Open access peer-reviewed chapter

The Influence of Microbial Metabolites in the Gastrointestinal Microenvironment on Anticancer Immunity

Written By

Silke Neumann, Estelle M. Peyroux, Matt J. Woodall, Nick J. Shields, Sarah L. Young and Sharon T. Pattison

Submitted: 03 May 2019 Reviewed: 18 June 2019 Published: 02 August 2019

DOI: 10.5772/intechopen.88137

From the Edited Volume

Current Cancer Treatment

Edited by Mirjana Rajer and Eva Segelov

Chapter metrics overview

1,067 Chapter Downloads

View Full Metrics

Abstract

The gastrointestinal (GI) tumour microenvironment is characterised by its unique colonisation with bacteria that are estimated to match the total number of cells in our body. It is becoming increasingly clear that the microbiome and its metabolites are important orchestrators of local and systemic immune responses, anticancer immunity and the host response to cancer therapy. Apart from their role as an energy source, metabolites have been shown to modulate inflammation, immune cell function and cancer cell survival. The polarisation of immune cell subsets by microbial metabolites towards either pro- or antitumorigenic functions strongly affects cancer progression and outcomes. In this chapter, we will discuss the link between microbial metabolites in the GI tumour microenvironment, anticancer immune responses and cancer progression.

Keywords

  • gastrointestinal tumour environment
  • host immune response
  • innate immunity
  • microbial metabolites
  • metabolism

1. Introduction

The GI tract is a complex ecosystem, populated by a large variety of bacteria, fungi and viruses that together form the intestinal microbiome. A surprising amount of local and systemic bodily functions are affected by the composition of the microbiome and its produced metabolites. This includes the generation of energy, metabolism of dietary components and synthesis of vitamins as well as regulation of immune responses, behaviour and mood. Perturbations of microbial populations, commonly referred to as dysbiosis, have been associated with a large number of diseases, such as inflammatory bowel disease [1], diabetes [2], obesity [3], autism [4], depression [5] and colorectal cancer [6, 7]. Understanding the reciprocal relationship between the microbiota and immunity has received great attention as it is becoming increasingly clear that inflammatory processes underlie many pathologies. The complexity of microbiome-immune interactions is staggering as not only the presence or absence of bacterial species shape immunity, but metabolites produced and modified by bacteria have a direct effect on the immune system’s ability to react to infectious and non-communicable diseases [8].

Microbial metabolites, as the sum of products modified and synthesised by microbiota, can be a useful tool to understand microbiota-driven immune modulation when analysis of bacterial lineages proves difficult. Diversity and abundance of microbial communities varies greatly amongst healthy individuals, whereas metabolic pathways are conserved and stable [9, 10]. Therefore, assessing changes of metabolic pathways and how they affect immunity may provide crucial insights into the role of the GI microenvironment in health and disease. Microbial metabolites are commonly divided into three categories, (1) metabolites produced by bacteria, derived from host products; (2) metabolites modified by bacteria, derived from host products; and (3) metabolites synthesised by bacteria directly.

In the following sections, we will briefly describe GI cancers and components of the GI tract that shape the tumour microenvironment. Furthermore, we will discuss the evidence for connecting changes in the microbiome and its metabolites with carcinogenesis and the role of bacterial metabolites in shaping immunity and in particular anticancer immunity.

Advertisement

2. The GI cancer microenvironment

2.1 The gastrointestinal tract

The gastrointestinal tract starts at the mouth, extends to the anus and includes the oesophagus, stomach, small intestine, large intestine, liver and pancreas. Its main functions are primarily the disruption and digestion of food, absorption of nutrients and elimination of waste products. With the diverse functions of the GI tract, it is not unsurprising that it has a number of diverse environments which are contributed to by various types of immune cells and the multiple bacteria that reside in the GI tract.

Movement of food down the GI tract is facilitated by muscular contractions. Much of the tube that makes up the GI tract is muscle lined to enable this to occur, with sphincters at particular junctures to enable control of food passage. The muscle layers are coated by a mucous membrane which varies depending on the function of that section of the GI tract.

The epithelium that lines the GI tract can be broadly divided into three subtypes, primarily based on their function. Squamous epithelium is found at the start (mouth and oesophagus) and end (anus) of the GI tract providing a protective covering. Secretory epithelium is found in the stomach. Absorptive epithelium is found in both the small and large intestines. The small intestine has numerous fingerlike projections, called villi, that increase the surface area to facilitate absorption of nutrients with interspersed crypts, or glands, which contain the stem cells that give rise to the epithelial cells. The absorptive epithelium of the large intestine is more closely packed with glands specialised for water absorption and mucus-secreting cells to lubricate the passage of faecal material down the GI tract.

The tube that forms the GI tract has a number of layers that lie between the outer muscular wall (the muscularis propria) and the innermost epithelium. The epithelium forms the innermost layer of the mucosa, which has two additional components, the lamina propria (composed of supportive connective tissue) and a thin layer of smooth muscle, the muscularis mucosae. Underneath the mucosa is the submucosa, which contains connective tissue, nerves and lymphatic and blood vessels. The submucosa is surrounded by the outer muscularis propria, the muscle layer whose contractions facilitate passage of material down the GI tract. The supporting tissue surrounding the GI tract is called the adventitia or serosa and contains major nerves and blood vessels.

The GI tract includes two large glands, the liver and pancreas. Both develop from the primitive foregut embryonically and have functions that contribute to digestion by generating digestive fluids. The liver produces bile, which can be stored and concentrated in the gallbladder. When lipids enter the duodenum, neuroendocrine cells of the duodenal mucosa are stimulated to release cholecystokinin-pancreozymin (CCK) causing contraction of the gallbladder releasing bile into the duodenum. Bile acids are emulsifying agents which aid in lipid digestion. Pancreatic secretions reach the duodenum via the pancreatic duct and contain a high content of alkaline bicarbonate ions which assist in neutralising the acidic fluid that has come from the stomach. The pancreas also produces a number of enzymes including trypsin, chymotrypsin, amylase, lipase and carboxypeptidases which are involved in the breakdown of proteins, carbohydrates and lipids.

2.2 Microbiota in the GI tract

Our lifestyle, including diet, exercise, childhood microbial exposure and the use of antibiotics strongly, influences the composition of our microbiota [9, 11, 12, 13, 14]. Two phyla of bacteria dominate the human gut Bacteroidetes and Firmicutes. Over decades the ability to classify bacteria into their genus and species has evolved with technology resulting in numerous reclassifications. Bacteria can be additionally classified into subspecies on the basis of small but relevant differences within a species. Further classifications into strains or serovars, indicating variable immune antigens present on their surface, can be allocated outside nomenclature rules. This level of complexity demonstrates the purpose of studying microbial metabolites in the context of gut immunity, thereby avoiding the complexities of bacterial species, focusing instead on their metabolic output.

Epidemiological data initially made links between bacteria and cancer development. However, identifying the role of bacteria in cancer development has been challenging due to the importance of host factors in cancer susceptibility combined with the ubiquitous nature of bacteria and the prolonged period between introduction of a bacterium and development of overt cancer [15]. This is further complicated by environmental factors which are thought to play a much larger role than genetic makeup in determining the makeup of an individual’s microbiota [16].

While the knowledge of outcomes from bacterial interactions with human cells is growing, there is enormous potential for further discovery when accommodating other microbes that populate different levels of the gastrointestinal tract such as fungi and viruses [17].

2.3 Gastrointestinal cancers

Gastrointestinal (GI) cancers are as diverse as the environments of the GI tract and the various cell types found in the GI tract. Squamous cell carcinomas arise in the squamous epithelium of the oral cavity, oesophagus and anus. Those that arise in the oral cavity are considered head and neck cancers rather than GI cancers. Adenocarcinomas are cancers that arise from glandular epithelium and can arise in the oesophagus, stomach, small intestine, pancreas and large intestine. Other cancers that can arise from the GI tract include cholangiocarcinoma, with origin from bile duct cells; hepatocellular carcinoma (HCC), originating from hepatocytes (liver cells); gastrointestinal stromal tumours, originating from the interstitial cells of Cajal which have a role in the control of peristaltic contractions [18]; and neuroendocrine cancers which can arise from neuroendocrine cells throughout the GI tract. Multiple studies examining these GI cancers have demonstrated diverse molecular alterations within cancers that arise from the same cell type in the same organ of the GI tract, highlighting the multitude of malignancies that can arise in the GI tract [19, 20, 21, 22, 23, 24, 25, 26, 27].

Chronic inflammation and infection are intimately associated with the development of cancer, with 15% of global cancer cases in 2012 being attributed to a carcinogenic infection [28]. Examples from the GI tract include HCC with hepatitis B and C virus infections contributing to more than 70% of global HCC diagnoses in 2012 [28]. In gastric adenocarcinoma four molecular subtypes were described by The Cancer Genome Atlas (TCGA) in 2014, one is characterised by Epstein-Barr virus positivity and shows extreme DNA hypermethylation [29]. Helicobacter pylori (H. pylori) is considered a class I carcinogen by the World Health Organization due to the association of chronic infection with the development of gastric adenocarcinoma and mucosa-associated lymphoid tissue (MALT) lymphoma, a form of B cell lymphoma in the stomach [30, 31]. The risk of developing gastric cancer with H. pylori is dependent on the virulence factors of the strain causing infection, other environmental factors and host genetics [32, 33, 34].

