Hostname: page-component-8448b6f56d-wq2xx Total loading time: 0 Render date: 2024-04-25T04:49:08.373Z Has data issue: false hasContentIssue false

Is resistant starch protective against colorectal cancer via modulation of the WNT signalling pathway?

Published online by Cambridge University Press:  20 February 2015

Fiona C. Malcomson*
Affiliation:
Human Nutrition Research Centre, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne NE4 5PL, UK
Naomi D. Willis
Affiliation:
Human Nutrition Research Centre, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne NE4 5PL, UK
John C. Mathers
Affiliation:
Human Nutrition Research Centre, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne NE4 5PL, UK
*
*Corresponding author: F. Malcomson, email f.c.malcomson@newcastle.ac.uk
Rights & Permissions [Opens in a new window]

Abstract

Epidemiological and experimental evidence suggests that non-digestible carbohydrates (NDC) including resistant starch are protective against colorectal cancer. These anti-neoplastic effects are presumed to result from the production of the SCFA, butyrate, by colonic fermentation, which binds to the G-protein-coupled receptor GPR43 to regulate inflammation and other cancer-related processes. The WNT pathway is central to the maintenance of homeostasis within the large bowel through regulation of processes such as cell proliferation and migration and is frequently aberrantly hyperactivated in colorectal cancers. Abnormal WNT signalling can lead to irregular crypt cell proliferation that favours a hyperproliferative state. Butyrate has been shown to modulate the WNT pathway positively, affecting functional outcomes such as apoptosis and proliferation. Butyrate's ability to regulate gene expression results from epigenetic mechanisms, including its role as a histone deacetylase inhibitor and through modulating DNA methylation and the expression of microRNA. We conclude that genetic and epigenetic modulation of the WNT signalling pathway may be an important mechanism through which butyrate from fermentation of resistant starch and other NDC exert their chemoprotective effects.

Type
Conference on ‘Carbohydrates in health: friends or foes’
Copyright
Copyright © The Authors 2015 

Colorectal cancer (CRC) is the fourth most common cancer in the UK, and incidence has risen by 6 % in the past 10 years( 1 ). This cancer arises in colonocytes, the single layer of columnar epithelial cells which line the large bowel. In common with all cancer types, CRC develops because of genomic damage which gives the neoplastic cell new, and more competitive, characteristics( Reference Hanahan and Weinberg 2 ) enabling sustained proliferative signalling, evasion of growth suppressors, resistance to cell death, replicative immortality, the ability to induce angiogenesis and eventually, the potential for metastasis. While genetic susceptibility contributes to CRC risk, most CRC develop because of genomic damage caused by exogenous and endogenous processes and events. It has been estimated that over half of the CRC in the UK in 2010 resulted from environmental factors( Reference Parkin, Boyd and Walker 3 ) such as diet and physical activity and this provides support for the hypothesis that a high proportion of CRC cases could be prevented through appropriate dietary and lifestyle choices or the development of suitable interventions. The World Cancer Research Fund /American Institute for Cancer Research Colorectal Cancer 2011 Report( 4 ) concluded that there is convincing evidence for increased risk of CRC with the consumption of red meat, processed meat and alcohol (in men) and also with increasing body and abdominal fatness. In addition, there is convincing evidence that more physical activity and the consumption of foods containing dietary fibre decreases CRC risk.

Dietary fibre and the modulation of CRC risk

Dietary fibre has been defined as carbohydrate polymers with ten or more monomeric units which are not hydrolysed by the endogenous enzymes in the small intestine of human subjects( 5 ). It encompasses non-digestible carbohydrates (NDC) such as resistant starch (RS) and polydextrose. RS is the portion of dietary starch that avoids enzymatic digestion and absorption in the small intestine and consequently reaches the large bowel undigested( Reference Cummings and Stephen 6 ). Here, it is the target for fermentation by resident colonic bacteria to produce SCFA such as acetate, propionate and butyrate( Reference Topping and Clifton 7 ). Four types of RS have been described: type 1 physically inaccessible RS, e.g. that are found in seeds; type 2 resistant granules, e.g. in uncooked potato or unripe banana; type 3 retrograded RS, found in cooked and cooled foods such as bread; type 4 chemically modified RS which is used widely for technological purposes in processed foods( Reference Topping and Clifton 7 ).

Based on clinical observations in Africa, Burkitt was one of the first to report an association between higher consumption of dietary fibre and reduced CRC incidence( Reference Burkitt 8 ). Since then, many epidemiological studies have quantified links between dietary fibre intake and CRC risk. A recent meta-analysis of the results from twenty-five prospective studies confirmed an inverse correlation between dietary fibre intake and CRC risk( Reference Aune, Chan and Lau 9 ). The authors concluded that CRC risk is 10 % lower for every 10 g/d increase in dietary fibre intake.

Early attempts to explain the protective effects of dietary fibre focused on the increased faecal bulk, reduced transit time and dilution of faecal content which were expected to reduce exposure of the large bowel to carcinogens( Reference Hylla, Gostner and Dusel 10 , Reference Lipkin, Reddy and Newmark 11 ). More recently, the focus has shifted to the beneficial effects of SCFA, in particular butyrate, which are produced by colonic fermentation of NDC( Reference Williams, Coxhead and Mathers 12 ). Butyrate is produced predominantly by bacterial species of the Clostridia, Roseburia and Eubacteria genera( Reference Pryde, Duncan and Hold 13 ). Butyrate is the preferred energy source for colonocytes and plays an important role within the large bowel mucosa where it regulates key cellular processes including proliferation, differentiation and apoptosis that contribute to the maintenance of homeostasis( Reference Hamer, Jonkers and Venema 14 ). Furthermore, butyrate has been reported to positively modulate WNT signalling( Reference Bordonaro, Lazarova and Sartorelli 15 ), a pathway that is frequently aberrantly activated in CRC, suggesting that this may be one of the mechanisms through which butyrate, and other NDC, are protective against CRC.

The WNT signalling pathway

WNT signalling comprises three different pathways: the canonical or β-catenin pathway, the non-canonical planar cell polarity pathway and the WNT/calcium pathway. The principal pathway, that is most relevant in human health and disease, is the canonical pathway. The canonical WNT signalling pathway is observed in several tissue and cell types( Reference Clevers 16 ), including colonic crypts, where it regulates cell proliferation, migration and differentiation( Reference Bienz and Clevers 17 ).

