Diet and Nutrients and Immune
Diet and Nutrients and Immune
Diet and Nutrients and Immune
Recent findings point toward diet having a major impact on human health. Diets can either
affect the gut microbiota resulting in alterations in the host’s physiological responses
or by directly targeting the host response. The microbial community in the mammalian
gut is a complex and dynamic system crucial for the development and maturation of
both systemic and mucosal immune responses. Therefore, the complex interaction
between available nutrients, the microbiota, and the immune system are central regu-
lators in maintaining homeostasis and fighting against invading pathogens at mucosal
sites. Westernized diet, defined as high dietary intake of saturated fats and sucrose
Edited by:
Raquel Hontecillas, and low intake of fiber, represent a growing health risk contributing to the increased
Virginia Tech, United States occurrence of metabolic diseases, e.g., diabetes and obesity in countries adapting a
Reviewed by: westernized lifestyle. Inflammatory bowel diseases (IBD) and asthma are chronic muco-
Mourad Aribi,
University of Tlemcen, Algeria
sal inflammatory conditions of unknown etiology with increasing prevalence worldwide.
Christopher Alan Jolly, These conditions have a multifactorial etiology including genetic factors, environmental
University of Texas at Austin, factors, and dysregulated immune responses. Their increased prevalence cannot solely
United States
be attributed to genetic considerations implying that other factors such as diet can be
*Correspondence:
Silvia Melgar a major contributor. Recent reports indicate that the gut microbiota and modifications
[email protected] thereof, due to a consumption of a diet high in saturated fats and low in fibers, can
trigger factors regulating the development and/or progression of both conditions. While
Specialty section:
This article was submitted to
asthma is a disease of the airways, increasing evidence indicates a link between the gut
Nutritional Immunology, and airways in disease development. Herein, we provide a comprehensive review on the
a section of the journal impact of westernized diet and associated nutrients on immune cell responses and the
Frontiers in Immunology
microbiota and how these can influence the pathology of IBD and asthma.
Received: 31 January 2017
Accepted: 03 July 2017 Keywords: westernized diet, inflammatory bowel disease, asthma, saturated fat, micronutrients, microbiota
Published: 28 July 2017
Citation:
GENERAL INTRODUCTION
Statovci D, Aguilera M, MacSharry J
and Melgar S (2017) The Impact
The prevalence of chronic inflammatory diseases affecting mucosal sites such as the intestine and the
of Western Diet and Nutrients
on the Microbiota and Immune
airways is increasing worldwide (1, 2). Among these, inflammatory bowel disease [IBD, mainly com-
Response at Mucosal Interfaces. prising ulcerative colitis (UC) and Crohn’s Disease (CD)] and allergic asthma are the most relevant.
Front. Immunol. 8:838. Recent findings point toward potential links between these two pathologies, e.g., histamine and mast
doi: 10.3389/fimmu.2017.00838 cell activity and immunoglobulin E (IgE) production, reviewed in Ref. (3). Both diseases have a
multifactorial cause, in which environmental factors such as diet and immune modulators are the current treatments of choice
and the commensal microbiota are gaining increased attention. for patients with IBD, with the overall goal being inflammation
In this regard, consumption of the so-called “Westernized” diet is reduction, increase of mucosal healing and symptom relief
associated with increased risk for IBD (4) and asthma morbidity (16, 44). New treatment strategies include inhibitors targeting
(5). Westernized diet is characterized by a high content of proteins leukocyte trafficking (α4β7 integrin, sphingosine-1-phosphate,
(derived from fatty domesticated and processed meats), saturated S1P), cytokine [interleukin (IL)-12p40, IL-23], janus kinase-
fats, refined grains, sugar, alcohol, salt, and corn-derived fructose pathway, inflammasome (NLRP3/IL-1β), to name a few (45, 46).
syrup, with an associated reduced consumption of fruits and The etiology of IBD is still unknown but the evidence points
vegetables (4, 6, 7). Research in the last decade has uncovered toward environmental factors, with the microbiota being of
that changes from a diet rich in fibers and low in fats to a diet particular interest. The microbiota can trigger and/or sustain a
low in fibers and high in saturated fats directly contributes to the tissue damaging immune response in genetically susceptible indi-
development of obesity, metabolic syndrome, and cardiovascular viduals. Research in the last decade has identified a crucial role for
diseases (8, 9). Macronutrients (carbohydrates, lipids, and protein) the commensal bacteria in the pathogenesis of IBD. Experimental
and micronutrients (vitamins and minerals) are required for our IBD was not evidenced in animals raised under germ-free (GF)
body to function and several of these are naturally obtained from conditions when compared to groups of conventionalized animals
our diets and from the resident microbiota. Both patients with (47). Antibiotics have proven beneficial to certain subgroups of
IBD and asthma present nutritional problems leading to several patients with CD but not in patients with UC (48) and inflamma-
complications including anemia, osteoporosis, acute respiratory tory lesions are more frequently located in areas with large bacte-
infections, etc. The link between diet, nutrients and immune rial burden, i.e., ileum and colon (49). Alterations in the enteric
responses is embedded in a complex network of signals and has microbial flora reported in patients with IBD include decreases in
to be considered in the light of other factors including microbial Firmicutes and Bacteroidetes and increases in Enterobacteriaceae
composition, genetic background, and lifestyle, to mention but a (e.g., Escherichia coli) (50). A reduction in anti-inflammatory
few. The advent of high-throughput Next-Generation Sequencing commensals, such as Faecalibacterium prausnitzii has been asso-
technologies has driven the discovery and dissection of regula- ciated with CD (51). Several pathogenic bacteria are suggested as
tory mechanisms involved in the disease state. In this review, etiological agents of IBD but to date none has been identified to
we will focus on the interactions between diets and nutrients cause IBD (47). In contrast, pathobionts, i.e., commensal bacteria
associated with Westernized regimes and their impact on the with potential pathological properties, have been isolated, includ-
microbiota and immune responses at mucosal interfaces, i.e., ing strains of adherent and invasive E. coli (AIEC), commonly
the intestines and lungs. We will outline the complex network identified in the mucosa of CD patients (52).
between nutrients, microbial alterations and abnormal immune Genome-wide association studies have so far identified over
responses associated with IBD and asthma. 160 genetic loci in IBD, with 30 loci being specific to CD, 23
Table 1 summarizes the impact of dietary factors on host loci to UC, and 110 loci are associated with both forms of IBD
responses. Figure 1 summarizes identified genes associated with (53). IBD susceptibility single-nucleotide polymorphisms were
IBD and asthma and immune responses. Figure 2 displays the identified in genes affecting innate and adaptive immune cell
regulatory interaction between diet, mucosal immunity, and function, bacterial recognition, etc. (Figure 1). Therefore, the role
commensal microbiota to maintain mucosal homeostasis and the of mononuclear phagocytes including monocytes/macrophages
resulting pathology upon loss of balance. Figures 3 and 4 outline and dendritic cells (DCs) in the development of IBD has been
the mechanisms targeted by nutrients in the healthy intestine extensively studied. Several mouse models of IBD, including
and lung and in the inflamed gut (IBD) and lung (asthma), dextran sodium sulfate (DSS)- and 2,4,6-trinitrobenzene sulfonic
respectively. acid-induced colitis and the TNFΔARE model of Crohn’s-like ile-
itis, have revealed that lamina propria mononuclear phagocytes
have protective as well as pathogenic roles during the disease
IBD—GENETICS, IMMUNE RESPONSE, progression (54–60). Three explanations have been postulated
AND MICROBIOTA to explain these findings—(1) an inappropriate response to non-
harmful commensal bacteria (i.e., NOD2, REL, CARD9); (2)
Inflammatory bowel diseases are multifactorial chronic immune- an inefficient clearance of microbes (commensals/pathobionts)
mediated diseases of the gastrointestinal (GI) tract. They often leading to chronic immune stimulation (i.e., ATG16L1, IRGM),
have an early onset and a course which is characterized by intermit- and (3) a failure to resolve inflammation by maintaining a pro-
tent phases of remission and relapses (42, 43). Nearly 2.2 million inflammatory phenotype (i.e., IL-12, IL-18RAP/IL-1R1, IFNGR/
people in Europe, 1.5 million Americans, and several 100,000 more IFNAR1) (39) (Figure 1). The intestinal epithelium functions as
individuals suffer from IBD. The prevalence and incidence of IBD a barrier between the host and its environment (microbes, non-
is increasing worldwide and its growth is not confined to Western self-antigens from diet, nutrients, etc.) and consists of highly
Europe or the USA; countries adapting a westernized lifestyle, specialized cells that fulfill this barrier task. Genes associated
such as Japan and South Africa, are seeing mounting numbers with epithelial cell function, such as HNF4A, ECM1, CDH1 have
of affected individuals (2). Patients suffering from IBD present a UC correlation (53) (Figure 1). Alterations in barrier integrity
symptoms such as abdominal pain, fever, and diarrhea with blood associated with IBD include decreased structure of tight-junction
and/or mucus excretion. Antibiotics, biologics, corticosteroids, (TJ) proteins, which regulate paracellular permeability, impaired
mucus production due to loss of goblet cells and an altered migrate to the lymph nodes and induce T cell activation (70)
production of antimicrobial peptides (61) (Figure 4). (Figure 4). The subsequent T cell immune response can result in
In terms of location, CD can affect any part of the GI tract either an allergenic Th2/eosinophilic IgE-mediated inflammation
from the mouth to rectum. However, in the majority of patients or an inflammatory Th1/neutrophilic cell influx into the airway.
with CD the inflammation is localized to the distal ileum and The Th2 allergenic response involves the interaction of DCs, Th2
proximal colon (62). The inflammation in CD is patchy and often cells, and IL-4 producing basophils which induce the expansion
transmural, which can lead to the development of fibrosis, fistu- of type 2 innate lymphoid cells (ILC2) which also produce the Th2
las, fissures, strictures, etc. A dense infiltration with macrophages cytokines, IL-5, IL-9, and IL-13, leading to eosinophil and mast
and lymphocytes and granuloma formation is a typical feature cell trafficking to the lung and goblet cell mucus secretion (70)
of the disease. Patients with CD present an imbalanced immune (Figure 4). IL-4 derived from Th2 cells also induces IgE produc-
response with high expression of innate pro-inflammatory tion by B cells. The mixed Th2 and Th1 neutrophilic or Th2 low
cytokines, including IL-1β, IL-6, and tumor necrosis factor asthma is induced by toll-like receptor (TLR) activation result-
(TNF)-α and a T helper (Th)1 (IL-12-mediated interferon ing in IL-1β secretion and activation of inflammatory Th1 and
(IFN)γ) and Th17 (IL-17a) profile resulting in an enhanced Th17 cells. These cells release IL-17a and IFNγ, which activate
and uncontrolled immune response (16, 43, 62) (Figure 4). In neutrophils and macrophages to release TNFα and induce inflam-
contrast to CD, UC is restricted to the mucosa of the colon and is matory signals. The resulting immune infiltrates induces the
associated with large infiltrates of neutrophils, T and B cells in the symptoms of asthma—bronchoconstriction, mucus production,
lamina propria. Characteristically, the inflammation originates in and the resultant tissue remodeling increasing smooth muscle
the rectum extending continuously in a proximal fashion. Crypt and collagen deposition (71, 72). The subsequent remodeling
abscesses, formed by extravasation of neutrophils through the results in airway wall thickening, compromised lung function
intestinal epithelium, ulcerations, and goblet cell loss are typical and changes in the lung microbiota (41, 73). The mechanisms
features of UC. Moreover, high levels of innate cytokines, includ- of asthma have been studied successfully over the decades using
ing IL-1β, IL-6, and TNFα, and chemokines, such as CXCL8 and murine models, such as the ovalbumin and the house dust mite
GROα/CXCL1 (neutrophil attractants), as well as an atypical Th2 challenge models (74). While all models have limitations, these
cytokine profile accompanied by high production of IL-5, IL-10, studies have led to successful therapy development and under-
transforming growth factor beta (TGFβ) and only initially IL-4 standing of the genetics of asthma (75). Asthma treatment targets
production, which is superseded by IL-13 production is associ- immune processes by using inhaled corticosteroids, to reduce
ated to patients with UC (62–65) (Figure 4). inflammation, and bronchodilators to counteract the effects of
bronchoconstriction induced by the release of histamine, prosta-
glandins, interleukins, or leukotrienes.
