Butyric acid: what is the future for this old substance?

DOI: https://doi.org/10.4414/smw.2012.13596

Paolo Sossai

Summary

In this brief review, we present some data from the literature on butyric acid and some of its more interesting potential uses, especially in the field of gastroenterology. Due to its principal characteristics, butyric acid is primarily used for pathologies of the colon (functional, inflammatory). Although only preliminary data are available, butyric acid may also have interesting extraintestinal applications, such as in the treatment of haematological, metabolic, and neurological pathologies.

Introduction

Butyric acid (BA) is a carboxylic acid with the formula CH3-CH2-CH2-COOH. It is frequently used in the veterinary field, especially in ruminant animals. Together with other short-chain fatty acids (propionic acid and acetic acid), BA is the principal source of energy produced by ruminal fermentation of cellulose and starch. In the field of zootechnics, butyric acid is used to improve the growth of bovine animals [1]. In humans, BA is synthesised by the colonic microflora (microbiota) during fermentation of digestible fiber, such as cereal flour, inulin, and psyllium [2].

In humans, the effects of BA can be subdivided into intestinal and extraintestinal. Intestinal effects include: regulating transepithelial transport, improving the inflammatory and oxidative states of the intestinal mucosa, reinforcing the mucosal barrier, modulating visceral sensitivity and motility, and preventing and inhibiting colon carcinoma. Extraintestinal effects are less well known; they have been studied in vitro and in animal models and sometimes even in humans. Currently investigated effects include: haemoglobinopathies, hypercholesterolaemia, reducing resistance to insulin (in animal studies), and reducing ischemic stroke (in animal studies).

Intestinal effects

The intestinal ecosystem is comprised of epithelium, immune system cells, enteric neurons, microbiota, and prebiotics. Our knowledge of the intestinal microbiota remains quite limited due to the difficulty of identifying the numerous bacterial strains present [3]. The human intestine hosts a large quantity of bacteria (approximately 1013–1014) and could thus be termed a “superorgan” as the human body is comprised of approximately 1014 cells [4, 5].

BA is one of the short-chain fatty acids produced by the colon, in particular, by the proximal colonic microbiota and constitutes one of the primary sources of energy for colonocytes [2]. It enhances the absorptive and antisecretive capabilities of the intestinal mucosa. Clausen et al. [6] demonstrated that some cases of antibiotic-associated diarrhea are related to inhibited bacterial fermentation to short-chain fatty acids, leading to reduced absorption of water and sodium. BA has also been shown to stimulate the production of mucin – a specific defense of intestinal mucosa – through increased expression of mucin genes, such as MUC2 [2].

BA has a double effect on cell growth, defined as the “butyrate paradox”; it stimulates physiological proliferation of normal enterocytes, whereas it inhibits cell proliferation in a colon carcinoma cell line in vitro by enhancing histone acetylation [3]. The possible preventive effects of BA acid are attractive, especially considering the adverse effects of drugs used to prevent colorectal cancer, such as aspirin and cyclooxygenase type 2 (COX 2) inhibitors. The administration of aspirin and nonsteroidal anti-inflammatory drugs (NSAIDs), such as celecoxib, has resulted in significant gastrointestinal and cardiovascular toxicity. Furthermore, COX2 inhibitors are very expensive. These issues with other colorectal cancer preventative measures make it very appealing to think that BA could be used in high-risk patients (those with familial adenomatous polyps or hereditary non-polyposis colorectal cancer) and those over age 60 (who are at higher risk due to age) [7].

BA has anti-inflammatory effects through inhibition of NF-κB, which reduces the expressions of cytokine genes, including TNFα, IL1β, IL2, IL6, IL8, and IL12. Clinical experiences indicate that antioxidant activity of BA reduces reactive oxygen species and increases the amount of reduced glutathione [8]. BA’s anti-inflammatory action has been the subject of some interesting studies regarding its use in inflammatory bowel diseases (IBD) as well as in radiation proctitis. Almost all available data is derived from small studies on ulcerative colitis, where it has been administered primarily by enema (table 1) [9–16]. Sodium butyrate is usually the form administered in conjunction with mesalazine for mild to moderate IBD; however, Assisi et al. utilised a slow-release preparation of calcium butyrate and inulin (NMX formulation) [9]. The majority of reported studies performed post-treatment evaluation using clinical, endoscopic, and histologic data, but Assisi et al. [9] and Steinhart et al. [15] used only a clinical score (ulcerative colitis disease activity index; UDCAI).

The studies in table 1 have several limitations, including the fact that not all are randomised, the small numbers of patients studied, the different methods of administering BA (oral/enema), and the different criteria for endpoint evaluation. It should also be noted that in the randomised studies reported in table 1, the number of patients treated with BA is only 50% of the total number of patients treated, except for in the cross-over study by Scheppach et al. [16].

