Abstract

Background and Aims:

Both science and patients associate diet with inflammatory bowel disease [IBD]. There is no doubt that links between IBD and diet are numerous, based on both epidemiological studies and experimental studies. However, scientific evidence to support dietary advice is currently lacking, and dietary counselling for IBD patients is often limited in clinical practice to the improvement of nutrient intake. This review aimed to focus on both patient’s beliefs about and molecular mechanisms for crosstalk between nutrients and inflammation.

Methods:

A literature search using PubMed was performed to identify relevant studies on diet and/or nutrients and their role in IBD. Pubmed [from inception to January 20, 2016] was searched using the terms: ‘Crohn’, ‘colitis’,’ intestinal epithelial cells’, and a list of terms relating to diet or numerous specific nutrients. Terms associated with nutrients were individually tested in the context of IBD. Reference lists from studies selected were manually searched to identify further relevant reports. Manuscripts about diet in the context of IBD from basic science, epidemiological studies, or clinical trials were selected and reviewed. Only articles published in English were included.

Results:

Epidemiological studies highlight the key role of diet in IBD development, and many IBD patients report diet as a triggering factor in relapse of disease. In addition, we present research on the impact of nutrients on innate immunity.

Conclusion:

Diet may offer an alternative approach to restoring deficient innate immunity in IBD, and this may be the scientific rationale for providing dietary counselling for IBD patients.

1. Introduction

Links between inflammatory bowel disease [IBD] and diet are numerous, ranging from clinical evidence of frequent multifactorial nutritional deficiencies in IBD patients to efficacy of nutrition therapy either as sole therapy or as adjunctive therapy.1 Moreover, IBD patients frequently report that specific foods cause aggravation of their symptoms, and recent reports have highlighted the importance of these dietary beliefs from the patient’s perspective.2,3 However, scientific evidence to support specific dietary advice is currently lacking, and dietary counseling [apart from perhaps in paediatrics] is almost limited in clinical practice to the improvement of nutrient intake in IBD patients, or the avoidance of foods that may precipitate obstructive symptoms in selected patients. Nevertheless, enteral nutrition is a dietary therapy for IBD. In active Crohn’s disease [CD] in children, exclusive enteral nutrition was as effective as corticosteroids in inducing remissions.4 In adult patients with CD, exclusive enteral nutrition can lead to mucosal healing and quality of life improvement.5

IBD is a complex chronic inflammatory condition, and compelling studies have confirmed the concept that IBD results from an unbalanced relationship within the triad: environment, genes, and innate immunity.6 Nutrients seem to be involved in these interactions, and among environmental factors, a Western diet is considered to be a possible trigger for chronic intestinal inflammation, by altering the microbiome and by other mechanisms. Epidemiological observations suggest an association between IBD incidence and dietary patterns.7 In an experimental IBD model, exposure to a Western diet induced dysbiosis accompanied by an intestinal barrier dysfunction in mice.8 The pathways implicated in the nutrient-driven proinflammatory effects are numerous: nutrients may modulate innate immunity and consequently promote inflammation and influence the microbiota.9

This review will focus on interactions between nutrients and innate immunity, and attempt to relate patient’s beliefs to molecular mechanisms.

1.1. Dietary beliefs of IBD patients

IBD patients frequently report that specific food items are able to influence their disease by worsening or alleviating their symptoms.10 During the last 2 years, studies have investigated associations between diet and IBD patient symptoms.2, 3, 10–12 Hou et al. focused on patient-designed diets.11 Indeed, patients have access to numerous non-medical resources for dietary advice, such as IBD support groups or unverified sources on the internet. Hou et al. observed that patient-devised diets are mainly based on food restrictions, and advice from patients’ various sources is highly conflicting.11 Similarly, a French team investigated dietary beliefs and behavior in adult IBD patients and reported that the majority of IBD patients [66%] are avoiding certain foods.3 Patients base their choices mainly on personal experience of symptoms worsening and on their beliefs about a relapse prevention effect by specific food elimination.3 In a similar study performed in the UK, 60% of patients felt they expressed a desire for more dietary advice.2

Consequently, IBD patients are frequently on restricted diets without a scientific rationale.13 The most documented example is avoidance of dairy products. Contrary to the belief frequently held by IBD patients, Jowett et al. did not find any association between relapse and increased intake of milk and dairy products.14 Also, such dietary practice is not taking into account the evidence of links between low dairy products intake and increased nutritional deficiencies. An Italian study using a food questionnaire reported that the diet of IBD patients contained less calcium than that of healthy controls.15 This might potentially increase the risk of osteoporosis through inadequate dietary calcium intake. In addition, a survey from New Zealand reported that the influence of dairy products on CD symptoms was more influenced by fat content than lactose content.16 Furthermore, reduced dairy product intake is associated with low vitamin D intake. Vitamin D impact on innate immunity will be discussed later in this review—compelling evidence suggests it has a role in IBD physiopathology. Indeed, vitamin D deficiency correlates with disease activity.17 In an IBD model, vitamin D protects from colitis through microbiome modifications.18 More recently, a low level of the vitamin D receptor [VDR] was associated with innate immunity dysfunction: abnormal Paneth cells, impaired autophagy function, and dysbiosis.19

Another very popular exclusion diet, especially in the USA [0.5% of the US population], is a gluten-free diet [GFD] initiated without a diagnosis of coeliac disease. Herfarth et al. have performed a cross-sectional study on 1647 IBD patients from the Crohn’s & Colitis Foundation of America [CCFA] partners cohort, demonstrating the high prevalence of a GFD [19.1%].12 Among patients who attempted a GFD, two out of three described an improvement in their gastrointestinal symptoms, and 38% reported fewer or less-severe IBD flares.12 These observations are yet to be confirmed by appropriate controlled intervention trials. Inflammatory bowel disease patients need dietary advice based on strong scientific rationale to avoid them using unnecessarily restrictive diets that may impair their nutritional status. Richman et al. have recently reviewed the literature in order to provide dietary guidance based on ‘best available evidence’ and have pointed out an urgent need for further evidence.20

1.2. Epidemiological evidence of the role of diet in IBD

It has long been suspected that environment, in particular dietary habits, contributes to the development of IBD [Table 1, Figure 1]. Migration studies have demonstrated the role of dietary patterns, particularly Westernization of diet, in IBD development.21 The Western dietary pattern [high fat, high n-6 polyunsaturated fatty acids [PUFAs], high meat, low fruits and vegetables] is associated with an increased IBD risk7 and also with exacerbated colitis [red meat,22 dietary fats23]. In addition, an increased incidence of IBD has been associated with diets high in animal protein.24 In Japan, dietary pattern [fat/protein] was found to be associated with CD risk,7 while increased consumption of animal protein has been associated with higher IBD risk in France.25 The Western diet is also characterized by an unbalanced ratio of both types of PUFA [n-3:n-6 ratio]. Linoleic acid [LA, n-6 PUFA] consumption has markedly increased [3-fold throughout the 20th century].26 Recent epidemiological studies have demonstrated the role of dietary intake of mono-unsaturated fatty acids [MUFAs] and PUFAs in ulcerative colitis [UC] development: Higher intake of LA, an n-6 PUFA,27 or higher intake of trans-unsaturated fat28 is associated with an increased risk of UC, whereas oleic acid [n-9 MUFA]29 or n-3 PUFA consumption28, 30 is beneficial. The Western diet is associated with a low intake of fruit and vegetables, but a long-term high intake of dietary fibre reduces CD risk.31 Western dietary components can affect mucosal immunology by affecting numerous mechanisms (such as gut barrier function, pattern recognition receptors [PRRs] or mucin expression). For example, we previously demonstrated that n-3 PUFAs can downregulate intestinal inflammation through peroxisome proliferator-activated receptor gamma [PPARγ].32 Dimerization of Toll-like receptor-4 [TLR-4], a PRR, can be activated or inhibited according to the fatty acid type.33

Table 1.

Epidemiological evidence of the role of diet in IBD.

BeneficialDeleteriousHypothesized mechanism/action
LipidsOleic acid29 Anti-inflammatory properties129
Inhibition of adhesion molecules
Competition with n-6 PUFA
DHA28, 130 Linoleic acid27
Arachidonic acid29
Unbalanced n-3/n-6 ratio:
n-6 PUFA:
Pro-inflammatory effects of arachidonic acid–derived eicosanoids
n-3 PUFA: 
- Competition with n-6 PUFA 
- PPARγ activation 
- TLR-4 inhibition
Vitamin D100, 101 Anti-inflammatory effect of VDR signalling
ProteinsAnimal protein25 Main source of n-6 PUFA
Dietary FibreDietary fibre31 Anti-inflammatory effect of short-chain fatty acid
BeneficialDeleteriousHypothesized mechanism/action
LipidsOleic acid29 Anti-inflammatory properties129
Inhibition of adhesion molecules
Competition with n-6 PUFA
DHA28, 130 Linoleic acid27
Arachidonic acid29
Unbalanced n-3/n-6 ratio:
n-6 PUFA:
Pro-inflammatory effects of arachidonic acid–derived eicosanoids
n-3 PUFA: 
- Competition with n-6 PUFA 
- PPARγ activation 
- TLR-4 inhibition
Vitamin D100, 101 Anti-inflammatory effect of VDR signalling
ProteinsAnimal protein25 Main source of n-6 PUFA
Dietary FibreDietary fibre31 Anti-inflammatory effect of short-chain fatty acid
Table 1.

Epidemiological evidence of the role of diet in IBD.

