transition and colorectum cancer metastasis [26]. To determine whether the tumor-suppressive effect of B.adolescentis on CRC relies on DCN+ macrophages, we established the Dcn-KD Raw264.7 cells by lentivirus-based Cas9 system (Fig. 3H, I). We then injected CT26 cells combined with Dcn-KD or wildtype Raw264.7 cells (CT26: Raw264.7 = 10:1) treated with or without B.adolescentis (MOI = 10:1) into BALB/c nude mice, and found that B.adolescentis-treated Raw264.7 obviously impeded tumor growth, whereas knockdown of Dcn notably diminished the tumor suppressive effect of B.adolescentis on CRC (Fig. 3J-L). We found B.adolescentis-treated macrophages had stronger cytotoxic activity against tumor cells than E. coli or vehicle (PBS) groups in vitro (Supplementary Fig. 4D, E). The levels of Il6 and Tnf were elevated in B.adolescentis-treated macrophages, while Dcn deficiency did not influence the expression of Il6 and Tnf (Supplementary Fig. 4F, G). Taken together, these data suggested that B.adolescentis induced DCN+ macrophages to suppress CRC growth.

Fig. 3
figure 3

B.adolescentis facilitated the infiltration of Decorin+ macrophages to suppress CRC. A The heatmap of RNA-seq of macrophages treated with B.adolescentis or vehicle (PBS). B The volcano plot showed differentially expressed genes in scRNA-seq of macrophages treated with B.adolescentis or vehicle (PBS). C 3852 differentially expressed genes in RNA-seq and 1036 differentially expressed genes of macrophages in scRNA-seq were subjected to Venn diagram analysis (|log2fold change|> 0.5, p.adj < 0.05), and 11 shared differentially expressed genes were acquired. D The levels of 11 differentially expressed genes in BMDMs treated with B.adolescentis were determined by qRT-PCR. EF The number of DCN+F4/80+ cells was examined by immunofluorescence in the colorectal tumor of AOM/DSS mice treated with B.adolescentis, E. coli or vehicle (PBS); The yellow arrows indicate the positively stained cells. scale bars, 20 μm. G BMDMs or human macrophage THP-1 cells were incubated with B.adolescentis or vehicle (PBS) for 24 h. Protein level of DCN was measured by Western blot. H Raw264.7 cells was incubated with B.adolescentis or vehicle (PBS) for 24 h. Protein level of DCN was measured by Western blot. The protein level of DCN in Raw264.7 Dcn-knockdown cells or Raw264.7 control cells. J-L Raw264.7 Dcn-knockdown cells or control cells were pretreated with B.adolescentis or vehicle (PBS), and combined with CT26 were injected into BALB/c nude mice. Tumor volume was recorded after 6 days. The independent experiment was repeated three times. Data are shown as mean ± SD. ns: No statistical difference, * P < 0.05, ** P < 0.01, *** P < 0.001, **** P < 0.0001; Student t test (D), ANOVA test (F, K, L)

The activation of TLR2 is essential for inducing DCN+ macrophages by B.adolescentis

Pattern recognition receptors (PRRs) play a central role in innate immune response [27, 28]. As Toll-like receptors (TLRs) are a major class of PPRs that recognize extracellular signals, we screened out TLRs that were differentially expressed in RNA-seq of B.adolescentis-treated BMDMs (Fig. 4A) and checked them by qRT-PCR, and discovered that TLR2 was most significantly upregulated (Fig. 4B). We then confirmed the upregulation of TLR2 in BMDMs treated with B.adolescentis at the protein level (Fig. 4C). Mechanically, to verify that B.adolescentis induced the DCN+ macrophages via TLR2, we performed TLR2 inhibition assays by Cu-CPT22 and found that Cu-CPT22 administration abolished the upregulation of DCN in BMDMs treated with B.adolescentis (Fig. 4D, E). Similar results were reproduced in THP-1 cells (Fig. 4F-I). We further combined injected HCT116 and THP-1 cells with B.adolescentis (MOI = 10:1) or vehicle into BALB/c nude mice along with intraperitoneally injecting Cu-CPT22 every two days. We found that TLR2 inhibition reduced the number of DCN+ macrophages in tumor and attenuated tumor suppressive effect of B.adolescentis-treated macrophages (Fig. 4J-M, Supplementary Fig. 5A-C). These results suggested that B.adolescentis induced the DCN+ macrophages in a TLR2-dependent way.