In the inactive state, or when activation of the pathway is inhibited by antagonists such as secreted frizzled-related proteins (SFRP), the β-catenin destruction complex, comprising adenomatous polyposis coli, AXIN, glycogen synthase kinase 3β and casein kinase 1, phosphorylates β-catenin (Fig. 1a)( Reference Bienz and Clevers 17 , Reference Schneikert and Behrens 18 ). Phosphorylated β-catenin is recognised by β-transducin repeat-containing protein which targets β-catenin for ubiquitination by the E3 ubiquitin ligase complex and, consequently, proteasomal degradation( Reference Clevers 16 , Reference Schneikert and Behrens 18 ). With low cytoplasmic concentrations of β-catenin, there is no translocation to the nucleus, transcription is not activated and the T cell factor and lymphoid enhancer factor-1 transcription factors are bound to, and inhibited by, Groucho and transducin-like enhancer of split. Activation of the canonical pathway occurs when a WNT ligand binds to a frizzled receptor, resulting in an interaction with co-receptors of the LDL receptor-related protein family (Fig. 1b). The β-catenin destruction complex is inactivated following inhibition of AXIN by dishevelled. Consequently, cytoplasmic levels of β-catenin rise and the protein is translocated to the nucleus where it binds to, and activates, T cell factor/lymphoid enhancer factor-1 transcription factors( Reference Sierra, Yoshida and Joazeiro 19 ), which switch on transcription of target genes such as c-MYC and c-JUN.

Fig. 1. The canonical WNT signalling pathway. (a) In the inactive state, when there are no WNT ligands bound to frizzled receptors or when WNT antagonists, such as WIF1, inhibit activation of the WNT pathway, the β-catenin destruction complex (adenomatous polyposis coli (APC), AXIN, casein kinase (CK1) and glycogen synthase kinase 3β (GSK3β)) binds to and phosphorylates β-catenin. This targets β-catenin for ubiquitination by β-transducin 117 repeat-containing protein (β-TrCP) and, consequently, proteasomal degradation. In the nucleus, lymphoid enhancer factor-1 (LEF) and T cell factor (TCF) transcription factors are repressed by Groucho. (b) When the WNT pathway is activated by binding of WNT ligands to frizzled receptors, the β-catenin destruction complex is inhibited by dishevelled (Dvl). Levels of dephosphorylated, active β-catenin rise and β-catenin translocates to the nucleus where it binds to LEF and TCF transcription factors to activate transcription of target genes such as c-MYC and c-JUN.

The WNT signalling pathway is also implicated in several cancers including CRC( Reference Luo, Chen and Deng 20 ), where it is constitutively activated in approximately 90 % of sporadic cases( Reference Klaus and Birchmeier 21 ). Aberrant expression of the WNT pathway results mainly from the loss of the APC tumour suppressor gene( Reference Kinzler and Vogelstein 22 ). However, genetic and epigenetic mechanisms also contribute to altered expression of other components of the pathway e.g. activating mutations in β-catenin( Reference Morin, Sparks and Korinek 23 ) and down-regulation of SFRP1 due to promoter hypermethylation( Reference Caldwell, Jones and Gensberg 24 ) and, therefore, to tumour development.

The effects of resistant starch on colorectal carcinogenesis

Studies investigating the effects of RS on colorectal carcinogenesis have yielded inconsistent results. While some studies have indicated a chemoprotective effect of RS, others have reported no effect and some have observed adverse effects.

In rats treated with 1,2-dimethylhydrazine dihydrochloride (DMH; a carcinogen used widely in experimental studies of CRC formation), incidence of tumours was reduced when the animals were fed Novelose 330 (a type 3 RS) compared with those fed a standard diet( Reference Bauer-Marinovic, Florian and Muller-Schmehl 25 ). High-amylose maize starch (HAMS; a type 2 RS) has also been shown to reduce incidence and the number of adenocarcinomas in rats when the HAMS diet was provided for 4 weeks prior to injection with the carcinogen azoxymethane( Reference Le Leu, Brown and Hu 26 ).

The formation of aberrant crypt foci (ACF), which are the earliest detectable pre-neoplastic lesions in CRC( Reference Alrawi, Schiff and Carroll 27 ), is a marker of early-stage colorectal carcinogenesis and has been used as a surrogate endpoint in several studies investigating colorectal carcinogenesis in animal models. Using an azoxymethane-induced model of ACF, rats given raw potato starch (a type 2 RS) for 3 weeks post carcinogen treatment had significantly reduced ACF formation compared with controls( Reference Liu and Xu 28 ). In contrast, rats fed moderate and high doses of RS prior to injection with azoxymethane had a significantly greater number of ACF compared with those on the control diet and those on the low dose of RS. These results suggest that RS may be protective if given during the promotion stage of colorectal carcinogenesis i.e. following carcinogen treatment, but may have adverse effects at the pre-initiation stage( Reference Liu and Xu 28 ). Consistent with findings from this study, two earlier animal studies reported protective effects of RS given during the promotion stage( Reference Thorup, Meyer and Kristiansen 29 , Reference Cassand, Maziere and Champ 30 ). Thorup et al. ( Reference Thorup, Meyer and Kristiansen 29 ) reported that there were significantly fewer ACF in azoxymethane-treated rats fed a potato starch-containing (RS) diet compared with those fed control or corn starch-containing diets. Similarly, rats fed RS (with or without a vitamin A supplement) for 12 weeks post-DMH injection had significantly reduced ACF formation( Reference Cassand, Maziere and Champ 30 ).

Some studies have also reported no effect of RS on CRC in animal models. Although DMH-treated rats fed RS (3 or 10 %) had significantly increased butyrate concentrations compared with those fed control diets, there were no effects of RS on the development of CRC( Reference Sakamoto, Nakaji and Sugawara 31 ). Similarly, there was no effect on ACF formation in rats fed HAMS for 4 weeks prior to DMH injection( Reference Maziere, Meflah and Tavan 32 ).

In contrast, adverse effects of RS in promoting colorectal carcinogenesis have also been reported. Increased ACF density and colonic tumour formation were observed by Young et al. in DMH-injected rats fed potato starch for 31 weeks( Reference Young, McIntyre and Albert 33 ). It has been argued that carcinogen-treatment in animals may not be the best model for studies of human colorectal carcinogenesis and that genetic models which recapitulate aberrant WNT signalling because of Apc mutation may be more appropriate. Using the Apc1638N mouse model of sporadic CRC( Reference Smits, Kartheuser and Jagmohan-Changur 34 ), we observed that feeding RS (a 1:1 blend of raw potato starch and Hylon VII, both type 2 RS) for 5 months increased tumour formation within the small intestine( Reference Williamson, Kartheuser and Coaker 35 ). However, this adverse effect was prevented with the synergistic administration of aspirin, which suggests that reducing inflammation may reduce CRC risk.

