Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2015 Feb 4.
Published in final edited form as: Dig Dis Sci. 2013 Jul 10;58(10):2756–2766. doi: 10.1007/s10620-013-2744-4

Pathobiology and Potential Therapeutic Value of Intestinal Short-Chain Fatty Acids in Gut Inflammation and Obesity

Jessica Soldavini 1, Jonathan D Kaunitz 2,3,4,5,6,7,
PMCID: PMC4317286  NIHMSID: NIHMS504066  PMID: 23839339

Abstract

Background

The lumen of the gastrointestinal tract contains many substances produced from the breakdown of foodstuffs, from salivary, esophageal, intestinal, hepatic, and pancreatic secretions, and from sloughed cells present in the gastrointestinal lumen. Although these substances were traditionally regarded as waste products, there is increasing realization that many can be biologically active, either as signalling compounds or as nutrients. For example, proteins are broken down into amino acids, which are then sensed by nutrient receptors. The gut microbiome, which is at highest abundance in the ileocecum, has powerful metabolic activity, digesting and breaking down unabsorbed carbohydrates, proteins, and other ingested nutrients into phenols, amines, volatile organic compounds, methane, carbon dioxide, hydrogen, and hydrogen sulfide into volatile fatty acids, also called short-chain fatty acids (SCFAs).

Conclusion

These latter substances are the topic of this review. In this review, we will briefly discuss recent advances in the understanding SCFA production, signalling, and absorption, followed by a detailed description and discussion of trials of SCFAs, probiotics, and prebiotics in the treatment of gastrointestinal disease, in particular ulcerative colitis (UC), pouchitis, short bowel syndrome, and obesity.

Keywords: Probiotics, Short-chain fatty acids, Prebiotics, Synbiotics, Obesity, Colitis

Introduction

The lumen of the gastrointestinal tract contains many substances produced from the breakdown of foodstuffs, from salivary, esophageal, intestinal, hepatic, and pancreatic secretions, and from sloughed cells present in the gastrointestinal lumen. Although these substances were traditionally regarded as waste products, there is increasing realization that many can be biologically active, either as signalling compounds or as nutrients. For example, proteins are broken down into amino acids, which are then sensed by nutrient receptors. The gut microbiome, which is at highest abundance in the ileocecum, has powerful metabolic activity, digesting and breaking down unabsorbed carbohydrates, proteins, and other ingested nutrients into phenols, amines, volatile organic compounds, methane, carbon dioxide, hydrogen, and hydrogen sulfide into volatile fatty acids, also called short-chain fatty acids (SCFAs) [1-3]. These latter substances are the topic of this review.

In this review, we will briefly discuss recent advances in the understanding of SCFA production, signalling, and absorption, followed by a detailed description and discussion of trials of SCFAs, probiotics, and prebiotics in the treatment of gastrointestinal disease, in particular ulcerative colitis (UC), diversion colitis, pouchitis, short bowel syndrome, and obesity.

Short Chain Fatty Acids

SCFA are defined as 1–6 carbon volatile fatty acids existing in straight- and branched-chain conformations. Common SCFAs include formic, acetic, proprionic, butyric, isobutyric, valeric, isovaleric, and caproic acids [4, 5]. We will focus on SCFA production, absorption by the intestinal mucosa, and function, with discussion of selected disease entities, such as inflammatory bowel disease, diversion colitis, pouchitis, short bowel syndrome, and obesity which relate to SCFA production or may be ameliorated by SCFA therapy. In this fashion, we hope to inform the reader on their importance for the maintenance of mucosal integrity, their nutritional value, and also about their use in difficult-to-treat clinical situations.

Short Chain Fatty Acids in the Colon

Acetate, propionate, and butyrate constitute 90–95 % of the SCFA present in the colon [1]. The approximate molar ratios of acetate:propionate:butyrate are 60:20:20. Throughout the different regions of the colon, although absolute SCFA concentrations may vary, molar ratios appear to remain constant. Concentrations of SCFA are 40 % lower in the left than in right colon and are highest in the cecum and ascending colon [6]. In autopsies, Cummings et al. reported total SCFA concentrations of 123 ± 12 mmol/kg (ascending), 117 ± 9 mmol/kg (transverse), 80 ± 17 mmol/kg (descending) and 100 ± 30 mmol/kg (sigmoid and rectum), which agrees with the relative ratios found in another autopsy study by Macfarlane et al. [6, 7]. The pKa of SCFA is ~4.8, qualifying them as weak acids. Because the colonic pH is 6.0–7.5, 99 % of SCFA are anionic (unprotonated) in this pH range [1].

Short Chain Fatty Acid Production

A variety of substrates may be metabolized by the intestinal microbiome, including resistant starch, dietary fiber, simple sugars, sugar alcohols, sloughed off epithelial cells, mucus, intestinal enzymes and other secretions, and unabsorbed or undigested proteins [1, 2, 8]. Resistant starch refers to starch and products of starch degradation that enter the colon after resisting digestion in the small intestine [3]. In humans, estimates of the amount of dietary starch entering the large intestine range from 5 to 20 % [9]. Fecal SCFA concentrations in humans increase in response to ingestion of resistant starch, which are accordingly considered prebiotics [3], defined as indigestible fermentable fibers that have beneficial effects on health and well being by altering the composition or activity of the gut microbiota [10]. Other prebiotics include fructo-oligosaccharides (FOS), galacto-oligosaccharides (GOS), lactulose, and inulin [10].

Differences in the rate and ratio of SCFA production depend primarily on the type of substrate as opposed to the composition of the intestinal microbiota [1]. Englyst et al. reported that starch and pectin were degraded faster than xylan and arabinogalactan. The study also reported molar ratios of acetate, propionate and butyrate to be 50:22:29, 50:42:8, 82:15:3, and 84:14:2 from the fermentation of starch arabinogalactan, xylan and pectin, respectively [11]. Inulin and oligofructose produce low levels of butyrate, with oligofructose predominately producing acetate [12]. Branched-chain fatty acids are produced from proteins rather than from carbohydrates [1]. Substrates have varying levels of fermentability, with soluble fibers fermented more completely than insoluble fibers [9]. In vitro, 97 % of pectin, 6–7 % of cellulose, <50 % of wheat bran, and 20–50 % of psyllium are fermentable. Highly fermentable substrates produce greater amounts of SCFA [3].

Short Chain Fatty Acid Absorption

The colon absorbs more than 95 % of SCFA [3]. Although the mechanism of SCFA absorption is not entirely understood, several possibilities have been proposed. In the colon, SCFA absorption is coupled to HCO3 secretion [13, 14]. Evidence has been reported for apical SCFA/HCO3 in colonocytes [14-16], which is electroneutral with a coupling ratio of 1:1 [16]. The monocarboxylate transporter 1 (MCT1), which belongs to the solute carrier gene family SLC16 [17], is another proposed plasma membrane SCFA transport protein, expressed primarily in colonic epithelial cells [18-20]. MCT1 co-transports H+ with monocarboxylic acids, such as SCFA, with an H+:monocarboxylic acid ratio of 1:1 [17, 21]. Since this process is electroneutral, membrane potential is not affected, with the transmembrane SCFA concentration gradient driving SCFA to enter colonocytes from the lumen [21].

The SLC5A8 gene encodes another transporter with a similar functional identity to MCT1, although they do not share genetic similarity. While they both transport monocarboxylic acids, including SCFA, the transporter encoded by SLC5A8 is coupled to Na+ as opposed to H+, referred to as the sodium-coupled monocarboxylate transporter 1 (SMCT1) [21]. SMCT1 expression was reported on the apical membrane of human colonic epithelial cells [22, 23]. Unlike the electroneutral transport involved with MCT1, transport via SMCT1 is electrogenic with the stoichiometry of Na+:monocarboxylic acid varying from 2:1 to 4:1 depending on the substrate [17, 24-26]. SMCT1 has a fairly high affinity for monocarboxylates, with affinity for butyrate >propionate > l-lactate > d-lactate > acetate [25].

