Editorial Focus

A novel facet to consider for the effects of butyrate on its target cells. Focus on “The short-chain fatty acid butyrate is a substrate of breast cancer resistance protein”

Published Online:https://doi.org/10.1152/ajpcell.00290.2011

short-chain fatty acids (SCFAs), acetate, propionate, and butyrate are produced by bacterial fermentation of dietary fiber in the colonic lumen. In the normal human large intestine, SCFA concentrations range from 70 to 100 mM; these SCFAs account for ∼80% of the anions present in the lumen (5). The relative ratio of acetate:propionate:butyrate is considered to remain stable at 60:25:15 (11). Butyrate has received the most attention due to its multiple beneficial roles in health and disease states. Butyrate is considered the preferred energy source for colonocytes and helps maintain the epithelial integrity (5, 17). Numerous beneficial roles of butyrate within and outside the intestine have been reported. In the intestine, butyrate has multiple effects (5), which include 1) increases in NaCl absorption via activating Na+-H+ and Cl-HCO3 exchangers (6, 9); 2) influences on cell proliferation and differentiation (8, 17); 3) plays an anti-inflammatory role via modulating the release of prostaglandin E2, cytokines, and chemokines from immune cells (10); 4) alters gut barrier function by inducing mucin synthesis and antimicrobial peptide production, and by decreasing intestinal tight junction permeability via AMP-activated protein kinase (5); 5) affects colonic motility by modulating acetylation in the myenteric plexus and via release of 5-HT (13, 27); and 6) helps prevent and inhibit colonic carcinogenesis. Extra-intestinal beneficial effects of butyrate include 1) increasing fetal hemoglobin production; 2) lowering serum cholesterol levels; 3) stimulating neurogenesis in brain after ischemic injury; and 4) providing positive effects in the treatment of obesity, insulin resistance, cystic fibrosis, urea cycle enzyme deficiency, and sickle cell disease (5). The role of butyrate in the inhibition and prevention of colon cancer is likely one of its most important beneficial effects.

Butyrate and colon cancer.

Butyrate has been shown to act as both preventive and inhibitory in carcinogenesis of the colon. The chemopreventive effect is mediated by upregulation of detoxifying enzymes for xenobiotics and oxidants, an effect that derives from rather complex actions of butyrate on cell proliferation and differentiation, termed the “butyrate paradox” (5, 8). This term derives from the ability of butyrate to inhibit cell proliferation and induce apoptosis in colon cancer cell lines, whereas under normal conditions butyrate induces cell proliferation in colonic crypts (1). The mechanisms of the effects of butyrate on colon cancer mainly include its absorption into colonocytes followed by its multiple effects on cell proliferation/differentiation via its inhibition of histone deacetylases (HDACs) (Fig. 1). In human colonic cell lines, butyrate increases p21 gene expression, thereby inducing cell cycle arrest via inhibition of HDACs (7). However, a recent report showed that this effect of butyrate on p21 gene expression occurs by two mechanisms: HDAC inhibition and decreased expression of the miR-106b gene family (19). Additional effects of butyrate on cell apoptosis involve effects on Bcl2 family proteins, e.g., upregulation of (pro-apoptotic) BAK and downregulation of (anti-apoptotic) BclxL (24, 25). Another anticancer effect of butyrate is its effects on canonical Wnt signaling pathway, which is constitutively activated in most colonic tumors (5). Butyrate may also induce autophagy in colonic epithelial cells (28). Indirect effects of butyrate, e.g., upregulating MDR1 expression or conversion of estrone to 17β-estradiol may also underlie the decreased incidence of colon cancer (2, 22). Two very recent studies, however, showed that the anticancer effects of butyrate involved its interactions with cell surface G-protein-coupled receptors: GPR109a and GPR43 (29, 30), effects that appear to be independent of its inhibition of HDACs (30). Thus the anticarcinogenic effects of butyrate are rather complex and could involve participation of SCFA receptors as well as the uptake of butyrate into the colonocytes and subsequent effects on HDACs (Fig. 1).

