You are on page 1of 12

Cancer Letters 469 (2020) 456–467

Contents lists available at ScienceDirect

Cancer Letters
journal homepage: www.elsevier.com/locate/canlet

Original Articles

Clostridium butyricum, a butyrate-producing probiotic, inhibits intestinal T


tumor development through modulating Wnt signaling and gut microbiota
Danfeng Chena,1, Duochen Jina,1, Shumin Huanga,1, Jingyi Wua, Mengque Xua,b, Tianyu Liua,
Wenxiao Donga, Xiang Liua, Sinan Wanga, Weilong Zhonga, Yi Liuc,d, Ruihuan Jiangd,
Meiyu Piaoa, Bangmao Wanga,∗∗, Hailong Caoa,d,∗
a
Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin Institute of Digestive Disease, PR China
b
Department of Gastroenterology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, PR China
c
Department of Gastroenterology and Hepatology, Tianjin Third Central Hospital, Tianjin, PR China
d
Department of Gastroenterology and Hepatology, Hotan District People's Hospital, Xinjiang Uygur Autonomous Region, Xinjiang, PR China

A R T I C LE I N FO A B S T R A C T

Keywords: Gut microbiota dysbiosis is closely involved in intestinal carcinogenesis. A marked reduction in butyrate-pro-
Clostridium butyricum ducing bacteria has been observed in patients with colorectal cancer (CRC); nevertheless, the potential benefit of
Gut microbiota butyrate-producing bacteria against intestinal tumor development has not been fully investigated. We found that
Short-chain fatty acids Clostridium butyricum (C. butyricum, one of the commonly used butyrate-producing bacteria in clinical settings)
Wnt signaling pathway
significantly inhibited high-fat diet (HFD)-induced intestinal tumor development in Apcmin/+ mice. Moreover,
Apcmin/+ mouse
intestinal tumor cells treated with C. butyricum exhibited decreased proliferation and increased apoptosis.
Additionally, C. butyricum suppressed the Wnt/β-catenin signaling pathway and modulated the gut microbiota
composition, as demonstrated by decreases in some pathogenic bacteria and bile acid (BA)-biotransforming
bacteria and increases in some beneficial bacteria, including short-chain fatty acid (SCFA)-producing bacteria.
Accordingly, C. butyricum decreased the fecal secondary BA contents, increased the cecal SCFA quantities, and
activated G-protein coupled receptors (GPRs), such as GPR43 and GPR109A. The anti-proliferative effect of C.
butyricum was blunted by GPR43 gene silencing using small interfering RNA (siRNA). The analysis of clinical
specimens revealed that the expression of GPR43 and GPR109A gradually decreased from human normal colonic
tissue to adenoma to carcinoma. Together, our results show that C. butyricum can inhibit intestinal tumor de-
velopment by modulating Wnt signaling and gut microbiota and thus suggest the potential efficacy of butyrate-
producing bacteria against CRC.

1. Introduction contrast, dietary fiber has been negatively correlated with the risk of
CRC [6].
Colorectal cancer (CRC) is considered one of the most serious public Numerous studies have revealed that gut microbiota dysbiosis is
health problems and is the third most commonly diagnosed cancer in related to CRC [7–9]. Compared with healthy individuals, patients with
both males and females [1]. In addition to genetic factors, various en- CRC often suffer from a reduction in butyrate-producing bacteria
vironmental and lifestyle factors have been implicated in the develop- [10–14], which suggests that butyrate-producing bacteria might po-
ment of CRC [2]. A high-fat diet (HFD) has been recognized as a major tentially exhibit anti-tumor properties in CRC. Short-chain fatty acids
environmental risk factor for CRC, and the associated mechanisms in- (SCFAs), primarily acetate, propionate, and butyrate, are the major
clude disturbance of the bile acid (BA) metabolism, induction of chronic products of the colonic microbial fermentation of undigested dietary
low-grade inflammation, and disruption of the gut microbiota [3–5]. In fiber [15,16]. Butyrate has potential anti-inflammatory and anti-


Corresponding author. Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin Institute of Digestive Disease,
Anshan Road No.154, Heping District, Tianjin, 300052, PR China. Tel.: +86 022 60362608.
∗∗
Corresponding author. Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin Institute of Digestive Disease,
Anshan Road No.154, Heping District, Tianjin, 300052, PR China. Tel.: +86 022 60362608.
E-mail addresses: tjmughgi@hotmail.com (B. Wang), caohailong@tmu.edu.cn (H. Cao).
1
These authors contributed equally to this work.

https://doi.org/10.1016/j.canlet.2019.11.019
Received 23 July 2019; Received in revised form 10 November 2019; Accepted 11 November 2019
0304-3835/ © 2019 Elsevier B.V. All rights reserved.
D. Chen, et al. Cancer Letters 469 (2020) 456–467

Abbreviations GC gas chromatography


GPRs G-protein coupled receptors
ANOSIM analysis of similarities HFD high-fat diet
AOM azoxymethane IBD inflammatory bowel disease
APC adenomatous polyposis coli LCA lithocholic acid
ATCC American Type Culture Collection LEfSe linear discriminant analysis effect size
BA bile acid MEM Minimum Essential Medium
BD basal diet OTUs operational taxonomic units
CA cholic acid PBS phosphate-buffered saline
CB C. butyricum, Clostridium butyricum PCoA principal coordinate analysis
CCK-8 Cell Counting Kit-8 SCFA short-chain fatty acid
CDCA chenodeoxycholic acid siRNA small interfering RNA
CRC colorectal cancer SPF specific pathogen-free
DCA deoxycholic acid TdT terminal deoxynucleotidyl transferase
DMH 1,2-dimethylhydrazine dihydrochloride TLR4 Toll-like receptor 4
FBS et al. bovine serum TNBS 2,4,6-trinitrobenzene sulfonic acid
F/B Firmicutes/Bacteroidetes TUNEL TdT-mediated dUTP nick end-labeling