Short Chain Fatty Acid Chemosensing

In the wake of the Human Genome Project and other large-scale gene sequencing efforts, thousands of gene sequences that corresponded to known protein families were identified that had no known function. These “orphans” were ascribed function in many cases via subsequent experimentation through the process of “de-orphanization.” One of the largest protein families is the 7 transmembrane (7TM) receptor family, also termed G-protein coupled receptors (GPCRs), which include most of the well-described receptors for neurotransmitters, hormones, and drugs, in addition to other endogenous and exogenous ligands. About ten of the de-orphanized GPCRs have been identified as nutrient receptors, and of those, five have been identified as fat receptors, more specifically, for long chain, medium chain, and SCFAs. The two SCFA GPCRs, termed GPR43 [free fatty acid receptor2 (FFAR2) and GPR41 (FFAR3)] were formally described in 2008 [27] and only recently have been attributed with intestinal chemosensing function, such as in mediating the release of the incretin glucagon-like peptide 1 (GLP-1) from colonic enteroendocrine L-cells [28]. Expression of these SCFA sensing receptors on the apical membrane of L-cells, expressed in particular in intestinal segments exposed to high SCFA concentrations such as the ileocecum and the bovine rumen [29, 30], suggests a plausible mechanism in which luminal SCFAs signal hormonal responses.

Functions of Short Chain Fatty Acids

SCFA produced in the colon contribute approximately 5–10 % towards human energy requirements [31], presumably through their uptake by specific apical solute carriers such as SMCT1 with subsequent hepatic metabolism to CO2, the tricarboxylic acid cycle and other metabolic pathways. In the colon, preference for oxidation is butyrate > acetate > propionate, glucose and glutamine [32]. By providing additional calories, SCFA maintain energy homeostasis, in particular with loss of small intestinal surface area or function, with resultant insufficient capability to absorb adequate nutrients [21]. SCFA affect cell proliferation and differentiation and can evoke hormone release [1, 33-36]. In colon cancer cell lines, SCFAs induce apoptosis [37-39]. SCFA function as histone deacetylase (HDAC) inhibitors, which are pro-differentiation, pro-apoptosis, and can induce cycle growth arrest in cancer cells [40].

SCFA also modulate inflammation and can affect several leukocyte functions. They suppress production of pro-inflammatory mediators such as TNF-α, IL-6 and NO. Butyrate can enhance the release of the anti-inflammatory cytokine IL-10. SCFA are involved with leukocyte chemotaxis, affecting migration to inflammatory sites. The anti-inflammatory effects of SCFA may be related to the activation of their cognate G protein-coupled receptors GPR41 and GPR43 in addition to their function as HDAC inhibitors [41].

Disease States Linked with SCFAS

Several intestinal diseases, in particular those of mucosal inflammation, are thought to result from SCFA dysbiosis. Accordingly, therapies have been proposed to restore SCFA production or to provide exogenous SCFAs in an attempt to reduce inflammation.

Ulcerative Colitis

Ulcerative colitis (UC) is a type of inflammatory bowel disease (IBD) that predominantly affects the colon. In patients with UC, bloody diarrhea and abdominal cramping often occur [42, 43]. Although the pathogenesis of UC is incompletely understood, a combination of factors including genetics, abnormal intestinal microflora, an anomalous immune response to intestinal bacteria, diet, and intestinal barrier dysfunction are thought to contribute [10]. Patients with IBD have an altered composition of the intestinal microbiota compared to healthy individuals [44-46]. Differences in the intestinal microbiota have also been reported between active and inactive IBD and within patients when comparing mucosal samples between inflamed with non-inflamed mucosa [45, 46]. Nuclear factor kappa B (NF-κB) may also contribute to UC pathogenesis. Genes induced during inflammation of the mucosa, such as TNF-α, IL-1β, IL-6, and IL-8, are regulated by NF-κB whereas products of some of these genes can activate NF-κB [47].

The standard treatment for mild UC is 5-aminosalicylic acid (5-ASA), also known as mesalamine, which can be given topically, either orally or rectally. Corticosteroids, such as prednisone, or other immunomodulators such as thiopurines or cytokine blocking antibodies are recommended for patients who do not respond to mesalamine [42]. Some data, however, suggests that altering the gut microbiome may have a salutary effect on IBD. For example, in UC patients, decreased fecal concentrations of butyrate and propionate have been reported [44]. Impaired butyrate oxidation was also reported in patients with UC [48, 49].

In order to restore the hypothesized microbial dysbiosis associated with UC, several therapies have been proposed which are directed at either restoring “beneficial” flora, generating endogenous SCFAs, or providing exogenous SCFAs.

Endogenous Short Chain Fatty Acids: Probiotics and Prebiotics

Probiotics, defined as ingestible bacterial cultures which can survive transit through the GI tract, are believed to promote human health. Prebiotics, which are believed to increase the preponderance of beneficial bacteria by supplying specific nutrients, increase cecal and fecal concentrations of SCFA [50-58]. Several human studies have reported increases in fecal SCFA after probiotic or prebiotic supplementation [54-58]. In one study, patients with active mild to moderate UC were randomized to receive either a bifidobacteria-fermented milk (BFM) or placebo for 12 weeks. Of the 18 patients whose fecal SCFA acid concentrations were collected and analyzed at the end of the 12 weeks, participants in the BFM group experienced significant increases in fecal total SCFA, butyrate, and propionate concentrations over baseline, whereas the placebo group did not [54]. Probiotics and prebiotics can alter intestinal microbiota while having beneficial effects for the host. Probiotics are living microorganisms whereas prebiotics are usually indigestible fermentable fibers. Synbiotics are a combination of probiotics and prebiotics [10]. While prebiotics may benefit animal models of colitis, results remain inconclusive, with effects dependant on the type of prebiotics and model of colitis used [10, 50, 51, 53, 59-69]. Similarly, the beneficial effects of probiotics in colitis models are strain dependent [52, 53, 70-73]. Very few studies of prebiotics and synbiotics in patients with IBD have been carried out in humans. While some of these studies report reduced inflammation and other protective indicators, small sample sizes and a short treatment duration impair interpretability, with inconclusive data supporting the claimed benefits for UC patients [74-76].

A recent Cochrane publication reviewed the efficacy of probiotics in the treatment of UC. Only four studies that met inclusion criteria were included in the review. These studies included between 32 and 327 patients and were carried out for 3–12 months. Probiotics were compared to mesalamine in three of the studies and to placebo in the fourth. The authors reported no statistically significant differences in the maintenance of remission of probiotics versus mesalamine or placebo. In a pooled analysis of the three studies examining probiotics versus mesalamine, 40.1 % of patients in the probiotic group and 34.1 % of patients in the mesalamine group reported relapse (OR 1.33, 95 % CI 0.94–1.90). In a study of 32 patients, 75 % versus 92 % of patients experienced relapse in the probiotic versus placebo group, respectively (OR 0.27, 95 % CI 0.03–2.68). The authors concluded that there was insufficient evidence to support the efficacy of probiotics in ulcerative colitis for the maintenance of remission [77].

Another Cochrane review evaluated publications designed to assess the efficacy of probiotics for inducing remission in UC. Perhaps due to stringent quality criteria, only four studies again were included. No statistically significant differences were present between the probiotic and the comparison groups. Like the evidence for the use of probiotics in the maintenance of remission in UC, there is insufficient evidence to recommend the use of probiotics in UC for inducing remission [78].

Being a live material and nonstandardized, comparison among probiotic preparations can be difficult. A recent meta-analysis evaluated the use of probiotics in UC according to strain and UC activity. In patients with inactive UC, E. coli Nissle 1917 was comparable to mesalamine in preventing relapse. Promising evidence was also presented supporting the induction of remission with the commercial probiotic preparation VSL#3—a proprietary mixture containing viable lyophilized bacteria of four strains of lactobacillus (L. casei, L. plantarum, L. acidophilus, and L. delbrueckii subsp. bulgaricus), three strains of bifidobacterium (B. longum, B. breve, and B. infantis), and one strain of Streptococcus salivarius subsp in patients with mild to moderately active UC. Interpretation of these studies is however limited by small sample size, high dropout rates, and inadequate controls [43].

Exogenous Short Chain Fatty Acids

Exogenous SCFA administration has been studied in experimental models of colitis in addition to small clinical trials.