Fig. 4
figure 4

The activation of TLR2 is essential for inducing DCN+ macrophages by B.adolescentis. The heatmap of differentially expressed TLRs genes in RNA-seq of BMDMs treated with B.adolescentis or vehicle (PBS). B The levels of differentially expressed TLRs genes in BMDMs treated with B.adolescentis were determined by qRT-PCR. C BMDMs were incubated with B.adolescentis or vehicle (PBS) for 24 h. Protein levels of TLR2 and DCN were tested by Western blot. DE BMDMs were incubated with B.adolescentis or vehicle (PBS) for 24 h with or without 25 μM Cu-CPT22. Protein levels of TLR2 and DCN were tested by Western blot and mRNA level of Dcn was tested by qRT-PCR. F-I THP-1 cells were incubated with B.adolescentis or vehicle (PBS) for 24 h (H-G). THP-1 cells were incubated with B.adolescentis or vehicle (PBS) for 24 h with or without 25 μM Cu-CPT22 (HI). Protein levels of TLR2 and DCN were tested by Western blot and mRNA level of DCN was tested by qRT-PCR. J-L HCT116 cells were injected into BALB/c nude mice combined with THP-1 cells pretreated with B.adolescentis or vehicle (PBS) for 24 h (n = 5 per group). From the beginning of tumor inoculation until sacrifice, 3 mg/kg Cu-CPT22 or vehicle (5% DMSO) were injected intraperitoneally to mice every two days. Tumor volume was recorded after 6 days. M The positive ratio of Ki67 in mice tumor tissue. The independent experiment was repeated three times. Data are shown as mean ± SD. * P < 0.05, ** P < 0.01, *** P < 0.001, **** P < 0.0001; Student t test (B, G), ANOVA test (E, I, K, L, M)

B.adolescentis regulated DCN+ macrophages through TLR2/YAP axis

Previous studies have demonstrated that TLR2/YAP axis regulated cancer innate immunity [29, 30], yet the function of TLR2/YAP axis in macrophages was unclear. We discovered that the level of YAP was upregulated in BMDMs and THP-1 cells (Fig. 5A, B) and TLR2 inhibition diminished the regulation of B.adolescentis on YAP (Supplementary Fig. 6A). The nuclear expression of YAP increased in BMDMs and THP-1 cells after B.adolescentis treatment (Fig. 5C-E). YAP inhibitor verteporfin markedly reduced the expression of YAP and DCN in B.adolescentis-treated macrophages but it did not influence the expression of TLR2 (Fig. 5F, G, Supplementary Fig. 6B, C). And overexpression of YAP in BMDMs and THP-1 cells increased the level of DCN (Fig. 5H, I). We then injected HCT116 combined with B.adolescentis (MOI = 10:1) or vehicle treated THP-1 cells into BALB/c nude mice, and intraperitoneally injected with verteporfin every days. YAP inhibitor administration attenuated the tumor suppressive effect of B.adolescentis-treated THP-1 cells and reduced the number of DCN+ macrophages in tumors on BALB/c nude mice. (Fig. 5J-L, Supplementary Fig. 6D-F). Furthermore, we found that the expression levels of DCN, TLR2 and YAP were elevated after treated with B.adolescentis in human primary macrophages (Supplementary Fig. 7A, B).

Fig. 5
figure 5

B.adolescentis regulated DCN+ macrophages through TLR2/YAP axis. AB BMDMs and THP-1 cells were incubated with B.adolescentis or vehicle (PBS) for 24 h. Protein levels of TLR2 and YAP were tested by Western blot. C Immunofluorescence assay of YAP distribution in BMDMs. BMDMs were stained with specific antibody against YAP (green), and the nuclei were counterstained with DAPI (blue). Scale bar, 10 μm. DE Nuclear and cytoplasmic separation assay was performed in B.adolescentis-treated BMDMs and THP-1 cells, then the protein level of YAP in the nuclear and cytoplasm was detected by Western blot. FG BMDMs and THP-1 cells were incubated with B.adolescentis or vehicle (PBS) for 24 h with or without 1 μM verteporfin. Protein levels of TLR2, YAP and DCN were tested by Western blot. HI YAP was overexpressed in BMDMs and THP-1 cells, and protein levels of YAP and DCN were tested by Western blot. J-L HCT116 cells were injected into BALB/c nude mice combined with THP-1 cells pretreated with B.adolescentis or vehicle (PBS) for 24 h (n = 5 per group). From the beginning of tumor inoculation until sacrifice, 50 mg/kg verteporfin or vehicle (5% DMSO) were injected intraperitoneally to mice every day. Tumor volume was recorded after 6 days. The independent experiment was repeated three times. Data are shown as mean ± SD. * P < 0.05, ** P < 0.01, **** P < 0.0001; ANOVA test (K, L)