In colorectal cancer studies, Fusobacterium subspecies were consistently identified as being differentially present in tumour samples; however, these findings are still limited by small sample sizes [35]. A study of the microbiome in a hereditary form of CRC has implicated oncotoxins produced by co-colonisation with Bacteroides fragilis (B. fragilis) and Escherichia coli (E. coli) subspecies in mucosal biofilms. Further animal studies have revealed the bacterial synergy involved in carcinogenesis whereby the B. fragilis toxin increases expression of the pro-inflammatory cytokine interleukin (IL)-17 enabling the oncotoxin-producing E. coli to invade the mucosa inducing DNA damage in epithelial cells [36].

In addition to bacteria and viruses being implicated in carcinogenesis, parasitic infections have also been implicated in cancer development in the GI tract, with liver fluke infection, particularly Opisthorchis viverrini, being associated with the development of cholangiocarcinoma [37].

2.4 Inflammation in the GI tract

Acute inflammation is an integral part of the host defence against pathogens and tissue damage and is also required for the initiation of beneficial antitumour immunity [38, 39]. In contrast, it is ongoing ‘smouldering’ inflammation that contributes to tumour development, progression, invasion and metastasis [40]. Low-grade inflammation affects the function of immune cells and promotes an immune-suppressive, tumour-promoting phenotype [41, 42]. This in turn is associated with reduced immune surveillance and clearance of tumour cells by the immune system.

Chronic inflammation can be induced through a variety of mechanisms, including chronic infections [43], autoimmunity [44], metabolic disorders [45] and altered microbiota [46, 47]. In the GI tract in particular, the host immune system has to maintain a delicate balance, and pathogens and malignant cells need to be cleared, whereas normal flora has to be tolerated. Disruption of immune tolerance or dysbiosis may result in loss of epithelial barrier function and overstimulation of immune cells, leading to tissue damage and chronic inflammation.

Conditions associated with recurrent or chronic inflammation, such as inflammatory bowel disease (IBD), have been shown to contribute to the risk of developing small and large intestine cancers [48, 49]. Mechanistically, this has been related to increased stimulation of inflammation-promoting immune cells by altered microbiota [46]. As a result, pro-inflammatory cytokines and chemokines are secreted, attract further immune cells into the tissue and polarise them towards tumour-promoting functions [50, 51, 52]. Particularly the presence of pathogenic T-cell subsets, induced by pro-inflammatory cytokines, has been shown to be a predictor of poor prognosis in colorectal cancer patients [53]. Chronic inflammation also contributes to the expansion of oncogenic bacteria thereby re-enforcing disease progression [54]. See Box 1 for an overview of the immune cell populations involved in intestinal antitumour immune responses.

Box 1.

Overview of immune cell populations involved in intestinal antitumour immune responses.

Dendritic cells
DCs are innate immune cells that develop from myeloid precursors in the bone marrow. They are capable of detecting pathogens or tissue disturbances and initiate an inflammatory response. In the tumour microenvironment, they are thought to engulf dead tumour cells and debris and present fragments thereof to T cells, thus initiating anticancer immune responses.
Macrophages
Macrophages are antigen-presenting cells that arise from either circulating monocytes or embryonic progenitors that persist into adulthood, both giving rise to tissue-specific macrophage populations that are capable of self-renewal [186, 187]. Macrophages serve important functions in immunity, cancer, metabolism and tissue repair. Macrophages play an important role in in the antitumour immune response but can also adopt a pro-tumour phenotype in the tumour environment [188, 189].
Cytotoxic T cells (CD8+T cells)
CTLs express the CD8 receptor and recognise antigens presented on the surface of antigen-presenting cells. Once primed by this encounter, CTLs are capable of recognising the same antigens and kill target cells expressing the antigen. Tumour-specific CTL responses are crucial for controlling tumour growth.
Helper T cells (CD4+T cells)
Th cells express the CD4 receptor and support functions of innate and adaptive immune cells by secreting cytokines. Depending on the environment they encounter, Th cells develop into subsets with a wide range of functions [190].
Cytokines, such as IL-12, promote the development of Th1 cells, which are efficient at secreting IFN-γ and TNF-α, important cytokines for antitumour immunity. When naïve CD4+ T cells reside an environment high in IL-6 and TGF-β, they develop into Th17 cells, which promote autoimmunity and are a negative prognostic marker for colorectal cancer [49]. Th cells can develop into immune-suppressive Tregs in the presence of TGF-β when expressing the transcription factor FOXP3.
Mucosa-activated invariant T cells (MAIT cells)
MAIT cells reside at mucosal surfaces in the lung and the intestine [163] and are widely distributed in tissue and the systemic blood circulation [164, 165]. They have innate immune cell features but also express a semi-invariant T-cell receptor, which can recognise antigens presented on a monomorphic MHC class 1-related protein (MR1) expressed by antigen-presenting cells [166]. MAIT cells are first responders to a variety of infections caused by bacteria, fungi and viruses through detection of microbial B vitamin antigens.
Innate lymphoid cells (ILCs)
ILCs stem from the lymphoid lineage but have innate immune cell characteristics [109]. They are quick responders and contribute to elimination of pathogens and tissue homeostasis by producing a variety of cytokines. Based on their specific cytokine secretion, ILCs are grouped into different classes that resemble their T-cell counterparts, for example, group 3 ILCs (ILC3s) resemble Th17 cells and produce IL-17 and IL-22 [109].

Advertisement

3. Microbial metabolites that shape antitumour immunity

Metabolites produced or modified by bacteria significantly impact health and disease by acting locally on GI tract cells but can also have systemic effects by influencing the function and activation states of immune cells. The ‘metabolome’ constitutes the sum of small molecules produced by a biological system and is a powerful approach to explore the current condition of that system [55]. Metabolomics refers to the analysis of metabolites using techniques, such as mass spectrometry, nuclear magnetic resonance analysis, high-performance liquid chromatography and gas chromatography coupled with mass spectrometry. Obtained peak patterns can be compared against spectral databases for identification of metabolites. Metabolomics can be combined with metagenomics, investigating the genetic material of the entire community, and metatranscriptomics, exploring which genes are expressed, to increase our understanding of microbiomes. The benefits, disadvantages and technical challenges of these omics techniques are reviewed extensively elsewhere [56, 57, 58, 59].

In the following sections, we will discuss the impact of microbial metabolites on immune cell function, focusing on how these metabolites shape the immune response. The anatomical components of the intestinal immune system including immune and epithelial cell populations and the mechanisms employed by these cell populations to discriminate between commensal and pathogenic bacteria have been reviewed extensively recently [60, 61, 62, 63, 64, 65, 66].

3.1 Metabolites produced by bacteria from dietary components

3.1.1 Short-chain fatty acids

Short-chain fatty acids (SCFAs) are 1–6 carbon volatile fatty acids which can either be in straight or branched chain conformation [67]. They are end products of fermentation of indigestible carbohydrates such as starch and fibre, by anaerobic microbiota in the caecum and large intestine [68]. SCFAs are the most abundant metabolite in the colon and consist almost entirely of acetate (C2), propionate (C3) and butyrate (C4) [69]. Acetate is the most common SCFA (60% of total SCFAs) in the colon and can also reach the systemic circulation after absorption from the GI tract. Propionate and butyrate make up roughly 20% of the SCFAs in faeces each [68, 70]. Propionate is mainly metabolised in the liver after draining into the portal vein after absorption from the gut mucosa, while butyrate is the preferred energy source of colonocytes and is digested locally [71].

SCFAs affect host physiology and pathology through a multitude of local and systemic mechanisms of action (Figure 1). In the GI tract, they act through binding to transmembrane G protein-coupled receptors (GPRs) and diffusion into epithelial and immune cells where they modify post-translational gene expression and function as energy source.

Figure 1.

Effects of the SCFA butyrate on epithelial and immune cell function. SCFAs are produced through fermentation of non-digestible fibre and starch by microbiota. (a) Cancer cells switch their metabolism to glycolysis and are less efficient at metabolising SCFAs such as butyrate, leading to accumulation of butyrate in the cell. Increased concentrations of butyrate inhibit HDAC activity and induce apoptosis, reduce proliferation and increase immunogenicity of cancer cells. (b) In healthy epithelial cells, butyrate is metabolised through oxidative phosphorylation and used as energy source by the cell. Butyrate also activates NLRP3 and NLRP6 inflammasomes through binding to GPRs, resulting in secretion of cytokines IL-1β and IL-18. In turn, IL-18 strengthens intestinal barrier integrity and promotes diversity of intestinal microbiota. (c) The effects of butyrate on immune cells in the lamina propria can be described as promoting the development and activity of anti-inflammatory populations, such as Tregs, while suppressing immune cell functions contributing to inflammation. Butyrate suppresses the maturation of DCs, limits their ability to prime CTLs and reduces the production of pro-inflammatory cytokines in DCs and macrophages. Together this reduces inflammation and the development of inflammatory Th subsets, such as Th17, which contribute to intestinal carcinogenesis.

GPRs implicated in SCFA signalling are free fatty acid receptors GPR41, GPR43 and GPR109a. The SCFAs acetate, propionate and butyrate have differing selectivity for these receptors with all three binding to GPR43, expressed on the GI epithelium and immune cells [72, 73]. Propionate and butyrate bind to GPR41, expressed by lamina propria cells in the large intestine, immune cells and cells of the peripheral nervous system [72]. Butyrate has also been found to ligate GPR109a expressed by large intestinal epithelium and certain subsets of immune cells [73].