Fig. 1.

Fig. 1.Mechanisms of butyrate (BT) entry, efflux, and its effects on colon cancer. Ac, acetylation; MCT1, monocarboxylate transporter1; SMCT1, sodium dependent MCT1; BCRP, breast cancer resistance protein; HDAC, histone deacetylase.


Mechanisms of butyrate uptake and efflux from the colonocytes.

It was assumed for a long time that non-ionic diffusion of protonated SCFAs was the major mechanism of SCFA absorption in the intestine (9). However, other studies showed involvement of carrier-mediated processes, e.g., SCFA/HCO3 or a SCFA/Cl exchangers (9, 12). It is now accepted that monocarboxylate transporter 1 (MCT1) plays a major role in carrier-mediated SCFA transport in colonocytes (3, 12) (Fig. 1). However, its localization in polarized colonocytes has been controversial. Although many studies indicate it localizes to the apical membrane, its basolateral localization has also been reported (15, 20). SLC5A8 (SMCT1), a sodium-dependent monocarboxylate transporter also localizes to the apical membranes of colonocytes (4, 14). However, its functionality in the human colon has not yet been demonstrated.

Very little information is available with respect to the efflux of the absorbed SCFAs from the colonocytes, although basolateral MCTs (e.g., MCT1, MCT4, or MCT5) may play a role in this process (Fig. 1) (15). In other cell types (e.g., hepatocytes), butyrate efflux has been linked to the uptake of extracellular sulfate-conjugated steroids via organic anion transporter-7 (OATP-7) (26). A suppression in the expression of both MCT1 and SMCT1 appears to be an adaptive mechanism of colon cancer cells to avert the anticancer effect of butyrate (14, 21). Interestingly, Glut1 expression is upregulated in colon cancer, indicating that, during malignancy, colonic tissue switches from utilizing butyrate to glucose for energy needs (21). It is important to note that colonic cancer cells and normal cells also express other mechanisms that may lower butyrate levels in the cells: multi-drug resistance (MDR)-related transporters [e.g., MDR1, MRPs or BCRP (members of the ABC superfamily of transporters)] that can reduce the intracellular availability of anticancer agents and other xenobiotics. Interestingly, butyrate can upregulate the expression of MDR1 expression (22). However, whether any of these transporters play a role in transporting butyrate has not been investigated.

A novel role of BCRP in butyrate efflux from colonocytes.

In this issue of the American Journal of Physiology–Cell Physiology, Gonçalves et al. (16) demonstrate that butyrate is a substrate for BCRP but not for MDR1 or MRPs. The authors use pharmacological approaches with known inhibitors of these proteins to study the uptake or efflux of butyrate in two intestinal cell lines and one breast cancer cell line in which butyrate serves as a substrate of BCRP and can be pumped out of the cell via this protein. The authors utilized a transformed intestinal cell line (Caco2) and a non-transformed rat intestinal cell line (IEC-6) to examine the potential role of these transporters in both normal and transformed cells. Interestingly, authors showed that IEC-6 cells, but not Caco2 cells, have BCRP-mediated efflux of butyrate. The absence of an efflux process for butyrate through BCRP was shown to result from the absence of BCRP expression in Caco2 cells. However, in butyrate-treated (48 h) Caco2 cells, BCRP expression was increased and resulted in the transport of butyrate. The authors also demonstrated that this role of BCRP in butyrate efflux was not cell line-specific: MDA-MB-231, a breast cancer line known to express high levels of BCRP, also showed a role for BCRP in efflux of butyrate. The functionality of BCRP in butyrate transport was further confirmed by examining the effects of butyrate on cell proliferation in IEC-6 cells: butyrate combined with a BCRP inhibitor potentiated the inhibitory effect of butyrate on cell proliferation. The authors also showed a role for butyrate in inhibiting the efflux of [3H]folate, a known substrate for BCRP, in IEC-6 cells. Gonçalves et al. (16) speculate that, since butyrate is a substrate for BCRP, it may compete with chemotherapeutic drugs (e.g., 5-fluorouracil, methotrexate, etc.) for exit from the cells and thus might increase their intracellular concentration and enhance their therapeutic effect. These studies, therefore, provide a new facet to consider in understanding the mechanisms that underlie beneficial effects of SCFAs as well as in designing butyrate-based therapeutic modalities.