carcinogenic properties [17]. However, the oral administration of bu- receptors (GPRs), such as GPR43 and GPR109A. Collectively, these
tyrate is challenging due to its rancid smell and unpleasant taste [18], findings reveal that C. butyricum might exhibit potential efficacy against
and a butyrate enema promotes colonic hypersensitivity [19]. Another CRC.
class of microbial metabolites, BAs, promotes CRC [20,21]. Primary
BAs, such as cholic acid (CA) and chenodeoxycholic acid (CDCA), are 2. Materials and methods
synthesized from cholesterol in hepatocytes and then biotransformed
by bacteria of the genus Clostridium into secondary BAs, such as 2.1. Bacterial preparation
deoxycholic acid (DCA) and lithocholic acid (LCA), respectively
[22,23]. C. butyricum (ATCC 19398) was supplied by the China General
The characteristics and mechanism of action of Clostridium butyr- Microbiological Culture Collection Center and incubated in brain heart
icum (C. butyricum), which are one of the most studied butyrate-pro- infusion medium under anaerobic conditions for 24 h at 37 °C until the
ducing bacteria, have been reported since 1880 [24]. Moreover, C. logarithmic phase of growth with a bacterial density of 0.5 at A600. The
butyricum has been clinically used for the treatment of several gastro- bacteria were harvested by centrifugation (3000 g × 5 min) and re-
intestinal diseases, such as antimicrobial-associated diarrhea and in- suspended in sterile phosphate-buffered saline (PBS) to a final experi-
flammatory bowel disease (IBD), without serious side effects in Asia mental concentration of 2 × 109 CFU/0.2 mL, which was selected
(particularly in Japan and China) [25–28]. A study conducted by Wang based on previous research and a preliminary experiment [39]. The
et al. revealed the stronger anti-inflammatory activities of combination supernatant was collected, filtered through 0.22-μm pore-size filters
of Lactobacillus acidophilus CGMCC 7282 with C. butyricum CGMCC and then diluted with complete culture medium.
7281 than either strain alone in both 2,4,6-trinitrobenzene sulfonic acid
(TNBS)-induced acute colitis in Sprague-Dawley rats and oxazolone-
2.2. Cell culture and treatment
induced chronic colitis in BALB/c mice [29]. C. butyricum has been
implicated in exacerbating cerebral ischemia/reperfusion injury [30]
Human CRC cell lines (HCT116, Caco-2, and HCT8) were purchased
and improving depression [31]. Moreover, its antitumor properties from the American Type Culture Collection (ATCC, Manassas, VA,
have also been observed in 1,2-dimethylhydrazine dihydrochloride
USA). HCT116 cells were cultured in Dulbecco's Modified Eagle
(DMH)-induced CRC and azoxymethane (AOM)+TNBS-induced colitis- Medium containing 10% fetal bovine serum (FBS) [40,41]; Caco-2 cells
associated cancer models [32,33]. Nevertheless, to our knowledge, the
were cultured in Minimum Essential Medium (MEM) with 20% FBS and
effect of C. butyricum on intestinal tumor development in a genetically 1.0% nonessential amino acids [42]; and HCT8 cells were cultured in
modified mouse model remains to be assessed, and its efficacy in reg-
Roswell Park Memorial Institute 1640 medium with 10% FBS [43]. All
ulating the gut microbiota has not been examined. More than 95% of the cells were incubated in a humidified atmosphere with 5% CO2 at
CRC develops from adenomas over a number of years, and this pro-
37 °C.
gression is known as the adenoma-adenocarcinoma sequence. A muta- The cell viability was determined using Cell Counting Kit-8 (CCK-8,
tion in the adenomatous polyposis coli (Apc) gene, which is a negative Beyotime, China). HCT116 and Caco-2 cells were plated in 96-well
regulator of Wnt/β-catenin signaling, is often an early event in the plates (4 × 103 cells/well [44]) and then treated with C. butyricum
adenoma-adenocarcinoma sequence [34]. The Apcmin/+ mouse model supernatant or sodium butyrate at different concentrations for various
carrying a mutation of the Apc gene can develop multiple intestinal times. After the treatments, 10 μL of CCK-8 assay solution was added to
adenomas and is therefore of particular advantage in intestinal tumor each well, and the cells were further incubated for 1 h at 37 °C. The
studies [35,36], and several studies have revealed that HFD-treated absorbance of each well was then measured at 450 nm.
Apcmin/+ mice develop more intestinal tumors [37,38]; therefore, the
HFD-treated Apcmin/+ mouse model was selected for our study.
2.3. Animal experiments
We addressed the efficacy and associated mechanisms of C. butyr-
icum on intestinal tumor development both in vitro and in vivo. Our
Female four-week-old Apcmin/+ mice purchased from the Animal
results show that C. butyricum suppresses intestinal tumor development,
Model Institute of Nanjing University were maintained in specific pa-
partly by modulating Wnt/β-catenin signaling and gut microbiota, and
thogen-free (SPF) facilities. All the mice (n = 30) were acclimatized for
these effects are accompanied by the repression of cell proliferation, the
7 days and then administered an antibiotic cocktail containing 500 mg
stimulation of apoptosis, a decrease in the fecal secondary BA contents,
of ampicillin, 250 mg of vancomycin, 500 mg of neomycin and 250 mg
increased SCFA production, and the activation of G-protein coupled
of metronidazole (Sigma-Aldrich, St. Louis, MO, USA) for 3 days to

457
D. Chen, et al. Cancer Letters 469 (2020) 456–467

deplete the indigenous microbiota and thus allow C. butyricum coloni- adenomas of intestinal tissues were stored at −80 °C.
zation. The experimental design is shown in Fig. 1A. The mice were
randomly divided into three groups: HFD-CB group, HFD-PBS group,
and BD-PBS group. The CB group was gavaged with C. butyricum at a 2.5. Histological and immunohistochemical analyses
dosage of 2 × 109 CFU/0.2 mL 3 times a week for 12 weeks, whereas
the PBS groups were orally administered 0.2 mL of sterile PBS 3 times a Formalin-fixed intestinal tissues were embedded in paraffin as
week for 12 weeks. In addition, the HFD groups were fed an HFD (60% previously described [3,46]. The degree of tumor dysplasia was as-
fat, 20% protein, and 20% carbohydrates), whereas the basal diet (BD) sessed based on previous research [21]. For immunohistochemistry, the
group was fed a BD (16% fat, 20% protein, and 64% carbohydrates). tissue slides were incubated with the following primary antibodies: Ki-
The rodents were monitored daily for any signs of illness and weighed 67 (ab16667, Abcam, Cambridge, MA, USA), β-catenin (ab32572,
weekly. The rodents were fasted for 12 h and euthanized by CO2 in- Abcam, Cambridge, MA, USA), cyclin D1 (ab134175, Abcam, Cam-
halation, and the cecal contents were harvested for SCFA analysis. Our bridge, MA, USA), GPR43 (ab118449, Abcam, Cambridge, MA, USA),
study was approved by the Institutional Animal Care and Use Com- and GPR109A (ab198693, Abcam, Cambridge, MA, USA). The sections
mittee at Tianjin Medical University, Tianjin, P. R. China. were then washed, incubated with horseradish peroxidase-labeled
secondary antibodies and counterstained with hematoxylin. Ki-67 and
cyclin D1 are expressed in the nucleus, inactivated β-catenin is located
2.4. Measurement of intestinal tumors and tissue collection in the cytomembrane, and ectopic β-catenin is located in the cytoplasm
and nucleus. GPR43 and GPR109A are expressed on the surface of in-
The intestinal tissues were collected as previously described testinal epithelial cells. Five random fields from a single section were
[38,45]. The small intestine was divided into three equal portions viewed to calculate the numbers of positive cells, and the data were
(proximal, middle and distal sections). The intestinal tumors were quantified based on the average percentages of positive cells.
classified as small (< 1 mm), middle (1–2 mm), or large (> 2 mm).
Intestinal Swiss rolls were fixed in 10% buffered formalin, and the

Fig. 1. Clostridium butyricum inhibited HFD-in-


duced intestinal tumor development in Apcmin/+
mice. (A) Experimental flow chart. (B) Total
number of intestinal tumors in mice after the dif-
ferent treatments. One-way ANOVA. (C) Number of
colonic tumors in Apcmin/+ mice. One-way ANOVA.
(D) CB decreased the intestinal tumor burden. One-
way ANOVA. (E) Numbers of intestinal tumors of
different sizes in the three groups. Two-way
ANOVA. (F–I) Representative and histological ap-
pearance of colonic tumors in the mice after the
different treatments. BD, basal diet. HFD, high-fat
diet. CB, Clostridium butyricum. Scale bars, 50 μm
*P < 0.05, **P < 0.01, ***P < 0.001. n = 10 for
each group.

458
D. Chen, et al. Cancer Letters 469 (2020) 456–467

2.6. TUNEL assay 2.12. Gene silencing of GPR43 using siRNA

Terminal deoxynucleotidyl transferase (TdT)-mediated dUTP nick To knock down GPR43 expression, the cells were transiently
end-labeling (TUNEL) was used for the detection of apoptosis in in- transfected with small interfering RNA that targets GPR43 (GPR43
testinal tumors [47]. Five areas in each section were selected randomly siRNA; sense strand, 5′-GGCCUGGGUUAUGUCCUUUTT-3′, antisense
to calculate the percentages of apoptotic cells. strand, 5′-AAAGGACAUAACCCAGGCCTT-3′; GenePharma, Shanghai,
China) and negative control siRNA (nontargeting siRNA; sense strand,
2.7. Realtime-polymerase chain reaction 5′-UUCUCCGAACGUGUCACGUTT-3′, antisense strand, 5′-ACGUGACA
CGUUCGGAGAATT-3′) as described previously [51]. CRC cells in 6-
Total RNA was isolated using the RNeasy mini kit (TIANGEN, well plates were transfected with 2 μg of GPR43 siRNA or control siRNA
Carlsbad, CA, USA) and reverse-transcribed using the TIANScript RT Kit using Lipofectamine 2000 (Invitrogen) in Opti-MEM according to the
(TIANGEN, Inc. Beijing, China). The oligonucleotide primer sequences manufacturer's instructions. After 6 h, the cells were refreshed with
for the target genes are shown in Table 1. The relative mRNA expres- complete medium for 24 h and then stimulated with C. butyricum su-
sion was calculated using the 2-ΔΔCT method. pernatant or sodium butyrate.