Experimental UC Models

A study of male Wistar rats with 2,4,6-trinitrobenzene sulfonic acid (TNBS)-induced colitis reported clinical and functional recovery in rats treated with butyrate enemas. Compared to rats that were untreated or treated with saline enemas, rats in the butyrate group had improvements in inflammation, diarrhea, colonic damage score, tissue myeloperoxidase (MPO) activity, and electrolyte absorption [79]. In male Wistar rats with TNBS-induced colitis, rats treated with sodium butyrate enemas, 5-ASA, and a combination of sodium butyrate and 5-ASA all showed improvements in diarrhea, colonic damage score, and MPO activity. These improvements were accompanied by markers of mucosal inflammation and integrity such as increased trefoil factor3 (TFF3) mRNA expression, which is believed to stabilize colonic mucus, decreased production of the serum cytokine IL1β and tissue expression of the pro-inflammatory protein NF-κB, with the greatest effects in the combination treatment group [80]. Not all studies of SCFA enemas in experimental colitis have shown benefit. For example, bowel thickness and lesion scores increased in Sprague–Dawley rats with TNBS-induced colitis following administration of SCFA enemas compared to saline enemas [81].

Uncontrolled Trials

Small, uncontrolled clinical studies dating from the 1990s have reported beneficial effects of SCFA on UC (Table 1). Breuer et al. administered 100 ml SCFA (80 mM sodium acetate, 30 mM sodium propionate, 40 mM sodium butyrate) intra-rectally twice a day for 6 weeks in 12 patients with active UC. Nine out of the ten subjects who completed the trial were at least “much improved” by trial’s end. Significant improvements were reported for stool frequency, rectal bleeding, and lifestyle impact; improvements in endoscopic mucosal appearance, erosions, and exudates, with decreased frequency of crypt abscesses and mucin depletion were also reported [82]. In ten patients with ulcerative proctosigmoiditis, 60 ml of 80 mM sodium butyrate was administered nightly for 6 weeks. Six of these patients responded with a fall in the ulcerative colitis disease activity index (UCDAI) score by 3 or more points, with four judged to have a complete response, with a UCDAI of 3 or less at the end of the trial [83]. Nine patients with distal UC received an enema twice a day of 100 ml 80 mM sodium butyrate plus 2 g 5-ASA for four weeks. Seven of the nine patients had significant improvement in clinical scores and decreases of reported blood in stool, stool frequency and endoscopic scores [84].

Table 1.

Uncontrolled trials of SCFA and colitis

Patients, n Treatment Results Reference
12 SCFA enema Improvements in stool frequency, rectal bleeding, mucosal appearance, lifestyle impact, erosions exudate, cryptitis abscesses, and mucin depletion [82]
10 NaB enema Six out of ten patients responded to treatment and four of these had complete response [83]
9 5-ASA and NaB Improvements in clinical scores and decreases in blood in stool, number of bowel movements, and endoscopic scores [84]
216 Butyrate and inulin tablets Improvement in UCDAI scores [85]

SCFA short-chain fatty acids, UCDAI ulcerative colitis disease activity index

Controlled Studies

Several controlled studies, listed in Table 2, address the administration of exogenous SCFAs on UC. A double-blind placebo-controlled trial by Scheppach et al. randomized 47 patients with distal UC to receive a mixed SCFA enema (60 mM sodium acetate, 30 mM sodium propionate, 40 mM sodium acetate), 100 mM sodium butyrate alone, or a saline placebo twice a day for 8 weeks. UCDAI decreased significantly in all groups with no significant differences between groups. Remission was 47 % for SCFA, 38 % for butyrate, and 25 % for placebo [86]. Breuer et al. randomized 103 patients with distal UC to receive an enema of mixed SCFA (80 mM sodium acetate, 30 mM sodium propionate, 40 mM sodium butyrate) or saline for 6 weeks. While more patients receiving the SCFA enema showed improvement, the results were not statistically significant, possibly related to poor compliance. Every patient in the SCFA group who improved, compared to 37 % who did not, was compliant [87]. A double-blind placebo controlled trial by Vernia et al. of 51 patients with chronically active mild to moderate distal UC randomized patients to receive 2 g topical mesalamine with 80 mM sodium butyrate or 2 g topical mesalamine with 80 mM NaCl b.i.d. for 6 weeks. A significant difference in the rate of remission, with 25 % of the treatment group compared to 4 % of the control group responding, was reported. Compared to baseline values, both groups had significant improvement in clinical scores and improvement in UCDAI scores. The butyrate group had significant improvements compared to the control group for clinical scores, stool frequency, urgency, and self-assessment [88]. Lührs et al. randomized 11 patients to receive a 60 ml b.i.d. enema of 100 mM sodium butyrate or saline for 8 weeks. DAI scores were significantly lower in the butyrate group compared to the control group at trial completion, accompanied by reduced NF-κB translocation in lamina propria macrophages [89]. In 35 UC patients in clinical remission randomized to receive a 60 ml enema daily for 20 days containing either 100 mM sodium butyrate or saline, butyrate had only minor effects on inflammatory measures, with significant increases in colonic IL-10/IL-12 ratios and increased colonic concentrations of chemokine (C–C motif) ligand 5 (CCL5) in the butyrate group compared to placebo. No significant differences were found on oxidative stress parameters [90].

Table 2.

Controlled trials of SCFA and colitis

Patients, n Treatment Results Reference
47 SCFA enema versus placebo No significant differences [86]
103 SCFA enema versus placebo No significant differences [87]
51 5-ASA and NaB enema versus 5-ASA and placebo Significant increase in remission and improvements in clinical scores, stool frequency, urgency, and self-assessment [88]
11 NaB enema versus placebo Significant decrease in DAI scores, reduction of NF-κB translocation in lamina propria macrophages [89]
335 NaB enema versus placebo Minor effects on inflammatory parameters and no significant effects on oxidative stress parameters [90]
39 NaB enema versus placebo No significant differences [91]
45 Corticosteroids versus 5-ASA versus SCFA enema Similar improvement, although SCFA more cost-effective [92]
30 Oral mesalazine and butyrate versus oral mesalazine and placebo Improvement in clinical and UCDAI scores [93]

SCFA short-chain fatty acids, UCDAI ulcerative colitis disease activity index

In 39 patients with distal UC treated with a 60 ml daily enema of 80 mM sodium butyrate or control solution of NaCl (with 0.8 mM sodium butyrate added to produce a butyrate odor), no statistically significant differences were found in clinical improvement or remission, mucosal appearance scores, or histological scores [91]. Finally, in a randomized administration of hydrocortisone, 5-ASA, and SCFA enemas, patients with proctosigmoiditis had similar improvements in endoscopic scores, histological scores, and symptom improvement, although SCFA were more cost effective [92].

Since drug administration by enema is cumbersome, inconvenient and disliked by many, oral SCFA administration has been advocated. For example, mice with DSS-induced colitis fed a control diet plus sodium butyrate had improvements in mucosal inflammation, diarrhea, crypt length, and the inflammatory profile of the intestinal mucosa, with improvements in the cytokine pattern of the small intestine [94]. In an uncontrolled trial of 216 patients with mild to moderate UC, patients were given three tablets containing 307 mg butyrate and 250 mg of the prebiotic inulin t.i.d. in addition to 300 mg of mesalamine t.i.d. for 6 months. At the study end, of the 196 subjects completing the trial, compliance was 92 %, 110 (56 %) patients had complete clinical and endoscopic remission, 46 (23 %) had clinical remission and endoscopic improvement, 14 (7 %) had complete clinical remission and slight endoscopic improvement, and 26 (13 %) had no clinical or endoscopic improvement, with lower ulcerative colitis disease activity index (UCDAI) scores [85]. In a randomized double-blind placebo-controlled trial, 30 patients with mild to moderate UC colitis received 2.4-g oral mesalamine combined with either 4 g butyrate or placebo daily for 6 weeks. Both groups showed significant decreases in UCDAI scores and improvement in clinical index and histological scores. When considering clinical and UCDAI scores, there was significantly greater improvement in the butyrate versus placebo group [93].

Pouchitis

The ileal pouch-anal anastomosis procedure is performed in some patients with UC or familial adenomatous polyposis (FAP) in order to avoid ostomy creation. Pouchitis, or inflammation of the ileoanal reservoir, is a common complication occurring in 20–50 % of UC and 0–11 % of FAP patients [95]. Fecal SCFA are reduced in patients with pouchitis [96, 97]. Since the pathogenesis of pouchitis is thought to be similar to UC, probiotics and SCFA have been studied as treatments.

Using meta-analysis, probiotics were reported to be effective in the prevention and treatment of pouchitis [98, 99]. VSL#3 was the most effective probiotic in the treatment of pouchitis [99]. SCFA enemas do not improve symptoms, although only a very few small studies have been published [97, 100].

