Minireviews Open Access
Copyright ©The Author(s) 2023. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. Jan 28, 2023; 29(4): 682-691
Published online Jan 28, 2023. doi: 10.3748/wjg.v29.i4.682
Gaseous metabolites as therapeutic targets in ulcerative colitis
Chu K Yao, Department of Gastroenterology, Monash University, Melbourne 3004, Australia
Chen Sarbagili-Shabat, Pediatric Gastroenterology Unit, PIBD Research Center, Wolfson Medical Center, Holon 5822012, Israel
ORCID number: Chu K Yao (0000-0002-5301-2457).
Author contributions: Yao CK and Sarbagili-Shabat C conducted the literature search; Yao CK devised headings for the article; and all authors drafted, wrote the article and approved of the final content.
Conflict-of-interest statement: Yao CK has received support for investigator-initiated grants from Atmo Biosciences. She also works in a department that financially benefits from the sales of a digital application and booklets on the low fermentable oligosaccharides, disaccharides, monosaccharides and polyols diet. Funds raised contribute to research of the Department of Gastroenterology and to the University. She does not receive personal remuneration. Sarbagili-Shabat C has no conflicts of interest to report.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Chu K Yao, PhD, RN, Senior Research Fellow, Department of Gastroenterology, Monash University, Level 6, Alfred Centre, 99 Commercial Road, Melbourne 3004, Australia. chu.yao@monash.edu
Received: September 20, 2022
Peer-review started: September 20, 2022
First decision: November 15, 2022
Revised: December 19, 2022
Accepted: January 10, 2023
Article in press: January 10, 2023
Published online: January 28, 2023

Abstract

Diet therapies are currently under-utilised in optimising clinical outcomes for patients with active ulcerative colitis (UC). Furthermore, existing dietary therapies are framed by poorly defined mechanistic targets to warrant its success. There is good evidence to suggest that microbial production of gaseous metabolites, hydrogen sulfide (H2S) and nitric oxide (NO) are implicated in the development of mucosal inflammation in UC. On a cellular level, exposure of the colonic epithelium to excessive concentrations of these gases are shown to promote functional defects described in UC. Hence, targeting bacterial production of these gases could provide an opportunity to formulate new dietary therapies in UC. Despite the paucity of evidence, there is epidemiological and clinical data to support the concept of reducing mucosal inflammation in UC via dietary strategies that reduce H2S. Several dietary components, namely sulphur-containing amino acids and inorganic sulphur have been shown to be influential in enhancing colonic H2S production. More recent data suggests increasing the supply of readily fermentable fibre as an effective strategy for H2S reduction. Conversely, very little is known regarding how diet alters microbial production of NO. Hence, the current evidence suggest that a whole diet approach is needed. Finally, biomarkers for assessing changes in microbial gaseous metabolites in response to dietary interventions are very much required. In conclusion, this review identifies a great need for high quality randomised-controlled trials to demonstrate the efficacy of a sulphide-reducing dietary therapy for patients with active UC.

Key Words: Diet, Ulcerative colitis, Hydrogen sulfide, Nitric oxide, Sulphide-reducing diet

Core Tip: There is room to develop efficacious dietary therapies in ulcerative colitis (UC) by targeting underlying pathogenic mechanisms. Emerging data indicates that dietary factors play a significant role in modulating two gaseous metabolites, hydrogen sulphide and nitric oxide, that affect the integrity of the colonic mucosal barrier in UC. These gases are produced by the colonic microbiota in response to sulphur-containing protein and to a lesser extent, inorganic sulphur (sulphates and sulphites), but suppressed by the presence of fermentable fibre. Preliminary work suggests that a multi-prong diet that targets reduction of these gases have therapeutic potential and further randomised-controlled trials are underway.



INTRODUCTION

Ulcerative colitis (UC) is characterised by chronic inflammation of the colonic epithelium as a result of an aberrant immune response to poorly understood initiating triggers[1]. Diet is a well-recognised environmental factor in the development of UC[1,2], but remains an under-utilised therapeutic tool amongst physicians and dietitians alike. Dietary management is currently directed at providing supportive symptomatic management. However, in the recent years, there has been a dogma shift towards harvesting dietary therapies with mechanistic targets for the induction of disease remission, as evidenced by the growing number of review articles in the area[3-5].

Whilst most research have been focused on altered immune regulation in the early initiative events, there is now a good body of evidence generated over the last 20 years suggesting that UC is an epithelial disease[6]. Metabolic defects in the colonic epithelium are central in its pathogenesis and may be responsible for mucosal barrier breakdown[7]. In turn, microbial metabolites such as hydrogen sulfide (H2S) and nitric oxide (NO) that are toxic at excessive concentrations, may further exert injurious effects on the epithelium[8]. Diet is a major factor in colonic production of these metabolites. Hence, dietary strategies that minimise their production mechanistically may have therapeutic benefits in UC. This review aims to examine the evidence for H2S and NO as causative agents in UC, the influence of diet on their colonic metabolism and to explore the rationale as well as evidence to date for dietary strategies targeting these gaseous metabolites as a therapy in UC.

COLONIC PRODUCTION OF H2S & NO

Luminal H2S is derived solely from metabolic activities of the microbiota, namely from fermentation of sulfur-containing amino acids and dissimilatory sulfate reduction[9]. Approximately 6-18 g/d of proteinaceous substrates are delivered to the colon for fermentation, the bulk of this originating from undigested dietary protein and a smaller proportion from endogenous protein secretions[10]. A range of protein-fermenting microbes with the capacity to generate H2S have been reported including Escherichia coli, Clostridium spp., Bacteroides spp. and Klebsiella pneumonia[10]. In contrast, the capacity to reduce sulfate within the microbiota appears to be limited. A smaller proportion of malabsorbed dietary inorganic sulfur (0.3-8 mmol/d)[11] reach the colon as substrates for dissimilatory sulfate reduction. Sulfate- and sulfite-reducing bacteria such as Desulfovibrio spp. and Bilophila wadsworthia are highly specialised microbes with capacity for sulfate reduction[11].