To investigate the relationship between DCN+ macrophages and macrophage polarization state, we examined the related markers of macrophages after co-culturing with B.adolescentis and found that the number of M1 macrophages increased while M2 macrophages decreased in B.adolescentis-treated BMDMs (Supplementary Fig. 8A-D). B.adolescentis upregulated the expression levels of DCN, TLR2 and YAP in M1 macrophages (Supplementary Fig. 8E, F), which reflected that the function of DCN+ macrophages might be similar with traditional M1-like macrophages. These results reflected that B.adolescentis increased DCN+ macrophages through the TLR2/YAP axis.

B.adolescentis abundance correlated with TLR2 and DCN+ macrophages in patients with CRC

To further investigate the clinical relevance, we quantified B.adolescentis and the expression of TLR2 and DCN with quantitative qRT-PCR analysis in paired fresh CRC and adjacent non-tumor tissue. According to the level of B.adolescentis in CRC tissue, we divided these patients (n = 65) into high and low B.adolescentis group. We found that compared with the low B.adolescentis group, the number of macrophages in high B.adolescentis group was higher (Fig. 6A), and the abundance of B.adolescentis was related to the number of DCN+ macrophages (Fig. 6B). Moreover, the expression of DCN and TLR2 was decreased in CRC tissue compared with their adjacent non-tumor tissue (Fig. 6C), and the abundance of B.adolescentis was positively correlated with the expression of TLR2 and DCN in CRC tissue (Fig. 6D). In addition, the expression of DCN was correlated with the expression of TLR2 (Fig. 6E). Lastly, we confirmed the above results using TCGA GTEx datasets (Fig. 6F, G). These data indicated that B.adolescentis abundance correlated with the expression of TLR2 and the number of DCN+ macrophages in patients with CRC.

Fig. 6
figure 6

B.adolescentis abundance correlated with TLR2 and DCN+ macrophages in patients with CRC. A Representative images of immunofluorescence assay for CD68 in tumor tissue with low or high B.adolescentis abundance from cohort; scale bars, 50 μm. B Representative images of immunofluorescence staining of DCN+ macrophages (CD68+DCN+ dual positive) in tumor tissue of CRC; scale bars, 50 μm. The yellow and white arrows indicate the positively stained cells. C qRT-PCR analysis of DCN and TLR2 mRNA expression in tumor or para-tumor tissue of CRC. D The correlation of B.adolescentis with DCN or TLR2 was analyzed in tumor tissue of CRC. E The correlation of DCN and TLR2 was analyzed in tumor tissue of CRC. F Transcript levels of DCN and TLR2 in colorectal cancer and normal tissue from TCGA database. G The correlation of DCN and TLR2 was analyzed in TCGA database. The independent experiment was repeated three times. Data are shown as mean ± SD. * P < 0.05, **** P < 0.0001; Mann–Whitney test (A, C, F), Linear Regression (D, E, G)

Discussion

Several infant-type species of human-derived bifidobacterial (HRB) are associated with strengthening host immunity and suppressing tumorigenesis. However, the role and mechanism of adult-type HRB in cancer remains unclear [31]. In this study, we revealed that B.adolescentis suppressed colorectal tumorigenesis in AOM/DSS mice.

Macrophages are a major component of TME whose functional plasticity leads to anti-tumor and pro-tumor function in different environment [32]. Furthermore, targeting macrophages is an emerging field of interest due to synergies with immune checkpoint inhibitors, chemotherapy, and radiation therapy in preclinical studies [33, 34]. Our groups have reported that some specific microbes were closely related to macrophages functions [20, 35]. In this study, we discovered that B.adolescentis suppressed colorectal tumorigenesis through inducing DCN+ macrophages, and targeting macrophages with B.adolescentis maybe a potential novel strategy for cancer therapy.