BeneficialDeleteriousHypothesized mechanism/action
LipidsOleic acid29 Anti-inflammatory properties129
Inhibition of adhesion molecules
Competition with n-6 PUFA
DHA28, 130 Linoleic acid27
Arachidonic acid29
Unbalanced n-3/n-6 ratio:
n-6 PUFA:
Pro-inflammatory effects of arachidonic acid–derived eicosanoids
n-3 PUFA: 
- Competition with n-6 PUFA 
- PPARγ activation 
- TLR-4 inhibition
Vitamin D100, 101 Anti-inflammatory effect of VDR signalling
ProteinsAnimal protein25 Main source of n-6 PUFA
Dietary FibreDietary fibre31 Anti-inflammatory effect of short-chain fatty acid
BeneficialDeleteriousHypothesized mechanism/action
LipidsOleic acid29 Anti-inflammatory properties129
Inhibition of adhesion molecules
Competition with n-6 PUFA
DHA28, 130 Linoleic acid27
Arachidonic acid29
Unbalanced n-3/n-6 ratio:
n-6 PUFA:
Pro-inflammatory effects of arachidonic acid–derived eicosanoids
n-3 PUFA: 
- Competition with n-6 PUFA 
- PPARγ activation 
- TLR-4 inhibition
Vitamin D100, 101 Anti-inflammatory effect of VDR signalling
ProteinsAnimal protein25 Main source of n-6 PUFA
Dietary FibreDietary fibre31 Anti-inflammatory effect of short-chain fatty acid
Dietary pattern modulates IBD risk.
Figure 1.

Dietary pattern modulates IBD risk.

Results from epidemiological studies have shown that dietary patterns are associated with inflammatory bowel disease [IBD] risk. Features of a healthy diet included high fibre and prudent unsaturated fatty acid consumption decreased its risk, while the hallmarks of a Western diet [red meat, saturated fatty acids, n-6 PUFA] increased its risk.

While we mainly discuss changes directly affecting host immunology, diet can also shape the microbiota, leading to a host immunity response. Comparison of the microbiota from rural Burkina Faso children with that of urban European children from Italy demonstrated how diet can profoundly modify the microbiota composition.9

1.3. Innate immunity sensors: a target for nutrients?

A family of intracellular danger-sensing molecules called NLRs [NOD-like receptors or nucleotide-binding domain leucine–rich repeat-containing genes] has been implicated in the pathogenesis of IBD. Within this family are the NOD [nucleotide-binding oligomerization domain] and NLRP [pyrin domain containing NLR] subfamilies [Figure 2]. The link between NOD2 mutation and CD has been well established, and it is the most commonly found gene variant associated with CD.34 In addition, alteration of another family of bacterial sensors, the Toll-like receptors [TLRs, Figure 2], is well described in IBD studies. IBD patients also exhibit an abnormal composition and activity of gut microbiota. Interactions between intestinal microbes and food may influence IBD pathogenesis. Compelling studies demonstrate the role of diet in shaping gut microbiota.35 Defective innate immunity could contribute to IBD pathogenesis by enabling a tolerance breakdown to intestinal microbiota in genetically predisposed individuals.

Dietary targets of intestinal innate immunity.
Figure 2.

Dietary targets of intestinal innate immunity.

Pathogen-associated molecular patterns [PAMPs] are microbial motifs that enable the innate immune system to recognize groups of pathogens within the luminal environment. The host recognizes PAMPs with the help of pattern recognition receptors, such as the toll-like receptors [TLRs] and the intracellular nucleotide-binding oligomerization domain [NOD] receptors. Upon ligand recognition, TLRs undergo conformational changes that allow recruitment of adaptor molecules such as CD14 or MD2. Most TLRs then recruit the MyD88 adaptor molecule, leading to free NF-kB transcription factor being sequestered in the cytosol, causing activation of inflammatory cytokines such as TNF-α or IL-6. NOD2 recognizes the peptidoglycans [PGNs], the breakdown products of intracellular bacteria. Upon stimulation, NOD2 can activate the MAPK pathway or the release of the NF-kB, both leading to the upregulation of proinflammatory cytokines. Inflammasomes play a critical role in orchestrating the innate immune response in response to pathogens and cellular stress such as reactive oxygen species [ROS]. The inflammasome is a multimolecular complex, and one of its main functions is to activate caspase-1 and cleave pro-forms of IL-1β and IL-18 into their active secreted forms. Inflammasome formation is a two-step process: [i] TLR-induced NF-kB transcription of pro-IL-1β, pro-IL-18 and inflammasome components such as NLRP3, and [ii] inflammasome oligomerization, leading to caspase-1 activation and a subsequent cytokine cleavage. These molecular compounds can be targeted by nutrients. Phytochemicals can downregulate inflammasome, NOD2 and TLR pathways. Fish oil or DHA can regulate TLR and NOD2 signalling. By contact, TLR signalling can be activated by some FODMAPS [inulin, levan] or saturated fatty acids. These mechanisms could contribute to explaining how food can interact with intestinal inflammation.

These findings demonstrate innate immunity control of the adaptive immune response and the crucial role of the gene–environment interplay in IBD induction. Until now, this emerging theory has not taken into account the role of diet in IBD development. There is considerable interest in modulating IBD development through dietary therapy in an attempt to rectify innate immunity defects resulting from genetic mutations. Given this, we have investigated how nutrients may affect innate immunity and whether diet modification offers an alternative therapeutic approach.

The mucosal surface of the intestine plays a key role in control of the host immune response by acting as a barrier against bacterial and dietary antigens. In addition, the gut barrier is a first defence mechanism strongly influenced by nutrients. Nevertheless, interactions between nutrients and the gut barrier could represent a review in themselves. We chose to focus our review on the innate immune cell components.

1.4. Dietary habits and innate immunity in chronic diseases

Obesity and diabetes studies have demonstrated that dietary compounds are able to modulate innate immunity components. For example, a high-fat, high-carbohydrate [HFHC] meal induced TLR expression and increased plasma endotoxin concentrations in monocytes from normal-weight subjects.36 Interestingly, orange juice intake with the HFHC meal prevented meal-induced inflammatory stress, including the increase in endotoxin and TLR expression.36 The authors of this study have also investigated the effect of saturated fat [as cream intake] in healthy volunteers, and they found that cream was able to induce TLR-4 expression and lipopolysaccharide [LPS] concentration in mononuclear cells.37 Similarly, splenocytes from mice with high-fat–induced obesity exhibited a 60% higher expression of NOD2 compared with the control group.38 Exacerbation of colitis has been reported in IL-10-/- mice receiving a high saturated fat diet.23 The authors of that study demonstrated that the high saturated fat diet modulated the composition of the bile acids, leading to a modified composition of intestinal microbiota and a subsequent perturbation of gut homeostasis.23 Similarly, a Western diet increased the NOD-2 and TLR-5 mRNA levels in an experimental CD model.8 Thus, consumption of a high-fat diet was associated with changes in the gut microbiota and with upregulated PRR expression in murine models. While the influence of a Western diet on gut microbiota has already been established, more research is required in order to identify the underlying mechanisms by which nutrients can modify intestinal microbiota. Nevertheless, a metabolomics study recently reported that intestinal microbiota-derived metabolites can be correlated with CD—that group found a strong correlation between the most abundant bacteria in ileal CD and fatty acids.39

A Western diet is also associated with added ingredients, such as emulsifiers or added sugars. Maltodextrin is a common dietary polysaccharide used as an emulsifier in the Western diet. McDonald et al. have demonstrated its deleterious role in defence mechanisms. Indeed, E. coli adhesion to intestinal epithelial cells (IECs) was improved in the presence of maltodextrins in vitro.40 They also observed that consumption of maltodextrins in mice leads to better Salmonella survival and higher mucosal colonization.41 In a recent study from Chassaing et al., mice received the emulsifiers at a low concentration via drinking water for 12 weeks. The authors found that consumption of emulsifiers leads to a low-grade inflammation and obesity/metabolic syndrome in wild-type mice, while it promoted colitis in IL-10-/- mice.42

Red meat is also a hallmark of the Western dietary pattern. Its consumption is associated with an increased IBD risk,25 while a diet rich in red meat exacerbates dextran sulfate sodium [DSS]-induced colitis.22 In addition, Erridge (from the UK), who has studied the ability of numerous foodstuffs to induce innate immunity, reported that numerous unspoiled foods can contain TLR-2 or -4 stimulants.43, 44

In conclusion, a Western diet may exacerbate intestinal inflammation through changes in the gut microbiota leading to an upregulation of the TLR- and NOD-pathways. In addition, obese patients have a more severe disease course.45 These new and exciting observations directly link diet and obesity to activation of the innate immune system, inflammation, and end organ dysfunction.

2. Effect of specific nutrients on innate immunity compounds [Table 2]

2.1. Fatty acids

Butyrate and propionate are short-chain fatty acids that are produced through the fermentation of dietary fibre by intestinal microbiota. In peptidoglycan [PGN]-treated intestinal epithelial cell line Caco-2, increasing concentration of butyrate enhanced production of the chemokines IL-8 and GRO-α through NF-κB activation.46 The authors of that study demonstrated that butyrate enhanced NOD-2 expression by activating histone acetylation in the Nod2 promoter region, thus promoting chemokine secretion.46 Interestingly, they have shown that silencing NOD2 and TLR2 significantly inhibited PGN-mediated chemokine production, suggesting that both NOD2 and TLR2 are required for maximal response.46 In the bovine mammary gland, short-chain fatty acids such as propionate and hexanoate have demonstrated antimicrobial properties by inhibiting internalization of Staphylococcus aureus and by producing antimicrobial peptides.47 In the intestine, the effect of digested and fermented green kiwifruit has been evaluated in intestinal epithelial cell line HT-29, and those authors found that only fermented compounds were able to increase human β-defensin 1 and 2 production48; they speculated that this effect was probably mediated by the short-chain fatty acid content of the fermenta, because short-chain fatty acids were able to increase hBD-1 and hBD2 production.48 More recently, butyrate treatment was able to restore VDR expression and inhibit inflammation in a colitis model.49

Table 2.

Dietary targets of innate immunity.

