Recent Progress in Nutrition (ISSN 2771-9871) is an international peer-reviewed Open Access journal published quarterly online by LIDSEN Publishing Inc. This periodical is devoted to publishing high-quality papers that describe the most significant and cutting-edge research in all areas of nutritional sciences. Its aim is to provide timely, authoritative introductions to current thinking, developments and research in carefully selected topics. Also, it aims to enhance the international exchange of scientific activities in nutritional science and human health.

Recent Progress in Nutrition publishes high quality intervention and observational studies in nutrition. High quality systematic reviews and meta-analyses are also welcome as are pilot studies with preliminary data and hypotheses generating studies. Emphasis is placed on understanding the relationship between nutrition and health and of the role of dietary patterns in health and disease.

Topics contain but are not limited to:

  • Macronutrients
  • Micronutrients
  • Essential nutrients
  • Bioactive nutrients
  • Nutrient requirements
  • Nutrient sources
  • Human nutrition aspects
  • Functional foods
  • Nutraceuticals
  • Health claims
  • Public health
  • Diet-related disorders
  • Metabolic syndrome
  • Malnutrition
  • Nutritional supplements
  • Sport nutrition

It publishes a variety of article types: Original Research, Review, Communication, Opinion, Comment, Conference Report, Technical Note, Book Review, etc.

There is no restriction on paper length, provided that the text is concise and comprehensive. Authors should present their results in as much detail as possible, as reviewers are encouraged to emphasize scientific rigor and reproducibility.

Publication Speed (median values for papers published in 2023): Submission to First Decision: 6.7 weeks; Submission to Acceptance: 16.1 weeks; Acceptance to Publication: 6 days (1-2 days of FREE language polishing included)

Current Issue: 2024  Archive: 2023 2022 2021
Open Access Review

Dietary Modulation of the Gut Microbiome in Inflammatory Bowel Disease

Lindsey Cundra 1, Michael Saadeh 2, Manasa Vallabhaneni 2, Kevin Houston 3, Steve D’Souza 2, David A Johnson 4,*

  1. Department of Internal Medicine, MedStar Georgetown University Hospital, Washington DC, USA

  2. Department of Internal Medicine, Eastern Virginia Medical School, Norfolk VA, USA

  3. Department of Internal Medicine, Virginia Commonwealth University, Richmond VA, USA

  4. Division of Gastroenterology, Department of Internal Medicine, Eastern Virginia Medical School, Norfolk VA, USA

Correspondence: David A Johnson

Academic Editor: Célia C.G. Silva

Special Issue: Role of Diets, Vitamins, and Minerals in Cancers and Various Diseases

Received: April 28, 2022 | Accepted: August 04, 2022 | Published: August 09, 2022

Recent Progress in Nutrition 2022, Volume 2, Issue 3, doi:10.21926/rpn.2203019

Recommended citation: Cundra L, Saadeh M, Vallabhaneni M, Houston K, D'Souza S, Johnson DA. Dietary Modulation of the Gut Microbiome in Inflammatory Bowel Disease. Recent Progress in Nutrition 2022;2(3):22; doi:10.21926/rpn.2203019.

© 2022 by the authors. This is an open access article distributed under the conditions of the Creative Commons by Attribution License, which permits unrestricted use, distribution, and reproduction in any medium or format, provided the original work is correctly cited.

Abstract

Inflammatory bowel disease (IBD), which includes ulcerative colitis, and Crohn’s disease, is a chronic relapsing-remitting inflammatory state of the gastrointestinal (GI) system. The cause of IBD is multifactorial, encompassing factors of genetics, environment, and the host immune system, initiating a complex interplay of maladaptive host immune responses and subsequent chronic inflammation. Aberrant host immune responses are at least in part due to changes in the commensal microbiome, which can in turn affect the development and progression of IBD. Diet is known to directly affect the composition of the microbiota which in turn affects the host immune system. Herein, we review the impact of dietary macronutrients, notable supplements, and selected dietary interventions on the gut microbiota and its effect on the microbiome and host metabolism in patients with IBD. Having diet available as an adjunctive, manageable intervention for patients with IBD will remain a promising area of research for the foreseeable future.

Keywords

Diet; inflammatory bowel disease; Crohn’s disease; ulcerative colitis; Mediterranean diet; dietary supplements

1. Inflammatory Bowel Disease and the Microbiota

1.1 Introduction

The two main manifestations of IBD are ulcerative colitis (UC), and Crohn’s disease (CD). It is estimated there are over 6.8 million individuals affected with IBD worldwide, largely increased from 1990 when the prevalence was 3.7 million [1]. Both the incidence and prevalence of IBD are increasing globally at various rates [2,3,4]. The etiology of IBD remains elusive yet involves aberrant immune responses to environmental factors, such as dietary antigens. The maladaptive responses are at least in part due to changes in the commensal microbiome, which can in turn affect the development, and progression of IBD. The GI microbiome has been postulated to play a key role in IBD, both as a cause, but also as a propagator of IBD. Clearly, dietary components play a profound role in the function, and composition of the gut microbiota, and thus in the pathogenesis and treatment of IBD.

It is inherently difficult to investigate the complex interactions between dietary components and the microbiota. Several overarching dietary patterns, however, have been investigated in both epidemiological studies and clinical trials. Herein, we examine dietary macronutrients, notable supplements, and selected dietary interventions on the microbiome in patients with IBD and animal models of colitis. We will provide a background on CD and UC, discuss dietary components and their effect on the microbiota in IBD, as well as selected dietary patterns, such as the Mediterranean diet (MD), low fiber diets, and exclusion diets.

1.2 Crohn’s Disease

Crohn’s disease is characterized by transmural inflammation and skip lesions. While it may occur at any location of the GI tract, roughly 30% of patients have small bowel involvement, most commonly at the terminal ileum. Approximately 20% of patients have disease localized to the colon, and nearly 50% of patients have involvement of the small and large intestine [5]. Individuals with CD often experience periods of symptomatic relapse and remission with variation in disease severity. This typically follows a bimodal pattern at age of onset, with the first peak occurring between 15 and 30 years of age, and the second peak occurring mainly in women between the ages of 60 and 70 [6].

There are many genes at play, most notably the NOD2/CARD15 gene on chromosome 16, which recently has been shown to be associated with ileal involvement and earlier age of onset [7]. Variants at the NOD2 and CDH1 loci confer the largest increase in relative risk of IBD, and have been associated with UC, specifically [8,9,10]. However, in non-Caucasian populations, the incidence of IBD does not correlate with NOD2 variation [11]. The pathophysiology involves a multitude of factors including genetic, immunologic, dietary, and infectious. It begins with focal inflammatory infiltrate around intestinal crypts, before progressing to ulceration of the mucosal layer. Non-caseating granulomas soon develop, and inflammation of the entire intestinal wall follows shortly thereafter. Chronic inflammation may lead to strictures and fibrosis of the intestine, increasing the risk for obstruction. Additionally, due to transmural inflammation, enterocutaneous, enterovesical, enterovaginal, and enteroenteric fistulas may occur [6].

1.3 Ulcerative Colitis

Ulcerative colitis is the other major type of IBD, and the more common form worldwide. It follows a bimodal distribution, with the first incidence peak occurring around 20 to 30 years of age, and the second peak occurring around 50 to 80 years of age [12]. It is characterized by continuous mucosal inflammation beginning in the rectum and extending proximally throughout the colon. Inflammatory infiltrate, primarily consisting of lymphocytes, invade the lamina propria and crypts of Lieberkühn, leading to crypt branching, and abscess formation. This further progresses to ulceration of the mucosa and submucosa, loss of normal vascular pattern, friability, bleeding, and granulation tissue [6,12,13].

Genetic susceptibility is the most significant risk factor, with family history encompassing 8% to 14% of patients, and leading to four times the risk of developing UC [13]. Additionally, it is hypothesized that genetically susceptible individuals who develop the disease undergo alterations in gut microbiota thereby affecting mucosal immunity. Ulcerative Colitis is strongly associated with primary sclerosing cholangitis and poses an increased risk for colorectal carcinoma with 5% of patients developing colon cancer [6,12,14].

1.4 IBD and the Microbiome: Dysbiosis

The microbiota of each person is unique with significant variability of taxa between persons. The intestinal microbiota comprises a multitude of phyla, the most common of which are Bacteroidetes, Firmicutes, Proteobacteria, and Actinobacteria [15,16]. Diet is known to directly influence the composition of these phyla into distinct phenotypes, which directly affects the host immune system. Given the gut microbiota acts symbiotically to produce energy substrates, facilitate host metabolism, as well as modulate the host immune system. Specifically, the microbiota helps to modulate host immunity by protecting against pathogens.

The significant inter-individual variation in the microbiome composition can be attributed to multiple factors that affect development of the ‘normal’ microbiome. Several prenatal and childhood factors (childhood exposome), including amniotic fluid microbial composition, cesarean section versus vaginal delivery, breast versus formula feeding, and diet and local environment during childhood, are all associated with variation in composition of commensal flora [17]. Additional work has also demonstrated several phenotype associations in addition to childhood exposome, including diet, current exposome, socioeconomic factors, and general state of health [18]. Of the phenotype groups, diet seems to have a greater associative effect on commensal microbial composition than socioeconomics, general medical factors, and current and childhood exposomes [18].