Diversion Colitis

Diversion colitis, an inflammatory condition that occurs in nonfunctioning segments of the colon, develops in most patients after surgical diversion of the fecal stream. Several mechanisms for the development of diversion colitis have been proposed, including a nutritional deficiency of SCFA in the colonic lumen [101, 102]. Studies in Wistar rats suggest that the use of SCFA enemas may improve symptoms [102, 103]. Harig et al. [101] successfully treated patients with diversion colitis using SCFA enemas. Not all studies, however, have found improvements with the use of SCFA enemas in diversion colitis. A study by Guillemot et al. [104] found no endoscopic or histological improvements, although this study was shorter in duration than the study by Harig et al.

Short Bowel Syndrome

Short bowel syndrome (SBS) is associated with numerous complications including dehydration, diarrhea, electrolyte disturbances, malabsorption and malnutrition. Patients with a resected bowel may undergo intestinal adaptation, where the residual intestine undergoes structural and functional enhancements. Total parenteral nutrition is sometimes used in patients with SBS, although this may inhibit intestinal adaptation as the residual gut is not being stimulated [105]. In rat and neonatal pig models, supplementation of TPN solutions with SCFA may enhance intestinal adaptation [106-109]. Possible mechanisms for the effects of SCFA on intestinal adaptation include upregulation of glucose transporter 2 (GLUT2) [107], upregulation of proglucan and ornithine decarboxylase [106], and increases of the plasma concentrations of the intestinotrophic hormone GLP-2 [105, 109]. SCFA absorption in the colon may also have beneficial effects for SBS patients by serving as a source of calories [105]. A recent study in patients with SBS reported that the primary carbohydrate substrate for fermentation by the colonic microbiota is starch, with pectin also increasing SCFA production by the colonic microbiota [110].

Obesity

Exogenous SCFA, and endogenous SCFA derived from dietary fiber and prebiotics, may participate in the pathogenesis of obesity. As discussed above, SCFA are ligands for the SCFA GPCRs GPR41 and GPR43. GPR43 may be involved in appetite regulation by increasing secretion of peptide YY (PYY) and GLP-1, which regulate digestive enzymes and satiety [28, 111, 112]. In mice, exogenous SCFA prevented the occurrence of diet-induced obesity, with butyrate and propionate being more effective than acetate. Butyrate and propionate supplementation prevented weight gain induced by a high-fat diet, whereas acetate prevented 40 % of this weight gain. SCFA also inhibited food intake and stimulated the anorexigenic peptides GLP-1, PYY and amylin, with butyrate > propionate > acetate [113]. Through the activation of GPR43, acetate and propionate inhibit lipolysis [114, 115], reduce plasma levels of FFA [114], and stimulate adipogenesis in mice [115]. Xiong et al. [35] reported that propionate activates GPR41, stimulating leptin production in mouse adipocytes. Propionate and butyrate stimulated leptin secretion in mouse epididymal adipocytes whereas acetate stimulated leptin secretion in mesenteric adipocytes in a study by Zaibi et al. They suggest that GPR43 as opposed to GPR41 is involved, as they were unable to detect GPR41 mRNA in mouse adipose tissue sites [34]. In human adipose tissue, propionate increased leptin expression while reducing the expression of the proinflammatory factor resistin [36]. Propionate is anti-inflammatory, increasing lipogenesis and glucose uptake in human omental adipose tissue [116]. These functions of SCFA, in particular with regard to their effects on satiety-related hormones and on adipocytes, may find therapeutic utility in the prevention of obesity.

Conclusions

SCFAs, a major component of the intestinal luminal content, can act as signalling molecules or nutrients. Recent advances in the understanding of intestinal epithelial biology have included the identification of a likely SCFA uptake mechanism and of two SCFA GPCRs that likely transduce the presence of luminal SCFA into neurohormonal signals that can affect appetite, glycemic control, intestinal growth and gut motility. Due to their anti-inflammatory effects, SCFAs may be beneficial for treating patients with UC. Although probiotics, prebiotics, and synbiotics enhance gut SCFA production, it remains inconclusive whether or not they are beneficial in the treatment of UC. Exogenously administered SCFA also have had variable success in UC treatment, with uncontrolled studies reporting benefit, but many randomized controlled trials reporting no significant improvements, although SCFA may be more cost effective than are mesalamine compounds due to comparable efficacy at lower cost [92]. Although the majority of studies were designed to study rectally-administered SCFA, trials of oral SCFA are promising, offering the possibility of increased efficacy and convenience. Evidence also suggests that SCFA have beneficial effects in pouchitis, SBS and obesity, although present results are preliminary.

Footnotes

Conflict of interest The authors declare no conflict of interest with any products or commercial entities discussed herein.

Contributor Information

Jessica Soldavini, Nutrition and Food, Greater Los Angeles Veteran Affairs Healthcare System, WLAVA Medical Center, Los Angeles, CA 90073, USA.

Jonathan D. Kaunitz, Email: jake@ucla.edu, Medical Services, Greater Los Angeles Veteran Affairs Healthcare System, WLAVA Medical Center, Los Angeles, CA 90073, USA; Department of Medicine, UCLA School of Medicine, Los Angeles, CA 90024, USA; Department of Surgery, UCLA School of Medicine, Los Angeles, CA 90024, USA; Department of Medicine, CURE: Digestive Diseases Research Center, Los Angeles, CA 90073, USA; Brentwood Biomedical Research Institute, Los Angeles, CA 90073, USA; West Los Angeles VAMC, Building 114, Room 217E, Los Angeles, CA 90073, USA.