TargetsThe goodThe evil
DefensinAmino acids [arginine, isoleucine]
Fermented green kiwi fruit
Probiotics
InflammasomePhytochemicals [EGCG*, quercetin*, rutin*]
NOD2Fish oil
Phytochemicals [curcumin]
Butyrate
TLRPUFA [DHA, fish oil]
Amino acids [alanyl-glutamine, arginine]
Phytochemicals [curcumin, 1D10DG, garlic extract, cinnamaldehyde, resveratrol, EGCG]
FODMAP [oligolactan]
Probiotics
Saturated fatty acids
FODMAPs [inulin, levan, β1–4 mannobiose]
TargetsThe goodThe evil
DefensinAmino acids [arginine, isoleucine]
Fermented green kiwi fruit
Probiotics
InflammasomePhytochemicals [EGCG*, quercetin*, rutin*]
NOD2Fish oil
Phytochemicals [curcumin]
Butyrate
TLRPUFA [DHA, fish oil]
Amino acids [alanyl-glutamine, arginine]
Phytochemicals [curcumin, 1D10DG, garlic extract, cinnamaldehyde, resveratrol, EGCG]
FODMAP [oligolactan]
Probiotics
Saturated fatty acids
FODMAPs [inulin, levan, β1–4 mannobiose]

*Effects of these nutrients have not been documented in the intestine.

Table 2.

Dietary targets of innate immunity.

TargetsThe goodThe evil
DefensinAmino acids [arginine, isoleucine]
Fermented green kiwi fruit
Probiotics
InflammasomePhytochemicals [EGCG*, quercetin*, rutin*]
NOD2Fish oil
Phytochemicals [curcumin]
Butyrate
TLRPUFA [DHA, fish oil]
Amino acids [alanyl-glutamine, arginine]
Phytochemicals [curcumin, 1D10DG, garlic extract, cinnamaldehyde, resveratrol, EGCG]
FODMAP [oligolactan]
Probiotics
Saturated fatty acids
FODMAPs [inulin, levan, β1–4 mannobiose]
TargetsThe goodThe evil
DefensinAmino acids [arginine, isoleucine]
Fermented green kiwi fruit
Probiotics
InflammasomePhytochemicals [EGCG*, quercetin*, rutin*]
NOD2Fish oil
Phytochemicals [curcumin]
Butyrate
TLRPUFA [DHA, fish oil]
Amino acids [alanyl-glutamine, arginine]
Phytochemicals [curcumin, 1D10DG, garlic extract, cinnamaldehyde, resveratrol, EGCG]
FODMAP [oligolactan]
Probiotics
Saturated fatty acids
FODMAPs [inulin, levan, β1–4 mannobiose]

*Effects of these nutrients have not been documented in the intestine.

3. Short-chain fatty acids are key bacterial products and can target innate immunity through antimicrobial peptide production

Unsaturated fatty acids are essential components of the intestinal inflammatory response. Indeed, they modulate cell membrane composition with a subsequent modification of the activation of receptors. They can also act as signalling molecules, and numerous nuclear receptors are molecular targets of fatty acids: the outcome is altered gene expression.

TLR-4 is a cell-surface receptor and is activated by ligand-induced dimerization. Opposing effects of fatty acids on TLR-4 activation have been demonstrated in vitro: TLR-4 dimerization is facilitated by saturated fatty acids, while unsaturated fatty acids (like docosahexaenoic acid [DHA]) inhibit its dimerization.33 Inhibition of TLR-4 expression by DHA has been confirmed in IL-1–treated human intestinal microvascular endothelial cells.50

Similarly, fish oil supplementation for 21 days led to an enrichment of intestinal mucosa into eicosapentaenoic acid [EPA], DHA, and total [n-3] PUFA [an improved intestinal integrity], but also downregulated mRNA levels of TLR-4 and NOD2 signalling pathways in LPS-treated weaned pigs.51 In contrast, both n-3 PUFA and n-6 PUFA treatment at 15 µM resulted in an increase in the internalization of the microorganisms by the murine macrophage cell line RAW.52

4. In IBD models, n-3 PUFA can regulate intestinal innate immunity through PRR inhibition

4.1. Amino acids

Glutamine and arginine are non-essential amino acids, but they are considered essential in stress situations, and both demonstrate anti-inflammatory properties in IBD models.53–63 Glutamine is the preferential substrate for enterocytes, and its effect has been evaluated in numerous experimental models of gut barrier dysfunction. In a neonatal deprivation model, TLR-4 expression was upregulated in separated rats, and from our laboratory we reported the specific effect of alanyl-glutamine supplementation in downregulating its colonic expression.62 Similarly, glutamine supplementation decreased TLR-4 mRNA levels in the jejunum and the colon of LPS-treated rats.64 Intraperitoneal injection of alanyl-glutamine decreased colon TLR-4 in a DSS colitis model.65 No potential mechanisms have yet been demonstrated to explain the effect of glutamine on TLR-4. As TLR-4 activation66 involves reactive oxygen species, we hypothesized that the antioxidant properties of glutamine may be responsible for TLR-4 inhibition.

Arginine is a nitric oxide precursor. Supplementation of diets with 0.5–1% L-arginine increased serum levels of arginine and inhibited the TLR-4–Myd88 signalling pathway in Salmonella-infected weaned piglets.67 It has been demonstrated that nitric oxide can downregulate TLR-4 gene expression,68 and bacterial infection is also associated with inducible nitric oxide synthase [iNOS] overexpression, with subsequent nitric oxide formation as a defence mechanism. Thus, arginine supplementation may decrease TLR-4 expression through the nitric oxide pathway. Treatment with arginine or isoleucine also upregulated secretion of human beta-defensin-1 [hBD1] in the HCT-118 intestinal cell line.69 Surprisingly, the authors tested a range of arginine concentrations from 25 to 500 µg/mL, and only the particular concentration of 250 µg/mL of arginine was able to induce hBD1.69 Dietary nutrients such as tryptophan are also involved in host–microbiome interactions. Tryptophan is an amino acid and its metabolites regulate intestinal homeostasis. The authors of an elegant study70 have investigated the potential action of tryptophan metabolites on resident microbiota, and they describe a novel metabolic pathway whereby Lactobacilli are able to switch their fuel from sugar to tryptophan to produce indoles. Upon aryl hydrocarbon receptor activation by indoles, IL-22 was produced, conferring resistance to Candida albicans in mice. The authors demonstrated that dietary tryptophan affected host–fungal symbiosis via the microbiota.70

Fermented milks are a source of bioactive peptides with a beneficial effect on the intestinal mucosa. For example, beta-casomorphin-7, a peptide produced from bovine β-casein, strongly stimulates mucin production in human intestinal mucus-producing cell line HT-29MTX.71 A French team has evaluated the effect of the total peptide pool from fermented milk in the same cell line, and they found that mucin gene expression [MUC2–4] and production increased.72 They then identified the peptide responsible for the biological activity by high-performance liquid chromatography, and the identified peptide was then administrated by gavage in rat pups.72 They reported an enhanced number of goblet and Paneth cells along the small intestine, associated with a higher expression of intestinal mucins and antibacterial factors such as lysozyme.72

Therefore, amino acids can target innate immunity by numerous mechanisms: [i] PRR inhibition [glutamine, arginine], [ii] antimicrobial production [arginine, isoleucine], and [iii] by acting on microbiota. Further studies in IBD are required.

4.1. Phytochemicals

Curcumin derived from Curcuma longa is the yellow pigment of curry. It exerted anti-inflammatory properties in experimental models of IBD73–76 and also in IBD clinical trials.77, 78 In rats with 2,4,6-trinitrobenzenesulfonic acid [TNBS]-induced colitis, administration of curcumin at the dose of 100mg/kg inhibited inflammatory mediators [such as myeloperoxidase] and also downregulated colon TLR-4 and Myd88 expression.79 Phytochemicals can also inhibit NOD2 activation by interfering with NOD2 dimerization.80 This has been reported for curcumin in the colonic epithelial cell line HCT-116.81In vitro, it has been demonstrated that curcumin is able to counteract the effects of SLC22A4 and IL-10 variants associated with IBD.82 A novel mechanism behind the anti-inflammatory effect of curcumin has been described in the lung83 and involves the cell surface receptor Triggering Receptor Expressed on Myeloid cells 1 [TREM-1]. TREM is expressed on macrophages and dendritic cells [DCs] and is believed to be an amplifier of the TLR-induced inflammatory response. Curcumin at 10 µM inhibited TREM-1 expression and mRNA levels in primary mice macrophages and prevented the binding of NF-κB to TREM-1 promoter.83 The authors confirmed the downregulation of TREM-1 by curcumin at 200mg/kg in the lung from septic mice.83 Very recently, Lang et al. have reported in an international multicenter prospective double-blind study that 53.8% of patients receiving 0.3g per day of a 95% pure curcumin compound added to 4 g of maintenance mesalamine achieved a clinical remission in patients with mild-to-moderate ulcerative colitis compared with none in the control group at the end of a 1-month study.78

It has also been demonstrated that a derivative of ginger, 1-dehydro-10-gingerdione, acts on TLR-4 expression in macrophages.84 1-dehydro-10-gingerdione at from 10 to 100 µM inhibited LPS binding to myeloid differentiation 2 [MD2], a TLR-4 co-receptor, and also downregulated both TLR-4–mediated expression of NF-κB and activating protein-1 in LPS-treated murine macrophage cell line RAW264.7.84

Phytochemicals can interfere with TLR4 dimerization [Figure 2]. For example, garlic extract downregulated NF-κB activity, with a subsequent decreased expression of iNOS and COX-2 by inhibiting TLR-4 dimerization in LPS-treated RAW264.7 cells.85 The same team described a similar mechanism with cinnamaldehyde, a cinnamon extract.86 Similarly, resveratrol, a polyphenol from grapes, also inhibited the TLR-4 signalling pathway in the same cell line RAW264.7 in response to LPS.87