The microbiome and the host have a mutual symbiotic relationship where the host provides a diet rich in nutrients, while the gut microbiome can affect many physiologic mechanisms and cascade pathways. More importantly, evidence has shown that certain microbiome antigens play an immune-mediated destructive or protective role in inducing the differentiation of mucosal CD4+ T cells into T-helper 1 cells, T-helper 17 (Th17) cells, or T-regulatory (Treg) cells [19]. The balance of Th17 and Treg cells, which is characterized by levels of pro-inflammatory and anti-inflammatory cytokines, is critical for the host’s intestinal homeostasis, and is directly affected by the normal gut microbiota content. Moreover, the mucosal immune cells act in concert with the intestinal barrier to maintain a balanced flora. A misbalanced flora, or dysbiosis of the flora, can result in damage to the intestinal microbial barrier.

Dysbiosis encompasses a change in the composition of the commensal microbiome as compared to those of healthy individuals. It has been viewed as a risk factor in the pathogenesis of many GI diseases including GI reflux disease, peptic ulcer disease, infections, and various cancers [20,21,22]. Describing changes in the gut microbiome taxonomy generally includes describing changes in the composition of the major phyla Bacteroides and Firmicutes. The ratio of these phyla has important influence in maintaining normal intestinal homeostasis, expressed as the Firmicutes/Bacteroidetes (F/B) ratio [23]. It is generally accepted that decreases in the F/B ratio have been associated with IBD [23]. Bacillus, Clostridium, Enterococcus, Lactobacillus, and Ruminicoccus are the most common genera in the Firmicutes phylum. Whereas in the phylum Bacteroidetes, the most common genera include Bacteroides, Parabacteroides, and Prevotella.

Specific alterations in the genera have been associated with IBD. For instance, Fecalibacterium prausnitzii, Clostridium, and Bacteroides have been associated with a decreased abundance in IBD [24,25]. On the contrary, Escherichia coli, Fusobacterium, and Veillonella have been associated with an increased abundance in patients with IBD [24,25]. Further, these changes and modulations in the genera composition were associated with disease activity in patients with IBD. In a study comparing active UC and inactive UC, there was a significant difference in the abundance of Firmicutes [26]. In the study, there was similarly a significant difference in those with severe CD as compared to mild to moderate CD [26]. The equilibrium of the gut microbiota with the host is highly important to overall systemic physiology, intestinal barrier integrity, and host immunity. Next, we will discuss how dietary components can directly affect the composition of the intestinal microbiota.

2. Dietary Macronutrients and the Gut Microbiota

2.1 Fats

Dietary fats play essential and critical roles in metabolism, cell function, and comprise cell membranes. Dietary fatty acids can be classified into saturated fatty acids and unsaturated fatty acids. Unsaturated fatty acids include monounsaturated fatty acids (MUFAs) and polyunsaturated fatty acids (PUFAs). PUFAs can be further classified into omega-3 (n-3) and omega-6 (n-6) fatty acids. The intake of dietary fats has significant and varied effects on the immune system through changes in gut microbiota composition and changes in paracellular permeability by interacting with epithelial cells directly [27]. Evidence supports proliferative changes in the colonic microbiome lead to cytokine production, with a subsequent effect on the host immune system.

Key players in the inflammatory pathways include Toll-like receptors (TLRs) and NF-κB. Activation of NF-κB leads to the production of pro-inflammatory cytokines [28]. PUFAs inhibit the NF-κB pathway through protectins. The influence of dietary PUFAs on inflammation may be due to increased endotoxemia as evidence supports a direct correlation between plasma endotoxin levels and dietary fat intake, affecting epithelial gut permeability [29]. These changes are demonstrated in germ-free mouse models colonized by dysbiotic flora and with high fat microbiota transfers. Several studies on murine and human models have demonstrated several possible etiologies-increased endotoxemia [30], increased gut permeability [31], and a shift in the gut microbiota composition [32].

The F/B ratio, as discussed previously, influences intestinal homeostasis [23]. High fat (HF) diets have been shown to induce changes in both murine models and human trials [33]. HF diet changes in animal and human models were characterized by a decrease in Bacteroides, and an increase in Firmicutes, a possible hallmark for obesity [23]. These changes in taxonomy affect the gut metabolome and modulate the expression of tight junctions in the mucosal gut barrier [33,34]. The microbiota of mice fed a HF diet were transferred to germ-free mice, with a notable increase in body fat. The microbiota of obese individuals, and of those fed a HF diet, are characterized by a decrease in Bacteroides and an increase in Firmicutes [23,33]. Depletion of Bacteroides results in dysbiosis via depletion of short-chain fatty acids. HF diets induce a pro-inflammatory state, decrease microbial diversity, promote intestinal colonization with pathobionts, exacerbate colitis severity in murine models, and are associated risk factors in patients with IBD [29,35,36,37,38,39,40,41,42].

The effect of dietary fatty acids on microbiota changes differ according to the type as each type has varied effects on the immune system and in­flammatory cascades. MUFAs, n-6 PUFAs, and n-3 PUFAs each play a different role in IBD pathogenesis [43].

2.2 MUFAs and PUFAs

The different roles of n-3 and n-6 PUFAs are demonstrated by their metabolism by-products. The metabolism of n-6 and n-3 fatty acids results in different effects on inflammatory cascades. Although there is some disagreement, effects of n-3 PUFAs on inflammation are generally considered anti-inflammatory, whereas n-6 PUFAs are pro-inflammatory [44]. N-3 PUFAs include eicosapentaenoic acid (EPA, C20:5) and docosahexaenoic acid (DHA, C22:6). PUFAs are important constituents in cell membranes and phospholipid bilayers. Increasing intake of PUFAs modifies cell membranes by increasing EPA and DHA [45]. By influencing the cell membrane constituents, PUFAs influence the function of cells involved in inflammation as well as alter the microbiome composition.

A large majority of the gut microbiota belongs to the phyla Bacteroidetes and Firmicutes, as well as the genera Lactobacillus and Bacteroides. The type of dietary fat is a major driver of these phyla and genera structures. In one study exploring the differences in the microbiota of murine models fed differing fat diets, the microbiota of the fish oil diet had higher levels of Lactobacillus as compared to those fed a lard diet [46]. The species Lactobacillus has been linked to reduced gut inflammation in IBD [35]. N-3 PUFA supplementation produces a positive impact on gut microbiota through changes in its common genera as evidenced in a recent randomized, open-label, crossover trial [47]. The study was 8 weeks in duration and supplementation consisted of 4 grams of mixed EPA/DHA supplementation with a washout period of 3 months. The study demonstrated a reversible increased abundance of several genera, including Bifidobacterium, Roseburia, and Lactobacillus with the n-3 PUFA interventions, all of which are butyrate-producing bacteria. No statistically significant changes were observed in the F/B phyla ratio. Butyrate-producing bacteria have been linked to favorable effects due to short-chain fatty acid (SCFA) production by the microbiota and restoration of epithelial barrier integrity in patients with IBD [48,49,50].

Other studies have considered whether certain ratios of fatty acids have more favorable effects on microbiome changes in an effort to optimize immune function. An increased n-6/n-3 ratio and a correlation to an increased incidence of IBD have been noted in several studies [51,52]. One study demonstrated an increased risk of developing UC in patients with high n-6 PUFA intake and a reduced risk with n-3 PUFA intake [51]. In a prospective study of women enrolled in the Nurses' Health Study cohorts, comprised of 269 incident cases of CD, and 338 incident cases of UC, greater intake of n-3 PUFAs was associated with a lower risk of UC.

An optimal ratio of n-3 to n-6 PUFAs is desirable as the former is associated with benefit in patients with IBD, and the latter with risk. One such study examined the relationship between multiple oil blends, to include both MUFAs and PUFAs [53]. The authors sought to investigate how multiple oil blends affect the intestinal microbiota in 25 persons after 1 month of treatment. The treatments included three diets with differing MUFA:PUFA ratios. The study demonstrated microbiota shifts at the lower taxonomical levels rather than at the phyla level, but the effect was more pronounced in the obese subjects. The MUFA-rich treatments increased the populations of Prevotella and Parabacteroides, and decreased Isobaculum. The two PUFA-rich diets favored an increase in Isobaculum. These genera have not been well studied so their function in an increased capacity has not been elucidated. However, these results suggest a profound effect of BMI on treatment effect in gut microbiota at lower taxonomical levels.

Higher BMI has been associated with a greater abundance of Firmicutes, and a lesser abundance of Bacteroidetes in phyla ratios. Moreover, a recent study investigating the influence of a fish oil enriched diet on intestinal tissues, taken from patients with IBD, found a significantly increased ratio of interleukin-1ra (the receptor antagonist of IL-1) resulting in a decrease in the pro-inflammatory IL-1 activation [54]. Bifidobacteria, Lactobacilli, Prevotella spp., and Roseburia spp., all known to be beneficial gut bacteria, were significantly increased whereas harmful gut bacteria were significantly decreased.

In summary, most studies indicate a beneficial effect of dietary n-3 PUFAs on the gut microbiome. Dietary n-3 PUFAs have shown to affect gut integrity, improve epithelial barrier function, optimize the F/B ratio, and increase the abundance of butyrate-producing bacteria thus favoring anti-inflammatory activity.

2.3 Omega-3 Supplementation in IBD

Supplementation of n-3 fatty acids has been employed in multiple RCTs in an attempt to replicate its perceived effectiveness in ameliorating inflammation. In a Cochrane meta-analysis, n-3 PUFA supplementation was suggested to aid in the maintenance of remission in CD [55]. It also noted an inverse relationship between n-3 consumption and the risk of IBD. However, when analyzing several of the larger studies to exclude inherent bias, the authors concluded n-3 PUFA supplementation was unlikely to be effective for the maintenance of remission in CD and was not better than placebo for preventing relapse [52]. In summary, dietary fish oil-derived n-3 PUFAs may affect disease outcomes in a beneficial manner for those with IBD. However, there is clearly a need for further study to better characterize how n-3 PUFAs may improve biomarkers, clinical outcomes, and restore a homeostatic microbiome.