2.8. Western blot analysis


2.13. Collection of colonic specimens
Total proteins were extracted using RIPA buffer containing protease
Human colonic biopsies were obtained via colonoscopy at the
inhibitors (Solarbio, Beijing, China). The nuclear proteins were ex-
Digestive Endoscopy Center of Tianjin Medical University General
tracted for β-catenin analysis using the Nuclear Extraction Kit
Hospital, Tianjin, China. We analyzed 30 colonic biopsy samples, in-
(Beyotime, Shanghai, China). The membranes were incubated with
cluding normal tissue, adenoma tissues, and carcinoma tissues (10 pa-
appropriate primary antibodies against β-catenin (1:5000), cyclin D1
tients in each group). All the subjects were unrelated individuals of both
(1:10000), and c-Myc (ab32072, Abcam, Cambridge, MA, USA; 1:1000)
sexes aged 50–75 years and had consistent dietary habits. The exclusion
and then with appropriate secondary antibodies. β-actin (1:5000) and
criteria were as follows: i) prior surgery, chemotherapy, or radio-
histone 2A (1:5000) were selected as internal controls for total and
therapy; ii) diagnosis of IBD; and iii) presence of other malignant tu-
nuclear proteins, respectively. The proteins were quantified by densi-
mors. The participants were diagnosed by colonoscopy and patholo-
tometry using ImageJ software.
gical examination of colonic biopsies. Normal colonic tissues were
randomly obtained from a cluster of age- and sex-matched individuals
2.9. Illumina sequencing analysis
during health examinations involving colonoscopy of normal colonic
mucosa. The expression of GPR43 and GPR109A was examined by
16S rRNA gene sequencing was performed by the Realbio Genomics
immunohistochemical staining. Written informed consent was obtained
Institute (Shanghai, China). Total genomic DNA from frozen stool
from all the individuals, and the study was approved by the Ethics
samples was isolated using the QIAamp DNA Stool Mini Kit (Omega
Committee of General Hospital, Tianjin Medical University, China.
Bio-Tek, Norcross, GA, USA), and the V3–V4 hypervariable region of
the 16S rRNA genes was then amplified. The PCR products were col-
lected and sequenced using the Illumina HiSeq platform (Illumina, San
2.14. Statistical analysis
Diego, CA, USA). The high-quality reads were clustered into operational
taxonomic units (OTUs) based on sequences with ≥97% similarity and
The statistical analyses were performed using GraphPad Prism
then analyzed using the QIIME platform.
version 7.00. The measurement data are presented as the
means ± SDs. The data were statistically analyzed by one-way or two-
2.10. Fecal bile acid analysis way ANOVA. A P-value less than 0.05 was considered significant.

The main BA profiles in feces were determined by liquid chroma-


tography-mass spectrometry as previously described [48]. CA
(Aladdin), LCA (Aladdin), DCA (Sigma), and CDCA (Aladdin) were
added to the fecal samples as reference standards, and an external
Table 1
standard method was performed. Briefly, the fecal BAs were thoroughly
Primer sequences used for Realtime-PCR.
ground, suspended in chromatographic ethanol, and ultrasonically
processed at 30 °C for 60 min. All fecal samples were centrifuged at Primers Sequence
10,000 g and 4 °C for 10 min. The supernatant was concentrated to
hGPR41 Forward 5′- TGCGGTCAATGGGATCTG -3′
dryness under nitrogen, redissolved with chromatographic methanol, Reverse 5′- CAAGCACAGAGACGTAAGAGG -3′
and then filtered with a 0.22-μm pore-size filter. The BA concentration hGPR43 Forward 5′- TGTATGGAGTGATTGCAGCTC -3′
was quantified using a liquid chromatograph (Agilent 1260) combined Reverse 5′- TGGTTATCGGTGAAGTTCTCG -3′
with a 6120B mass spectrometer. hGPR109A Forward 5′- CACTCTCAGCTTCACCTACATG -3′
Reverse 5′- CTCACCTGTCATCTTCCTCTG -3′
hP21WAF1 Forward 5′- TGTCCGTCAGAACCCATGC -3′
2.11. Cecal short-chain fatty acid quantification Reverse 5′- AAAGTCGAAGTTCCATCGCTC -3′
hGAPDH Forward 5′- ACATCGCTCAGACACCATG -3′
Reverse 5′- TGTAGTTGAGGTCAATGAAGGG -3′
The SCFA concentrations were determined by gas chromatography
mGPR41 Forward 5′- GTGTAGTCTGTTGGTTCCTGG -3′
(GC) as previously described [49,50]. Briefly, the cecal contents were Reverse 5′- TGTAGGTTGCATTTCCCCAG -3′
diluted, acidified, and extracted ultrasonically on ice for 10 min. The mGPR43 Forward 5′- AGGTTTGCTACTGATCCGC -3′
samples were then centrifuged at 12,000 g and 4 °C for 15 min. After the Reverse 5′- GTACCCCTTCTGCTTGACTTC -3′
supernatant was mixed with ethyl acetate (1:1), the extract was filtered mGPR109A Forward 5′- TTTCTCCAGCCCATCTTTCC -3′
Reverse 5′- ATACTTCTGGTTGTGCTGGG -3′
through a 0.22-μm pore-size filter and poured into an Agilent 7890A
mGAPDH Forward 5′- GGAGAAACCTGCCAAGTATG -3′
Series GC. The SCFA standards were purchased from Sigma-Aldrich (St. Reverse 5′- TGGGAGTTGCTGTTGAAGTC -3′
Louis, MO, USA).

459
D. Chen, et al. Cancer Letters 469 (2020) 456–467

+
3. Results mice treated with C. butyricum developed adenoma with or without
low-grade dysplasia (Fig. 1F–I). In the animal experiments, we used PBS
3.1. Clostridium butyricum inhibited HFD-induced intestinal tumor as a control instead of heat-killed C. butyricum, which was a weakness of
development in Apcmin/+ mice our study. Isono A et al. reported that C. butyricum TO-A downregulated
the mRNA level of Toll-like receptor 4 (TLR4) in human colonic epi-
At the end of the experiment, the body weight of the HFD-PBS group thelial cells compared with untreated controls, but the mRNA level of
was increased compared with that of the BD-PBS group (P < 0.001; TLR4 was not significantly altered by heat-killed C. butyricum TO-A
Supplementary Fig. S1); however, no significant change in body weight [52]. Concordantly, we found that heat-killed C. butyricum supernatant
was observed between the HFD-CB and HFD-PBS groups. As shown in exerted no inhibitory effect on colon cancer cells, which suggests that
Fig. 1B, an HFD led to a significant increase in the total number of heat-killed C. butyricum has no anti-CRC actions (Supplementary Fig.
intestinal tumors (20.9 ± 3.78 vs 11.6 ± 2.2, P < 0.01), whereas C. S2). Collectively, these results demonstrate that C. butyricum prevents
butyricum prevented the formation of intestinal tumors (15 ± 2.10, HFD-induced intestinal tumor development.
P < 0.01). The number of colonic tumors in the HFD-CB group was
lower than that in the HFD-PBS group (1.2 ± 0.87 vs 2.8 ± 1.33,
P < 0.01; Fig. 1C). The intestinal tumor burden in the group fed the 3.2. Clostridium butyricum suppressed intestinal tumor cell proliferation
HFD alone was higher than that in the control group (28.9 ± 3.45 vs and stimulated their apoptosis
15.3 ± 2.69, P < 0.001; Fig. 1D); however, C. butyricum significantly
decreased the intestinal tumor burden compared with that found in the We subsequently evaluated whether C. butyricum supernatant could
HFD group (21.7 ± 4.27 vs 28.9 ± 3.45, P < 0.01). Moreover, the inhibit the proliferation of CRC cells using the CCK-8 assay. At a con-
number of intestinal tumors of different sizes in the HFD-PBS group was centration of 20%, C. butyricum supernatant significantly decreased the
also increased compared with that in the BD-PBS group; however, C. viability of HCT116 cells in a time-dependent manner compared with
butyricum significantly decreased the number of tumors sized > 1 mm that of the controls (P < 0.001; Fig. 2A); however, at a relatively low
(Fig. 1E). High-grade dysplasia in the colon, including intramucosal concentration, that is, 10%, C. butyricum supernatant exerted almost no
carcinoma, was observed in 70% of mice fed the HFD, whereas Apcmin/ effects on cell viability. The experiment with Caco-2 cells revealed that
C. butyricum supernatant at a concentration of 20% decreased the cell