Epigallocatechin-3-gallate [EGCG] is a green tea polyphenol and it has been demonstrated to exert anti-inflammatory effects in IBD models.80, 88 A cell surface receptor called 67kDa Laminin Receptor [67LR] is considered to be a polyphenol-sensing molecule, and it has been recently identified as a regulator of proliferation and adhesion in normal intestinal epithelial cells.89 Epigallocatechin-3-gallate is able to downregulate the inflammatory response in immune cells,90, 91 and it inhibited the TLR-signalling pathway through 67LR in LPS-treated DCs.91 Similar effects on the TLR-2 signalling pathway have been found in PGN-treated macrophages.90

Effects of flavonoids on inflammasome have been described for other organs, but their effects on intestinal inflammation have not yet been documented. Inhibition of NLRP1 inflammasome by EGCG at 0.1 to 10 µM has been reported in human metastatic melanoma cells.92 The effects of quercetin and its derivative rutin have been evaluated in rats with fructose-induced renal injury, and both blocked NLRP3 inflammasome activation.93 Very recently, it has been reported that resveratrol inhibited NLRP1 and NLRP3 inflammasome in LPS-treated human macrophages.94

Phytochemicals are able to downregulate Janus-kinase pathways. In mesenchymal stromal cells, treatment with EGCG at 30 µM downregulated STAT3 phosphorylation.95 Malvidin, a red wine polyphenol, inhibited NF-κB activation through the JNK MAPK pathway in LPS-stimulated RAW 264.7 macrophages.96 Resveratrol, a polyphenolic compound found in red wine, downregulated nitric oxide production and chemokine secretion by blocking JAK/STAT-1 IFNγ-treated macrophages.97 Treatment with curcumin at 50mg/kg/D ameliorated the inflammatory score in mice with DSS-induced colitis by targeting the STAT3 pathway.98

5. Phytochemicals can target innate immunity, and their main documented action is their inhibitory effect on TLR-4

5.1. Vitamins

Vitamin D influences innate immunity,99 and Ananthakrishnan et al. have recently reported that higher predicted plasma levels of 25[OH]D significantly reduce the risk of incident CD and non-significantly reduce the risk of UC in women.100 Similarly, vitamin D metabolism is dependent on sun exposure, and a French team has recently found an association between high residential sun exposure with a decreased risk of incident CD in a prospective French cohort.101 In human peripheral blood mononuclear cells exposed to M. tuberculosis, antimicrobial peptide cathelicidin was upregulated 16-fold by 25[OH]D at 100nM, while TLR-2 and TLR-4 expression were downregulated.102 A recent study highlighted the link between its receptor signalling and a key component of innate immunity: intestinal autophagy.49 They found that a low intestinal epithelial VDR alters intestinal autophagy [reduces ATG16L1 expression and impairs antimicrobial function of Paneth cells].49 The authors were able to restore VDR signalling by a dietary approach. Indeed, butyrate administration can restore intestinal VDR expression, leading to colitis inhibition.49

6. Vitamin D deficiencies are very common in IBD patients and may partly contribute to an altered innate immunity response through reduced antimicrobial peptide production by Paneth dysfunction or PRR upregulation

Retinoic acid is a vitamin A derivative that plays a crucial role in maintaining immune homestasis, and it has recently been highlighted that retinoic acid also regulates the homeostasis of pre-DC–derived DC subsets.103

6.1. Probiotics

The main single dietary approach to influencing IBD development is the use of probiotics. A recent case–control study demonstrated that more IBD patients than controls use probiotics to manage their health. There is considerable variability among probiotic strains, and not all probiotics will be beneficial for IBD management. Compelling studies have demonstrated the inhibitory effect of probiotics on TLR signalling pathways in vitro in intestinal epithelial cells and DCs.104–106 While a strain of Lactobacillus exerts beneficial effects in experimental necrotizing enterocolitis through modulation of TLR-4 and NF-κB,107 a different strain acts through TLR2 in a radiation injury model.108 In addition, production of defensins was also upregulated in vitro in response to probiotic treatment.105 Another strain of probiotic, Bifidobacterium breve, was able to reduce susceptibility to colitis in Nod1-/- and Nod2-/- mice.109 While probiotic studies have been promising in experimental models of IBD, clinical trials have failed to prove their efficacy for postoperative prophylaxis.110, 111 Nevertheless, the last systematic review from the Cochrane database studies suggests that VSL#3 appears more effective than placebo for maintenance of remission in chronic pouchitis and prevention of pouchitis.112 The dietary targets of innate immunity are illustrated in Figure 2.

6.2. Starch

Dietary starch is able to modulate intestinal inflammation in numerous IBD models. Resistant starch–fed IL-10-/- mice exhibited a lower cytokine secretion [IL-6, CXCL1] than control diet–fed mice.113 Interestingly, resistant starch can counterbalance the deleterious effect of red meat consumption in mice with DSS-induced colitis.22 Dietary starch also protects against colitis-associated colorectal cancer in rats by reducing tumor multiplicity and adenocarcinoma formation through intestinal microbiota changes.114

6.3. FODMAPs

FODMAPs are food items that are rich in fermentable oligo-, di- and monosacharides, and polyols. Low FODMAP diet has been proposed to limit functional symptom in irritable bowel syndrome patients.115 FODMAPs are also able to target innate immunity compounds [Figure 2].

Inulin is a polysaccharide, and treatment with inulin at 1mg/mL increased phagocytosis in phorbol myristate acetate [PMA]-stimulated macrophages; this effect was inhibited by using a blocking antibody specific to TLR4.116 Similarly, levan treatment at 10 µg/mL induced production of IL-12 p40 and TNF-α by macrophage cell lines in vitro through the TLR4 pathway.117 Treatment with beta-1,4-mannobiose induced TNF-α and IL-6 secretion in RAW 264.7 macrophages, and this effect was abrogated by inhibiting TLR4.118 Nevertheless, the same study reported that beta-1,4-mannobiose was able to reduce the inflammatory process in endotoxemic mice.118 In contrast, an apple oligolactan was able to downregulate TLR4 levels in mice with colitis-associated colon cancer and in LPS-treated intestinal cell line HT-29.119 Honey is able to stimulate cytokine production in macrophages and human monocytes.120, 121 The TLR4 signalling pathway mediated the pro-inflammatory effect of honey. Both studies considered that there may be factors other than TLR4 stimulants [such as LPS] that may explain this effect.120, 121

Interestingly, all the FODMAPs cited above are also considered as prebiotics, meaning that they should confer benefits upon host well-being and health. In a randomized, double-blind, placebo-controlled trial, active CD patients received 15 g of fructo-oligosaccharides for 4 weeks, but there was no significant benefit in clinical response.122 In UC patients, 12 g/day of oligofructose-enriched inulin for 2 weeks was associated with a reduced fecal calprotectin.123

7. Combination treatment studies

Most of the studies presented above use a single nutrient, but a combination of individual nutrients may be better. For example, we have previously shown that only the combination of both amino acids [glutamine and arginine] was able to decrease TNF-α production in CD intestinal biopsies; use of a single amino acid was not effective.59 Similarly, the combination of EPA and resveratrol had a stronger anti-inflammatory effect than either of these molecules separately in LPS-treated macrophages.124 We also reported that rats with TNBS-induced colitis had a lower inflammatory score when they received a combination of fish oil with conventional treatment 5-aminosalicylic acid [5-ASA] than when they received a higher dose of 5-ASA alone.125

8. The interest of a multidisciplinary team

Inflammatory bowel disease patients often question their gastroenterologists about diet, and they believe that diet can influence their disease course.2, 3 While studies have highlighted that IBD patients have a strong interest in diet, their nutritional knowledge is mainly limited to unofficial sources such as lay websites.11, 13, 126 A multidisciplinary team including gastroenterologists, dietitians and nutritionists is mandatory for providing relevant dietary advice and adequate expert clinical dietitian support for IBD patients. A review about the existing dietary guidelines is available,127 but individualized dietetic intervention should be encouraged, because it may enable patient-tailored dietary advice based on current therapy and the natural history of the disease. Indeed, dietary intake is dependent on gender, age and disease course,126 and a one-size-fits-all approach is unwise, especially in the face of a paucity of data. Individualized dietetic advice may also limit the use of inappropriate patient-devised exclusion diets13 and the subsequent accentuating of nutrient deficiencies or even potential eating disorders [Table 3]. Instead, it will contribute to improving their quality of life. Some dietary restriction, such as low lactose, may be suitable for patients suffering from active inflammation.

Table 3.

Popular exclusion diets in IBD patients, their potential benefits and scientific evidence regarding these diets.

Exclusion dietBenefitScientific evidence
Specific carbohydrate dietClinical and mucosal improvements in paediatric CDSmall monocentric prospective pilot study: 9 to 7 patients for 12, 52 weeks of specific carbohydrate diet131
Low-FODMAP dietDecreased functional gastrointestinal symptomsPilot study: 52 CD and 20 UC patients [p < 0.02 for all]132
69/88 IBD patients [p < 0.001]133
Gluten-free diet [GFD]Improved self-reported gastrointestinal symptoms [65.6%]
Fewer or less severe IBD flares [38.3%]
Crohn’s & Colitis Foundation of America internet-based cohort: 449/1647 participants had previously tried a GFD or reported current use of GFD12
Paleo dietA literature search using PubMed from inception to April 2016 was performed to identify relevant studies using the terms: ‘Crohn’ OR ‘ulcerative colitis’,’ OR ‘inflammatory bowel diseases’ and ‘paleo’, and no records were found.
Exclusion dietBenefitScientific evidence
Specific carbohydrate dietClinical and mucosal improvements in paediatric CDSmall monocentric prospective pilot study: 9 to 7 patients for 12, 52 weeks of specific carbohydrate diet131
Low-FODMAP dietDecreased functional gastrointestinal symptomsPilot study: 52 CD and 20 UC patients [p < 0.02 for all]132
69/88 IBD patients [p < 0.001]133
Gluten-free diet [GFD]Improved self-reported gastrointestinal symptoms [65.6%]
Fewer or less severe IBD flares [38.3%]
Crohn’s & Colitis Foundation of America internet-based cohort: 449/1647 participants had previously tried a GFD or reported current use of GFD12
Paleo dietA literature search using PubMed from inception to April 2016 was performed to identify relevant studies using the terms: ‘Crohn’ OR ‘ulcerative colitis’,’ OR ‘inflammatory bowel diseases’ and ‘paleo’, and no records were found.
Table 3.