References

  • 1.Mortensen PB, Clausen MR. Short-chain fatty acids in the human colon: relation to gastrointestinal health and disease. Scand J Gastroenterol Suppl. 1996;216:132–148. doi: 10.3109/00365529609094568. [DOI] [PubMed] [Google Scholar]
  • 2.Cummings JH, Englyst HN, Wiggins HS. The role of carbohydrates in lower gut function. Nutr Rev. 1986;44:50–54. doi: 10.1111/j.1753-4887.1986.tb07586.x. [DOI] [PubMed] [Google Scholar]
  • 3.Topping DL, Clifton PM. Short-chain fatty acids and human colonic function: roles of resistant starch and nonstarch polysaccharides. Physiol Rev. 2001;81:1031–1064. doi: 10.1152/physrev.2001.81.3.1031. [DOI] [PubMed] [Google Scholar]
  • 4.Nahon S, Lahmek P, Lesgourgues B, et al. Predictive factors of GI lesions in 241 women with iron deficiency anemia. Am J Gastroenterol. 2002;97:590–593. doi: 10.1111/j.1572-0241.2002.05534.x. [DOI] [PubMed] [Google Scholar]
  • 5.Bergman EN. Energy contributions of volatile fatty acids from the gastrointestinal tract in various species. Physiol Rev. 1990;70:567–590. doi: 10.1152/physrev.1990.70.2.567. [DOI] [PubMed] [Google Scholar]
  • 6.Macfarlane GT, Gibson GR, Cummings JH. Comparison of fermentation reactions in different regions of the human colon. J Appl Bacteriol. 1992;72:57–64. doi: 10.1111/j.1365-2672.1992.tb04882.x. [DOI] [PubMed] [Google Scholar]
  • 7.Cummings JH, Pomare EW, Branch WJ, Naylor CP, Macfarlane GT. Short chain fatty acids in human large intestine, portal, hepatic and venous blood. Gut. 1987;28:1221–1227. doi: 10.1136/gut.28.10.1221. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Cummings JH, Englyst HN. Fermentation in the human large intestine and the available substrates. Am J Clin Nutr. 1987;45:1243–1255. doi: 10.1093/ajcn/45.5.1243. [DOI] [PubMed] [Google Scholar]
  • 9.Wong JM, de Souza SR, Kendall CW, Emam A, Jenkins DJ. Colonic health: fermentation and short chain fatty acids. J Clin Gastroenterol. 2006;40:235–243. doi: 10.1097/00004836-200603000-00015. [DOI] [PubMed] [Google Scholar]
  • 10.Looijer-van Langen MA, Dieleman LA. Prebiotics in chronic intestinal inflammation. Inflamm Bowel Dis. 2009;15:454–462. doi: 10.1002/ibd.20737. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Englyst HN, Hay S, Macfarlane GT. Polysaccharide breakdown by mixed populations of human faecal bacteria. FEMS Microbiol Lett. 1987;45:163–171. [Google Scholar]
  • 12.Cummings JH, Macfarlane GT, Englyst HN. Prebiotic digestion and fermentation. Am J Clin Nutr. 2001;73:415S–420S. doi: 10.1093/ajcn/73.2.415s. [DOI] [PubMed] [Google Scholar]
  • 13.Umesaki Y, Yajima T, Yokokura T, Mutai M. Effect of organic acid absorption on bicarbonate transport in rat colon. Pflügers Arch. 1979;379:43–47. doi: 10.1007/BF00622903. [DOI] [PubMed] [Google Scholar]
  • 14.Hunt JN, Knox MT. A relation between the chain length of fatty acids and the slowing of gastric emptying. J Physiol. 1968;194:327–336. doi: 10.1113/jphysiol.1968.sp008411. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Vidyasagar S, Rajendran VM, Binder HJ. Three distinct mechanisms of HCO3-secretion in rat distal colon. Am J Physiol Cell Physiol. 2004;287:C612–C621. doi: 10.1152/ajpcell.00474.2003. [DOI] [PubMed] [Google Scholar]
  • 16.Kawamata K, Hayashi H, Suzuki Y. Propionate absorption associated with bicarbonate secretion in vitro in the mouse cecum. Pflügers Arch. 2007;454:253–262. doi: 10.1007/s00424-006-0200-4. [DOI] [PubMed] [Google Scholar]
  • 17.Ganapathy V, Thangaraju M, Gopal E, et al. Sodium-coupled monocarboxylate transporters in normal tissues and in cancer. AAPS J. 2008;10:193–199. doi: 10.1208/s12248-008-9022-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Ritzhaupt A, Wood IS, Ellis A, Hosie KB, Shirazi-Beechey SP. Identification of a monocarboxylate transporter isoform type 1 (MCT1) on the luminal membrane of human and pig colon. Biochem Soc Trans. 1998;26:S120. doi: 10.1042/bst026s120. [DOI] [PubMed] [Google Scholar]
  • 19.Ritzhaupt A, Ellis A, Hosie KB, Shirazi-Beechey SP. The characterization of butyrate transport across pig and human colonic luminal membrane. J Physiol. 1998;507:819–830. doi: 10.1111/j.1469-7793.1998.819bs.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Ritzhaupt A, Wood IS, Ellis A, Hosie KB, Shirazi-Beechey SP. Identification and characterization of a monocarboxylate transporter (MCT1) in pig and human colon: its potential to transport L-lactate as well as butyrate. J Physiol. 1998;513:719–732. doi: 10.1111/j.1469-7793.1998.719ba.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Gupta N, Martin PM, Prasad PD, Ganapathy V. SLC5A8 (SMCT1)-mediated transport of butyrate forms the basis for the tumor suppressive function of the transporter. Life Sci. 2006;78:2419–2425. doi: 10.1016/j.lfs.2005.10.028. [DOI] [PubMed] [Google Scholar]
  • 22.Gopal E, Miyauchi S, Martin PM, et al. Transport of nicotinate and structurally related compounds by human SMCT1 (SLC5A8) and its relevance to drug transport in the mammalian intestinal tract. Pharm Res. 2007;24:575–584. doi: 10.1007/s11095-006-9176-1. [DOI] [PubMed] [Google Scholar]
  • 23.Paroder V, Spencer SR, Paroder M, et al. Na+/monocarboxylate transport (SMCT) protein expression correlates with survival in colon cancer: molecular characterization of SMCT. Proc Natl Acad Sci USA. 2006;103:7270–7275. doi: 10.1073/pnas.0602365103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Gopal E, Fei YJ, Sugawara M, et al. Expression of slc5a8 in kidney and its role in Na+-coupled transport of lactate. J Biol Chem. 2004;279:44522–44532. doi: 10.1074/jbc.M405365200. [DOI] [PubMed] [Google Scholar]
  • 25.Miyauchi S, Gopal E, Fei YJ, Ganapathy V. Functional identification of SLC5A8, a tumor suppressor down-regulated in colon cancer, as a Na+-coupled transporter for short-chain fatty acids. J Biol Chem. 2004;279:13293–13296. doi: 10.1074/jbc.C400059200. [DOI] [PubMed] [Google Scholar]
  • 26.Coady MJ, Chang MH, Charron FM, et al. The human tumour suppressor gene SLC5A8 expresses a Na+-monocarboxylate cotransporter. J Physiol. 2004;557:719–731. doi: 10.1113/jphysiol.2004.063859. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Stoddart LA, Smith NJ, Milligan G. International Union of Pharmacology. LXXI. Free fatty acid receptors FFA-1,-2, and - 3: pharmacology and pathophysiological functions. Pharmacol Rev. 2008;60:405–417. doi: 10.1124/pr.108.00802. [DOI] [PubMed] [Google Scholar]
  • 28.Karaki SI, Tazoe H, Kaji I, Otomo Y, Yajima T, Kuwahara A. Contractile and secretory responses of luminal short-chain fatty acids and the expression of these receptors, GPR41 and GPR43, in the human small and large intestines. Gastroenterology. 2008;134:A368–A368. [Google Scholar]
  • 29.Wang A, Akers RM, Jiang H. Short communication: presence of G protein-coupled receptor 43 in rumen epithelium but not in the islets of Langerhans in cattle. J Dairy Sci. 2012;95:1371–1375. doi: 10.3168/jds.2011-4886. [DOI] [PubMed] [Google Scholar]
  • 30.Kaji I, Karaki S, Tanaka R, Kuwahara A. Density distribution of free fatty acid receptor 2 (FFA2)-expressing and GLP-1-producing enteroendocrine L cells in human and rat lower intestine, and increased cell numbers after ingestion of fructo-oligosaccharide. J Mol Histol. 2011;42:27–38. doi: 10.1007/s10735-010-9304-4. [DOI] [PubMed] [Google Scholar]
  • 31.McNeil NI. The contribution of the large intestine to energy supplies in man. Am J Clin Nutr. 1984;39:338–342. doi: 10.1093/ajcn/39.2.338. [DOI] [PubMed] [Google Scholar]
