Elsevier

Microbial Pathogenesis

Volume 149, December 2020, 104569
Microbial Pathogenesis

Gut microbiota-derived metabolites and colorectal cancer: New insights and updates

https://doi.org/10.1016/j.micpath.2020.104569Get rights and content

Highlights

  • Until now, several gut microbiota-derived metabolites have been identified.

  • Short-Chain Fatty Acids are the most important metabolites of gut microbiota.

  • Gut microbial metabolites play vital roles in the initiation or inhibition of CRC.

  • Elucidating the role of metabolites may be promising strategies for CRC management.

Abstract

Abundant evidence from in-vitro as well as in-vivo studies supports the gut microbiota-derived metabolites as crucial executors of diet effect on the host physiology. As such, a number of microbiota-derived metabolites produced from diet have been connected to complex forms of human diseases such as colorectal cancer (CRC). Despite current unresolved questions concerning molecular mechanisms between metabolites, host signaling pathways, and CRC, some new progresses promise continued advancement of the field. Therefore, clarification of the molecular events underlying which metabolites may regulate proliferation of colonocytes will hopefully open up new avenues for seeking the possibilities affecting host health and exploitation of these capabilities for therapeutic purpose. In this Review, we will discuss recent insights into contributions of the gut microbiota-derived metabolites to CRC and argue that the cumulative effects of metabolites should be considered with the intention of better predict and prevent cancer progression. We will also discuss the signaling pathways induced by specific metabolites toward down-regulation and/or up-regulation of immune system that eventually trigger progression and/or inhibition of CRC.

Introduction

The human gut microbiota is composed of a variety of microorganisms that contribute to the host's health, physiology, and disease by synthesizing unique enzymes and through biochemical pathways relating to their versatile metabolic genes [1]. Several studies showed that diet has predominant role in formation of gut microbiota composition. Host diet impacts the gut microbial community and their metabolites (protective or harmful), making microbes a link between diet and different physiological states of host. It is assumed that colorectal cancer (CRC) may be closely correlated with dietary regime and life style of the patients [2,3].

Scientific evidences suggest that the innate immune cells can translate the signals and metabolites from gut microbiota into physiological responses [4]. Consistent with this finding, the more studies revealed that the gut microbiota and their metabolites strongly influence the host's immune system [5]. Multiple studies have shown that metabolites such as butyrate, propionate, acetate, and niacin contribute to protection of host against CRC, compared to metabolites like secondary bile acids, lactate, trimethylamine N-oxide (TMAO), N-nitroso compounds (NOC), Acetaldehyde, 4-hydroxyphenylacetic acid (HO-PAA), Phenylacetic acid (PAA), phenol, and bacterial toxin, which are correlated with CRC development (Fig. 1). Our current knowledge supports the idea that cross talk between the host, gut microbiota, and their metabolites can offer an innovative strategy for prevention and control of several diseases and cancers [6]. In this review, we will discuss about the current understanding of the role of specialized gut microbiota-derived metabolites on various aspects of immunity and cancer with a special focus on CRC.

Section snippets

Search strategy

We conducted a comprehensive search strategy of the literatures to review best available evidence and identify studies relevant to this review. We searched PubMed, Web of Science, Scopus, and Google Scholar for available papers on this subject with the following search terms: gut microbiota, metabolites, short-chain fatty acids (SCFAs), butyrate, propionate, acetate, bile acid, lactate, succinate, protein-derived metabolites, TMAO, NOC, bacterial toxin, niacin, diet, CRC, and the results were

Short-chain fatty acids (SCFAs)

The most important fermentative products of microbial activity in the gut are SCFAs containing butyrate, propionate, and acetate [7]. It was shown that bacterial fermentation of complex non-digestible polysaccharide can produce SCFAs through distinct pathways [8]. Also, it is reported that the number of SCFA-producing bacteria in mice treated with a high-fiber was higher than that in mice treated with low-fiber [9]. G protein-coupled receptors (GPR) such as GPR40, GPR41, GPR43, and GPR109A are

Butyrate

Butyrate is undeniably the most important SCFA. Butyrate activates GPR109A and inhibits the protein kinase B or Akt (PKB/Akt) and nuclear transcription factor-kappa beta p65 (NF-κB) signaling pathways to improve the intestinal epithelium barrier dysfunction and inflammatory response [18]. Growing evidence has demonstrated the positive role of butyrate in suppressing the expression of the essential molecules for gut homeostasis such as indoleamine 2, 3-dioxygenase-1 (IDO-1) by two separate

Propionate

Propionate can show an equivalent influence as butyrate as an anti-inflammatory agent in the gut and in colon cancer [30]. Accumulating in-vitro and in-vivo evidence has indicated that propionate as well as butyrate could directly increase the expression of gene related to Tc17 cells and CD8+ CTLs and promote antitumor effects [20]. Remarkably, Furusawa et al. showed that butyrate and propionate play critical roles in controlling intestinal inflammation by stimulating the differentiation of