Decorin is a member of the small leucine-rich proteoglycans (SLRPs) which are an important subset of extracellular matrix [36]. Soluble DCN engages multiple receptor tyrosine kinases within the target-rich environment of the tumor stroma and tumor parenchyma [25]. DCN possesses a multitude of oncosuppressive functions including growth suppressing, angiostasis, and tumor cells mitophagy arrest [37]. Several research groups have discovered that DCN deficiency promotes epithelial-mesenchymal transition and hepatic metastasis of colorectal carcinoma [26, 37]. DCN derived from T cells mediates inhibition of carcinogenesis in microglia and reduces glioma formation [38]. However, the function of macrophage-derived DCN has not been reported. We found that B.adolescentis increased the DCN+ macrophages and then suppressed colorectal tumorigenesis. B.adolescentis also boosted the cytotoxicity of macrophages against tumor cells and increased the expression of Il6 and Tnf in macrophages in a Dcn-independent way.

Toll-like receptors (TLRs) are an important component family of pattern recognition receptors (PRRs) responsible for early recognition in the innate immune system. TLR2 recognizes a vast of ligands derived from microbes such as lipoproteins/lipopeptides and peptidoglycans [39]. Bifidobacterium breve has been reported to induce dendritic cell maturation, activation, and survival via TLR2 [40]. Here, we found that B.adolescentis elevated the expression of TLR2 in macrophages and the activation of TLR2 is essential for B.adolescentis regulating DCN+ macrophages. However, how B.adolescentis was recognized by TLR2 in macrophages needs to be further explored.

Emerging evidences suggested that the differentiation, metabolism, and functions of innate immune cells were extensively regulated by YAP [41, 42]. Microbes are reported to regulate the activation of YAP in cancer cells to inhibits colorectal tumorigenesis [43, 44]. YAP in macrophages promoted the LPS/IFN-γ-triggered activation of M1 macrophages, while impaired the IL-4/IL-13 induced M2 macrophages polarization [45]. Prior studies suggested that TLR2/YAP axis played an important role in innate immunity and the activation of TLR2/YAP axis in tumor cells suppressed tumor growth [29, 30]. However, the underlying mechanism of B.adolescentis interacted with macrophages and the function of TLR2/YAP axis in macrophages remain largely unknown. Indeed, we observed that TLR2/YAP axis was activated by B.adolescentis and inhibition of TLR2/YAP axis significantly impaired the induction of DCN+ macrophages by B.adolescentis. Consistent with our findings, B.adolescentis, TLR2 and DCN were reduced in CRC tissue. However, the relationship between the expression of TLR2 and DCN in macrophages and the abundance of B.adolescentis needs to be further explored.

In conclusion, our results provided a novel insight into the interaction and molecular mechanism of Bifidobacterium with macrophages. Our findings suggested that B.adolescentis could act as probiotics to regulate macrophages in colorectal cancer, thus becoming a potential target for tumor vaccine delivery and providing a novel strategy for tumor therapy.

Availability of data and materials

All data generated or analyzed during the study are included in this published article and its additional files.

Change history

Abbreviations

B.a :

Bifidobacterium adolescentis

CRC:

Colorectal cancer

qRT-PCR:

Quantitative real-time PCR

TME:

Tumor microenvironment

DCN:

Decorin

References 

  1. Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer statistics, 2022. CA Cancer J Clin. 2022;72(1):7–33. https://doi.org/10.3322/caac.21708.

    Article  PubMed  Google Scholar 

  2. Cai J, Sun L, Gonzalez FJ. Gut microbiota-derived bile acids in intestinal immunity, inflammation, and tumorigenesis. Cell Host Microbe. 2022;30(3):289–300. https://doi.org/10.1016/j.chom.2022.02.004.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Wang L, Tang L, Feng Y, Zhao S, Han M, Zhang C, Yuan G, Zhu J, Cao S, Wu Q, et al. A purified membrane protein from Akkermansia muciniphila or the pasteurised bacterium blunts colitis associated tumourigenesis by modulation of CD8(+) T cells in mice. Gut. 2020;69(11):1988–97. https://doi.org/10.1136/gutjnl-2019-320105.

    Article  CAS  PubMed  Google Scholar 

  4. Chung L, Thiele Orberg E, Geis AL, Chan JL, Fu K, DeStefano Shields CE, Dejea CM, Fathi P, Chen J, Finard BB et al. Bacteroides fragilis Toxin Coordinates a Pro-carcinogenic Inflammatory Cascade via Targeting of Colonic Epithelial Cells. Cell Host Microbe 2018, 23(2):203–214 e205.https://doi.org/10.1016/j.chom.2018.01.007.

