Hostname: page-component-7c8c6479df-ws8qp Total loading time: 0 Render date: 2024-03-28T13:20:56.878Z Has data issue: false hasContentIssue false

A review of the potential mechanisms for the lowering of colorectal oncogenesis by butyrate

Published online by Cambridge University Press:  07 June 2012

Kim Y. C. Fung*
Affiliation:
CSIRO Preventative Health National Research Flagship, Adelaide, SA5000, Australia CSIRO Food and Nutritional Sciences, Adelaide, SA5000, Australia
Leah Cosgrove
Affiliation:
CSIRO Preventative Health National Research Flagship, Adelaide, SA5000, Australia CSIRO Food and Nutritional Sciences, Adelaide, SA5000, Australia
Trevor Lockett
Affiliation:
CSIRO Preventative Health National Research Flagship, Adelaide, SA5000, Australia CSIRO Food and Nutritional Sciences, North Ryde, NSW2113, Australia
Richard Head
Affiliation:
CSIRO Preventative Health National Research Flagship, Adelaide, SA5000, Australia
David L. Topping
Affiliation:
CSIRO Preventative Health National Research Flagship, Adelaide, SA5000, Australia CSIRO Food and Nutritional Sciences, Adelaide, SA5000, Australia CSIRO Food and Nutritional Sciences, North Ryde, NSW2113, Australia CSIRO Food Futures National Research Flagship, North Ryde, NSW2113, Australia
*
*Corresponding author: Dr K. Y. C. Fung, fax +61 8 8303 8899, email kim.fung@csiro.au
Rights & Permissions [Opens in a new window]

Abstract

Colorectal cancer (CRC) is a leading cause of preventable cancer deaths worldwide, with dietary factors being recognised as key risk modifiers. Foods containing dietary fibre are protective to a degree that the World Cancer Research Fund classifies the evidence supporting their consumption as ‘convincing’. The mechanisms by which fibre components protect against CRC remain poorly understood, especially their interactions with the gut microbiome. Fibre is a composite of indigestible plant polysaccharides and it is emerging that fermentable fibres, including resistant starch (RS), are particularly important. RS fermentation induces SCFA production, in particular, relatively high butyrate levels, and in vitro studies have shown that this acid has strong anti-tumorigenic properties. Butyrate inhibits proliferation and induces apoptosis of CRC cell lines at physiological concentrations. These effects are attributed to butyrate's ability to alter gene transcription by inhibiting histone deacetylase activity. However, the more recent discovery of G-protein coupled receptors that bind butyrate and other SCFA and data obtained from proteomic and genomic experiments suggest that alternative pathways are involved. Here, we review the mechanisms involved in butyrate-induced apoptosis in CRC cells and, additionally, the potential role this SCFA may play in mediating key processes in tumorigenesis including genomic instability, inflammation and cell energy metabolism. This discussion may help to inform the development of strategies to lower CRC risk at the individual and population levels.

Type
Review Article
Copyright
Copyright © The Authors 2012

Colorectal cancer (CRC) has emerged as one of the most prevalent types of cancer worldwide and is the fourth most common cause of cancer mortality(Reference Ferlay, Shin and Bray1). CRC incidence varies considerably across geographical regions. It is the highest (approximately 20–45 per 100 000) among affluent societies such as Australia/New Zealand, Europe, the USA and the UK, and the lowest among African and Asian countries (approximately 5–20 per 100 000)(Reference Ferlay, Shin and Bray1). Japan currently records the highest incidence of CRC and this appears likely to persist and continue over time(Reference Center, Jemal and Smith2, Reference Parkin, Bray and Ferlay3). In countries such as Korea, Singapore and Eastern Europe, the incidence of disease is approaching that of high-risk countries with a longer history of affluence(Reference Center, Jemal and Smith2, Reference Haggar and Boushey4, Reference Wong and Eu5). This increase in the risk of CRC has been attributed to industrialisation and accompanying environmental influences associated with a transition from a low- to high-income economy(Reference Center, Jemal and Smith2). Epidemiological studies have drawn strong correlations between CRC incidence and modifiable lifestyle factors such as body weight, diet, physical activity, smoking and alcohol consumption(Reference Haggar and Boushey4). These associations indicate the strong possibility of CRC prevention and it is believed that 30–60 % of cases can be prevented with appropriate nutrition and diet(6, Reference Platz, Willett and Colditz7).

Dietary fibre is one of the most promising candidates for a protective role in CRC, with strong support from epidemiological and experimental animal studies. However, there is a degree of ambiguity in the population data, with some studies showing no significant effect(Reference Bingham, Day and Luben8, Reference Park, Hunter and Spiegelman9). This may reflect the food sources of fibre. Recently, the European Prospective Investigation into Cancer and Nutrition reported that although total dietary fibre was associated with a 30 % reduction in CRC risk, no one food source offered more protection than another(Reference Bingham, Day and Luben8). While fibre is derived largely from plant foods, it must be recognised that any protective effects of particular fibre-containing food subgroups (e.g. fruits and vegetables) can also be contributed by other nutrients present such as β-carotene, lycopene and polyphenols(Reference McCullough, Robertson and Chao10). Dietary fibre consists largely of plant polysaccharides that resist human small-intestinal enzymes and some of the protection against colorectal tumorigenesis may reflect this bulking action. Greater stool mass is expected to lower the exposure of colonocytes to carcinogens and mutagens through physical dilution and also through reduction in transit time. There is also a strong case for protection through the interactions between the large-bowel microbiome and fibre polysaccharides, which are emerging as a critical factor in the promotion of optimal colonic function(Reference Brouns, Kettlitz and Arrigoni11, Reference Topping and Clifton12). As in obligate herbivores, so too in humans, there is substantial microbial fermentation of fibre with SCFA, primarily acetate, propionate and butyrate, as significant end products(Reference Topping and Clifton12, Reference Bugaut13). The rate of fermentation of fibre varies according to type and food source; e.g. fibre derived from grains is fermented much more slowly and less completely than that from fruits and vegetables(Reference Freudenheim, Graham and Horvath14, Reference Stephen and Cummings15). Cellulose is a major constituent of plant cell walls in both cereals and fruits and vegetables, but there are major differences between the two groups. Soluble fibre polysaccharides are generally higher in fruits and vegetables, reflecting their higher content of uronic acids. In contrast, cereal grains contain more arabinoxylans, mixed-linkage glucans and oligosaccharides(Reference Marlett16, Reference Topping17). There is also a major difference between the various fibre polysaccharides (e.g. NSP and resistant starch (RS)) in the profile of SCFA which are produced. In the diets of industrialised countries at high risk of CRC (e.g. Australia), fibre intake is largely as cereal NSP with relatively little RS. In contrast, it appears that in traditional agrarian societies at low risk, NSP intakes are comparatively low but RS intakes are high. This is likely to be a significant factor in disease as RS fermentation favours butyrate production over that of NSP and large-bowel butyrate levels are higher when foods common to such populations are consumed(Reference Ahmed, Segal and Hassan18). Importantly, butyrate is the primary energy source for colonocytes where its oxidation contributes to at least 60 % of the cell's energy requirements(Reference Cummings19) and it is the SCFA most associated with protection against colorectal carcinogenesis(Reference Perrin, Pierre and Patry20).

This review focuses on how butyrate, a SCFA produced in the colon by the fermentation of dietary fibre, potentially prevents colorectal oncogenesis. Here, we review the experimental data derived from in vitro and animal model systems describing the mechanisms, in addition to histone deacetylase (HDAC) inhibition, influencing butyrate's anti-tumorigenic actions. Taken together, these data provide compelling evidence to support human intervention studies to determine the true potential of RS in lowering the risk of colorectal oncogenesis in the wider population.

Anti-tumorigenic properties of butyrate

SCFA play a significant role in maintaining the normal physiological functions of the colonic mucosa(Reference Topping and Clifton12) (Fig. 1). Although acetate is the most abundant colonic SCFA, butyrate has been studied the most due to its potent anti-tumorigenic properties. Butyrate inhibits proliferation and induces differentiation and apoptosis of CRC cells in vitro at concentrations similar to those found in the large bowel in vivo. Increased butyrate supply reduces the incidence of carcinogen-induced colon tumours in rodent models, partly through induction of apoptosis(Reference Clarke, Topping and Bird21, Reference Le Leu, Hu and Brown22). It also opposes diet-induced colonocyte DNA damage in animals, supporting its potential to promote genetic stability(Reference Toden, Bird and Topping23). Animal experiments have shown that consumption of red meat induces the formation of N-nitroso-compounds and DNA adducts, in particular the O6-methyl-adduct of 2′-deoxyguanosine(Reference Lewin, Bailey and Bandaletova24). Moreover, it has been shown that tissues display different abilities in the removal of these adducts(Reference Tan, Gerber and Cosgrove25). Failure to remove these adducts via either intrinsic DNA repair mechanisms or apoptosis results in the elevation of mutation rates. Where mutations occur in key oncogenes (e.g. Kirsten rat sarcoma viral oncogene homolog; KRAS) or tumour suppressor genes (e.g. p53), the risk for CRC development can rise dramatically(Reference Kampman, Voskuil and van Kraats26, Reference Slattery, Curtin and Anderson27).