On the other hand, two major sources of luminal NO are known: (1) Mucosal production from arginine; or (2) Anaerobic bacterial denitrification which reduces nitrates to nitrites and to NO[8]. To date, little work has been done to examine microbial populations capable of denitrification. Hence, the understanding of microbial pathways for gaseous production has important implications not only as potential therapeutic targets but has significant relevance for manipulation of dietary substrates.

ROLE AS LUMINAL TOXINS IN PATHOGENESIS OF UC

The most compelling argument for the colonic epithelium as the primary defect in UC has been derived from ex vivo studies showing diffuse structural and functional abnormalities in the absence of histological or endoscopic inflammation[12-14]. A key functional defect identified is the impaired uptake and oxidation of butyrate by colonocytes for energy[15,16]. As a result, the energy-starved colonic epithelium has limited ability to perform other metabolic functions including the maintenance of barrier function. Furthermore, reduced structural integrity of the colonic mucus layer was reported by van der Post et al[13]. This was characterised by a marked decrease in core mucus components in both inflamed and non-inflamed biopsy samples, with similar findings reported previously[14]. Hence, the induction of mucosal inflammation may occur as a secondary response to the increased intestinal permeability[6].

Several lines of observations support the involvement of luminal H2S and NO in perpetuating functional defects of the colonic epithelium. These concepts are summarised in Figure 1. First, Levine et al[17] showed that faecal release of H2S was three-fold higher and more rapid in UC patients (both active and quiescent) compared to controls. Additionally, a greater relative abundance and activity of sulfate-reducing microbes, Desulfovibrio, has been documented in faecal or mucosal biopsy samples of patients with UC compared to non-UC controls[18,19]. Gut dysbiosis may be the main pathogenic factor of UC, and the higher dominance of sulphate-reducing microbes may potentially contribute to the dysbiosis hypothesised in the pathogenesis of UC. No data currently exists of potential alterations to the abundance of protein-fermenting microbes in UC. Furthermore, in contrast to a healthy colonic epithelium where H2S is effectively detoxified, enzymatic detoxification activity of H2S have been shown to be significantly depressed in UC[20]. Finally, elevated luminal H2S concentrations are shown to be directly proportional to the severity of disease[16,17], providing an evidence base for a pathogenic link with UC. Likewise, direct assessment of luminal NO using a rectal balloon in patients with active UC demonstrated markedly higher rectal NO levels in these patients compared to those with irritable bowel syndrome and healthy controls[21].

Figure 1
Figure 1 Proposed mechanisms of microbial metabolites, hydrogen sulfide and nitric oxide, in the pathogenesis of ulcerative colitis via a dysfunctional colonic epithelium and breakdown in mucosal barrier function. Figure summarised from references[8,55-57]. CHO: Carbohydrate; H2S: Hydrogen sulfide; SRB: Sulfate-reducing bacteria; H2S: Hydrogen sulfide; IL: Interleukin.

Secondly, reduced carbohydrate fermentative ability, as was recently reported[22], and decreased accessibility to short-chain fatty acids[23] may have lead-on effects on altered sulfur metabolism. Insights gained by assessment of intestinal pH responses to dietary manipulation of fermentable fibres suggest that abnormalities in carbohydrate fermentative ability may be region specific[24]. Reduced butyrate utilisation may increase luminal accumulation of H2S as its regulatory role on detoxification pathways are affected[25]. On the other hand, fibre deprivation may act synergistically with H2S to increase breakdown of the mucous layer[26].

Thirdly, at excessive concentrations, continuous exposure of isolated colonocytes to combined H2S and NO in vitro can produce extensive disruption of the epithelial barrier by interfering with cell membrane synthesis[8], impeding butyrate oxidation and subsequently, cellular respiration, producing an energy-deficient state as described earlier. This theory was confirmed by Leung et al[27] who induced a histological state that was similar to the pathology of UC in the colon of rats administered with sulfates (carrageenan). Furthermore, excessive H2S and NO may exert other pathogenic effects, including direct immune effects and these are summarised in Figure 1.

Hence, restricting epithelial exposure to luminal H2S and NO via reduced microbial production may hypothetically improve epithelial function and reduce mucosal inflammation in UC, a novel therapeutic strategy that was proposed two decades ago[28] but has only achieved some progress in the last two years. Progress is hampered by difficulties in accurate measurements of luminal H2S and NO to provide a biomarker for assessing the efficacy of interventions on these metabolites. These challenges are discussed further in the subsequent sections.

DIET AS PRIMARY STRATEGY FOR COLONIC H2S & NO MANIPULATION

From discussions above, it can be hypothesised that a key strategy in reducing microbial H2S and NO production is by reducing substrate availability. Food choice represents a rationale candidate for manipulation as substrate delivery to the colon is strongly influenced by dietary intake. Indeed, several lines of evidence exist supporting the efficacy of dietary manipulation on colonic H2S production. In contrast, the influence of diet on the extent of bacterial denitrification has been inconsistently shown.