  5. Gur C, Ibrahim Y, Isaacson B, Yamin R, Abed J, Gamliel M, Enk J, Bar-On Y, Stanietsky-Kaynan N, Coppenhagen-Glazer S, et al. Binding of the Fap2 protein of Fusobacterium nucleatum to human inhibitory receptor TIGIT protects tumors from immune cell attack. Immunity. 2015;42(2):344–55. https://doi.org/10.1016/j.immuni.2015.01.010.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Overacre-Delgoffe AE, Bumgarner HJ, Cillo AR, Burr AHP, Tometich JT, Bhattacharjee A, Bruno TC, Vignali DAA, Hand TW: Microbiota-specific T follicular helper cells drive tertiary lymphoid structures and anti-tumor immunity against colorectal cancer. Immunity 2021, 54(12):2812–2824 e2814.https://doi.org/10.1016/j.immuni.2021.11.003.

  7. Yang Y, Li L, Xu C, Wang Y, Wang Z, Chen M, Jiang Z, Pan J, Yang C, Li X, et al. Cross-talk between the gut microbiota and monocyte-like macrophages mediates an inflammatory response to promote colitis-associated tumourigenesis. Gut. 2020;70(8):1495–506. https://doi.org/10.1136/gutjnl-2020-320777.

    Article  CAS  PubMed  Google Scholar 

  8. Hezaveh K, Shinde RS, Klotgen A, Halaby MJ, Lamorte S, Ciudad MT, Quevedo R, Neufeld L, Liu ZQ, Jin R et al: Tryptophan-derived microbial metabolites activate the aryl hydrocarbon receptor in tumor-associated macrophages to suppress anti-tumor immunity. Immunity 2022, 55(2):324–340 e328.https://doi.org/10.1016/j.immuni.2022.01.006.

  9. Jiang J, Mei J, Yi S, Feng C, Ma Y, Liu Y, Liu Y, Chen C. Tumor associated macrophage and microbe: The potential targets of tumor vaccine delivery. Adv Drug Deliv Rev. 2022;180:114046. https://doi.org/10.1016/j.addr.2021.114046.

    Article  CAS  PubMed  Google Scholar 

  10. Henrick BM, Rodriguez L, Lakshmikanth T, Pou C, Henckel E, Arzoomand A, Olin A, Wang J, Mikes J, Tan Z et al: Bifidobacteria-mediated immune system imprinting early in life. Cell 2021, 184(15):3884–3898 e3811.https://doi.org/10.1016/j.cell.2021.05.030.

  11. Grangette C. Bifidobacteria and subsets of dendritic cells: friendly players in immune regulation! Gut. 2012;61(3):331–2. https://doi.org/10.1136/gutjnl-2011-301476.

    Article  CAS  PubMed  Google Scholar 

  12. Groeger D, O’Mahony L, Murphy EF, Bourke JF, Dinan TG, Kiely B, Shanahan F, Quigley EM. Bifidobacterium infantis 35624 modulates host inflammatory processes beyond the gut. Gut Microbes. 2013;4(4):325–39. https://doi.org/10.4161/gmic.25487.

    Article  PubMed  PubMed Central  Google Scholar 

  13. Matson V, Fessler J, Bao R, Chongsuwat T, Zha Y, Alegre ML, Luke JJ, Gajewski TF. The commensal microbiome is associated with anti-PD-1 efficacy in metastatic melanoma patients. Science. 2018;359(6371):104–8. https://doi.org/10.1126/science.aao3290.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Goc J, Lv M, Bessman NJ, Flamar AL, Sahota S, Suzuki H, Teng F, Putzel GG, Eberl G, Withers DR, et al. Dysregulation of ILC3s unleashes progression and immunotherapy resistance in colon cancer. Cell. 2021;184(19):5015-5030.e5016. https://doi.org/10.1016/j.cell.2021.07.029.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Sivan A, Corrales L, Hubert N, Williams JB, Aquino-Michaels K, Earley ZM, Benyamin FW, Lei YM, Jabri B, Alegre ML, et al. Commensal Bifidobacterium promotes antitumor immunity and facilitates anti-PD-L1 efficacy. Science. 2015;350(6264):1084–9. https://doi.org/10.1126/science.aac4255.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Shi Y, Zheng W, Yang K, Harris KG, Ni K, Xue L, Lin W, Chang EB, Weichselbaum RR, Fu YX: Intratumoral accumulation of gut microbiota facilitates CD47-based immunotherapy via STING signaling. J Exp Med 2020, 217(5).https://doi.org/10.1084/jem.20192282.