Activation of the GPRs leads to changes in intracellular potassium concentrations [K+], which directly activate intracellular danger-sensing molecular complexes, called inflammasomes. Integral components of inflammasomes are Nod-like receptors, which are cytosolic pattern recognition receptors (PRRs). Particularly changes in the NLRP3 and NLRP6 inflammasomes (containing Nod-like receptors 3 and 6, respectively) have been implicated in exacerbating intestinal inflammation [74, 75, 76]. Inflammasome complexes can be activated through a two-step process. The first signal is considered the ‘priming signal’, which induces nuclear factor (NF)-kB-mediated transcription of inflammasome components and pro-IL-1β and IL-18 in epithelial and immune cells [77]. The second signal leads to the assembly of the inflammasome complex and caspase-1-dependent processing of pro-IL-1β and IL-18 into their biologically active forms.

IL-1β and IL-18 are important signalling molecules for gut homeostasis and immune effector function. IL-1β can have pro- and anti-homeostatic functions, whereas IL-18 is generally regarded as a crucial cytokine for maintaining gut barrier integrity and a healthy microbiome composition. A reduction in IL-18 secretion has been found to be associated with a shift in microbiota towards the expansion of Bacteroidetes, which promote colonic inflammation and carcinogenesis in mouse models [46, 76]. This aligns with findings that describe a decreased expression of NLRP6 in gastric cancer, correlated with a reduced survival time in patients [78]. When the NLRP6 inflammasome was overexpressed, gastric cancer cell proliferation and development were inhibited, and migration and invasion of cancer cells were decreased. Furthermore, NLRP6 activity has been linked with intact epithelial barrier function and prevention of colorectal cancer development [79]. Even though NLRP3 inflammasome activation has been shown to contribute to tumour-promoting inflammation and immune infiltrate in several ways [11, 80, 81, 82, 83], many reports highlight the beneficial functions of the NLRP3 inflammasome in preventing intestinal cancer development. For example, activation of NLRP3 inflammasomes has been demonstrated to protect from intestinal carcinogenesis via IL-18-mediated epithelial repair [84] and suppression of metastatic colon cancer growth via maturation of natural killer (NK) cells and stimulation of their tumoricidal activity [85]. The complex biology of intestinal inflammasome signalling and its role in tumorigenesis have been reviewed recently [74, 86]. It remains to be investigated how the often overlapping and controversial findings regarding inflammasome functions orchestrate induction and resolution of inflammation.

SCFAs, particularly butyric acid and β-hydroxybutyrate, stimulate NLRP3 and NLRP6 inflammasomes through binding to GPR43 and GPR109a, leading to increased production of IL-1β and IL-18 [87, 88]. Subsequently it was shown that dietary supplementation with sodium butyrate or increased consumption of dietary fibre protected mice against colitis [87] and colonic carcinogenesis [89] through production of IL-18 and promotion of gut homeostasis. Interestingly, even though activation of the NLRP3 inflammasome was mediated via activation of GPRs, stimulation of GPRs with synthetic agonists did not recapitulate these findings, indicating that SCFAs must act on additional targets that influence cytokine secretion [90].

A prominent target of SCFAs is histone deacetylases (HDAC) and acetyltransferases (HAT), which regulate gene expression by allowing or preventing access of the transcription machinery to DNA. HDAC inhibitors have been used in cancer therapy for their ability to induce cancer cell death, reduce proliferation and increase immunogenicity of cancer cells as well as stimulate anticancer immune function [91, 92, 93]. Cancer cells utilise glucose as their primary energy source, and thus SCFAs, such as butyrate, accumulate and due to increased concentration inhibit HDAC activity [94]. In contrast, healthy cells are capable of metabolising butyrate into small molecules required for energy generation, thereby preventing accumulation of butyrate and HDAC inhibition [95, 96].

Interestingly, a similar mechanism may explain the diverging effects of butyrate on immune cell populations in the gut. In order to retain intestinal homeostasis, immune cells have to remain passive when challenged with host microbiota and food antigens yet remain responsive to fight pathogenic bacteria. This diversity of function is supported by SCFAs that induce a hypo-responsive state in immune cell populations, which are capable of promoting inflammation, such as macrophages, dendritic cells (DCs) and T cells [47], yet cells involved in containing inflammation are induced and expanded by SCFAs [97, 98].

DCs and macrophages are professional antigen-presenting cells, highly proficient at scanning the environment for invaders or tissue disturbances. Once detected, pathogens or abnormal cells are engulfed, processed and presented in small fragments to T helper (Th) cells. These cells, in turn, differentiate into populations of effector Th cells, directed by cytokines from DCs. Secretion of pro-inflammatory cytokines by DCs, such as IL-6, IL-12 and IL-23 in particular, supports the polarisation of Th cells towards effector and inflammatory subsets Th1 and Th17, respectively. This is important for removal of pathogens but can be detrimental for tissue homeostasis if not regulated tightly. Th cells also facilitate the full activation and memory development of cytotoxic T cells (CTLs), which are able to kill antigen specifically and react swiftly in the case of a second encounter. SCFAs, butyrate and propionate, but not acetate, have been shown to reduce production of pro-inflammatory cytokines, such as IL-12 and IL-23, and chemokines in DCs and also impair the maturation of DCs [90, 99, 100]. Changes in cytokine secretion are associated with impaired ability of DCs to prime CTLs [101], reduced polarisation of Th-cell subsets towards effector and inflammatory subsets and induction of regulatory T cells (Tregs) [102].

Regulatory T cells have an important role in control of inflammation. Tissue inflammation and autoimmunity are promoted if Tregs are not present or dysfunctional. In contrast to many other cancers where Tregs are thought to suppress effective antitumour immunity, GI cancer patients benefit from the presence of Tregs in the tumour microenvironment [103]. Tregs limit inflammatory processes, induce tolerance towards food and microbial antigens and promote stem cell renewal in the intestine through a variety of mechanisms. This includes production of anti-inflammatory cytokines, such as IL-10 and transforming growth factor beta (TGF-β)1, expression of inhibitory molecules and restriction of nutrients required by effector T cells, particularly Th1 and Th17 cells [64, 104, 105, 106].

Interestingly, due to the high rates of glycolysis in effector and pro-inflammatory cells, such as CTLs, Th1 and Th17 subsets, butyrate accumulates in these cells, leading to an inhibitory effect mediated by both HDAC inhibition and binding to GPR109a [90, 93, 102]. In contrast, anti-inflammatory cells, such as Tregs, which rely on oxidative phosphorylation can process butyrate for energy consumption, circumventing these effects [107]. It has been demonstrated comprehensively that SCFAs drive Treg development via HDAC inhibition and GPR activation in the intestine and periphery, thereby protecting mice against colonic inflammation, colitis and colorectal cancer [97, 98, 106, 108].

3.1.2 Indole derivatives

Indoles are aromatic heterocyclic compounds, produced by gut bacteria from the degradation of tryptophan via several enzymes [109]. Tryptophan is an essential amino acid, which cannot be produced by the host and is taken up in the diet. Dietary tryptophan can be metabolised by microbiota and host cells to indole derivatives that have important immune modulatory functions in the gut [110]. Indole derivatives, such as kynurenines, are ligands for the aryl hydrocarbon receptor (AHR), an intracellular ligand-activated transcription factor with important roles in detecting environmental changes and alerting cells to them. Microbial AHR ligands are thought to play an important role in maintaining intestinal homeostasis and limiting inflammation [111]. The importance of AHR signalling has been demonstrated in AHR−/− mice where clearance of pathogenic bacteria was impaired while intestinal inflammation was elevated and associated with an increased risk of developing colitis [112, 113, 114].

Mechanistically, bacterial AHR ligands have been shown to induce the production of IL-22 in innate lymphoid cells (ILCs), which promotes diversity of gut microbiota and protects mucosal barrier functions [115, 116]. ILCs stem from the lymphoid lineage but have innate immune cell characteristics [117]. They are quick responders and contribute to elimination of pathogens and tissue homeostasis by producing a variety of cytokines. Based on their specific cytokine secretion, ILCs are grouped into different classes that resemble their T-cell counterparts, for example, group 3 ILCs (ILC3s) resemble Th17 cells and produce IL-17 and IL-22 [117]. Even though production of IL-22 by ILC3s is vital for mucosal homeostasis, elevated levels of ILC3s and increased production of IL-17 have been associated with IBD pathology [118, 119]. Furthermore, it has been found that IL-22 contributes to tumorigenesis in the colon when elevated chronically. This was mediated via an inflammasome-dependent reduction of IL-22 binding protein and chronic elevated IL-22 levels [50, 51].

Genetic induction of constitutively active AHR signalling in mouse models has been found to be associated with stomach and liver cancer development [120, 121], whereas the absence of AHR in AHR−/− mice protected from prostate cancer [122]. As AHR is crucially involved in early development, maintenance of stem cells and cell differentiation, it is difficult to discern if stable genetic induction or ablation of these signalling pathways may promote carcinogenesis directly or through disturbances in early development.

3.1.3 Polyamines

Polyamines are small polycationic molecules, derived either from the diet or synthesised by gut bacteria or host cells [123]. While they are found in almost all living cells, the method of production in mammalian and bacterial cells differs. Intestinal bacteria use inducible or constitutive forms of amino acid decarboxylase enzymes in order to produce polyamines with arginine as a precursor. Mammalian synthesis involves a series of steps to convert arginine to polyamines, with ornithine decarboxylase being the rate-limiting enzyme. Putrescine, spermidine and spermine are the major polyamines secreted by both the gut microbiota and mammalian cells and have important immune modulatory functions [124].