Acetate

Acetate is one of the most abundant produced SCFA, followed by propionate and butyrate. There are few studies on acetate regarding its implication in CRC as compared to butyrate and propionate. Therefore, the exact mechanism of anti-inflammatory effect of acetate on CRC development is poorly understood. One study demonstrated that by increasing the expression of anti-inflammatory cytokine and reducing the generation of pro-inflammatory factors and NF-κB pathway in CRC cells, acetate can inhibit

Bile acid

Another important metabolite of gut microbiota is bile acids, which can be classified into two groups: i-primary bile acids; ii-secondary bile acids. The interaction between primary bile acid like cholic acid (CA) and gut microbiota can transform it into secondary bile acids like lithocholic acid (LCA) and deoxycholic acid (DCA). Secondary bile acids are increase due to intake of high-fat diets and can prompt the development of CRC. The association between bile acids and gut microbiota was

Lactate

Lactate is an intermediate and/or terminal metabolite produced by the gut microbiota. Based on the angiogenic effect of lactate, the delivery of nutrients and growth factors to the tumor microenvironment (TME) can be related to the functions of glycolytic tumor cells [52]. In this way, lactate can promote tumor invasion by providing environmental conditions through acidification of TME [53]. Moreover, it can stimulate angiogenic responses for transferring oxygen, glucose, and other nutrients to

Succinate

Another intermediate metabolite of the gut microbiota is succinate, which can be significantly produced by gut microbiota during fermentation of dietary fiber. At present, there is both supporting and conflicting literature focused on the role of succinate in the regulation of cancerous cell and CRC. The in-vitro and ex-vivo results have shown that polyphenols can result in high amounts of succinate production in the cecum and impact CRC by inhibiting the growth and proliferation of colon

Protein-derived metabolites

Fermentation of protein by the gut microbiota produces numerous amino acid metabolites. Hydrogen sulfide (H2S) is one of the amino acid derived metabolites which has usually been used as an energy source for colonic epithelial metabolism. The evidence outlining the effects of H2S on CRC has been summarized in a few studies. Interestingly, high concentration of H2S in the colon due to consumption of high protein diet (HPD) can form the adaptive response in colonocytes by stimulating the

Trimethylamine-N-oxide (TMAO)

Another intestinal microbiota-dependent metabolite is TMAO that can play an important role in the development of CRC by reacting with flavin monooxygenase (FMO) [70]. In accordance with the results obtained by Xu et al. the relationship between CRC risk and plasma factors of choline metabolism showed that TMAO is a potential indicator of CRC [71]. Although its molecular function still remains to be identified.

N-nitroso compounds (NOC)

Consumptions of high red meats and/or processed meats diet as primary source of protein and other nitrogenous residues have been previously shown as a risk factor for development of CRC. It is well known that host diet with high-protein content can increase the production of NOC by gut microbiota. Taken together, NOC by G—>A transitions in K-ras gene could mediate the progression of CRC [72]. It is generally accepted notion that activated human neutrophils could produce NOC during chronic

Bacterial toxin

Microbes can promote cancer by producing toxic metabolites or carcinogenic products that increase disease incidences and susceptibility. Bacterial toxin is another type of CRC-related metabolite which can be produced by gut microbiota such as Bacteroides fragilis (B. fragilis) and Escherichia coli (E. coli). The B. fragilis toxin plays an important role in the development of CRC through several mechanisms such as activation of β-catenin signaling and E-cadherin cleavage, induction of NF-κB

Niacin

Niacin is another bacterial product that can inhibit inflammation and tumorigenesis by connecting to the GPR109A. The valuable effect of niacin was discovered by Singh et al. in an in-vivo study, showing its role in the induction of anti-inflammatory activities in macrophages and dendritic cells and also in differentiation of Treg and IL-10-producing T cells is possible through GPR109A signaling. The end result is suppression of inflammation and tumor progression [87]. Confirming these results,

Correlation between gut microbiota-derived metabolites, diet, and CRC

Over recent years, it is generally accepted that there is an obvious relationship between the gut microbiota composition and their metabolites, diet, and the risk of CRC (Table 1). Dietary fibers are commonly indigestible complex carbohydrates for the host, which can be fermented by gut bacteria into SCFA [89]. In recent years, studies have aimed to address how fibers or fat alone or combined with each other can affect CRC risk. In this respect, an elegant clinical trial has found that higher

Potential role of the gut microbiota-derived metabolites in the early screening of CRC