Popular exclusion diets in IBD patients, their potential benefits and scientific evidence regarding these diets.

Exclusion dietBenefitScientific evidence
Specific carbohydrate dietClinical and mucosal improvements in paediatric CDSmall monocentric prospective pilot study: 9 to 7 patients for 12, 52 weeks of specific carbohydrate diet131
Low-FODMAP dietDecreased functional gastrointestinal symptomsPilot study: 52 CD and 20 UC patients [p < 0.02 for all]132
69/88 IBD patients [p < 0.001]133
Gluten-free diet [GFD]Improved self-reported gastrointestinal symptoms [65.6%]
Fewer or less severe IBD flares [38.3%]
Crohn’s & Colitis Foundation of America internet-based cohort: 449/1647 participants had previously tried a GFD or reported current use of GFD12
Paleo dietA literature search using PubMed from inception to April 2016 was performed to identify relevant studies using the terms: ‘Crohn’ OR ‘ulcerative colitis’,’ OR ‘inflammatory bowel diseases’ and ‘paleo’, and no records were found.
Exclusion dietBenefitScientific evidence
Specific carbohydrate dietClinical and mucosal improvements in paediatric CDSmall monocentric prospective pilot study: 9 to 7 patients for 12, 52 weeks of specific carbohydrate diet131
Low-FODMAP dietDecreased functional gastrointestinal symptomsPilot study: 52 CD and 20 UC patients [p < 0.02 for all]132
69/88 IBD patients [p < 0.001]133
Gluten-free diet [GFD]Improved self-reported gastrointestinal symptoms [65.6%]
Fewer or less severe IBD flares [38.3%]
Crohn’s & Colitis Foundation of America internet-based cohort: 449/1647 participants had previously tried a GFD or reported current use of GFD12
Paleo dietA literature search using PubMed from inception to April 2016 was performed to identify relevant studies using the terms: ‘Crohn’ OR ‘ulcerative colitis’,’ OR ‘inflammatory bowel diseases’ and ‘paleo’, and no records were found.

This will require much work and clinical research, and these studies are challenging to perform. Indeed, large prospective controlled clinical trials are required for providing accurate dietary recommendations to patients. Many of the current recommendations are empiric. A specific opportunity might be to conduct interventional studies in patients undergoing resection and anastomosis due to complications of CD—such patients most closely replicate evolution of the disease with histological recurrence, endoscopic recurrence, clinical recurrence, complications and surgery. A further opportunity is to study patients with UC who are in remission to determine whether adjunctive dietary intervention [in addition to pharmacological therapy] in a randomized controlled study may provide benefits in terms of maintenance of remission.

However, as previously mentioned, restriction of dairy products is frequent in IBD patients.15, 128 This may contribute to calcium deficiency. Appointment with a dietitian may clarify why the patient is limiting his/her dairy intake. In the case of lactose malabsorption or dairy fear, the dietitian can suggest alternative sources of calcium other than from a dairy source according to individual patient taste or dietary lifestyle, for example, calcium-rich water or calcium-enriched orange juice. Since the effects of dairy products on symptoms are linked to their fat content,16 low fat cheese can be advised. Calcium supplementation can also be proposed.

While red meat and processed meat intake are associated with potential relapse,14, 25 iron deficiency is frequent in IBD patients.126 At first, IBD patients often consume protein and total fat to excess,126 and the dietitian can translate nutritional dietary guidelines into practical advice to the IBD patients to limit their meat, especially red meat consumption; meat alternatives may be an option. A personalized menu adapted to individual IBD patient taste, individual nutritional needs, and disease course can be programmed and regularly revised as required.

9. Conclusions

Nutrients are very involved in the innate immunity processes implicated in IBD pathogenesis, and the evidence presented here provides a rationale for pushing clinical research forward on this topic. Patients and physicians need to overcome the common ‘if it hurts, don’t do it’ method of guiding diet and reach a more evidence-based nutritional counselling practice.

Funding

No specific funding has been received—this study is part of our routine work.

Conflict of Interest

None.

Author Contributions

Rachel Marion-Letellier: literature search, figures, writing; Guillaume Savoye: literature search, writing; Subrata Ghosh: literature search, writing.

References

1.

Chapman-Kiddell
CA
Davies
PS
Gillen
L
Radford-Smith
GL
.
Role of diet in the development of inflammatory bowel disease
.
Inflamm Bowel Dis
2010
;
16
:
137
51
.

2.

Limdi
JK
Aggarwal
D
McLaughlin
JT
.
Dietary practices and beliefs in patients with inflammatory bowel disease
.
Inflamm Bowel Dis
2016
;
22
:
164
70
.

3.

Zallot
C
Quilliot
D
Chevaux
JB
et al. .
Dietary beliefs and behavior among inflammatory bowel disease patients
.
Inflamm Bowel Dis
2013
;
19
:
66
72
.

4.

Dziechciarz
P
Horvath
A
Shamir
R
Szajewska
H
.
Meta-analysis: enteral nutrition in active Crohn’s disease in children
.
Aliment Pharmacol Ther
2007
;
26
:
795
806
.

5.

Guo
Z
Wu
R
Zhu
W
et al. .
Effect of exclusive enteral nutrition on health-related quality of life for adults with active Crohn’s disease
.
Nutr Clin Pract
2013
;
28
:
499
505
.

6.

Baumgart
DC
Carding
SR
.
Inflammatory bowel disease: cause and immunobiology
.
Lancet
2007
;
369
:
1627
40
.

7.

Shoda
R
Matsueda
K
Yamato
S
Umeda
N
.
Epidemiologic analysis of Crohn disease in Japan: increased dietary intake of n-6 polyunsaturated fatty acids and animal protein relates to the increased incidence of Crohn disease in Japan
.
Am J Clin Nutr
1996
;
63
:
741
5
.

8.

Martinez-Medina
M
Denizot
J
Dreux
N
et al. .
Western diet induces dysbiosis with increased E coli in CEABAC10 mice, alters host barrier function favouring AIEC colonisation
.
Gut
2014
;
63
:
116
24
.

9.

De Filippo
C
Cavalieri
D
Di Paola
M
et al. .
Impact of diet in shaping gut microbiota revealed by a comparative study in children from Europe and rural Africa
.
Proc Natl Acad Sci U S A
2010
;
107
:
14691
6
.

10.

Cohen
AB
Lee
D
Long
MD
et al. .
Dietary patterns and self-reported associations of diet with symptoms of inflammatory bowel disease
.
Dig Dis Sci
2013
;
58
:
1322
8
.

11.

Hou
JK
Lee
D
Lewis
J
.
Diet and inflammatory bowel disease: review of patient-targeted recommendations
.
Clin Gastroenterol Hepatol
2014
;
12
:
1592
600
.

12.

Herfarth
HH
Martin
CF
Sandler
RS
Kappelman
MD
Long
MD
.
Prevalence of a gluten-free diet and improvement of clinical symptoms in patients with inflammatory bowel diseases
.
Inflamm Bowel Dis
2014
;
20
:
1194
7
.

13.

Hwang
C
Ross
V
Mahadevan
U
.
Popular exclusionary diets for inflammatory bowel disease: the search for a dietary culprit
.
Inflamm Bowel Dis
2014
;
20
:
732
41
.

14.

Jowett
SL
Seal
CJ
Pearce
MS
et al. .
Influence of dietary factors on the clinical course of ulcerative colitis: a prospective cohort study
.
Gut
2004
;
53
:
1479
84
.

15.

Vernia
P
Loizos
P
Di Giuseppantonio
I
Amore
B
Chiappini
A
Cannizzaro
S
.
Dietary calcium intake in patients with inflammatory bowel disease
.
J Crohns Colitis
2014
;
8
:
312
7
.

16.

Nolan-Clark
D
Tapsell
LC
Hu
R
Han
DY
Ferguson
LR
.
Effects of dairy products on Crohn’s disease symptoms are influenced by fat content and disease location but not lactose content or disease activity status in a New Zealand population
.
J Am Diet Assoc
2011
;
111
:
1165
72
.

17.

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
17
.

18.

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
86
.

19.

Wu
S
Zhang
YG
Lu
R
Xia
Y
Zhou
D
Petrof
EO
Claud
EC
Chen
D
Chang
EB
Carmeliet
G
Sun
J
.
Intestinal epithelial vitamin D receptor deletion leads to defective autophagy in colitis
.
Gut
2015;
64
(
7
):
1082
94
.

20.

Richman
E
Rhodes
JM
.
Review article: evidence-based dietary advice for patients with inflammatory bowel disease
.
Aliment Pharmacol Ther
2013
;
38
:
1156
71
.

21.

Cosnes
J
Gower-Rousseau
C
Seksik
P
Cortot
A
.
Epidemiology and natural history of inflammatory bowel diseases
.
Gastroenterology
2011
;
140
:
1785
94
.

22.

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
82
.

23.

Devkota
S
Wang
Y
Musch
MW
et al. .
Dietary-fat-induced taurocholic acid promotes pathobiont expansion and colitis in Il10-/- mice
.
Nature
2012
;
487
:
104
8
.

24.

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
73
.

25.

Jantchou
P
Morois
S
Clavel-Chapelon
F
Boutron-Ruault
M-C
Carbonnel
F
.
Animal protein intake and risk of inflammatory bowel disease: the E3N prospective study
.
Am J Gastroenterol
2010
;
105
:
2195
201
.

26.