Along with other polyamines, spermine directly regulates cells in the innate arm of the immune system and has an anti-inflammatory effect. Spermine inhibits lipopolysaccharide-induced expression of pro-inflammatory cytokines in monocytes and macrophages [125]. In macrophages, spermine is able to increase the expression of IL-10 and suppress production of inflammatory cytokines such as IFN-γ [126]. These functions were shown to have anti-inflammatory and protective effects in animal models of local and systemic inflammation [127].

Conversely, spermine inhibits the activation of the NLRP6 inflammasome and reduces the amount of IL-1β and IL-18 released. This is counteracted by taurine, another microbial metabolite, which is discussed below. The inhibitory effect of spermine on NLRP6 activity may be counteracted by the role polyamines play in maintenance of the gut epithelial lining. Many studies have found that intestinal mucosal repair is associated with an increase in levels of spermine, spermidine and putrescine [128]. Furthermore, when the synthesis of polyamines is blocked, migration and proliferation of intestinal epithelial cells to the site of injury as well as in regular turnover of mucosal cells are significantly reduced. Polyamines promote the transcription of E-cadherin, which is important for the formation of tight junctions. In this regard, they play a role in stabilising the gut epithelium, so it is able to act as a barrier between the external and internal environment [128].

3.2 Metabolites modified by bacteria, derived from host products

3.2.1 Bile acids and derivatives

Bile acids are physiological surfactants, produced in the liver and secreted into the duodenum or stored in the gall bladder. Bile acid molecules contain a hydrophobic hemisphere and a hydrophilic one, enabling them to associate around dietary fats and fat-soluble vitamins into micelles [129]. This promotes the breakdown and absorption of these molecules in the hydrophilic environment of the GI tract [130]. Approximately 95% of bile acids are reabsorbed via active transport by the apical sodium-dependent bile acid transporter in the ileum of the small intestine [131]. Microbial bile salt hydrolases catalyse the hydrolysis of amide bond linkage in bile acids, releasing an unconjugated bile acid. The de-conjugation of bile acids causes the release of glycine and taurine, which can then be used for further metabolism and growth.

Bile acids modulate innate immune cell function by inhibiting NF-kB activity, resulting in reduced production of pro-inflammatory cytokines and molecules (TNF-α, IL-1β, IL-6, IL-12, cyclooxygenase-1 and cyclooxygenase-2, and inducible nitric oxide synthase) in stimulated monocytes, macrophages, DCs and intestinal epithelial cells [132, 133, 134, 135]. In human macrophages, administration of bile acids leads to increased production of IL-10 and a decrease in phagocytosis [136]. The reduction in pro-inflammatory cytokines combined with the increase in anti-inflammatory cytokine production induces the development, recruitment and expansion of Tregs in the colon. Together, the properties of bile acids improve barrier integrity and outcomes in mouse models of experimental colitis, which lead to the development of inflammatory bowel disease and colorectal cancer [133]. The effects occur via bile acid-mediated activation of the farnesoid X receptor (FXR), a ligand-activated nuclear receptor, and the G protein-coupled bile acid receptor 1 (GPBAR1). These receptors also play a crucial role in bile acid-induced inhibition of the NLRP3 inflammasome, which is associated with reduced levels of secreted IL-1β and IL-18 [137, 138].

In contrast to primary bile acids, the bile acid-derivative taurine stimulates NLRP6 inflammasome activity, leading to increased production of IL-18 [139]. Levy et al. found high taurine and associated IL-18 concentrations maintained and restored functional microbiota typically present in healthy flora.

3.3 Metabolites synthesised by bacteria directly

Microbiota are able to synthesise metabolites that are either unique to prokaryotic organisms, such as capsule polysaccharides and certain vitamins, or that can also be produced by host cells, for example, adenosine triphosphate (ATP).

3.3.1 ATP

In addition to its role as universal energy source, ATP is an important signalling molecule that directly impacts immune cell function when released into the extracellular space. ATP is not only produced by living organisms but has been found to be secreted by a variety of commensal and pathogenic bacteria [140, 141]. Generally, increased levels of ATP are produced and secreted by host cells under inflammatory stress conditions and injury, often associated with inflammatory cell death [142]. Furthermore, the tumour microenvironment has high concentrations of extracellular ATP, at least partly induced by hypoxia, an activator of ATP secretion, and necrotic cell death [143, 144]. The chronic presence of ATP in the tumour microenvironment supports cancer cell proliferation, survival and metastasis as reviewed elsewhere [144]. In the immune context, most of the actions of ATP have been described to be pro-inflammatory; however, its hydrolysis product adenosine has immune-suppressive functions.

Host-, tumour- and microbial-derived ATP binds to purinergic-type receptor P2, while adenosine, the downstream product of hydrolysed ATP, binds to P1 receptors [144]. Purinergic P2 receptors are expressed highly by immune cells, and ATP exerts most of its pro-inflammatory effects through binding to P2X(1–7) ion channels and P2Y(1, 4, 6, 11–14) metabotropic purinergic receptors [145]. Activation of purinergic receptor P2X7 by ATP increases intracellular potassium and calcium concentrations [146]. Together with a priming signal, ATP is an important inducer of NLRP3 inflammasome activity [147]. As discussed previously, activation of the NLRP3 inflammasome and the subsequent secretion of IL-1β and IL-18 have important roles in shaping the magnitude of inflammatory responses, gut homeostasis and barrier function and have a controversial role in tumour progression [86].

Besides its role as inflammasome activator, ATP modulates migration of innate and adaptive immune cell subsets [148]. After release of ATP into the extracellular space, innate immune cells such as monocytes, mature DCs, neutrophils, macrophages and microglia are mobilised via activation of P2X and P2Y receptors and migrate to the source of the high ATP concentration. This migratory response is further amplified through autocrine activation of pannexin 1 channels in the membrane of innate immune cells [149, 150, 151, 152, 153]. Interestingly, ATP has been shown to affect migration of CD4+ T-cell subsets differently, depending on their function and activation status. While activated CD4+ T cells respond to high ATP concentrations and stimulation of P2X7 and P2X4 receptors with induction of apoptosis, immune-suppressive Tregs increase proliferation and migration via their P2Y2 receptor [154].

In the context of intestinal inflammation and carcinogenesis, ATP drives the polarisation of CD4+ T cells towards IL-17-producing CD4+ T cells, associated with a higher susceptibility to develop colitis and exacerbation of existing colitis in experimental mouse models [155, 156]. A Th17 signature in colorectal cancer patients is associated with disease progression and worse outcomes [53]. Polymorphism of the ATP-converting enzyme CD39 (hydrolysis of ATP to adenosine diphosphate (ADP)) in IBD patients and increased expression of P2X7 receptors in the inflamed epithelium of Crohn’s patients have been found, suggesting another role of ATP in disease pathology [156, 157].

ATP is hydrolysed by CD39 and CD73 to adenosine, which have been widely investigated and reviewed for their immune-suppressive functions in the tumour environment [144, 151, 152, 158]. Therapeutic inhibition of ATP and adenosine receptors as well as targeting of CD39 and CD73, alone and in combination with traditional chemotherapy, has shown great promise to prevent tumour growth by overturning adenosine-induced immune suppression [159, 160, 161, 162, 163]. However, recent evidence demonstrates that extracellular ATP is required for the formation of long-term, antigen-specific CTL responses, which are crucial for immunological memory [164]. It remains to be determined if therapeutic targeting of purinergic receptors and conversion enzymes affects development of immunological memory in cancer, which is desirable to prevent cancer occurrence.

3.3.2 Vitamins

Humans lack the ability to produce most essential vitamins and rely on vitamins to be supplied with the diet and produced by gut bacteria. Microbiota are able to synthesise vitamin K and a large number of B vitamins, such as folate (vitamin B9), riboflavin (vitamin B2), pyridoxine (vitamin B6), cobalamin (vitamin B12) and methionine [165]. B vitamins have achieved great attention for their cancer-preventing properties, with folate being the most investigated B vitamin in the cancer context [166]. The cancer-preventing mechanisms have been attributed to the role of B vitamins as cofactors in metabolic processes related to energy generation and gene regulation [166, 167]. Folate (B9) and pyridoxine (B6) have also been found to modulate intestinal immunity by increasing CD4+ T-cell proliferation, trafficking and survival of Treg subsets and NK cell cytotoxicity [168, 169, 170].

Interestingly, bacteria that synthesise vitamins B2 and B9 are recognised by mucosa-activated invariant T (MAIT) cells. MAIT cells reside at mucosal surfaces in the lung and the intestine [171] and are also widely distributed in tissue and the systemic blood circulation [172, 173]. They have innate immune cell features but also express a semi-invariant T-cell receptor, which can recognise antigens presented on a monomorphic MHC class 1-related protein (MR1) expressed by antigen-presenting cells [174]. MAIT cells are first responders to a variety of infections caused by bacteria, fungi and viruses through detection of microbial B vitamin antigens.

Upon activation, MAIT cells are able to proliferate and produce cytotoxic molecules, capable of destroying infected cells displaying microbial B vitamin antigens on their MR1 protein [175, 176, 177]. Furthermore, MAIT cells produce immune modulatory cytokines, including IFN-γ, IL-2, IL-17, IL-10 and TNF-α [178]. While IFN-γ production is highly desirable to promote antitumour immunity, an IL-17 signature has been found to be associated with worse outcomes in CRC patients [53]. Numbers of MAIT cells decrease in the peripheral circulation but accumulate in intestinal tumours [179, 180]. Several groups report a diminished ability of tumour-infiltrating MAIT cells to produce IFN-γ combined with increased secretion of IL-17 [180, 181]. Even though it appears that MAIT cells may develop a tumour-promoting phenotype in the tumour microenvironment and thus contribute to cancer progression, further studies are needed to elucidate the role of these recently discovered cells.