Fig. 1 The effects of butyrate in the normal colon and in colorectal tumour cells. HDAC, histone deacetylase.

The effects of butyrate are highly selective for cancer cells and its ability to modulate numerous cellular processes has contributed to the difficulty in identifying the precise mechanisms underlying each of its anti-tumorigenic properties, in particular the ability to induce apoptosis. Some of the key molecules mediating butyrate's action are summarised in Table 1. Many proteomic and transcriptomic studies have been conducted to understand the signalling pathways involved in butyrate-induced apoptosis(Reference Daly and Shirazi-Beechey28Reference Tan, Seow and Liang33) and also to understand the mechanisms involved in the development of butyrate resistance; i.e. how a sub-population of cancer cells circumvents apoptosis in a butyrate-rich environment to form tumours(Reference Fung, Brierley and Henderson29, Reference Fung, Lewanowitsch and Henderson30, Reference Olmo, Turnay and Perez-Ramos34). Studies have shown that CRC cells that are glycolytic and have adapted to metabolise butyrate in a high-butyrate environment; i.e. butyrate-resistant cells have a growth advantage and potentially form more aggressive cancers(Reference Lopez de Silanes, Olmo and Turnay35, Reference Serpa, Caiado and Carvalho36). Although the underlying mechanisms and signalling pathways leading to these observed changes remain elusive, the recent discovery of receptors with affinity for butyrate and other SCFA strengthens the possibility that butyrate-induced apoptosis is mediated by mechanisms in addition to HDAC inhibition.

Table 1 Summary of the genes and proteins involved in the anti-tumorigenic effects of butyrate

JNK, c-Jun N-terminal kinase; ERK, extracellular signal-regulated kinase; MCP1, monocyte chemoattractant protein 1; GM-CSF, granulocyte-macrophage colony stimulating factor; VEGF, vascular endothelial growth factor; CRC, colorectal cancer.

Mechanisms of butyrate-induced apoptosis in colorectal cancer cells

Studies focused on the mechanisms involved in butyrate-induced apoptosis have consistently demonstrated a rapid release of cytochrome c into the cytosol and activation of the caspase cascade as key features. In vitro studies have demonstrated that butyrate (>0·5 mmol/l) induces apoptosis in CRC cell lines(Reference Kim, Park and Lee37). Butyrate is reported to activate the intrinsic pathway of apoptosis, to sensitise cancer cells to apoptosis mediated by the extrinsic pathway and, more recently, butyrate has been shown to induce autophagic cell death(Reference Pajak, Gajkowska and Orzechowski38Reference Wang, Luo and Xia41). These pathways appear to be activated in parallel to amplify the apoptotic response. Dysregulated expressions, resulting from butyrate treatment of pro- and anti-apoptotic proteins such as those belonging to the Bcl-2 protein family(Reference Avivi-Green, Polak-Charcon and Madar42, Reference Ruemmele, Dionne and Qureshi43) or the TNF receptor superfamily have also been reported(Reference Kim, Park and Lee37, Reference Pajak, Gajkowska and Orzechowski38).

Although activation of these cascades by butyrate has been demonstrated consistently, the triggers responsible for their initiation remain elusive. Butyrate (at 2–4 mmol/l) elicits a cell stress response in vitro characterised by the activation of genes such as those belonging to the growth arrest and DNA damage-inducible (GADD) family and activation of the mitogen-activated protein kinase (MAPK) signalling pathway(Reference Scott, Longpre and Loo44Reference Zhang, Zhou and Bao46). A report by Scott et al. (Reference Scott, Longpre and Loo44) correlated MAPK activation by butyrate with induction of GADD153 in HCT116 cells. In the RKO CRC cell line, butyrate activated the c-Jun N-terminal kinase (JNK) but not the p38 arm of the MAPK signalling pathway and this correlated with caspase activation and apoptosis(Reference Zhang, Zhou and Bao46). Tong et al. (Reference Tong, Yin and Song45) reported the loss of expression in both mouse and human intestinal tumours, lending further support to its role in intestinal tumour formation. Butyrate is also reported to increase phosphorylation of p38 and its downstream target heat shock protein 27 in both HCT116 CRC cells and MCF7 breast cancer cells(Reference Fung, Brierley and Henderson29, Reference Yonezawa, Kobayashi and Obara47). Although the apoptotic response to butyrate is dependent on the cell line being studied, in each case, the activation of MAPK signalling occurs within minutes of butyrate exposure. This indicates that induction of a cell stress response occurs as an early event in butyrate-induced apoptosis.

Effect of butyrate on processes involved in tumorigenesis

Genomic instability and epigenetic regulation

Histone deacetylase inhibition

Regulation of gene expression via inhibition of HDAC activity is the primary mechanism associated with butyrate-induced apoptosis. HDAC inhibitors (including butyrate) target the transcription of less than 10 % of the human genome selectively and their cellular effects are also mediated by modulating the acetylation state of both histone and non-histone proteins including transcription factors, structural proteins and proteins involved in signal transduction(Reference Wilson, Chueh and Togel48). Our knowledge of the number of non-histone proteins modified as a result of butyrate treatment is limited but is known to include the Sp1 and Sp3 transcription factors(Reference Waby, Chirakkal and Yu49, Reference White, Mulligan and King50). Although the functional consequences of Sp1 and Sp3 acetylation by butyrate are not known, acetylation of Sp1 has been hypothesised to increase p21 expression and mediate p53-dependent cell cycle arrest in CRC cell lines(Reference Waby, Chirakkal and Yu49). Identification of other non-histone protein targets acetylated by butyrate treatment may provide new insights into butyrate's mechanisms of action.

Role of p53 in cell cycle arrest and apoptosis

In vitro studies have shown consistently that butyrate induces cell cycle arrest and apoptosis in both a p53-dependent and -independent manner at physiologically relevant concentrations (0·6–5 mmol/l)(Reference Janson, Brandner and Siegel51). In CRC cell lines, butyrate down-regulates the expression of p53 mRNA and protein and also directly increases the expression of p53 target genes (e.g. p21WAF1, p27 and cyclin-dependent kinases) to induce cell cycle arrest(Reference Gope and Gope52, Reference Nakano, Mizuno and Sowa53). Activation of p53 and its translocation to the nucleus is regulated by post-translational modifications, including acetylation by histone acetyltransferases such as p300, that can increase both the stability and pro-apoptotic activity of p53(Reference Yuan, Huang and Ishiko54). Prolonged or transient hyper-acetylation of p53 by HDAC inhibitors such as butyrate may represent an additional mechanism making an impact on p53-dependent apoptosis(Reference Terui, Murakami and Takimoto55).

Butyrate and micro-RNA regulation

Butyrate also alters gene expression independently of HDAC inhibition by regulating the expression of micro-RNA (miRNA)(Reference Hu, Dong and Dalal56, Reference Zhang, Li and Nan57). micro-RNA are non-protein coding RNA species that regulate translation of their respective mRNA targets. Over 1000 human miRNA have been identified (www.miBase.org accessed June 2011). Despite the intensity of effort, only a small number of miRNA, notably miR-31 and those of the miR-194/-215 and miR-143/-145 clusters, have consistently been associated with colorectal tumorigenesis(Reference Necela, Carr and Asmann58). In vitro studies have demonstrated that butyrate regulates the expression of miRNA in HCT116 cells and in human CRC stem cells characterised by CD133 cell surface expression when compared to respective control cells, including those mentioned previously(Reference Hu, Dong and Dalal56, Reference Zhang, Li and Nan57). Establishing the role of miRNA in the regulation of gene transcription is still in its infancy and the true potential that butyrate may have on miRNA expression, or regulation of its function, remains to be elucidated.