Blasbalg
TL
Hibbeln
JR
Ramsden
CE
Majchrzak
SF
Rawlings
RR
.
Changes in consumption of omega-3 and omega-6 fatty acids in the United States during the 20th century
.
Am J Clin Nutr
2011
;
93
:
950
62
.

27.

Tjonneland
A
Overvad
K
Bergmann
MM
et al. .
Linoleic acid, a dietary n-6 polyunsaturated fatty acid, and the aetiology of ulcerative colitis: a nested case–control study within a European prospective cohort study
.
Gut
2009
;
58
:
1606
11
.

28.

Ananthakrishnan
AN
Khalili
H
Konijeti
GG
et al. .
Long-term intake of dietary fat and risk of ulcerative colitis and Crohn’s disease
.
Gut
2014
;
63
:
776
84
.

29.

de Silva
PS
Luben
R
Shrestha
SS
Khaw
KT
Hart
AR
.
Dietary arachidonic and oleic acid intake in ulcerative colitis etiology: a prospective cohort study using 7-day food diaries
.
Eur J Gastroenterol Hepatol
2013
;
26
:
11
8
.

30.

John
SA
Luben
RB
Shrestha
SSB
Welch
AC
Khaw
K-TD
Hart
AR
.
Dietary n-3 polyunsaturated fatty acids and the aetiology of ulcerative colitis: a UK prospective cohort study
.
Eur J Gastroenterol Hepatol
2010
;
22
:
602
6
.

31.

Ananthakrishnan
AN
Khalili
H
Konijeti
GG
et al. .
A prospective study of long-term intake of dietary fiber and risk of Crohn’s disease and ulcerative colitis
.
Gastroenterology
2013
;
145
:
970
7
.

32.

Marion-Letellier
R
Butler
M
Dechelotte
P
Playford
RJ
Ghosh
S
.
Comparison of cytokine modulation by natural peroxisome proliferator-activated receptor gamma ligands with synthetic ligands in intestinal-like Caco-2 cells and human dendritic cells—potential for dietary modulation of peroxisome proliferator-activated receptor gamma in intestinal inflammation
.
Am J Clin Nutr
2008
;
87
:
939
48
.

33.

Lee
JY
Plakidas
A
Lee
WH
et al. .
Differential modulation of Toll-like receptors by fatty acids: preferential inhibition by n-3 polyunsaturated fatty acids
.
J Lipid Res
2003
;
44
:
479
86
.

34.

Hugot
J-P
Chamaillard
M
Zouali
H
et al. .
Association of NOD2 leucine-rich repeat variants with susceptibility to Crohn’s disease
.
Nature
2001
;
411
:
599
603
.

35.

Lee
D
Albenberg
L
Compher
C
et al. .
Diet in the pathogenesis and treatment of inflammatory bowel diseases
.
Gastroenterology
2015
;
148
:
1087
106
.

36.

Ghanim
H
Sia
CL
Upadhyay
M
et al. .
Orange juice neutralizes the proinflammatory effect of a high-fat, high-carbohydrate meal and prevents endotoxin increase and Toll-like receptor expression
.
Am J Clin Nutr
2010
;
91
:
940
9
.

37.

Deopurkar
R
Ghanim
H
Friedman
J
et al. .
Differential effects of cream, glucose, and orange juice on inflammation, endotoxin, and the expression of Toll-like receptor-4 and suppressor of cytokine signaling-3
.
Diabetes Care
2010
;
33
:
991
7
.

38.

Kim
MS
Choi
MS
Han
SN
.
High fat diet–induced obesity leads to proinflammatory response associated with higher expression of NOD2 protein
.
Nutr Res Pract
2011
;
5
:
219
23
.

39.

Baur
P
Martin
FP
Gruber
L
et al. .
Metabolic phenotyping of the Crohn’s disease–like IBD etiopathology in the TNFΔRE/WT Mouse Model
.
J Proteome Res
2014
;
10
:
5523
35
.

40.

Nickerson
KP
McDonald
C
.
Crohn’s disease-associated adherent-invasive Escherichia coli adhesion is enhanced by exposure to the ubiquitous dietary polysaccharide maltodextrin
.
PLoS One
2012
;
7
:
e52132
.

41.

Nickerson
KP
Homer
CR
Kessler
SP
et al. .
The dietary polysaccharide maltodextrin promotes Salmonella survival and mucosal colonization in mice
.
PLoS One
2014
;
9
:
e101789
.

42.

Chassaing
B
Koren
O
Goodrich
JK
et al. .
Dietary emulsifiers impact the mouse gut microbiota promoting colitis and metabolic syndrome
.
Nature
2015
;
519
:
92
6
.

43.

Erridge
C
.
Accumulation of stimulants of Toll-like receptor (TLR)-2 and TLR4 in meat products stored at 5 °C
.
J Food Sci
2011
;
76
:
H72
9
.

44.

Erridge
C
.
Stimulants of Toll-like receptor (TLR)-2 and TLR-4 are abundant in certain minimally-processed vegetables
.
Food Chem Toxicol
2011
;
49
:
1464
7
.

45.

Moran
GW
Dubeau
M-F
Kaplan
GG
Panaccione
R
Ghosh
S
.
The increasing weight of Crohn’s disease subjects in clinical trials: a hypothesis-generatings time-trend analysis
.
Inflamm Bowel Dis
2013
;
19
:
2949
56
.

46.

Leung
C-H
Lam
W
Ma
D-L
Gullen
EA
Cheng
Y-C
.
Butyrate mediates nucleotide-binding and oligomerisation domain (NOD) 2-dependent mucosal immune responses against peptidoglycan
.
Eur J Immunol
2009
;
39
:
3529
37
.

47.

Alva-Murillo
N
Ochoa-Zarzosa
A
López-Meza
JE
.
Short chain fatty acids (propionic and hexanoic) decrease Staphylococcus aureus internalization into bovine mammary epithelial cells and modulate antimicrobial peptide expression
.
Vet Microbiol
2012
;
155
:
324
31
.

48.

Bentley-Hewitt
K
Blatchford
P
Parkar
S
Ansell
J
Pernthaner
A
.
Digested and fermented green kiwifruit increases human β-defensin 1 and 2 production in vitro
.
Plant Foods Hum Nutr
2012
;
67
:
208
14
.

49.

Wu
S
Zhang
YG
Lu
R
et al. .
Intestinal epithelial vitamin D receptor deletion leads to defective autophagy in colitis
.
Gut
2015
;
64
:
1082
94
.

50.

Ibrahim
A
Mbodji
K
Hassan
A
et al. .
Anti-inflammatory and anti-angiogenic effect of long chain n-3 polyunsaturated fatty acids in intestinal microvascular endothelium
.
Clin Nutr
2011
;
30
:
678
87
.

51.

Liu
Y
Chen
F
Odle
J
et al. .
Fish oil enhances intestinal integrity and inhibits TLR4 and NOD2 signaling pathways in weaned pigs after LPS challenge
.
J Nutr
2012
;
142
:
2017
24
.

52.

Adolph
S
Schoeniger
A
Fuhrmann
H
Schumann
J
.
Unsaturated fatty acids as modulators of macrophage respiratory burst in the immune response against Rhodococcus equi and Pseudomonas aeruginosa
.
Free Radic Biol Med
2012
;
52
:
2246
53
.

53.

Bertrand
J
Goichon
A
Chan
P
et al. .
Enteral glutamine infusion modulates ubiquitination of heat shock proteins, Grp-75 and Apg-2, in the human duodenal mucosa
.
Amino Acids
2014
;
46
:
1059
67
.

54.

Coeffier
M
Le Pessot
F
Leplingard
A
et al. .
Acute enteral glutamine infusion enhances heme oxygenase-1 expression in human duodenal mucosa
.
J Nutr
2002
;
132
:
2570
3
.

55.

Coeffier
M
Marion
R
Ducrotte
P
Dechelotte
P
.
Modulating effect of glutamine on IL-1beta–induced cytokine production by human gut
.
Clin Nutr
2003
;
22
:
407
13
.

56.

Coeffier
M
Marion
R
Leplingard
A
Lerebours
E
Ducrotte
P
Dechelotte
P
.
Glutamine decreases interleukin-8 and interleukin-6 but not nitric oxide and prostaglandins e(2) production by human gut in-vitro
.
Cytokine
2002
;
18
:
92
7
.

57.

Coeffier
M
Marion-Letellier
R
Dechelotte
P
.
Potential for amino acids supplementation during inflammatory bowel diseases
.
Inflamm Bowel Dis
2010
;
16
:
518
24
.

58.

Deniel
N
Marion-Letellier
R
Charlionet
R
et al. .
Glutamine regulates the human epithelial intestinal HCT-8 cell proteome under apoptotic conditions
.
Mol Cell Proteomics
2007
;
6
:
1671
9
.

59.

Lecleire
S
Hassan
A
Marion-Letellier
R
et al. .
Combined glutamine and arginine decrease proinflammatory cytokine production by biopsies from Crohn’s patients in association with changes in nuclear factor-kappaB and p38 mitogen-activated protein kinase pathways
.
J Nutr
2008
;
138
:
2481
6
.

60.

Marion
R
Coeffier
M
Leplingard
A
Favennec
L
Ducrotte
P
Dechelotte
P
.
Cytokine-stimulated nitric oxide production and inducible NO-synthase mRNA level in human intestinal cells: lack of modulation by glutamine
.
Clin Nutr
2003
;
22
:
523
8
.

61.

Marion
R
Coeffier
MM
Gargala
G
Ducrotte
P
Dechelotte
PP
.
Glutamine and CXC chemokines IL-8, Mig, IP-10 and I-TAC in human intestinal epithelial cells
.
Clin Nutr
2004
;
23
:
579
85
.

62.

Mbodji
K
Torre
S
Haas
V
Dechelotte
P
Marion-Letellier
R
.
Alanyl-glutamine restores maternal deprivation–induced TLR4 levels in a rat neonatal model
.
Clin Nutr
2011
;
30
:
672
7
.