It is tempting to speculate that MAIT cells may impact intestinal cancer development and progression through recognition of B vitamin antigens produced by dysbiotic and carcinogenic bacteria. Since MAIT cells can be activated or inhibited depending on the B vitamin antigen presented on MR1 proteins, MAIT cells have been suggested as attractive targets for cancer immunotherapy [182]. This is in part related to their potential to be targeted in combination with chemotherapy, due the expression of drug resistance proteins that allows their survival and activation during and post-chemotherapy [172].

3.3.3 Bacterial polysaccharides

Commensal bacteria contribute to intestinal homeostasis through production of capsular polysaccharides. Polysaccharide A (PSA), the most studied bacterial polysaccharide, is produced by B. fragilis and plays an important role in regulating intestinal inflammation. Exogenous administration or bacterial production of PSA can prevent the development of experimental colitis by activating Treg and inhibiting Th17 responses [183, 184, 185]. This is mediated by PSA binding to PRRs expressed by DCs, which in turn secrete IL-10 that promotes the development and activation of Tregs. Furthermore, PSA influences the polarisation of Th subsets towards IFN-γ-producing Th cells, an important effector population for anticancer immunity. B. fragilis are not the only commensals that regulate inflammation; many other strains, for example, a large number of Clostridium strains [186], have been shown to have anti-inflammatory functions. This highlights the fact that the sum of commensals and their metabolites, rather than a defined strain or metabolite, shapes the functionality of the immune system by impacting the polarisation of immune subsets, crucial for clearance of diseases.

Advertisement

4. Conclusions

The link between microbiome disturbances and the development of inflammatory diseases highlights the importance of studying the effects of microbes and their metabolites on immune cell function. Deciphering the effects of microbial metabolites on the immune system in a highly dynamic organ system, such as the GI tract, is inherently difficult. The actions of individual metabolites need to be considered before the complex interplay of microbes, metabolites and cellular components, such as epithelial and immune cells can be investigated. The GI tumour microenvironment is unique in that immunological tolerance is required to maintain a healthy intestinal environment, including maintenance of the “normal” microbiome, yet the presence of regulatory immune cells may impede antitumour immune responses and promote carcinogenesis.

There is increasing evidence from preclinical mouse model systems and human studies that GI tract microbiota, such as B. fragilis, Bifidobacterium, Faecalibacterium prausnitzii and Akkermansia muciniphila, can directly influence response to treatment including immunotherapies and survival in some malignancies [187, 188, 189, 190, 191, 192, 193]. This effect is potentially mediated by bacteria stimulating activation of innate immune cells and downstream polarisation of Th-cell subsets towards Th1 cells [194]. The species and diversity of bacterium identified as influencing treatment response and survival vary, likely reflecting the complexity of the interactions involved, the diverse malignancies and populations within which those malignancies had arisen, and the number of bacterial species that have immunomodulatory effects mediated through the GI tract.

The influence of infections on initiation and promotion of cancer has been long recognised, but our understanding of the complex network of interactions between the host, the microbiome, the genetics of both the host and microbiome and the metabolome remains superficial. These interactions are not static, which, with the diversity of the GI tract environment, add to the challenge of deciphering what microbial species may be influencing the immune response in a tumour-promoting or tumour-suppressive manner. The complexity of microbial species and indeed the complexity of immune cells and their function mean that practically assaying and identifying individual species of bacteria, or subsets of immune cells, clinically in a prognostic or predictive sense is challenging. The more readily measureable microbial metabolome may provide a more clinically accessible read-out of this interaction. The wide-ranging impact that products of microbial metabolism have on immune cell function and polarity and therefore anticancer immunity has been underappreciated to this point. A greater understanding of how microbial metabolites influence the GI tumour microenvironment has the potential to expand therapeutic options and improve survival of patients with GI cancers.

Advertisement

Conflict of interest

The authors declare no conflict of interest.