The effects of resistant starch and butyrate on the expression of WNT pathway components

The effects of RS on the expression of WNT pathway components have been investigated in only one published study in which carcinogen-treated rats were fed diets containing three corn maize varieties: an Argentinian strain, a Guatemalan strain and a hybrid of the two( Reference Cray 36 ). Those animals fed the Argentinian strain, which yielded the lowest RS content, had enhanced β-catenin expression compared with those rats fed the hybrid diet. In addition, expression of the WNT antagonist, SFRP4, was significantly lower in rats fed the diets (Guatemalan strain and hybrid) with the highest RS content. Altogether, these results suggest that RS reduced WNT pathway activity in carcinogen-treated rats. However, this study also found that expression of two other negative regulators of WNT signalling, AXIN2 and WISP1, was significantly reduced following the consumption of the two higher RS diets, which would be indicative of reduced WNT pathway inhibition with RS. These observations underline the complexity of regulation of WNT signalling and the difficulty in drawing unequivocal conclusions.

In contrast with the paucity of studies with RS and WNT signalling, there are several studies, notably from the Bordonaro group, which have investigated the effects of butyrate on the expression of WNT pathway components. These investigators observed that treatment of SW620 colon carcinoma cells with 5 mm butyrate (a physiological concentration) induced apoptosis and that this was associated with increased activity of the T cell factor transcription factor indicating that the apoptotic effect may have been mediated through effects on WNT signalling( Reference Bordonaro, Mariadason and Aslam 37 ). Furthermore, butyrate treatment increased formation of β-catenin–T cell factor complexes, which would activate transcription of target genes. A later study by the same group found that butyrate increased the levels of unphosphorylated, and therefore active, β-catenin following butyrate treatment in eight CRC cell lines( Reference Bordonaro, Lazarova and Sartorelli 38 ).

Other studies have investigated effects of butyrate on another functional outcome of the WNT pathway i.e. differentiation. There is an evidence that butyrate treatment of LIM2537 colon cancer cells induces WNT signalling and that this is paralleled by greater differentiation of the, usually, poorly differentiated tumour cells( Reference Vincan, Leet and Reyes 39 ). The activity of glycogen synthase kinase 3β, a member of the β-catenin destruction complex, was significantly reduced in the butyrate-treated cells and, indeed, this led to stabilised pools of β-catenin within the cytoplasm which would be expected to increase WNT activity. There was an inverse correlation between glycogen synthase kinase 3β expression and differentiation, suggesting that the induction of differentiation by butyrate resulted from an increase in WNT signalling. Unexpectedly, this was not paralleled by an increase in the expression of two target genes of the WNT pathway, c-MYC and CCND1.

Germann et al. observed that 4·5 mm butyrate treatment of CC531 rat colon carcinoma cells resulted in positive modulation of expression of four WNT pathway target genes (CCND1, c-MYC, FOSL1 and FST), which have been reported to be up-regulated in CRC( Reference Wong and Pignatelli 40 Reference Germann, Dihlmann and Hergenhahn 42 ). However, the effects of butyrate on c-MYC expression are complex. While increased c-MYC transcription is observed with butyrate-induced WNT signalling, this treatment may result in less c-MYC expression through inhibition of elongation causing a transcriptional block( Reference Wilson, Velcich and Arango 43 ).

Studies using LT97 (a cell line representative of an early stage in the progression from the normal mucosa to adenomas) showed greater up-regulation of WNT signalling and, consequently, induction of apoptosis with butyrate treatment suggesting that early-stage neoplasms may be more responsive to the effects of butyrate( Reference Lazarova, Lee and Wong 44 ).

A recent study by the Bordonaro group utilised total human microarray analyses to identify a total of 1587 direct and indirect targets of the WNT pathway whose expression was modulated by a physiologically-relevant concentration of butyrate (5 mm) in HCT-116 CRC cells( Reference Lazarova, Chiaro and Bordonaro 45 ). The differentially-expressed genes included those encoding proteins that are involved in several key processes including differentiation, migration and DNA replication.

Modulation of WNT signalling by resistant starch and butyrate via epigenetic mechanisms

Gene expression may be regulated by epigenetic mechanisms, such as histone modifications, DNA methylation and the expression of microRNA (miRNA)( Reference Mathers, Strathdee and Relton 46 ). These mechanisms result in heritable changes in gene expression and function without alterations in the DNA sequence itself( Reference Dupont, Armant and Brenner 47 ).

The role of butyrate as a histone deacetylase inhibitor

Histone acetylation refers to the addition of acetyl groups to the lysine residues on histone tails by histone acetyltransferase, removing the positive charge( Reference Struhl 48 ). Acetylated, relaxed DNA, known as euchromatin, is more easily accessible to transcription factors and may lead to increased transcription of the corresponding gene. On the contrary, histone deacetylation by histone deacetylase results in a more condensed DNA structure, known as heterochromatin, whereby transcription is reduced. Altered acetylation of histones associated with genes involved in regulation of the cell cycle, and particularly deacetylation of histone 4, has been linked with cancer development and progression( Reference Esteller 49 ).

The role of butyrate as a histone deacetylase inhibitor has been studied extensively and is well established( Reference Berni Canani, Di Costanzo and Leone 50 ) and butyrate treatment may restore expression of silenced genes leading to restoration of normal levels of proliferation, differentiation and apoptosis. Furthermore, butyrate affects acetylation of other non-histone targets, such as transcription factor Sp1( Reference Fung, Cosgrove and Lockett 51 ).

Resistant starch, butyrate and DNA methylation

DNA methylation describes the addition of a methyl group to the C5 position of a cytosine residue that is followed by a guanine residue, known as a CpG site, resulting in 5-methylcytosine( Reference Bird 52 ). DNA methylation is catalysed by a small family of DNA methyltransferases, primarily DNMT1, DNMT3a and DNMT3b( Reference Robertson 53 ). CpG islands refer to regions rich in CpG dinucleotides that are not normally methylated( Reference Robertson 54 ). Hypermethylation of CpG islands close to, or within, the promoter region is associated with repressed transcription principally through preventing transcription factor binding. DNA methylation can also inhibit transcription indirectly through steric hindrance by methyl-CpG-binding proteins which impede transcription factor binding( Reference Robertson 54 , Reference Baylin and Herman 55 ). In CRC and other cancers, global demethylation of DNA is observed frequently( Reference Baylin and Herman 55 ). In addition, promoter hypermethylation and, consequently, silencing of tumour suppressor genes is common in CRC as is hypomethylation and, therefore, up-regulation of oncogenes.

To date, there is only one published study of the effects of RS on DNA methylation of WNT pathway-related genes in a double-blind, randomised, placebo-controlled crossover trial with intervention periods lasting 4 weeks. In this study, DNA methylation was quantified using MethyLight, a qPCR-based method, in rectal mucosal biopsies collected from seventeen healthy participants( Reference Worthley, Le Leu and Whitehall 56 ). Methylation of the promoter regions of sixteen genes, including a member of the WNT pathway, SFRP1, was quantified. SFRP1 encodes a member of the SFRP family of WNT inhibitors and the down-regulation of SFRP1, associated with promoter methylation, is implicated in colorectal carcinogenesis( Reference Caldwell, Jones and Gensberg 24 ). However, Worthley et al. ( Reference Worthley, Le Leu and Whitehall 56 ) observed a significant (P = 0·040) effect of treatment on the methylation of MINT2 only and concluded that this was likely due to chance.