Fig. 2. Clostridium butyricum suppressed the proliferation and promoted the apoptosis of intestinal tumor cells. (A) HCT116 and Caco-2 cells were treated
with different concentrations of CB supernatant or sodium butyrate for different times, and their viability was analyzed using the CCK-8 assay. Two-way ANOVA. (B)
The intestinal sections were processed for Ki-67 immunohistochemistry. One-way ANOVA. (C) The TUNEL assay was used for the assessment of apoptosis in intestinal
tumors. One-way ANOVA. NC, normal control. CB, Clostridium butyricum. NaBu, sodium butyrate. BD, basal diet. HFD, high-fat diet. Scale bars, 50 μm *P < 0.05,
**P < 0.01, ***P < 0.001. n = 10 for each group.

460
D. Chen, et al. Cancer Letters 469 (2020) 456–467

viability for 48 and 72 h (P < 0.001; Fig. 2A). Sodium butyrate, which P < 0.05; Fig. 3A and B). Furthermore, a Western blot analysis re-
was used as a positive control, concentration-dependently (0, 0.1, 1, vealed decreased expression of β-catenin, cyclin D1, and c-Myc in C.
and 10 mmol/L) decreased the viability of CRC cells with an unclear butyricum-treated HCT116 cells compared with the controls
dependence on time. Because sodium butyrate at a concentration of (P < 0.001; Fig. 3C and D). Thus, the downregulation of Wnt signaling
1 mmol/L was enough to exert obvious inhibitory effects on the pro- might be a critical mechanism involved in its anticarcinogenic effects.
liferation of HCT116 cells, and according to our GC data
(Supplementary Fig. S3), C. butyricum supernatant at a concentration of
20% contained approximately 0.5 mmol/L butyrate, which was on the 3.4. Clostridium butyricum altered the HFD-induced gut microbiota
same order of magnitude as 1 mmol/L sodium butyrate. Thus, C. bu- dysbiosis in Apcmin/+ mice
tyricum supernatant at a concentration of 20% and 1 mmol/L sodium
butyrate were selected for cell stimulation in the subsequent experi- The comparison of the OTUs among the three groups revealed 294
ments. The proliferation and apoptosis of intestinal tumor cells in mice OTUs in the HFD-CB group, 332 OTUs in the HFD-PBS group, and 380
were quantified by Ki-67 immunohistochemistry and the TUNEL assay, OTUs in the BD-PBS group, and a total of 222 OTUs were shared by the
respectively. Compared with the HFD-PBS group, the C. butyricum- different samples (Fig. 4A). The gut microbiota of all the samples was
treated group exhibited a significant decreased Ki-67-positive rate dominated by four major phyla: Bacteroidetes, Firmicutes, Proteobacteria,
(20.84 ± 3.50 vs 38.38 ± 4.25, P < 0.01; Fig. 2B) and an increased and Verrucomicrobia. Notably, compared with the control, a lower
percentage of apoptotic cells (7.99 ± 0.24 vs 1.81 ± 0.12, abundance of Bacteroidetes and a higher abundance of Firmicutes were
P < 0.001; Fig. 2C) within the colon tumors. These results show that C. observed in mice fed an HFD (Fig. 4B), which resulted in an increased
butyricum significantly suppresses the proliferation of intestinal tumor Firmicutes/Bacteroidetes (F/B) ratio in the HFD feeding alone group
cells and stimulates their apoptosis. (1.2) compared with that in the control group (0.4). However, C. bu-
tyricum did not statistically affect the F/B ratio. The genus-level analysis
revealed that HFD increased the relative abundance of pathogenic
3.3. Clostridium butyricum modulated the Wnt/β-catenin signaling pathway bacteria, such as Desulfovibrio, Odoribacter, and Helicobacter, whereas C.
butyricum weakened this increase (Fig. 4C and Supplementary Figs.
An aberrant Wnt/β-catenin signaling pathway is positively relevant S4A–S4C). Interestingly, the relative abundance of BA-biotransforming
to intestinal carcinogenesis, and this pathway has been considered a bacteria in mice fed the HFD alone was higher than that in mice fed a
potential therapeutic target [53]. Compared with the HFD-PBS mice, control diet, such as Clostridium XlVb and Clostridium XI, whereas C.
the HFD-CB mice showed lower ectopic expression of β-catenin in the butyricum decreased the abundance of these bacteria, but this decrease
nucleus (28.98 ± 1.55 vs 63.12 ± 6.04, P < 0.05) and a decreased was not statistically significant (Fig. 4C and Supplementary Figs. S4D
proportion of cyclin D1-positive cells (27.73 ± 1.79 vs 42.80 ± 5.58, and S4E). Additionally, a relatively lower abundance of Lactobacillus

Fig. 3. Clostridium butyricum suppressed the Wnt/β-catenin signaling pathway. (A–B) An immunohistochemistry analysis of intestinal tumors revealed that the
Wnt/β-catenin pathway was inhibited in Apcmin/+ mice that received CB. The β-catenin-positive cells in A indicate nuclear staining indicative of Wnt activation. One-
way ANOVA. (C–D) The protein levels of total and nuclear β-catenin, cyclin D1, and c-Myc in HCT116 cells were analyzed by western blotting using anti-β-actin and
anti-histone 2A antibodies as internal controls for total and nuclear protein, respectively. The proteins were quantified densitometrically using ImageJ software. Two-
way ANOVA. BD, basal diet. HFD, high-fat diet. CB, Clostridium butyricum. NaBu, sodium butyrate. Scale bars, 50 μm *P < 0.05, ***P < 0.001. n = 10 for each
group.

461
D. Chen, et al. Cancer Letters 469 (2020) 456–467

Fig. 4. Clostridium butyricum modulated the composition of the gut microbiota in Apcmin/+ mice. (A) Venn diagrams of bacterial OTUs. (B) Bar charts of the gut
microbiota composition at the phylum level in the three groups. (C) Bar charts of the gut microbiota composition at the genus level in each mouse. (D–E) Shannon
and Simpson diversity indexes of the gut microbiota. Kruskal-Wallis test. BD, basal diet. HFD, high-fat diet. CB, Clostridium butyricum. NS, not significant. *P < 0.05.
n = 8–10. The analyses were performed at the end of the experiment.