First, acute dietary studies in healthy controls changing from low to a high animal protein diet consistently raised faecal H2S levels[29,30]. Magee et al[29] reported this increase in H2S levels to be linear with increasing intake of red meat (from 0 to 600 g/d). In another study, a four-day animal-based diet specifically increased a sulfite-reducing species, Bilophila wadsworthia, while a plant-based diet reduced this cluster[31]. Similarly, the animal-based diet significantly increased sulfide reductases needed for H2S production[31]. Another source of inorganic sulfur in the diet occurs naturally in the form of glucosinolates in the Brassica vegetables family. However, a two-week diet high in brassica was associated with a reduction in the abundance of sulphate-reducing bacteria in a randomized crossover study with ten healthy adults[32], which seems to indicate that natural inorganic sulfur is not a determining factor in H2S production associated with sulfate-reducing bacteria. Thirdly, whilst assessment of sulfate-reducing bacteria may be useful, it does not provide a comprehensive picture of functional alterations in microbial H2S metabolism in vivo. Preliminary insights were gained with the use of a gas-sensing technology incorporating real-time, accurate measurements of H2S to enable further understanding of the extent of dietary influence on microbial H2S production[33]. A comparison between faecal slurries spiked with cysteine, a sulfur-containing amino acid, and sodium sulfate showed marked differences in faecal H2S generation, with cysteine vigorously stimulating H2S over sulfate. This finding indicates that protein fermentation may be a major pathway for H2S production than dissimilatory sulfate reduction. Furthermore, faecal H2S was effectively reduced by readily fermentable fibres, resistant starch and fructo-oligosaccharides, both of which are prebiotics, and even in the presence of excessive faecal H2S production using cysteine[33]. The likely mechanism for H2S suppression by fermentable fibre in the presence of cysteine is the shift from protein to carbohydrate fermentation as microbes preferentially ferment fibre than protein[10]. Suppression of H2S has been reported by another study where a 1.5-fold increase in total dietary fibre that accompanied the reduction in animal protein had a negative impact on H2S production[30] and in a second study, the addition of resistant starch to a high meat diet reduced markers of protein fermentation including H2S[34]. Both inulin and fructo-oligosaccharides, well-established prebiotics were also shown to reduce H2S levels in pigs[35].

In addition, one of the strategies targeting the microbiota is probiotics with specific probiotic strains shown to be effective in inducing remission in active UC[36]. However, its properties on the gut microbiota warrants further investigation, particularly with regards to the influence of different probiotic strains on H2S production. On the other hand, a promising probiotic treatment for UC is recent development of a ‘smart probiotic’ where E. coli Nissle 1917 was genetically engineered to detect colonic NO and would theoretically be able to release biologic therapy at the site of elevated colonic NO[37]. This engineered probiotic had previously been shown to have positive impact on the intestinal barrier function, and were able to reduce inflammation in dextran sulfate sodium -induced colitis mice model[38]. Prebiotics are another key player in microbiome manipulation that have been suggested to have a positive effect on the microbiome. Their mechanisms in modulating microbial H2S have already been discussed earlier. However, randomized controlled trials (RCTs) in UC patients that evaluated the efficacy of prebiotic supplementation alone demonstrated limited weak effect[39] which indicates that a multi-prong approach, not just prebiotic supplementation, is required to achieve clinical effects.

Secondly, there is some evidence from epidemiological studies that provide clues for the influence of dietary sulfur-containing protein, sulfates and sulfites on the clinical course of UC. One study reported a correlation between a high protein intake and increased risk of developing UC[40] whilst only one study has shown an association between a high intake of sulfur amino acids and sulfate with a three-fold greater risk of disease relapse[41]. Subsequently, the potential clinical efficacy of a sulfur-restricted diet was first described from a small open-label study in eight UC patients. The low sulfur diet combined with stable salazopyrin therapy was associated with histological and clinical improvement[9]. Changes in colonic H2S production was unfortunately, not measured as a mechanism for efficacy but the promise of this dietary approach warrant further investigation in a controlled trial.

Furthermore, there has been growing interest in the role of carrageenan, a sulphated polysaccharide food additive that escapes digestion in the small intestine almost intact and is fermented to release sulphates[42], which is then metabolised to produce H2S. In 2017, a RCT by Bhattacharyya et al[43] assessed the role of carrageenan, a sulphated polysaccharide food additive, in maintaining relapse in 14 UC patients in remission. Following a year of no-carrageenan diet, relapse rates appeared to be higher in the five patients receiving 200 mg carrageenan, a dose slightly below average intakes of carrageenan in the US diet, than those who received placebo capsules. Unfortunately, significant recruitment issues impacted on the sample size of the study, making it difficult to ascertain whether this was a real clinical effect.

Finally, the major sources of nitrites in the diet are food preservatives in cured and processed meat, while the major source of dietary nitrates is vegetables[44]. Thus far, the effect of different sources of nitrite and nitrate in the diet on microbial NO production are unknown and should be further investigated in clinical studies.

TRANSLATING PROPOSED DIETARY STRATEGY INTO CLINICAL APPLICATION

Several dietary strategies can therefore be implied for future clinical application from studies thus far. First, a multi-prong intervention targeting dietary substrates with H2S-modulating abilities, expanding on Roediger[9]’s earlier work, is warranted in active UC patients. This approach should consider reducing intake of sulfur-containing protein such as methionine, cysteine and taurine, and added sources of inorganic sulfur to reduce excessive/chronic H2S exposure to the colonic epithelium. Major sources of these foods are listed in Table 1. Inorganic sulfur exist as food additives in several forms, sulfur dioxide (E220), sulfites (E221-E227) and as a sulphated polysaccharide, carrageenan (E407). In Australia and Europe, food labelling requirements mandate only the labelling of added sulphites (in amounts > 10 mg/kg) in food product without specifying the amount used. Inorganic sulfur intake by foods and beverages has been showed to be six-fold higher in the western diet in comparison to a typical African rural diet[45,46]. However, food composition tables on sulfur-containing protein, inorganic sulfur and carrageenan are far from complete to adequately assess habitual intake of UC patients, to ensure successful design of the dietary therapy. More importantly, there are grounds that increasing a combination of fibre, rather than restricting total fibre, maybe an efficacious strategy for H2S suppression whilst improving nutrient delivery to the colonic epithelium in UC. Resistant starch and fructo-oligosaccharides, whilst efficacious, are fermented in the proximal colon[10]. Hence, a strategy that will carry the fermentation of these fibres across the entire colon by combining with a minimally fermentable fibre is required.