There are no published studies of the effects of butyrate in CRC cells on the methylation state of members of the WNT pathway. However, in human gastric cancer cells, where, as in CRC, aberrant activation of WNT signalling is observed frequently, Shin et al. ( Reference Shin, Kim and Lee 57 ) showed that butyrate treatment restored SFRP1 expression following promoter demethylation.

Resistant starch, butyrate and microRNA expression

miRNA are small, non-coding RNA that down-regulate the expression of their target genes by degrading mRNA or inhibiting translation. Approximately 1000 miRNA have been identified in human subject( Reference Bentwich, Avniel and Karov 58 ), with each single miRNA being able to target several genes and, likewise, a single gene may be targeted by many miRNA( Reference Lewis, Burge and Bartel 59 ). Aberrant expression of miRNA, resulting in altered expression (usually down-regulation) of target genes involved in the regulation of proliferation, migration and differentiation, may contribute to carcinogenesis( Reference Calin and Croce 60 ). Due to their recent discovery and the complexity in deciphering their many putative targets, investigations of the effects of NDC or butyrate on expression of miRNA that target genes from the WNT pathway specifically are limited.

A very recent randomised, controlled, crossover trial by Humphreys et al. ( Reference Humphreys, Conlon and Young 61 ) was the first reported human study to investigate the effects of RS on miRNA expression. Twenty-three participants were randomised to either a high red meat diet or a high red meat diet plus butyrylated RS for 4 weeks. Red meat has been associated with increased CRC risk, and so the aim of the study was to examine whether RS could reverse detrimental effects of the high red meat diet, particularly altered expression of miRNA, in the colorectum. Following consumption of the high red meat diet, the investigators observed an increase of approximately 30 % in expression of miRNA from the miR-17–92 cluster, an oncogenic cluster that is overexpressed in CRC. However, the effect was miRNAs specific and expression of five miRNA from this cluster, namely miR-17, miR-19a, miR-19b, miR-20a and miR-92a were significantly reduced in those fed butyrylated RS.

Microarray analysis of miRNA expression in HT29 colorectal adenocarcinoma cells treated with up to 10 mm sodium butyrate for 48 h revealed that a total of thirty-nine miRNA were up-regulated and thirty down-regulated( Reference Humphreys, Cobiac and Le Leu 62 ). Subsequent validation by qPCR confirmed that expression of the selected miRNA belonging to the miR-17–92 and miR-106a-363 clusters was reduced significantly following treatment with 5 mm butyrate.

The effects of resistant starch and butyrate on colonic crypt cell proliferation

Studies on the effects of RS and butyrate on crypt cell proliferation have yielded conflicting results. This divergence in findings is due, in part, to differences in the health status of the tissue under study. In some cases, mucosal cells from the normal colon respond to butyrate (and other SCFA) by increasing proliferation. In contrast, there is almost universal agreement that butyrate treatment of cancer cells suppresses proliferation( Reference Williams, Coxhead and Mathers 12 ). Furthermore, differences in RS or butyrate dose, type of RS, participant/cell characteristics and length of treatment may influence responses and contribute to difficulties in drawing unambiguous conclusions from available data. For example, we have shown that the DNA mismatch repair status of cells determines their cell proliferative/apoptotic response to butyrate treatment( Reference Dronamraju, Coxhead and Kelly 63 ).

Findings from studies that have investigated the effects of RS on colonic crypt cell proliferation are summarised in Table 1. The majority of the studies that have investigated the effects of RS supplementation on cell proliferation in the colon of healthy subjects have found no effect on proliferation. This includes the study by Wacker et al. ( Reference Wacker, Wanek and Eder 64 ) which administered the largest dose of RS (up to 59·7 g /d). In contrast, a number of studies have reported reduced cell proliferation in the colorectum of individuals with neoplasia( Reference Bauer-Marinovic, Florian and Muller-Schmehl 25 , Reference Le Leu, Brown and Hu 26 ). Importantly, we have also shown that supplementation of CRC patients with a 1:1 blend of Novelose 240 and Novelose 330 (RS types 2 and 3) reduced the proportion of mitotic cells in the top half of the crypt( Reference Dronamraju, Coxhead and Kelly 65 ). Study of the distribution of mitotic cells within the crypt (rather than measuring total proliferation within the whole crypt) may be a better indicator of CRC risk because alterations in the distribution of mitotic cells within the crypt have been observed to be one of the earliest detectable pre-malignant alterations in the apparently-normal mucosa of those at higher risk( Reference Mills, Shepherd and Hall 66 , Reference Terpstra, van Blankenstein and Dees 67 ).

Table 1. Studies that have investigated the effects of resistant starch (RS) on colonic crypt cell proliferation

HAMS, high amylose maize starch; HAMSB, butyrylated high amylose maize starch.

Likewise, the findings from investigations of the effects of butyrate on colonic crypt cell proliferation are inconsistent. While there is an evidence that butyrate may increase cell proliferation in the healthy colon in specific circumstances ( Reference Scheppach, Bartram and Richter 68 , Reference Kien, Blauwiekel and Bunn 69 , Reference Mortensen, Langkilde and Joergensen 70 ), based on our earlier observations in rats, we concluded that there is no convincing evidence that SCFA (or butyrate, specifically) are responsible for raising crypt cell proliferation above normal. In those instances where greater SCFA supply has been associated with increased crypt cell proliferation, the increase may be (1) from a hypoproliferative state towards a normal proliferative state, (2) a transient phenomenon accompanying tissue hypertrophy or (3) a homeostatic response to increased cell loss by cell sloughing or apoptosis( Reference Key, McClean and Mathers 71 ).

The differential responses of normal and cancer cells to butyrate treatment are referred to as the butyrate paradox. Comalada et al. ( Reference Comalada, Bailon and de Haro 72 ) compared the effects of butyrate treatment on healthy fetal human colon cells and on HT-29 colorectal adenocarcinoma cells. Butyrate inhibited cell proliferation of the HT-29 cells but had no effect on the normal cells. Significantly reduced cell proliferation of HT-29 cells treated with 5 mm butyrate for 48 h has also been reported by Hodin et al. ( Reference Hodin, Meng and Archer 73 ).

Bordonaro et al. ( Reference Bordonaro, Lazarova and Sartorelli 15 ) have suggested that the differences in the effects of butyrate observed on proliferation in healthy compared with cancerous cells may be due to the sensitivity and responsiveness of these cells to butyrate. They proposed that, in cancerous cells where the WNT pathway is hyperactive, butyrate further induces WNT signalling and consequently apoptosis. However, in healthy cells where normal, moderate levels of WNT activity are found, butyrate contributes to the normal regulation of processes within the colon by the WNT pathway, such as the induction of proliferation. However, the latter is likely to increase only if starting from an abnormally low level( Reference Key, McClean and Mathers 71 ).