ASTHMA—GENETICS, IMMUNE Several genetic studies have identified asthma susceptibility
RESPONSE, AND MICROBIOTA genes, including IRAK3, SMAD3, ORMDL3, IL-1RL1, IL-13,
IL-33, TNFAIP3, and TSLP (76) (Figure 1). Recent studies
Asthma is an increasingly common heterogeneous chronic inflam- have highlighted the role of the site of the mutation and allele
matory disease, which places substantial burden on patients, their frequency and the particular site of functionality, such as the
families, and the community (66). Asthma is characterized by asthmatic epithelium, and the epigenetic regulation thereof
airway immune hyper responsiveness to inhaled environmental as being key factors contributing to asthma (77, 78). Studies
particles leading to wheezing, breathlessness, chest tightness, on DNA methylation, and microRNA modulation of gene
and coughing effecting airway function (http://ginasthma.org/). expression, are now shedding light on the pathogenesis of this
Worldwide the incidence of asthma, is increasing, with an esti- multifactorial disease.
mated 300 million affected individuals (http://ginasthma.org/). There is increasing evidence that the gut plays a key role in
Once thought to be a childhood disease it is now presenting in effecting the allergic immune response. Murine models have
respiratory clinics as first time adult onset asthma. demonstrated how feeding of gut commensals can reduce
Asthma presents with airway inflammation following expo- allergy symptoms by inducing T regulatory cells (Tregs) which
sure to insults such as allergens, pollutants, and microbes (67). migrate to the lung and reduce the immune response (79, 80).
The primary site of immune induction is initially the lung epithe- Indeed, antibiotic-mediated disruption of the gut microbiota
lium, which interacts with the underlying antigen presenting cells and mycobiota has been shown to exacerbate allergic asthma
such as DCs, inducing an immune response (Figure 4). Alveolar symptoms in mice (81, 82). Recently, an elegant study by Arrieta
macrophages in the airway lumen act as clearance and immune and colleagues found that the relative abundance of the bacterial
sampling mechanisms at the interface between the mucosa and genera Faecalibacterium, Lachnospiria, Veillonella, and Rothia
the external environment (68). The immune signaling from and Clostridium neonatale are decreased in the gut of children
epithelial cells and macrophages results in secretion of first order at risk of asthma development (83, 84). These microbes were
cytokines, such as CXCL8, IFNα, IL-1β, IL-33, TGFβ, and thymic significantly different between the groups at 3 months of age
stromal lymphopoietin (TSLP), which induce a rapid immune and the difference decreased as children reached 1 year of age
trafficking and a clearance response which subsequently results highlighting a colonization window of opportunity and of an
in second order cytokine secretion by T cells (69). Activated DCs appropriate immune education.
Figure 1 | Shared and individual inflammatory bowel disease (IBD) and Asthma susceptibility genes/loci. The outlined genes are grouped according to function. In
green color are the genes associated to asthma, in blue color are the genes associated to ulcerative colitis only, in orange color are the genes associated to Crohn’s
disease only, in purple color are the genes associated to IBD, in black color are the genes associated to asthma and Crohn’s disease or asthma and IBD,
respectively. Adapted from Ref. (37–40). Abbreviations: AK2, adenylate kinase 2; ATG16L1, autophagy related 16 like 1; CARD9, caspase recruitment domain family
member 9; CD14, cluster of differentiation 14; CDH1, cadherin 1; CREM, CAMP responsive element modulator; CTLA4, cytotoxic T-lymphocyte associated protein
4; DENND1B, DENN domain containing 1B; ECM1, extracellular matrix protein 1; FCGR2A, Fc fragment of IgG receptor IIa; FCGR2B, Fc fragment of IgG receptor
IIb; FLG, filaggrin; GNA12, G-protein subunit alpha 12; GSTM1, glutathione S-transferase mu 1; GSTP1, glutathione S-transferase pi 1; GSTT1, glutathione
S-transferase theta 1; HAVCR1, hepatitis A virus cellular receptor 1; HNF4A, hepatocyte nuclear factor 4 alpha; IL-13, interleukin 13; IL-4, interleukin 4; IL-10,
interleukin 10; IL-12B, interleukin 12B; IL-1R1, interleukin 1 receptor type 1; IL-1R2, interleukin 1 receptor type 2; IL23R, interleukin 23 receptor; IL-27, interleukin
27; IL-4R, interleukin 4 receptor; CXCR1, C–X–C motif chemokine receptor 1; CXCR2, C–X–C motif chemokine receptor 2; IRGM, immunity related GTPase M;
LAMB1, laminin subunit beta 1; LRRK2, leucine rich repeat kinase 2; LTA, lymphotoxin alpha; LTC4S, leukotriene C4 synthase; NOD2, nucleotide binding
oligomerization domain containing 2; ORMDL3, ORMDL sphingolipid biosynthesis regulator 3; REL, REL proto-oncogene, NF-κB subunit; SBNO2, strawberry notch
homolog 2; SLC11A1, solute carrier family 11 member 1; SLC22A5, solute carrier family 22 member 5; SMAD3, SMAD Family Member 3; STAT3, signal transducer
and activator of transcription 3; Th, T helper cell; TGFB1, transforming growth factor beta 1; TNF, tumor necrosis factor; TRAF1, TNF receptor associated factor 1;
TYK2, tyrosine kinase 2.
REGULATION OF MICROBIOTA BY DIET and protection of the host (Figure 2A). The commensal micro-
biota, consisting of up to 1 × 1014 bacteria, confers coloniza-
Diet has a major impact on human health, whether by affecting tion resistance against pathogens, which is a key host defense
the host directly or through changes of the microbial community. mechanism against enteric infections. Commensal microbes
The microbial community in the mammalian gut is a complex and occupy niches and exhaust nutrients thereby limiting the growth
dynamic system with a steady state (85), which can be perturbed of newcomers. Dietary changes, inflammation, and antibiotics
by many environmental factors, including diet, lifestyle, drugs, can disrupt the commensal microbial community and hence
thereby changing the host’s physiology (10, 86) (Figure 2). As a increase the risk of colonization and expansion of incoming
response to dietary changes, shifts in the composition of the gut pathogens (90). The commensal microbiota is also essential for
microbiota occur. Especially during early-life events like weaning the elimination of pathogens from the gut. For example, unlike
off and introduction to solid food, the dynamics of colonization conventional specific pathogen free mice, GF mice are unable to
are critically involved in educating as well as training the immune eradicate enteric pathogens such as Citrobacter rodentium from
system (87). The consumption of a Westernized diet containing the gut (19). Therefore, the abundance and diversity of micro-
excessive amounts of refined and processed foods, red meats, bial members plays a crucial role in fulfilling these functions,
and sugary beverages, accompanied with a low consumption i.e., symbiosis, colonization resistance, clearance of pathogens,
of fibers, fruits, and vegetables, is associated with the increased etc. Decreased microbial diversity or altered composition,
occurrence of metabolic diseases, such as diabetes and obesity, e.g., increased Firmicutes to Bacteroidetes ratio, entail various
both of which are associated with systemic low-grade inflamma- health risks for the host and are generally associated to poor health
tion attributed to endotoxemia (88, 89) (Figure 2B). A healthy (91). Feeding high-fat and high sucrose diet to wild-type (WT)
gut microbiota maintains a symbiotic relationship within the gut mice leads to decreased gut microbiota diversity and an increase
mucosa offering essential functions in metabolism, immunology, in opportunistic pathogens, resulting in a decreased prevalence
of specific gut barrier-protective bacteria (92). However, revert- and asthma, with increased asthma disease severity, has been
ing the animals to regular chow food reverses the dietary pertur- specifically identified in children. Obesity appears to increase
bations thereby confirming the structural resilience of the gut injury in the lungs of asthmatic patients by increasing eosino-
microbiota. Similarly, the human microbiota can rapidly adapt phil numbers to the airway wall and the systemic production
to dietary changes (93, 94). A comparative analysis of vegan, of pro-inflammatory cytokines TNFα, IL-6, IL-1 (5). A recent
vegetarian, and omnivore diets and the corresponding shifts publication highlighted a pathogenic role for IL-17a produced
in microbial communities revealed that a significant increase from ILC3 cells in airways disease in mice with diet-induced
in β-diversity was present as quickly as 24 h post switch to the obesity. This was accompanied by NLRP3 inflammasome and
animal-based diet (93). These findings suggest that changes IL-17a activation in the adipose tissue and the lungs, leading to
in environmental conditions of gut microbiota, e.g., through the identification of a new inflammasome/Th17 mechanism in
changes in diet, put selective pressure on various species, which asthma (108). Similarly to IBD, a reduction in adiponectin levels
in turn leads to competition for the most adaptable bacteria to has also been reported, which can affect the anti-inflammatory
survive and replicate (7). immune function in asthma patients (5).
Figure 2 | Interaction between diet, microbiota, and immune response at mucosal sites. (A) To keep a healthy state, the local microbiota and mucosal immune
system are in homeostasis at mucosal sites. The microbiota educates and promotes the maturation of the immune system by induction of pro-inflammatory and
anti-inflammatory immune cells, e.g., Th17 (SFB), T regulatory cells (Clostridia spp.), and Th1 (Bacteroides fragilis). Moreover, the immune system surveys microbial
activities (e.g., antigen sampling at the mucosal barrier) and responds in a controlled fashion by producing, e.g., antimicrobial peptides, sIgA to prevent tissue
damage. The integrity of the mucosal barrier is sustained by bacteria-produced metabolites (e.g., SCFA) such as butyrate resulting in high expression of tight-
junction proteins and mucus production, thereby restricting interaction of microbes to the lumen and luminal epitheliums. The diet is involved in all processes,
serving the microbiome with fermentable fibers and the immune system and epithelium with essential nutrients, e.g., vitamins and minerals. (B) During pathological
conditions, such as inflammatory bowel disease and asthma, the homeostasis at the mucosal barrier is disrupted. A westernized diet, i.e., high in SFA, high ω-6/ω-3
ratio, high sucrose and iron (oral iron supplements), and low in fiber promotes inflammation and growth of pathogenic/pathobiont (disease causing) bacteria in the
gut. The microbiota, which is rich in non-beneficial bacteria, favorably induces the maturation of pro-inflammatory immune cells, leading to uncontrolled inflammation
resulting in tissue damage of the mucosal compartment. The damaged mucosa and shifted immune response fail to control the microbiota, which exaggerates the
pathophysiological state. Under certain conditions, bacteria-derived LPS enters the systemic circulation and further stimulates the immune system toward a
pro-inflammatory state. Abbreviations: LPS, lipopolysaccharide; SCFAs, short-chain fatty acids; SFAs, saturated fatty acids; SFB, segmented filamentous bacteria;
sIgA, secretory immunoglobulin A; ω-6/ω-3, omega-6/omega-3 fatty acid ratio; Th, T helper.
for foods with high calories, i.e., from fat and sucrose, has been sucrose, resulted in altered microbiota composition, par-
linked to the worldwide increase in metabolic diseases (119). ticularly in the expansion of and colonization of AIEC and
In IBD, one of the first studies that related sucrose to the devel- reduction in protective bacteria, increased permeability and
opment of CD was published four decades ago (120). More colonic pro-inflammatory mediators (Figure 4), and reduction
recently, the EPIC study uncovered an association between in short-chain fatty acids (SCFA) concentrations and the SCFA
high consumption of sugar and soft drinks accompanied by receptor GPR43, supporting a role of fat and sucrose in exacer-
a low vegetable intake to a higher UC risk (121). Similarly, bating inflammation due to changes in microbial composition
high sugar consumption in sweetened beverages has been (20, 123). Overall, the studies to date indicate that a diet rich in
linked to asthma and dental caries which is also increased saturated fat and high sucrose content presents a risk factor for
in asthmatics (18, 21, 122). Studies in experimental models, IBD and asthma and is associated to inflammation while results
e.g., transgenic CEABAC10 mice fed a diet high in fat and from MUFA-containing diets are contradictory.
Figure 3 | Schematic illustrating the nutrient factors regulating microbial and host responses in the healthy gut and lung. Homeostatic balance at the mucosa due
to a balanced diet rich in fiber allows for regulated interactions between the epithelia and the microbiome. This dialog with the microbiome allows for appropriate
epithelial barrier function, mucus secretion, and underlying immune sensing. In the gut, a balanced microbiome generates SCFAs and dietary long chain FAs and the
fat-soluble vitamins A and D which induce a tolerogenic mucosal immune state locally at the gut but also systemically and particularly in the lung. The gut-derived
SCFAs acetate and propionate enhance DCs, ILC, and macrophage phagocytic function and Tregs balance resulting in the control of lung microbiota and efficient
mucocillary clearance of inhaled microbes and particulates. Lung figure adapted from Ref. (41). Abbreviations: CCR9, C–C motif chemokine receptor 9; DCs,
dendritic cells; DHA, docosahexaenoic acid; EPA, eicosapentaenoic acid; FAs, fatty acids; GC, goblet cells; GPR, G-protein coupled receptor; ILC, innate lymphoid
cells; α4β7, integrin α4β7; IL-1β, interleukin 1 beta; IL-4, interleukin 4; IL-5, interleukin 5; IL-10, interleukin 10; Fe2+, iron; MΦ, macrophage; NFκB, nuclear factor
kappa-light-chain-enhancer of activated B cells; PPARγ, peroxisome proliferator-activated receptor gamma; RA, retinoic acid; SCFAs, short-chain fatty acids; TGFβ,
transforming growth factor beta; Th, T helper; Tregs, T regulatory cells; TLR4, toll-like receptor 4; VitA, vitamin A; VitD, vitamin D; VDR, vitamin D receptor; healthy
bacteria phyla— , bacteroides; , firmicutes ; , barrier integrity.