  17. Mager LF, Burkhard R, Pett N, Cooke NCA, Brown K, Ramay H, Paik S, Stagg J, Groves RA, Gallo M, et al. Microbiome-derived inosine modulates response to checkpoint inhibitor immunotherapy. Science. 2020;369(6510):1481–9. https://doi.org/10.1126/science.abc3421.

    Article  CAS  PubMed  Google Scholar 

  18. Fan L, Qi Y, Qu S, Chen X, Li A, Hendi M, Xu C, Wang L, Hou T, Si J et al: B. adolescentis ameliorates chronic colitis by regulating Treg/Th2 response and gut microbiota remodeling. Gut Microbes 2021, 13(1):1–17.https://doi.org/10.1080/19490976.2020.1826746.

  19. Neufert C, Heichler C, Brabletz T, Scheibe K, Boonsanay V, Greten FR, Neurath MF: Inducible mouse models of colon cancer for the analysis of sporadic and inflammation-driven tumor progression and lymph node metastasis. Nat Protoc 2021, 16(1):61–85.https://doi.org/10.1038/s41596-020-00412-1.

  20. Fan L, Xu C, Ge Q, Lin Y, Wong CC, Qi Y, Ye B, Lian Q, Zhuo W, Si J et al: A. Muciniphila Suppresses Colorectal Tumorigenesis by Inducing TLR2/NLRP3-Mediated M1-Like TAMs. Cancer Immunol Res 2021, 9(10):1111–1124.https://doi.org/10.1158/2326-6066.Cir-20-1019.

  21. Xiao R, Zeng J, Bressler EM, Lu W, Grinstaff MW: Synthesis of bioactive (1→6)-β-glucose branched poly-amido-saccharides that stimulate and induce M1 polarization in macrophages. Nat Commun 2022, 13(1):4661.https://doi.org/10.1038/s41467-022-32346-5.

  22. Shan X, Hu P, Ni L, Shen L, Zhang Y, Ji Z, Cui Y, Guo M, Wang H, Ran L et al: Serine metabolism orchestrates macrophage polarization by regulating the IGF1-p38 axis. Cell Mol Immunol 2022, 19(11):1263–1278.https://doi.org/10.1038/s41423-022-00925-7.

  23. Kopp A, Smeets R, Gosau M, Kröger N, Fuest S, Köpf M, Kruse M, Krieger J, Rutkowski R, Henningsen A et al: Effect of process parameters on additive-free electrospinning of regenerated silk fibroin nonwovens. Bioact Mater 2020, 5(2):241–252.https://doi.org/10.1016/j.bioactmat.2020.01.010.

  24. Zhang L, Li Z, Skrzypczynska KM, Fang Q, Zhang W, O'Brien SA, He Y, Wang L, Zhang Q, Kim A et al: Single-Cell Analyses Inform Mechanisms of Myeloid-Targeted Therapies in Colon Cancer. Cell 2020, 181(2):442–459.e429.https://doi.org/10.1016/j.cell.2020.03.048.

  25. Neill T, Schaefer L, Iozzo RV: Decorin as a multivalent therapeutic agent against cancer. Adv Drug Deliv Rev 2016, 97:174–185.https://doi.org/10.1016/j.addr.2015.10.016.

  26. Mao L, Yang J, Yue J, Chen Y, Zhou H, Fan D, Zhang Q, Buraschi S, Iozzo RV, Bi X. Decorin deficiency promotes epithelial-mesenchymal transition and colon cancer metastasis. Matrix Biol. 2021;95:1–14. https://doi.org/10.1016/j.matbio.2020.10.001.

    Article  CAS  PubMed  Google Scholar 

  27. Si H, Yang Q, Hu H, Ding C, Wang H, Lin X. Colorectal cancer occurrence and treatment based on changes in intestinal flora. Semin Cancer Biol. 2021;70:3–10. https://doi.org/10.1016/j.semcancer.2020.05.004.