Table 1 Content of sulfur, nitrate and nitrite in selected foods.
Food category
Specific food
Sulfur amino acids (cysteine + methionine)1, mg/100 g
Sulfates2, mg/100 g/mL
Nitrate3, mg/kg
Nitrite3, mg/kg
High sulfur amino acids foodsBeef239---
Chicken291---
Turkey269---
Tuna268---
Prawns189---
Eggs162---
Cheese, hard174---
High sulfites foodsDried apricots-300--
Dried apples-490--
Commercial bread-80-150--
Wine-38--
High sulfates foodsCabbage-84--
Broccoli-90--
Cauliflower-50--
Brussels sprouts-93--
High nitrates foodsLettuce--2351-
Celery--2110-
Spinach--1509-
Leek--841-
High nitrites foodsSausages, boiled---40
Poultry meat---32
Beef---59
Bacon---86

Indeed, tolerability and the potential clinical effects of a dietary approach incorporating strategies discussed above have already been evaluated. In an open-label dietary advice study, Day et al[47] reported excellent tolerability of dietary strategy called the 4-strategies to a sulphide-reducing (4-SURE) diet by patients with mild to moderately active UC. This was despite the 38% increase in dietary fibre and the four-fold increase in resistant starch intake by these patients. Food-related quality of life also increased markedly. Whilst it was impractical to assess colonic H2S in this study, markers of protein fermentation, namely faecal branched chain fatty acids were used as a surrogate. The significant reduction in faecal branched to short-chain fatty acid ratio following the 4-SURE study indicated that protein fermentation being the major pathway for luminal H2S production was reduced. Whilst the study did not intend to primarily assess clinical end-points due to the uncontrolled study design, there were indicators for the diet to positively affect clinical outcomes and mucosal healing. Data supported by a significant reduction in faecal calprotectin. A second dietary approach, called the Ulcerative Colitis Exclusion Diet (UCED), also incorporated a similar exclusion of decreasing intake of total protein, sulphur-containing amino acids, food additives along with additional restrictions of animal and saturated fat, haeme, whilst increasing intake of tryptophan, pectin and resistant starch[48]. In a RCT comparing a combination of UCED with faecal transplant, faecal transplant or diet alone, Sarbagili Shabat et al[48] observed that clinical response and endoscopic remission were the greatest for the UCED diet. Furthermore, the promising outcomes of the UCED was supported by an earlier open-label study in paediatric patients with mild to moderate active UC on stable maintenance therapy, where the diet treatment showed that patients had a significant decrease in sulfur-containing amino acids consumption as well as a significant increase in total fiber consumption[49].

Whilst these proposed dietary approaches are not quite ready for clinical application until RCTs have been performed (currently underway) to replicate the promising findings, it does suggest that patients with active inflammation do tolerate a certain increase in high fibre foods and builds on the suggestion to minimise intake of processed foods. Moreover, the limitations of the available reported clinical trials targeting reduction of H2S production as a treatment strategy for UC (Table 2) suggest the need for larger, high quality dietary studies incorporating gut microbiome composition and function assessment including changes in microbial H2S metabolism.

Table 2 Summary of studies clinical outcomes by dietary interventions for ulcerative colitis as a possible strategy to modify hydrogen sulfide production.
Ref.
Dietary intervention
Study design
Main outcomes
Limitations
Roediger[9], 1998Low sulfur dietOpen-label, prospective pilot study. Patients were instructed to follow low sulfur diet + stable dose of salazopyrin for 12 mo (n = 4 adults)All patients showed clinical and histological improvement and no relapse attacks were observedVery small sample size
Bhattacharyya et al[43], 2017No-carrageenan dietDouble-blind RCT: Carrageenan capsules versus placebo. Patients with remission were followed up until relapse or of 12 mo (n = 12 adults)The carrageenan group demonstrated significant higher relapse rate and an increase in FC and IL-6 values from study onsetSmall sample size in each group. The effects on the microbiome were not addressed and precise measurements of compliance with the diet were not performed
Chiba et al[58], 2019Lacto-ovo-semivegetarian diet-PBDProspective single arm study. Patients were followed after induction therapy incorporating PBD (n = 92 children and adults)The cumulative relapse rates at 1 and 5 yr were 14% and 27% respectively, which is indicated by the authors to be lower than those previously reportedSmall sample size without control group. The mechanistic effect of the diet was not addressed
Sarbagili Shabat et al[48], 2022UCEDSingle-blind RCT in adults with active refractory UC: Group1: FT alone; group2: FT with UCED; group3: UCED alone. The primary endpoint was week 8 clinical remission (n = 51)UCED alone demonstrated the greatest clinical and endoscopic remission rates compared to single donor FT with or without dietSmall sample size in each group. Eligibility criteria include patients with severe UC, of whom none obtain remission. The effects on the microbiome were not addressed
Sarbagili-Shabat et al[49], 2021UCEDOpen-label, prospective pilot study in children with active UC. The primary endpoint was week 6 clinical remission (n = 24)UCED lead to 38% clinical remission and FC improvementSmall sample size without control group. The effects on the microbiome were not addressed
Day et al[47], 20224-SUREOpen-label, prospective pilot study in adults with active UC. The primary endpoint was week 8 tolerability (n = 28)The 4-SURE diet was well tolerated and lead to 46% clinical response and 36% endoscopic improvement. Fecal excretion of SCFAs increased while BCFAs decreasedChanges in colonic H2S not able to be measured. Lack of control and inadequate power for interpretation of secondary clinical end-points
BIOMARKERS FOR ASSESSING RESPONSE OF DIETARY THERAPY