Dairy Products effect of raw milk has been speculated to be due to improvement
Dairy products are a major source of SFA present in our diets. in nutrition, prevention of lactose intolerance, or the presence
In a Japanese study, an increased incidence of CD was strongly of “good” bacteria. However, the topic is still debatable and more
correlated to milk protein (124). Furthermore, an increase in studies are needed.
cheese consumption is associated with an increased risk of both
UC and CD (125). In line with these findings, IL-10−/− mice fed a Emulsifiers
saturated milk fat-derived diet resulted in an increased severity of Processed foods have been identified as a risk factor for IBD
colitis associated with a colonic Th1 profile and presence of CD4+ (4, 115) and in 2013, it was hypothesized that the increased
IFN-γ+ cells in the mesenteric lymph nodes (MLNs) as a result incidence in CD was the result of a higher consumption of
of blooming of an opportunistic bacteria Bilophila wadsworthia emulsifiers in processed foods (131). Indeed, using an animal
(126) (Figure 4). Interestingly, WT mice fed milk fat diet and model of colitis (IL-10−/− mice), it was shown that exposure
presenting a blooming of B. wadsworthia did not develop colitis, to two common emulsifiers, carboxymethylcellulose (CMC)
indicating the impact of genetic predisposition on the subsequent and polysorbate-80 (P80), aggravated colitis by increasing gut
inflammatory response. Of note, 20% of patients with UC ben- permeability, reducing mucus thickness, promoting higher
efited from excluding milk and cheese from their diet (127). These penetration of intestinal bacteria, and altered microbial compo-
collected data indicate that dairy products may play a role in IBD sition, particularly by enrichment in Bilophila spp. (Figure 4).
pathology. In contrast to IBD, a decreased asthma risk is associated Exacerbation in obesity/metabolic syndrome was also observed
to milk fat (117, 118). Additionally, it has been reported children in emulsifier-treated TLR5−/− mice (132). Supporting these
who consumed raw milk during childhood show a reduced risk findings, it was shown in vitro that addition of P80 induced a
of developing atopy and/or asthma (128–130). The protective higher translocation of E. coli across M-cells (133) (Figure 4).
More recently, using an ex vivo model of human microbiota NUTRIENTS AND THEIR IMPACT ON
culture which was exposed to CMC and P80 revealed an altered MICROBIOTA AND IMMUNE RESPONSES
gene expression and microbial composition by, e.g., induction
of bioactive flagellin. However, transfer of emulsifier-treated
AT MUCOSAL SITES
microbiota to GF mice resulted in a low-grade inflammation Recent evidence has identified the existence of a cross talk between
and metabolic syndrome features but not colitis (134). The rel- the host and the commensal microbiota within the gut. In this
evance of these findings for the development of the microbiota dialog, nutrients play an important role either by directly interact-
and immune system and in other chronic conditions is yet to ing with the host via the epithelium or the intestinal immune system
be addressed. or indirectly, by modulating the composition of the commensal
microbiota which in turn will interact with the immune system, and
vice versa (Figure 2A). The immune system will react promptly and
Fibers, Vegetables, Fruits, and Fish adapt depending on the microbiota (commensal, pathobionts, and
In contrast to dietary fats, diets rich in fish (n-3 PUFAs), fer- pathogens) and the diet (prebiotics, supplements, or detrimental
mentable fibers and vegetables and fruits lower the risk for IBD nutrients). The worldwide increased incidence of IBD and asthma
(4, 115). For example, an inverse correlation of eicosapentaenoic has been hypothesized to be associated with changes in dietary
acid (EPA) and docosapentaenoic acid with colonic cytokine habits, i.e., westernized life style. In the following sections, we will
levels was identified in UC patients (115). Several studies have outline several important macro- and micronutrients associated
suggested that a diet low in refined carbohydrates could be ben- with diets and their effect on immune responses at mucosal sites
eficial in the treatment of CD, reviewed in Ref. (135); however, important in health and in IBD and asthma.
clinical trials are needed for this view to be proven (136, 137).
Findings from a prospective study investigating the long-term
intake of dietary fiber and risk of incidence of CD and UC in
Macronutrients
In this section, we will summarize the impact of the main macro-
women revealed a 40% reduction in risk of developing CD while
nutrients fat, carbohydrates and proteins on immune responses
no association to UC was observed (138). Much of the fiber
and microbiota (Table 1).
intake in this study originated from fruits. The Aryl hydrocarbon
receptor (AhR) was identified as a potential mechanism linking
Fats
the positive effects of fruit-derived fiber. AhR is expressed ubiq-
Under this section, we will outline the impact that saturated-,
uitously in vertebrate cells and mediates the toxicity of xenobiotic
monounsaturated-, and PUFAs have on immune and microbial
molecules by binding to the AhR nuclear translocator and acti-
responses associated with IBD and asthma.
vating dioxin- or xenobiotic-response element sequences (139).
Indole-3-carbinol, a major component of cruciferous vegetables Saturated Fatty Acids
(e.g., broccoli, cabbage, and cauliflower) can activate AhR in the Fatty acids belonging to SFAs and containing 12 or less carbon
intestine (140), inducing maintenance and expansion of intestinal (CX) atoms, include carprylic acid (C8:0), capric acid (C10:0),
intraepithelial lymphocytes (IELs) and IL-22-producing ILCs and lauric acid (C12:0), are found in vegetable oils, cocoa butter,
(141). IELs are T cells expressing αE (CD103)/β7 integrin, local- palm oil. SFAs containing more than 12 carbon atoms include
ized in between epithelial cells and are characterized as either myristic (C14:0), palmitic acid (C16:0), stearic acid (C18:0)
conventional (CD4+ or CD8αβ+TCRαβ+) or unconventional which can be found in lard, butter, beef, pork, chicken fats, eggs,
(TCRγδ+ and CD8αα+TCRαβ+) IELs. IELs have been regarded and vegetable oils (147).
as a first line of defense against infected/damaged epithelial cells Evidence exists that SFA can act as pro-inflammatory media-
and are implicated in the regeneration of the intestinal epithelium tors, e.g., as ligands for TLR4 (148, 149). Potential mechanisms
(142). Indeed, recent studies have demonstrated that IELs play by which SFAs elicit a TLR4-induced inflammatory response
a hitherto underappreciated role in gut epithelial homeostasis have been recently reviewed (12). Briefly, it is proposed that
(143). In addition, aberrant IEL phenotype and lineages are now similar to lipopolysaccharide (LPS), the SFA lauric acid can
evident in pathologies such as celiac disease and enteropathy- trigger TLR4 via CD14/MD2 activation thereby promoting
associated T lymphoma (144). The roles that these differing IELs the expression of the transcription factor NF-κB, which plays
play in sensing and interacting with the gut microbiota in IBD a crucial role in the induction of the pro-inflammatory media-
and other GI conditions warrants further investigation. tors COX2, TNFα, IL-1β, IL-6, CXCL8, IL-12, and IFNγ (150)
In asthma, a high-fiber intake in late pregnant mothers was (Figure 4). Comparably, palmitic acid and stearate can induce
correlated with high serum acetate levels and resulted in lower pro-inflammatory cytokine production from macrophages via
infant GP visits for cough or wheeze (17). In children aged 10–14, degranulation of Ikappa B alpha (IκBα) and phosphorylation of
a dietary intake of fruits, but not vegetables, was negatively related c-Jun N-terminal kinases, MAPKs, and extracellular signal-regu-
to wheeze, while no protective effect was identified in adults (27). lated kinases (ERK). LPS together with palmitate activate reactive
Intake of oily fish, such as salmon, sardines, herring, tuna, oxygen species and the NLRP3 inflammasome leading to IL-1β
and mackerel, which are rich in n-3 FAs, have shown a potential maturation in macrophages (95) (Figure 4). Moreover, a high
benefit in preventing asthma in children and in patients with intake of SFA, i.e., given in western diet, leads to the modification
UC, while no beneficial effect in adults with asthma was reported of the gut microbiota raising the proportion of Gram-negative
(145, 146). bacteria and thereby the natural ligand for TLR4, LPS, as well as
Figure 4 | Schematic illustrating the nutrient factors affecting microbial and host responses in the inflamed gut in inflammatory bowel disease (IBD) and the lung in
Asthma. In both IBD and Asthma genetic susceptibility, microbiota, and dietary changes result in disease development and inflammation. Dysfunctional epithelia
barrier function allows for malabsorption of nutrients, inappropriate immune sampling, and colonization of the gut by pathobionts and subsequent disease
exacerbation. In the lung, environmental triggers stimulate inflammatory and allergic reactions resulting in mucus hypersecretion, epithelia, and tissue remodeling
and resulting compromised of lung function. This microenvironment change allows for microbial changes which allow for increased respiratory infections in asthmatic
patients. Both IBD and asthma pathogenesis is related to reduced microbiota-derived SCFAs, malabsorption of iron and Vitamins and reduced gut-derived SCFA
result in a trend toward an inflammatory sensing of the mucosa associated microbiota. A diet high in SFA increases TLR4 sensing and subsequent inflammatory
reactions to the microbiota resulting in disease progression. This dysregulated mucosal inflammation changes the epithelia barrier function and subsequently alters
the microbiota of both sensitive immune sites displaying the characteristic phenotypes associated with both IBD and asthma. Lung figure adapted from Ref. (41).
Abbreviations: CXCL8, C–X–C Motif Chemokine Ligand 8; DCs, dendritic cells; Emul, emulsifier; GC, goblet cell; HF/HS, high-fat/high sucrose; ILC, innate lymphoid
cells; IL-1β, interleukin 1 beta; IL-6, interleukin 6; IL-12, interleukin 12; Fe2+, iron; LPS, lipopolysaccharide; MΦ, macrophage; MAPK/ERK, mitogen-activated protein
kinase/extracellular signal-regulated kinase; MF, milk fat diet; N, neutrophils; NFκB, nuclear factor kappa-light-chain-enhancer of activated B cells; PPARγ,
peroxisome proliferator-activated receptor gamma; SFA, saturated fatty acids; SCFAs, short-chain fatty acids; TGFβ, transforming growth factor beta; Th, T helper;
TNF, tumor necrosis factor; TLR4, toll-like receptor 4; VitA, vitamin A; VitD, vitamin D; healthy bacteria phyla— , bacteroides; , firmicutes; , adherent and
invasive Escherichia coli (AIEC); , Bilophila wadsworthia; , other altered bacterial spp; , compromised barrier integrity.
an increase in intestinal permeability, which in itself induces a propria but without the development of obesity or obesity-
state of metabolic endotoxemia (151) (Figure 4). Intake of SFAs associated metabolic features (154). Similarly, Mdr1a−/− mice
can also increase plasma low-density lipoprotein cholesterol by fed a lard-based high-fat diet for 12 weeks led to an exacerbation
inducing the formation of low-density Lipoprotein (LDL) and of spontaneous colitis associated with elongated crypts, loss
by reducing LDL turn-over leading to the generation of oxidized of goblet cells, and infiltration of immune cells. Contrary to
LDL (152). Both oxidized LDL and phospholipids are damage- TNFΔARE mice, Mdr1a−/− mice developed obesity as character-
associated molecular patterns, which are also recognized by TLR4 ized by increased adiposity and presence of foamy macrophages,
and can trigger a CD36–TLR4–TLR6-mediated inflammatory while WT mice did not (155). Rats fed a diet containing capric
response (153). These findings support a pro-inflammatory effect and lauric acid followed by DSS-induced colitis, developed worse
of SFA on the microbiota on innate responses in macrophages. colitis associated with a higher colonic myeloperoxidase activity
To date, mechanistic studies on saturated fats in human IBD and a pro-inflammatory cytokine profile as well as a reduction
are scarce and, therefore, much of our knowledge on SFAs and in goblet cells (156). Overall, these findings indicate that the
intestinal inflammation emanates from studies in experimental type of SFA diet, microbiota status, diet regimen, and/or the
models. TNFΔARE mice fed a palm oil-based high-fat diet for genetic background of the animals determine the development
up to 12 weeks resulted in an initial acceleration of ileitis fol- of intestinal inflammation and obesity, suggesting that different
lowed by worsening of proximal colitis associated with loss of dietary-induced mechanisms regulate these two conditions. In
TJ protein occludin in the distal ileum, endotoxin translocation, asthma, SFA have been shown to effect symptoms and immune
and increased infiltration of DCs and Th17 cells into the lamina activation, e.g., by inducing a neutrophilic inflammation and
suppressing bronchodilator recovery in asthmatic patients (11, role, e.g., it is produced by macrophages and neutrophils as a
27) (Figure 4). response to inflammatory stimuli and (2) a regulatory role, by
inducing immune tolerance, independent of IL-10 or Tregs (167).