2.4 Carbohydrates

Carbohydrates serve as a key source of nutrition for the microbiota as well as the host. Carbohydrates can be categorized as digestible and non-digestible. Digestible carbohydrates include both mono- and disaccharides as well as starches. Non-digestible carbohydrates are mainly found as components of dietary fiber. In general, high sugar diets are associated with an increased risk of UC and CD [56]. Large epidemiological studies have defined an association between both refined sugar and artificial sweeteners with IBD incidence [51]. A high sugar diet can alter the gut microbial composition and diminish SCFAs, a proposed mechanism to exacerbate colitis [56]. The proposed mechanism suggests as different carbohydrates are digested, an imbalance in intestinal absorption is created in the lumen, leading to growth of specific pathobionts [57]. In patients with IBD, carbohydrate malabsorption is a common observation and lends support to high carbohydrate diets favoring dysbiosis [36,57,58,59].

2.5 Short-Chain Fatty Acids

Fiber consists of soluble and insoluble fiber. Although non-digestible carbohydrates do resist digestion, they are able to provide the body with energy through fermentation. For instance, fiber can be fermented into SCFAs by the gut microbiota to produce carbon and energy. The SCFAs produced by gut bacteria are acetate, propionate, and butyrate. When SCFAs are produced by the gut microbiota, through a variety of anti-inflammatory local effects, they protect the host and have beneficial effects on energy metabolism. Butyrate and other SCFAs have anti-inflammatory effects that improve host immunity, mainly though promotion of optimal intestinal barrier function and integrity. Consequently, a lack of fiber may compromise intestinal barrier function and perpetuate IBD, the mechanism of which is still to be completely understood.

The gut microbiome in patients with IBD is associated with a depletion of SCFA-producing bacteria as evidenced in fecal and mucosal samples [56]. Among the dominant butyrate-producing taxa, the Ruminococcaceae and Lachnospiraceae families of the Firmicutes phylum have been noted to be depleted in those with IBD, as well as Faecalibacterium prausnitzii, and Roseburia hominis [60,61]. In IBD, other notable alterations have been noted in Eubacterium, Roseburia, Anaerostipes, and Coprococcus spp [62,63]. Research suggests a key genetic microbial inability to produce butyrate or a significant decrease of which is involved in IBD pathogenesis [62].

2.6 SCFA Supplementation in IBD

SCFAs have been employed in multiple RCTs with various delivery methods in an attempt to ameliorate IBD-related inflammation [64,65,66,67,68,69]. In a recent RCT, the authors sought to evaluate the colonic-delivery formulation of butyrate, and its effect on the fecal microbiota [68]. The study was designed as a double-blinded placebo-controlled study of 49 patients with IBD. Patients were randomized to receive microencapsulated sodium-butyrate or placebo for 2 months. Prior to treatment, patients with CD had a lower abundance of Feacalibacterium, Akkermansia, and Lachnospiraceae as compared to healthy controls. After treatment, in patients with CD, there was an increase in butyrogenic colonic bacteria Butyricicoccus. In patients with UC, there was a notable increase in Lachnospiraceae spp. However, the authors did not find any difference in terms of clinical outcome or disease activity. The authors also noted limitations of the study to include a small sample size, and the majority of the patients were not in active UC or CD. However, the study concluded oral butyrate did modulate the composition of the microbiome to more favorable ratios and increased SCFA production. This study establishes a possible directed intervention to optimizing outcomes in IBD through SCFA supplementation or butyrogenic bacteria.

2.7 Protein

Dietary proteins are derived from many different foods and represent a significant component in both animal and plant products. Animal-based proteins differ from plant-based proteins in terms of amino acid composition, nutritional value, bioavailability, quality, and quantity. Animal-based proteins tend to have a higher level of bioavailability and essential amino acids but also more saturated fat. High protein intake, mainly animal protein such as meat or fish but not eggs or dairy, is associated with an increased risk of UC and CD [70,71]. Animal-derived foods have consistently been associated with higher abundances of Firmicutes, Ruminococcus and pathways favoring endotoxin synthesis [72].

Consumption of certain amino acids has been studied in terms of immunomodulation, and pro- and anti- inflammatory cascades. Whereas consumption of glutamine, arginine, histidine, tryptophan, and threonine has been demonstrated in murine colitis models to improve clinical and biochemical markers [73,74,75,76,77,78,79,80,81]. High consumption of branched-chain amino acids (BCAAs) has been associated with oxidative stress while low consumption has an anti-inflammatory effect [82,83,84]. Moreover, consumption of certain amino acids may lead to dysbiosis and worsening of colitis [85].

3. Noteworthy Dietary Supplements and the Gut Microbiota: Curcumin, Vitamin D, and Polyphenols

3.1 Curcumin

Curcumin, found in the Indian spice turmeric, has long been purported to have anti-inflammatory medicinal properties [86]. Its antioxidant properties are largely due to potent inhibition of NF-κB activation and inhibition of TNF-mediated actions [87,88,89,90]. In the microbiome, its effects have been investigated in murine models as well as human models as to protect gut barrier integrity and increase species diversity [91,92]. In one study, curcumin supplementation ameliorated endotoxemia induced by a HF diet in rat models [93]. Another study illuminated how curcumin can increase the abundance of butyrate-producing bacteria, which led to improved gut barrier integrity [91]. The study found a significant increase in the species Clostridium. Another study found curcumin to increase the relative abundance of Lactobacillales [94].

In human models, a double-blinded, randomized pilot study used turmeric, curcumin, or a placebo (turmeric tablets contained 1000 mg turmeric root and the curcumin tablets contained 1000 mg of curcumin) for a total of 8 weeks and determined subsequent changes in the gut microbiota community architecture [92]. Among the individual participants, microbiome species diversity did increase although patterns in increase were not uniform among the subjects. Although both turmeric and curcumin altered the gut microbiota in a similar pattern. The authors describe the subjects in terms of “responders” and “non-responders.” The “responders” had an increase in Clostridium spp., Bacteroides spp., Citrobacter spp., Cronobacter spp., Enterobacter spp., Enterococcus spp., Klebsiella spp., Parabacteroides spp., and Pseudomonas spp. The authors postulated that these results suggest individual response to curcumin dependent on host characteristics. In previous studies [51], host BMI was a significant factor in phyla shift dynamics, although the authors did not comment on this characteristic. The results suggest subject-to-subject variation in response dynamics. These studies support the anti-inflammatory and antioxidant effects of curcumin, and their effect on the microbiome through down regulation of pro-inflammatory cytokines.

3.2 Curcumin Supplementation in IBD

In a randomized, placebo-controlled, double-blinded study, curcumin was given in combination with mesalamine to patients with mild to moderate UC [95]. The treatment group consisted of mesalamine and curcumin capsules (3 g/day) whereas the placebo group consisted of mesalamine and an identical placebo. Clinical remission rates were significant in the group with curcumin (53.8% versus 0%; P = 0.01). In addition, clinical response (65.3% versus 12.5%; P < 0.001), and endoscopic remission (38% versus 0%; P = 0.043) were also found to be significant as compared to mesalamine and placebo. The study demonstrates the efficacy of curcumin as an adjuvant treatment to standard therapy. These findings have been replicated and validate that the adjunctive use of curcumin with mesalamine versus the use of mesalamine alone in the treatment of UC increased the odds of clinical remission by threefold [96]. Studies have also demonstrated that curcumin has the ability to significantly improve rates of maintenance of remission in patients [97]. A recent meta-analysis showed curcumin significantly improved clinical and endoscopic remissions in patients with IBD [97]. These findings have not been replicated in those with CD. Further, although these studies demonstrate the ability of curcumin to induce remission in patients with active mild to moderate UC, demonstration of these findings in concordance with microbiome changes has not been studied.

3.3 Vitamin D

Vitamin D is a fat-soluble vitamin that is metabolized to form 25(OH)D3, and then hydroxylated to generate 1,25(OH)2D3, one of the active metabolites. The active vitamin D hormone binds to the vitamin D receptor (VDR). The VDR is highly expressed in the intestinal tract. Vitamin D has been found to be deficient in up to 60% of patients with IBD [98,99]. Vitamin D plays an important role in gut microbiota modulation [98,100,101]. Downregulation of the VDR has been associated with a decrease in Lactobacillus, and an increase in Proteobacteria [100]. Vitamin D mechanisms clearly modulate the gut microbiota, the significance of which will be discussed herein.

Vitamin D has been shown to correlate with disease activity in those with IBD. In a retrospective longitudinal study, serum vitamin D levels were inversely associated with inflammatory markers and clinical disease activity in patients with IBD [101]. Vitamin D protects the intestinal epithelial barrier and modulates the gut microbiota [100,101,102]. In addition, the VDR acts as a transcription regulator and is inversely correlated with markers of colonic inflammation [103]. VDR expression has been shown to affect the gut microbiome by promoting SCFA production and protecting intestinal barrier integrity. In murine models, vitamin D supplementation has demonstrated the ability to increase the microbiota species of Clostridium, Bacteroides, and Proteobacteria ssp [104]. In a large clinical study, there was a notable correlation between the active metabolite of vitamin D and butyrate producers [105]. Thus, it is possible that VDR, in combination with the active vitamin D metabolite, may inhibit the inflammatory response seen in IBD.