It is key that a biomarker for assessing diet response is incorporated early on after dietary therapy is administered as a way of assessing whether the diet is achieving its intended mechanistic effect. An example of this is the reduction in breath hydrogen production after introduction of a diet low in fermentable carbohydrates as a biomarker of intervention success[50]. However, in the case of dietary approaches targeting colonic H2S and NO, there are difficulties with accessing reliable measurement techniques for these volatile gases, particularly with ex vivo measurements often requiring freshly passed faecal samples[17,33], which introduces practical issues for trial patients. Currently, measurements for H2S mainly involve faecal sulphide, urinary sulphate or breath H2S[51]. Sensitivity of these measurements are impacted by its adsorption or susceptibility to oxidation, yielding low concentrations[51]. In contrast, the only reported assessment of luminal NO has been using direct sampling (via a tonometric balloon) and measurement via a rapid-response chemiluminescence technique[21]. While the method has good sensitivity, it is unknown whether this biomarker is directly responsive to alterations in dietary nitrate and nitrite intake. Finally, there is potential for direct intestinal gas sampling, such as the gas-sensing capsule[52], but these do not yet measure H2S or NO. In the absence of reliable direct measurements, indirect assessments could target markers of protein fermentation for H2S, quantification of sulphate- or sulphite-reducing bacteria which are dependent on availability of proteinaceous substrates for growth[53], and have capacities for denitrification and sulphate-reduction[54]. Hence, an effective biomarker for monitoring the success of sulphide- and NO-reducing dietary approaches remains elusive and is very much needed to support the development of the proposed dietary therapies. Therefore, as in most studies, assessment of dietary response is primarily assessed by different questionnaires such as dietary intake questionnaires, food-related quality of life or health-related quality of life questionnaire. Combined biomarker measurements with assessment by questionnaires can be the ideal tool for estimating the effect of specific dietary exposure.

CONCLUSION

Microbial H2S and NO metabolites have causative roles in the pathogenesis of UC via their damaging effects on the colonic epithelium. Modulation of their production within the colonic lumen in order to reduce colonic epithelial exposure to these luminal stressors presents an attractive therapeutic target that has yet to be adequately explored. The current evidence suggests that dietary manipulation is likely to be an effective strategy to modify colonic H2S production whereas little is known regarding dietary modulation of NO. It is also clear that sulfur-containing amino acids are major substrates that promote H2S production over inorganic sulphur but data has emerged suggesting that increasing fermentable fibre is highly efficacious in reducing H2S production. These findings have been utilised to inform the design of multi-prong dietary approaches which have yielded promising therapeutic efficacy in mild to moderate active UC. However, key to advancing the success of this research is the urgent need for better technology to accurately assess luminal concentrations of these volatile gases. Finally, before implementation into dietary practice can be pursued, further investigations into their efficacy on altering disease activity using robust dietary trial designs (which are currently underway), expansion of food composition data and mechanisms of H2S reduction are highly warranted.

Footnotes

Provenance and peer review: Invited article; Externally peer reviewed.

Peer-review model: Single blind

Corresponding Author’s Membership in Professional Societies: Gastroenterological Society of Australia, 100610

Specialty type: Gastroenterology and hepatology

Country/Territory of origin: Australia

Peer-review report’s scientific quality classification

Grade A (Excellent): 0

Grade B (Very good): B

Grade C (Good): C, C

Grade D (Fair): 0

Grade E (Poor): 0

P-Reviewer: Coşkun MG, Turkey; Morozov S, Russia; Zhao G, China S-Editor: Wang JJ L-Editor: A P-Editor: Wang JJ