Monounsaturated Fatty Acids and Derived Oils n-3 PUFAs are primarily sourced from the human diet, with
Monounsaturated fatty acids, including palmitoleic acid (C16:1) DHA and EPA especially sourced from fish (e.g., salmon) and
and oleic acid (C18:1, OA) are normally found in macadamia ALA from seed oils (e.g., walnut, linseed oil). Several reports
nuts, blue-green algae, olive oil, canola oil, beef tallow, lard, and have highlighted their effect in preventing and/or treatment of
avocado. different inflammatory diseases in animals and humans, includ-
Diets rich in MUFA appear to reduce LDL cholesterol and ing IBD and asthma. n-3 PUFAs can inhibit TLR4 signaling
potentially increase high-density lipoprotein (HDL) cholesterol and the subsequent gene transcription of pro-inflammatory
(157), and palmitoleate treatment of M1 macrophages induce an mediators (168), a process partly mediated via GPR120 (169)
anti-inflammatory M2 profile (158) indicating an anti-inflamma- (Figure 3). n-3 PUFAs can also activate the anti-inflammatory
tory capacity of MUFAs. The specific role of MUFAs in IBD and transcription factor PPAR-γ and inhibit NF-κB (Figure 3) and
asthma remains inconclusive. For example, a prospective study the subsequent pro-inflammatory cytokine production includ-
by de Silva and colleagues showed that a dietary oleic acid was ing TNFα, activity which is highly expressed in the mucosa of
inversely associated with UC development (114), while palmi- patients with IBD and asthma subtypes (13, 170, 171). DHA
toleic and oleic acid treatment of polarized intestinal epithelial can also improve epithelial barrier integrity by increasing the
cells impaired epithelial barrier function (159). MUFA and oleic expression of the TJ proteins occludin and claudin-1 (Figure 3)
acid intake indicated an increased risk of wheeze and non-atopic (172), as the integrity of the epithelium is critical in prevent-
asthma, respectively (160). Extra virgin olive oil, high in MUFAs, ing paracellular translocation of LPS into systemic circulation.
contains highly bioactive components which are present in the PUFA supplementation in particular those related to fish oils can
unsaponifiable fraction (UF). Beneficial effects of UF and olive oil also modulate asthma symptoms (173, 174), with reduction of
were demonstrated in an acute DSS model of colitis and in mice with wheeze, improved pulmonary function and reduced pro-inflam-
C. rodentium induced colitis. Disease amelioration included matory mediators in sputum. However, it should be noted that
alleviation of oxidative stress, reduction of pro-inflammatory many trials are designed to measure different outcomes which
proteins and increased levels of intestinal alkaline phosphatase, can deliver conflicting findings (27, 175). With the evolution in
which can de-phosphorylate bacterial LPS (161–163). In line food technology and modern agriculture in the last 100 years the
with these findings, isolated blood and intestinal T cells from amount of n-6 FAs, e.g., LA present in our food has increased
UC patients treated with UF resulted in a reduction in T cell due to change in animal feed from grass to grains (176, 177).
activation, β7 integrin expression and IFNγ production as well as Therefore, the ratio of n-6/n-3 PUFAs present in the food has
induction of apoptosis (164). Findings from these studies indicate changed from 1:1 to 16:1 in USA/Europe (177) (Figure 2). This
MUFA exert both pro- and anti-inflammatory activities in these ratio has increased dramatically due to food processing, less fish
mucosal conditions. and fiber consumption, and the dietary habits of farm animals. In
line with this, a recent study identified a higher ARA:EPA ratio in
Polyunsaturated Fatty Acids the inflamed mucosa of UC patients, which correlated with the
Polyunsaturated fatty acids are FAs containing more than one severity of the disease (178). No association of n-6/n-3 ratio was
double carbon bonds. Therefore, naturally, they are more prone found in individuals with hay fever or allergic sensitization (179).
to oxidation and oxidized LDL synthesis. Long chain PUFAs The relevance of a lack of n-6/n-3 ratio association to asthma is
are divided in two main groups; omega-3 (n-3) PUFAs includ- still to be uncovered.
ing—alpha-linolenic acid (ALA, C18:2), docosahexaenoic acid
(DHA, C22:6), and EPA (C20:5); and omega-6 (n-6) PUFAs Carbohydrates
including—linoleic acid (LA, C18:3), and arachidonic acid (ARA, Carbohydrates are divided into four groups: monosaccharides,
20:4). LA and ALA are referred to as essential FAs as they are disaccharides, oligosaccharides and polysaccharides. Generally,
the precursors of ARA, EPA, and DHA. DHA and EPA compete monosaccharides and disaccharides are referred to as sugar. In
for the enzymes and products of ARA metabolism whereby they the western diet a large amount of calories are ingested in form
can antagonize the formation of inflammation related eicosanoid of refined carbohydrates, i.e., sucrose, starch, fructose syrup,
mediators (165, 166). etc.—obtained from soft drinks, pastries and desserts, and white
Arachidonic acid is the primary n-6 PUFA found in inflam- bread. This energy-dense but nutrient-poor diet is a risk factor
matory cells and is important for the production of inflammatory for obesity, type 2 diabetes, cardiovascular diseases and more.
eicosanoids. ARA is formed out of LA which is further converted Therefore, the biological plausibility exists that it has impact on
to prostaglandins [e.g., prostaglandin E2 (PGE2), leukotrienes intestinal inflammation and asthma. Moreover, related to the
(LTBD4), and other lipoxygenase or cyclooxygenase products carbohydrates that can be metabolically used by gut microbes, the
(COX1/-2)], the so-called eicosanoids, all of which have pro- and term “microbiota-accessible carbohydrate” has been proposed.
anti-inflammatory in addition to atherogenic and pro-thrombotic The term refers to the ability of microbial carbohydrates to modify
effects (166). PGE2 is one of the key prostaglandins produced in the composition of the microbiota, and dictate the functionality
the intestine where it has dual functions: (1) a pro-inflammatory and metabolic output (180).
Fibers and acetate and propionate to GPR43 also supported the reduc-
Dietary fiber is a plant-based nutrient and a type of carbohy- tion in airway inflammation (17, 23, 194, 195). These microbiota-
drate, which due to its biochemical structures resists digestion mediated effects are also linked to maternal influences of asthma
by intestinal and pancreatic enzymes in the human GI tract. risk, where high serum acetate concentrations, but not propion-
Therefore, the fiber passes through the GI tract relatively intact. ate, in pregnant mothers’ correlate with reduced asthma risk and
Fermentable carbohydrate substrates such as non-starch polysac- are thought to modulate Tregs biology of the fetus (17). These
charides, resistant starch and oligosaccharides serve as important findings clearly point to a crosstalk between the gut and the lung
substrates for the gut microbiota. The microbes located in the via SCFA in asthma and between the microbiota and the intestine
human colon use fermentation to produce SCFAs, lactate and in IBD, indicating that treatments aiming to increase SCFAs and
gas (181). These fermentation products selectively promote the SCFA producing bacteria should be further investigated.
growth of beneficial Bifidobacteria and Lactobacilli and exert
anti-inflammatory (inhibition of NFκB transcription via GPR41) Proteins
and anti-carcinogenic functions (182). Proteins consist of carbon, hydrogen, oxygen, and nitrogen
elements. They are essential nutrients and are involved in virtu-
Short-Chain Fatty Acids. Upon fermentation of dietary fiber, ally all physiological functions. High protein intake, especially
bacterial metabolites such as SCFAs are produced in the colon. animal derived protein, is associated with an increased risk of
SCFA mediates the communication between the commensal CD (196). In asthmatics high ingestion of cured meats is linked
microbiota and the immune system affecting the balance between with worsening of symptoms (24).
pro- and anti-inflammatory responses. The beneficial effects of Carnitine is an amino acid derivative synthesized primar-
butyrate on colonic health are particularly well established (183). ily in the liver and kidneys from lysine and methionine and
Microbial-derived butyrate and to a lesser extent acetate and is involved in lipid metabolism in eukaryotic cells (197). In
propionate, can facilitate the generation of extrathymic Foxp3+ humans, the main source of carnitine is red meat. Humans
Tregs which are crucial for limiting intestinal inflammation with an omnivorous diet following ingestion of l-carnitine,
(17, 184, 185) (Figure 3). Presence of butyrate during human DC presented increased plasma trimethylamine-N-oxide (TMAO)
maturation results in a tolerogenic phenotype, with an increased levels, which was dependent on microbiota mechanisms, when
expression of IL-10 (186, 187), further supporting the regulatory compared to vegans or vegetarians. Elevated plasma levels of
potential of SCFAs (Figure 3). SCFA can signal through the acti- TMAO are positively correlated with an increased risk for major
vation of specific G-protein coupled receptors (GPCRs), GPR41, adverse cardiovascular events (198). However, TMAO has also
GPR43, GPR109, which are particularly expressed in immune protective functions, e.g., by protecting cells from osmotic and
cells, e.g., polymorphonuclear leukocytes, and are suggested to hydrostatic damage, and therefore, is essential for all organisms
participate in immune surveillance of the colonic mucosa affect- (25). A recent study proposed plasma TMAO as a non-invasive
ing the balance between pro- and anti-inflammatory responses biomarker for IBD, as decreased TMAO levels are seen in these
(188). GPR43 is additionally expressed in the colonic epithe- patients (199). The reduced TMAO was likely related to altera-
lium where it can mediate SCFA-regulated effects on epithelial tions in the gut microbiome (the abundance of anaerobes or fac-
barrier and proliferation (Figure 3). Butyrate is also a natural ultative anaerobes) in these patients. TMAO is also an oxidation
ligand for PPARγ, which is expressed in colonic epithelial cells, product of trimethylamine which can be found in seafood, fish,
macrophages and lymphocytes. Colonic PPARγ expression is etc. Consumption of TMAO-containing food, e.g., oily fish can
linked to host–microbe interactions with natural (e.g., SCFA— lead to accumulation of TMAO and protection against asthma
butyrate, conjugated LA) and synthetic (e.g., 5-aminosalicylic in childhood (200). Further studies are necessary to shed a light
acid) PPARγ ligands preventing inflammation in experimental into mechanisms that underlie the association of high intake of
colitis (189). animal protein in these conditions.
A significant decrease in the number of butyrate-producing Other studies have also shown how dietary peptides and amino
bacteria including Eubacterium rectale/Roseburia spp (which acids can modulate intestinal immune functions and influence
belong to Clostridium coccoides) and F. prausnitzii (which belong inflammatory responses. Supplementation with the dipeptide
to Clostridium leptum cluster), both within the Firmicutes phylum alanine-glutamine, led to decreased expression of inflammatory
was revealed in patients with UC and CD (190, 191). In patients mediators and increased expression of mucin 2 (MUC2) promot-
with UC, colonic irrigation with butyrate is able to limit inflam- ing mucosal recovery in the DSS-induced colitis mouse model
mation and in experimental models it ameliorates inflammation (26). Moreover, lower serum tryptophan is associated to active
and modifies microbial composition (183, 192). CD (201). Supplementing tryptophan presented beneficial effects
In the Canadian CHILD study the SCFA acetate was reduced in the DSS-induced porcine IBD model, by inducing T cell apop-
in the feces of infants with atopy and wheeze (83). Feeding a tosis and thereby inhibiting Th1-mediated immune responses
high fiber diet to mice has resulted in gut microbiota alteration and subsequently reducing inflammation (202).
which concomitantly leads to increased serum SCFAs, such as
acetate and propionate, and alleviates allergic asthma symptoms Micronutrients
(Figure 3). These SCFAs induced an enhanced DC and mac- Micronutrients or trace elements are nutrients required by
rophage phagocytosis and reduced Th2 responses in the murine organisms in small quantities to maintain a variety of physi-
lung (17, 23, 193). Furthermore, binding of propionate to GPR41 ological functions and as most of them are essential, they need
to be obtained from the diet. These minerals include iron, cobalt, shift in Th17/Tregs in UC biopsies (216–218). VitA appears to
chromium, copper, iodine, manganese, selenium (Se), zinc (Zn) influence the microbiota, as its deficiency seems to favor a non-
and vitamins include Vitamin A (VitA), vitamin B1 (VitB1), symptomatic reservoir of E. coli-like enteric infections (219, 220).
Vitamin B6 (VitB6), Vitamin B9 (VitB9), VitB12, VitD, and VitK.