This exciting report also raises many questions. For example: 1) What is the physiological significance of BCRP in butyrate efflux in view of multiple beneficial effects of butyrate in healthy subjects? This argument should also consider the fact that BCRP expression is highest in the human duodenum and lowest in the colon (18); however, butyrate is produced mainly in the colon. 2) Does BCRP function as an efflux protein for other SCFAs (e.g., acetate or propionate), which also have beneficial effects? 3) BCRP expression is suppressed in cancer cells (23), consistent with the low expression seen in Caco-2 cells; however, the present study shows upregulation of BCRP in Caco2 cells by butyrate. Thus it is not clear whether the findings of Gonçalves et al. (16) will occur under in vivo conditions where high concentrations of luminal butyrate exist. 4) Will BCRP-knockout (KO) mice be more susceptible to butyrate for its therapeutic potential for colon cancer or other diseases? 5) Which signaling pathways regulate BCRP function and its expression in the colon? 6) Since both the MCT1 and SMCT1 are downregulated in colon cancer, hence limiting the availability of butyrate inside the cells (14, 21), what role will butyrate play by itself or by enhancing the therapeutic potential of other chemotherapeutic drugs through inhibition of their efflux from the cells by BCRP, as discussed above? The answers to many of these questions in future studies utilizing BCRP KO mice as well as molecular approaches utilizing siRNA for BCRP or its overexpression should be important to further establish this novel role of BCRP in butyrate transport and its implications in health and disease.

GRANTS

R. K. Gill's work is supported by National Institute of Diabetes and Digestive and Kidney Diseases Grant DK-74458, and P. K. Dudeja's laboratory is supported by the department of Veterans Affairs and the National Institute of Diabetes and Digestive and Kidney Diseases Grants DK-54016, DK-81858, and P01 DK-067887.

DISCLOSURES

No conflicts of interest, financial or otherwise, are declared by the author(s).