3.4 Vitamin D Supplementation in IBD

Vitamin D supplementation has been employed in several clinical trials with perceived potential to modulate the microbiota. In one study in patients with CD in remission, vitamin D supplementation resulted in significant shifts in gut microbiota composition [106]. In the study, an increase in the abundance of beneficial bacteria like Alistipes, Parabacteroides, Roseburia, and Faecalibacterium was observed. As previously mentioned, in patients with IBD, a reduced abundance of Faecalibacterium has been noted. It is an important commensal microbe that produces SCFAs. Moreover, in a prospective pilot study aimed to evaluate markers of intestinal inflammation and changes in the fecal microbiota, patients with active and inactive UC were supplemented with 40000 IU of vitamin D once weekly for 8 weeks [107]. The study showed an improvement in objective markers of inflammation following vitamin D replacement in patients with active UC. Across all participants, an increase in Clostridium, and Enterobacteriacae spp. was noted. However no overall change in fecal microbial diversity occurred following vitamin D supplementation. The authors suggested it is possible that vitamin D supplementation does not affect the gut microbiome, or that the difference between fecal and mucosal microbiotas was not observed in the small study. Optimization of vitamin D clearly may have effects on the immune system by improving the composition of the microbiome through a series of dynamic interactions, however larger clinical trials are warranted to draw this conclusion.

3.5 Polyphenols

Polyphenols are classified as organic chemicals and into five major classes (flavonoids, phenolic acids, tannins, lignans, and stilbenes). Their potential mechanism of ameliorating oxidative stress via anti-inflammatory properties makes them an attractive option for potential therapeutics. The link between IBD and dietary polyphenol has been variable in current literature. A large cohort study did not show an association of IBD and polyphenol intake, while other work has associated daily polyphenol intake (primarily in green tea) with decreased risk of IBD [108]. Resveratrol is a stilbene found in grapes and red wine associated with a lower risk for CD [109]. Epigallocatechin-3-gallate (EGCG), a flavonol found in teas, has been associated with a lower risk of both UC and CD [110,111,112].

It has been postulated that polyphenols exert antioxidant and anti-inflammatory effects mainly via mechanisms of regulating gut microbes [113]. EGCG has been associated with microbiome changes that may provide protection from IBD. One study evaluating efficacy of EGCG found an increase in abundance of Akkermansia and SCFAs, especially butyrate, in murine colitis models [112]. FMT from these treated mice into separate murine colitis models was demonstrated to reduce colitis and reproduce the microbiome compositional change and increase in SCFAs.

Resveratrol was found to increase microbial diversity, alter the F/B ratio, decrease the abundance of Enterococcus faecalis, and increase the abundance of Lactobacillus and Bifidobacterium [113]. In a study to explore the therapeutic efficacy of resveratrol on murine IBD models, resveratrol was found to significantly reduce disease activity indices and improve histologic alterations [114]. In another murine study, resveratrol treatment improved gut barrier integrity and ameliorated UC colitis models [115].

3.6 Polyphenol Supplementation in IBD

Polyphenol supplementation has been employed in two studies with potential to regulate disease activity [116]. In a pilot study, oral EGCG supplementation was associated with decreased UC activity index in comparison to controls [112]. In a randomized, double-blinded, placebo-controlled study, 50 patients with active UC supplemented their diets with 500 mg resveratrol or placebo for 6 weeks [116]. Serum inflammatory markers were assessed at baseline, and at the end of the study. Resveratrol supplementation led to a significant reduction in inflammatory markers. The authors concluded that supplementation with 500 mg resveratrol can modify disease activity in patients with UC. However, demonstration of these findings in concordance with microbiota changes has not been studied. Future studies and clinical trials are clearly needed to confirm the perceived potential of resveratrol, and its likely effect on the gut microbiome in humans. It has been noted that oral polyphenols have low bioavailability, when undergoing metabolism in the intestine the bioavailability of resveratrol is less than 1% [117,118,119]. It has also been suggested that rectal administration may provide benefit due to high direct mucosal exposure, although further study is needed to explore the clinical applications.

4. Diets and the Gut Microbiota

4.1 Notable Diets in IBD

Emerging data, through large epidemiolocal studies, demonstrate a typical Western diet (WD) is associated with the rising prevalence of IBD and systemic low-grade inflammation [120]. The WD is characterized by a high intake of processed and pre-packaged food items, as well as animal protein, dairy, grains, refined sugar, and low fiber foods. Such food items contain high amounts of n-6 PUFAs, saturated fatty acids, sugar, and sugar substitutes. Importantly, these food items also lack fiber, n-3 fatty acids, and plant polyphenols. As discussed previously, these dietary components are important for gut barrier integrity, intestinal microecology, and immune function. On the contrary, there is a negative association of IBD rates with the consumption of food items characteristic of the Mediterranean diet (MD) [121]. Although there are strong epidemiological associations, few interventional trials and clinical studies have been pursued given the inherent difficulty in studying dietary patterns. However, few are underway and will greatly add to our understanding, which will be discussed herein.

Historically, exclusive enteral nutrition (EEN) has proven efficacy in IBD [38,122,123,124,125,126]. EEN is a first line therapy in pediatric CD but is not routinely used as first-line therapy in adults as its application varies around the world [123,126,127,128,129]. EEN may induce remission via modulation of the gut microbiota, however the mechanisms by which are still not well understood. In adult patients with mild to moderate biologic naive CD, a dietary modification of EEN, titled the Crohn's disease exclusion diet (CDED) with or without partial enteral nutrition, was effective for induction and maintenance of remission [130]. The diet is highly restrictive and is designed to exclude food items that adversely affect the microbiome [130,131,132]. The specific carbohydrate diet (SCD) is a dietary intervention focused on consumption of specific carbohydrates, like monosaccharides, and excludes disaccharides, and most polysaccharides [133]. The SCD has been demonstrated to induce and maintain remission in pediatric patients with IBD albeit the majority of evidence is based on small case studies and adherence can be challenging [133,134,135,136,137,138].

In review of the ESPEN guidelines, there is no diet that is generally recommended for adult patients with IBD [139]. However, several diets may prove to be an effective and a viable option for adults. The low fermentable, oligo-, di-, monosaccharides, and polyols (FODMAPs) diets have shown promise in improving functional symptoms [140,141]. The low FODMAP diet is associated with improvement in IBD disease symptom severity according to a recent Cochrane review. However, the research does not support the diet affecting disease activity or inflammatory markers [140,141,142,143]. Large clinical studies have been completed and are also underway for the MD, which will be discussed herein [144,145,146,147].

The MD is characterized by vegetables, fish, nuts, seeds, extra-virgin olive oil, fruits, whole grains, dairy, as well as low amounts of red meat [148]. These food items are abundant in fiber, n-3 fatty acids, and plant polyphenols. These dietary components and high-level adherence to the MD influence the composition and function of the gut microbiome with a beneficial impact on inflammation and the metabolome [149,150]. Studies have found an interconnection between the MD and microbial metabolites, such as an increase in SCFAs and butyrate-producing bacteria [149,150]. The studies demonstrated an increase in Firmicutes, Bacteroidetes, Clostridium, and a decrease in the abundance of Proteobacteria and Bacillaceae. A recent controlled interventional study investigated dietary intake and gut microbiota in patients with CD in remission [151]. Fifteen patients with quiescent CD consuming a low plant-based, and high meat-based, diet received a 12-week structured diversified dietary intervention based on the MD. The study noted an increase in Faecalibacterium, and Bifidobacterium, and a decrease in Proteobacteria. These findings suggest the MD can induce positive changes in the gut microbiota, implying a mechanism by which diet induces remission in IBD.

4.2 Conclusion

While the exact etiology of IBD is unknown, there exists an imbalance of pro-inflammatory and anti-inflammatory mediators which modulate immune responses to environmental factors in a genetically susceptible host. The wide-ranging effect of the microbiome is imperative in this illustration as diet influences the evolution of host-specific changes in the microbial community. Interestingly, the specific dietary factors responsible are not yet well defined. However, certain dietary patterns such as the MD have been shown to direct toward immunologic optimization, intestinal integrity, and a balanced microbiome. Further, several noteworthy dietary supplements also appear to hold significant therapeutic potential for IBD management approaches.

Author Contributions

Dr. Johnson DA contributed to the construction of the project; all authors wrote and edited the manuscript.

Competing Interests

The authors have declared that no competing interests exist.