63.

Thebault
S
Deniel
N
Marion
R
et al. .
Proteomic analysis of glutamine-treated human intestinal epithelial HCT-8 cells under basal and inflammatory conditions
.
Proteomics
2006
;
6
:
3926
37
.

64.

Kessel
A
Toubi
E
Pavlotzky
E
et al. .
Treatment with glutamine is associated with down-regulation of Toll-like receptor-4 and myeloid differentiation factor 88 expression and decrease in intestinal mucosal injury caused by lipopolysaccharide endotoxaemia in a rat
.
Clin Exp Immunol
2008
;
151
:
341
7
.

65.

Hou
YC
Liu
JJ
Pai
MH
Tsou
SS
Yeh
SL
.
Alanyl-glutamine administration suppresses Th17 and reduces inflammatory reaction in dextran sulfate sodium–induced acute colitis
.
Int Immunopharmacol
2013
;
17
:
1
8
.

66.

Asehnoune
K
Strassheim
D
Mitra
S
Kim
JY
Abraham
E
.
Involvement of reactive oxygen species in Toll-Like Receptor 4-dependent activation of NF-κB
.
J Immunol
2004
;
172
:
2522
9
.

67.

Chen
Y
Chen
D
Tian
G
et al. .
Dietary arginine supplementation alleviates immune challenge induced by Salmonella enterica serovar Choleraesuis bacterin potentially through the Toll-like receptor 4-myeloid differentiation factor 88 signalling pathway in weaned piglets
.
Br J Nutr
2012
;
108
:
1069
76
.

68.

Wu
HS
Zhang
L
Chen
Y
et al. .
Effect of nitric oxide on toll-like receptor 2 and 4 gene expression in rats with acute lung injury complicated by acute hemorrhage necrotizing pancreatitis
.
Hepatobiliary Pancreat Dis Int
2005
;
4
:
609
13
.

69.

Sherman
H
Chapnik
N
Froy
O
.
Albumin and amino acids upregulate the expression of human beta-defensin 1
.
Mol Immunol
2006
;
43
:
1617
23
.

70.

Zelante
T
Iannitti
RG
Cunha
C
et al. .
Tryptophan catabolites from microbiota engage aryl hydrocarbon receptor and balance mucosal reactivity via interleukin-22
.
Immunity
2013
;
39
:
372
85
.

71.

Martinez-Maqueda
D
Miralles
B
De Pascual-Teresa
S
Reveron
I
Munoz
R
Recio
I
.
Food-derived peptides stimulate mucin secretion and gene expression in intestinal cells
.
J Agric Food Chem
2012
;
60
:
8600
5
.

72.

Plaisancie
P
Claustre
J
Estienne
M
et al. .
A novel bioactive peptide from yoghurts modulates expression of the gel-forming MUC2 mucin as well as population of goblet cells and Paneth cells along the small intestine
.
J Nutr Biochem
2012
;
24
:
213
21
.

73.

Binion
DG
Otterson
MF
Rafiee
P
.
Curcumin inhibits VEGF-mediated angiogenesis in human intestinal microvascular endothelial cells through COX-2 and MAPK inhibition
.
Gut
2008
;
57
:
1509
17
.

74.

Deguchi
Y
Andoh
A
Inatomi
O
et al. .
Curcumin prevents the development of dextran sulfate Sodium (DSS)-induced experimental colitis
.
Dig Dis Sci
2007
;
52
:
2993
8
.

75.

Ukil
A
Maity
S
Karmakar
S
Datta
N
Vedasiromoni
JR
Das
PK
.
Curcumin, the major component of food flavour turmeric, reduces mucosal injury in trinitrobenzene sulphonic acid-induced colitis
.
Br J Pharmacol
2003
;
139
:
209
18
.

76.

Zhang
M
Deng
C
Zheng
J
Xia
J
Sheng
D
.
Curcumin inhibits trinitrobenzene sulphonic acid–induced colitis in rats by activation of peroxisome proliferator-activated receptor gamma
.
Int Immunopharmacol
2006
;
6
:
1233
42
.

77.

Holt
PR
Katz
S
Kirshoff
R
.
Curcumin therapy in inflammatory bowel disease: a pilot study
.
Dig Dis Sci
2005
;
50
:
2191
3
.

78.

Lang
A
Salomon
N
Wu
JCY
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
9.e1
.

79.

Lubbad
A
Oriowo
MA
Khan
I
.
Curcumin attenuates inflammation through inhibition of TLR-4 receptor in experimental colitis
.
Mol Cell Biochem
2009
;
322
:
127
35
.

80.

Zhao
L
Lee
JY
Hwang
DH
.
Inhibition of pattern recognition receptor–mediated inflammation by bioactive phytochemicals
.
Nutr Rev
2011
;
69
:
310
20
.

81.

Huang
S
Zhao
L
Kim
K
Lee
DS
Hwang
DH
.
Inhibition of Nod2 signaling and target gene expression by curcumin
.
Mol Pharmacol
2008
;
74
:
274
81
.

82.

McCann
MJ
Johnston
S
Reilly
K
et al. .
The effect of turmeric (Curcuma longa) extract on the functionality of the solute carrier protein 22 A4 (SLC22A4) and interleukin-10 (IL-10) variants associated with inflammatory bowel disease
.
Nutrients
2014
;
6
:
4178
90
.

83.

Yuan
Z
Syed
MA
Panchal
D
Rogers
D
Joo
M
Sadikot
RT
.
Curcumin mediated epigenetic modulation inhibits TREM-1 expression in response to lipopolysaccharide
.
Int J Biochem Cell Biol
2012
;
44
:
2032
43
.

84.

Park
SH
Kyeong
MS
Hwang
Y
Ryu
SY
Han
SB
Kim
Y
.
Inhibition of LPS binding to MD-2 co-receptor for suppressing TLR4-mediated expression of inflammatory cytokine by 1-dehydro-10-gingerdione from dietary ginger
.
Biochem Biophys Res Commun
2012
;
419
:
735
40
.

85.

Youn
HS
Lim
HJ
Lee
HJ
et al. .
Garlic (Allium sativum) extract inhibits lipopolysaccharide-induced Toll-like receptor 4 dimerization
.
Biosci Biotechnol Biochem
2008
;
72
:
368
75
.

86.

Youn
HS
Lee
JK
Choi
YJ
et al. .
Cinnamaldehyde suppresses toll-like receptor 4 activation mediated through the inhibition of receptor oligomerization
.
Biochem Pharmacol
2008
;
75
:
494
502
.

87.

Jakus
PB
Kalman
N
Antus
C
et al. .
TRAF6 is functional in inhibition of TLR4-mediated NF-κB activation by resveratrol
.
J Nutr Biochem
2013
;
24
:
819
23
.

88.

Ran
ZH
Chen
C
Xiao
SD
.
Epigallocatechin-3-gallate ameliorates rats colitis induced by acetic acid
.
Biomed Pharmacother
2008
;
62
:
189
96
.

89.

Khalfaoui
T
Groulx
JF
Sabra
G
et al. .
Laminin receptor 37/67LR regulates adhesion and proliferation of normal human intestinal epithelial cells
.
PLoS One
2013
;
8
:
e74337
.

90.

Byun
EH
Omura
T
Yamada
K
Tachibana
H
.
Green tea polyphenol epigallocatechin-3-gallate inhibits TLR2 signaling induced by peptidoglycan through the polyphenol sensing molecule 67-kDa laminin receptor
.
FEBS Lett
2011
;
585
:
814
20
.

91.

Byun
E-B
Choi
H-G
Sung
N-Y
Byun
E-H
.
Green tea polyphenol epigallocatechin-3-gallate inhibits TLR4 signaling through the 67-kDa laminin receptor on lipopolysaccharide-stimulated dendritic cells
.
Biochem Biophys Res Commun
2012
;
426
:
480
5
.

92.

Ellis
LZ
Liu
W
Luo
Y
et al. .
Green tea polyphenol epigallocatechin-3-gallate suppresses melanoma growth by inhibiting inflammasome and IL-1β2 secretion
.
Biochem Biophys Res Commun
2011
;
414
:
551
6
.

93.

Hu
Q-H
Zhang
X
Pan
Y
Li
Y-C
Kong
L-D
.
Allopurinol, quercetin and rutin ameliorate renal NLRP3 inflammasome activation and lipid accumulation in fructose-fed rats
.
Biochem Pharmacol
2012
;
84
:
113
25
.

94.

Huang
TT
Lai
HC
Chen
YB
et al. .
cis-Resveratrol produces anti-inflammatory effects by inhibiting canonical and non-canonical inflammasomes in macrophages
.
Innate Immun
2014
;
20
:
735
50
.

95.

Zgheib
A
Lamy
S
Annabi
B
.
Epigallocatechin gallate targeting of membrane type 1 matrix metalloproteinase-mediated Src and Janus kinase/signal transducers and activators of transcription 3 signaling inhibits transcription of colony-stimulating factors 2 and 3 in mesenchymal stromal cells
.
J Biol Chem
2013
;
288
:
13378
86
.

96.

Bognar
E
Sarszegi
Z
Szabo
A
et al. .
Antioxidant and anti-inflammatory effects in RAW264.7 macrophages of malvidin, a major red wine polyphenol
.
PLoS One
2013
;
8
:
e65355
.

97.

Chung
EY
Kim
BH
Hong
JT
et al. .
Resveratrol down-regulates interferon-gamma-inducible inflammatory genes in macrophages: molecular mechanism via decreased STAT-1 activation
.
J Nutr Biochem
2010
;
22
:
902
9
.

98.

Liu
L
Liu
YL
Liu
GX
et al. .
Curcumin ameliorates dextran sulfate sodium-induced experimental colitis by blocking STAT3 signaling pathway
.
Int Immunopharmacol
2013
;
17
:
314
20
.

99.