One interesting application of BA involves its use in irritable bowel syndrome (IBS), of which knowledge is still fragmentary, especially regarding the enteric nervous system and its regulation. IBS has a prevalence of about 11.5% in the European population and therefore has significant impact on healthcare [17]. Preliminary in vitro and animal studies demonstrate activity of BA on colonic motility through choline acetyltransferase [1], but there is limited data in humans. Vanhoutvin et al. [18] administered sodium butyrate as enema versus placebo to assess pain, sensation of urgent defecation, and discomfort as measured by a barostat in 11 healthy volunteers (3 males and 8 females). The authors conclude that butyrate administered as enema (especially in doses of 100 mmol/L) increases the pressure threshold before causing pain or discomfort – although the study did not report the effect of patient age, and the results only refer to healthy subjects. In contrast, studies in animals (rats) show visceral hypersensitivity to butyrate enemas; thus, there is a need for randomised, controlled studies measuring the impact of BA on IBS [19].

One pharmaceutical technical problem regarding the use of butyrate is that most commercially available formulations do not have standardised release. The study by Assisi et al. [9] used an oral formulation that releases BA in the form of calcium butyrate in the colon, which permitted a more practical (oral) and standardised administration.

This brief overview illustrates the favourable effects of BA that have been observed in the clinic. The etiopathogenetic mechanisms are not yet well understood. Although there are a few reports in the literature of the use of BA in IBD, new clinical studies are needed to confirm these data and to clarify its use for other conditions, such as IBS and cancer prevention. The possibility of using BA to prevent colorectal cancer is obviously of great clinical interest and merits systematic investigation.

Table 1
Author Year N. Pts. Randomised U.C./ Crohn Enema/ oral Mesalazine/ Sulfasalazine Duration Butyrate dose Outcome
Assisi RF(9) 2008 216 No U.C. Oral Yes 6 months 921 mg/day 82.4% (improved)
Di Sabotino A(10) 2005 13 No Crohn Oral Yes 8 weeks 4 gr/day 69% (improved)
Vernia P(11) 2003 51 Yes U.C. Enema Yes 6 weeks 160 mmol/day S.
Vernia P(12) 2000 30 Yes U.C. Oral Yes 6 weeks 4 gr/day N.S.
Steinhart AH(13) 1996 38 Yes U.C. Enema Yes (28/38) 6 weeks 80 mmol/day N.S.
Vernia P(14) 1995 40 Yes U.C. Enema Yes 6 weeks 200 ml/day (mixture) S.
Steinhart AH(15) 1994 10 No U.C. Enema Yes 6 weeks 80 mmol/day 60% (improved)
Scheppach W(16) 1992 10 Yes U.C. Enema Yes (3/10) 2 weeks 100 mmol/day S.
N.S. = not aignificant; S. = significant

Extraintestinal effects

BA increases the production of fetal haemoglobin (HbF) in β-thalassemia patients and in those with sickle cell anaemia, reducing ineffective erythropoiesis. Generally speaking, an increase in HbF reduces the α/non-α chain imbalance, thus improving the anaemia. BA is a histone deacetylase inhibitor that, in vitro, stimulates synthesis of the γ haemoglobin chains and sometimes of α chains; this occurs in a different manner, according to the type of disease (β-thalassemia or sickle cell anaemia) [20]. In 2011, Perrine et al. published a phase I, randomised, double-blind study that tested the safety and blood effects (reticulocytes and HbF assays) of a BA derivative (sodium 2,2-dimethylbutyrate) given orally in healthy volunteers. The results are quite encouraging in terms of safety and statistically significant increases in reticulocytes, indicating favourable prospects for use in patients with β-thalassemia and sickle cell disease [21, 22].

BA can also function as an ammonia scavenger in patients with enzymatic deficit of the urea cycle. These patients have an ornithine transcarbamylase (mitochondrial enzyme) deficiency, which develops at different ages, in both newborns and adults, and involves predominantly neurological symptoms that range from headache to coma [23, 24]. Ornithine transcarbamylase is expressed in the liver and intestine, and is encoded by a gene located on chromosome Xp21.1. The initial diagnosis of deficiency is based on hyperammonaemia, and can later be confirmed by testing of amino acids, such as citrulline (lower) and glutamine and alanine (higher). BA acts on the conjugation of glutamine and successive excretion in urine. Burlina et al. administered sodium phenylbutyrate (median dose of 352 mg/kg/day in three to four divided doses) to 9 patients with ornithine transcarbamylase deficit, and followed them for 26 months. During the study, no episodes of hyperammonaemia requiring hospitalisation occurred, it was possible to increase the patients’ intake of protein, and no side effects were attributable to the therapy [25].