  • 32.Fleming SE, Fitch MD, DeVries S, Liu ML, Kight C. Nutrient utilization by cells isolated from rat jejunum, cecum and colon. J Nutr. 1991;121:869–878. doi: 10.1093/jn/121.6.869. [DOI] [PubMed] [Google Scholar]
  • 33.Scheppach W, Bartram P, Richter A, et al. Effect of short-chain fatty acids on the human colonic mucosa in vitro. JPEN J Parenter Enteral Nutr. 1992;16:43–48. doi: 10.1177/014860719201600143. [DOI] [PubMed] [Google Scholar]
  • 34.Zaibi MS, Stocker CJ, O’Dowd J, et al. Roles of GPR41 and GPR43 in leptin secretory responses of murine adipocytes to short chain fatty acids. FEBS Lett. 2010;584:2381–2386. doi: 10.1016/j.febslet.2010.04.027. [DOI] [PubMed] [Google Scholar]
  • 35.Xiong Y, Miyamoto N, Shibata K, et al. Short-chain fatty acids stimulate leptin production in adipocytes through the G protein-coupled receptor GPR41. Proc Natl Acad SciUSA. 2004;101:1045–1050. doi: 10.1073/pnas.2637002100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Al-Lahham SH, Roelofsen H, Priebe M, et al. Regulation of adipokine production in human adipose tissue by proprionic acid. Eur J Clin Invest. 2010;40:401–407. doi: 10.1111/j.1365-2362.2010.02278.x. [DOI] [PubMed] [Google Scholar]
  • 37.Hague A, Manning AM, Hanlon KA, Huschtscha LI, Hart D, Paraskeva C. Sodium butyrate induces apoptosis in human colonic tumour cell lines in a p53-independent pathway: implications for the possible role of dietary fibre in the prevention of large-bowel cancer. Int J Cancer. 1993;55:498–505. doi: 10.1002/ijc.2910550329. [DOI] [PubMed] [Google Scholar]
  • 38.Hague A, Elder DJ, Hicks DJ, Paraskeva C. Apoptosis in colorectal tumour cells: induction by the short chain fatty acids butyrate, propionate and acetate and by the bile salt deoxycholate. Int J Cancer. 1995;60:400–406. doi: 10.1002/ijc.2910600322. [DOI] [PubMed] [Google Scholar]
  • 39.Deniz M, Bozkurt A, Kurtel H. Mediators of glucagon-like peptide 2-induced blood flow: responses in different vascular sites. Regul Pept. 2007;142:7–15. doi: 10.1016/j.regpep.2007.01.002. [DOI] [PubMed] [Google Scholar]
  • 40.Marks P, Rifkind RA, Richon VM, Breslow R, Miller T, Kelly WK. Histone deacetylases and cancer: causes and therapies. Nat Rev Cancer. 2001;1:194–202. doi: 10.1038/35106079. [DOI] [PubMed] [Google Scholar]
  • 41.Vinolo MA, Rodrigues HG, Hatanaka E, Sato FT, Sampaio SC, Curi R. Suppressive effect of short-chain fatty acids on production of proinflammatory mediators by neutrophils. J Nutr Biochem. 2011;22:849–855. doi: 10.1016/j.jnutbio.2010.07.009. [DOI] [PubMed] [Google Scholar]
  • 42.Baumgart DC, Sandborn WJ. Inflammatory bowel disease: clinical aspects and established and evolving therapies. Lancet. 2007;369:1641–1657. doi: 10.1016/S0140-6736(07)60751-X. [DOI] [PubMed] [Google Scholar]
  • 43.Jonkers D, Penders J, Masclee A, Pierik M. Probiotics in the management of inflammatory bowel disease: a systematic review of intervention studies in adult patients. Drugs. 2012;72:803–823. doi: 10.2165/11632710-000000000-00000. [DOI] [PubMed] [Google Scholar]
  • 44.Takaishi H, Matsuki T, Nakazawa A, et al. Imbalance in intestinal microflora constitution could be involved in the pathogenesis of inflammatory bowel disease. Int J Med Microbiol. 2008;298:463–472. doi: 10.1016/j.ijmm.2007.07.016. [DOI] [PubMed] [Google Scholar]
  • 45.Sokol H, Seksik P, Furet JP, et al. Low counts of Faecalibacterium prausnitzii in colitis microbiota. Inflamm Bowel Dis. 2009;15:1183–1189. doi: 10.1002/ibd.20903. [DOI] [PubMed] [Google Scholar]
  • 46.Mylonaki M, Rayment NB, Rampton DS, Hudspith BN, Brostoff J. Molecular characterization of rectal mucosa-associated bacterial flora in inflammatory bowel disease. Inflamm Bowel Dis. 2005;11:481–487. doi: 10.1097/01.mib.0000159663.62651.4f. [DOI] [PubMed] [Google Scholar]
  • 47.Atreya I, Atreya R, Neurath MF. NF-κB in inflammatory bowel disease. J Intern Med. 2008;263:591–596. doi: 10.1111/j.1365-2796.2008.01953.x. [DOI] [PubMed] [Google Scholar]
  • 48.De Preter V, Geboes KP, Bulteel V, et al. Kinetics of butyrate metabolism in the normal colon and in ulcerative colitis: the effects of substrate concentration and carnitine on the betaoxidation pathway. Aliment Pharmacol Ther. 2011;34:526–532. doi: 10.1111/j.1365-2036.2011.04757.x. [DOI] [PubMed] [Google Scholar]
  • 49.De Preter V, Bulteel V, Suenaert P, et al. Pouchitis, similar to active ulcerative colitis, is associated with impaired butyrate oxidation by intestinal mucosa. Inflamm Bowel Dis. 2009;15:335–340. doi: 10.1002/ibd.20768. [DOI] [PubMed] [Google Scholar]
  • 50.Hino S, Ito H, Bito H, Kawagishi H, Morita T. Ameliorating effects of short-chain inulin-like fructans on the healing stage of trinitrobenzene sulfonic acid-induced colitis in rats. Biosci Biotechnol Biochem. 2011;75:2169–2174. doi: 10.1271/bbb.110460. [DOI] [PubMed] [Google Scholar]
  • 51.Komiyama Y, Andoh A, Fujiwara D, et al. New prebiotics from rice bran ameliorate inflammation in murine colitis models through the modulation of intestinal homeostasis and the mucosal immune system. Scand J Gastroenterol. 2011;46:40–52. doi: 10.3109/00365521.2010.513062. [DOI] [PubMed] [Google Scholar]
  • 52.Hong YS, Ahn YT, Park JC, et al. 1H NMR-based metabonomic assessment of probiotic effects in a colitis mouse model. Arch Pharm Res. 2010;33:1091–1101. doi: 10.1007/s12272-010-0716-1. [DOI] [PubMed] [Google Scholar]
  • 53.Osman N, Adawi D, Molin G, Ahrne S, Berggren A, Jeppsson B. Bifidobacterium infantis strains with and without a combination of oligofructose and inulin (OFI) attenuate inflammation in DSS-induced colitis in rats. BMC Gastroenterol. 2006;6:31. doi: 10.1186/1471-230X-6-31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Kato K, Mizuno S, Umesaki Y, et al. Randomized placebo-controlled trial assessing the effect of bifidobacteria-fermented milk on active ulcerative colitis. Aliment Pharmacol Ther. 2004;20:1133–1141. doi: 10.1111/j.1365-2036.2004.02268.x. [DOI] [PubMed] [Google Scholar]
  • 55.Schneider SM, Girard-Pipau F, Filippi J, et al. Effects of Saccharomyces boulardii on fecal short-chain fatty acids and microflora in patients on long-term total enteral nutrition. World J Gastroenterol. 2005;11:6165–6169. doi: 10.3748/wjg.v11.i39.6165. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Damen B, Cloetens L, Broekaert WF, et al. Consumption of breads containing in situ-produced arabinoxylan oligosaccharides alters gastrointestinal effects in healthy volunteers. J Nutr. 2012;142:470–477. doi: 10.3945/jn.111.146464. [DOI] [PubMed] [Google Scholar]
  • 57.Holscher HD, Faust KL, Czerkies LA, et al. Effects of prebioticcontaining infant formula on gastrointestinal tolerance and fecal microbiota in a randomized controlled trial. JPEN J Parenter Enteral Nutr. 2012;36:95S–105S. doi: 10.1177/0148607111430087. [DOI] [PubMed] [Google Scholar]
  • 58.Riezzo G, Orlando A, D’Attoma B, et al. Randomised clinical trial: efficacy of Lactobacillus paracasei-enriched artichokes in the treatment of patients with functional constipation—a doubleblind, controlled, crossover study. Aliment Pharmacol Ther. 2012;35:441–450. doi: 10.1111/j.1365-2036.2011.04970.x. [DOI] [PubMed] [Google Scholar]
  • 59.Moreau NM, Martin LJ, Toquet CS, et al. Restoration of the integrity of rat caeco-colonic mucosa by resistant starch, but not by fructo-oligosaccharides, in dextran sulfate sodium-induced experimental colitis. Br J Nutr. 2003;90:75–85. doi: 10.1079/bjn2003867. [DOI] [PubMed] [Google Scholar]