Inflammation and the immune response

SCFA have anti-inflammatory effects in the large bowel. In patients with distal ulcerative colitis, rectal administration of either SCFA mixtures(Reference Breuer, Buto and Christ59, Reference Breuer, Soergel and Lashner60) or butyrate alone(Reference Scheppach, Sommer and Kirchner61) has been shown to be effective at ameliorating the clinical symptoms of the disease. In intestinal epithelial cells, butyrate modulates colonic inflammation by reducing the expression of IL-8(Reference Huang, Katz and Martin62) and inhibiting inducible NO synthase expression(Reference Stempelj, Kedinger and Augenlicht63). Butyrate also alleviates oxidative stress and protects against oxidative DNA damage in cultured CRC cells and in colonic mucosal cells(Reference Rosignoli, Fabiani and De Bartolomeo64, Reference Sauer, Richter and Pool-Zobel65). This is supported further by reports of altered expression of proteins involved in free-radical scavenging and elevated activity of glutathione S-transferase, a protein responsible for metabolising potential carcinogens(Reference Fung, Lewanowitsch and Henderson30, Reference Ebert, Klinder and Peters66). Furthermore, incubation of HT29 cells with 4 mmol/l butyrate suppresses cyclo-oxygenase-2 expression and activity(Reference Tong, Yin and Giardina67). However, data derived from in vivo experiments where colorectal tumour tissue was exposed to butyrate have been inconclusive, possibly due to the size of the patient cohorts tested, tumour tissue heterogeneity and potential confounding by clinical parameters(Reference Jahns, Wilhelm and Jablonowski68). This is a critical factor in validating the relevance of data derived from model systems to human CRC and it is highly desirable to invest in such an effort.

Activation of NF-κB is one of the primary contributors to the development of inflammation-associated carcinogenesis, including CRC arising from chronic inflammatory conditions such as ulcerative colitis(Reference Greten, Eckmann and Greten69). Although butyrate regulates the activity of NF-κB, the signalling mechanisms involved in this process are not known. Constitutive activation of NF-κB has been reported in approximately 40 % of colorectal tumour tissues(Reference Sakamoto, Maeda and Hikiba70) and NF-κB activation in tumour cells promotes survival both by potentiating the inflammatory response through the activation of signalling pathways regulated by pro-inflammatory cytokines and by regulating the expression of anti-apoptotic genes(Reference Chen, Edelstein and Gelinas71, Reference Zong, Edelstein and Chen72).

Role of the SLC5A8 transporter in the inflammatory response and colorectal cancer

The Na-linked solute transporter SLC5A8 (solute carrier family 5, member 8) is silenced epigenetically in a number of different cancers(Reference Li, Myeroff and Smiraglia73). This transporter is expressed in the colon, kidney and thyroid and recognises (and transports) monocarboxylic acids, including butyrate. Methylation of the SLC5A8 gene and its subsequent loss of expression have been detected in 59 % of CRC adenoma and tumour tissues(Reference Li, Myeroff and Smiraglia73). Silencing of SLC5A8 in CRC has been associated with mutant and inactive adenomatous polyposis coli (APC) protein and aberrant Wnt signalling(Reference Thangaraju, Cresci and Itagaki74). In vitro studies have shown that butyrate transport by SLC5A8 inhibits HDAC activity and the growth of tumour cells(Reference Thangaraju, Cresci and Itagaki74, Reference Thangaraju, Gopal and Martin75). Thangaraju et al. (Reference Thangaraju, Cresci and Itagaki74) also reported that loss of SLC5A8 expression on CRC cell lines was inversely linked with apoptosis, and that apoptosis occurred as a result of inhibition of HDAC activity and activation of the caspase cascade by butyrate.

Involvement of SCFA receptors in the inflammatory response and colorectal cancer

SCFA augment the immune and inflammatory response by influencing immune cell functions such as chemotaxis, phagocytosis, reactive oxygen species production and cytokine/chemokine release. Butyrate reduces reactive oxygen species production and cytokine release in activated neutrophils (at approximately 1·6 mmol/l)(Reference Vinolo, Rodrigues and Hatanaka76) and plays a role in immune cell migration in vivo (Reference Bailon, Cueto-Sola and Utrilla77, Reference Vinolo, Rodrigues and Hatanaka78). Recently, production of acetate by bifidobacteria was found to improve the immune defence function of intestinal epithelial cells in vivo (Reference Fukuda, Toh and Hase79). These effects have been attributed, in part, to the activation of G-protein coupled receptors (GPR), in particular GPR109A and GPR43. The GPR40 family comprises receptors for SCFA and medium-chain fatty acids, with GPR41 and GPR43 having the highest affinity for SCFA ( < 5 carbons)(Reference Brown, Goldsworthy and Barnes80Reference Nilsson, Kotarsky and Owman82). Although these receptors display millimolar affinity for the SCFA, these concentrations are readily achievable in the colon. The role of GPR43 in inflammatory conditions has been studied widely using isolated immune cells and in mouse models of intestinal inflammation(Reference Le Poul, Loison and Struyf81, Reference Aoyama, Kotani and Usami83Reference Sina, Gavrilova and Forster85). In mouse models of both acute and chronic colitis induced by dextran sulphate sodium, absence of GPR43 expression resulted in greater colonic inflammation and compromised mucosal integrity when compared to wild-type littermates expressing the receptor(Reference Maslowski, Vieira and Ng84). Consumption of acetate in the drinking water resulted in improved inflammatory indices in wild-type mice but not in GPR43 knockout mice, indicating that activation of this receptor by SCFA plays a role in modulating the inflammatory response. Conversely, Sina et al. (Reference Sina, Gavrilova and Forster85) reported that loss of GPR43 expression reduced colonic inflammation in the chronic dextran sulphate sodium mouse model of colitis. These apparently contradictory results may be attributed to differences in the dose and duration of dextran sulphate sodium administration between these two studies. Nevertheless, both studies report that stimulation of GPR43 by SCFA (acetate(Reference Maslowski, Vieira and Ng84) and propionate and butyrate(Reference Sina, Gavrilova and Forster85)) is essential for immune cell recruitment and that this chemotactic response is mediated by the MAPK pathway.

GPR43 has been implicated in CRC prevention where it may have a tumour-suppressive role(Reference Tang, Chen and Jiang86). Loss of GPR43 expression occurs in colorectal adenocarcinoma tissue when compared to normal mucosa and reduced expression has been noted in colorectal hyperplasia and benign colorectal disease including polyps(Reference Tang, Chen and Jiang86). In a panel of nine CRC cell lines, Tang et al. (Reference Tang, Chen and Jiang86) determined that GPR43 was expressed in the HT29 CRC cell line only. They established also that re-expression of this receptor in CRC cells and activation by either butyrate (50% inhibitory concentration (IC50) 0·8 mmol/l) or propionate (IC50 2 mmol/l) inhibited proliferation and induced apoptosis and cell cycle arrest, providing further support for the role of GPR43 in maintaining normal cellular function. This effect of propionate is important, as it has been reported to induce some effects which are similar to butyrate, albeit at much higher concentrations. GPR43 expression has been studied in the MCF7 breast cancer cell line where its activation by each of the SCFA (at 10 mmol/l) induced a cell stress response mediated specifically by p38 MAPK signalling(Reference Yonezawa, Kobayashi and Obara47). These data indicate that GPR43 plays a pivotal role in activating the signalling pathways associated with the reported cellular and anti-tumorigenic effects of butyrate (and other SCFA). Despite this, GPR43-mediated cell signalling events in both the normal colon, colonic inflammatory conditions and in the development of CRC need to be determined.

GPR109A was initially identified as the receptor for nicotinic acid(Reference Soga, Kamohara and Takasaki87Reference Wise, Foord and Fraser89) and belongs to a receptor subfamily that includes GPR81 and GPR109B(Reference Ahmed, Tunaru and Offermanns90). Although GPR109A and GPR109B are similar structurally and have a similar expression pattern, they differ in their ligand specificity(Reference Wise, Foord and Fraser89, Reference Taggart, Kero and Gan91). GPR109A is activated by nicotinic acid, 3-hydroxybutyrate and butyrate, whereas GPR109B displays much low affinity for them. Butyrate binds and activates the GPR109A receptor (50% effective concentration (EC50) 1·6 mmol/l), but has no effect on GPR109B and neither receptor displays affinity for acetate or propionate(Reference Taggart, Kero and Gan91). GPR109A is expressed in the human colon and the expression of this receptor is dysregulated in CRC(Reference Thangaraju, Cresci and Liu92). A similar expression pattern was also determined in a mouse model of CRC and in a panel of human CRC cell lines. The authors determined also that silencing of GPR109A in CRC occurs as a result of DNA methylation. Subsequent re-expression of GPR109A in CRC cell lines and its activation by 1 mmol/l butyrate abolished NF-κB activation and induced apoptosis. Apoptosis occurred independently of HDAC inhibition, lending further support to the existence of alternate mechanisms involved in butyrate-induced apoptosis. Although GPR109A is also expressed on immune and inflammatory cells, its role in these cells has yet to be determined.