Micronutrient deficiencies impair immune function and increase Vitamin D
the severity of disease (203). Micronutrient deficiencies occur in Vitamin D belongs to a group of fat-soluble vitamins which are
more than half of patients with IBD, with CD patients present- essential for bone mineralization and optimal intestinal absorp-
ing more deficiencies than UC patients, with the most common tion of calcium, iron, magnesium, phosphate, and Zn. VitD
being VitB1, VitB6, VitB12, VitD, VitK, iron, folic acid, Se, and Zn regulates the epithelial integrity/barrier function and is involved
(204). This deficiency in vitamins (hypovitaminosis) is thought in the detoxification and protection against infection as well as in
to be the result of malabsorption, altered microbial composition controlling of the commensal microbiota (221, 222).
and impaired host mucosal system. Low dietary intakes of VitA Vitamin D can be obtained from the diet or by dermal synthesis,
and VitC are associated with asthmatics (205). The next section e.g., in the skin where it is produced from 7-dehydrocholesterol
summarizes the most relevant micronutrients in relation to IBD (222). 1,25-dihydroxyvitamin D3 [1,25(OH)2D3] is the active
and asthma pathology. form, which arises from the bloodstream (endocrine action) or
it can be locally produced from circulating 25(OH)D3 within
Vitamins intestinal cells (intracrine, autocrine and paracrine action). VitD
Vitamins can be absorbed from the diet but the gut commensal is ubiquitously expressed in several human tissues including
microbiota play an important role in their production and bio- immune cells, but its expression is higher in intestinal epithelial
availability (206). Indeed, the diet of germ-free mice requires cells (223). VitD deficiency has been associated with a greater dis-
supplementation with dietary VitK and B Vitamins to maintain a ease activity and extended disease duration in patients with IBD
normal function (207). (Figure 4) – why a supplementation of VitD is often required.
Human polymorphisms in the vitamin D receptor (VDR) are
Vitamin A also associated with susceptibility to IBD (224). Vitamin D3 has
Vitamin A is a group of unsaturated organic compounds been linked to beneficial effects in asthma; however, the benefits
including retinol, retinoic acid (RA), and several pro-vitamin A are mainly observed in children or via maternal supplementa-
carotenoids including beta-carotene. They are fat-soluble sub- tion (225). Recent studies have identified that VitD3 appear to
stances obtained from animal food sources. RA is a metabolite modify VEGF function and reduction in airway smooth muscle
of VitA, which is produced by CD103+ DCs and epithelial cells proliferation (226).
and acts as ligand for RA receptors (RARs) and Retinoic-X- The biological actions of 1,25(OH)2D3 are mediated via
Receptor (RXR), transcription factors regulating gene expres- the VDR, which acts as an heterodimer with RXR to acti-
sion. Lymphoid cells express RAR, RXRs and RA, which are vate VitD target genes (222, 227). VDR targeted pathways
known to regulate IgA and mucosal homeostasis (Figure 3). regulating inflammatory responses include TLR and NF-κB
DCs from Gut associated lymphoid tissue (GALT) produce RA signaling, Th17/Tregs response, apoptosis, cell proliferation
to sustain gut tropism and in synergy with GALT-DC-derived and differentiation, barrier function, etc. (Figure 3). The anti-
IL-6 or IL-5, induce IgA production (208) (Figure 3). RA can inflammatory role of 1,25(OH)2D3 is based on the suppressive
also control the presence of RORγ+ ILCs, the formation of effect of NFκB activity, as NFκB-induced pathways are enhanced
lymphoid tissue in the small intestine (209) and appears to be a in VDR−/− mice exposed to bacterial and chemically induced
cofactor in IgA class switch recombination (210). RA can also colitis (228). 1,25(OH)2D3 regulates intestinal barrier trough
induce the gut-homing capacity on T cells by the up-regulation the up-regulation of TJ proteins including occludin, ZO1, clau-
of the integrin α4β7 and the chemokine receptor CCR9 (211) din 2 and E-cadherin (221, 229) (Figure 3). In support of this,
(Figure 3). A diet deficient in VitA can lead to a systemic pro- IL-10−/− mice expressing the human VDR in intestinal epithelial
inflammatory state, due to a lack of homing integrins in MLN cells resulted in a reduced development of spontaneous colitis
activated T- and B-cells, which then go into systemic circulation (230). VitD has been linked to the modulation and control of
instead of migrating back to the gut (212, 213). RA, together the gut commensal microbial composition (Figure 3), since
with TGFβ, promotes naïve CD4+ to become Foxp3+ Tregs and VDR−/− mice present an altered microbiota with more abundance
RA alone has also inhibitory effects on Th17 cell differentiation on Bacteroidetes and Proteobacteria phyla and less abundance on
(Figure 3). Others have also described that VitA can impair the Firmicutes phyla (231). Interestingly, analysis of mice treated
the reprogramming of Tregs into IL-17-producing cells during with 1,25(OH)2D3 revealed an increased C. rodentium load in
intestinal inflammation (214). the colon and spleen doubtless due to the suppression of a Th17
Patients with IBD have been reported to be deficient in VitA response, which is essential for C. rodentium clearance (232). In
(Figure 4). Cytochrome P450 26 B1 (CYP26B1) participates in contrast, following infection with C. rodentium, a diet deficient
the degradation of RA, and homozygous carriers of the CYP26B1 in VitD aggravated barrier function, microbiota composition and
polymorphism rs2241057 have been associated as risk factor of inflammation (233). Correspondingly, mice fed a high-fat and
CD development, linking an elevated catabolic function of RA to VitD deficient diet presented increased ileal antimicrobial pep-
IBD (215). Supplementation of VitA/RA seems to attenuate intes- tides and Helicobacter hepaticus and reductions in TJ proteins,
tinal inflammation in experimental models and even induces a MUC2 expression and abundance of beneficial bacteria such as
Intestine Lung
Macronutrients
Fat
High-fat diet Permeability of epithelial barrier ↑ Neutrophil ↑ (10, 11)
TLR4 mRNA ↑
Saturated fatty acids Activation of TLR4↑ (12)
n-3 PUFA, e.g., EPA, DHA PG (series-3) ↑ Maternal supplementation reduced Childhood (13)
LK (series-5) ↑ Asthma (14, 15)
Leukocyte chemotaxis↓ IL-13 cord blood ↓
IL-1β ↓, TNFα ↓ Mucus (murine) ↓
Resolvins, Maresins and Protectins↑ CD45+ inflammatory cell infiltrates (murine) ↓
n-6 PUFA, e.g., ARA, linoleic acid PG (series-2) ↑ Mucus (murine) ↓ (14)
LK (series-4) ↑
SCFA, e.g., butyrate Energy source for colonocytes Allergy ↓ (16, 17)
Barrier function ↑
Peroxisome proliferator-activated receptor γ activation ↑
Carbohydrates
Sucrose Permeability of epithelial barrier ↑ Asthma and dental caries ↑ (18–21)
Fermentable carbohydrates, e.g., fiber Butyrate production ↑ SCFA levels ↑ (22, 23)
DC maturation ↓
TH2 response ↓
Proteins
Animal-derived proteins, e.g., carnitine TMAO synthesis ↑ Asthma exacerbation (cured meats) (24, 25)
Dipeptides, e.g., alanine–glutamine Mucin 2 expression ↑ (26)
Micronutrients
Vitamins
Vitamin A, e.g., RA, β-carrotene Induction of tolerogenic DC and Tregs ↑ (27, 28)
Vitamin D Cathelicidin production Maternal supplementation reduction in airway (27, 29, 30)
Innate defense toward regulatory state ↑ smooth muscle
Ca2+absorption ↑
Vitamin B, e.g., thiamine, folate, Vitamin B9 deficiency—colonic Foxp3 + Tregs ↓ Folate deficiency—asthma exacerbations (31–34)
cobalamine, pyridoxine
Minerals
Iron Incorporated into iron–sulfur clusters, redox cofactors, or used (35, 36)
metalloenzymes
ARA, arachidonic acid; Ca2+, calcium; DHA, docosahexaenoic acid; EPA, eicosapentaenoic acid; Foxp3, Forkhead-Box-Protein P3; LK, leukotrienes; PG, prostaglandins; RA, retinoic
acid; TLR4, toll-like receptor 4; PUFA, polyunsaturated fatty acid; SCFA, short-chain fatty acid; Tregs, T regulatory cells; CD, cluster of differentiation; DC, dendritic cells;
IL, interleukin; TMAO, trimethylamine-N-oxide; Th, T helper cell.
Akkermansia muciniphila developing insulin resistance and fatty highlight a potential link between the microbiota, glucose, FAs
liver (234). and mucosal alteration leading to intestinal inflammation in
which thiamine may have a key role.
Other Vitamins
Vitamin B1. Vitamin B1, also known as thiamine, is mainly Vitamin B6. Vitamin B6 is a water-soluble vitamin, also known
obtained from whole grains, trout, pork, peas and beans. It has as pyridoxine, which exists in several forms and can be obtained
an important role in the catabolism of sugars and amino acids from fruits, vegetables, grains, fish and meat. The biologically
(235, 236). Thiamine is a component of the pyruvate dehydroge- active form of VitB6, Pyridoxal 5′-phosphate (PLP), is involved
nase that catalyzes the formation of Acetyl CoA in FA synthesis, in the synthesis or metabolism of proteins, lipids and carbohy-
a pathway which is altered in IBD (237). Pediatric IBD patients drates and important in the modulation of immune pathways
presented alterations in cellular transport of thiamine and FAs (236, 239). PLP treatment ameliorated colitis in IL-10−/− mice due
synthesis (e.g., bile acids) (237, 238). Altogether, these findings to a reduction in colonic TNFα, IL-6, IFNγ, COX-2 and nitric oxide
synthase (iNOS) expression and modulation of the chemotactic spp. and Clostridium cluster IV, leading to an increased butyrate
lipid S1P (240). Eubacterium rectale, a dominant non-pathogenic concentration in the gut, without the induction of colitis. Thus,
fecal Gram-positive commensal bacterium has been described as iron supplementation can increase the proportion of beneficial
one of the important bacterial group synthetizing PLP (241). gut microbiota metabolites which may contribute to gut health
in IBD individuals (256).
Vitamin B9. Vitamin B9 is a water-soluble vitamin also known
as folic acid or folate, it can be obtained from vegetables and fruits Other Minerals
and is important in DNA repair and methylation aiding rapid Selenium. Selenium is an essential antioxidant trace min-
cell division and growth during, e.g., infancy and pregnancy. eral which can be obtained from proteins or vegetables and is
Bacteria linked to folate biosynthesis include Bifidobacteria and used in the body to synthetize the amino acid selenocysteine
Lactobacilli groups (207, 242, 243). A deficiency in VitB9 has been (selenoproteins). Large amounts of Se can cause toxicity (257).
more commonly ascribed to CD, especially in patients with ileal The amino acid transporters SLC3A1 and SLC1A4 have been
disease, than in UC patients (244, 245). VitB9 deficiency has also suggested as Se transporters (236) and play a role in intesti-
been associated with a reduction in colonic Foxp3+ Tregs (33). nal epithelial permeability and barrier function. Lower Se
Folate supplementation during pregnancy has been linked to serum levels have been described in children with IBD (258).
an increased risk of infants developing asthma; however, this is A possible therapeutic role for Se has been described, whereby
controversial with conflicting studies providing little clear evi- macrophage derived selenoproteins enhance 15-hydroxypros-
dence (34, 246). taglandine dehydrogenase (15-PGDH) and protects mice from
DSS-induced colitis (259). A potential role for Se in asthma
Vitamin K. Vitamin K is a fat-soluble vitamin required for pathogenesis has been hypothesized; however, data from
synthesis of certain proteins involved in blood coagulation human studies are conflicting. A benefit of Se supplementation
and is linked to calcium pathways and calcification. VitK1 can in animal asthma studies has been identified by regulating Th
be obtained from, e.g., meats, cheeses, and eggs or synthetized cell differentiation (260).
(VitK2) by the microbiota of the colon de novo or from VitK1
(236, 247, 248). VitK deficiency has been associated with both Zinc. Zinc is an important mineral involved in wound repair,
adult and pediatric CD patients (249). A protective role of VitK tissue regeneration, and the immune response. Low serum Zn
was shown in a model of DSS-induced colitis associated with a levels have been reported in children with IBD (258). Zn given
reduction in IL-6 production from B cells (250). orally to CD patients restored intestinal permeability by modu-
lating TJ proteins in both the small intestine and colon (261). Zn
Minerals concentrations have been found to be significantly lower in asth-
Minerals are obtained from the diet, and are essential nutrients matic patients and it is thought to result in reduced antioxidant
needed by organisms for synthesis of common organic molecules. function increasing asthma risk (262).