was observed in mice fed an HFD compared with that of the control, Taken together, the results indicate that HFD induces a major alteration
whereas the abundance of Lactobacillus was restored by C. butyricum, in the gut microbiota composition, whereas C. butyricum at least partly
but this effect was not statistically significant (Fig. 4C and alters the gut microbiota dysbiosis caused by HFD.
Supplementary Fig. S4F).
HFD led to a significant decrease in the Shannon and Simpson in- 3.5. Clostridium butyricum altered the levels of microbial-derived
dices with respect to the control values (Shannon: 5.05 ± 0.22 vs metabolites
5.52 ± 0.34, P < 0.05; Simpson: 0.94 ± 0.01 vs 0.95 ± 0.01,
P < 0.05; Fig. 4D–E); however, the effect of C. butyricum on those two Numerous studies have revealed the relationships between micro-
indexes was not statistically significant, which suggested that HFD ex- bial-derived metabolites (BAs and SCFAs) and CRC [54–56]. The HFD
erted stronger negative effects on the α diversity of the gut microbiota. induced a subtle increase in the concentration of CA compared with the
An analysis of similarities (ANOSIM) based on weighted UniFrac dis- BD, but no difference in the level of CDCA was found between these
tances indicated the existence of quite different gut microbiota struc- groups (Fig. 6A). In addition, primary BAs were not affected by C. bu-
tures in the three groups (R = 0.63, P = 0.001; Fig. 5A). A principal tyricum treatment (Fig. 6A). The levels of the fecal secondary BAs DCA
coordinate analysis (PCoA) based on weighted UniFrac distances re- and LCA were markedly increased in the HFD-treated mice compared
vealed a more similar structure between HFD-CB and HFD-PBS mice with the control (DCA: 0.63 ± 0.16 vs 0.17 ± 0.03 mg/g, P < 0.001;
(Fig. 5B). We compared the gut microbiota among the three groups LCA: 0.14 ± 0.02 vs 0.10 ± 0.02 mg/g, P < 0.01; Fig. 6A). Sur-
through linear discriminant analysis effect size (LEfSe) to identify the prisingly, the DCA and LCA levels in the C. butyricum group were de-
specific microbiota linked to C. butyricum treatment. Ruminococcaceae creased significantly (DCA: 0.46 ± 0.04 vs 0.63 ± 0.16 mg/g,
and Eubacterium, whose members are generally associated with buty- P < 0.05; LCA: 0.10 ± 0.01 vs 0.14 ± 0.02 mg/g, P < 0.01;
rate production, were more abundant in the HFD-CB group (Fig. 5C). Fig. 6A). The concentrations of cecal SCFAs in the HFD-fed group were

462
D. Chen, et al. Cancer Letters 469 (2020) 456–467

Fig. 5. Clostridium butyricum altered specific bacterial taxa. (A) ANOSIM based on weighted UniFrac distances. (B) A PCoA based on weighted UniFrac distances
indicated a clear difference in the gut microbiota structure between the HFD-treated and non-HFD-treated groups. (C) Cladogram generated from the LEfSe. BD, basal
diet. HFD, high-fat diet. CB, Clostridium butyricum. n = 8–10. The analyses were performed at the end of the experiment.

increased compared with those in the control group. Notably, the levels treated HCT116 cells compared with the group treated with 1 mmol/L
of acetic acid (1671.6 ± 3199.5 vs 238.9 ± 72.3 μmol/g, sodium butyrate (P < 0.01; Fig. 7A), but no difference in the mRNA
P < 0.001), propionic acid (447.9 ± 112.5 vs 34.9 ± 8.8 μmol/g, levels of GPR43 and GPR41 was found between these groups. Similar
P < 0.001), and butyric acid (373.7 ± 96.2 vs 27.7 ± 6.8 μmol/g, results were found in Caco-2 and HCT8 cells (Supplementary Fig. S5).
P < 0.001) in the C. butyricum-treated mice were significantly higher Furthermore, we investigated whether the anticarcinogenic effect of C.
than those in the HFD-fed mice (Fig. 6B). These results suggest that the butyricum was dependent on GPR43 through siRNA-mediated silencing
anticarcinogenic effect of C. butyricum might be partly due to altera- of the gene encoding the GPR43 receptor and found that the gene si-
tions in microbial-derived metabolites. lencing of GPR43 blunted the anti-proliferative effect of C. butyricum
(Fig. 7B). In addition, marked decreases in the mRNA levels of GPR43
were found in HCT116 cells treated with GPR43 siRNA (Fig. 7C). The
3.6. Clostridium butyricum activated G-protein coupled receptors cyclin-dependent kinase inhibitor P21WAF1 acts as an important in-
hibitor of cell cycle progression [59]. The mRNA level of P21WAF1 was
SCFAs induce multiple signaling pathways partly through their increased in HCT116 cells treated with C. butyricum, and the silencing
binding to GPRs such as GPR41, GPR43, and GPR109A with varying of GPR43 blocked the C. butyricum-mediated induction of P21WAF1 ex-
affinities [57]. GPR43 and GPR109A recognize SCFAs and have been pression (Fig. 7C). The mRNA levels of GPRs in the HFD-PBS group
implicated in the suppression of intestinal inflammation and CRC [58]. exhibited no significant change compared with those in the BD-PBS
Compared with the control, the stimulation of HCT116 cells with C. group (Fig. 7D). However, the mRNA levels of GPR43 and GPR109A in
butyricum supernatant and sodium butyrate induced 4.5-fold and 4.2- the HFD-CB group were upregulated by 3.89-fold and 3.8-fold, re-
fold upregulation of the GPR43 mRNA levels, 2.6-fold and 7.0-fold in- spectively, compared with those in the HFD-PBS group (P < 0.05), but
creases in the GPR109A mRNA levels, and no significant change in no change in the GPR41 mRNA level was found between these groups
GPR41 mRNA expression, respectively (Fig. 7A). The GPR109A mRNA (Fig. 7D). We then examined the expression of GPR43 and GPR109A in
level was downregulated 2.7-fold in the C. butyricum supernatant-

463
D. Chen, et al. Cancer Letters 469 (2020) 456–467

Fig. 6. Clostridium butyricum altered the levels of microbial-derived metabolites. (A) Effect of CB on primary BAs and secondary BAs in the fecal contents. One-
way ANOVA. (B) Alterations in the SCFA levels in the cecal contents of mice after the different treatments. One-way ANOVA. BD, basal diet. HFD, high-fat diet. CB,
Clostridium butyricum. BAs, bile acids. CA, cholic acid. CDCA, chenodeoxycholic acid. DCA, deoxycholic acid. LCA, lithocholic acid. SCFAs, short-chain fatty acids.
*P < 0.05, **P < 0.01, ***P < 0.001. n = 8–10. The analyses were performed at the end of the experiment.

human colonic tissues and found that the expression of GPR43 and which is different from the results obtained by Chen et al. using C57BL/
GPR109A showed a gradual decrease from human normal colonic tissue 6 mice treated with DMH [32]. C. butyricum represses the proliferation
to adenoma to carcinoma (Fig. 7E). The percentages of GPR43-and and enhances the apoptosis of intestinal tumor cells both in vitro and in
GPR109A-positive cells in carcinoma tissues were decreased compared vivo. Moreover, to our knowledge, our study is the first to focus on the
with those in adenoma tissues (GPR43: 13.84 ± 2.86% vs efficacy of C. butyricum in regulating the gut microbiota in a CRC mouse
27.50 ± 3.75%, P < 0.05; GPR109A: 19.46 ± 3.22% vs model. The novel finding of our study is that C. butyricum can meta-
30.70 ± 3.74%, P < 0.05; Fig. 7E). These results indicate that the bolize fat to produce SCFAs, which have been implicated in inhibiting
activation of GPR43 and GPR109A plays a critical role in the antic- CRC [66], and thus significantly alleviate the tumor burden caused by
arcinogenic effects of C. butyricum. HFD.
The dysregulation of Wnt signaling is considered a crucial event in
4. Discussion CRC progression [67–69]. A mutation in the Apc gene leads to increased
cytoplasmic accumulation and nuclear translocation of β-catenin [70].
Currently, the restoration of an altered gut microbiota using pro- β-catenin interacts with the T cell factor/lymphoid enhancer factor and
biotics is considered a potential strategy for the prevention and treat- thereby increases the expression of its target genes, such as cyclin D1
ment of CRC [60,61]. Butyrate-producing bacteria exhibit therapeutic and c-Myc, which have been further linked to aberrant cell proliferation
potential for CRC because these bacteria are diminished in the fecal and apoptosis [71]. This study shows that Apcmin/+ mice fed an HFD
microbiota of patients with CRC [62,63]. However, whether C. butyr- exhibit increased ectopic accumulation of β-catenin and upregulated
icum can inhibit intestinal tumor development in genetically modified expression of cyclin D1 and that these effects are significantly decreased
models and its efficacy on the gut microbiota remain uncertain. As by C. butyricum. Recent studies demonstrated that some harmful bac-
demonstrated in our study, C. butyricum inhibits intestinal tumor de- teria can activate Wnt/β-catenin through bacterial effectors and toxins,
velopment in Apcmin/+ mice induced by HFD. Moreover, the mechan- such as Helicobacter pylori, Salmonella enterica, Fusobacterium nucleatum,
isms involved in the protective effects of C. butyricum are related to and Bacteroides fragilis [72,73]. In our study, HFD increases the relative
suppressing the proliferation and promoting the apoptosis of tumor abundance of Helicobacter, and C. butyricum weakens this increase, but
cells, modulating the gut microbiota, and inhibiting Wnt/β-catenin further studies are needed to investigate whether C. butyricum sup-
signaling. Importantly, C. butyricum treatment alters microbial-derived presses Wnt signaling directly or through other mediators.
metabolites such as secondary BAs and SCFAs and activates the G- Numerous studies have described gut dysbiosis in CRC. Hence, we
protein coupled receptors GPR43 and GPR109A. Therefore, C. butyr- analyzed the microbial diversity of fecal samples from the different
icum might exert protective effects against CRC. treatment groups and found that C. butyricum partly alters the gut mi-
The antitumor activity of C. butyricum in CRC cell lines and a che- crobiota dysbiosis caused by HFD. In agreement with previous reports
mically induced CRC mouse model was recently reported [32,64]. C. [74,75], the present study shows that HFD feeding results in an in-
butyricum increases P21WAF1 expression in C57BL/6 mice in- creased F/B ratio. C. butyricum not only inhibits the increase in pa-
traperitoneally injected with DMH, which suggests that it exerts an thogenic bacteria but also enhances the growth of probiotic bacteria.
antiproliferative effect [32]. The efficacy of C. butyricum for the treat- Previous studies have revealed that members of Clostridium biotrans-
ment of colitis-associated cancer induced by AOM/TNBS was also ob- form primary BAs into secondary BAs via 7α-dehydroxylation [76–78],
served in a previous study [33]. HFD has been recognized as a major and intestinal Bacteroidetes are negatively correlated with secondary
environmental risk factor for CRC [65]. Concordantly, our study reveals BAs [79]. Consistent with these findings, we found a decreased abun-
that C. butyricum ameliorates the development of intestinal tumors and dance of Clostridium and an increased abundance of Bacteroidetes in the
improves the morphological abnormalities in HFD-fed Apcmin/+ mice, mice administered C. butyricum. Interestingly, we also observed