References
1.  Du L, Ha C. Epidemiology and Pathogenesis of Ulcerative Colitis. Gastroenterol Clin North Am. 2020;49:643-654.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 86]  [Cited by in F6Publishing: 197]  [Article Influence: 49.3]  [Reference Citation Analysis (0)]
2.  Hou JK, Abraham B, El-Serag H. Dietary intake and risk of developing inflammatory bowel disease: a systematic review of the literature. Am J Gastroenterol. 2011;106:563-573.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 590]  [Cited by in F6Publishing: 607]  [Article Influence: 46.7]  [Reference Citation Analysis (0)]
3.  Lewis JD, Abreu MT. Diet as a Trigger or Therapy for Inflammatory Bowel Diseases. Gastroenterology. 2017;152:398-414.e6.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 202]  [Cited by in F6Publishing: 215]  [Article Influence: 30.7]  [Reference Citation Analysis (0)]
4.  Sarbagili-Shabat C, Sigall-Boneh R, Levine A. Nutritional therapy in inflammatory bowel disease. Curr Opin Gastroenterol. 2015;31:303-308.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 19]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
5.  Sigall-Boneh R, Levine A, Lomer M, Wierdsma N, Allan P, Fiorino G, Gatti S, Jonkers D, Kierkus J, Katsanos KH, Melgar S, Yuksel ES, Whelan K, Wine E, Gerasimidis K. Research Gaps in Diet and Nutrition in Inflammatory Bowel Disease. A Topical Review by D-ECCO Working Group [Dietitians of ECCO]. J Crohns Colitis. 2017;11:1407-1419.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 67]  [Cited by in F6Publishing: 70]  [Article Influence: 10.0]  [Reference Citation Analysis (0)]
6.  Gibson PR. Ulcerative colitis: an epithelial disease? Baillieres Clin Gastroenterol. 1997;11:17-33.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 14]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
7.  Roediger WEW. Causation of human ulcerative colitis: A lead from an animal model that mirrors human disease. JGH Open. 2019;3:277-280.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 2]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
8.  Roediger WE. Review article: nitric oxide from dysbiotic bacterial respiration of nitrate in the pathogenesis and as a target for therapy of ulcerative colitis. Aliment Pharmacol Ther. 2008;27:531-541.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 25]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
9.  Roediger WE. Decreased sulphur aminoacid intake in ulcerative colitis. Lancet. 1998;351:1555.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 31]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
10.  Yao CK, Muir JG, Gibson PR. Review article: insights into colonic protein fermentation, its modulation and potential health implications. Aliment Pharmacol Ther. 2016;43:181-196.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 222]  [Cited by in F6Publishing: 244]  [Article Influence: 30.5]  [Reference Citation Analysis (0)]
11.  Florin T, Neale G, Gibson GR, Christl SU, Cummings JH. Metabolism of dietary sulphate: absorption and excretion in humans. Gut. 1991;32:766-773.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 161]  [Cited by in F6Publishing: 165]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
12.  Delpre G, Avidor I, Steinherz R, Kadish U, Ben-Bassat M. Ultrastructural abnormalities in endoscopically and histologically normal and involved colon in ulcerative colitis. Am J Gastroenterol. 1989;84:1038-1046.  [PubMed]  [DOI]  [Cited in This Article: ]
13.  van der Post S, Jabbar KS, Birchenough G, Arike L, Akhtar N, Sjovall H, Johansson MEV, Hansson GC. Structural weakening of the colonic mucus barrier is an early event in ulcerative colitis pathogenesis. Gut. 2019;68:2142-2151.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 177]  [Cited by in F6Publishing: 234]  [Article Influence: 46.8]  [Reference Citation Analysis (0)]
14.  Podolsky DK, Isselbacher KJ. Glycoprotein composition of colonic mucosa. Specific alterations in ulcerative colitis. Gastroenterology. 1984;87:991-998.  [PubMed]  [DOI]  [Cited in This Article: ]
15.  Roediger WE. The colonic epithelium in ulcerative colitis: an energy-deficiency disease? Lancet. 1980;2:712-715.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 469]  [Cited by in F6Publishing: 436]  [Article Influence: 9.9]  [Reference Citation Analysis (0)]
16.  De Preter V, Arijs I, Windey K, Vanhove W, Vermeire S, Schuit F, Rutgeerts P, Verbeke K. Decreased mucosal sulfide detoxification is related to an impaired butyrate oxidation in ulcerative colitis. Inflamm Bowel Dis. 2012;18:2371-2380.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 32]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
17.  Levine J, Ellis CJ, Furne JK, Springfield J, Levitt MD. Fecal hydrogen sulfide production in ulcerative colitis. Am J Gastroenterol. 1998;93:83-87.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 147]  [Cited by in F6Publishing: 145]  [Article Influence: 5.6]  [Reference Citation Analysis (0)]
18.  Rowan F, Docherty NG, Murphy M, Murphy B, Calvin Coffey J, O'Connell PR. Desulfovibrio bacterial species are increased in ulcerative colitis. Dis Colon Rectum. 2010;53:1530-1536.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 197]  [Cited by in F6Publishing: 218]  [Article Influence: 15.6]  [Reference Citation Analysis (0)]
19.  Loubinoux J, Bronowicki JP, Pereira IA, Mougenel JL, Faou AE. Sulfate-reducing bacteria in human feces and their association with inflammatory bowel diseases. FEMS Microbiol Ecol. 2002;40:107-112.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 241]  [Cited by in F6Publishing: 254]  [Article Influence: 16.9]  [Reference Citation Analysis (0)]
20.  De Preter V, Arijs I, Windey K, Vanhove W, Vermeire S, Schuit F, Rutgeerts P, Verbeke K. Impaired butyrate oxidation in ulcerative colitis is due to decreased butyrate uptake and a defect in the oxidation pathway. Inflamm Bowel Dis. 2012;18:1127-1136.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 83]  [Cited by in F6Publishing: 78]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
21.  Reinders CI, Herulf M, Ljung T, Hollenberg J, Weitzberg E, Lundberg JO, Hellström PM. Rectal mucosal nitric oxide in differentiation of inflammatory bowel disease and irritable bowel syndrome. Clin Gastroenterol Hepatol. 2005;3:777-783.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 42]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