References

  1. Alatab S, Sepanlou SG, Ikuta K, Vahedi H, Bisignano C, Safiri S, et al. The global, regional, and national burden of inflammatory bowel disease in 195 countries and territories, 1990-2017: A systematic analysis for the global burden of disease study 2017. Lancet Gastroenterol Hepatol. 2020; 5: 17-30. [CrossRef]
  2. Shivashankar R, Tremaine WJ, Harmsen WS, Loftus Jr EV. Incidence and prevalence of Crohn’s disease and ulcerative colitis in Olmsted County, Minnesota from 1970 through 2010. Clin Gastroenterol Hepatol. 2017; 15: 857-863. [CrossRef]
  3. Xu F, Liu Y, Wheaton AG, Rabarison KM, Croft JB. Trends and factors associated with hospitalization costs for inflammatory bowel disease in the United States. Appl Health Econ Health Policy. 2019; 17: 77-91. [CrossRef]
  4. Ye Y, Pang Z, Chen W, Ju S, Zhou C. The epidemiology and risk factors of inflammatory bowel disease. Int J Clin Exp Med. 2015; 8: 22529-22542.
  5. Ranasinghe IR, Hsu R. Crohn Disease. In: Pediatric Gastroenterology. StatPearls Publishing; 2021. pp.205-214.
  6. Weisiger RA, Bilhartz LE, Sleisenger MH, Fordtran JS. Sleisenger & Fordtran's Gastrointestinal and liver disease: review and assessment.‎ Philadelphia: W.B. Saunders Ltd.; 1999.
  7. Targan SR, Hanauer SB, van Deventer SJ, Mayer L, Present DH, Braakman T, et al. A short-term study of chimeric monoclonal antibody cA2 to tumor necrosis factor alpha for Crohn’s disease. Crohn's Disease cA2 Study Group. N Engl J Med. 1997; 337: 1029-1035. [CrossRef]
  8. Ogura Y, Bonen DK, Inohara N, Nicolae DL, Chen FF, Ramos R, et al. A frameshift mutation in NOD2 associated with susceptibility to Crohn’s disease. Nature. 2001; 411: 603-606. [CrossRef]
  9. Parolini C, Chiesa G, Gong E, Caligari S, Cortese MM, Koga T, et al. Apolipoprotein A-I and the molecular variant apoA-I (Milano): Evaluation of the antiatherogenic effects in knock-in mouse model. Atherosclerosis. 2005; 183: 222-229. [CrossRef]
  10. Barrett JC, Lee JC, Lees CW, Prescott NJ, Anderson CA, Phillips A, et al. Genome-wide association study of ulcerative colitis identifies three new susceptibility loci, including the HNF4A region. Nat Genet. 2009; 41: 1330-1334. [CrossRef]
  11. Cavanaugh J. NOD2: Ethnic and geographic differences. World J Gastroenterol. 2006; 12: 3673-3677. [CrossRef]
  12. Gajendran M, Loganathan P, Jimenez G, Catinella AP, Ng N, Umapathy C, et al. A comprehensive review and update on ulcerative colitis. Dis Mon. 2019; 65: 100851. [CrossRef]
  13. Hedayat KM, Lapraz JC, Schuff B. Ulcerative Colitis. In: The theory of endobiogeny: Volume 4: Bedside handbook. London: Academic Press; 2019. [CrossRef]
  14. Kappelman MD, Rifas-Shiman SL, Kleinman K, Ollendorf D, Bousvaros A, Grand RJ, et al. The prevalence and geographic distribution of Crohn’s disease and ulcerative colitis in the United States. Clin Gastroenterol Hepatol. 2007; 5: 1424-1429. [CrossRef]
  15. Rinninella E, Raoul P, Cintoni M, Franceschi F, Miggiano GAD, Gasbarrini A, et al. What is the healthy gut microbiota composition? A changing ecosystem across age, environment, diet, and diseases. Microorganisms. 2019; 7: 14. [CrossRef]
  16. Qin J, Li R, Raes J, Arumugam M, Burgdorf KS, Manichanh C, et al. A human gut microbial gene catalogue established by metagenomic sequencing. Nature. 2010; 464: 59-65. [CrossRef]
  17. Arrieta MC, Stiemsma LT, Amenyogbe N, Brown EM, Finlay B. The intestinal microbiome in early life: Health and disease. Front Immunol. 2014; 5: 427. [CrossRef]
  18. Gacesa R, Kurilshikov A, Vich Vila A, Sinha T, Klaassen MAY, Bolte LA, et al. Environmental factors shaping the gut microbiome in a Dutch population. Nature. 2022; 604: 732-739. [CrossRef]
  19. Chen H, Li H, Liu Z. Interplay of intestinal microbiota and mucosal immunity in inflammatory bowel disease: A relationship of frenemies. Therap Adv Gastroenterol. 2020; 13: 1756284820935188. [CrossRef]
  20. D'Souza SM, Houston K, Keenan L, Yoo BS, Parekh PJ, Johnson DA. Role of microbial dysbiosis in the pathogenesis of esophageal mucosal disease: A paradigm shift from acid to bacteria? World J Gastroenterol. 2021; 27: 2054-2072. [CrossRef]
  21. Carding S, Verbeke K, Vipond DT, Corfe BM, Owen LJ. Dysbiosis of the gut microbiota in disease. Microb Ecol Health Dis. 2015; 26: 26191. [CrossRef]
  22. Petersen C, Round JL. Defining dysbiosis and its influence on host immunity and disease. Cell Microbiol. 2014; 16: 1024-1033. [CrossRef]
  23. Stojanov S, Berlec A, Štrukelj B. The influence of probiotics on the firmicutes/bacteroidetes ratio in the treatment of obesity and inflammatory bowel disease. Microorganisms. 2020; 8: 1715. [CrossRef]
  24. Serban DE. Microbiota in inflammatory bowel disease pathogenesis and therapy. Nutr Clin Pract. 2015; 30: 760-779. [CrossRef]
  25. Manzini S, Pinna C, Busnelli M, Cinquanta P, Rigamonti E, Ganzetti GS, et al. Beta2-adrenergic activity modulates vascular tone regulation in lecithin: Cholesterol acyltransferase knockout mice. Vascul Pharmacol. 2015; 74: 114-121. [CrossRef]
  26. Vester-Andersen MK, Mirsepasi-Lauridsen HC, Prosberg MV, Mortensen CO, Träger C, Skovsen K, et al. Increased abundance of proteobacteria in aggressive Crohn’s disease seven years after diagnosis. Sci Rep. 2019; 9: 13473. [CrossRef]
  27. Usuda H, Okamoto T, Wada K. Leaky gut: Effect of dietary fiber and fats on microbiome and intestinal barrier. Int J Mol Sci. 2021; 22: 7613. [CrossRef]
  28. Silverberg MS, Satsangi J, Ahmad T, Arnott ID, Bernstein CN, Brant SR, et al. Toward an integrated clinical, molecular and serological classification of inflammatory bowel disease: Report of a working party of the 2005 Montreal world congress of gastroenterology. Can J Gastroenterol. 2005; 19 Suppl A: 5A-36A. [CrossRef]
  29. Cani PD, Bibiloni R, Knauf C, Waget A, Neyrinck AM, Delzenne NM, et al. Changes in gut microbiota control metabolic endotoxemia-induced inflammation in high-fat diet-induced obesity and diabetes in mice. Diabetes. 2008; 57: 1470-1481. [CrossRef]
  30. Cani PD, Amar J, Iglesias MA, Poggi M, Knauf C, Bastelica D, et al. Metabolic endotoxemia initiates obesity and insulin resistance. Diabetes. 2007; 56: 1761-1772. [CrossRef]
  31. Turnbaugh PJ, Ley RE, Mahowald MA, Magrini V, Mardis ER, Gordon JI. An obesity-associated gut microbiome with increased capacity for energy harvest. Nature. 2006; 444: 1027-1031. [CrossRef]
  32. Velasquez MT. Altered gut microbiota: A link between diet and the metabolic syndrome. Metab Syndr Relat Disord. 2018; 16: 321-328. [CrossRef]
  33. Hildebrandt MA, Hoffmann C, Sherrill-Mix SA, Keilbaugh SA, Hamady M, Chen YY, et al. High-fat diet determines the composition of the murine gut microbiome independently of obesity. Gastroenterology. 2009; 137: 1716-1724.e2. [CrossRef]
  34. Fujisaka S, Avila-Pacheco J, Soto M, Kostic A, Dreyfuss JM, Pan H, et al. Diet, genetics, and the gut microbiome drive dynamic changes in plasma metabolites. Cell Rep. 2018; 22: 3072-3086. [CrossRef]
  35. David LA, Maurice CF, Carmody RN, Gootenberg DB, Button JE, Wolfe BE, et al. Diet rapidly and reproducibly alters the human gut microbiome. Nature. 2014; 505: 559-563. [CrossRef]
  36. Martinez-Medina M, Denizot J, Dreux N, Robin F, Billard E, Bonnet R, et al. Western diet induces dysbiosis with increased E coli in CEABAC10 mice, alters host barrier function favouring AIEC colonisation. Gut. 2014; 63: 116-124. [CrossRef]
  37. Levine A, Sigall Boneh R, Wine E. Evolving role of diet in the pathogenesis and treatment of inflammatory bowel diseases. Gut. 2018; 67: 1726-1738. [CrossRef]
  38. Levine A, Milo T, Buller H, Markowitz J. Consensus and controversy in the management of pediatric Crohn disease: An international survey. J Pediatr Gastroenterol Nutr. 2003; 36: 464-469. [CrossRef]
  39. Malesza IJ, Malesza M, Walkowiak J, Mussin N, Walkowiak D, Aringazina R, et al. High-fat, western-style diet, systemic inflammation, and gut microbiota: A narrative review. Cells. 2021; 10: 3164. [CrossRef]