464
D. Chen, et al. Cancer Letters 469 (2020) 456–467

Fig. 7. Clostridium butyricum activated G-protein coupled receptors and increased P21WAF1 expression. (A) Relative expression of GPR41, GPR43, and
GPR109A in HCT116 cells. One-way ANOVA. (B) The anti-proliferative activity of C. butyricum was inhibited by siRNA-mediated GPR43 silencing. One-way ANOVA.
(C) Expression of GPR43 and P21WAF1 in GPR43-knockdown HCT116 cells relative to the negative control. One-way ANOVA. (D) Relative expression of GPR41,
GPR43, and GPR109A in colonic tumor tissues. One-way ANOVA. (E) Immunohistochemical detection of GPR43 and GPR109A in human colonic tissues. One-way
ANOVA. n = 10 for each group. CB, Clostridium butyricum. NaBu, sodium butyrate. BD, basal diet. HFD, high-fat diet. ns, not significant. Scale bars, 50 μm
*P < 0.05, **P < 0.01, ***P < 0.001. The analyses were performed at the end of the experiment.

decreased levels of secondary BAs in C. butyricum-treated mice. Taken inhibits cell proliferation and induces apoptosis following butyrate
together, the results indicate that the positive effect exerted by C. bu- treatment [85,86]. GPR109A suppresses NF-κB activation, down-
tyricum on the gut microbiota might contribute to its inhibitory impact regulates cyclin D1, and upregulates the death receptor pathway [85].
on CRC. Interestingly, we also found that C. butyricum increases the expression
An accumulating body of evidence demonstrates that a high dietary of GPR43 and GPR109A in colonic tissue. The siRNA-mediated gene
fiber intake is related to a lower risk of colorectal carcinogenesis [80]. silencing of GPR43 blunts the anti-proliferative effect of C. butyricum
This protective effect of dietary fiber might be attributable to the an- and blocks the C. butyricum-mediated induction of P21WAF1 expression
ticarcinogenic properties of beneficial microbial metabolic products, in CRC cells, and this finding indicates that the protective role of C.
such as SCFAs [81]. Notably, Ruminococcaceae and Eubacterium, which butyricum might be related to the activation of GPR43. Moreover, the
are well known to produce SCFAs, are increased after C. butyricum results from the immunohistochemical analysis of human colonic tis-
treatment. In addition, C. butyricum obviously increases the con- sues also suggest that GPRs play a role in inhibiting carcinoma pro-
centration of each acid, as expected. Butyrate can influence multiple gression. Further studies are needed to investigate whether C. butyricum
intracellular signaling pathways in the intestine partly through binding exerts anti-tumor effects in GPR43/109A-knockout Apcmin/+ mice.
to specific butyrate receptors, namely, GPR41, GPR43, and GPR109A Taken together, our results suggest that C. butyricum suppresses
[82–84]. GPR41, GPR43, and GPR109A can be activated by butyrate, intestinal tumor development at least partly by modulating Wnt/β-ca-
although with varying affinities. The activation of GPR43 and GPR109A tenin signaling and the gut microbiota. Thus, C. butyricum might be