22.  James SL, Christophersen CT, Bird AR, Conlon MA, Rosella O, Gibson PR, Muir JG. Abnormal fibre usage in UC in remission. Gut. 2015;64:562-570.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 74]  [Cited by in F6Publishing: 81]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
23.  Khalil NA, Walton GE, Gibson GR, Tuohy KM, Andrews SC. In vitro batch cultures of gut microbiota from healthy and ulcerative colitis (UC) subjects suggest that sulphate-reducing bacteria levels are raised in UC and by a protein-rich diet. Int J Food Sci Nutr. 2014;65:79-88.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 32]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
24.  Yao CK, Burgell RE, Taylor KM, Ward MG, Friedman AB, Barrett JS, Muir JG, Gibson PR. Effects of fiber intake on intestinal pH, transit, and predicted oral mesalamine delivery in patients with ulcerative colitis. J Gastroenterol Hepatol. 2021;36:1580-1589.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 3]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
25.  Ramasamy S, Singh S, Taniere P, Langman MJ, Eggo MC. Sulfide-detoxifying enzymes in the human colon are decreased in cancer and upregulated in differentiation. Am J Physiol Gastrointest Liver Physiol. 2006;291:G288-G296.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 103]  [Cited by in F6Publishing: 107]  [Article Influence: 5.9]  [Reference Citation Analysis (0)]
26.  Desai MS, Seekatz AM, Koropatkin NM, Kamada N, Hickey CA, Wolter M, Pudlo NA, Kitamoto S, Terrapon N, Muller A, Young VB, Henrissat B, Wilmes P, Stappenbeck TS, Núñez G, Martens EC. A Dietary Fiber-Deprived Gut Microbiota Degrades the Colonic Mucus Barrier and Enhances Pathogen Susceptibility. Cell. 2016;167:1339-1353.e21.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1285]  [Cited by in F6Publishing: 1572]  [Article Influence: 224.6]  [Reference Citation Analysis (0)]
27.  Leung FW, Heng MC, Allen S, Seno K, Leung JW, Heng MK. Involvement of luminal bacteria, heat shock protein 60, macrophages and gammadelta T cells in dextran sulfate sodium-induced colitis in rats. Dig Dis Sci. 2000;45:1472-1479.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 14]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
28.  Roediger WE, Moore J, Babidge W. Colonic sulfide in pathogenesis and treatment of ulcerative colitis. Dig Dis Sci. 1997;42:1571-1579.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 198]  [Cited by in F6Publishing: 201]  [Article Influence: 7.4]  [Reference Citation Analysis (0)]
29.  Magee EA, Richardson CJ, Hughes R, Cummings JH. Contribution of dietary protein to sulfide production in the large intestine: an in vitro and a controlled feeding study in humans. Am J Clin Nutr. 2000;72:1488-1494.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 234]  [Cited by in F6Publishing: 227]  [Article Influence: 9.5]  [Reference Citation Analysis (0)]
30.  Teigen L, Mathai PP, Lopez S, Matson M, Elkin B, Kozysa D, Kabage AJ, Hamilton M, Vaughn BP, Sadowsky MJ, Khoruts A. Differential hydrogen sulfide production by a human cohort in response to animal- and plant-based diet interventions. Clin Nutr. 2022;41:1153-1162.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 1]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
31.  David LA, Maurice CF, Carmody RN, Gootenberg DB, Button JE, Wolfe BE, Ling AV, Devlin AS, Varma Y, Fischbach MA, Biddinger SB, Dutton RJ, Turnbaugh PJ. Diet rapidly and reproducibly alters the human gut microbiome. Nature. 2014;505:559-563.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5625]  [Cited by in F6Publishing: 5986]  [Article Influence: 544.2]  [Reference Citation Analysis (0)]
32.  Kellingray L, Tapp HS, Saha S, Doleman JF, Narbad A, Mithen RF. Consumption of a diet rich in Brassica vegetables is associated with a reduced abundance of sulphate-reducing bacteria: A randomised crossover study. Mol Nutr Food Res. 2017;61.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 50]  [Article Influence: 7.1]  [Reference Citation Analysis (0)]
33.  Yao CK, Rotbart A, Ou JZ, Kalantar-Zadeh K, Muir JG, Gibson PR. Modulation of colonic hydrogen sulfide production by diet and mesalazine utilizing a novel gas-profiling technology. Gut Microbes. 2018;9:510-522.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 18]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
34.  Le Leu RK, Young GP, Hu Y, Winter J, Conlon MA. Dietary red meat aggravates dextran sulfate sodium-induced colitis in mice whereas resistant starch attenuates inflammation. Dig Dis Sci. 2013;58:3475-3482.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 56]  [Cited by in F6Publishing: 52]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
35.  Deng YF, Di Liao X, Wang Y, Liang JB, Tufarelli V. Prebiotics Mitigate In Vitro Sulfur-Containing Odour Generation in Caecal Content of Pigs. Italian J Animal Sci. 2015;14:132-137.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 10]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
36.  Derwa Y, Gracie DJ, Hamlin PJ, Ford AC. Systematic review with meta-analysis: the efficacy of probiotics in inflammatory bowel disease. Aliment Pharmacol Ther. 2017;46:389-400.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 224]  [Cited by in F6Publishing: 227]  [Article Influence: 32.4]  [Reference Citation Analysis (1)]
37.  McKay R, Hauk P, Quan D, Bentley WE. Development of Cell-Based Sentinels for Nitric Oxide: Ensuring Marker Expression and Unimodality. ACS Synth Biol. 2018;7:1694-1701.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 20]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
38.  Praveschotinunt P, Duraj-Thatte AM, Gelfat I, Bahl F, Chou DB, Joshi NS. Engineered E. coli Nissle 1917 for the delivery of matrix-tethered therapeutic domains to the gut. Nat Commun. 2019;10:5580.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 193]  [Cited by in F6Publishing: 158]  [Article Influence: 31.6]  [Reference Citation Analysis (0)]
39.  Wedlake L, Slack N, Andreyev HJ, Whelan K. Fiber in the treatment and maintenance of inflammatory bowel disease: a systematic review of randomized controlled trials. Inflamm Bowel Dis. 2014;20:576-586.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 109]  [Cited by in F6Publishing: 108]  [Article Influence: 10.8]  [Reference Citation Analysis (0)]
40.  Jantchou P, Morois S, Clavel-Chapelon F, Boutron-Ruault MC, Carbonnel F. Animal protein intake and risk of inflammatory bowel disease: The E3N prospective study. Am J Gastroenterol. 2010;105:2195-2201.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 268]  [Cited by in F6Publishing: 269]  [Article Influence: 19.2]  [Reference Citation Analysis (0)]
