Transporters and receptors for short-chain fatty acids as the molecular link between colonic bacteria and the host

https://doi.org/10.1016/j.coph.2013.08.006Get rights and content

Highlights

  • The relationship between colonic bacteria and the host is not commensal but mutual.

  • Bacterial fermentation products serve as the messengers in this relationship.

  • The fermentation products protect against inflammation and cancer in colon.

  • SLC5A8 is critical for bacteria–host communication only when fiber intake is low.

  • GPR109A and GPR43 are critical for the communication even with optimal fiber intake.

The mutually beneficial relationship between colonic bacteria and the host has been recognized but the molecular aspects of the relationship remain poorly understood. Dietary fiber is critical to this relationship. The short-chain fatty acids acetate, propionate and butyrate, generated by bacterial fermentation of dietary fiber, serve as messengers between colonic bacteria and the host. The beneficial effects of these bacterial metabolites in colon include, but are not limited to, suppression of inflammation and prevention of cancer. Recent studies have identified the plasma membrane transporter SLC5A8 and the cell-surface receptors GPR109A and GPR43 as essential for the biologic effects of short-chain fatty acids in colon. These three proteins coded by the host genome provide the molecular link between colonic bacteria and the host.

Introduction

The human body harbors trillions (∼1014) of microorganisms, most of them present in the large intestine, playing an important role in the health of the host. Though called ‘commensal bacteria’, the relationship between gut-associated bacteria and the host is not commensal. The term ‘commensalism’ was based on the belief that only bacteria benefit from this co-habitation while the host neither benefits nor is harmed. From what we know now, nothing can be farther from the truth. Normal bacteria in colon have profound effects on the host, including protection against enteric infections, production of vitamins, maturation of intestinal function, assistance in digestion and absorption, modulation of the mucosal immune system in the gut, and maintenance of energy homeostasis [1, 2, 3]. Thus, the term ‘mutualism’ is more justifiable than ‘commensalism’ to describe the relationship between gut bacteria and the host. The well-deserved recognition of the importance of gut bacteria to our health has been one of the hallmarks of the last decade, highlighted by the initiation of the Human Microbiome Project by the National Institutes of Health in the United States. This project has delineated the range of structural and functional diversity of the microbial communities that reside in different parts, including the large intestine, of a healthy human body [4••]. The recent progress relating to the microbial ecology of the mammalian gut in health and disease has been hailed as one of the ten major breakthroughs of the year 2011 [5].

The health benefits of colonic bacteria are principally related to dietary fiber, which includes resistant starch and non-starch polysaccharides such as cellulose, hemicellulose, pectins, and gums. There are two classes of dietary fiber: soluble and insoluble. Most of the soluble fibers are fermentable by colonic bacteria and used as the nutrient source to support the growth of these microorganisms in the colon. It has been known for decades that the short-chain fatty acids (SCFAs: acetate, propionate, and butyrate), produced by bacterial fermentation of dietary fiber, are the mediators of the beneficial effects of colonic bacteria on the host [6, 7, 8], but the molecular mechanisms underlying the process remained poorly understood. This has changed dramatically in recent years. In this review, we summarize the recent discoveries on the role of three genes in the host genome, namely SLC5A8, GPR109A, and GPR43, as the critical determinants of the bacteria–host communication.

Section snippets

SCFAs as bacterial metabolites and their function in the colon

Most of the bacteria in the mammalian large intestine are anaerobes, which ferment dietary carbohydrates, particularly resistant starch and dietary fiber, into acetate, propionate, and butyrate. The relative proportions of these three bacterial metabolites in the colonic lumen are 6:3:1, and the total concentration is approximately 100 mm. These metabolites are readily absorbed in the colon and serve as the preferred energy substrates for colonocytes. Only small amounts of these SCFAs leave the

SLC5A8, a plasma membrane transporter, and its relevance to intracellular actions of SCFAs in colonocytes and immune cells

SLC5A8 was originally identified as a tumor suppressor that is silenced in colon cancer by DNA methylation [13]. When re-expressed in colon cancer cells, SLC5A8 causes growth arrest and apoptosis. However, the identity of the substrate that is transported by the protein was not known. How the transport function could be linked to tumor suppression also remained unclear. Studies from our laboratory showed for the first time that SLC5A8 is a Na+-coupled transporter for a variety of SCFAs,

GPR109A, a cell-surface G-protein-coupled receptor, and its relevance to the biologic actions of butyrate in colon

GPR109A (hydroxycarboxylic acid receptor 2 or HCA2) was first identified as a G-protein-coupled receptor that is induced by IFN-γ in macrophages [26] and thus named PUMA-G (Protein Upregulated in MAcrophages by IFN-Gamma). GPR109A in humans corresponds to PUMA-G in rodents [27]. GPR109A is expressed not only in macrophages but also in adipocytes. Two independent groups of investigators, while searching for the mechanism underlying the lipid-lowering effects of niacin, discovered that niacin