Cancer cell metabolism

Tumour cells display an altered cellular metabolism that can be viewed as an adaptive response to an hypoxic microenvironment or as a result of mutations in oncogenes and tumour-suppressor genes leading to higher glycolytic activity and enhanced energy production – a phenomenon known as the ‘Warburg effect’(Reference Warburg93). Although this characteristic of cancer cells has been studied widely, the mechanisms linking this switch in cell metabolism from a normal phenotype to cancer cell survival are uncertain. Transcription factors such as hypoxia inducible factor (HIF), tumour protein p53, octamer-binding protein 1 (OCT1), NF-KB and avian myelocytomatosis viral oncogene homolog (MYC) have been linked to dysregulated expression of nutrient transporters, glycolytic enzymes, and proteins involved in mitochondrial function(Reference Cairns, Harris and Mak94) and it is possible that butyrate may target these transcription factors to alter cell metabolism. In vitro studies and those involving isolated human colonocytes have indicated that butyrate modulates the levels and activity of transcription factors. In the case of c-myc, 5 mmol/l butyrate inhibits its transcription and protein expression in malignant cells, possibly via its ability to inhibit HDAC activity(Reference Emenaker and Basson95, Reference Wilson, Velcich and Arango96). In vitro studies also demonstrate that, while butyrate increases transcription of the HIF-1α gene, it represses the transcriptional regulatory activity of HIF-1α protein by inhibiting its translocation to the nucleus in Caco2 cells(Reference Pellizzaro, Coradini and Daidone97, Reference Zgouras, Wachtershauser and Frings98). These studies demonstrate further that inhibition of VEGF expression and angiogenesis occurs with butyrate incubation, but the effects on cellular metabolism were not investigated. Regulation of HIF-1α translocation by butyrate presents a potential mechanism contributing to its effects on angiogenesis.

Regulation of cancer cell metabolism by butyrate

Recent studies have shown that CRC cells, which are highly dependent on glycolysis, can acquire a unique ability to utilise both butyrate and glucose as their energy source and that this metabolic switch can be induced by the former(Reference Serpa, Caiado and Carvalho36). This capacity for both butyrate and glucose metabolism by colorectal tumour cells is supported by an elevated expression of solute transporters with high affinity for either substrate. In particular, monocarboxylate transporter 1 (MCT1, SLC16A1 (solute carrier family 16, member 1)) and GLUT type 1 (GLUT1, SLC2A1 (solute carrier family 2, member 1)) are elevated in CRC tissue when compared to adjacent normal mucosa(Reference Koukourakis, Giatromanolaki and Harris99, Reference Pinheiro, Longatto-Filho and Scapulatempo100). MCT1 is expressed widely in many different cell types and has been characterised as the primary butyrate transporter in the colon. It is expressed in healthy colon tissue and in many different cultured CRC cell lines(Reference Andriamihaja, Chaumontet and Tome101, Reference Tamai, Takanaga and Maeda102). Reports on the expression of MCT1 in CRC tissue have been conflicting with reported increases(Reference Koukourakis, Giatromanolaki and Harris99, Reference Pinheiro, Longatto-Filho and Scapulatempo100) or decreases(Reference Lambert, Wood and Ellis103) in expression. In addition to facilitating butyrate entry into the cell, MCT1 functions to remove lactate, a potentially cytotoxic metabolic by-product of glycolysis, indicating that it plays a dual role to enhance the survival of CRC cells(Reference Koukourakis, Giatromanolaki and Harris99). Butyrate also increases the expression of MCT1 on CRC cells in a dose-dependent manner and this is partially attributed to NF-κB activity(Reference Borthakur, Saksena and Gill104).

GLUT1 is expressed widely and its expression is elevated in cells from many different types of cancer. Its level of expression in tumours, including CRC, has been correlated with poor clinical outcomes(Reference Haber, Rathan and Weiser105). The mechanisms involved in the regulation of GLUT1 expression in cancer cells are not completely understood. Induction of its expression has been linked with an hypoxic microenvironment and regulation by HIF-1. However, there are alternative mechanisms(Reference Kim, Gao and Dang106). Using a panel of CRC cell lines, Yun et al. (Reference Yun, Rago and Cheong107) recently showed that increased GLUT1 expression is independent of HIF-1 activity and may instead be a downstream consequence of dysregulated signalling pathways caused by mutations in the KRAS or BRAF (v-raf murine sarcoma viral oncogene homolog B1) genes. It is not known if butyrate is able to regulate the expression of GLUT1 in CRC cells. Although these findings are contradictory, they can be reconciled if this altered metabolic profile is due to the presence of a butyrate-resistant sub-population of CRC cells. These cells would not succumb to apoptosis with butyrate exposure, but could proliferate to establish tumours. In the Caco2 CRC cell line, butyrate inhibits the pyruvate dehydrogenase complex, reducing the capacity for glycolysis and inducing a switch to a butyrate-oxidising phenotype providing a potential explanation for the development of a butyrate-resistant cell population(Reference Blouin, Penot and Collinet108). This is an established feature of the regulation of pyruvate dehydrogenase in a number of tissues. Glutamine metabolism was increased to favour the production of precursors for fatty acid synthesis, a process essential for cell proliferation. The group further demonstrated that this metabolic switch occurred as a result of HDAC inhibition by butyrate.

Recently, a link was established between HIF-1α and APC, further supporting the role of HIF-1α in the early stages of carcinogenesis(Reference Newton, Kenneth and Appleton109). In this study, HIF-1α was found to bind directly to the promoter region of APC, inhibiting its expression and (potentially) enhancing tumour cell survival(Reference Newton, Kenneth and Appleton109). This study also showed that depletion of wild-type APC protein, but not a mutant one, resulted in elevated HIF-1α activity. Mutations in APC are common to CRC, and thus up-regulation of HIF-1α in these cells may provide a further competitive advantage to tumour cells by altering their metabolism to promote adaptation to an hypoxic environment. Limitations in O2 supply could be an important factor in CRC development and also help to explain some of the unanswered questions in the aetiology of the disease. Cigarette smoking is an established risk factor for CRC and heavy use leads to the significant accumulation of erythrocyte carbonmonoxyhaemoglobin(Reference Williamson, Pols and Illman110). Blood carbonmonoxyhameoglobin leads to substantial changes in metabolic processes (e.g. ethanol metabolism(Reference Topping, Fishlock and Trimble111) and lipoprotein catabolism(Reference Gardner, Topping and Mayes112)) through decreased tissue O2 consumption. In contrast, greater SCFA production (through fermentation) leads to greater visceral perfusion and, hence, increased O2 supply although infusion studies suggest that the effect of butyrate is rather less than the other SCFA(Reference Topping and Clifton12).

Butyrate and butyrate analogues as chemotherapeutic agents

HDAC inhibitors are being investigated seriously as potential chemotherapeutic agents. Suberoylanilide hydroxamic acid was recently approved for the treatment of cutaneous T-cell lymphoma(Reference Mann, Johnson and Cohen113). Experiments with various cancer cell lines have been conducted to determine if combined treatment with DNA methyltransferase inhibitors (e.g. 5-aza-2′-deoxycytidine) and prototype HDAC inhibitors (e.g. trichostatin A, butyrate) could restore the function of key tumour-suppressor genes(Reference Fang, Chen and Lu114). Studies such as these are aimed primarily at determining the potential efficacy of combined treatments for cancer therapy. Butyrate itself is disqualified as a candidate drug on several counts. These include its ready uptake and metabolism by many cell types in the body, leading to rapid clearance and short half-life in the circulation. To overcome these limitations, many studies have explored the potential of butyrate derivatives, with 4-phenylbutyrate and tributyrin being the most promising(Reference Kang, Lee and Lee115Reference Miyoshi, Sakaki and Usami118). In vitro studies comparing the structural analogues of butyrate have shown that the apoptotic properties of butyrate are dependent on the lack of substitution at the 2- and 3-positions of the carboxylate backbone(Reference Ooi, Good and Williams119, Reference Ooi, Good and Williams120). Furthermore, these studies identified 4-benzoylbutyrate and 4-phenylbutyrate to be the most potent of the thirty-two analogues compared, indicating that a three-atom spacer between any bulky moiety and carboxyl group may also be essential for the anti-tumorigenic properties of any butyrate-based pharmaceutical. Although these studies did not identify analogues that were more potent than butyrate in vitro, they provide a starting point for the development of novel therapeutic agents.