  40. Rohr MW, Narasimhulu CA, Rudeski-Rohr TA, Parthasarathy S. Negative effects of a high-fat diet on intestinal permeability: A review. Adv Nutr. 2020; 11: 77-91. [CrossRef]
  41. Schulz MD, Atay C, Heringer J, Romrig FK, Schwitalla S, Aydin B, et al. High-fat-diet-mediated dysbiosis promotes intestinal carcinogenesis independently of obesity. Nature. 2014; 514: 508-512. [CrossRef]
  42. Kim KA, Gu W, Lee IA, Joh EH, Kim DH. High fat diet-induced gut microbiota exacerbates inflammation and obesity in mice via the TLR4 signaling pathway. PLoS One. 2012; 7: e47713. [CrossRef]
  43. Patterson E, O’Doherty RM, Murphy EF, Wall R, O’Sullivan O, Nilaweera K, et al. Impact of dietary fatty acids on metabolic activity and host intestinal microbiota composition in C57BL/6J mice. Br J Nutr. 2014; 111: 1905-1917. [CrossRef]
  44. Bagga D, Wang L, Farias-Eisner R, Glaspy JA, Reddy ST. Differential effects of prostaglandin derived from omega-6 and omega-3 polyunsaturated fatty acids on COX-2 expression and IL-6 secretion. Proc Natl Acad Sci U S A. 2003; 100: 1751-1756. [CrossRef]
  45. Chiesa G, Busnelli M, Manzini S, Parolini C. Nutraceuticals and bioactive components from fish for dyslipidemia and cardiovascular risk reduction. Mar Drugs. 2016; 14: 113. [CrossRef]
  46. Caesar R, Tremaroli V, Kovatcheva-Datchary P, Cani PD, Bäckhed F. Crosstalk between gut microbiota and dietary lipids aggravates wat inflammation through TLR signaling. Cell Metab. 2015; 22: 658-668. [CrossRef]
  47. Watson H, Mitra S, Croden FC, Taylor M, Wood HM, Perry SL, et al. A randomised trial of the effect of omega-3 polyunsaturated fatty acid supplements on the human intestinal microbiota. Gut. 2018; 67: 1974-1983. [CrossRef]
  48. Geirnaert A, Calatayud M, Grootaert C, Laukens D, Devriese S, Smagghe G, et al. Butyrate-producing bacteria supplemented in vitro to Crohn’s disease patient microbiota increased butyrate production and enhanced intestinal epithelial barrier integrity. Sci Rep. 2017; 7: 11450. [CrossRef]
  49. Sun M, Wu W, Liu Z, Cong Y. Microbiota metabolite short chain fatty acids, GPCR, and inflammatory bowel diseases. J Gastroenterol. 2017; 52: 1-8. [CrossRef]
  50. Costantini L, Molinari R, Farinon B, Merendino N. Impact of omega-3 fatty acids on the gut microbiota. Int J Mol Sci. 2017; 18: 2645. [CrossRef]
  51. Racine A, Carbonnel F, Chan SSM, Hart AR, Bueno-de-Mesquita HB, Oldenburg B, et al. Dietary patterns and risk of inflammatory bowel disease in Europe: Results from the epic study. Inflamm Bowel Dis. 2016; 22: 345-354. [CrossRef]
  52. Feagan BG, Sandborn WJ, Mittmann U, Bar-Meir S, D'Haens G, Bradette M, et al. Omega-3 free fatty acids for the maintenance of remission in Crohn disease: The epic randomized controlled trials. JAMA. 2008; 299: 1690-1697. [CrossRef]
  53. Pu S, Khazanehei H, Jones PJ, Khafipour E. Interactions between obesity status and dietary intake of monounsaturated and polyunsaturated oils on human gut microbiome profiles in the canola oil multicenter intervention trial (COMIT). Front Microbiol. 2016; 7: 1612. [CrossRef]
  54. Meister D, Ghosh S. Effect of fish oil enriched enteral diet on inflammatory bowel disease tissues in organ culture: Differential effects on ulcerative colitis and Crohn’s disease. World J Gastroenterol. 2005; 11: 7466-7472. [CrossRef]
  55. Lev-Tzion R, Griffiths AM, Leder O, Turner D. Omega 3 fatty acids (fish oil) for maintenance of remission in Crohn’s disease. Cochrane Database Syst Rev. 2014; 2014: CD006320. [CrossRef]
  56. Laffin M, Fedorak R, Zalasky A, Park H, Gill A, Agrawal A, et al. A high-sugar diet rapidly enhances susceptibility to colitis via depletion of luminal short-chain fatty acids in mice. Sci Rep. 2019; 9: 12294. [CrossRef]
  57. Dixon LJ, Kabi A, Nickerson KP, McDonald C. Combinatorial effects of diet and genetics on inflammatory bowel disease pathogenesis. Inflamm Bowel Dis. 2015; 21: 912-922. [CrossRef]
  58. Barrett JS, Irving PM, Shepherd SJ, Muir JG, Gibson PR. Comparison of the prevalence of fructose and lactose malabsorption across chronic intestinal disorders. Aliment Pharmacol Ther. 2009; 30: 165-174. [CrossRef]
  59. Mentella MC, Scaldaferri F, Pizzoferrato M, Gasbarrini A, Miggiano GAD. Nutrition, IBD and gut microbiota: A review. Nutrients. 2020; 12: 944. [CrossRef]
  60. Machiels K, Joossens M, Sabino J, De Preter V, Arijs I, Eeckhaut V, et al. A decrease of the butyrate-producing species Roseburia hominis and Faecalibacterium prausnitzii defines dysbiosis in patients with ulcerative colitis. Gut. 2014; 63: 1275-1283. [CrossRef]
  61. Gill PA, van Zelm MC, Muir JG, Gibson PR. Review article: Short chain fatty acids as potential therapeutic agents in human gastrointestinal and inflammatory disorders. Aliment Pharmacol Ther. 2018; 48: 15-34. [CrossRef]
  62. Paramsothy S, Nielsen S, Kamm MA, Deshpande NP, Faith JJ, Clemente JC, et al. Specific bacteria and metabolites associated with response to fecal microbiota transplantation in patients with ulcerative colitis. Gastroenterology. 2019; 156: 1440-1454.e2. [CrossRef]
  63. Deleu S, Machiels K, Raes J, Verbeke K, Vermeire S. Short chain fatty acids and its producing organisms: An overlooked therapy for IBD? EBioMedicine. 2021; 66: 103293. [CrossRef]
  64. Banasiewicz T, Krokowicz Ł, Stojcev Z, Kaczmarek BF, Kaczmarek E, Maik J, et al. Microencapsulated sodium butyrate reduces the frequency of abdominal pain in patients with irritable bowel syndrome. Colorectal Dis. 2013; 15: 204-209. [CrossRef]
  65. Vanhoutvin SALW, Troost FJ, Kilkens TOC, Lindsey PJ, Hamer HM, Jonkers DMAE, et al. The effects of butyrate enemas on visceral perception in healthy volunteers. Neurogastroenterol Motil. 2009; 21: 952-e976. [CrossRef]
  66. Kannampalli P, Shaker R, Sengupta JN. Colonic butyrate- algesic or analgesic? Neurogastroenterol Motil. 2011; 23: 975-979. [CrossRef]
  67. Vernia P, Annese V, Bresci G, d'Albasio G, D'Incà R, Giaccari S, 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. [CrossRef]
  68. Facchin S, Vitulo N, Calgaro M, Buda A, Romualdi C, Pohl D, et al. Microbiota changes induced by microencapsulated sodium butyrate in patients with inflammatory bowel disease. Neurogastroenterol Motil. 2020; 32: e13914. [CrossRef]
  69. Di Sabatino A, Morera R, Ciccocioppo R, Cazzola P, Gotti S, Tinozzi FP, et al. Oral butyrate for mildly to moderately active Crohn’s disease. Aliment Pharmacol Ther. 2005; 22: 789-794. [CrossRef]
  70. 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. [CrossRef]
  71. Forbes A, Escher J, Hébuterne X, Kłęk S, Krznaric Z, Schneider S, et al. Espen guideline: Clinical nutrition in inflammatory bowel disease. Clin Nutr. 2017; 36: 321-347. [CrossRef]
  72. Bolte LA, Vich Vila A, Imhann F, Collij V, Gacesa R, Peters V, et al. Long-term dietary patterns are associated with pro-inflammatory and anti-inflammatory features of the gut microbiome. Gut. 2021; 70: 1287-1298. [CrossRef]
  73. Giriş M, Erbil Y, Doğru-Abbasoğlu S, Yanik BT, Aliş H, Olgaç V, et al. The effect of heme oxygenase-1 induction by glutamine on TNBS-induced colitis. The effect of glutamine on TNBS colitis. Int J Colorectal Dis. 2007; 22: 591-599. [CrossRef]
  74. Xue H, Sufit AJD, Wischmeyer PE. Glutamine therapy improves outcome of in vitro and in vivo experimental colitis models. JPEN J Parenter Enteral Nutr. 2011; 35: 188-197. [CrossRef]
  75. Ren W, Yin J, Wu M, Liu G, Yang G, Xion Y, et al. Serum amino acids profile and the beneficial effects of L-arginine or L-glutamine supplementation in dextran sulfate sodium colitis. PLoS One. 2014; 9: e88335. [CrossRef]
  76. Gao J, Xu K, Liu H, Liu G, Bai M, Peng C, et al. Impact of the gut microbiota on intestinal immunity mediated by tryptophan metabolism. Front Cell Infect Microbiol. 2018; 8: 13. [CrossRef]
  77. Islam J, Sato S, Watanabe K, Watanabe T, Ardiansyah, Hirahara K, et al. Dietary tryptophan alleviates dextran sodium sulfate-induced colitis through aryl hydrocarbon receptor in mice. J Nutr Biochem. 2017; 42: 43-50. [CrossRef]