465
D. Chen, et al. Cancer Letters 469 (2020) 456–467

considered a promising option in the treatment of CRC. promotes intestinal carcinogenesis, Mol. Carcinog. 58 (2019) 1155–1167.
[22] S.L. Long, G. CGM, S.A. Joyce, Interactions between gut bacteria and bile in health
and disease, Mol. Asp. Med. 56 (2017) 54–65.
Author contributions [23] T. Fu, S. Coulter, E. Yoshihara, et al., FXR regulates intestinal cancer stem cell
proliferation, Cell 176 (2019) 1098–1112 e18.
Chen DF, Jin DC, Wang BM, and Cao HL designed the research [24] N. Cassir, S. Benamar, B. La Scola, Clostridium butyricum: from beneficial to a new
emerging pathogen, Clin. Microbiol. Infect. 22 (2016) 37–45.
study; Chen DF, Jin DC, Huang SM, Wu JY, Xu MQ, Liu TY, Dong WX, [25] H. Seki, M. Shiohara, T. Matsumura, et al., Prevention of antibiotic-associated
Liu X, Wang SN, Zhong WL, Liu Y, Jiang RH, and Piao MY performed diarrhea in children by Clostridium butyricum MIYAIRI, Pediatr. Int. 45 (2003)
the experiments; Chen DF, Jin DC, Huang SM, and Cao HL wrote the 86–90.
[26] A. Hayashi, T. Sato, N. Kamada, et al., A single strain of Clostridium butyricum
paper; and all the authors approved the final draft of the manuscript. induces intestinal IL-10-producing macrophages to suppress acute experimental
colitis in mice, Cell Host Microbe 13 (2013) 711–722.
Funding sources [27] S. Mo, B.S. Kim, S.J. Yun, et al., Genome sequencing of Clostridium butyricum DKU-
01, isolated from infant feces, Gut Pathog. 7 (2015) 8.
[28] A. Yasueda, T. Mizushima, R. Nezu, et al., The effect of Clostridium butyricum
This study was supported by grants (81570478, 81741075, and MIYAIRI on the prevention of pouchitis and alteration of the microbiota profile in
81700456) from the National Natural Science Foundation of China, a patients with ulcerative colitis, Surg. Today 46 (2016) 939–949.
grant (17JCYBJC24900) from the Natural Science Foundation of [29] Y. Wang, Y. Gu, K. Fang, et al., Lactobacillus acidophilus and Clostridium butyricum
ameliorate colitis in murine by strengthening the gut barrier function and de-
Tianjin, a grant (2019M651049) from the Postdoctoral Science creasing inflammatory factors, Benef. Microbes 9 (2018) 775–787.
Foundation of China, and a grant (2018242755) from the Medical and [30] J. Sun, F. Wang, Z. Ling, et al., Clostridium butyricum attenuates cerebral ischemia/
Health Science and Technology Project of Zhejiang. reperfusion injury in diabetic mice via modulation of gut microbiota, Brain Res.
1642 (2016) 180–188.
[31] T. Miyaoka, M. Kanayama, R. Wake, et al., Clostridium butyricum MIYAIRI 588 as
Declaration of competing interest adjunctive therapy for treatment-resistant major depressive disorder: a prospective
open-label trial, Clin. Neuropharmacol. 41 (2018) 151–155.
[32] Z.F. Chen, L.Y. Ai, J.L. Wang, et al., Probiotics Clostridium butyricum and Bacillus
The authors declare no conflicts of interest. subtilis ameliorate intestinal tumorigenesis, Future Microbiol. 10 (2015)
1433–1445.
Appendix A. Supplementary data [33] Y. Xiao, X. Dai, K. Li, et al., Clostridium butyricum partially regulates the devel-
opment of colitis-associated cancer through miR-200c, Cell. Mol. Biol. (Noisy-Le-
Grand) 63 (2017) 59–66.
Supplementary data to this article can be found online at https:// [34] A. Klaus, W. Birchmeier, Wnt signalling and its impact on development and cancer,
doi.org/10.1016/j.canlet.2019.11.019. Nat. Rev. Cancer 8 (2008) 387–398.
[35] Y. Yamada, H. Mori, Multistep carcinogenesis of the colon in Apc(Min/+) mouse,
Cancer Sci. 98 (2007) 6–10.
References [36] R. Jackstadt, O.J. Sansom, Mouse models of intestinal cancer, J. Pathol. 238 (2016)
141–151.
[1] R.L. Siegel, K.D. Miller, A. Jemal, Cancer statistics, CA A Cancer J. Clin. 2018 (68) [37] K.A. Baltgalvis, F.G. Berger, M.M. Peña, et al., The interaction of a high-fat diet and
(2018) 7–30. regular moderate intensity exercise on intestinal polyp development in Apc Min/+
[2] D. Wang, R. Zhao, Y.Y. Qu, et al., Colonic lysine homocysteinylation induced by mice, Cancer Prev. Res. 2 (2009) 641–649.
high-fat diet suppresses DNA damage repair, Cell Rep. 25 (2018) 398–412 e6. [38] A. Saxena, R. Tammali, K.V. Ramana, et al., Aldose reductase inhibitor, fidarestat
[3] H. Cao, M. Xu, W. Dong, et al., Secondary bile acid-induced dysbiosis promotes prevents high-fat diet-induced intestinal polyps in ApcMin/+ mice, Curr. Cancer
intestinal carcinogenesis, Int. J. Cancer 140 (2017) 2545–2556. Drug Targets 18 (2018) 905–911.
[4] D. Dermadi, S. Valo, S. Ollila, et al., Western diet deregulates bile acid homeostasis, [39] Y. Shi, L.Z. Xu, K. Peng, et al., Specific immunotherapy in combination with
cell proliferation, and tumorigenesis in colon, Cancer Res. 77 (2017) 3352–3363. Clostridium butyricum inhibits allergic inflammation in the mouse intestine, Sci.
[5] Yilmaz ÖH, Dietary regulation of the origins of cancer, Sci. Transl. Med. (2018) 10. Rep. 5 (2015) 17651.
[6] K. Makki, E.C. Deehan, J. Walter, et al., The impact of dietary fiber on gut micro- [40] J. Liu, C. Mu, W. Yue, et al., A diterpenoid derivate compound targets selenocys-
biota in host health and disease, Cell Host Microbe 23 (2018) 705–715. teine of thioredoxin reductases and induces Bax/Bak-independent apoptosis, Free
[7] S.V. Lynch, O. Pedersen, The human intestinal microbiome in health and disease, N. Radic. Biol. Med. 63 (2013) 485–494.
Engl. J. Med. 375 (2016) 2369–2379. [41] T. Lu, Z. Zhu, J. Wu, et al., DRAM1 regulates autophagy and cell proliferation via
[8] D. Chen, J. Wu, D. Jin, et al., Fecal microbiota transplantation in cancer manage- inhibition of the phosphoinositide 3-kinase-Akt-mTOR-ribosomal protein S6
ment: current status and perspectives, Int. J. Cancer (2018). pathway, Cell Commun. Signal. 17 (2019) 28.
[9] A.M. Thomas, P. Manghi, F. Asnicar, et al., Metagenomic analysis of colorectal [42] T. Satoh, S. Sugiura, K. Shin, et al., A multi-throughput multi-organ-on-a-chip
cancer datasets identifies cross-cohort microbial diagnostic signatures and a link system on a plate formatted pneumatic pressure-driven medium circulation plat-
with choline degradation, Nat. Med. 25 (2019) 667–678. form, Lab Chip 18 (2017) 115–125.
[10] H.M. Chen, Y.N. Yu, J.L. Wang, et al., Decreased dietary fiber intake and structural [43] C. Yan, L. Luo, C.Y. Guo, et al., Doxorubicin-induced mitophagy contributes to drug
alteration of gut microbiota in patients with advanced colorectal adenoma, Am. J. resistance in cancer stem cells from HCT8 human colorectal cancer cells, Cancer
Clin. Nutr. 97 (2013) 1044–1052. Lett. 388 (2017) 34–42.
[11] S.J. Bultman, Emerging roles of the microbiome in cancer, Carcinogenesis 35 [44] Y. Peng, L. Qiu, D. Xu, et al., M4IDP, a zoledronic acid derivative, induces G1 arrest,
(2014) 249–255. apoptosis and autophagy in HCT116 colon carcinoma cells via blocking PI3K/Akt/
[12] M.B. Burns, J. Lynch, T.K. Starr, et al., Virulence genes are a signature of the mi- mTOR pathway, Life Sci. 185 (2017) 63–72.
crobiome in the colorectal tumor microenvironment, Genome Med. 7 (2015) 55. [45] H. Cao, S. Luo, M. Xu, et al., The secondary bile acid, deoxycholate accelerates
[13] S. Hu, L. Liu, E.B. Chang, et al., Butyrate inhibits pro-proliferative miR-92a by di- intestinal adenoma-adenocarcinoma sequence in Apc (min/+) mice through en-
minishing c-Myc-induced miR-17-92a cluster transcription in human colon cancer hancing Wnt signaling, Fam. Cancer 13 (2014) 563–571.
cells, Mol. Cancer 14 (2015) 180. [46] R. Xie, Y. Sun, J. Wu, et al., Maternal high fat diet alters gut microbiota of offspring
[14] S. Bullman, C.S. Pedamallu, E. Sicinska, et al., Analysis of Fusobacterium persis- and exacerbates DSS-induced colitis in adulthood, Front. Immunol. 9 (2018) 2608.
tence and antibiotic response in colorectal cancer, Science 358 (2017) 1443–1448. [47] H. Esen, F. Erdi, B. Kaya, et al., Tissue thioredoxin reductase-1 expression in as-
[15] Y. Furusawa, Y. Obata, S. Fukuda, et al., Commensal microbe-derived butyrate trocytomas of different grades, J. Neuro Oncol. 121 (2015) 451–458.
induces the differentiation of colonic regulatory T cells, Nature 504 (2013) [48] M. Hagio, M. Matsumoto, M. Fukushima, et al., Improved analysis of bile acids in
446–450. tissues and intestinal contents of rats using LC/ESI-MS, J. Lipid Res. 50 (2009)
[16] E.E. Canfora, M. RCR, K. Venema, et al., Gut microbial metabolites in obesity, 173–180.
NAFLD and T2DM, Nat. Rev. Endocrinol. 15 (2019) 261–273. [49] C. Zhang, J.M. Monk, J.T. Lu, et al., Cooked navy and black bean diets improve
[17] J. Yang, J. Yu, The association of diet, gut microbiota and colorectal cancer: what biomarkers of colon health and reduce inflammation during colitis, Br. J. Nutr. 111
we eat may imply what we get, Protein Cell 9 (2018) 474–487. (2014) 1549–1563.
[18] R. Simeoli, R.G. Mattace, C. Pirozzi, et al., An orally administered butyrate-re- [50] J. Yang, C. Ding, X. Dai, et al., Soluble dietary fiber ameliorates radiation-induced
leasing derivative reduces neutrophil recruitment and inflammation in dextran intestinal epithelial-to-mesenchymal transition and fibrosis, JPEN - J. Parenter.
sulphate sodium-induced murine colitis, Br. J. Pharmacol. 174 (2017) 1484–1496. Enter. Nutr. 41 (2017) 1399–1410.
[19] X. Long, M. Li, L.X. Li, et al., Butyrate promotes visceral hypersensitivity in an IBS- [51] Z. Xu, Y. Zhang, J. Jiang, et al., Epidermal growth factor induces HCCR expression
like model via enteric glial cell-derived nerve growth factor, Neuro Gastroenterol. via PI3K/Akt/mTOR signaling in PANC-1 pancreatic cancer cells, BMC Canc. 10
Motil. 30 (2018) e13227. (2010) 161.
[20] W. Wang, J. Wang, J. Li, et al., Cholecystectomy damages aging-associated in- [52] A. Isono, T. Katsuno, T. Sato, et al., Clostridium butyricum TO-A culture super-
testinal microbiota construction, Front. Microbiol. 9 (2018) 1402. natant downregulates TLR4 in human colonic epithelial cells, Dig. Dis. Sci. 52
[21] S. Wang, W. Dong, L. Liu, et al., Interplay between bile acids and the gut microbiota (2007) 2963–2971.
[53] R. Nusse, H. Clevers, Wnt/β-Catenin signaling, disease, and emerging therapeutic