The effects of resistant starch and butyrate on apoptosis in the large bowel

A number of studies, mostly in vivo, have shown that increased WNT activity is associated with induction of apoptosis( Reference Bordonaro, Mariadason and Aslam 37 , Reference Romagnolo, Berrebi and Saadi-Keddoucci 74 , Reference Wong, Huelsken and Birchmeier 75 ). Furthermore, the Bordonaro group have reported a linear relationship between WNT activity and levels of apoptosis in ten CRC cell lines( Reference Lazarova, Bordonaro and Carbone 76 ). However, several in vitro studies have reported the opposite effect( Reference Groden, Joslyn and Samowitz 77 Reference Zhang, Otevrel and Gao 79 ).

In sixteen pigs supplemented with raw potato starch for 16 weeks, Nofrarias et al. ( Reference Nofrarias, Martinez-Puig and Pujols 80 ) observed a significant reduction in apoptosis within the crypts of pigs fed the RS diet compared with controls. A similar earlier study also reported a reduction in apoptosis in the colon of pigs fed a potato starch diet and, interestingly, these researchers reported significantly higher faecal butyrate concentrations following the RS diet( Reference Claus, Losel and Lacorn 81 ).

The majority of studies have agreed that butyrate induces apoptosis( Reference Armstrong and Mathers 82 ), primarily via induction of the intrinsic and extrinsic apoptotic pathways. Furthermore, butyrate has been reported to modulate the expression of apoptotic genes including down-regulation of the anti-apoptotic gene BCL-2 ( Reference Avivi-Green, Polak-Charcon and Madar 83 ) and up-regulation of the pro-apoptotic gene BAX ( Reference Purwani, Iskandriati and Suhartono 84 ). In vivo, increased apoptosis has been observed in both the distal and proximal colon of carcinogen-treated rats fed a diet with RS type 3( Reference Bauer-Marinovic, Florian and Muller-Schmehl 25 ). Furthermore, significantly greater levels of apoptosis have been found in the distal colon of carcinogen-treated rats fed a butyrylated high amylose maize starch, which produces significantly greater concentrations of butyrate in the colon, compared with HAMS and a low RS diet( Reference Clarke, Young and Topping 85 ). In addition, this increase in apoptosis correlated positively with distal colonic luminal butyrate concentrations, suggesting that the enhanced apoptosis was a consequence of the extra butyrate production. However, some studies have reported no effect of butyrate on levels of apoptosis in the colon( Reference Winter, Young and Hu 86 , Reference Le Leu, Hu and Brown 87 ).

Conclusions

The WNT signalling pathway is central to normal function of the colorectal epithelium and aberrant WNT signalling is a cardinal feature of most CRC. Since high intakes of NDC are associated with lower CRC risk, this review investigated the evidence that NDC such as RS may have this protective effect through impacts on WNT signalling. Such effects may be mediated by butyrate, a major SCFA endproduct of RS fermentation in the large bowel. Several studies have observed positive modulation of WNT pathway components by RS and by butyrate and, in some cases, these have correlated with protective effects on functional outcomes such as apoptosis and differentiation. The effects of RS and butyrate on the expression of WNT pathway-related genes may result from epigenetic mechanisms including inhibition of histone deacetylation, reduction of DNA methylation and altered expression of miRNA. In particular, butyrate reduces the methylation state of SFRP1, which is frequently silenced in CRC as a consequence of hypermethylation, in cancer cells. In addition, RS and butyrate reduce expression of miRNA from the oncogenic miR-17–92 cluster both in vitro and in vivo. Numerous studies have reported effects of RS and its fermentation product, butyrate, on cell proliferation and apoptosis, which may be regulated by the WNT pathway. However, the effects of both RS and butyrate on proliferation and apoptosis appear to differ markedly between normal and tumour cells. In the healthy crypt, RS and butyrate contribute to the maintenance of homeostasis by WNT signalling through promoting (initially low) levels of proliferation and by reducing apoptosis. However, in cancerous cells, where the crypt is in a hyperproliferative state and has high levels of WNT signalling, RS and butyrate reduce proliferation and induce apoptosis. It must be noted, however, that the effects of RS and butyrate on these two processes may not be due exclusively to effects on WNT signalling and that modulation of additional pathways including the Notch and MAPK signalling pathways is also likely to be important( Reference Bordonaro, Tewari and Atamna 88 , Reference Zuo, Lu and Zhou 89 ). To confirm the chemoprotective effects of RS and butyrate, and to better understand the mechanisms through which these effects are mediated, well-designed, randomised, placebo-controlled dietary intervention studies are required. In the only such human randomised controlled trial study to date with cancer as the endpoint, there was no evidence that supplemental RS affected the development of CRC( Reference Mathers, Movahedi and Macrae 90 ) and we concluded that dietary supplementation with RS does not emulate the apparently protective effects of diets rich in dietary fibre against CRC.

Financial Support

This work was supported by an award from the BBSRC Diet and Health Research Industry Club (DRINC) (grant number BB/H005013/1).

Conflicts of Interest

None.

Authorship

This manuscript was prepared by F. C. M., edited by N. D. W. and J. C. M. and final version agreed by all authors.