Accordingly, mineral deficiencies in the westernized diet have a
major impact on host health. The next section summarizes the CONCLUSION AND FUTURE
most relevant minerals in relation to IBD and asthma pathology. PERSPECTIVES
Iron The increasing incidence of IBD and asthma implies a funda-
Anemia is one of the most common extra-intestinal manifesta- mental role for environmental factors. Several lines of evidence
tions of IBD. The deficiency, results from either an absolute state, have identified the consumption of a diet high in saturated fat
i.e., poor dietary intake of iron, reduced iron absorption, and/ and high in sucrose increases a person’s risk for several chronic
or increased blood loss from chronically inflamed intestinal conditions including IBD and asthma (4, 27). Studies in animal
mucosa; and/or functional iron state, i.e., deficiency in VitB12 models, human cells and tissues for both conditions have also
and insufficient availability of iron for incorporation into eryth- highlighted the complex interaction between diet, the micro-
roid precursors despite normal or increased body iron stores biota and the host response, especially regarding aggravation of
(251, 252). It is estimated that up to 80% of patients with IBD inflammatory responses upon consumption of certain trigger-
present with anemia (253). Consequently, oral or i.v. iron supple- ing nutrients and diets. Dietary ingredients in the western diet
mentation is important in treatment of IBD patients. Nonetheless, are not limited to the ones listed in this review. Apart from the
caution is needed as non-absorbed iron can be toxic to intestinal increased consumption of saturated fats and sucrose, there is an
epithelial cells, since it can stimulate growth and virulence of increased consumption of food additives including sweeteners,
bacteria and appears to worsen disease activity in the patients emulsifiers, thickeners, preservatives and food colorings. Some
(178, 254). In support of this data, rats supplemented with iron of these have already been associated to asthma and IBD (122,
and exposed to DSS-induced colitis revealed an increased neu- 132, 134) but there is a need for larger epidemiological stud-
trophil infiltration, TNFα and IL-1 expression and NF-κB acti- ies to identify the effect that these products may have on both
vation – all of which could be prevented by supplementing the microbial composition and host responses (e.g., inflammation,
diet with VitE (dl-alpha-tocopherol acetate) (255). In contrast, metabolism, etc.) in disease development. In an elegant study by
rats with humanized gut microbiota and fed dietary iron sup- Arrieta and colleagues it was shown that changes in the micro-
plementation exhibited an increased abundance of Bacteroides biota especially at a young age, can help to identify a window of
opportunity for bacteria colonization and an appropriate immune metabolomics or systems biology, will reveal a clearer picture of
education in asthmatics (83). Future studies aiming to identify the components contributing to the pathology of these chronic
whether a similar new widow also exists prior to IBD onset inflammatory conditions. Furthermore, the impact of current
and whether the education of the mucosal immune system can therapeutic treatments and diets on disease progression and
be regulated by specific diet combinations and corresponding the microbiota are currently lacking, therefore, future studies
microbial alterations are required. Such studies would likely addressing this question could lead to new beneficial treatment
lead to the discovery of new target/regulatory mechanisms for strategies.
both conditions.
Individuals with IBD and asthma present with a deficiency in AUTHOR CONTRIBUTIONS
certain essential nutrients, e.g., VitD and Iron. The reductionist
approach undertaken whereby the effect of one single nutrient DS, MA, and JM wrote the manuscript; SM wrote and edited the
has been investigated in animal models, has so far provided major manuscript. All authors read and approved the final manuscript.
insights into the regulatory mechanisms associated with inflam-
mation and metabolism. In addition, the majority of animal ACKNOWLEDGMENTS
studies and diets, employs standard “high fat” diet containing a
nutrient content designed to sustain rodent health and are, there- The APC Microbiome Institute and the authors in this publication
fore, not relevant to a human and patient dietary intake (263). receive financial support from Science Foundation Ireland (SFI)
Studies using more complex systems, e.g., humanized mice (con- under Grant Number SFI/12/RC/2273. The authors would like
sisting of both a human immune system and human microbiota) to acknowledge SERVIER INTERNATIONAL for the Servier
fed with a humanized diet will provide a deeper mechanistic medical art illustrations that were used in this review. The authors
understanding of the complex diet-microbiota-host network. want to thanks Dr. Amanda Lohan for proofreading of the
Integration of various approaches, e.g., genomic, transcriptomic, manuscript.
REFERENCES 14. Hall JA, Hartman J, Skinner MM, Schwindt AR, Fischer KA, Vorachek WR,
et al. Dietary enrichment with 20% fish oil decreases mucus production
1. El-Gabalawy H, Guenther LC, Bernstein CN. Epidemiology of immune-me- and the inflammatory response in mice with ovalbumin-induced allergic
diated inflammatory diseases: incidence, prevalence, natural history, and lung inflammation. PLoS One (2016) 11:e0163819. doi:10.1371/journal.
comorbidities. J Rheumatol Suppl (2010) 85:2–10. doi:10.3899/jrheum.091461 pone.0163819
2. Molodecky NA I, Soon S, Rabi DM, Ghali WA, Ferris M, Chernoff G, et al. 15. Klemens CM, Berman DR, Mozurkewich EL. The effect of perina-
Increasing incidence and prevalence of the inflammatory bowel diseases tal omega-3 fatty acid supplementation on inflammatory markers
with time, based on systematic review. Gastroenterology (2012) 142:46–54. and allergic diseases: a systematic review. BJOG (2011) 118:916–25.
e42; quiz e30. doi:10.1053/j.gastro.2011.10.001 doi:10.1111/j.1471-0528.2010.02846.x
3. Kotlyar DS, Shum M, Hsieh J, Blonski W, Greenwald DA. Non-pulmonary 16. Engel MA, Neurath MF. New pathophysiological insights and mod-
allergic diseases and inflammatory bowel disease: a qualitative review. World ern treatment of IBD. J Gastroenterol (2010) 45:571–83. doi:10.1007/
J Gastroenterol (2014) 20:11023–32. doi:10.3748/wjg.v20.i32.11023 s00535-010-0219-3
4. Uranga JA, Lopez-Miranda V, Lombo F, Abalo R. Food, nutrients and nutra- 17. Thorburn AN, McKenzie CI, Shen S, Stanley D, Macia L, Mason LJ, et al.
ceuticals affecting the course of inflammatory bowel disease. Pharmacol Rep Evidence that asthma is a developmental origin disease influenced by mater-
(2016) 68:816–26. doi:10.1016/j.pharep.2016.05.002 nal diet and bacterial metabolites. Nat Commun (2015) 6:7320. doi:10.1038/
5. Leiria LO, Martins MA, Saad MJ. Obesity and asthma: beyond T(H)2 inflam- ncomms8320
mation. Metabolism (2015) 64:172–81. doi:10.1016/j.metabol.2014.10.002 18. Alavaikko S, Jaakkola MS, Tjaderhane L, Jaakkola JJ. Asthma and caries: a
6. Park Y, Subar AF, Hollenbeck A, Schatzkin A. Dietary fiber intake and systematic review and meta-analysis. Am J Epidemiol (2011) 174:631–41.
mortality in the NIH-AARP diet and health study. Arch Intern Med (2011) doi:10.1093/aje/kwr129
171:1061–8. doi:10.1001/archinternmed.2011.18 19. Kamada N, Kim YG, Sham HP, Vallance BA, Puente JL, Martens EC, et al.
7. Tilg H, Moschen AR. Food, immunity, and the microbiome. Gastroenterology Regulated virulence controls the ability of a pathogen to compete with the
(2015) 148:1107–19. doi:10.1053/j.gastro.2014.12.036 gut microbiota. Science (2012) 336:1325–9. doi:10.1126/science.1222195
8. Mozaffarian D, Hao T, Rimm EB, Willett WC, Hu FB. Changes in diet and 20. Martinez-Medina M, Denizot J, Dreux N, Robin F, Billard E, Bonnet R,
lifestyle and long-term weight gain in women and men. N Engl J Med (2011) et al. Western diet induces dysbiosis with increased E coli in CEABAC10
364:2392–404. doi:10.1056/NEJMoa1014296 mice, alters host barrier function favouring AIEC colonisation. Gut (2014)
9. Shoelson SE, Herrero L, Naaz A. Obesity, inflammation, and insulin resistance. 63:116–24. doi:10.1136/gutjnl-2012-304119
Gastroenterology (2007) 132:2169–80. doi:10.1053/j.gastro.2007.03.059 21. Park S, Akinbami LJ, McGuire LC, Blanck HM. Association of sugar-sweet-
10. Ley RE, Peterson DA, Gordon JI. Ecological and evolutionary forces ened beverage intake frequency and asthma among U.S. adults, 2013. Prev
shaping microbial diversity in the human intestine. Cell (2006) 124:837–48. Med (2016) 91:58–61. doi:10.1016/j.ypmed.2016.08.004
doi:10.1016/j.cell.2006.02.017 22. Louis P, Scott KP, Duncan SH, Flint HJ. Understanding the effects of diet
11. Wood LG, Garg ML, Gibson PG. A high-fat challenge increases airway on bacterial metabolism in the large intestine. J Appl Microbiol (2007)
inflammation and impairs bronchodilator recovery in asthma. J Allergy Clin 102:1197–208. doi:10.1111/j.1365-2672.2007.03322.x
Immunol (2011) 127:1133–40. doi:10.1016/j.jaci.2011.01.036 23. Trompette A, Gollwitzer ES, Yadava K, Sichelstiel AK, Sprenger N, Ngom-
12. Rocha DM, Caldas AP, Oliveira LL, Bressan J, Hermsdorff HH. Saturated Bru C, et al. Gut microbiota metabolism of dietary fiber influences allergic
fatty acids trigger TLR4-mediated inflammatory response. Atherosclerosis airway disease and hematopoiesis. Nat Med (2014) 20:159–66. doi:10.1038/
(2016) 244:211–5. doi:10.1016/j.atherosclerosis.2015.11.015 nm.3444
13. Calder PC. Fatty acids and inflammation: the cutting edge between food 24. Li Z, Rava M, Bedard A, Dumas O, Garcia-Aymerich J, Leynaert B, et al.
and pharma. Eur J Pharmacol (2011) 668(Suppl 1):S50–8. doi:10.1016/j. Cured meat intake is associated with worsening asthma symptoms. Thorax
ejphar.2011.05.085 (2017) 72:206–12. doi:10.1136/thoraxjnl-2016-208375
25. Ufnal M, Zadlo A, Ostaszewski R. TMAO: a small molecule of great expecta- 47. Sartor RB. Microbial influences in inflammatory bowel diseases.
tions. Nutrition (2015) 31:1317–23. doi:10.1016/j.nut.2015.05.006 Gastroenterology (2008) 134:577–94. doi:10.1053/j.gastro.2007.11.059
26. Hou YC, Chu CC, Ko TL, Yeh CL, Yeh SL. Effects of alanyl-glutamine 48. Perencevich M, Burakoff R. Use of antibiotics in the treatment of inflam-
dipeptide on the expression of colon-inflammatory mediators during the matory bowel disease. Inflamm Bowel Dis (2006) 12:651–64. doi:10.1097/01.
recovery phase of colitis induced by dextran sulfate sodium. Eur J Nutr MIB.0000225330.38119.c7
(2013) 52:1089–98. doi:10.1007/s00394-012-0416-3 49. Eckburg PB, Bik EM, Bernstein CN, Purdom E, Dethlefsen L,
27. Garcia-Larsen V, Del Giacco SR, Moreira A, Bonini M, Charles D, Reeves T, Sargent M, et al. Diversity of the human intestinal microbial flora. Science
et al. Asthma and dietary intake: an overview of systematic reviews. Allergy (2005) 308:1635–8. doi:10.1126/science.1110591
(2016) 71:433–42. doi:10.1111/all.12800 50. Sun L, Nava GM, Stappenbeck TS. Host genetic susceptibility, dysbiosis, and
28. Kang SG, Wang C, Matsumoto S, Kim CH. High and low vitamin A viral triggers in inflammatory bowel disease. Curr Opin Gastroenterol (2011)
therapies induce distinct FoxP3+ T-cell subsets and effectively control 27:321–7. doi:10.1097/MOG.0b013e32834661b4
intestinal inflammation. Gastroenterology (2009) 137:1391–402.e1391–6. 51. Sokol H, Pigneur B, Watterlot L, Lakhdari O, Bermudez-Humaran LG,
doi:10.1053/j.gastro.2009.06.063 Gratadoux JJ, et al. Faecalibacterium prausnitzii is an anti-inflammatory
29. Abdo J, Rai V, Agrawal DK. Interplay of immunity and vitamin D: inter- commensal bacterium identified by gut microbiota analysis of Crohn dis-
actions and implications with current IBD therapy. Curr Med Chem (2017) ease patients. Proc Natl Acad Sci U S A (2008) 105:16731–6. doi:10.1073/
24:852–67. doi:10.2174/0929867323666161026124951 pnas.0804812105
30. Yuk JM, Shin DM, Lee HM, Yang CS, Jin HS, Kim KK, et al. Vitamin D3 52. Darfeuille-Michaud A, Boudeau J, Bulois P, Neut C, Glasser AL,
induces autophagy in human monocytes/macrophages via cathelicidin. Cell Barnich N, et al. High prevalence of adherent-invasive Escherichia coli
Host Microbe (2009) 6:231–43. doi:10.1016/j.chom.2009.08.004 associated with ileal mucosa in Crohn’s disease. Gastroenterology (2004)
31. Bailey LB, Stover PJ, McNulty H, Fenech MF, Gregory JF III, Mills JL, 127:412–21. doi:10.1053/j.gastro.2004.04.061
et al. Biomarkers of nutrition for development-folate review. J Nutr (2015) 53. Jostins L, Ripke S, Weersma RK, Duerr RH, McGovern DP, Hui KY,
145:1636S–80S. doi:10.3945/jn.114.206599 et al. Host-microbe interactions have shaped the genetic architecture of
32. Blatter J, Brehm JM, Sordillo J, Forno E, Boutaoui N, Acosta-Perez E, inflammatory bowel disease. Nature (2012) 491:119–24. doi:10.1038/
et al. Folate deficiency, atopy, and severe asthma exacerbations in Puerto nature11582
Rican children. Ann Am Thorac Soc (2016) 13:223–30. doi:10.1513/ 54. Abe K, Nguyen KP, Fine SD, Mo JH, Shen C, Shenouda S, et al. Conventional
AnnalsATS.201508-549OC dendritic cells regulate the outcome of colonic inflammation independently
33. Kinoshita M, Kayama H, Kusu T, Yamaguchi T, Kunisawa J, Kiyono H, et al. of T cells. Proc Natl Acad Sci U S A (2007) 104:17022–7. doi:10.1073/
Dietary folic acid promotes survival of Foxp3+ regulatory T cells in the pnas.0708469104
colon. J Immunol (2012) 189:2869–78. doi:10.4049/jimmunol.1200420 55. Arranz A, Doxaki C, Vergadi E, Martinez de la Torre Y, Vaporidi K,
34. Thuesen BH, Husemoen LL, Ovesen L, Jorgensen T, Fenger M, Lagoudaki ED, et al. Akt1 and Akt2 protein kinases differentially contribute
Gilderson G, et al. Atopy, asthma, and lung function in relation to folate and vita- to macrophage polarization. Proc Natl Acad Sci U S A (2012) 109:9517–22.