Raman
M
Milestone
AN
Walters
JR
Hart
AL
Ghosh
S
.
Vitamin D and gastrointestinal diseases: inflammatory bowel disease and colorectal cancer
.
Therap Adv Gastroenterol
2011
;
4
:
49
62
.

100.

Ananthakrishnan
AN
Khalili
H
Higuchi
LM
et al. .
Higher predicted vitamin D status is associated with reduced risk of Crohn’s disease
.
Gastroenterology
2011
;
142
:
482
9
.

101.

Jantchou
P
Clavel-Chapelon
F
Racine
A
Kvaskoff
M
Carbonnel
F
Boutron-Ruault
MC
.
High residential sun exposure is associated with a low risk of incident Crohn’s disease in the prospective E3N cohort
.
Inflamm Bowel Dis
2013
;
20
:
75
81
.

102.

Khoo
A-L
Chai
LYA
Koenen
HJPM
et al. .
Vitamin D3 down-regulates proinflammatory cytokine response to Mycobacterium tuberculosis through pattern recognition receptors while inducing protective cathelicidin production
.
Cytokine
2011
;
55
:
294
300
.

103.

Klebanoff
CA
Spencer
SP
Torabi-Parizi
P
et al. .
Retinoic acid controls the homeostasis of pre-cDC-derived splenic and intestinal dendritic cells
.
J Exp Med
2013
;
210
:
1961
76
.

104.

Rizzo
A
Losacco
A
Carratelli
CR
Domenico
MD
Bevilacqua
N
.
Lactobacillus plantarum reduces Streptococcus pyogenes virulence by modulating the IL-17, IL-23 and Toll-like receptor 2/4 expressions in human epithelial cells
.
Int Immunopharmacol
2013
;
17
:
453
61
.

105.

Rizzo
A
Losacco
A
Carratelli
CR
.
Lactobacillus crispatus modulates epithelial cell defense against Candida albicans through Toll-like receptors 2 and 4, interleukin 8 and human beta-defensins 2 and 3
.
Immunol Lett
2013
;
156
:
102
9
.

106.

Bermudez-Brito
M
Munoz-Quezada
S
Gomez-Llorente
C
et al. .
Human intestinal dendritic cells decrease cytokine release against Salmonella infection in the presence of Lactobacillus paracasei upon TLR activation
.
PLoS One
2012
;
7
:
e43197
.

107.

Liu
Y
Fatheree
NY
Mangalat
N
Rhoads
JM
.
Lactobacillus reuteri strains reduce incidence and severity of experimental necrotizing enterocolitis via modulation of TLR4 and NF-kappaB signaling in the intestine
.
Am J Physiol Gastrointest Liver Physiol
2011
;
302
:
G608
17
.

108.

Ciorba
MA
Riehl
TE
Rao
MS
et al. .
Lactobacillus probiotic protects intestinal epithelium from radiation injury in a TLR-2/cyclo-oxygenase-2–dependent manner
.
Gut
2011
;
61
:
829
38
.

109.

Natividad
JM
Petit
V
Huang
X
et al. .
Commensal and probiotic bacteria influence intestinal barrier function and susceptibility to colitis in Nod1-/-; Nod2-/- mice
.
Inflamm Bowel Dis
2011
;
18
:
1434
46
.

110.

Doherty
GA
Bennett
GC
Cheifetz
AS
Moss
AC
.
Meta-analysis: targeting the intestinal microbiota in prophylaxis for post-operative Crohn’s disease
.
Aliment Pharmacol Ther
2010
;
31
:
802
9
.

111.

Marteau
P
Lemann
M
Seksik
P
et al. .
Ineffectiveness of Lactobacillus johnsonii LA1 for prophylaxis of postoperative recurrence in Crohn’s disease: a randomised, double blind, placebo controlled GETAID trial
.
Gut
2006
;
55
:
842
7
.

112.

Singh
S
Stroud
AM
Holubar
SD
Sandborn
WJ
Pardi
DS
.
Treatment and prevention of pouchitis after ileal pouch–anal anastomosis for chronic ulcerative colitis
.
Cochrane Database Syst Rev
2015
;
11
:
CD001176
.

113.

Valcheva
R
Hotte
N
Gillevet
P
Sikaroodi
M
Thiessen
A
Madsen
KL
.
Soluble dextrin fibers alter the intestinal microbiota and reduce proinflammatory cytokine secretion in male IL-10–deficient mice
.
J Nutr
2015
;
145
:
2060
6
.

114.

Hu
Y
Le Leu
RK
Christophersen
CT
et al. .
Manipulation of the gut microbiota using resistant starch is associated with protection against colitis-associated colorectal cancer in rats
.
Carcinogenesis
2016
;
37
:
366
75
.

115.

Gibson
PR
Barrett
JS
Muir
JG
.
Functional bowel symptoms and diet
.
Intern Med J
2013
;
43
:
1067
74
.

116.

Nagahara
Y
Nagamori
T
Tamegai
H
et al. .
Inulin stimulates phagocytosis of PMA-treated THP-1 macrophages by involvement of PI3-kinases and MAP kinases
.
BioFactors
2011
;
37
:
447
54
.

117.

Xu
Q
Yajima
T
Li
W
Saito
K
Ohshima
Y
Yoshikai
Y
.
Levan (β-2, 6-fructan), a major fraction of fermented soybean mucilage, displays immunostimulating properties via Toll-like receptor 4 signalling: induction of interleukin-12 production and suppression of T-helper type 2 response and immunoglobulin E production
.
Clin Exp Allergy
2006
;
36
:
94
101
.

118.

Kovacs-Nolan
J
Kanatani
H
Nakamura
A
Ibuki
M
Mine
Y
.
beta-1,4-mannobiose stimulates innate immune responses and induces TLR4-dependent activation of mouse macrophages but reduces severity of inflammation during endotoxemia in mice
.
J Nutr
2013
;
143
:
384
91
.

119.

Liu
L
Li
YH
Niu
YB
et al. .
An apple oligogalactan prevents against inflammation and carcinogenesis by targeting LPS/TLR4/NF-κB pathway in a mouse model of colitis-associated colon cancer
.
Carcinogenesis
2010
;
31
:
1822
32
.

120.

Raynaud
A
Ghezali
L
Gloaguen
V
Liagre
B
Quero
F
Petit
JM
.
Honey-induced macrophage stimulation: AP-1 and NF-kappaB activation and cytokine production are unrelated to LPS content of honey
.
Int Immunopharmacol
2013
;
17
:
874
9
.

121.

Tonks
AJ
Dudley
E
Porter
NG
et al. .
A 5.8-kDa component of manuka honey stimulates immune cells via TLR4
.
J Leukoc Biol
2007
;
82
:
1147
55
.

122.

Benjamin
JL
Hedin
CRH
Koutsoumpas
A
et al. .
Randomised, double-blind, placebo-controlled trial of fructo-oligosaccharides in active Crohn’s disease
.
Gut
2011
;
60
:
923
9
.

123.

Casellas
F
Borruel
N
Torrejon
A
et al. .
Oral oligofructose-enriched inulin supplementation in acute ulcerative colitis is well tolerated and associated with lowered faecal calprotectin
.
Aliment Pharmacol Ther
2007
;
25
:
1061
7
.

124.

Pallares
V
Calay
D
Cedo
L
et al. .
Enhanced anti-inflammatory effect of resveratrol and EPA in treated endotoxin-activated RAW 264.7 macrophages
.
Br J Nutr
2012
;
108
:
1562
73
.

125.

Mbodji
K
Charpentier
C
Guerin
C
et al. .
Adjunct therapy of n-3 fatty acids to 5-ASA ameliorates inflammatory score and decreases NF-kappaB in rats with TNBS-induced colitis
.
J Nutr Biochem
2012
;
24
:
700
5
.

126.

Walton
M
Alaunyte
I
.
Do patients living with ulcerative colitis adhere to healthy eating guidelines? A cross-sectional study
.
Br J Nutr
2014
;
112
:
1628
35
.

127.

Brown
AC
Rampertab
SD
Mullin
GE
.
Existing dietary guidelines for Crohn’s disease and ulcerative colitis
.
Expert Rev Gastroenterol Hepatol
2011
;
5
:
411
25
.

128.

Brasil Lopes
M
Rocha
R
Castro Lyra
A
et al. .
Restriction of dairy products; a reality in inflammatory bowel disease patients
.
Nutr Hosp
2014
;
29
:
575
81
.

129.

Carrillo
C
Cavia Mdel
M
Alonso-Torre
S
.
Role of oleic acid in immune system; mechanism of action; a review
.
Nutr Hosp
2012
;
27
:
978
90
.

130.

John
S
Luben
R
Shrestha
SS
Welch
A
Khaw
KT
Hart
AR
.
Dietary n-3 polyunsaturated fatty acids and the aetiology of ulcerative colitis: a UK prospective cohort study
.
Eur J Gastroenterol Hepatol
2010
;
22
:
602
6
.

131.

Cohen
SA
Gold
BD
Oliva
S
et al. .
Clinical and mucosal improvement with specific carbohydrate diet in pediatric Crohn disease
.
J Pediatr Gastroenterol Nutr
2014
;
59
:
516
21
.

132.

Gearry
RB
Irving
PM
Barrett
JS
Nathan
DM
Shepherd
SJ
Gibson
PR
.
Reduction of dietary poorly absorbed short-chain carbohydrates (FODMAPs) improves abdominal symptoms in patients with inflammatory bowel disease—a pilot study
.
J Crohns Colitis
2009
;
3
:
8
14
.

133.

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
36
.

Author notes

Corresponding author: Dr Rachel Marion-Letellier, PhD, INSERM Unit UMR1073, Faculté de Médecine-Pharmacie, Institut Fédératif de Recherches Multidisciplinaires sur les Peptides [I.F.R. 23], 22, boulevard Gambetta, 76183 Rouen cedex, France. Tel: 33-2-35-14-82-45; Fax: 33-2-35-14-82-26; Email: [email protected]