466
D. Chen, et al. Cancer Letters 469 (2020) 456–467

modalities, Cell 169 (2017) 985–999. 469–480.


[54] P. Louis, G.L. Hold, H.J. Flint, The gut microbiota, bacterial metabolites and col- [71] E. Ashihara, T. Takada, T. Maekawa, Targeting the canonical Wnt/β-catenin
orectal cancer, Nat. Rev. Microbiol. 12 (2014) 661–672. pathway in hematological malignancies, Cancer Sci. 106 (2015) 665–671.
[55] A.N. Thorburn, L. Macia, C.R. Mackay, Diet, metabolites, and "western-lifestyle" [72] A. Gagnaire, B. Nadel, D. Raoult, et al., Collateral damage: insights into bacterial
inflammatory diseases, Immunity 40 (2014) 833–842. mechanisms that predispose host cells to cancer, Nat. Rev. Microbiol. 15 (2017)
[56] J. Suez, E. Elinav, The path towards microbiome-based metabolite treatment, Nat 109–128.
Microbiol 2 (2017) 17075. [73] M.R. Rubinstein, J.E. Baik, S.M. Lagana, et al., Fusobacterium nucleatum promotes
[57] A. Koh, F. De Vadder, P. Kovatcheva-Datchary, et al., From dietary fiber to host colorectal cancer by inducing Wnt/β-catenin modulator Annexin A1, EMBO Rep.
physiology: short-chain fatty acids as key bacterial metabolites, Cell 165 (2016) (2019) 20.
1332–1345. [74] A. Cotillard, S.P. Kennedy, L.C. Kong, et al., Dietary intervention impact on gut
[58] Y. Tian, Q. Xu, L. Sun, et al., Short-chain fatty acids administration is protective in microbial gene richness, Nature 500 (2013) 585–588.
colitis-associated colorectal cancer development, J. Nutr. Biochem. 57 (2018) [75] Z. Gao, B. Guo, R. Gao, et al., Microbiota disbiosis is associated with colorectal
103–109. cancer, Front. Microbiol. 6 (2015) 20.
[59] A.L. Gartel, S.K. Radhakrishnan, Lost in transcription: p21 repression, mechanisms, [76] J.M. Ridlon, D.J. Kang, P.B. Hylemon, et al., Bile acids and the gut microbiome,
and consequences, Cancer Res. 65 (2005) 3980–3985. Curr. Opin. Gastroenterol. 30 (2014) 332–338.
[60] P. Ambalam, M. Raman, R.K. Purama, et al., Probiotics, prebiotics and colorectal [77] N. Hartmann, M. Kronenberg, Cancer immunity thwarted by the microbiome,
cancer prevention, Best Pract. Res. Clin. Gastroenterol. 30 (2016) 119–131. Science 360 (2018) 858–859.
[61] E. Jacouton, F. Chain, H. Sokol, et al., Probiotic strain Lactobacillus casei BL23 [78] Y. Xiao, K. Zhou, Y. Lu, et al., Administration of antibiotics contributes to choles-
prevents colitis-associated colorectal cancer, Front. Immunol. 8 (2017) 1553. tasis in pediatric patients with intestinal failure via the alteration of FXR signaling,
[62] T. Wang, G. Cai, Y. Qiu, et al., Structural segregation of gut microbiota between Exp. Mol. Med. 50 (2018) 155.
colorectal cancer patients and healthy volunteers, ISME J. 6 (2012) 320–329. [79] N. Keren, F.M. Konikoff, Y. Paitan, et al., Interactions between the intestinal mi-
[63] P.D. Cani, Gut cell metabolism shapes the microbiome, Science 357 (2017) crobiota and bile acids in gallstones patients, Environ Microbiol Rep 7 (2015)
548–549. 874–880.
[64] H. Arimochi, K. Morita, S. Nakanishi, et al., Production of apoptosis-inducing [80] M. Song, K. Wu, J.A. Meyerhardt, et al., Fiber intake and survival after colorectal
substances from soybean protein by Clostridium butyricum: characterization of cancer diagnosis, JAMA Oncol 4 (2018) 71–79.
their toxic effects on human colon carcinoma cells, Cancer Lett. 277 (2009) [81] S.J. O'Keefe, Diet, microorganisms and their metabolites, and colon cancer, Nat.
190–198. Rev. Gastroenterol. Hepatol. 13 (2016) 691–706.
[65] M.D. Schulz, C. Atay, J. Heringer, et al., High-fat-diet-mediated dysbiosis promotes [82] N. Singh, A. Gurav, S. Sivaprakasam, et al., Activation of Gpr109a, receptor for
intestinal carcinogenesis independently of obesity, Nature 514 (2014) 508–512. niacin and the commensal metabolite butyrate, suppresses colonic inflammation
[66] A. Górska, D. Przystupski, M.J. Niemczura, et al., Probiotic bacteria: a promising and carcinogenesis, Immunity 40 (2014) 128–139.
tool in cancer prevention and therapy, Curr. Microbiol. 76 (2019) 939–949. [83] D. Bolognini, A.B. Tobin, G. Milligan, et al., The pharmacology and function of
[67] L. Ai, Y. Ren, Y. Li, et al., Synbindin deficiency inhibits colon carcinogenesis by receptors for short-chain fatty acids, Mol. Pharmacol. 89 (2016) 388–398.
attenuating Wnt cascade and balancing gut microbiome, Int. J. Cancer 145 (2019) [84] S. Sivaprakasam, P.D. Prasad, N. Singh, Benefits of short-chain fatty acids and their
206–220. receptors in inflammation and carcinogenesis, Pharmacol. Ther. 164 (2016)
[68] P. Antas, L. Novellasdemunt, A. Kucharska, et al., SH3BP4 regulates intestinal stem 144–151.
cells and tumorigenesis by modulating β-catenin nuclear localization, Cell Rep. 26 [85] M. Thangaraju, G.A. Cresci, K. Liu, et al., GPR109A is a G-protein-coupled receptor
(2019) 2266–2273 e4. for the bacterial fermentation product butyrate and functions as a tumor suppressor
[69] D.M. Gay, R.A. Ridgway, M. Müller, et al., Loss of BCL9/9l suppresses Wnt driven in colon, Cancer Res. 69 (2009) 2826–2832.
tumourigenesis in models that recapitulate human cancer, Nat. Commun. 10 (2019) [86] Y. Tang, Y. Chen, H. Jiang, et al., G-protein-coupled receptor for short-chain fatty
723. acids suppresses colon cancer, Int. J. Cancer 128 (2011) 847–856.
[70] H. Clevers, Wnt/beta-catenin signaling in development and disease, Cell 127 (2006)

467

You might also like