Conclusion and future perspectives

CRC is a disease where there is clear potential for lowering the risk through dietary and lifestyle changes, with 30–60 % of tumours being preventable. The evidence available suggests that the consumption of diets high in fibre, in particular RS, and low in fats and proteins is protective against CRC development. The role of fibre fermentation is an area of growing significance in disease aetiology, with increasing attention being given to SCFA(Reference Cassidy, Bingham and Cummings121). It appears that SCFA mediate many of the effects previously ascribed to fibre alone. Butyrate is central to the proposed link between diet and protection against CRC. There appears to be a clear pathway that integrates, at a population level, the beneficial effects of a high-fibre diet (especially fermentable fibre), the formation of SCFA by colonic microbiota and demonstration of a role for the SCFA in maintaining physiological function in the colon including cell growth and regulation. Key to this hypothesis is the ability of butyrate to influence cellular processes to support a normal cell population and induce apoptosis and inhibit tumorigenesis (Fig. 2). While in vitro experiments indicate that HDAC inhibition is the primary mechanism for butyrate's anti-tumorigenic effects, the evidence for additional cellular pathways contributing to butyrate's pro-apoptotic and anti-proliferative activities in tumour cells is mounting. The current body of knowledge offers promise for containing CRC, but it is a clear priority to delineate these mechanisms in experimental systems and move on to human population and intervention studies to determine their true potential for risk reduction. Nutritional trials in free-living subjects have shown that it is possible to raise faecal SCFA (including butyrate) to levels found in low-risk populations such as native Africans consuming traditional foods(Reference Bird, Vuaran and King122, Reference McOrist, Miller and Bird123). Although these studies demonstrate that such interventions are feasible, it appears that some individuals do not show the expected rise in SCFA on increasing RS consumption(Reference McOrist, Miller and Bird123). This may reflect differences in the capacity of their large-bowel microbiome to ferment starch(Reference Cummings, Beatty and Kingman124). Clearly, it is imperative to show that any increase in colonic butyrate supply can be effective and sustained.

Fig. 2 Summary of the anti-tumorigenic effects of butyrate. HDAC, histone deacetylase; miRNA, micro-RNA; GPR43, G-protein coupled receptor 43; GPR109A, G-protein coupled receptor 109A; ROS, reactive oxygen species; COX2, cyclo-oxygenase-2; HIF-1α, hypoxia inducible factor.

Acknowledgements

The authors would like to thank Dr Leanne Purins and Dr Julie Clarke for helpful discussions. The present work was supported by the CSIRO Preventative Health and Food Futures National Research Flagships. The authors declare no conflicts of interest. K. Y. C. F., D. L. T. and R. H. wrote the manuscript. K. Y. C. F., L. C., T. L., R. H. and D. L. T. contributed to the ideas and participated in the editing. All authors read and approved the final manuscript.