References

  1. 1. Frank DN et al. Molecular-phylogenetic characterization of microbial community imbalances in human inflammatory bowel diseases. PNAS. 2007;104(34):13780-13785
  2. 2. Wen L et al. Innate immunity and intestinal microbiota in the development of Type 1 diabetes. Nature. 2008;455(7216):1109-1113
  3. 3. Ley RE et al. Microbial ecology: Human gut microbes associated with obesity. Nature. 2006;444(7122):1022-1023
  4. 4. Hsiao EY et al. Microbiota modulate behavioral and physiological abnormalities associated with neurodevelopmental disorders. Cell. 2013;155(7):1451-1463
  5. 5. Valles-Colomer M et al. The neuroactive potential of the human gut microbiota in quality of life and depression. Nature Microbiology. 2019;4(4):623-632
  6. 6. Kostic AD et al. Genomic analysis identifies association of fusobacterium with colorectal carcinoma. Genome Research. 2012;22(2):292-298
  7. 7. Allen-Vercoe E et al. Fusobacterium and Enterobacteriaceae: Important players for CRC? Immunology Letters. 2014;162(2 Pt A):54-61
  8. 8. Levy M et al. Metabolites: Messengers between the microbiota and the immune system. Genes & Development. 2016;30(14):1589-1597
  9. 9. The Human Microbiome Project C et al. Structure, function and diversity of the healthy human microbiome. Nature. 2012;486:207
  10. 10. Turnbaugh PJ et al. A core gut microbiome in obese and lean twins. Nature. 2009;457(7228):480-484
  11. 11. Zitvogel L et al. Nutrition, inflammation and cancer. Nature Immunology. 2017;18(8):843-850
  12. 12. Proctor LM et al. The integrative human microbiome project. Nature. 2019;569(7758):641-648
  13. 13. Zmora N et al. You are what you eat: Diet, health and the gut microbiota. Nature Reviews Gastroenterology & Hepatology. 2019;16(1):35-56
  14. 14. Macpherson AJ et al. How nutrition and the maternal microbiota shape the neonatal immune system. Nature Reviews. Immunology. 2017;17(8):508-517
  15. 15. Zur Hausen H. The search for infectious causes of human cancers: Where and why. Virology. 2009;392(1):1-10
  16. 16. Dąbrowska K et al. Correlations of host genetics and gut microbiome composition. Frontiers in Microbiology. 2016;7:1357
  17. 17. Hillman ET et al. Microbial ecology along the gastrointestinal tract. Microbes and Environments. 2017;32(4):300-313
  18. 18. Corless CL et al. Gastrointestinal stromal tumours: Origin and molecular oncology. Nature Reviews. Cancer. 2011;11(12):865-878
  19. 19. Cancer Genome Atlas Research N. Comprehensive molecular characterization of gastric adenocarcinoma. Nature. 2014;513(7517):202-209
  20. 20. Cancer Genome Atlas Research N et al. Integrated genomic characterization of oesophageal carcinoma. Nature. 2017;541(7636):169-175
  21. 21. Cancer Genome Atlas Research Network. Electronic address aadhe, et al. Integrated genomic characterization of pancreatic ductal adenocarcinoma. Cancer Cell. 2017;32(2):185-203 e13
  22. 22. Farshidfar F et al. Integrative genomic analysis of cholangiocarcinoma identifies distinct IDH-mutant molecular profiles. Cell Reports. 2017;18(11):2780-2794
  23. 23. Hoshida Y et al. Integrative transcriptome analysis reveals common molecular subclasses of human hepatocellular carcinoma. Cancer Research. 2009;69(18):7385-7392
  24. 24. Lawrence B et al. Recurrent loss of heterozygosity correlates with clinical outcome in pancreatic neuroendocrine cancer. NPJ Genomic Medicine. 2018;3:18
  25. 25. Liu Y et al. Comparative molecular analysis of gastrointestinal adenocarcinomas. Cancer Cell. 2018;33(4):721-735 e8
  26. 26. Scarpa A et al. Whole-genome landscape of pancreatic neuroendocrine tumours. Nature. 2017;543(7643):65-71
  27. 27. Yao JC et al. Genomic profiling of NETs: A comprehensive analysis of the RADIANT trials. Endocrine Related Cancer. 2019;26(4):391-403
  28. 28. Plummer M et al. Global burden of cancers attributable to infections in 2012: A synthetic analysis. Lancet Global Health. 2016;4(9):e609-e616
  29. 29. Cancer Genome Atlas Research Consortium. Comprehensive molecular characterization of gastric adenocarcinoma. Nature. 2014;513(7517):202-209
  30. 30. World Health Organization, International Agency for Research on Cancer. Schistosomes, liver flukes and Helicobacter pylori. IARC Working Group on the Evaluation of Carcinogenic Risks to Humans. Lyon, 7-14 June 1994. ARC Mongr Eval Carcinog Risks Hum. 1994;61:1-241
  31. 31. Stolte M et al. Helicobacter and gastric MALT lymphoma. Gut. 2002;50(Suppl 3):III19-III24
  32. 32. Peek RM Jr et al. Helicobacter pylori and gastrointestinal tract adenocarcinomas. Nature Reviews. Cancer. 2002;2(1):28-37
  33. 33. Fuccio L et al. Gastric cancer, Helicobacter pylori infection and other risk factors. World Journal of Gastrointestinal Oncology. 2010;2(9):342-347
  34. 34. Conteduca V et al. H. pylori infection and gastric cancer: State of the art (review). International Journal of Oncology. 2013;42(1):5-18
  35. 35. Shang F-M et al. Fusobacterium nucleatum and colorectal cancer: A review. World Journal of Gastrointestinal Oncology. 2018;10(3):71-81
  36. 36. Dejea CM et al. Patients with familial adenomatous polyposis harbor colonic biofilms containing tumorigenic bacteria. Science. 2018;359:592-597
  37. 37. Sripa B et al. Liver fluke induces cholangiocarcinoma. PLoS Medicine. 2007;4(7):e201
  38. 38. Chen DS et al. Oncology meets immunology: The cancer-immunity cycle. Immunity. 2013;39(1):1-10
  39. 39. Schreiber RD et al. Cancer immunoediting: Integrating Immunity’s roles in cancer suppression and promotion. Science. 2011;331(6024):1565-1570
  40. 40. Mantovani A et al. Cancer-related inflammation. Nature. 2008;454(7203):436-444
  41. 41. Ostrand-Rosenberg S et al. Cross-talk between myeloid-derived suppressor cells (MDSC), macrophages, and dendritic cells enhances tumor-induced immune suppression. Seminars in Cancer Biology. 2012;22(4):275-281
  42. 42. Gabrilovich D et al. Myeloid-derived suppressor cells as regulators of the immune system. Nature Reviews Immunology. 2009;9:162-174
  43. 43. Hand TW et al. Linking the microbiota, chronic disease, and the immune system. Trends in Endocrinology & Metabolism. 2016;27(12):831-843
  44. 44. Piconese S et al. Viral hepatitis, inflammation, and cancer: A lesson for autoimmunity. Journal of Autoimmunity. 2018;95:58-68
  45. 45. O’Sullivan J et al. Obesity and gastrointestinal cancer: The interrelationship of adipose and tumour microenvironments. Nature reviews. Gastroenterology & Hepatology. 2018;15(11):699-714
  46. 46. Hu B et al. Microbiota-induced activation of epithelial IL-6 signaling links inflammasome-driven inflammation with transmissible cancer. PNAS. 2013;110(24):9862-9867
  47. 47. Levy M et al. Dysbiosis and the immune system. Nature Reviews. Immunology. 2017;17(4):219-232
  48. 48. Ekbom A et al. Ulcerative colitis and colorectal cancer. New England Journal of Medicine. 1990;323(18):1228-1233
  49. 49. Karin M et al. Innate immunity gone awry: Linking microbial infections to chronic inflammation and cancer. Cell. 2006;124(4):823-835
  50. 50. Grivennikov SI et al. Adenoma-linked barrier defects and microbial products drive IL-23/IL-17-mediated tumour growth. Nature. 2012;491(7423):254-258
  51. 51. Huber S et al. IL-22BP is regulated by the inflammasome and modulates tumorigenesis in the intestine. Nature. 2012;491(7423):259-263
  52. 52. Garrett WS et al. Colitis-associated colorectal cancer driven by T-bet deficiency in dendritic cells. Cancer Cell. 2009;16(3):208-219
  53. 53. Tosolini M et al. Clinical impact of different classes of infiltrating T cytotoxic and helper cells (Th1, Th2, Treg, Th17) in patients with colorectal cancer. Cancer Research. 2011;71(4):1263-1271
  54. 54. Arthur JC et al. Intestinal inflammation targets cancer-inducing activity of the microbiota. Science. 2012;338(6103):120-123
  55. 55. Fiehn O. Metabolomics—The link between genotypes and phenotypes. Plant Molecular Biology. 2002;48(1-2):155-171
  56. 56. Aguiar-Pulido V et al. Metagenomics, metatranscriptomics, and metabolomics approaches for microbiome analysis. Evolutionary Bioinformatics Online. 2016;12(Suppl 1):5-16
  57. 57. Vernocchi P et al. Gut microbiota profiling: Metabolomics based approach to unravel compounds affecting human health. Frontiers in Microbiology. 2016;7(1144)
  58. 58. Kumar R et al. Getting started with microbiome analysis: Sample acquisition to bioinformatics. Current Protocols in Human Genetics. 2014;82:18.8.1-18.8.29
  59. 59. Aldridge BB et al. Microbial metabolomics: Innovation, application, insight. Current Opinion in Microbiology. 2014;19:90-96
  60. 60. Artis D. Epithelial-cell recognition of commensal bacteria and maintenance of immune homeostasis in the gut. Nature Reviews. Immunology. 2008;8(6):411-420
  61. 61. Mowat AM. Anatomical basis of tolerance and immunity to intestinal antigens. Nature Reviews. Immunology. 2003;3(4):331-341
  62. 62. Izcue A et al. Regulatory T cells suppress systemic and mucosal immune activation to control intestinal inflammation. Immunological Reviews. 2006;212:256-271
  63. 63. Bain CC et al. Macrophages in intestinal homeostasis and inflammation. Immunological Reviews. 2014;260(1):102-117
  64. 64. Whibley N et al. Regulatory T cell adaptation in the intestine and skin. Nature Immunology. 2019;20(4):386-396
  65. 65. Stagg AJ. Intestinal dendritic cells in health and gut inflammation. Frontiers in Immunology. 2018;9:2883
  66. 66. Mann ER et al. Intestinal antigen-presenting cells in mucosal immune homeostasis: Crosstalk between dendritic cells, macrophages and B-cells. World Journal of Gastroenterology. 2014;20(29):9653-9664
  67. 67. Rios-Covian D et al. Intestinal short chain fatty acids and their link with diet and human health. Frontiers in Microbiology. 2016;7:185
  68. 68. den Besten G et al. The role of short-chain fatty acids in the interplay between diet, gut microbiota, and host energy metabolism. Journal of Lipid Research. 2013;54(9):2325-2340
  69. 69. Flint HJ. Gut microbial metabolites in health and disease. Gut Microbes. 2016;7(3):187-188
  70. 70. Louis P et al. Understanding the effects of diet on bacterial metabolism in the large intestine. Journal of Applied Microbiology. 2007;102(5):1197-1208