References

1. Cancer Research UK. Bowel cancer key facts. (2014) http://www.cancerresearchuk.org/cancer-info/cancerstats/keyfacts/bowel-cancer/ (accessed August 2014).Google Scholar
2. Hanahan, D, Weinberg, RA (2011) Hallmarks of cancer: the next generation. Cell 144, 646674.Google Scholar
3. Parkin, DM, Boyd, L, Walker, LC (2011) 16. The fraction of cancer attributable to lifestyle and environmental factors in the UK in 2010. Br J Cancer 105 Suppl 2, S77S81.Google Scholar
4. World Cancer Research Fund/American Institute for Cancer Research (2011) Continuous update project report. Food, nutrition, physical activity, and the prevention of colorectal cancer.Google Scholar
5. CAC (2009) Codex Alimentarius Commission; Food and Agriculture Organization; World Health Organization. Report of the 30th session of the Codex Commitee on nutrition and foods for special dietary uses. http://www.codexalimentarius.net/download/report/710/al32_26e.pdf (accessed August 2014).Google Scholar
6. Cummings, JH & Stephen, AM (2007) Carbohydrate terminology and classification. Eur J Clin Nutr 61 Suppl 1, S518.Google Scholar
7. Topping, DL & Clifton, PM (2001) Short-chain fatty acids and human colonic function: roles of resistant starch and nonstarch polysaccharides. Physiol Rev 81, 10311064.Google Scholar
8. Burkitt, DP (1971) Epidemiology of cancer of the colon and rectum. Cancer 28, 313.Google Scholar
9. Aune, D, Chan, DS, Lau, R et al. (2011) Dietary fibre, whole grains, and risk of colorectal cancer: systematic review and dose-response meta-analysis of prospective studies. Br Med J 343, d6617.Google Scholar
10. Hylla, S, Gostner, A, Dusel, G et al. (1998) Effects of resistant starch on the colon in healthy volunteers: possible implications for cancer prevention. Am J Clin Nutr 67, 136142.Google Scholar
11. Lipkin, M, Reddy, B, Newmark, H et al. (1999) Dietary factors in human colorectal cancer. Annu Rev Nutr 19, 545586.Google Scholar
12. Williams, EA, Coxhead, JM & Mathers, JC (2003) Anti-cancer effects of butyrate: use of micro-array technology to investigate mechanisms. Proc Nutr Soc 62, 107115.Google Scholar
13. Pryde, SE, Duncan, SH, Hold, GL et al. (2002) The microbiology of butyrate formation in the human colon. FEMS Microbiol Lett 217, 133139.Google Scholar
14. Hamer, HM, Jonkers, D, Venema, K et al. (2008) Review article: the role of butyrate on colonic function. Aliment Pharmacol Ther 27, 104119.Google Scholar
15. Bordonaro, M, Lazarova, DL & Sartorelli, AC (2008) Butyrate and Wnt signaling: a possible solution to the puzzle of dietary fiber and colon cancer risk? Cell Cycle 7, 11781183.CrossRefGoogle Scholar
16. Clevers, H (2006) Wnt/beta-catenin signaling in development and disease. Cell 127, 469480.Google Scholar
17. Bienz, M & Clevers, H (2000) Linking colorectal cancer to Wnt signaling. Cell 103, 311320.Google Scholar
18. Schneikert, J & Behrens, J (2007) The canonical Wnt signalling pathway and its APC partner in colon cancer development. Gut 56, 417425.Google Scholar
19. Sierra, J, Yoshida, T, Joazeiro, CA et al. (2006) The APC tumor suppressor counteracts beta-catenin activation and H3K4 methylation at Wnt target genes. Genes Dev 20, 586600.Google Scholar
20. Luo, J, Chen, J, Deng, ZL et al. (2007) Wnt signaling and human diseases: what are the therapeutic implications? Lab Invest 87, 97103.Google Scholar
21. Klaus, A & Birchmeier, W (2008) Wnt signalling and its impact on development and cancer. Nat Rev Cancer 8, 387398.Google Scholar
22. Kinzler, KW & Vogelstein, B (1996) Lessons from hereditary colorectal cancer. Cell 87, 159170.Google Scholar
23. Morin, PJ, Sparks, AB, Korinek, V et al. (1997) Activation of beta-catenin-Tcf signaling in colon cancer by mutations in beta-catenin or APC. Science 275, 17871790.Google Scholar
24. Caldwell, GM, Jones, C, Gensberg, K et al. (2004) The Wnt antagonist sFRP1 in colorectal tumorigenesis. Cancer Res 64, 883888.Google Scholar
25. Bauer-Marinovic, M, Florian, S, Muller-Schmehl, K et al. (2006) Dietary resistant starch type 3 prevents tumor induction by 1,2-dimethylhydrazine and alters proliferation, apoptosis and dedifferentiation in rat colon. Carcinogenesis 27, 18491859.Google Scholar
26. Le Leu, RK, Brown, IL, Hu, Y et al. (2007) Suppression of azoxymethane-induced colon cancer development in rats by dietary resistant starch. Cancer Biol Ther 6, 16211626.Google Scholar
27. Alrawi, SJ, Schiff, M, Carroll, RE et al. (2006) Aberrant crypt foci. Anticancer Res 26, 107119.Google Scholar
28. Liu, R & Xu, G (2008) Effects of resistant starch on colonic preneoplastic aberrant crypt foci in rats. Food Chem Toxicol 46, 26722679.CrossRefGoogle ScholarPubMed
29. Thorup, I, Meyer, O & Kristiansen, E (1995) Effect of potato starch, cornstarch and sucrose on aberrant crypt foci in rats exposed to azoxymethane. Anticancer Res 15, 21012105.Google Scholar
30. Cassand, P, Maziere, S, Champ, M et al. (1997) Effects of resistant starch- and vitamin A-supplemented diets on the promotion of precursor lesions of colon cancer in rats. Nutr Cancer 27, 5359.Google Scholar
31. Sakamoto, J, Nakaji, S, Sugawara, K et al. (1996) Comparison of resistant starch with cellulose diet on 1,2-dimethylhydrazine-induced colonic carcinogenesis in rats. Gastroenterology 110, 116120.Google Scholar
32. Maziere, S, Meflah, K, Tavan, E et al. (1998) Effect of resistant starch and/or fat-soluble vitamins A and E on the initiation stage of aberrant crypts in rat colon. Nutr Cancer 31, 168177.Google Scholar
33. Young, GP, McIntyre, A, Albert, V et al. (1996) Wheat bran suppresses potato starch–potentiated colorectal tumorigenesis at the aberrant crypt stage in a rat model. Gastroenterology 110, 508514.Google Scholar
34. Smits, R, Kartheuser, A, Jagmohan-Changur, S et al. (1997) Loss of Apc and the entire chromosome 18 but absence of mutations at the Ras and Tp53 genes in intestinal tumors from Apc1638N, a mouse model for Apc-driven carcinogenesis. Carcinogenesis 18, 321327.Google Scholar
35. Williamson, SL, Kartheuser, A, Coaker, J et al. (1999) Intestinal tumorigenesis in the Apc1638N mouse treated with aspirin and resistant starch for up to 5 months. Carcinogenesis 20, 805810.Google Scholar
36. Cray, NL (2013) Effects of diets containing digestion-resistant starch on Wnt pathway control of proliferation and differentiation of the colorectal mucosa. Ames, Iowa: Iowa State University.Google Scholar