Apart from mechanistic evaluation of the gut microbiota-derived metabolites, in terms of CRC detection, several metabolomics studies strongly supported the association between the levels of metabolites present in the gut and progression of CRC. Data proved that suitable CRC screening through identification and elimination of precancerous lesions can potentially result in early detection and reduction of incidence and mortality rates of CRC. Based on the recommendations from the American Cancer

Perspectives and future directions

It is now becoming apparent that the gut microbiota-derived metabolites have a significant role in host physiology and CRC. Over the past decades, elegant experiments exploiting the effect of the gut microbiota-derived metabolites on the intestine found that changing the eating habits could result in the progression or reduction of CRC. Metabolically, SCFAs as well as niacin cause opposing effects compared to secondary BAs, lactate, TMAO, and NOC on the course of colonic inflammation and CRC,

Conclusion

Recent researches have begun to focus attention on the role of gut microbiota-derived metabolites in the development, prevention, and screening of CRC. Dysbiosis is characterized by starvation of the gut microbiota which can be caused by dietary imbalance. This leads to increased incidences of CRC as a consequence of secondary bile acid and/or bacterial toxin production by gut microbiota. Contrastingly, gut microbiota-derived SCFAs have been shown to have inhibitory impacts on colonic

Funding/support

This work was supported by the National Science Foundation of China (grant number 81972315).

Authors' contributions

All the authors equally contributed to this work. Also, all authors read and approved the final manuscript.

Declaration of competing interest

The authors declare no conflicts of interest with respect to authorship and publication of this article.

Acknowledgements

We apologize to researchers whose work was not cited because of limitations for publication.

References (112)

  • M.K. Baek et al.

    Lithocholic acid upregulates uPAR and cell invasiveness via MAPK and AP-1 signaling in colon cancer cells

    Canc. Lett.

    (2010)
  • S.M. Centuori et al.

    Deoxycholic acid mediates non-canonical EGFR-MAPK activation through the induction of calcium signaling in colon cancer cells

    Biochim. Biophys. Acta Mol. Cell Biol. Lipids

    (2016)
  • H. Zeng et al.

    Butyrate and deoxycholic acid play common and distinct roles in HCT116 human colon cell proliferation

    J. Nutr. Biochem.

    (2015)
  • E. Im et al.

    Ursodeoxycholic acid (UDCA) can inhibit deoxycholic acid (DCA)-induced apoptosis via modulation of EGFR/Raf-1/ERK signaling in human colon cancer cells

    J. Nutr.

    (2004)
  • M. Beaumont et al.

    Detrimental effects for colonocytes of an increased exposure to luminal hydrogen sulfide: the adaptive response

    Free Radic. Biol. Med.

    (2016)
  • M. Libiad et al.

    Hydrogen sulfide perturbs mitochondrial bioenergetics and triggers metabolic reprogramming in colon cells

    J. Biol. Chem.

    (2019)
  • W. Wang et al.

    H(2)S induces Th1/Th2 imbalance with triggered NF-κB pathway to exacerbate LPS-induce chicken pneumonia response

    Chemosphere

    (2018)
  • W.R. Russell et al.

    High-protein, reduced-carbohydrate weight-loss diets promote metabolite profiles likely to be detrimental to colonic health

    Am. J. Clin. Nutr.

    (2011)
  • I.C. McCall et al.

    Effects of phenol on barrier function of a human intestinal epithelial cell line correlate with altered tight junction protein localization

    Toxicol. Appl. Pharmacol.

    (2009)
  • L. Armand et al.

    In vitro impact of amino acid-derived bacterial metabolites on colonocyte mitochondrial activity, oxidative stress response and DNA integrity

    Biochim. Biophys. Acta Gen. Subj.

    (2019)
  • M. Beaumont et al.

    Quantity and source of dietary protein influence metabolite production by gut microbiota and rectal mucosa gene expression: a randomized, parallel, double-blind trial in overweight humans

    Am. J. Clin. Nutr.

    (2017)
  • I.T. Vermeer et al.

    Neutrophil-mediated formation of carcinogenic N-nitroso compounds in an in vitro model for intestinal inflammation

    Toxicol. Lett.

    (2004)
  • D.G. Hebels et al.

    N-nitroso compound exposure-associated transcriptomic profiles are indicative of an increased risk for colorectal cancer

    Canc. Lett.

    (2011)
  • B.E. Dutilh et al.

    Screening metatranscriptomes for toxin genes as functional drivers of human colorectal cancer

    Best Pract. Res. Clin. Gastroenterol.

    (2013)
  • B.K. Thakur et al.

    Unveiling the mutational mechanism of the bacterial genotoxin colibactin in colorectal cancer

    Mol. Cell

    (2019)
  • N. Singh et al.