References

1Ferlay, J, Shin, HR, Bray, F, et al. (2010) GLOBOCAN 2008 v1.2, Cancer Incidence and Mortality Worldwide: IARC CancerBase No. 10 [Internet]. Lyon: International Agency for Research on Cancer.Google Scholar
2Center, MM, Jemal, A, Smith, RA, et al. (2009) Worldwide variations in colorectal cancer. CA Cancer J Clin 59, 366378.CrossRefGoogle ScholarPubMed
3Parkin, DM, Bray, F, Ferlay, J, et al. (2005) Global cancer statistics, 2002. CA Cancer J Clin 55, 74108.CrossRefGoogle ScholarPubMed
4Haggar, FA & Boushey, RP (2009) Colorectal cancer epidemiology: incidence, mortality, survival, and risk factors. Clin Colon Rectal Surg 22, 191197.CrossRefGoogle ScholarPubMed
5Wong, MT & Eu, KW (2007) Rise of colorectal cancer in Singapore: an epidemiological review. ANZ J Surg 77, 446449.CrossRefGoogle ScholarPubMed
6World Cancer Research Fund & American Institute for Cancer Research (2007) Food, Nutrition, Physical Activity, and the Prevention of Cancer: A Global Perspective. Washington, DC: AICR.Google Scholar
7Platz, EA, Willett, WC, Colditz, GA, et al. (2000) Proportion of colon cancer risk that might be preventable in a cohort of middle-aged US men. Cancer Causes Control 11, 579588.CrossRefGoogle Scholar
8Bingham, SA, Day, NE, Luben, R, et al. (2003) Dietary fibre in food and protection against colorectal cancer in the European Prospective Investigation into Cancer and Nutrition (EPIC): an observational study. Lancet 361, 14961501.CrossRefGoogle ScholarPubMed
9Park, Y, Hunter, DJ, Spiegelman, D, et al. (2005) Dietary fiber intake and risk of colorectal cancer: a pooled analysis of prospective cohort studies. JAMA 294, 28492857.CrossRefGoogle ScholarPubMed
10McCullough, ML, Robertson, AS, Chao, A, et al. (2003) A prospective study of whole grains, fruits, vegetables and colon cancer risk. Cancer Causes Control 14, 959970.CrossRefGoogle ScholarPubMed
11Brouns, F, Kettlitz, B & Arrigoni, E (2002) Resistant starch and ‘the butyrate revolution’. Trends Food Sci Technol 13, 251261.CrossRefGoogle Scholar
12Topping, DL & Clifton, PM (2001) Short-chain fatty acids and human colonic function: roles of resistant starch and nonstarch polysaccharides. Physiol Rev 81, 10311064.CrossRefGoogle ScholarPubMed
13Bugaut, M (1987) Occurrence, absorption and metabolism of short chain fatty acids in the digestive tract of mammals. Comp Biochem Physiol B 86, 439472.CrossRefGoogle ScholarPubMed
14Freudenheim, JL, Graham, S, Horvath, PJ, et al. (1990) Risks associated with source of fiber and fiber components in cancer of the colon and rectum. Cancer Res 50, 32953300.Google ScholarPubMed
15Stephen, AM & Cummings, JH (1980) Mechanism of action of dietary fibre in the human colon. Nature 284, 283284.CrossRefGoogle ScholarPubMed
16Marlett, JA (1992) Content and composition of dietary fiber in 117 frequently consumed foods. J Am Diet Assoc 92, 175186.CrossRefGoogle ScholarPubMed
17Topping, D (2007) Cereal complex carbohydrates and their contribution to human health. J Cer Sci 46, 220229.CrossRefGoogle Scholar
18Ahmed, R, Segal, I & Hassan, H (2000) Fermentation of dietary starch in humans. Am J Gastroenterol 95, 10171020.CrossRefGoogle ScholarPubMed
19Cummings, JH (1984) Colonic absorption: the importance of short chain fatty acids in man. Scand J Gastroenterol Suppl 93, 8999.Google ScholarPubMed
20Perrin, P, Pierre, F, Patry, Y, et al. (2001) Only fibres promoting a stable butyrate producing colonic ecosystem decrease the rate of aberrant crypt foci in rats. Gut 48, 5361.CrossRefGoogle ScholarPubMed
21Clarke, JM, Topping, DL, Bird, AR, et al. (2008) Effects of high-amylose maize starch and butyrylated high-amylose maize starch on azoxymethane-induced intestinal cancer in rats. Carcinogenesis 29, 21902194.CrossRefGoogle ScholarPubMed
22Le Leu, RK, Hu, Y, Brown, IL, et al. (2009) Effect of high amylose maize starches on colonic fermentation and apoptotic response to DNA-damage in the colon of rats. Nutr Metab (Lond) 6, 11.CrossRefGoogle ScholarPubMed
23Toden, S, Bird, AR, Topping, DL, et al. (2007) Dose-dependent reduction of dietary protein-induced colonocyte DNA damage by resistant starch in rats correlates more highly with caecal butyrate than with other short chain fatty acids. Cancer Biol Ther 6, 253258.CrossRefGoogle ScholarPubMed
24Lewin, MH, Bailey, N, Bandaletova, T, et al. (2006) Red meat enhances the colonic formation of the DNA adduct O6-carboxymethyl guanine: implications for colorectal cancer risk. Cancer Res 66, 18591865.CrossRefGoogle ScholarPubMed
25Tan, SL, Gerber, JP, Cosgrove, LJ, et al. (2011) Is the tissue persistence of O(6)-methyl-2′-deoxyguanosine an indicator of tumour formation in the gastrointestinal tract? Mutat Res 721, 119126.CrossRefGoogle Scholar
26Kampman, E, Voskuil, DW, van Kraats, AA, et al. (2000) Animal products and K-ras codon 12 and 13 mutations in colon carcinomas. Carcinogenesis 21, 307309.CrossRefGoogle ScholarPubMed
27Slattery, ML, Curtin, K, Anderson, K, et al. (2000) Associations between dietary intake and Ki-ras mutations in colon tumors: a population-based study. Cancer Res 60, 69356941.Google ScholarPubMed
28Daly, K & Shirazi-Beechey, SP (2006) Microarray analysis of butyrate regulated genes in colonic epithelial cells. DNA Cell Biol 25, 4962.CrossRefGoogle ScholarPubMed
29Fung, KY, Brierley, GV, Henderson, ST, et al. (2011) Butyrate-induced apoptosis in HCT116 colorectal cancer cells includes induction of a cell stress response. J Proteome Res 10, 18601869.CrossRefGoogle ScholarPubMed
30Fung, KY, Lewanowitsch, T, Henderson, ST, et al. (2009) Proteomic analysis of butyrate effects and loss of butyrate sensitivity in HT29 colorectal cancer cells. J Proteome Res 8, 12201227.CrossRefGoogle ScholarPubMed
31Iacomino, G, Tecce, MF, Grimaldi, C, et al. (2001) Transcriptional response of a human colon adenocarcinoma cell line to sodium butyrate. Biochem Biophys Res Commun 285, 12801289.CrossRefGoogle ScholarPubMed
32Tan, HT, Tan, S, Lin, Q, et al. (2008) Quantitative and temporal proteome analysis of butyrate-treated colorectal cancer cells. Mol Cell Proteomics 7, 11741185.CrossRefGoogle ScholarPubMed
33Tan, S, Seow, TK, Liang, RC, et al. (2002) Proteome analysis of butyrate-treated human colon cancer cells (HT-29). Int J Cancer 98, 523531.CrossRefGoogle ScholarPubMed
34Olmo, N, Turnay, J, Perez-Ramos, P, et al. (2007) In vitro models for the study of the effect of butyrate on human colon adenocarcinoma cells. Toxicol In Vitro 21, 262270.CrossRefGoogle Scholar
35Lopez de Silanes, I, Olmo, N, Turnay, J, et al. (2004) Acquisition of resistance to butyrate enhances survival after stress and induces malignancy of human colon carcinoma cells. Cancer Res 64, 45934600.CrossRefGoogle ScholarPubMed
36Serpa, J, Caiado, F, Carvalho, T, et al. (2010) Butyrate-rich colonic microenvironment is a relevant selection factor for metabolically adapted tumor cells. J Biol Chem 285, 3921139223.CrossRefGoogle ScholarPubMed
37Kim, YH, Park, JW, Lee, JY, et al. (2004) Sodium butyrate sensitizes TRAIL-mediated apoptosis by induction of transcription from the DR5 gene promoter through Sp1 sites in colon cancer cells. Carcinogenesis 25, 18131820.CrossRefGoogle ScholarPubMed
38Pajak, B, Gajkowska, B & Orzechowski, A (2009) Sodium butyrate sensitizes human colon adenocarcinoma COLO 205 cells to both intrinsic and TNF-alpha-dependent extrinsic apoptosis. Apoptosis 14, 203217.CrossRefGoogle ScholarPubMed
39Shao, Y, Gao, Z, Marks, PA, et al. (2004) Apoptotic and autophagic cell death induced by histone deacetylase inhibitors. Proc Natl Acad Sci U S A 101, 1803018035.CrossRefGoogle ScholarPubMed
40Tang, Y, Chen, Y, Jiang, H, et al. (2011) The role of short-chain fatty acids in orchestrating two types of programmed cell death in colon cancer. Autophagy 7, 235237.CrossRefGoogle ScholarPubMed
41Wang, L, Luo, HS & Xia, H (2009) Sodium butyrate induces human colon carcinoma HT-29 cell apoptosis through a mitochondrial pathway. J Int Med Res 37, 803811.CrossRefGoogle ScholarPubMed
42Avivi-Green, C, Polak-Charcon, S, Madar, Z, et al. (2002) Different molecular events account for butyrate-induced apoptosis in two human colon cancer cell lines. J Nutr 132, 18121818.CrossRefGoogle ScholarPubMed
43Ruemmele, FM, Dionne, S, Qureshi, I, et al. (1999) Butyrate mediates Caco-2 cell apoptosis via up-regulation of pro-apoptotic BAK and inducing caspase-3 mediated cleavage of poly-(ADP-ribose) polymerase (PARP). Cell Death Differ 6, 729735.CrossRefGoogle ScholarPubMed
44Scott, DW, Longpre, JM & Loo, G (2008) Upregulation of GADD153 by butyrate: involvement of MAPK. DNA Cell Biol 27, 607614.CrossRefGoogle ScholarPubMed