    Article  CAS  PubMed  Google Scholar 

  28. Wong SH, Yu J. Gut microbiota in colorectal cancer: mechanisms of action and clinical applications. Nat Rev Gastroenterol Hepatol. 2019;16(11):690–704. https://doi.org/10.1038/s41575-019-0209-8.

    Article  CAS  PubMed  Google Scholar 

  29. Zhang L, Shi H, Chen H, Gong A, Liu Y, Song L, Xu X, You T, Fan X, Wang D, et al. Dedifferentiation process driven by radiotherapy-induced HMGB1/TLR2/YAP/HIF-1alpha signaling enhances pancreatic cancer stemness. Cell Death Dis. 2019;10(10):724. https://doi.org/10.1038/s41419-019-1956-8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Liu B, Zheng Y, Yin F, Yu J, Silverman N, Pan D. Toll Receptor-Mediated Hippo Signaling Controls Innate Immunity in Drosophila. Cell. 2016;164(3):406–19. https://doi.org/10.1016/j.cell.2015.12.029.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Wong CB, Odamaki T, Xiao JZ. Insights into the reason of Human-Residential Bifidobacteria (HRB) being the natural inhabitants of the human gut and their potential health-promoting benefits. FEMS Microbiol Rev. 2020;44(3):369–85. https://doi.org/10.1093/femsre/fuaa010.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Anderson NR, Minutolo NG, Gill S, Klichinsky M. Macrophage-Based Approaches for Cancer Immunotherapy. Cancer Res. 2021;81(5):1201–8. https://doi.org/10.1158/0008-5472.Can-20-2990.

    Article  CAS  PubMed  Google Scholar 

  33. Pathria P, Louis TL, Varner JA. Targeting Tumor-Associated Macrophages in Cancer. Trends Immunol. 2019;40(4):310–27. https://doi.org/10.1016/j.it.2019.02.003.

    Article  CAS  PubMed  Google Scholar 

  34. Xiang X, Wang J, Lu D, Xu X. Targeting tumor-associated macrophages to synergize tumor immunotherapy. Signal Transduct Target Ther. 2021;6(1):75. https://doi.org/10.1038/s41392-021-00484-9.

    Article  PubMed  PubMed Central  Google Scholar 

  35. Xu C, Fan L, Lin Y, Shen W, Qi Y, Zhang Y, Chen Z, Wang L, Long Y, Hou T, et al. Fusobacterium nucleatum promotes colorectal cancer metastasis through miR-1322/CCL20 axis and M2 polarization. Gut Microbes. 2021;13(1):1980347. https://doi.org/10.1080/19490976.2021.1980347.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Neill T, Schaefer L, Iozzo RV. Decorin: a guardian from the matrix. Am J Pathol. 2012;181(2):380–7. https://doi.org/10.1016/j.ajpath.2012.04.029.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Reszegi A, Horváth Z, Karászi K, Regős E, Postniková V, Tátrai P, Kiss A, Schaff Z, Kovalszky I, Baghy K: The Protective Role of Decorin in Hepatic Metastasis of Colorectal Carcinoma. Biomolecules 2020, 10(8).https://doi.org/10.3390/biom10081199.

  38. Chatterjee J, Sanapala S, Cobb O, Bewley A, Goldstein AK, Cordell E, Ge X, Garbow JR, Holtzman MJ, Gutmann DH. Asthma reduces glioma formation by T cell decorin-mediated inhibition of microglia. Nat Commun. 2021;12(1):7122. https://doi.org/10.1038/s41467-021-27455-6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Fitzgerald KA, Kagan JC. Toll-like Receptors and the Control of Immunity. Cell. 2020;180(6):1044–66. https://doi.org/10.1016/j.cell.2020.02.041.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Hoarau C, Lagaraine C, Martin L, Velge-Roussel F, Lebranchu Y. Supernatant of Bifidobacterium breve induces dendritic cell maturation, activation, and survival through a Toll-like receptor 2 pathway. J Allergy Clin Immunol. 2006;117(3):696–702. https://doi.org/10.1016/j.jaci.2005.10.043.

    Article  CAS  PubMed  Google Scholar 

  41. Zhang Q, Zhou R, Xu P: The Hippo Pathway in Innate Anti-microbial Immunity and Anti-tumor Immunity. Front Immunol 2020, 11:1473.https://doi.org/10.3389/fimmu.2020.01473.