  78. Nikolaus S, Schulte B, Al-Massad N, Thieme F, Schulte DM, Bethge J, et al. Increased tryptophan metabolism is associated with activity of inflammatory bowel diseases. Gastroenterology. 2017; 153: 1504-1516.e2. [CrossRef]
  79. Andou A, Hisamatsu T, Okamoto S, Chinen H, Kamada N, Kobayashi T, et al. Dietary histidine ameliorates murine colitis by inhibition of proinflammatory cytokine production from macrophages. Gastroenterology. 2009; 136: 564-574.e2. [CrossRef]
  80. Coburn LA, Gong X, Singh K, Asim M, Scull BP, Allaman MM, et al. L-arginine supplementation improves responses to injury and inflammation in dextran sulfate sodium colitis. PLoS One. 2012; 7: e33546. [CrossRef]
  81. Faure M, Mettraux C, Moennoz D, Godin JP, Vuichoud J, Rochat F, et al. Specific amino acids increase mucin synthesis and microbiota in dextran sulfate sodium-treated rats. J Nutr. 2006; 136: 1558-1564. [CrossRef]
  82. Zhenyukh O, Civantos E, Ruiz-Ortega M, Sánchez MS, Vázquez C, Peiró C, et al. High concentration of branched-chain amino acids promotes oxidative stress, inflammation and migration of human peripheral blood mononuclear cells via mTORC1 activation. Free Radic Biol Med. 2017; 104: 165-177. [CrossRef]
  83. Lee JH, Park E, Jin HJ, Lee Y, Choi SJ, Lee GW, et al. Anti-inflammatory and anti-genotoxic activity of branched chain amino acids (BCAA) in lipopolysaccharide (LPS) stimulated RAW 264.7 macrophages. Food Sci Biotechnol. 2017; 26: 1371-1377. [CrossRef]
  84. He L, Zhang J, Zhao J, Ma N, Kim SW, Qiao S, et al. Autophagy: The last defense against cellular nutritional stress. Adv Nutr. 2018; 9: 493-504. [CrossRef]
  85. Ni J, Shen TCD, Chen EZ, Bittinger K, Bailey A, Roggiani M, et al. A role for bacterial urease in gut dysbiosis and Crohn’s disease. Sci Transl Med. 2017; 9: eaah6888. [CrossRef]
  86. Priyadarsini KI. The chemistry of curcumin: From extraction to therapeutic agent. Molecules. 2014; 19: 20091-20112. [CrossRef]
  87. Lestari MLAD, Indrayanto G. Curcumin. Profiles Drug Subst Excip Relat Methodol. 2014; 39: 113-204. [CrossRef]
  88. Wright LE, Frye JB, Gorti B, Timmermann BN, Funk JL. Bioactivity of turmeric-derived curcuminoids and related metabolites in breast cancer. Curr Pharm Des. 2013; 19: 6218-6225. [CrossRef]
  89. Singh S, Aggarwal BB. Activation of transcription factor NF-kappa B is suppressed by curcumin (diferuloylmethane) [corrected]. J Biol Chem. 1995; 270: 24995-25000. [CrossRef]
  90. Xu YX, Pindolia KR, Janakiraman N, Noth CJ, Chapman RA, Gautam SC. Curcumin, a compound with anti-inflammatory and anti-oxidant properties, down-regulates chemokine expression in bone marrow stromal cells. Exp Hematol. 1997; 25: 413-422.
  91. Ohno M, Nishida A, Sugitani Y, Nishino K, Inatomi O, Sugimoto M, et al. Nanoparticle curcumin ameliorates experimental colitis via modulation of gut microbiota and induction of regulatory T cells. PLoS One. 2017; 12: e0185999. [CrossRef]
  92. Peterson CT, Vaughn AR, Sharma V, Chopra D, Mills PJ, Peterson SN, et al. Effects of turmeric and curcumin dietary supplementation on human gut microbiota: A double-blind, randomized, placebo-controlled pilot study. J Evid Based Integr Med. 2018; 23: 2515690x18790725. [CrossRef]
  93. Feng W, Wang H, Zhang P, Gao C, Tao J, Ge Z, et al. Modulation of gut microbiota contributes to curcumin-mediated attenuation of hepatic steatosis in rats. Biochim Biophys Acta Gen Subj. 2017; 1861: 1801-1812. [CrossRef]
  94. Larmonier CB, Laubitz D, Hill FM, Shehab KW, Lipinski L, Midura-Kiela MT, et al. Reduced colonic microbial diversity is associated with colitis in NHE3-deficient mice. Am J Physiol Gastrointest Liver Physiol. 2013; 305: G667-G677. [CrossRef]
  95. Lang A, Salomon N, Wu JCY, Kopylov U, Lahat A, Har-Noy O, et al. Curcumin in combination with mesalamine induces remission in patients with mild-to-moderate ulcerative colitis in a randomized controlled trial. Clin Gastroenterol Hepatol. 2015; 13: 1444-1449.e1. [CrossRef]
  96. Chandan S, Mohan BP, Chandan OC, Ahmad R, Challa A, Tummala H, et al. Curcumin use in ulcerative colitis: Is it ready for prime time? A systematic review and meta-analysis of clinical trials. Ann Gastroenterol. 2020; 33: 53-58. [CrossRef]
  97. Shahinfar H, Payandeh N, ElhamKia M, Abbasi F, Alaghi A, Djafari F, et al. Administration of dietary antioxidants for patients with inflammatory bowel disease: A systematic review and meta-analysis of randomized controlled clinical trials. Complement Ther Med. 2021; 63: 102787. [CrossRef]
  98. Caviezel D, Maissen S, Niess JH, Kiss C, Hruz P. High prevalence of vitamin D deficiency among patients with inflammatory bowel disease. Inflamm Intest Dis. 2018; 2: 200-210. [CrossRef]
  99. Krasinski SD, Russell RM, Furie BC, Kruger SF, Jacques PF, Furie B. The prevalence of vitamin K deficiency in chronic gastrointestinal disorders. Am J Clin Nutr. 1985; 41: 639-643. [CrossRef]
  100. Battistini C, Ballan R, Herkenhoff ME, Saad SMI, Sun J. Vitamin D modulates intestinal microbiota in inflammatory bowel diseases. Int J Mol Sci. 2020; 22: 362. [CrossRef]
  101. López-Muñoz P, Beltrán B, Sáez-González E, Alba A, Nos P, Iborra M. Influence of vitamin D deficiency on inflammatory markers and clinical disease activity in IBD patients. Nutrients. 2019; 11: 1059. [CrossRef]
  102. Del Pinto R, Pietropaoli D, Chandar AK, Ferri C, Cominelli F. Association between inflammatory bowel disease and vitamin D deficiency: A systematic review and meta-analysis. Inflamm Bowel Dis. 2015; 21: 2708-2717. [CrossRef]
  103. Garg M, Royce SG, Tikellis C, Shallue C, Sluka P, Wardan H, et al. The intestinal vitamin D receptor in inflammatory bowel disease: Inverse correlation with inflammation but no relationship with circulating vitamin D status. Therap Adv Gastroenterol. 2019; 12: 1756284818822566. [CrossRef]
  104. Ooi JH, Li Y, Rogers CJ, Cantorna MT. Vitamin D regulates the gut microbiome and protects mice from dextran sodium sulfate-induced colitis. J Nutr. 2013; 143: 1679-1686. [CrossRef]
  105. Kado D. Vitamin D metabolites and the gut microbiome [Internet]. Nature Portfolio Microbiology Community; 2020 [cited date 2022 April 17]. Available from: https://microbiologycommunity.nature.com/posts/vitamin-d-metabolites-and-the-gut-microbiome.
  106. Schäffler H, Herlemann DP, Klinitzke P, Berlin P, Kreikemeyer B, Jaster R, et al. Vitamin D administration leads to a shift of the intestinal bacterial composition in Crohn’s disease patients, but not in healthy controls. J Dig Dis. 2018; 19: 225-234. [CrossRef]
  107. Garg M, Hendy P, Ding JN, Shaw S, Hold G, Hart A. The effect of vitamin D on intestinal inflammation and faecal microbiota in patients with ulcerative colitis. J Crohns Colitis. 2018; 12: 963-972. [CrossRef]
  108. Lu Y, Zamora-Ros R, Chan S, Cross AJ, Ward H, Jakszyn P, et al. Dietary polyphenols in the aetiology of Crohn’s disease and ulcerative colitis-a multicenter European prospective cohort study (EPIC). Inflamm Bowel Dis. 2017; 23: 2072-2082. [CrossRef]
  109. Xiao Q, Zhu W, Feng W, Lee SS, Leung AW, Shen J, et al. A review of resveratrol as a potent chemoprotective and synergistic agent in cancer chemotherapy. Front Pharmacol. 2019; 9: 1534. [CrossRef]
  110. Brückner M, Westphal S, Domschke W, Kucharzik T, Lügering A. Green tea polyphenol epigallocatechin-3-gallate shows therapeutic antioxidative effects in a murine model of colitis. J Crohns Colitis. 2012; 6: 226-235. [CrossRef]
  111. Oz HS, Chen T, de Villiers WJS. Green tea polyphenols and sulfasalazine have parallel anti-inflammatory properties in colitis models. Front Immunol. 2013; 4: 132. [CrossRef]
  112. Wu Z, Huang S, Li T, Li N, Han D, Zhang B, et al. Gut microbiota from green tea polyphenol-dosed mice improves intestinal epithelial homeostasis and ameliorates experimental colitis. Microbiome. 2021; 9: 184. [CrossRef]
  113. Qiao Y, Sun J, Xia S, Tang X, Shi Y, Le G. Effects of resveratrol on gut microbiota and fat storage in a mouse model with high-fat-induced obesity. Food Funct. 2014; 5: 1241-1249. [CrossRef]