  71. 71. Cummings JH et al. The control and consequences of bacterial fermentation in the human colon. The Journal of Applied Bacteriology. 1991;70(6):443-459
  72. 72. Brown AJ et al. The Orphan G protein-coupled receptors GPR41 and GPR43 are activated by propionate and other short chain carboxylic acids. The Journal of Biological Chemistry. 2003;278(13):11312-11319
  73. 73. Le Poul E et al. Functional characterization of human receptors for short chain fatty acids and their role in polymorphonuclear cell activation. The Journal of Biological Chemistry. 2003;278(28):25481-25489
  74. 74. Zmora N et al. Inflammasomes and intestinal inflammation. Mucosal immunology. 2017;10(4):865-883
  75. 75. Elinav E et al. Inflammation-induced cancer: Crosstalk between tumours, immune cells and microorganisms. Nature Reviews. Cancer. 2013;13(11):759-771
  76. 76. Elinav E et al. NLRP6 inflammasome regulates colonic microbial ecology and risk for colitis. Cell. 2011;145(5):745-757
  77. 77. Hornung V et al. Silica crystals and aluminum salts activate the NALP3 inflammasome through phagosomal destabilization. Nature Immunology. 2008;9(8):847-856
  78. 78. Wang Q et al. NLRP6, decreased in gastric cancer, suppresses tumorigenicity of gastric cancer cells. Cancer Managment and Research. 2018;10:6431-6444
  79. 79. Normand S et al. Nod-like receptor pyrin domain-containing protein 6 (NLRP6) controls epithelial self-renewal and colorectal carcinogenesis upon injury. PNAS. 2011;108(23):9601-9606
  80. 80. Du Q et al. Dietary cholesterol promotes AOM-induced colorectal cancer through activating the NLRP3 inflammasome. Biochemical Pharmacology. 2016;105:42-54
  81. 81. Zitvogel L et al. Inflammasomes in carcinogenesis and anticancer immune responses. Nature Immunology. 2012;13(4):343-351
  82. 82. van Deventer HW et al. The inflammasome component NLRP3 impairs antitumor vaccine by enhancing the accumulation of tumor-associated myeloid-derived suppressor cells. Cancer Research. 2010;70(24):10161-10169
  83. 83. Chow MT et al. NLRP3 suppresses NK cell-mediated responses to carcinogen-induced tumors and metastases. Cancer Research. 2012;72(22):5721-5732
  84. 84. Zaki MH et al. The NLRP3 inflammasome protects against loss of epithelial integrity and mortality during experimental colitis. Immunity. 2010;32(3):379-391
  85. 85. Dupaul-Chicoine J et al. The Nlrp3 inflammasome suppresses colorectal cancer metastatic growth in the liver by promoting natural killer cell tumoricidal activity. Immunity. 2015;43(4):751-763
  86. 86. Karki R et al. Diverging inflammasome signals in tumorigenesis and potential targeting. Nature Reviews Cancer. 2019;19(4):197-214
  87. 87. Macia L et al. Metabolite-sensing receptors GPR43 and GPR109A facilitate dietary fibre-induced gut homeostasis through regulation of the inflammasome. Nature Communications. 2015;6:6734
  88. 88. Ohira H et al. Butyrate enhancement of interleukin-1β production via activation of oxidative stress pathways in lipopolysaccharide-stimulated THP-1 cells. Journal of Clinical Biochemistry and Nutrition. 2012;50(1):59-66
  89. 89. Singh N et al. Activation of Gpr109a, receptor for niacin and the commensal metabolite butyrate. Suppresses Colonic Inflammation and Carcinogenesis. Immunity. 2014;40(1):128-139
  90. 90. D’Souza WN et al. Differing roles for short chain fatty acids and GPR43 agonism in the regulation of intestinal barrier function and immune responses. PLoS One. 2017;12(7):e0180190
  91. 91. Falkenberg KJ et al. Histone deacetylases and their inhibitors in cancer, neurological diseases and immune disorders. Nature Reviews. Drug Discovery. 2014;13(9):673-691
  92. 92. Halsall JA et al. Histone deacetylase inhibitors for cancer therapy: An evolutionarily ancient resistance response may explain their limited success. BioEssays. 2016;38(11):1102-1110
  93. 93. Fellows R et al. Microbiota derived short chain fatty acids promote histone crotonylation in the colon through histone deacetylases. Nature Communications. 2018;9(1):105
  94. 94. Vander Heiden MG et al. Understanding the Warburg effect: The metabolic requirements of cell proliferation. Science. 2009;324(5930):1029-1033
  95. 95. Lupton JR. Microbial degradation products influence colon cancer risk: The butyrate controversy. The Journal of Nutrition. 2004;134(2):479-482
  96. 96. Donohoe DR et al. The Warburg effect dictates the mechanism of butyrate-mediated histone acetylation and cell proliferation. Molecular Cell. 2012;48(4):612-626
  97. 97. Furusawa Y et al. Commensal microbe-derived butyrate induces the differentiation of colonic regulatory T cells. Nature. 2013;504(7480):446-450
  98. 98. Arpaia N et al. Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation. Nature. 2013;504:451
  99. 99. Nastasi C et al. The effect of short-chain fatty acids on human monocyte-derived dendritic cells. Scientific Reports. 2015;5:16148
  100. 100. Qiang Y et al. Butyrate and retinoic acid imprint mucosal-like dendritic cell development synergistically from bone marrow cells. Clinical and Experimental Immunology. 2017;189(3):290-297
  101. 101. Nastasi C et al. Butyrate and propionate inhibit antigen-specific CD8(+) T cell activation by suppressing IL-12 production by antigen-presenting cells. Scientific Reports. 2017;7(1):14516
  102. 102. Kaisar MMM et al. Butyrate conditions human dendritic cells to prime type 1 regulatory T cells via both histone deacetylase inhibition and G protein-coupled receptor 109A signaling. Frontiers in Immunology. 2017;8:1429
  103. 103. Ward-Hartstonge KA et al. Regulatory T-cell heterogeneity and the cancer immune response. Clinical & Translational Immunology. 2017;6(9):e154
  104. 104. Barnes MJ et al. Regulatory T cells reinforce intestinal homeostasis. Immunity. 2009;31(3):401-411
  105. 105. Schmidt A et al. Molecular mechanisms of treg-mediated T cell suppression. Frontiers in Immunology. 2012;3:51
  106. 106. Zhang M et al. Butyrate inhibits interleukin-17 and generates Tregs to ameliorate colorectal colitis in rats. BMC Gastroenterology. 2016;16(1):84
  107. 107. Howie D et al. Foxp3 drives oxidative phosphorylation and protection from lipotoxicity. JCI insight. 2017;2(3):e89160
  108. 108. Smith PM et al. The microbial metabolites, short-chain fatty acids, regulate colonic Treg cell homeostasis. Science. 2013;341(6145):569-573
  109. 109. Jaglin M et al. Indole, a signaling molecule produced by the gut microbiota, negatively impacts emotional behaviors in rats. Frontiers in Neuroscience. 2018;12:216
  110. 110. Wikoff WR et al. Metabolomics analysis reveals large effects of gut microflora on mammalian blood metabolites. PNAS. 2009;106(10):3698-3703
  111. 111. Rothhammer V et al. The aryl hydrocarbon receptor: An environmental sensor integrating immune responses in health and disease. Nature Reviews Immunology. 2019;19(3):184-197
  112. 112. Kiss EA et al. Natural aryl hydrocarbon receptor ligands control organogenesis of intestinal lymphoid follicles. Science. 2011;334(6062):1561-1565
  113. 113. Shi LZ et al. The aryl hydrocarbon receptor is required for optimal resistance to listeria monocytogenes infection in mice. The Journal of Immunology. 2007;179(10):6952-6962
  114. 114. Qiu J et al. The aryl hydrocarbon receptor regulates gut immunity through modulation of innate lymphoid cells. Immunity. 2012;36(1):92-104
  115. 115. Lee JS et al. AHR drives the development of gut ILC22 cells and postnatal lymphoid tissues via pathways dependent on and independent of Notch. Nature Immunology. 2011;13:144
  116. 116. Zelante T et al. Tryptophan catabolites from microbiota engage aryl hydrocarbon receptor and balance mucosal reactivity via Interleukin-22. Immunity. 2013;39(2):372-385
  117. 117. Ebbo M et al. Innate lymphoid cells: Major players in inflammatory diseases. Nature Reviews. Immunology. 2017;17(11):665-678
  118. 118. Geremia A et al. IL-23-responsive innate lymphoid cells are increased in inflammatory bowel disease. The Journal of Experimental Medicine. 2011;208(6):1127-1133
  119. 119. Veldhoen M et al. The aryl hydrocarbon receptor links TH17-cell-mediated autoimmunity to environmental toxins. Nature. 2008;453:106
  120. 120. Andersson P et al. A constitutively active dioxin/aryl hydrocarbon receptor induces stomach tumors. PNAS. 2002;99(15):9990-9995
  121. 121. Moennikes O et al. A constitutively active dioxin/aryl hydrocarbon receptor promotes hepatocarcinogenesis in mice. Cancer Research. 2004;64(14):4707-4710
  122. 122. Fritz WA et al. The aryl hydrocarbon receptor inhibits prostate carcinogenesis in TRAMP mice. Carcinogenesis. 2007;28(2):497-505
  123. 123. Rooks MG et al. Gut microbiota, metabolites and host immunity. Nature Reviews. Immunology. 2016;16(6):341-352
  124. 124. Di Martino ML et al. Polyamines: Emerging players in bacteria-host interactions. International Journal of Medical Microbiology. 2013;303(8):484-491
  125. 125. Zhang M et al. Spermine inhibition of monocyte activation and inflammation. Molecular Medicine. 1999;5(9):595-605
  126. 126. Hasko G et al. Spermine differentially regulates the production of interleukin-12 p40 and interleukin-10 and suppresses the release of the T helper 1 cytokine interferon-gamma. Shock. 2000;14(2):144-149
  127. 127. Postler TS et al. Understanding the Holobiont: How microbial metabolites affect human health and shape the immune system. Cell Metabolism. 2017;26(1):110-130
  128. 128. Timmons J et al. Polyamines and gut mucosal homeostasis. Journal of Gastrointestinal and Digestive System. 2012;2(Suppl 7). pii: 001
  129. 129. Enright EF et al. Microbiome-mediated bile acid modification: Role in intestinal drug absorption and metabolism. Pharmacological Research. 2018;133:170-186
  130. 130. Wilson FA et al. Unstirred water layers in intestine: Rate determinant of fatty acid absorption from micellar solutions. Science (New York, N.Y.). 1971;174(4013):1031-1033
  131. 131. Copple BL et al. Pharmacology of bile acid receptors: Evolution of bile acids from simple detergents to complex signaling molecules. Pharmacological Research. 2016;104:9-21
  132. 132. Gadaleta RM et al. Activation of bile salt nuclear receptor FXR is repressed by pro-inflammatory cytokines activating NF-κB signaling in the intestine. Biochimica et Biophysica Acta (BBA)—Molecular Basis of Disease. 2011;1812(8):851-858