  • 60.Lara-Villoslada F, Debras E, Nieto A, et al. Oligosaccharides isolated from goat milk reduce intestinal inflammation in a rat model of dextran sodium sulfate-induced colitis. Clin Nutr. 2006;25:477–488. doi: 10.1016/j.clnu.2005.11.004. [DOI] [PubMed] [Google Scholar]
  • 61.Winkler J, Butler R, Symonds E. Fructo-oligosaccharide reduces inflammation in a dextran sodium sulphate mouse model of colitis. Dig Dis Sci. 2007;52:52–58. doi: 10.1007/s10620-006-9224-z. [DOI] [PubMed] [Google Scholar]
  • 62.Cherbut C, Michel C, Lecannu G. The prebiotic characteristics of fructooligosaccharides are necessary for reduction of TNBS-induced colitis in rats. J Nutr. 2003;133:21–27. doi: 10.1093/jn/133.1.21. [DOI] [PubMed] [Google Scholar]
  • 63.Rumi G, Tsubouchi R, Okayama M, Kato S, Mozsik G, Takeuchi K. Protective effect of lactulose on dextran sulfate sodium-induced colonic inflammation in rats. Dig Dis Sci. 2004;49:1466–1472. doi: 10.1023/b:ddas.0000042248.48819.ad. [DOI] [PubMed] [Google Scholar]
  • 64.Camuesco D, Peran L, Comalada M, et al. Preventative effects of lactulose in the trinitrobenzenesulphonic acid model of rat colitis. Inflamm Bowel Dis. 2005;11:265–271. doi: 10.1097/01.mib.0000160808.30988.d9. [DOI] [PubMed] [Google Scholar]
  • 65.Daddaoua A, Martinez-Plata E, Lopez-Posadas R, et al. Active hexose correlated compound acts as a prebiotic and is antiinflammatory in rats with hapten-induced colitis. J Nutr. 2007;137:1222–1228. doi: 10.1093/jn/137.5.1222. [DOI] [PubMed] [Google Scholar]
  • 66.Daddaoua A, Puerta V, Requena P, et al. Goat milk oligosaccharides are anti-inflammatory in rats with hapten-induced colitis. J Nutr. 2006;136:672–676. doi: 10.1093/jn/136.3.672. [DOI] [PubMed] [Google Scholar]
  • 67.Holma R, Juvonen P, Asmawi MZ, Vapaatalo H, Korpela R. Galacto-oligosaccharides stimulate the growth of bifidobacteria but fail to attenuate inflammation in experimental colitis in rats. Scand J Gastroenterol. 2002;37:1042–1047. doi: 10.1080/003655202320378239. [DOI] [PubMed] [Google Scholar]
  • 68.Hoentjen F, Welling GW, Harmsen HJ, et al. Reduction of colitis by prebiotics in HLA-B27 transgenic rats is associated with microflora changes and immunomodulation. Inflamm Bowel Dis. 2005;11:977–985. doi: 10.1097/01.mib.0000183421.02316.d5. [DOI] [PubMed] [Google Scholar]
  • 69.Schultz M, Munro K, Tannock GW, et al. Effects of feeding a probiotic preparation (SIM) containing inulin on the severity of colitis and on the composition of the intestinal microflora in HLA-B27 transgenic rats. Clin Diagn Lab Immunol. 2004;11:581–587. doi: 10.1128/CDLI.11.3.581-587.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Zhang HQ, Ding TT, Zhao JS, et al. Therapeutic effects of Clostridium butyricum on experimental colitis induced by oxazolone in rats. World J Gastroenterol. 2009;15:1821–1828. doi: 10.3748/wjg.15.1821. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Osman N, Adawi D, Ahrne S, Jeppsson B, Molin G. Probiotics and blueberry attenuate the severity of dextran sulfate sodium (DSS)-induced colitis. Dig Dis Sci. 2008;53:2464–2473. doi: 10.1007/s10620-007-0174-x. [DOI] [PubMed] [Google Scholar]
  • 72.Raz I, Gollop N, Polak-Charcon S, Schwartz B. Isolation and characterisation of new putative probiotic bacteria from human colonic flora. Br J Nutr. 2007;97:725–734. doi: 10.1017/S000711450747249X. [DOI] [PubMed] [Google Scholar]
  • 73.Peran L, Sierra S, Comalada M, et al. A comparative study of the preventative effects exerted by two probiotics, Lactobacillus reuteri and Lactobacillus fermentum, in the trinitrobenzenesulfonic acid model of rat colitis. Br J Nutr. 2007;97:96–103. doi: 10.1017/S0007114507257770. [DOI] [PubMed] [Google Scholar]
  • 74.Casellas F, Borruel N, Torrejon A, et al. Oral oligofructoseenriched inulin supplementation in acute ulcerative colitis is well tolerated and associated with lowered faecal calprotectin. Aliment Pharmacol Ther. 2007;25:1061–1067. doi: 10.1111/j.1365-2036.2007.03288.x. [DOI] [PubMed] [Google Scholar]
  • 75.Furrie E, Macfarlane S, Kennedy A, et al. Synbiotic therapy (Bifidobacterium longum/Synergy 1) initiates resolution of inflammation in patients with active ulcerative colitis: a randomised controlled pilot trial. Gut. 2005;54:242–249. doi: 10.1136/gut.2004.044834. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Ishikawa H, Matsumoto S, Ohashi Y, et al. Beneficial effects of probiotic bifidobacterium and galacto-oligosaccharide in patients with ulcerative colitis: a randomized controlled study. Digestion. 2011;84:128–133. doi: 10.1159/000322977. [DOI] [PubMed] [Google Scholar]
  • 77.Naidoo K, Gordon M, Fagbemi AO, Thomas AG, Akobeng AK. Probiotics for maintenance of remission in ulcerative colitis. Cochrane Database Syst Rev. 2011;12 doi: 10.1002/14651858.CD007443.pub2. CD007443. [DOI] [PubMed] [Google Scholar]
  • 78.Mallon P, McKay D, Kirk S, Gardiner K. Probiotics for induction of remission in ulcerative colitis. Cochrane Database Syst Rev. 2007;4 doi: 10.1002/14651858.CD005573.pub2. CD005573. [DOI] [PubMed] [Google Scholar]
  • 79.Butzner JD, Parmar R, Bell CJ, Dalal V. Butyrate enema therapy stimulates mucosal repair in experimental colitis in the rat. Gut. 1996;38:568–573. doi: 10.1136/gut.38.4.568. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Song M, Xia B, Li J. Effects of topical treatment of sodium butyrate and 5-aminosalicylic acid on expression of trefoil factor 3, interleukin 1beta, and nuclear factor kappaB in trinitrobenzene sulphonic acid induced colitis in rats. Postgrad Med J. 2006;82:130–135. doi: 10.1136/pgmj.2005.037945. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Tarrerias AL, Millecamps M, Alloui A, et al. Short-chain fatty acid enemas fail to decrease colonic hypersensitivity and inflammation in TNBS-induced colonic inflammation in rats. Pain. 2002;100:91–97. doi: 10.1016/s0304-3959(02)00234-8. [DOI] [PubMed] [Google Scholar]
  • 82.Breuer RI, Buto SK, Christ ML, et al. Rectal irrigation with short-chain fatty acids for distal ulcerative colitis. Preliminary report. Dig Dis Sci. 1991;36:185–187. doi: 10.1007/BF01300754. [DOI] [PubMed] [Google Scholar]
  • 83.Steinhart AH, Brzezinski A, Baker JP. Treatment of refractory ulcerative proctosigmoiditis with butyrate enemas. Am J Gastroenterol. 1994;89:179–183. [PubMed] [Google Scholar]
  • 84.Vernia P, Cittadini M, Caprilli R, Torsoli A. Topical treatment of refractory distal ulcerative colitis with 5-ASA and sodium butyrate. Dig Dis Sci. 1995;40:305–307. doi: 10.1007/BF02065414. [DOI] [PubMed] [Google Scholar]
  • 85.Assisi RF. Combined butyric acid/mesalazine treatment in ulcerative colitis with mild-moderate activity. Results of a multicentre pilot study. Minerva Gastroenterol Dietol. 2008;54:231–238. [PubMed] [Google Scholar]
  • 86.Scheppach W. Treatment of distal ulcerative colitis with shortchain fatty acid enemas. A placebo-controlled trial. German-Austrian SCFA Study Group. Dig Dis Sci. 1996;41:2254–2259. doi: 10.1007/BF02071409. [DOI] [PubMed] [Google Scholar]
  • 87.Breuer RI, Soergel KH, Lashner BA, et al. Short chain fatty acid rectal irrigation for left-sided ulcerative colitis: a randomised, placebo controlled trial. Gut. 1997;40:485–491. doi: 10.1136/gut.40.4.485. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Vernia P, Annese V, Bresci G, et al. Topical butyrate improves efficacy of 5-ASA in refractory distal ulcerative colitis: results of a multicentre trial. Eur J Clin Invest. 2003;33:244–248. doi: 10.1046/j.1365-2362.2003.01130.x. [DOI] [PubMed] [Google Scholar]