GPR43, a cell-surface G-protein-coupled receptor, and its relevance to the biologic actions of SCFAs

GPR43 (free fatty acid receptor 2; FFAR2), is also a cell-surface G-protein-coupled receptor expressed in the colon, which is activated by SCFAs, primarily acetate and propionate [33, 34]. Activation of GPR43 signaling by SCFAs plays a role in the regulation of appetite, electrolyte and fluid secretion, and inflammation [35, 36, 37••, 38, 39]. Gpr43−/− mice show reduced inflammatory responses against infection with the pathogenic bacterium C. rodentium while activation of the receptor with

Influence of gut bacteria on the intestinal expression of Slc5a8 and Gpr109a

Since butyrate, a bacterial fermentation product, is the substrate for SLC5A8 and agonist for GPR109A, we asked: Do gut bacteria influence the expression of SLC5A8 and GPR109A in colon? We addressed this question by comparing the expression of these two genes in the intestinal tract between conventional mice with normal bacteria in colon and germ-free mice with no bacteria in the intestine or anywhere else in the body [31••]. Slc5a8 mRNA and Gpr109a mRNA levels in colon and ileum were reduced

Dietary fiber content and its relationship to the functions of SLC5A8, GPR109A, and GPR43

Our hypothesis is that SLC5A8, GPR109A, and GPR43 are obligatory for the beneficial effects of SCFAs in colon. Knockout mouse models with deletion of Slc5a8, Gpr109a, and Gpr43 would be useful to test this hypothesis. Slc5a8−/− mice have been generated recently [41]. Surprisingly, treatment of Slc5a8−/− mice with carcinogens or crossing these mice with ApcMin/+ mice did not show any difference in tumor formation. Even though these findings seem to question the tumor-suppressive role of Slc5a8,

Nutritional and pharmacological implications

Optimal concentrations of SCFAs in the colonic lumen are essential for maintenance of colonic health. The concentrations of SCFAs in the colon can be reduced under various conditions such as intake of low dietary fiber and chronic use of antibiotics. Generation of SCFAs in colon can be optimized with nutritional interventions such as intake of appropriate prebiotics and probiotics. A recent study has shown that supplementation of the probiotic Lactobacillus GG and the butyrate ester tributyrin

Conclusions

On the basis of our current knowledge, we conclude that firstly the anti-inflammatory and tumor-suppressive functions of the high-affinity butyrate/propionate transporter SLC5A8 are dispensable under conditions of high-fiber intake but not dispensable under conditions of low-fiber intake, secondly the anti-inflammatory and tumor-suppressive signaling via the low-affinity SCFA receptors GPR109A and GPR43 is optimal only when the dietary fiber content is high but becomes suboptimal under

References and recommended reading

Papers of particular interest, published within the period of review, have been highlighted as:

  • • of special interest

  • •• of outstanding interest

References (44)

  • I. Cho et al.

    The human microbiome: at the interface of health and disease

    Nat Rev Genet

    (2012)
  • C.A. Lozupone et al.

    Diversity, stability and resilience of the human gut microbiota

    Nature

    (2012)
  • Structure, function and diversity of the healthy human microbiome

    Nature

    (2012)
  • Breakthrough of the Year: The Runners-Up. Microbes R Us

    Science

    (2011)
  • J.M. Wong et al.

    Colonic health: fermentation and short-chain fatty acids

    J Clin Gastroenterol

    (2006)
  • C.C. Roy et al.

    Short-chain fatty acids: ready for prime time?

    Nutr Clin Pract

    (2006)
  • R. Havenaar

    Intestinal health functions of colonic microbial metabolites: a review

    Benef Microbes

    (2011)
  • J.S. Chen et al.

    Short-chain fatty acid inhibitors of histone deacetylases: promising anticancer therapeutics?

    Curr Cancer Drug Targets

    (2003)
  • S. Kolida et al.

    Synbiotics in health and disease

    Annu Rev Food Sci Technol

    (2011)
  • M. Balakrishnan et al.

    Prebiotics, probiotics and digestive health

    Curr Opin Clin Nutr Metab Care

    (2012)
  • E.M. Quigley

    Prebiotics and probiotics: their role in the management of gastrointestinal disorders in adults

    Nutr Clin Pract

    (2012)
  • H. Li et al.

    SLC5A8, a sodium transporter, is a tumor suppressor gene silenced by methylation in human colon aberrant crypt foci and cancers

    Proc Natl Acad Sci U S A

    (2003)
  • Cited by (231)

    • The Promise of Niacin in Neurology

      2023, Neurotherapeutics
    View all citing articles on Scopus
    View full text