  133. 133. Gadaleta RM et al. Farnesoid X receptor activation inhibits inflammation and preserves the intestinal barrier in inflammatory bowel disease. Gut. 2011;60(4):463-472
  134. 134. Wang Y-D et al. Farnesoid X receptor antagonizes nuclear fjactor kappaB in hepatic inflammatory response. Hepatology (Baltimore, Md.). 2008;48(5):1632-1643
  135. 135. Vavassori P et al. The bile acid receptor FXR is a modulator of intestinal innate immunity. Journal of Immunology. 2009;183(10):6251-6261
  136. 136. Haselow K et al. Bile acids PKA-dependently induce a switch of the IL-10/IL-12 ratio and reduce proinflammatory capability of human macrophages. Journal of Leukocyte Biology. 2013;94(6):1253-1264
  137. 137. Hao H et al. Farnesoid X receptor regulation of the NLRP3 inflammasome underlies cholestasis-associated sepsis. Cell Metabolism. 2017;25(4):856-867.e5
  138. 138. Guo C et al. Bile acids control inflammation and metabolic disorder through inhibition of NLRP3 Inflammasome. Immunity. 2016;45(4):802-816
  139. 139. Levy M et al. Microbiota-modulated metabolites shape the intestinal microenvironment by regulating NLRP6 inflammasome signaling. Cell. 2015;163(6):1428-1443
  140. 140. Iwase T et al. Isolation and identification of ATP-secreting bacteria from mice and humans. Journal of Clinical Microbiology. 2010;48(5):1949-1951
  141. 141. Mempin R et al. Release of extracellular ATP by bacteria during growth. BMC Microbiology. 2013;13(1):301
  142. 142. Bergsbaken T et al. Pyroptosis: Host cell death and inflammation. Nature Reviews. Microbiology. 2009;7(2):99-109
  143. 143. Forrester T et al. Release of adenosine triphosphate from isolated adult heart cells in response to hypoxia. The Journal of Physiology. 1977;268(2):371-390
  144. 144. Di Virgilio F et al. Extracellular purines, purinergic receptors and tumor growth. Oncogene. 2017;36(3):293-303
  145. 145. Burnstock G et al. Cellular distribution and functions of P2 receptor subtypes in different systems. International Review of Cytology. 2004;240:31-304
  146. 146. Yaron JR et al. K+ regulates Ca2+ to drive inflammasome signaling: Dynamic visualization of ion flux in live cells. Cell Death and Disease. 2015;6:e1954
  147. 147. Ghiringhelli F et al. Activation of the NLRP3 inflammasome in dendritic cells induces IL-1beta-dependent adaptive immunity against tumors. Nature Medicine. 2009;15(10):1170-1178
  148. 148. Chen Y et al. ATP release guides neutrophil chemotaxis via P2Y2 and A3 receptors. Science (New York, N.Y.). 2006;314(5806):1792-1795
  149. 149. Saez PJ et al. ATP promotes the fast migration of dendritic cells through the activity of pannexin 1 channels and P2X7 receptors. Science Signaling. 2017;10(506). pii: eaah7107
  150. 150. Muller T et al. The purinergic receptor P2Y2 receptor mediates chemotaxis of dendritic cells and eosinophils in allergic lung inflammation. Allergy. 2010;65(12):1545-1553
  151. 151. Junger WG. Immune cell regulation by autocrine purinergic signalling. Nature Reviews. Immunology. 2011;11(3):201-212
  152. 152. Di Virgilio F et al. Extracellular ATP and P2 purinergic signalling in the tumour microenvironment. Nature Reviews Cancer. 2018;18(10):601-618
  153. 153. Schnurr M et al. ATP gradients inhibit the migratory capacity of specific human dendritic cell types: Implications for P2Y11 receptor signaling. Blood. 2003;102(2):613-620
  154. 154. Trabanelli S et al. Extracellular ATP exerts opposite effects on activated and regulatory CD4+ T cells via purinergic P2 receptor activation. Journal of Immunology (Baltimore, Md.: 1950). 2012;189(3):1303-1310
  155. 155. Atarashi K et al. ATP drives lamina propria T(H)17 cell differentiation. Nature. 2008;455(7214):808-812
  156. 156. Friedman DJ et al. CD39 deletion exacerbates experimental murine colitis and human polymorphisms increase susceptibility to inflammatory bowel disease. PNAS. 2009;106(39):16788-16793
  157. 157. Neves AR et al. Overexpression of ATP-activated P2X7 receptors in the intestinal mucosa is implicated in the pathogenesis of Crohn's disease. Inflammatory Bowel Diseases. 2014;20(3):444-457
  158. 158. Lee JS et al. Unfolding role of a danger molecule adenosine signaling in modulation of microbial infection and host cell response. International Journal of Molecular Sciences. 2018;19(1). pii: E199
  159. 159. Stagg J et al. CD73-deficient mice have increased antitumor immunity and are resistant to experimental metastasis. Cancer Research. 2011;71(8):2892-2900
  160. 160. Stagg J et al. Anti-CD73 antibody therapy inhibits breast tumor growth and metastasis. PNAS. 2010;107(4):1547-1552
  161. 161. Young A et al. Co-inhibition of CD73 and A2AR adenosine signaling improves anti-tumor immune responses. Cancer Cell. 2016;30(3):391-403
  162. 162. Vijayan D et al. Targeting immunosuppressive adenosine in cancer. Nature Reviews. Cancer. 2017;17(12):709-724
  163. 163. Allard B et al. The ectonucleotidases CD39 and CD73: Novel checkpoint inhibitor targets. Immunological Reviews. 2017;276(1):121-144
  164. 164. Borges da Silva H et al. The purinergic receptor P2RX7 directs metabolic fitness of long-lived memory CD8(+) T cells. Nature. 2018;559(7713):264-268
  165. 165. LeBlanc JG et al. Bacteria as vitamin suppliers to their host: A gut micro-biota perspective. Current Opinion in Biotechnology. 2013;24(2):160-168
  166. 166. Song M et al. Nutrients, foods, and colorectal cancer prevention. Gastroenterology. 2015;148(6):1244-60.e16
  167. 167. Eslami M et al. Importance of probiotics in the prevention and treatment of colorectal cancer. Journal of Cellular Physiology. 2019;234(10):17127-17143
  168. 168. Kunisawa J et al. A pivotal role of vitamin B9 in the maintenance of regulatory T cells in vitro and in vivo. PLoS One. 2012;7(2):e32094
  169. 169. Kunisawa J et al. Sphingosine 1-phosphate dependence in the regulation of lymphocyte trafficking to the gut epithelium. The Journal of Experimental Medicine. 2007;204(10):2335-2348
  170. 170. Schwab SR et al. Lymphocyte sequestration through S1P lyase inhibition and disruption of S1P gradients. Science. 2005;309(5741):1735-1739
  171. 171. Treiner E et al. Selection of evolutionarily conserved mucosal-associated invariant T cells by MR1. Nature. 2003;422(6928):164-169
  172. 172. Dusseaux M et al. Human MAIT cells are xenobiotic-resistant, tissue-targeted, CD161hi IL-17-secreting T cells. Blood. 2011;117(4):1250-1259
  173. 173. Tang XZ et al. IL-7 licenses activation of human liver intrasinusoidal mucosal-associated invariant T cells. Journal of Immunology. 2013;190(7):3142-3152
  174. 174. Kjer-Nielsen L et al. An overview on the identification of MAIT cell antigens. Immunology and Cell Biology. 2018;96(6):573-587
  175. 175. Le Bourhis L et al. MAIT cells detect and efficiently lyse bacterially-infected epithelial cells. PLoS Pathogens. 2013;9(10):e1003681
  176. 176. Gold MC et al. Human mucosal associated invariant T cells detect bacterially infected cells. PLoS Biology. 2010;8(6):e1000407
  177. 177. Kurioka A et al. MAIT cells are licensed through granzyme exchange to kill bacterially sensitized targets. Mucosal Immunology. 2014;8:429
  178. 178. Kumar V et al. Role of MAIT cells in the immunopathogenesis of inflammatory diseases: New players in old game. International Reviews of Immunology. 2018;37(2):90-110
  179. 179. Won EJ et al. Clinical relevance of circulating mucosal-associated invariant T cell levels and their anti-cancer activity in patients with mucosal-associated cancer. Oncotarget. 2016;7(46):76274-76290
  180. 180. Ling L et al. Circulating and tumor-infiltrating mucosal associated invariant T (MAIT) cells in colorectal cancer patients. Scientific Reports. 2016;6:20358
  181. 181. Sundstrom P et al. Human mucosa-associated invariant T cells accumulate in colon adenocarcinomas but produce reduced amounts of IFN-gamma. Journal of Immunology. 2015;195(7):3472-3481
  182. 182. Haeryfar SMM et al. Mucosa-associated invariant T cells in malignancies: A faithful friend or formidable foe? Cancer Immunology, Immunotherapy: CII. 2018;67(12):1885-1896
  183. 183. Mazmanian SK et al. A microbial symbiosis factor prevents intestinal inflammatory disease. Nature. 2008;453(7195):620-625
  184. 184. Dasgupta S et al. Plasmacytoid dendritic cells mediate anti-inflammatory responses to a gut commensal molecule via both innate and adaptive mechanisms. Cell Host & Microbe. 2014;15(4):413-423
  185. 185. Cobb BA et al. Polysaccharide processing and presentation by the MHCII pathway. Cell. 2004;117(5):677-687
  186. 186. Atarashi K et al. Induction of colonic regulatory T cells by indigenous clostridium species. Science. 2011;331(6015):337-341
  187. 187. Derosa L et al. Negative association of antibiotics on clinical activity of immune checkpoint inhibitors in patients with advanced renal cell and non-small-cell lung cancer. Annals of Oncology. 2018;29(6):1437-1444
  188. 188. Gopalakrishnan V et al. Gut microbiome modulates response to anti-PD-1 immunotherapy in melanoma patients. Science. 2018;359(6371):97-103
  189. 189. Iida N et al. Commensal bacteria control cancer response to therapy by modulating the tumor microenvironment. Science. 2013;342(6161):967-970
  190. 190. Matson V et al. The commensal microbiome is associated with anti-PD-1 efficacy in metastatic melanoma patients. Science. 2018;359(6371):104-108
  191. 191. Routy B et al. Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors. Science (New York, N.Y.). 2018;359(6371):91-97
  192. 192. Taur Y et al. The effects of intestinal tract bacterial diversity on mortality following allogeneic hematopoietic stem cell transplantation. Blood. 2014;124(7):1174-1182
  193. 193. Viaud S et al. The intestinal microbiota modulates the anticancer immune effects of cyclophosphamide. Science. 2013;342(6161):971-976
  194. 194. Routy B et al. The gut microbiota influences anticancer immunosurveillance and general health. Nature Reviews. Clinical Oncology. 2018;15(6):382-396

Written By

Silke Neumann, Estelle M. Peyroux, Matt J. Woodall, Nick J. Shields, Sarah L. Young and Sharon T. Pattison

Submitted: 03 May 2019 Reviewed: 18 June 2019 Published: 02 August 2019