  114. Yao J, Wei C, Wang JY, Zhang R, Li YX, Wang LS. Effect of resveratrol on Treg/Th17 signaling and ulcerative colitis treatment in mice. World J Gastroenterol. 2015; 21: 6572-6581. [CrossRef]
  115. Pan HH, Zhou XX, Ma YY, Pan WS, Zhao F, Yu MS, et al. Resveratrol alleviates intestinal mucosal barrier dysfunction in dextran sulfate sodium-induced colitis mice by enhancing autophagy. World J Gastroenterol. 2020; 26: 4945-4959. [CrossRef]
  116. Samsami-Kor M, Daryani NE, Asl PR, Hekmatdoost A. Anti-inflammatory effects of resveratrol in patients with ulcerative colitis: A randomized, double-blind, placebo-controlled pilot study. Arch Med Res. 2015; 46: 280-285. [CrossRef]
  117. Walle T, Hsieh F, DeLegge MH, Oatis Jr JE, Walle UK. High absorption but very low bioavailability of oral resveratrol in humans. Drug Metab Dispos. 2004; 32: 1377-1382. [CrossRef]
  118. Wenzel E, Somoza V. Metabolism and bioavailability of trans-resveratrol. Mol Nutr Food Res. 2005; 49: 472-481. [CrossRef]
  119. Walle T. Bioavailability of resveratrol. Ann N Y Acad Sci. 2011; 1215: 9-15. [CrossRef]
  120. Ng SC, Shi HY, Hamidi N, Underwood FE, Tang W, Benchimol EI, et al. Worldwide incidence and prevalence of inflammatory bowel disease in the 21st century: A systematic review of population-based studies. Lancet. 2017; 390: 2769-2778. [CrossRef]
  121. 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. [CrossRef]
  122. Narula N, Dhillon A, Zhang D, Sherlock ME, Tondeur M, Zachos M. Enteral nutritional therapy for induction of remission in Crohn’s disease. Cochrane Database Syst Rev. 2018; 4: CD000542. [CrossRef]
  123. Ashton JJ, Gavin J, Beattie RM. Exclusive enteral nutrition in Crohn’s disease: Evidence and practicalities. Clin Nutr. 2019; 38: 80-89. [CrossRef]
  124. Heuschkel RB, Menache CC, Megerian JT, Baird AE. Enteral nutrition and corticosteroids in the treatment of acute Crohn’s disease in children. J Pediatr Gastroenterol Nutr. 2000; 31: 8-15. [CrossRef]
  125. Alhagamhmad MH. Enteral nutrition in the management of Crohn’s disease: Reviewing mechanisms of actions and highlighting potential venues for enhancing the efficacy. Nutr Clin Pract. 2018; 33: 483-492. [CrossRef]
  126. Wall CL, Day AS, Gearry RB. Use of exclusive enteral nutrition in adults with Crohn’s disease: A review. World J Gastroenterol. 2013; 19: 7652-7660. [CrossRef]
  127. Senussi NH. Exclusive and partial enteral nutrition for Crohn’s disease. Lancet. 2017; 390: 1486. [CrossRef]
  128. Grover Z, Muir R, Lewindon P. Exclusive enteral nutrition induces early clinical, mucosal and transmural remission in paediatric Crohn’s disease. J Gastroenterol. 2014; 49: 638-645. [CrossRef]
  129. Pfeffer-Gik T, Yanai HA, Godny L, Ron Y, Maharshak N, Dotan I. Exclusive enteral nutrition in adults with active Crohn’s disease is associated with decreased disease activity. Gastroenterology. 2017; 152: S399. [CrossRef]
  130. Yanai H, Levine A, Hirsch A, Boneh RS, Kopylov U, Eran HB, et al. The Crohn’s disease exclusion diet for induction and maintenance of remission in adults with mild-to-moderate Crohn’s disease (CDED-AD): An open-label, pilot, randomised trial. Lancet Gastroenterol Hepatol. 2022; 7: 49-59. [CrossRef]
  131. Sigall-Boneh R, Pfeffer-Gik T, Segal I, Zangen T, Boaz M, Levine A. Partial enteral nutrition with a Crohn’s disease exclusion diet is effective for induction of remission in children and young adults with Crohn’s disease. Inflamm Bowel Dis. 2014; 20: 1353-1360. [CrossRef]
  132. Levine A, Wine E, Assa A, Sigall Boneh R, Shaoul R, Kori M, et al. Crohn's disease exclusion diet plus partial enteral nutrition induces sustained remission in a randomized controlled trial. Gastroenterology. 2019; 157: 440-450.e8. [CrossRef]
  133. Chitnavis MV, Braly KL. The specific carbohydrate diet in inflammatory bowel disease: The evidence and execution. Pract Gastroenterol. 2019; 43: 28-36.
  134. Suskind DL, Wahbeh G, Gregory N, Vendettuoli H, Christie D. Nutritional therapy in pediatric Crohn disease: The specific carbohydrate diet. J Pediatr Gastroenterol Nutr. 2014; 58: 87-91. [CrossRef]
  135. Kakodkar S, Farooqui AJ, Mikolaitis SL, Mutlu EA. The specific carbohydrate diet for inflammatory bowel disease: A case series. J Acad Nutr Diet. 2015; 115: 1226-1232. [CrossRef]
  136. Burgis JC, Nguyen K, Park KT, Cox K. Response to strict and liberalized specific carbohydrate diet in pediatric Crohn’s disease. World J Gastroenterol. 2016; 22: 2111-2117. [CrossRef]
  137. Cohen SA, Gold BD, Oliva S, Lewis J, Stallworth A, Koch B, et al. Clinical and mucosal improvement with specific carbohydrate diet in pediatric Crohn disease. J Pediatr Gastroenterol Nutr. 2014; 59: 516-521. [CrossRef]
  138. Haas SV, Haas MP. The treatment of celiac disease with the specific carbohydrate diet; report on 191 additional cases. Am J Gastroenterol. 1955; 23: 344-360.
  139. Bischoff SC, Escher J, Hébuterne X, Kłęk S, Krznaric Z, Schneider S, et al. ESPEN practical guideline: Clinical nutrition in inflammatory bowel disease. Clin Nutr. 2020; 39: 632-653. [CrossRef]
  140. Simões CD, Maganinho M, Sousa AS. FODMAPs, inflammatory bowel disease and gut microbiota: Updated overview on the current evidence. Eur J Nutr. 2022; 61: 1187-1198. [CrossRef]
  141. Prince AC, Myers CE, Joyce T, Irving P, Lomer M, Whelan K. Fermentable carbohydrate restriction (low fodmap diet) in clinical practice improves functional gastrointestinal symptoms in patients with inflammatory bowel disease. Inflamm Bowel Dis. 2016; 22: 1129-1136. [CrossRef]
  142. Limketkai BN, Iheozor-Ejiofor Z, Gjuladin-Hellon T, Parian A, Matarese LE, Bracewell K, et al. Dietary interventions for induction and maintenance of remission in inflammatory bowel disease. Cochrane Database Syst Rev. 2019; 2: CD012839. [CrossRef]
  143. Cox SR, Lindsay JO, Fromentin S, Stagg AJ, McCarthy NE, Galleron N, et al. Effects of low FODMAP diet on symptoms, fecal microbiome, and markers of inflammation in patients with quiescent inflammatory bowel disease in a randomized trial. Gastroenterology. 2020; 158: 176-188.e7. [CrossRef]
  144. Lewis JD, Sandler RS, Brotherton C, Brensinger C, Li H, Kappelman MD, et al. A randomized trial comparing the specific carbohydrate diet to a Mediterranean diet in adults with Crohn’s disease. Gastroenterology. 2021; 161: 837-852.e9. [CrossRef]
  145. Illescas O, Rodríguez-Sosa M, Gariboldi M. Mediterranean diet to prevent the development of colon diseases: A meta-analysis of gut microbiota studies. Nutrients. 2021; 13: 2234. [CrossRef]
  146. Schwingshackl L, Hoffmann G. Adherence to Mediterranean diet and risk of cancer: A systematic review and meta-analysis of observational studies. Int J Cancer. 2014; 135: 1884-1897. [CrossRef]
  147. Lewis JD. Trial of specific carbohydrate and Mediterranean diets to induce remission of Crohn’s disease (DINE-CD) [Internet]. Bethesda: ClinicalTrials.gov; 2021; NCT03058679. Available from:https://clinicaltrials.gov/ct2/show/NCT03058679.
  148. Trichopoulou A, Lagiou P. Healthy traditional Mediterranean diet: An expression of culture, history, and lifestyle. Nutr Rev. 1997; 55: 383-389. [CrossRef]
  149. Marlow G, Ellett S, Ferguson IR, Zhu S, Karunasinghe N, Jesuthasan AC, et al. Transcriptomics to study the effect of a Mediterranean-inspired diet on inflammation in Crohn’s disease patients. Hum Genomics. 2013; 7: 24. [CrossRef]
  150. De Filippis F, Pellegrini N, Vannini L, Jeffery IB, La Storia A, Laghi L, et al. High-level adherence to a Mediterranean diet beneficially impacts the gut microbiota and associated metabolome. Gut. 2016; 65: 1812-1821. [CrossRef]
  151. Zhang Z, Taylor L, Shommu N, Ghosh S, Reimer R, Panaccione R, et al. A diversified dietary pattern is associated with a balanced gut microbial composition of faecalibacterium and escherichia/shigella in patients with Crohn’s disease in remission. J Crohns Colitis. 2020; 14: 1547-1557. [CrossRef]
Newsletter
Download PDF Download Citation
0 0

TOP