37. Bordonaro, M, Mariadason, JM, Aslam, F et al. (1999) Butyrate-induced apoptotic cascade in colonic carcinoma cells: modulation of the beta-catenin-Tcf pathway and concordance with effects of sulindac and trichostatin A but not curcumin. Cell Growth Differ 10, 713720.Google Scholar
38. Bordonaro, M, Lazarova, DL & Sartorelli, AC (2007) The activation of beta-catenin by Wnt signaling mediates the effects of histone deacetylase inhibitors. Exp Cell Res 313, 16521666.Google Scholar
39. Vincan, E, Leet, CS, Reyes, NI et al. (2000) Sodium butyrate-induced differentiation of human LIM2537 colon cancer cells decreases GSK-3beta activity and increases levels of both membrane-bound and Apc/axin/GSK-3beta complex-associated pools of beta-catenin. Oncol Res 12, 193201.Google Scholar
40. Wong, NA & Pignatelli, M (2002) Beta-catenin–a linchpin in colorectal carcinogenesis? Am J Pathol 160, 389401.Google Scholar
41. Zhang, W, Hart, J, McLeod, HL et al. (2005) Differential expression of the AP-1 transcription factor family members in human colorectal epithelial and neuroendocrine neoplasms. Am J Clin Pathol 124, 1119.Google Scholar
42. Germann, A, Dihlmann, S, Hergenhahn, M et al. (2003) Expression profiling of CC531 colon carcinoma cells reveals similar regulation of beta-catenin target genes by both butyrate and aspirin. Int J Cancer 106, 187197.Google Scholar
43. Wilson, 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 Scholar
44. Lazarova, D, Lee, A, Wong, T et al. (2014) Modulation of Wnt activity and cell physiology by butyrate in LT97 microadenoma cells. J Cancer 5, 203213.CrossRefGoogle ScholarPubMed
45. Lazarova, DL, Chiaro, C & Bordonaro, M (2014) Butyrate induced changes in Wnt-signaling specific gene expression in colorectal cancer cells. BMC Res Notes 7, 226.CrossRefGoogle ScholarPubMed
46. Mathers, JC, Strathdee, G & Relton, CL (2010) Induction of epigenetic alterations by dietary and other environmental factors. Adv Genet 71, 339.Google Scholar
47. Dupont, C, Armant, DR & Brenner, CA (2009) Epigenetics: definition, mechanisms and clinical perspective. Semin Reprod Med 27, 351357.CrossRefGoogle ScholarPubMed
48. Struhl, K (1998) Histone acetylation and transcriptional regulatory mechanisms. Genes Dev 12, 599606.Google Scholar
49. Esteller, M (2007) Cancer epigenomics: DNA methylomes and histone-modification maps. Nat Rev Genet 8, 286298.Google Scholar
50. Berni Canani, R, Di Costanzo, M & Leone, L (2012) The epigenetic effects of butyrate: potential therapeutic implications for clinical practice. Clin Epigenetics 4, 4.Google Scholar
51. Fung, KY, Cosgrove, L, Lockett, T et al. (2012) A review of the potential mechanisms for the lowering of colorectal oncogenesis by butyrate. Br J Nutr 108, 820831.CrossRefGoogle ScholarPubMed
52. Bird, A (2002) DNA methylation patterns and epigenetic memory. Genes Dev 16, 621.CrossRefGoogle ScholarPubMed
53. Robertson, KD (2001) DNA methylation, methyltransferases, and cancer. Oncogene 20, 31393155.CrossRefGoogle ScholarPubMed
54. Robertson, KD (2005) DNA methylation and human disease. Nat Rev Genet 6, 597610.Google Scholar
55. Baylin, SB & Herman, JG (2000) DNA hypermethylation in tumorigenesis: epigenetics joins genetics. Trends Genet 16, 168174.Google Scholar
56. Worthley, DL, Le Leu, RK, Whitehall, VL et al. (2009) A human, double-blind, placebo-controlled, crossover trial of prebiotic, probiotic, and synbiotic supplementation: effects on luminal, inflammatory, epigenetic, and epithelial biomarkers of colorectal cancer. Am J Clin Nutr 90, 578586.CrossRefGoogle ScholarPubMed
57. Shin, H, Kim, JH, Lee, YS et al. (2012) Change in gene expression profiles of secreted frizzled-related proteins (SFRPs) by sodium butyrate in gastric cancers: induction of promoter demethylation and histone modification causing inhibition of Wnt signaling. Int J Oncol 40, 15331542.Google Scholar
58. Bentwich, I, Avniel, A, Karov, Y et al. (2005) Identification of hundreds of conserved and nonconserved human microRNAs. Nat Genet 37, 766770.Google Scholar
59. Lewis, BP, Burge, CB & Bartel, DP (2005) Conserved seed pairing, often flanked by adenosines, indicates that thousands of human genes are microRNA targets. Cell 120, 1520.Google Scholar
60. Calin, GA & Croce, CM (2006) MicroRNA signatures in human cancers. Nat Rev Cancer 6, 857866.Google Scholar
61. Humphreys, KJ, Conlon, MA, Young, GP et al. (2014) Dietary manipulation of oncogenic microRNA expression in human rectal Mucosa: a randomized trial. Cancer Prev Res (Phila) 7, 786795.Google Scholar
62. Humphreys, KJ, Cobiac, L, Le Leu, RK et al. (2013) Histone deacetylase inhibition in colorectal cancer cells reveals competing roles for members of the oncogenic miR-17–92 cluster. Mol Carcinog 52, 459474.Google Scholar
63. Dronamraju, SS, Coxhead, JM, Kelly, SB et al. (2010) Differential antineoplastic effects of butyrate in cells with and without a functioning DNA mismatch repair. Nutr Cancer 62, 105115.CrossRefGoogle ScholarPubMed
64. Wacker, M, Wanek, P, Eder, E et al. (2002) Effect of enzyme-resistant starch on formation of 1,N(2)-propanodeoxyguanosine adducts of trans-4-hydroxy-2-nonenal and cell proliferation in the colonic mucosa of healthy volunteers. Cancer Epidemiol Biomarkers Prev 11, 915920.Google ScholarPubMed
65. Dronamraju, SS, Coxhead, JM, Kelly, SB et al. (2009) Cell kinetics and gene expression changes in colorectal cancer patients given resistant starch: a randomised controlled trial. Gut 58, 413420.Google Scholar
66. Mills, SJ, Shepherd, NA, Hall, PA et al. (1995) Proliferative compartment deregulation in the non-neoplastic colonic epithelium of familial adenomatous polyposis. Gut 36, 391394.Google Scholar
67. Terpstra, OT, van Blankenstein, M, Dees, J et al. (1987) Abnormal pattern of cell proliferation in the entire colonic mucosa of patients with colon adenoma or cancer. Gastroenterology 92, 704708.Google Scholar
68. Scheppach, W, Bartram, P, Richter, A et al. (1992) Effect of short-chain fatty acids on the human colonic mucosa in vitro. JPEN J Parenter Enteral Nutr 16, 4348.Google Scholar
69. Kien, CL, Blauwiekel, R, Bunn, JY et al. (2007) Cecal infusion of butyrate increases intestinal cell proliferation in piglets. J Nutr 137, 916922.Google Scholar
70. Mortensen, FV, Langkilde, NC, Joergensen, JC et al. (1999) Short-chain fatty acids stimulate mucosal cell proliferation in the closed human rectum after Hartmann's procedure. Int J Colorectal Dis 14, 150154.Google Scholar