At another metabolic level, BA can down-regulate the expression of genes involved in the biosynthesis of cholesterol and triglycerides and modulate apolipoprotein biogenesis and lipoprotein assembly [26]. Metabolic activity of short-chain fatty acids (including BA) is mediated by individual intestinal microbiota. In both animals and humans, increasing evidence is emerging to support the relationship between obesity and type of intestinal microbiota [4, 27]. Sometimes the published articles present conflicting results. Obese individuals have a lower quantity of intestinal Bacteroides species and more Firmicutesbut some authors observed more Bacteroides in overweight and obese subjects [28]. BA binds receptors, including protein-coupled receptors (e.g., Gpr41), that are present on intestinal endocrine cell surfaces, and stimulate these cells to produce peptide YY, which in turn inhibits gut motility and increases satiety [29, 30].

Gpr41 is expressed also in adipose tissue and short-chain fatty acids stimulate leptin production both in cultured adipocytes and in the whole animal [31]. Animal studies demonstrate that BA and propionate can reduce obesity and insulin resistance in Gpr41-deficient mice with independent mechanism [32]. Furthermore, studies in animals (rats) showed that sodium butyrate can stimulate neuronal proliferation in zones that have undergone cerebral ischemia [33].

Conclusions

In conclusion, BA is a substance with interesting possibilities for gastroenterological therapies (e.g., IBD, IBS, and colorectal cancer) as well as for haematological diseases (e.g., β-thalassemia), metabolic diseases (e.g., enzymatic deficit urea cycle and obesity), and vascular stroke. Since BA cannot be patented, it is of little interest to the pharmaceutical industry except in regards to developing different types of release formulations, as has been done in the veterinary field.

References

  1 Gorka P, Kowalski ZM, Pietrzak P, Kotunia A, Jagusiak W, Holst JJ, et al. Effect of method of delivery of sodium butyrate on rumen development in newborn calves. J Diary Sci. 2011;94:5578–88.

  2 Berni Canani R, Di Costanzo M, Leone L, Pedata M, Meli R, Calignano A. Potential beneficial effects of butyrate in intestinal and extraintestinal disease. World J Gastroenterol. 2011;17:1519–28.

  3 Scheppach W, Weiler F. The butyrate story: old wine in new bottles? Curr Opin Clin Nutr Metab Care. 2004;7:563–7.

  4 Cani PD, Delzenne NM. The role of the gut microbiota in energy metabolism and metabolic disease. Curr Pharm Des. 2009;15:1546–58.

  5 Lederberg J. Infectious history. Science. 2000;288:287–93.

  6 Clausen MR, Bonnen H, Tvede M, Mortensen PB. Colonic fermentation to short-chain fatty acids is decreased in antibiotic-associated diarrhea. Gastroenterology. 1991;101:1497–504.

  7 Cooper K, Squires H, Carroll C, Papaioannou, Booth A, Logan RF, et al. Chemoprevention of colorectal cancer: systematic review and economic evaluation. Health Technology Assessment. 2010;14:1–206.

  8 Hamer HM, Jonkers DMAE, Vanhoutvin SALW, Troost FJ, Rijkers G, de Bruine A, et al. Effect of butyrate enemas on inflammation and antioxidant status in the colonic mucosa of patients with ulcerative colitis in remission. Clinical Nutrition. 2010;29:738–44.

  9 Assisi RF and GISDI Study Group. Combined butyric acid/mesalazine treatment in ulcerative colitis with mild-moderate activity. Minerva Gastroenterol Dietol. 2008;54:231–8.

10 Di Sabotino A, Morera R, Ciccocioppo R, Cazzola P, Gotti S, Tinozzi FP, et al. Oral butyrate for mild to moderately active Crohn’s disease. Aliment Pharmacol Ther. 2005;22:789–94.

11 Vernia P, Annese V, Bresci G, d’Albasio G, D’Incà R, Giaccari S, et al. Topical butyrate improbe efficacy of 5-ASA in refractory distal ulcerative colitis: results of a multicentre trial. Eur J Clin Invest. 2003;33:244–8.

12 Vernia P, Monteleone G, Grandinetti G, Villotti G, Di Giulio E, Frieri G, et al. Combined oral sodium butyrate and mesalazine treatment compared to oral mesalazine alone in ulcerative colitis. Randomized, double-blind, placebo-controlled pilot study. Dig Dis Sci. 2000;45:976–81.

13 Steinhart AH, Hiruki T, Brzezinski A, Baker JP. Treatment of left-sided ulcerative colitis with butyrate enemas: a controlled trial. Aliment Pharmacol Ther. 1996;10:729–36.

14 Vernia P, Marcheggiano A, Caprilli R, Frieri G, Corrao G, Valpini D, et al. Short-chain fatty acid topical treatment in distal ulcerative colitis. Aliment Pharmacol Ther. 1995;9:309–13.