REFERENCES

  • 1. Alrawi SJ , Schiff M , Carroll RE , Dayton M , Gibbs JF , Kulavlat M , Tan D , Berman K , Stoler DL , Anderson GR. Aberrant crypt foci. Anticancer Res 26: 107–119, 2006.
    PubMed | ISI | Google Scholar
  • 2. Anna Rawluszko A , Krokowicz P , Jagodzinski PP. Butyrate induces expression of 17beta-hydroxysteroid dehydrogenase type 1 in HT29 and SW707 colorectal cancer cells. DNA Cell Biol. In press.
    Google Scholar
  • 3. Borthakur A , Gill RK , Hodges K , Ramaswamy K , Hecht G , Dudeja PK. Enteropathogenic Escherischia coli inhibits butyrate uptake by altering the apical membrane MCT1 level. Am J Physiol Gastrointest Liver Physiol 290: G30–G35, 2006.
    Link | ISI | Google Scholar
  • 4. Borthakur A , Anbazhagan AN , Kumar A , Raheja G , Singh V , Ramaswamy K , Dudeja PK. The probiotic Lactobacillus plantarum counteracts TNF-alpha-induced downregulation of SMCT1 expression and function. Am J Physiol Gastrointest Liver Physiol 299: G928–G934, 2010.
    Link | ISI | Google Scholar
  • 5. Canani RB , Costanzo MD , Leone L , Pedata M , Meli R , Calignano A. Potential beneficial effects of butyrate in intestinal and extraintestinal diseases. World J Gastroenterol 17: 1519–1528, 2011.
    Crossref | PubMed | ISI | Google Scholar
  • 6. Canani RB , Terrin G , Cirillo P , Castaldo G , Salvatore F , Cardillo G , Coruzzo A , Troncone R. Butyrate as an effective treatment of congenital chloride diarrhea. Gastroenterology 127: 630–634, 2004.
    Crossref | PubMed | ISI | Google Scholar
  • 7. Chen YX , Fang JY , Lu J , Qiu DK. Regulation of histone acetylation on the expression of cell cycle-associated genes in human colon cancer cell lines. Zhonghua yi xue za zhi 84: 312–317, 2004.
    PubMed | Google Scholar
  • 8. Comalada M , Bailon E , de Haro O , Lara-Villoslada F , Xaus J , Zarzuelo A , Galvez J. The effects of short-chain fatty acids on colon epithelial proliferation and survival depend on the cellular phenotype. J Cancer Res Clin Oncol 132: 487–497, 2006.
    Crossref | PubMed | ISI | Google Scholar
  • 9. Cook SI , Sellin JH. Review article: short chain fatty acids in health and disease. Aliment Pharmacol Ther 12: 499–507, 1998.
    Crossref | PubMed | ISI | Google Scholar
  • 10. Cox MA , Jackson J , Stanton M , Rojas-Triana A , Bober L , Laverty M , Yang X , Zhu F , Liu J , Wang S , Monsma F , Vassileva G , Maguire M , Gustafson E , Bayne M , Chou CC , Lundell D , Jenh CH. Short chain fatty acids act as antiinflammatory mediators by regulating prostaglandin E(2) and cytokines. World J Gastroenterol 15: 5549–5557, 2009.
    Crossref | PubMed | ISI | Google Scholar
  • 11. D'Argenio G , Mazzacca G. Short-chain fatty acid in the human colon. Relation to inflammatory bowel diseases and colon cancer. Adv Exp Med Biol 472: 149–158, 1999.
    Crossref | PubMed | ISI | Google Scholar
  • 12. Dudeja PK , Ramaswamy K. Intestinal Anion Absorption. In: Physiology of the Gastrointestinal Tract. Oxford, UK: Elsevier, 2006, p. 1881–1915.
    Crossref | Google Scholar
  • 13. Fukumoto S , Tatewaki M , Yamada T , Fujimiya M , Mantyh C , Voss M , Eubanks S , Harris M , Pappas TN , Takahashi T. Short-chain fatty acids stimulate colonic transit via intraluminal 5-HT release in rats. Am J Physiol Regul Integr Comp Physiol 284: R1269–R1276, 2003.
    Link | ISI | Google Scholar
  • 14. Ganapathy V , Thangaraju M , Prasad PD. Nutrient transporters in cancer: relevance to Warburg hypothesis and beyond. Pharmacol Therap 121: 29–40, 2009.
    Crossref | PubMed | ISI | Google Scholar
  • 15. Gill RK , Saksena S , Alrefai WA , Sarwar Z , Goldstein JL , Carroll RE , Ramaswamy K , Dudeja PK. Expression and membrane localization of MCT isoforms along the length of the human intestine. Am J Physiol Cell Physiol 289: C846–C852, 2005.
    Link | ISI | Google Scholar