  • 89.Luhrs H, Gerke T, Muller JG, et al. Butyrate inhibits NF-κB activation in lamina propria macrophages of patients with ulcerative colitis. Scand J Gastroenterol. 2002;37:458–466. doi: 10.1080/003655202317316105. [DOI] [PubMed] [Google Scholar]
  • 90.Hamer HM, Jonkers DM, Vanhoutvin SA, et al. Effect of butyrate enemas on inflammation and antioxidant status in the colonic mucosa of patients with ulcerative colitis in remission. Clin Nutr. 2010;29:738–744. doi: 10.1016/j.clnu.2010.04.002. [DOI] [PubMed] [Google Scholar]
  • 91.Steinhart AH, Hiruki T, Brzezinski A, Baker JP. Treatment of left-sided ulcerative colitis with butyrate enemas: a controlled trial. Aliment Pharmacol Ther. 1996;10:729–736. doi: 10.1046/j.1365-2036.1996.d01-509.x. [DOI] [PubMed] [Google Scholar]
  • 92.Senagore AJ, MacKeigan JM, Scheider M, Ebrom JS. Short-chain fatty acid enemas: a cost-effective alternative in the treatment of nonspecific proctosigmoiditis. Dis Colon Rectum. 1992;35:923–927. doi: 10.1007/BF02253492. [DOI] [PubMed] [Google Scholar]
  • 93.Vernia P, Monteleone G, Grandinetti 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;45:976–981. doi: 10.1023/a:1005537411244. [DOI] [PubMed] [Google Scholar]
  • 94.Vieira EL, Leonel AJ, Sad AP, et al. Oral administration of sodium butyrate attenuates inflammation and mucosal lesion in experimental acute ulcerative colitis. J Nutr Biochem. 2012;23:430–436. doi: 10.1016/j.jnutbio.2011.01.007. [DOI] [PubMed] [Google Scholar]
  • 95.McLaughlin SD, Clark SK, Tekkis PP, Nicholls RJ, Ciclitira PJ. The bacterial pathogenesis and treatment of pouchitis. Therap Adv Gastroenterol. 2010;3:335–348. doi: 10.1177/1756283X10370611. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Clausen MR, Tvede M, Mortensen PB. Short-chain fatty acids in pouch contents from patients with and without pouchitis after ileal pouch-anal anastomosis. Gastroenterology. 1992;103:1144–1153. doi: 10.1016/0016-5085(92)91497-r. [DOI] [PubMed] [Google Scholar]
  • 97.Wischmeyer P, Pemberton JH, Phillips SF. Chronic pouchitis after ileal pouch-anal anastomosis: responses to butyrate and glutamine suppositories in a pilot study. Mayo Clin Proc. 1993;68:978–981. doi: 10.1016/s0025-6196(12)62270-8. [DOI] [PubMed] [Google Scholar]
  • 98.Ritchie ML, Romanuk TN. A meta-analysis of probiotic efficacy for gastrointestinal diseases. PLoS ONE. 2012;7:e34938. doi: 10.1371/journal.pone.0034938. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Elahi B, Nikfar S, Derakhshani S, Vafaie M, Abdollahi M. On the benefit of probiotics in the management of pouchitis in patients underwent ileal pouch anal anastomosis: a meta-analysis of controlled clinical trials. Dig Dis Sci. 2008;53:1278–1284. doi: 10.1007/s10620-007-0006-z. [DOI] [PubMed] [Google Scholar]
  • 100.de Silva HJ, Ireland A, Kettlewell M, Mortensen N, Jewell DP. Short-chain fatty acid irrigation in severe pouchitis. N Engl J Med. 1989;321:1416–1417. doi: 10.1056/nejm198911163212019. [DOI] [PubMed] [Google Scholar]
  • 101.Harig JM, Soergel KH, Komorowski RA, Wood CM. Treatment of diversion colitis with short-chain-fatty acid irrigation. N Engl J Med. 1989;320:23–28. doi: 10.1056/NEJM198901053200105. [DOI] [PubMed] [Google Scholar]
  • 102.Pacheco RG, Esposito CC, Müller LC, et al. Use of butyrate or glutamine in enema solution reduces inflammation and fibrosis in experimental diversion colitis. World J Gastroenterol. 2012;18:4278–4287. doi: 10.3748/wjg.v18.i32.4278. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Oliveira AJ, Pinto Júnior FE, Formiga MC, Melo SP, Brandao-Neto J, Ramos AM. Comparison of prophylactic and therapeutic use of short-chain fatty acid enemas in diversion colitis: a study in Wistar rats. Clinics. 2010;65:1351–1356. doi: 10.1590/S1807-59322010001200020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Guillemot F, Colombel JF, Neut C, et al. Treatment of diversion colitis by short-chain fatty acids. Prospective and double-blind study. Dis Colon Rectum. 1991;34:861–864. doi: 10.1007/BF02049697. [DOI] [PubMed] [Google Scholar]
  • 105.Tappenden KA, Albin DM, Bartholome AL, Mangian HF. Glucagon-like peptide-2 and short-chain fatty acids: a new twist to an old story. J Nutr. 2003;133:3717–3720. doi: 10.1093/jn/133.11.3717. [DOI] [PubMed] [Google Scholar]
  • 106.Tappenden KA, Thomson AB, Wild GE, McBurney MI. Shortchain fatty acids increase proglucagon and ornithine decarboxylase messenger RNAs after intestinal resection in rats. JPEN J Parenter Enteral Nutr. 1996;20:357–362. doi: 10.1177/0148607196020005357. [DOI] [PubMed] [Google Scholar]
  • 107.Tappenden KA, Thomson AB, Wild GE, McBurney MI. Short-chain fatty acid-supplemented total parenteral nutrition enhances functional adaptation to intestinal resection in rats. Gastroenterology. 1997;112:792–802. doi: 10.1053/gast.1997.v112.pm9041241. [DOI] [PubMed] [Google Scholar]
  • 108.Kripke SA, De Paula JA, Berman JM, Fox AD, Rombeau JL, Settle RG. Experimental short-bowel syndrome: effect of an elemental diet supplemented with short-chain triglycerides. Am J Clin Nutr. 1991;53:954–962. doi: 10.1093/ajcn/53.4.954. [DOI] [PubMed] [Google Scholar]
  • 109.Bartholome AL, Albin DM, Baker DH, Holst JJ, Tappenden KA. Supplementation of total parenteral nutrition with butyrate acutely increases structural aspects of intestinal adaptation after an 80% jejunoileal resection in neonatal piglets. JPEN J Parenter Enteral Nutr. 2004;28:210–222. doi: 10.1177/0148607104028004210. [DOI] [PubMed] [Google Scholar]
  • 110.Atia A, Girard-Pipau F, Hebuterne X, et al. Macronutrient absorption characteristics in humans with short bowel syndrome and jejunocolonic anastomosis: starch is the most important carbohydrate substrate, although pectin supplementation may modestly enhance short chain fatty acid production and fluid absorption. JPEN J Parenter Enteral Nutr. 2011;35:229–240. doi: 10.1177/0148607110378410. [DOI] [PubMed] [Google Scholar]
  • 111.Karaki S, Mitsui R, Hayashi H, et al. Short-chain fatty acid receptor, GPR43, is expressed by enteroendocrine cells and mucosal mast cells in rat intestine. Cell Tissue Res. 2006;324:353–360. doi: 10.1007/s00441-005-0140-x. [DOI] [PubMed] [Google Scholar]
  • 112.Karaki S, Tazoe H, Hayashi H, et al. Expression of the shortchain fatty acid receptor, GPR43, in the human colon. J Mol Histol. 2008;39:135–142. doi: 10.1007/s10735-007-9145-y. [DOI] [PubMed] [Google Scholar]
  • 113.Lin HV, Frassetto A, Kowalik EJ, Jr, et al. Butyrate and propionate protect against diet-induced obesity and regulate gut hormones via free fatty acid receptor 3-independent mechanisms. PLoS ONE. 2012;7:e35240. doi: 10.1371/journal.pone.0035240. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Ge H, Li X, Weiszmann J, et al. Activation of G protein-coupled receptor 43 in adipocytes leads to inhibition of lipolysis and suppression of plasma free fatty acids. Endocrinology. 2008;149:4519–4526. doi: 10.1210/en.2008-0059. [DOI] [PubMed] [Google Scholar]
  • 115.Hong YH, Nishimura Y, Hishikawa D, et al. Acetate and propionate short chain fatty acids stimulate adipogenesis via GPCR43. Endocrinology. 2005;146:5092–5099. doi: 10.1210/en.2005-0545. [DOI] [PubMed] [Google Scholar]
  • 116.Al-Lahham S, Roelofsen H, Rezaee F, et al. Propionic acid affects immune status and metabolism in adipose tissue from overweight subjects. Eur J Clin Invest. 2012;42:357–364. doi: 10.1111/j.1365-2362.2011.02590.x. [DOI] [PubMed] [Google Scholar]

RESOURCES