45Tong, C, Yin, Z, Song, Z, et al. (2007) c-Jun NH2-terminal kinase 1 plays a critical role in intestinal homeostasis and tumor suppression. Am J Pathol 171, 297303.CrossRefGoogle Scholar
46Zhang, Y, Zhou, L, Bao, YL, et al. (2010) Butyrate induces cell apoptosis through activation of JNK MAP kinase pathway in human colon cancer RKO cells. Chem Biol Interact 185, 174181.CrossRefGoogle ScholarPubMed
47Yonezawa, T, Kobayashi, Y & Obara, Y (2007) Short-chain fatty acids induce acute phosphorylation of the p38 mitogen-activated protein kinase/heat shock protein 27 pathway via GPR43 in the MCF-7 human breast cancer cell line. Cell Signal 19, 185193.CrossRefGoogle ScholarPubMed
48Wilson, AJ, Chueh, AC, Togel, L, et al. (2010) Apoptotic sensitivity of colon cancer cells to histone deacetylase inhibitors is mediated by an Sp1/Sp3-activated transcriptional program involving immediate-early gene induction. Cancer Res 70, 609620.CrossRefGoogle ScholarPubMed
49Waby, JS, Chirakkal, H, Yu, C, et al. (2010) Sp1 acetylation is associated with loss of DNA binding at promoters associated with cell cycle arrest and cell death in a colon cell line. Mol Cancer 9, 275.CrossRefGoogle Scholar
50White, NR, Mulligan, P, King, PJ, et al. (2006) Sodium butyrate-mediated Sp3 acetylation represses human insulin-like growth factor binding protein-3 expression in intestinal epithelial cells. J Pediatr Gastroenterol Nutr 42, 134141.CrossRefGoogle ScholarPubMed
51Janson, W, Brandner, G & Siegel, J (1997) Butyrate modulates DNA-damage-induced p53 response by induction of p53-independent differentiation and apoptosis. Oncogene 15, 13951406.CrossRefGoogle ScholarPubMed
52Gope, R & Gope, ML (1993) Effect of sodium butyrate on the expression of retinoblastoma (RB1) and P53 gene and phosphorylation of retinoblastoma protein in human colon tumor cell line HT29. Cell Mol Biol (Noisy-le-grand) 39, 589597.Google ScholarPubMed
53Nakano, K, Mizuno, T, Sowa, Y, et al. (1997) Butyrate activates the WAF1/Cip1 gene promoter through Sp1 sites in a p53-negative human colon cancer cell line. J Biol Chem 272, 2219922206.CrossRefGoogle Scholar
54Yuan, ZM, Huang, Y, Ishiko, T, et al. (1999) Role for p300 in stabilization of p53 in the response to DNA damage. J Biol Chem 274, 18831886.CrossRefGoogle ScholarPubMed
55Terui, T, Murakami, K, Takimoto, R, et al. (2003) Induction of PIG3 and NOXA through acetylation of p53 at 320 and 373 lysine residues as a mechanism for apoptotic cell death by histone deacetylase inhibitors. Cancer Res 63, 89488954.Google ScholarPubMed
56Hu, S, Dong, TS, Dalal, SR, et al. (2011) The microbe-derived short chain fatty acid butyrate targets miRNA-dependent p21 gene expression in human colon cancer. PLoS One 6, e16221.CrossRefGoogle ScholarPubMed
57Zhang, H, Li, W, Nan, F, et al. (2011) MicroRNA expression profile of colon cancer stem-like cells in HT29 adenocarcinoma cell line. Biochem Biophys Res Commun 404, 273278.CrossRefGoogle ScholarPubMed
58Necela, BM, Carr, JM, Asmann, YW, et al. (2011) Differential expression of microRNAs in tumors from chronically inflamed or genetic (APC) models of colon cancer. PLoS One 6, e18501.CrossRefGoogle ScholarPubMed
59Breuer, RI, Buto, SK, Christ, ML, et al. (1991) Rectal irrigation with short-chain fatty acids for distal ulcerative colitis. Preliminary report. Dig Dis Sci 36, 185187.CrossRefGoogle ScholarPubMed
60Breuer, RI, Soergel, KH, Lashner, BA, et al. (1997) Short chain fatty acid rectal irrigation for left-sided ulcerative colitis: a randomised, placebo controlled trial. Gut 40, 485491.CrossRefGoogle ScholarPubMed
61Scheppach, W, Sommer, H, Kirchner, T, et al. (1992) Effect of butyrate enemas on the colonic mucosa in distal ulcerative colitis. Gastroenterology 103, 5156.CrossRefGoogle ScholarPubMed
62Huang, N, Katz, JP, Martin, DR, et al. (1997) Inhibition of IL-8 gene expression in Caco-2 cells by compounds which induce histone hyperacetylation. Cytokine 9, 2736.CrossRefGoogle ScholarPubMed
63Stempelj, M, Kedinger, M, Augenlicht, L, et al. (2007) Essential role of the JAK/STAT1 signaling pathway in the expression of inducible nitric-oxide synthase in intestinal epithelial cells and its regulation by butyrate. J Biol Chem 282, 97979804.CrossRefGoogle ScholarPubMed
64Rosignoli, P, Fabiani, R, De Bartolomeo, A, et al. (2001) Protective activity of butyrate on hydrogen peroxide-induced DNA damage in isolated human colonocytes and HT29 tumour cells. Carcinogenesis 22, 16751680.CrossRefGoogle ScholarPubMed
65Sauer, J, Richter, KK & Pool-Zobel, BL (2007) Physiological concentrations of butyrate favorably modulate genes of oxidative and metabolic stress in primary human colon cells. J Nutr Biochem 18, 736745.CrossRefGoogle ScholarPubMed
66Ebert, MN, Klinder, A, Peters, WH, et al. (2003) Expression of glutathione S-transferases (GSTs) in human colon cells and inducibility of GSTM2 by butyrate. Carcinogenesis 24, 16371644.CrossRefGoogle ScholarPubMed
67Tong, X, Yin, L & Giardina, C (2004) Butyrate suppresses Cox-2 activation in colon cancer cells through HDAC inhibition. Biochem Biophys Res Commun 317, 463471.CrossRefGoogle ScholarPubMed
68Jahns, F, Wilhelm, A, Jablonowski, N, et al. (2011) Butyrate suppresses mRNA increase of osteopontin and cyclooxygenase-2 in human colon tumor tissue. Carcinogenesis 32, 913920.CrossRefGoogle ScholarPubMed
69Greten, FR, Eckmann, L, Greten, TF, et al. (2004) IKKbeta links inflammation and tumorigenesis in a mouse model of colitis-associated cancer. Cell 118, 285296.CrossRefGoogle Scholar
70Sakamoto, K, Maeda, S, Hikiba, Y, et al. (2009) Constitutive NF-kappaB activation in colorectal carcinoma plays a key role in angiogenesis, promoting tumor growth. Clin Cancer Res 15, 22482258.CrossRefGoogle Scholar
71Chen, C, Edelstein, LC & Gelinas, C (2000) The Rel/NF-kappaB family directly activates expression of the apoptosis inhibitor Bcl-x(L). Mol Cell Biol 20, 26872695.CrossRefGoogle ScholarPubMed
72Zong, WX, Edelstein, LC, Chen, C, et al. (1999) The prosurvival Bcl-2 homolog Bfl-1/A1 is a direct transcriptional target of NF-kappaB that blocks TNFalpha-induced apoptosis. Genes Dev 13, 382387.CrossRefGoogle ScholarPubMed
73Li, H, Myeroff, L, Smiraglia, D, et al. (2003) 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 100, 84128417.CrossRefGoogle ScholarPubMed
74Thangaraju, M, Cresci, G, Itagaki, S, et al. (2008) Sodium-coupled transport of the short chain fatty acid butyrate by SLC5A8 and its relevance to colon cancer. J Gastrointest Surg 12, 17731781, discussion 1781–1772.CrossRefGoogle ScholarPubMed
75Thangaraju, M, Gopal, E, Martin, PM, et al. (2006) SLC5A8 triggers tumor cell apoptosis through pyruvate-dependent inhibition of histone deacetylases. Cancer Res 66, 1156011564.CrossRefGoogle ScholarPubMed
76Vinolo, MA, Rodrigues, HG, Hatanaka, E, et al. (2011) Suppressive effect of short-chain fatty acids on production of proinflammatory mediators by neutrophils. J Nutr Biochem 22, 849855.CrossRefGoogle ScholarPubMed
77Bailon, E, Cueto-Sola, M, Utrilla, P, et al. (2010) Butyrate in vitro immune-modulatory effects might be mediated through a proliferation-related induction of apoptosis. Immunobiology 215, 863873.CrossRefGoogle ScholarPubMed
78Vinolo, MA, Rodrigues, HG, Hatanaka, E, et al. (2009) Short-chain fatty acids stimulate the migration of neutrophils to inflammatory sites. Clin Sci (Lond) 117, 331338.CrossRefGoogle ScholarPubMed
79Fukuda, S, Toh, H, Hase, K, et al. (2011) Bifidobacteria can protect from enteropathogenic infection through production of acetate. Nature 469, 543547.CrossRefGoogle ScholarPubMed
80Brown, AJ, Goldsworthy, SM, Barnes, AA, et al. (2003) The orphan G protein-coupled receptors GPR41 and GPR43 are activated by propionate and other short chain carboxylic acids. J Biol Chem 278, 1131211319.CrossRefGoogle ScholarPubMed
81Le Poul, E, Loison, C, Struyf, S, et al. (2003) Functional characterization of human receptors for short chain fatty acids and their role in polymorphonuclear cell activation. J Biol Chem 278, 2548125489.CrossRefGoogle ScholarPubMed
82Nilsson, NE, Kotarsky, K, Owman, C, et al. (2003) Identification of a free fatty acid receptor, FFA2R, expressed on leukocytes and activated by short-chain fatty acids. Biochem Biophys Res Commun 303, 10471052.CrossRefGoogle ScholarPubMed
83Aoyama, M, Kotani, J & Usami, M (2010) Butyrate and propionate induced activated or non-activated neutrophil apoptosis via HDAC inhibitor activity but without activating GPR-41/GPR-43 pathways. Nutrition 26, 653661.CrossRefGoogle ScholarPubMed
84Maslowski, KM, Vieira, AT, Ng, A, et al. (2009) Regulation of inflammatory responses by gut microbiota and chemoattractant receptor GPR43. Nature 461, 12821286.CrossRefGoogle ScholarPubMed