    Activation of Gpr109a, receptor for niacin and the commensal metabolite butyrate, suppresses colonic inflammation and carcinogenesis

    Immunity

    (2014)
  • D. Chen et al.

    Clostridium butyricum, a butyrate-producing probiotic, inhibits intestinal tumor development through modulating Wnt signaling and gut microbiota

    Canc. Lett.

    (2020)
  • S. Ocvirk et al.

    A prospective cohort analysis of gut microbial co-metabolism in Alaska Native and rural African people at high and low risk of colorectal cancer

    Am. J. Clin. Nutr.

    (2020)
  • I. Rowland et al.

    Gut microbiota functions: metabolism of nutrients and other food components

    Eur. J. Nutr.

    (2018)
  • F. Bishehsari et al.

    Dietary fiber treatment corrects the composition of gut microbiota, promotes SCFA production, and suppresses colon carcinogenesis

    Genes

    (2018)
  • T. Haraguchi et al.

    Cecal succinate elevated by some dietary polyphenols may inhibit colon cancer cell proliferation and angiogenesis

    J. Agric. Food Chem.

    (2014)
  • C.A. Thaiss et al.

    The microbiome and innate immunity

    Nature

    (2016)
  • M. Levy et al.

    Dysbiosis and the immune system

    Nat. Rev. Immunol.

    (2017)
  • Q. Li et al.

    Inter-kingdom signaling between gut microbiota and their host

    Cell. Mol. Life Sci.

    (2019)
  • G.T. Macfarlane et al.

    Fermentation in the human large intestine: its physiologic consequences and the potential contribution of prebiotics

    J. Clin. Gastroenterol.

    (2011)
  • H. Shuwen et al.

    Protective effect of the “food-microorganism-SCFAs” axis on colorectal cancer: from basic research to practical application

    J. Canc. Res. Clin. Oncol.

    (2019)
  • F. Bishehsari et al.

    Dietary fiber treatment corrects the composition of gut microbiota, promotes SCFA production, and suppresses colon carcinogenesis

    Genes

    (2018)
  • H.V. Lin et al.

    Butyrate and propionate protect against diet-induced obesity and regulate gut hormones via free fatty acid receptor 3-independent mechanisms

    PloS One

    (2012)
  • S. Lavoie et al.

    Expression of FFAR2 by dendritic cells prevents their expression of IL27 and is required for maintenance of mucosal barrier and immune response against colorectal tumors in mice

    Gastroenterology

    (2020)
  • U.-H. Jin et al.

    Short chain fatty acids enhance aryl hydrocarbon (Ah) responsiveness in mouse colonocytes and Caco-2 human colon cancer cells

    Sci. Rep.

    (2017)
  • B.S. Samuel et al.

    Effects of the gut microbiota on host adiposity are modulated by the short-chain fatty-acid binding G protein-coupled receptor, Gpr41

    Proc. Natl. Acad. Sci. Unit. States Am.

    (2008)
  • C. Martin-Gallausiaux et al.

    Butyrate produced by commensal bacteria down-regulates indolamine 2,3-dioxygenase 1 (Ido-1) expression via a dual mechanism in human intestinal epithelial cells

    Front. Immunol.

    (2018)
  • M. Luu et al.

    Regulation of the effector function of CD8+ T cells by gut microbiota-derived metabolite butyrate

    Sci. Rep.

    (2018)
  • L. Zhou et al.

    Faecalibacterium prausnitzii produces butyrate to maintain Th17/Treg balance and to ameliorate colorectal colitis by inhibiting histone deacetylase 1

    Inflamm. Bowel Dis.

    (2018)
  • C. Martin-Gallausiaux et al.

    Butyrate produced by gut commensal bacteria activates TGF-beta1 expression through the transcription factor SP1 in human intestinal epithelial cells

    Sci. Rep.

    (2018)
  • T. Xiao et al.

    Butyrate upregulates the TLR4 expression and the phosphorylation of MAPKs and NK-κB in colon cancer cell in vitro

    Oncol Lett

    (2018)
  • X. Sun et al.

    Butyrate inhibits indices of colorectal carcinogenesis via enhancing α‐ketoglutarate‐dependent DNA demethylation of mismatch repair genes

    Mol. Nutr. Food Res.

    (2018)
  • H.R.D. Alrafas et al.

    Effect of sodium butyrate supplementation on the gut microbiome during colorectal cancer

    Am Assoc Immnol

    (2019)
  • L. Boesmans et al.

    Butyrate producers as potential next-generation probiotics: safety assessment of the administration of Butyricicoccus pullicaecorum to healthy volunteers

    mSystems

    (2018)
  • S. Liang et al.

    Gut microbiome associated with APC gene mutation in patients with intestinal adenomatous polyps

    Int. J. Biol. Sci.

    (2020)
  • Cited by (0)

    View full text