  42. Wang S, Zhou L, Ling L, Meng X, Chu F, Zhang S, Zhou F: The Crosstalk Between Hippo-YAP Pathway and Innate Immunity. Front Immunol 2020, 11:323.https://doi.org/10.3389/fimmu.2020.00323.

  43. Li Q, Hu W, Liu WX, Zhao LY, Huang D, Liu XD, Chan H, Zhang Y, Zeng JD, Coker OO et al: Streptococcus thermophilus Inhibits Colorectal Tumorigenesis Through Secreting beta-Galactosidase. Gastroenterology 2021, 160(4):1179–1193 e1114.https://doi.org/10.1053/j.gastro.2020.09.003.

  44. Chen S, Zhang L, Li M, Zhang Y, Sun M, Wang L, Lin J, Cui Y, Chen Q, Jin C, et al. Fusobacterium nucleatum reduces METTL3-mediated m(6)A modification and contributes to colorectal cancer metastasis. Nat Commun. 2022;13(1):1248. https://doi.org/10.1038/s41467-022-28913-5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Zhou X, Li W, Wang S, Zhang P, Wang Q, Xiao J, Zhang C, Zheng X, Xu X, Xue S et al: YAP Aggravates Inflammatory Bowel Disease by Regulating M1/M2 Macrophage Polarization and Gut Microbial Homeostasis. Cell Rep 2019, 27(4):1176–1189 e1175.https://doi.org/10.1016/j.celrep.2019.03.028.

Download references

Acknowledgements

We thank Prof. Wei Liu (Zhejiang Academy of Agricultural Sciences) for providing microbial culture technical support.

Funding

This work was supported by the National Natural Science Foundation of China (grant no. 82072623, to L. Wang; grant no. 82270573, to S. Chen; grant no. 82203618, to L. Fan), Zhejiang Province Natural Science Foundation (grant no. LZ22H160002, to L. Wang), and Zhejiang Province Medicine and Health Science and Technology Project (grant no. 2023KY722, to L. Fan).

Author information

Authors and Affiliations

Authors

Contributions

LJ Wang conceived and designed the study. Yifeng Lin, Lina Fan, Yadong Qi Chaochao Xu, Dingjiacheng Jia and Yao jiang performed experiment. Yifeng Lin, Lina Fan, Yadong Qi analyzed and interpreted the data. Yifeng Lin drafted the manuscript. LJ Wang and SJ Chen critically revised the manuscript. All authors read and approved the final manuscript.

Corresponding authors

Correspondence to Shujie Chen or Liangjing Wang.

Ethics declarations

Ethics approval and consent to participate

For human specimens, Clinical Research Ethics Committee of the Sir Run Shaw Hospital, Zhejiang University School of Medicine approved the protocol (20211103–35). All aspects of the study were conducted in accordance with the principles of the Declaration of Helsinki. All animal studies were approved by the Institutional Animal Care and Use Committee (IACUC) of Zhejiang University (ZJU) (IACUC-02102214). All animal experiments strictly adhered to protocols, policies, and ethical guidelines formulated by our IACUC.

Consent for publication

Consent for publish was obtained from the participant to exhibit individual data.

Competing interests

The authors disclose no conflicts of interest related to this work.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

The original online version of this article was revised: authors identified an error in Fig. 4F. The images in Fig. 4F are mistakenly pasted.

Supplementary Information

Additional file 1: Figure S1.

Supplementation of B. adolescentis suppressed colorectal tumorigenesis and increased infiltration of macrophages. Figure S2. Gating strategy of macrophages and dendritic cells. Figure S3. B.adolescentis recruited macrophages to suppress colorectal tumorigenesis. Figure S4. B.adolescentis facilitated the infiltration of Decorin+ macrophages to suppress CRC. Figure S5.The activation of TLR2 is essential for inducing DCN+ macrophages by B.adolescentisFigure S6. B.adolescentis regulated DCN+ macrophages through TLR2/YAP axis. Figure S7. B.adolescentis activated TLR2/YAP/DCN in primary human macrophages. FigureS8. B.adolescentis activated TLR2/YAP/DCN in M1 macrophages. Table S1. Primers used for validation the gene expression level. 

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lin, Y., Fan, L., Qi, Y. et al. Bifidobacterium adolescentis induces Decorin+ macrophages via TLR2 to suppress colorectal carcinogenesis. J Exp Clin Cancer Res 42, 172 (2023). https://doi.org/10.1186/s13046-023-02746-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13046-023-02746-6

Keywords