15 Steinhart AH, Brzezinski A, Baker JP. Treatment of refractory ulcerative proctosigmoiditis with butyrate enemas. Am J Gastroenterol. 1994;89:179–83.

16 Scheppach W, Sommer H, Kirchner T, Paganelli GM, Bartram P, Christl S, et al. Effect of butyrate enemas on the colonic mucosa in distal ulcerative colitis. Gastroenterology. 1992;103:51–6.

17 Hungin AP, Whorwell PJ, Tack J, Mearin F. The prevalence, patterns and impact of irritable bowel syndrome: an international survey of 40,000 subjects. Aliment Pharmacol Ther. 2003;17:643–50.

18 Vanhoutvin SALW, Troost FJ, Kilkens TOC, Lindsey PJ, Hamer HM, Jonkers DMAE, et al. The effects of butyrate enemas on visceral perception in healthy volunteers. Neurogastroenterol Motil. 2009;21:952–e76.

19 Kannampalli P, Shaker R , Sengupta JN. Colonic butyrate- algesic or analgesic? Neurogastroenterol Motil. 2011;23:975–9.

20 Fathallah H, Taher A, Bazarbachi A, Atweh GF. Differences in response to fetal hemoglobin induction therapy in β-thalassemia and sickle cell disease. Blood Cells Mol Dis. 2009;43:58–62.

21 Perrine SP, Castaneda SA, Chui DHK, Faller DV, Berenson RJ, Siritanaratku N, et al. Fetal globin inducers: novel agents and new potential. Ann NY Acad Sci. 2010;1202:158–64.

22 Perrine SP, Wargin WA, Boosalis MS, Wallis WJ, Case S, Keefer JR, et al. Evaluation of safety and pharmacokinetics of sodium 2,2 dimethylbutyrate, a novel short chain fatty acid derivative, in a phase1, double-blind, placebo-controlled, single-dose, and repeat-dose studies in healthy volunteers. J Clin Pharmacol. 2011;51:1186–94.

23 Gordon N. Ornithine transcarbamylase deficiency: a urea cycle defect. Eur J Paediatr Neurol. 2003;7:115–21.

24 Walker V. Ammonia toxicity and its prevention in inherited defects of the urea cycle. Diabetes Obes Metab. 2009;11:823–35.

25 Burlina AB, Ogier H, Korall H, Trefz FK. Long-term treatment with sodium phenylbutyrate in ornithine transcarbamylase-deficient patients. Mol Genet Metab. 2001;72:351–5.

26 Marcil V, Delvin E, Seidman E, Poitras L, Zoltowska M, Garofalo C, et al. Modulation of lipid synthesis, apolipoprotein biogenesis, and lipoprotein assembly by butirate. Am J Physiol Gastrointest Liver Physiol. 2002;283:G340–G346.

27 Vrieze A, Holleman F, Zoetendal EG, deVos WM, Hoekstra JBL, Nieuwdorp M. The environment within: how gut microbiota may influence metabolism and body composition. Diabetologia. 2010;53:606–13.

28 Schwiertz A, Taras D, Schäfer K, Beijer S, Bos NA, Donus C, et al. Microbiota and SCFA in lean and overweight healthy subject. Obesity. 2009;18:190–5.

29 Samuel BS, Shaito A, Motoike T, Rey FE, Backhed F, Manchester JK, et al. Effects of the gut microbiota on host adiposity are modulated by the short-chain fatty-acid binding G protein-coupled receptor, Gpr41. Proc Natl Acad Sci USA 2008;105:16767–72.

30 Kootte RS, Vrieze A, Holleman F, Dallinga-Thie GM, Zoetendal EG, de Vos WM, et al. The therapeutic potential of manipulating gut microbiota in obesity and type 2 diabetes mellitus. Diabetes, Obesity and Metabolism. 2012;14:112–20.

31 Xiong Y, Miyamoto N, Shibata K, Valasek MA, Motoike T, Kedzierski RM, et al. Short-chain fatty acids stimulate leptin production in adypocites through the G protein-coupled receptor GPR41. Proc Natl Acad Sci USA 2004;101:1045–50.

32 Lin HV, Frassetto A, Kowalik EJ Jr, Nawrocki AR, Lu MM, Kosinsky JR, et al. Butyrate and propionate protect against diet-induced obesity and regulate gut hormones via free fatty acid receptor 3-indipendent mechanisms. PloS One. 2012;7:e35240.

33 Kim HJ, Leeds P, De-Maw Chuang. The HDCA inhibitor, sodium butyrate, stimulates neurogenesis in the ischemic brain. J Neurochem. 2009;110:1226–40.

Notes

Funding / potential competing interests: No financial support and no other potential conflict of interest relevant to this article was reported.