41.  Jowett SL, Seal CJ, Pearce MS, Phillips E, Gregory W, Barton JR, Welfare MR. Influence of dietary factors on the clinical course of ulcerative colitis: a prospective cohort study. Gut. 2004;53:1479-1484.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 293]  [Cited by in F6Publishing: 307]  [Article Influence: 15.4]  [Reference Citation Analysis (0)]
42.  Gibson G, Macfarlane S, Cummings J. The fermentability of polysaccharides by mixed human faecal bacteria in relation to their suitability as bulk‐forming laxatives. Lett Appl Microbiol. 1990;11:251-254.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 26]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
43.  Bhattacharyya S, Shumard T, Xie H, Dodda A, Varady KA, Feferman L, Halline AG, Goldstein JL, Hanauer SB, Tobacman JK. A randomized trial of the effects of the no-carrageenan diet on ulcerative colitis disease activity. Nutr Healthy Aging. 2017;4:181-192.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 61]  [Article Influence: 8.7]  [Reference Citation Analysis (0)]
44.  Temme EH, Vandevijvere S, Vinkx C, Huybrechts I, Goeyens L, Van Oyen H. Average daily nitrate and nitrite intake in the Belgian population older than 15 years. Food Addit Contam Part A Chem Anal Control Expo Risk Assess. 2011;28:1193-1204.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 26]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
45.  Florin THJ, Neale G, Goretski S, Cummings JH. The sulfate content of foods and beverages. J Food Comp Analysis. 1993;6:140-151.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 69]  [Cited by in F6Publishing: 71]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
46.  Magee EA, Curno R, Edmond LM, Cummings JH. Contribution of dietary protein and inorganic sulfur to urinary sulfate: toward a biomarker of inorganic sulfur intake. Am J Clin Nutr. 2004;80:137-142.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 33]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
47.  Day AS, Yao CK, Costello SP, Ruszkiewicz A, Andrews JM, Gibson PR, Bryant RV. Therapeutic Potential of the 4 Strategies to SUlfide-REduction (4-SURE) Diet in Adults with Mild to Moderately Active Ulcerative Colitis: An Open-Label Feasibility Study. J Nutr. 2022;152:1690-1701.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 8]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
48.  Sarbagili Shabat C, Scaldaferri F, Zittan E, Hirsch A, Mentella MC, Musca T, Cohen NA, Ron Y, Fliss Isakov N, Pfeffer J, Yaakov M, Fanali C, Turchini L, Masucci L, Quaranta G, Kolonimos N, Godneva A, Weinberger A, Kopylov U, Levine A, Maharshak N. Use of Faecal Transplantation with a Novel Diet for Mild to Moderate Active Ulcerative Colitis: The CRAFT UC Randomised Controlled Trial. J Crohns Colitis. 2022;16:369-378.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 39]  [Article Influence: 13.0]  [Reference Citation Analysis (0)]
49.  Sarbagili-Shabat C, Albenberg L, Van Limbergen J, Pressman N, Otley A, Yaakov M, Wine E, Weiner D, Levine A. A Novel UC Exclusion Diet and Antibiotics for Treatment of Mild to Moderate Pediatric Ulcerative Colitis: A Prospective Open-Label Pilot Study. Nutrients. 2021;13.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 7]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
50.  Halmos EP, Power VA, Shepherd SJ, Gibson PR, Muir JG. A diet low in FODMAPs reduces symptoms of irritable bowel syndrome. Gastroenterology. 2014;146:67-75.e5.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 810]  [Cited by in F6Publishing: 747]  [Article Influence: 74.7]  [Reference Citation Analysis (0)]
51.  Tangerman A. Measurement and biological significance of the volatile sulfur compounds hydrogen sulfide, methanethiol and dimethyl sulfide in various biological matrices. J Chromatogr B Analyt Technol Biomed Life Sci. 2009;877:3366-3377.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 180]  [Cited by in F6Publishing: 180]  [Article Influence: 12.0]  [Reference Citation Analysis (0)]
52.  Kalantar-Zadeh K, Berean KJ, Burgell RE, Muir JG, Gibson PR. Intestinal gases: influence on gut disorders and the role of dietary manipulations. Nat Rev Gastroenterol Hepatol. 2019;16:733-747.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 70]  [Cited by in F6Publishing: 82]  [Article Influence: 16.4]  [Reference Citation Analysis (0)]
53.  Gibson GR, Cummings JH, Macfarlane GT. Growth and activities of sulphate-reducing bacteria in gut contents of healthy subjects and patients with ulcerative colitis. FEMS Microbiol Lett. 1991;86:103-111.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 194]  [Cited by in F6Publishing: 193]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
54.  Moura I, Bursakov S, Costa C, Moura JJ. Nitrate and nitrite utilization in sulfate-reducing bacteria. Anaerobe. 1997;3:279-290.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 60]  [Cited by in F6Publishing: 55]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
55.  Roediger WE, Lawson MJ, Nance SH, Radcliffe BC. Detectable colonic nitrite levels in inflammatory bowel disease--mucosal or bacterial malfunction? Digestion. 1986;35:199-204.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 60]  [Cited by in F6Publishing: 62]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
56.  Beaumont M, Andriamihaja M, Lan A, Khodorova N, Audebert M, Blouin JM, Grauso M, Lancha L, Benetti PH, Benamouzig R, Tomé D, Bouillaud F, Davila AM, Blachier F. Detrimental effects for colonocytes of an increased exposure to luminal hydrogen sulfide: The adaptive response. Free Radic Biol Med. 2016;93:155-164.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 94]  [Cited by in F6Publishing: 72]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
57.  Ijssennagger N, Belzer C, Hooiveld GJ, Dekker J, van Mil SW, Müller M, Kleerebezem M, van der Meer R. Gut microbiota facilitates dietary heme-induced epithelial hyperproliferation by opening the mucus barrier in colon. Proc Natl Acad Sci U S A. 2015;112:10038-10043.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 226]  [Cited by in F6Publishing: 251]  [Article Influence: 27.9]  [Reference Citation Analysis (0)]
58.  Chiba M, Nakane K, Tsuji T, Tsuda S, Ishii H, Ohno H, Watanabe K, Obara Y, Komatsu M, Sugawara T. Relapse Prevention by Plant-Based Diet Incorporated into Induction Therapy for Ulcerative Colitis: A Single-Group Trial. Perm J. 2019;23.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 11]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]