85Sina, C, Gavrilova, O, Forster, M, et al. (2009) G protein-coupled receptor 43 is essential for neutrophil recruitment during intestinal inflammation. J Immunol 183, 75147522.CrossRefGoogle ScholarPubMed
86Tang, Y, Chen, Y, Jiang, H, et al. (2011) G-protein-coupled receptor for short-chain fatty acids suppresses colon cancer. Int J Cancer 128, 847856.CrossRefGoogle ScholarPubMed
87Soga, T, Kamohara, M, Takasaki, J, et al. (2003) Molecular identification of nicotinic acid receptor. Biochem Biophys Res Commun 303, 364369.CrossRefGoogle ScholarPubMed
88Tunaru, S, Kero, J, Schaub, A, et al. (2003) PUMA-G and HM74 are receptors for nicotinic acid and mediate its anti-lipolytic effect. Nat Med 9, 352355.CrossRefGoogle ScholarPubMed
89Wise, A, Foord, SM, Fraser, NJ, et al. (2003) Molecular identification of high and low affinity receptors for nicotinic acid. J Biol Chem 278, 98699874.CrossRefGoogle ScholarPubMed
90Ahmed, K, Tunaru, S & Offermanns, S (2009) GPR109A, GPR109B and GPR81, a family of hydroxy-carboxylic acid receptors. Trends Pharmacol Sci 30, 557562.CrossRefGoogle ScholarPubMed
91Taggart, AK, Kero, J, Gan, X, et al. (2005) (d)-Beta-hydroxybutyrate inhibits adipocyte lipolysis via the nicotinic acid receptor PUMA-G. J Biol Chem 280, 2664926652.CrossRefGoogle ScholarPubMed
92Thangaraju, M, Cresci, GA, Liu, K, et al. (2009) GPR109A is a G-protein-coupled receptor for the bacterial fermentation product butyrate and functions as a tumor suppressor in colon. Cancer Res 69, 28262832.CrossRefGoogle ScholarPubMed
93Warburg, O (1956) On the origin of cancer cells. Science 123, 309314.CrossRefGoogle ScholarPubMed
94Cairns, RA, Harris, IS & Mak, TW (2011) Regulation of cancer cell metabolism. Nat Rev Cancer 11, 8595.CrossRefGoogle ScholarPubMed
95Emenaker, NJ & Basson, MD (2001) Short chain fatty acids differentially modulate cellular phenotype and c-myc protein levels in primary human nonmalignant and malignant colonocytes. Dig Dis Sci 46, 96105.CrossRefGoogle ScholarPubMed
96Wilson, AJ, Velcich, A, Arango, D, et al. (2002) Novel detection and differential utilization of a c-myc transcriptional block in colon cancer chemoprevention. Cancer Res 62, 60066010.Google ScholarPubMed
97Pellizzaro, C, Coradini, D & Daidone, MG (2002) Modulation of angiogenesis-related proteins synthesis by sodium butyrate in colon cancer cell line HT29. Carcinogenesis 23, 735740.CrossRefGoogle ScholarPubMed
98Zgouras, D, Wachtershauser, A, Frings, D, et al. (2003) Butyrate impairs intestinal tumor cell-induced angiogenesis by inhibiting HIF-1alpha nuclear translocation. Biochem Biophys Res Commun 300, 832838.CrossRefGoogle ScholarPubMed
99Koukourakis, MI, Giatromanolaki, A, Harris, AL, et al. (2006) Comparison of metabolic pathways between cancer cells and stromal cells in colorectal carcinomas: a metabolic survival role for tumor-associated stroma. Cancer Res 66, 632637.CrossRefGoogle ScholarPubMed
100Pinheiro, C, Longatto-Filho, A, Scapulatempo, C, et al. (2008) Increased expression of monocarboxylate transporters 1, 2, and 4 in colorectal carcinomas. Virchows Arch 452, 139146.CrossRefGoogle Scholar
101Andriamihaja, M, Chaumontet, C, Tome, D, et al. (2009) Butyrate metabolism in human colon carcinoma cells: implications concerning its growth-inhibitory effect. J Cell Physiol 218, 5865.CrossRefGoogle ScholarPubMed
102Tamai, I, Takanaga, H, Maeda, H, et al. (1995) Participation of a proton-cotransporter, MCT1, in the intestinal transport of monocarboxylic acids. Biochem Biophys Res Commun 214, 482489.CrossRefGoogle ScholarPubMed
103Lambert, DW, Wood, IS, Ellis, A, et al. (2002) Molecular changes in the expression of human colonic nutrient transporters during the transition from normality to malignancy. Br J Cancer 86, 12621269.CrossRefGoogle ScholarPubMed
104Borthakur, A, Saksena, S, Gill, RK, et al. (2008) Regulation of monocarboxylate transporter 1 (MCT1) promoter by butyrate in human intestinal epithelial cells: involvement of NF-kappaB pathway. J Cell Biochem 103, 14521463.CrossRefGoogle ScholarPubMed
105Haber, RS, Rathan, A, Weiser, KR, et al. (1998) GLUT1 glucose transporter expression in colorectal carcinoma: a marker for poor prognosis. Cancer 83, 3440.3.0.CO;2-E>CrossRefGoogle ScholarPubMed
106Kim, JW, Gao, P & Dang, CV (2007) Effects of hypoxia on tumor metabolism. Cancer Metastasis Rev 26, 291298.CrossRefGoogle ScholarPubMed
107Yun, J, Rago, C, Cheong, I, et al. (2009) Glucose deprivation contributes to the development of KRAS pathway mutations in tumor cells. Science 325, 15551559.CrossRefGoogle Scholar
108Blouin, JM, Penot, G, Collinet, M, et al. (2011) Butyrate elicits a metabolic switch in human colon cancer cells by targeting the pyruvate dehydrogenase complex. Int J Cancer 128, 25912601.CrossRefGoogle ScholarPubMed
109Newton, IP, Kenneth, NS, Appleton, PL, et al. (2010) Adenomatous polyposis coli and hypoxia-inducible factor-1{alpha} have an antagonistic connection. Mol Biol Cell 21, 36303638.CrossRefGoogle ScholarPubMed
110Williamson, PA, Pols, RG, Illman, RJ, et al. (1987) Blood carbonmonoxyhaemoglobin levels are chronically elevated in alcoholics treated for detoxification. Atherosclerosis 67, 245250.CrossRefGoogle ScholarPubMed
111Topping, DL, Fishlock, RC, Trimble, RP, et al. (1981) Carboxyhaemoglobin inhibits the metabolism of ethanol by perfused rat liver. Biochem Int 3, 157163.Google Scholar
112Gardner, RS, Topping, DL & Mayes, PA (1978) Immediate effects of carbon monoxide on the metabolism of chylomicron remnants by perfused rat liver. Biochem Biophys Res Commun 82, 526531.CrossRefGoogle Scholar
113Mann, BS, Johnson, JR, Cohen, MH, et al. (2007) FDA approval summary: vorinostat for treatment of advanced primary cutaneous T-cell lymphoma. Oncologist 12, 12471252.CrossRefGoogle ScholarPubMed
114Fang, JY, Chen, YX, Lu, J, et al. (2004) Epigenetic modification regulates both expression of tumor-associated genes and cell cycle progressing in human colon cancer cell lines: Colo-320 and SW1116. Cell Res 14, 217226.CrossRefGoogle ScholarPubMed
115Kang, SN, Lee, E, Lee, MK, et al. (2011) Preparation and evaluation of tributyrin emulsion as a potent anti-cancer agent against melanoma. Drug Deliv 18, 143149.CrossRefGoogle ScholarPubMed
116Kuroiwa-Trzmielina, J, de Conti, A, Scolastici, C, et al. (2009) Chemoprevention of rat hepatocarcinogenesis with histone deacetylase inhibitors: efficacy of tributyrin, a butyric acid prodrug. Int J Cancer 124, 25202527.CrossRefGoogle ScholarPubMed
117Li, Y, Le Maux, S, Xiao, H, et al. (2009) Emulsion-based delivery systems for tributyrin, a potential colon cancer preventative agent. J Agric Food Chem 57, 92439249.CrossRefGoogle ScholarPubMed
118Miyoshi, M, Sakaki, H, Usami, M, et al. (2011) Oral administration of tributyrin increases concentration of butyrate in the portal vein and prevents lipopolysaccharide-induced liver injury in rats. Clin Nutr 30, 252258.CrossRefGoogle ScholarPubMed
119Ooi, CC, Good, NM, Williams, DB, et al. (2010) Structure–activity relationship of butyrate analogues on apoptosis, proliferation and histone deacetylase activity in HCT-116 human colorectal cancer cells. Clin Exp Pharmacol Physiol 37, 905911.CrossRefGoogle ScholarPubMed
120Ooi, CC, Good, NM, Williams, DB, et al. (2010) Efficacy of butyrate analogues in HT-29 cancer cells. Clin Exp Pharmacol Physiol 37, 482489.CrossRefGoogle ScholarPubMed
121Cassidy, A, Bingham, SA & Cummings, JH (1994) Starch intake and colorectal cancer risk: an international comparison. Br J Cancer 69, 937942.CrossRefGoogle ScholarPubMed
122Bird, AR, Vuaran, MS, King, RA, et al. (2008) Wholegrain foods made from a novel high-amylose barley variety (Himalaya 292) improve indices of bowel health in human subjects. Br J Nutr 99, 10321040.CrossRefGoogle ScholarPubMed
123McOrist, AL, Miller, RB, Bird, AR, et al. (2011) Fecal butyrate levels vary widely among individuals but are usually increased by a diet high in resistant starch. J Nutr 141, 883889.CrossRefGoogle ScholarPubMed
124Cummings, JH, Beatty, ER, Kingman, SM, et al. (1996) Digestion and physiological properties of resistant starch in the human large bowel. Br J Nutr 75, 733747.CrossRefGoogle ScholarPubMed
Figure 0

Fig. 1 The effects of butyrate in the normal colon and in colorectal tumour cells. HDAC, histone deacetylase.

Figure 1

Table 1 Summary of the genes and proteins involved in the anti-tumorigenic effects of butyrate

Figure 2

Fig. 2 Summary of the anti-tumorigenic effects of butyrate. HDAC, histone deacetylase; miRNA, micro-RNA; GPR43, G-protein coupled receptor 43; GPR109A, G-protein coupled receptor 109A; ROS, reactive oxygen species; COX2, cyclo-oxygenase-2; HIF-1α, hypoxia inducible factor.