min B(12) in adults. Allergy (2010) 65:1446–54. doi:10.1111/j.1398-9995.2010. doi:10.1073/pnas.1119038109
02378.x 56. Berndt BE, Zhang M, Chen GH, Huffnagle GB, Kao JY. The role of dendritic
35. Cassat JE, Skaar EP. Iron in infection and immunity. Cell Host Microbe (2013) cells in the development of acute dextran sulfate sodium colitis. J Immunol
13:509–19. doi:10.1016/j.chom.2013.04.010 (2007) 179:6255–62. doi:10.4049/jimmunol.179.9.6255
36. Perl DP, Fogarty U, Harpaz N, Sachar DB. Bacterial-metal interac- 57. Hunter MM, Wang A, Parhar KS, Johnston MJ, Van Rooijen N, Beck PL,
tions: the potential role of aluminum and other trace elements in et al. In vitro-derived alternatively activated macrophages reduce colonic
the etiology of Crohn’s disease. Inflamm Bowel Dis (2004) 10:881–3. inflammation in mice. Gastroenterology (2010) 138:1395–405. doi:10.1053/j.
doi:10.1097/00054725-200411000-00022 gastro.2009.12.041
37. Lees CW, Barrett JC, Parkes M, Satsangi J. New IBD genetics: common 58. Qualls JE, Kaplan AM, van Rooijen N, Cohen DA. Suppression of exper-
pathways with other diseases. Gut (2011) 60(12):1739–53. doi:10.1136/ imental colitis by intestinal mononuclear phagocytes. J Leukoc Biol (2006)
gut.2009.199679 80:802–15. doi:10.1189/jlb.1205734
38. Zhernakova A, van Diemen CC, Wijmenga C. Detecting shared pathogenesis 59. Qualls JE, Tuna H, Kaplan AM, Cohen DA. Suppression of experimental coli-
from the shared genetics of immune-related diseases. Nat Rev Genet (2009) tis in mice by CD11c+ dendritic cells. Inflamm Bowel Dis (2009) 15:236–47.
10(1):43–55. doi:10.1038/nrg2489 doi:10.1002/ibd.20733
39. Khor B, Gardet A, Xavier RJ. Genetics and pathogenesis of inflammatory 60. Watanabe N, Ikuta K, Okazaki K, Nakase H, Tabata Y, Matsuura M, et al.
bowel disease. Nature (2011) 474:307–17. doi:10.1038/nature10209 Elimination of local macrophages in intestine prevents chronic colitis in
40. Vercelli D. Discovering susceptibility genes for asthma and allergy. Nat Rev interleukin-10-deficient mice. Dig Dis Sci (2003) 48:408–14. doi:10.1023/
Immunol (2008) 8(3):169–82. doi:10.1038/nri2257 A:1021960401290
41. Sullivan A, Hunt E, MacSharry J, Murphy DM. The microbiome and 61. Coskun M. Intestinal epithelium in inflammatory bowel disease. Front Med
the pathophysiology of asthma. Respir Res (2016) 17:163. doi:10.1186/ (2014) 1:24. doi:10.3389/fmed.2014.00024
s12931-016-0479-4 62. Neurath MF. Cytokines in inflammatory bowel disease. Nat Rev Immunol
42. Huttenhower C, Kostic AD, Xavier RJ. Inflammatory bowel disease as a model (2014) 14:329–42. doi:10.1038/nri3661
for translating the microbiome. Immunity (2014) 40:843–54. doi:10.1016/j. 63. Fuss IJ, Heller F, Boirivant M, Leon F, Yoshida M, Fichtner-Feigl S, et al.
immuni.2014.05.013 Nonclassical CD1d-restricted NK T cells that produce IL-13 characterize an
43. Melgar S, Shanahan F. Inflammatory bowel disease-from mechanisms atypical Th2 response in ulcerative colitis. J Clin Invest (2004) 113:1490–7.
to treatment strategies. Autoimmunity (2010) 43:463–77. doi:10.3109/ doi:10.1172/JCI19836
08916931003674709 64. Inoue S, Matsumoto T, Iida M, Mizuno M, Kuroki F, Hoshika K, et al.
44. Lichtenstein GR, Hanauer SB, Sandborn WJ; Practice Parameters Committee Characterization of cytokine expression in the rectal mucosa of ulcerative
of American College of Gastroenterology. Management of Crohn’s disease in colitis: correlation with disease activity. Am J Gastroenterol (1999) 94:2441–6.
adults. Am J Gastroenterol (2009) 104:465–83; quiz 464, 484. doi:10.1038/ doi:10.1111/j.1572-0241.1999.01372.x
ajg.2008.168 65. Melgar S, Yeung MM, Bas A, Forsberg G, Suhr O, Oberg A, et al.
45. Neurath MF. Current and emerging therapeutic targets for IBD. Nat Rev Over-expression of interleukin 10 in mucosal T cells of patients
Gastroenterol Hepatol (2017) 14:269–78. doi:10.1038/nrgastro.2016.208 with active ulcerative colitis. Clin Exp Immunol (2003) 134:127–37.
46. Ozaki E, Campbell M, Doyle SL. Targeting the NLRP3 inflammasome in doi:10.1046/j.1365-2249.2003.02268.x
chronic inflammatory diseases: current perspectives. J Inflamm Res (2015) 66. Nunes C, Pereira AM, Morais-Almeida M. Asthma costs and social impact.
8:15–27. doi:10.2147/JIR.S51250 Asthma Res Pract (2017) 3:1. doi:10.1186/s40733-016-0029-3
67. Mitchell PD, O’Byrne PM. Biologics and the lung: TSLP and other epithelial 89. Wellen KE, Hotamisligil GS. Inflammation, stress, and diabetes. J Clin Invest
cell-derived cytokines in asthma. Pharmacol Ther (2017) 169:104–12. (2005) 115:1111–9. doi:10.1172/JCI200525102
doi:10.1016/j.pharmthera.2016.06.009 90. Kitamoto S, Nagao-Kitamoto H, Kuffa P, Kamada N. Regulation of viru-
68. Byrne AJ, Maher TM, Lloyd CM. Pulmonary macrophages: a new therapeu- lence: the rise and fall of gastrointestinal pathogens. J Gastroenterol (2016)
tic pathway in fibrosing lung disease? Trends Mol Med (2016) 22:303–16. 51:195–205. doi:10.1007/s00535-015-1141-5
doi:10.1016/j.molmed.2016.02.004 91. Jandhyala SM, Talukdar R, Subramanyam C, Vuyyuru H, Sasikala M,
69. Iwasaki A, Foxman EF, Molony RD. Early local immune defences in the Nageshwar Reddy D. Role of the normal gut microbiota. World J Gastroenterol
respiratory tract. Nat Rev Immunol (2017) 17:7–20. doi:10.1038/nri.2016.117 (2015) 21:8787–803. doi:10.3748/wjg.v21.i29.8787
70. Deckers J, Branco Madeira F, Hammad H. Innate immune cells in asthma. 92. Zhang C, Zhang M, Pang X, Zhao Y, Wang L, Zhao L. Structural resilience of
Trends Immunol (2013) 34:540–7. doi:10.1016/j.it.2013.08.004 the gut microbiota in adult mice under high-fat dietary perturbations. ISME
71. Barnes PJ. Immunology of asthma and chronic obstructive pulmonary J (2012) 6:1848–57. doi:10.1038/ismej.2012.27
disease. Nat Rev Immunol (2008) 8:183–92. doi:10.1038/nri2254 93. David LA, Maurice CF, Carmody RN, Gootenberg DB, Button JE, Wolfe BE,
72. Chang HS, Lee TH, Jun JA, Baek AR, Park JS, Koo SM, et al. Neutrophilic et al. Diet rapidly and reproducibly alters the human gut microbiome. Nature
inflammation in asthma: mechanisms and therapeutic considerations. (2014) 505:559–63. doi:10.1038/nature12820
Expert Rev Respir Med (2017) 11:29–40. doi:10.1080/17476348.2017.1268919 94. Walker AW, Ince J, Duncan SH, Webster LM, Holtrop G, Ze X, et al. Dominant
73. Budden KF, Gellatly SL, Wood DL, Cooper MA, Morrison M, Hugenholtz P, and diet-responsive groups of bacteria within the human colonic microbiota.
et al. Emerging pathogenic links between microbiota and the gut-lung axis. ISME J (2011) 5:220–30. doi:10.1038/ismej.2010.118
Nat Rev Microbiol (2017) 15:55–63. doi:10.1038/nrmicro.2016.142 95. Lyons CL, Kennedy EB, Roche HM. Metabolic inflammation-differential
74. Kumar RK, Herbert C, Foster PS. Mouse models of acute exacerbations of modulation by dietary constituents. Nutrients (2016) 8. doi:10.3390/
allergic asthma. Respirology (2016) 21:842–9. doi:10.1111/resp.12760 nu8050247
75. Debeuf N, Haspeslagh E, van Helden M, Hammad H, Lambrecht BN. Mouse 96. Harper JW, Zisman TL. Interaction of obesity and inflammatory bowel
models of asthma. Curr Protoc Mouse Biol (2016) 6:169–84. doi:10.1002/ disease. World J Gastroenterol (2016) 22:7868–81. doi:10.3748/wjg.v22.
cpmo.4 i35.7868
76. Ober C, Yao TC. The genetics of asthma and allergic disease: a 21st 97. Barbier M, Vidal H, Desreumaux P, Dubuquoy L, Bourreille A,
century perspective. Immunol Rev (2011) 242:10–30. doi:10.1111/j. Colombel JF, et al. Overexpression of leptin mRNA in mesenteric adipose
1600-065X.2011.01029.x tissue in inflammatory bowel diseases. Gastroenterol Clin Biol (2003)
77. Moheimani F, Hsu AC, Reid AT, Williams T, Kicic A, Stick SM, et al. The 27:987–91.
genetic and epigenetic landscapes of the epithelium in asthma. Respir Res 98. Bertin B, Desreumaux P, Dubuquoy L. Obesity, visceral fat and Crohn’s
(2016) 17:119. doi:10.1186/s12931-016-0434-4 disease. Curr Opin Clin Nutr Metab Care (2010) 13:574–80. doi:10.1097/
78. Sulovari A, Chen YH, Hudziak JJ, Li D. Atlas of human diseases influenced MCO.0b013e32833cf0f4
by genetic variants with extreme allele frequency differences. Hum Genet 99. Desreumaux P, Ernst O, Geboes K, Gambiez L, Berrebi D, Muller-Alouf
(2017) 136:39–54. doi:10.1007/s00439-016-1734-y H, et al. Inflammatory alterations in mesenteric adipose tissue in Crohn’s
79. Forsythe P, Inman MD, Bienenstock J. Oral treatment with live Lactobacillus disease. Gastroenterology (1999) 117:73–81. doi:10.1016/S0016-5085(99)
reuteri inhibits the allergic airway response in mice. Am J Respir Crit Care 70552-4
Med (2007) 175:561–9. doi:10.1164/rccm.200606-821OC 100. Rodrigues VS, Milanski M, Fagundes JJ, Torsoni AS, Ayrizono ML,
80. MacSharry J, O’Mahony C, Shalaby KH, Sheil B, Karmouty-Quintana H, Nunez CE, et al. Serum levels and mesenteric fat tissue expression of
Shanahan F, et al. Immunomodulatory effects of feeding with adiponectin and leptin in patients with Crohn’s disease. Clin Exp Immunol
Bifidobacterium longum on allergen-induced lung inflammation in the (2012) 170:358–64. doi:10.1111/j.1365-2249.2012.04660.x
mouse. Pulm Pharmacol Ther (2012) 25:325–34. doi:10.1016/j.pupt.2012. 101. Peyrin-Biroulet L, Gonzalez F, Dubuquoy L, Rousseaux C, Dubuquoy C,
05.011 Decourcelle C, et al. Mesenteric fat as a source of C reactive protein and as
81. Russell SL, Gold MJ, Reynolds LA, Willing BP, Dimitriu P, Thorson L, et al. a target for bacterial translocation in Crohn’s disease. Gut (2012) 61:78–85.