  • 16. Gonçalves P , Gregório I , Martel F. The short-chain fatty acid butyrate is a substrate of breast cancer resistance protein. Am J Physiol Cell Physiol (July 20, 2011). doi:10.1152/ajpcell.00146.2011.
    Google Scholar
  • 17. Greer JB , O'Keefe SJ. Microbial induction of immunity, inflammation, and cancer. Frontiers Physiol 1: 168, 2011.
    Crossref | PubMed | ISI | Google Scholar
  • 18. Gutmann H , Hruz P , Zimmermann C , Beglinger C , Drewe J. Distribution of breast cancer resistance protein (BCRP/ABCG2) mRNA expression along the human GI tract. Biochem Pharmacol 70: 695–699, 2005.
    Crossref | PubMed | ISI | Google Scholar
  • 19. Hu S , Dong TS , Dalal SR , Wu F , Bissonnette M , Kwon JH , Chang EB. The microbe-derived short chain fatty acid butyrate targets miRNA-dependent p21 gene expression in human colon cancer. PLoS One 6: e16221, 2011.
    Crossref | PubMed | ISI | Google Scholar
  • 20. Iwanaga T , Takebe K , Kato I , Karaki S , Kuwahara A. Cellular expression of monocarboxylate transporters (MCT) in the digestive tract of the mouse, rat, and humans, with special reference to slc5a8. Biomed Res (Tokyo) 27: 243–254, 2006.
    Crossref | Google Scholar
  • 21. Lambert DW , Wood IS , Ellis A , Shirazi-Beechey SP. Molecular changes in the expression of human colonic nutrient transporters during the transition from normality to malignancy. Br J Cancer 86: 1262–1269, 2002.
    Crossref | PubMed | ISI | Google Scholar
  • 22. Morrow CS , Nakagawa M , Goldsmith ME , Madden MJ , Cowan KH. Reversible transcriptional activation of mdr1 by sodium butyrate treatment of human colon cancer cells. J Biol Chem 269: 10739–10746, 1994.
    Crossref | PubMed | ISI | Google Scholar
  • 23. Robey RW , Polgar O , Deeken J , To KW , Bates SE. ABCG2: determining its relevance in clinical drug resistance. Cancer Metastasis Rev 26: 39–57, 2007.
    Crossref | PubMed | ISI | Google Scholar
  • 24. Ruemmele FM , Dionne S , Qureshi I , Sarma DS , Levy E , Seidman EG. 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 Differentiation 6: 729–735, 1999.
    Crossref | PubMed | ISI | Google Scholar
  • 25. Ruemmele FM , Schwartz S , Seidman EG , Dionne S , Levy E , Lentze MJ. Butyrate induced Caco-2 cell apoptosis is mediated via the mitochondrial pathway. Gut 52: 94–100, 2003.
    Crossref | PubMed | ISI | Google Scholar
  • 26. Shin HJ , Anzai N , Enomoto A , He X , Kim do K , Endou H , Kanai Y. Novel liver-specific organic anion transporter OAT7 that operates the exchange of sulfate conjugates for short chain fatty acid butyrate. Hepatology 45: 1046–1055, 2007.
    Crossref | PubMed | ISI | Google Scholar
  • 27. Soret R , Chevalier J , De Coppet P , Poupeau G , Derkinderen P , Segain JP , Neunlist M. Short-chain fatty acids regulate the enteric neurons and control gastrointestinal motility in rats. Gastroenterology 138: 1772–1782, 2010.
    Crossref | PubMed | ISI | Google Scholar
  • 28. Tang Y , Chen Y , Jiang H , Nie D. The role of short-chain fatty acids in orchestrating two types of programmed cell death in colon cancer. Autophagy 7: 235–237.
    Crossref | PubMed | ISI | Google Scholar
  • 29. Tang Y , Chen Y , Jiang H , Robbins GT , Nie D. G-protein-coupled receptor for short-chain fatty acids suppresses colon cancer. Int J Cancer 128: 847–856.
    Crossref | PubMed | ISI | Google Scholar
  • 30. Thangaraju M , Cresci GA , Liu K , Ananth S , Gnanaprakasam JP , Browning DD , Mellinger JD , Smith SB , Digby GJ , Lambert NA , Prasad PD , Ganapathy V. GPR109A is a G-protein-coupled receptor for the bacterial fermentation product butyrate and functions as a tumor suppressor in colon. Cancer Res 69: 2826–2832, 2009.
    Crossref | PubMed | ISI | Google Scholar

AUTHOR NOTES

  • Address for reprint requests and other correspondence: P. K. Dudeja, Univ. of Illinois at Chicago, 840 S. Wood St., Chicago, IL 60561 (e-mail: ).