71. Key, FB, McClean, D & Mathers, JC (1996) Tissue hypertrophy and epithelial proliferation rate in the gut of rats fed on bread and haricot beans (Phaseolus vulgaris). Br J Nutr 76, 273286.Google Scholar
72. Comalada, M, Bailon, E, de Haro, O et al. (2006) The effects of short-chain fatty acids on colon epithelial proliferation and survival depend on the cellular phenotype. J Cancer Res Clin Oncol 132, 487497.Google Scholar
73. Hodin, RA, Meng, S, Archer, S et al. (1996) Cellular growth state differentially regulates enterocyte gene expression in butyrate-treated HT-29 cells. Cell Growth Differ 7, 647653.Google Scholar
74. Romagnolo, B, Berrebi, D, Saadi-Keddoucci, S et al. (1999) Intestinal dysplasia and adenoma in transgenic mice after overexpression of an activated beta-catenin. Cancer Res 59, 38753879.Google Scholar
75. Wong, MH, Huelsken, J, Birchmeier, W et al. (2002) Selection of multipotent stem cells during morphogenesis of small intestinal crypts of Lieberkuhn is perturbed by stimulation of Lef-1/beta-catenin signaling. J Biol Chem 277, 1584315850.CrossRefGoogle ScholarPubMed
76. Lazarova, DL, Bordonaro, M, Carbone, R et al. (2004) Linear relationship between Wnt activity levels and apoptosis in colorectal carcinoma cells exposed to butyrate. Int J Cancer 110, 523531.Google Scholar
77. Groden, J, Joslyn, G, Samowitz, W et al. (1995) Response of colon cancer cell lines to the introduction of APC, a colon-specific tumor suppressor gene. Cancer Res 55, 15311539.Google Scholar
78. Morin, PJ, Vogelstein, B & Kinzler, KW (1996) Apoptosis and APC in colorectal tumorigenesis. Proc Natl Acad Sci U S A 93, 79507954.Google Scholar
79. Zhang, T, Otevrel, T, Gao, Z et al. (2001) Evidence that APC regulates survivin expression: a possible mechanism contributing to the stem cell origin of colon cancer. Cancer Res 61, 86648667.Google Scholar
80. Nofrarias, M, Martinez-Puig, D, Pujols, J et al. (2007) Long-term intake of resistant starch improves colonic mucosal integrity and reduces gut apoptosis and blood immune cells. Nutrition 23, 861870.Google Scholar
81. Claus, R, Losel, D, Lacorn, M et al. (2003) Effects of butyrate on apoptosis in the pig colon and its consequences for skatole formation and tissue accumulation. J Anim Sci 81, 239248.Google Scholar
82. Armstrong, F & Mathers, JC (2000) Kill and cure: dietary augmentation of immune defences against colon cancer. Proc Nutr Soc 59, 215220.Google Scholar
83. Avivi-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.Google Scholar
84. Purwani, E, Iskandriati, D & Suhartono, M (2012) Fermentation product of RS3 inhibited proliferation and induced apoptosis in colon cancer cell HCT-116. Adv. Biosci. Biotechnol. 3, 11891198.Google Scholar
85. Clarke, JM, Young, GP, Topping, DL et al. (2012) Butyrate delivered by butyrylated starch increases distal colonic epithelial apoptosis in carcinogen-treated rats. Carcinogenesis 33, 197202.Google Scholar
86. Winter, J, Young, GP, Hu, Y et al. (2014) Accumulation of promutagenic DNA adducts in the mouse distal colon after consumption of heme does not induce colonic neoplasms in the western diet model of spontaneous colorectal cancer. Mol Nutr Food Res 58, 550558.CrossRefGoogle Scholar
87. Le Leu, RK, Hu, Y, Brown, IL et al. (2010) Synbiotic intervention of Bifidobacterium lactis and resistant starch protects against colorectal cancer development in rats. Carcinogenesis 31, 246251.Google Scholar
88. Bordonaro, M, Tewari, S, Atamna, W et al. (2011) The Notch ligand Delta-like 1 integrates inputs from TGFbeta/Activin and Wnt pathways. Exp Cell Res 317, 13681381.Google Scholar
89. Zuo, L, Lu, M, Zhou, Q et al. (2013) Butyrate suppresses proliferation and migration of RKO colon cancer cells though regulating endocan expression by MAPK signaling pathway. Food Chem Toxicol 62, 892900.CrossRefGoogle ScholarPubMed
90. Mathers, JC, Movahedi, M, Macrae, F et al. (2012) Long-term effect of resistant starch on cancer risk in carriers of hereditary colorectal cancer: an analysis from the CAPP2 randomised controlled trial. Lancet Oncol 13, 12421249.Google Scholar
91. van Munster, IP, Tangerman, A & Nagengast, FM (1994) Effect of resistant starch on colonic fermentation, bile acid metabolism, and mucosal proliferation. Dig Dis Sci 39, 834842.Google Scholar
92. Burn, J, Bishop, DT, Chapman, PD et al. (2011) A randomized placebo-controlled prevention trial of aspirin and/or resistant starch in young people with familial adenomatous polyposis. Cancer Prev Res (Phila) 4, 655665.Google Scholar
93. van Gorkom, BA, Karrenbeld, A, van der Sluis, T et al. (2002) Calcium or resistant starch does not affect colonic epithelial cell proliferation throughout the colon in adenoma patients: a randomized controlled trial. Nutr Cancer 43, 3138.Google Scholar
94. Grubben, MJ, van den Braak, CC, Essenberg, M et al. (2001) Effect of resistant starch on potential biomarkers for colonic cancer risk in patients with colonic adenomas: a controlled trial. Dig Dis Sci 46, 750756.Google Scholar
95. Jacobasch, G, Schmiedl, D, Kruschewski, M et al. (1999) Dietary resistant starch and chronic inflammatory bowel diseases. Int J Colorectal Dis 14, 201211.Google Scholar
96. Mentschel, J & Claus, R (2003) Increased butyrate formation in the pig colon by feeding raw potato starch leads to a reduction of colonocyte apoptosis and a shift to the stem cell compartment. Metabolism 52, 14001405.Google Scholar
Figure 0

Fig. 1. The canonical WNT signalling pathway. (a) In the inactive state, when there are no WNT ligands bound to frizzled receptors or when WNT antagonists, such as WIF1, inhibit activation of the WNT pathway, the β-catenin destruction complex (adenomatous polyposis coli (APC), AXIN, casein kinase (CK1) and glycogen synthase kinase 3β (GSK3β)) binds to and phosphorylates β-catenin. This targets β-catenin for ubiquitination by β-transducin 117 repeat-containing protein (β-TrCP) and, consequently, proteasomal degradation. In the nucleus, lymphoid enhancer factor-1 (LEF) and T cell factor (TCF) transcription factors are repressed by Groucho. (b) When the WNT pathway is activated by binding of WNT ligands to frizzled receptors, the β-catenin destruction complex is inhibited by dishevelled (Dvl). Levels of dephosphorylated, active β-catenin rise and β-catenin translocates to the nucleus where it binds to LEF and TCF transcription factors to activate transcription of target genes such as c-MYC and c-JUN.

Figure 1

Table 1. Studies that have investigated the effects of resistant starch (RS) on colonic crypt cell proliferation