Perinatal antibiotic-induced shifts in gut microbiota have differential effects doi:10.1136/gutjnl-2011-300370
on inflammatory lung diseases. J Allergy Clin Immunol (2015) 135:100–9. 102. Pietsch J, Batra A, Stroh T, Fedke I, Glauben R, Okur B, et al. Toll-like
doi:10.1016/j.jaci.2014.06.027 receptor expression and response to specific stimulation in adipocytes and
82. Wheeler ML, Limon JJ, Bar AS, Leal CA, Gargus M, Tang J, et al. preadipocytes: on the role of fat in inflammation. Ann N Y Acad Sci (2006)
Immunological consequences of intestinal fungal dysbiosis. Cell Host 1072:407–9. doi:10.1196/annals.1326.021
Microbe (2016) 19:865–73. doi:10.1016/j.chom.2016.05.003 103. Sheehan AL, Warren BF, Gear MW, Shepherd NA. Fat-wrapping in Crohn’s
83. Arrieta MC, Stiemsma LT, Dimitriu PA, Thorson L, Russell S, Yurist- disease: pathological basis and relevance to surgical practice. Br J Surg (1992)
Doutsch S, et al. Early infancy microbial and metabolic alterations affect 79:955–8.
risk of childhood asthma. Sci Transl Med (2015) 7:307ra152. doi:10.1126/ 104. Borley NR, Mortensen NJ, Jewell DP, Warren BF. The relationship between
scitranslmed.aab2271 inflammatory and serosal connective tissue changes in ileal Crohn’s dis-
84. Stiemsma LT, Arrieta MC, Dimitriu PA, Cheng J, Thorson L, Lefebvre DL, ease: evidence for a possible causative link. J Pathol (2000) 190:196–202.
et al. Shifts in Lachnospira and Clostridium sp. in the 3-month stool micro- doi:10.1002/(SICI)1096-9896(200002)190:2<196::AID-PATH513>3.0.
biome are associated with preschool age asthma. Clin Sci (Lond) (2016) CO;2-5
130:2199–207. 105. Olivier I, Theodorou V, Valet P, Castan-Laurell I, Guillou H, Bertrand-Michel
85. Sonnenburg JL, Angenent LT, Gordon JI. Getting a grip on things: how J, et al. Is Crohn’s creeping fat an adipose tissue? Inflamm Bowel Dis (2011)
do communities of bacterial symbionts become established in our intestine? 17(3):747–57. doi:10.1002/ibd.21413
Nat Immunol (2004) 5:569–73. doi:10.1038/ni1079 106. Dubuquoy L, Rousseaux C, Thuru X, Peyrin-Biroulet L, Romano O,
86. De Filippo C, Cavalieri D, Di Paola M, Ramazzotti M, Poullet JB, Chavatte P, et al. PPARgamma as a new therapeutic target in inflammatory
Massart S, et al. Impact of diet in shaping gut microbiota revealed by a bowel diseases. Gut (2006) 55:1341–9. doi:10.1136/gut.2006.093484
comparative study in children from Europe and rural Africa. Proc Natl Acad 107. Mansen A, Guardiola-Diaz H, Rafter J, Branting C, Gustafsson JA.
Sci U S A (2010) 107:14691–6. doi:10.1073/pnas.1005963107 Expression of the peroxisome proliferator-activated receptor (PPAR) in the
87. Geuking MB, Koller Y, Rupp S, McCoy KD. The interplay between the mouse colonic mucosa. Biochem Biophys Res Commun (1996) 222:844–51.
gut microbiota and the immune system. Gut Microbes (2014) 5:411–8. doi:10.1006/bbrc.1996.0832
doi:10.4161/gmic.29330 108. Kim HY, Lee HJ, Chang YJ, Pichavant M, Shore SA, Fitzgerald KA, et al.
88. Cani PD, Amar J, Iglesias MA, Poggi M, Knauf C, Bastelica D, et al. Metabolic Interleukin-17-producing innate lymphoid cells and the NLRP3 inflam-
endotoxemia initiates obesity and insulin resistance. Diabetes (2007) masome facilitate obesity-associated airway hyperreactivity. Nat Med (2014)
56:1761–72. doi:10.2337/db06-1491 20:54–61. doi:10.1038/nm.3423
109. Gevers D, Kugathasan S, Denson LA, Vazquez-Baeza Y, Van Treuren W, review and meta-analysis. Clin Exp Allergy (2015) 45:744–57. doi:10.1111/
Ren B, et al. The treatment-naive microbiome in new-onset Crohn’s disease. cea.12429
Cell Host Microbe (2014) 15:382–92. doi:10.1016/j.chom.2014.02.005 129. House JS, Wyss AB, Hoppin JA, Richards M, Long S, Umbach DM, et al.
110. Halmos EP, Gibson PR. Dietary management of IBD – insights and advice. Early-life farm exposures and adult asthma and atopy in the Agricultural
Nat Rev Gastroenterol Hepatol (2015) 12:133–46. doi:10.1038/nrgastro. Lung Health Study. J Allergy Clin Immunol (2017) 140(1):249–56.e214.
2015.11 doi:10.1016/j.jaci.2016.09.036
111. Shivashankar R, Lewis JD. The role of diet in inflammatory bowel disease. 130. Sozanska B, Pearce N, Dudek K, Cullinan P. Consumption of unpasteurized
Curr Gastroenterol Rep (2017) 19:22. doi:10.1007/s11894-017-0563-z milk and its effects on atopy and asthma in children and adult inhabitants in
112. Willemsen LE. Dietary n-3 long chain polyunsaturated fatty acids in allergy rural Poland. Allergy (2013) 68:644–50. doi:10.1111/all.12147
prevention and asthma treatment. Eur J Pharmacol (2016) 785:174–86. 131. Roberts CL, Rushworth SL, Richman E, Rhodes JM. Hypothesis: increased
doi:10.1016/j.ejphar.2016.03.062 consumption of emulsifiers as an explanation for the rising incidence of
113. Chan SS, Luben R, Olsen A, Tjonneland A, Kaaks R, Teucher B, et al. Body Crohn’s disease. J Crohns Colitis (2013) 7:338–41. doi:10.1016/j.crohns.
mass index and the risk for Crohn’s disease and ulcerative colitis: data 2013.01.004
from a European Prospective Cohort Study (The IBD in EPIC study). Am 132. Chassaing B, Koren O, Goodrich JK, Poole AC, Srinivasan S, Ley RE, et al.
J Gastroenterol (2013) 108:575–82. doi:10.1038/ajg.2012.453 Dietary emulsifiers impact the mouse gut microbiota promoting colitis and
114. de Silva PS, Luben R, Shrestha SS, Khaw KT, Hart AR. Dietary arachidonic metabolic syndrome. Nature (2015) 519:92–6. doi:10.1038/nature14232
and oleic acid intake in ulcerative colitis etiology: a prospective cohort 133. Roberts CL, Keita AV, Duncan SH, O’Kennedy N, Soderholm JD, Rhodes JM,
study using 7-day food diaries. Eur J Gastroenterol Hepatol (2014) 26:11–8. et al. Translocation of Crohn’s disease Escherichia coli across M-cells: con-
doi:10.1097/MEG.0b013e328365c372 trasting effects of soluble plant fibres and emulsifiers. Gut (2010) 59:1331–9.
115. Wiese DM, Horst SN, Brown CT, Allaman MM, Hodges ME, doi:10.1136/gut.2009.195370
Slaughter JC, et al. Serum fatty acids are correlated with inflammatory 134. Chassaing B, Van de Wiele T, De Bodt J, Marzorati M, Gewirtz AT. Dietary
cytokines in ulcerative colitis. PLoS One (2016) 11:e0156387. doi:10.1371/ emulsifiers directly alter human microbiota composition and gene expres-
journal.pone.0156387 sion ex vivo potentiating intestinal inflammation. Gut (2017). doi:10.1136/
116. Ferreira P, Cravo M, Guerreiro CS, Tavares L, Santos PM, Brito M. Fat intake gutjnl-2016-313099
interacts with polymorphisms of Caspase9, FasLigand and PPARgamma 135. Hou JK, Lee D, Lewis J. Diet and inflammatory bowel disease: review of
apoptotic genes in modulating Crohn’s disease activity. Clin Nutr (2010) patient-targeted recommendations. Clin Gastroenterol Hepatol (2014)
29:819–23. doi:10.1016/j.clnu.2010.06.008 12:1592–600. doi:10.1016/j.cgh.2013.09.063
117. Nagel G, Linseisen J. Dietary intake of fatty acids, antioxidants and 136. Chan SS, Luben R, van Schaik F, Oldenburg B, Bueno-de-Mesquita HB,
selected food groups and asthma in adults. Eur J Clin Nutr (2005) 59:8–15. Hallmans G, et al. Carbohydrate intake in the etiology of Crohn’s disease
doi:10.1038/sj.ejcn.1602025 and ulcerative colitis. Inflamm Bowel Dis (2014) 20:2013–21. doi:10.1097/
118. Wijga AH, Smit HA, Kerkhof M, de Jongste JC, Gerritsen J, Neijens HJ, et al. MIB.0000000000000168
Association of consumption of products containing milk fat with reduced 137. Lucendo AJ, De Rezende LC. Importance of nutrition in inflammatory
asthma risk in pre-school children: the PIAMA birth cohort study. Thorax bowel disease. World J Gastroenterol (2009) 15:2081–8. doi:10.3748/wjg.
(2003) 58:567–72. doi:10.1136/thorax.58.7.567 15.2081
119. Yang ZH, Miyahara H, Takeo J, Katayama M. Diet high in fat and 138. Ananthakrishnan AN, Khalili H, Konijeti GG, Higuchi LM, de Silva P,
sucrose induces rapid onset of obesity-related metabolic syndrome Korzenik JR, et al. A prospective study of long-term intake of dietary fiber
partly through rapid response of genes involved in lipogenesis, insulin and risk of Crohn’s disease and ulcerative colitis. Gastroenterology (2013)
signalling and inflammation in mice. Diabetol Metab Syndr (2012) 4:32. 145:970–7. doi:10.1053/j.gastro.2013.07.050
doi:10.1186/1758-5996-4-32 139. Monteleone I, Pallone F, Monteleone G. Aryl hydrocarbon receptor and
120. Miller B, Fervers F, Rohbeck R, Strohmeyer G. [Sugar consumption in patients colitis. Semin Immunopathol (2013) 35:671–5. doi:10.1007/s00281-013-
with Crohn’s disease]. Verh Dtsch Ges Inn Med (1976) 82(Pt 1):922–4. 0396-2
121. Racine A, Carbonnel F, Chan SS, Hart AR, Bueno-de-Mesquita HB, 140. Li Y, Innocentin S, Withers DR, Roberts NA, Gallagher AR,
Oldenburg B, et al. Dietary patterns and risk of inflammatory bowel disease Grigorieva EF, et al. Exogenous stimuli maintain intraepithelial lympho-
in Europe: results from the EPIC study. Inflamm Bowel Dis (2016) 22:345–54. cytes via aryl hydrocarbon receptor activation. Cell (2011) 147:629–40.
doi:10.1097/MIB.0000000000000638 doi:10.1016/j.cell.2011.09.025
122. Berentzen NE, van Stokkom VL, Gehring U, Koppelman GH, Schaap LA, 141. Monteleone I, Rizzo A, Sarra M, Sica G, Sileri P, Biancone L, et al. Aryl
Smit HA, et al. Associations of sugar-containing beverages with asthma hydrocarbon receptor-induced signals up-regulate IL-22 production and
prevalence in 11-year-old children: the PIAMA birth cohort. Eur J Clin Nutr inhibit inflammation in the gastrointestinal tract. Gastroenterology (2011)
(2015) 69:303–8. doi:10.1038/ejcn.2014.153 141:237–248, 248.e231. doi:10.1053/j.gastro.2011.04.007
123. Agus A, Denizot J, Thevenot J, Martinez-Medina M, Massier S, Sauvanet P, 142. Qiu Y, Peng K, Liu M, Xiao W, Yang H. CD8alphaalpha TCRalphabeta
et al. Western diet induces a shift in microbiota composition enhancing sus- intraepithelial lymphocytes in the mouse gut. Dig Dis Sci (2016) 61:1451–60.
ceptibility to adherent-invasive E. coli infection and intestinal inflammation. doi:10.1007/s10620-015-4016-y
Sci Rep (2016) 6:19032. doi:10.1038/srep19032 143. Leibelt S, Friede ME, Rohe C, Gutle D, Rutkowski E, Weigert A, et al.
124. Shoda R, Matsueda K, Yamato S, Umeda N. Epidemiologic analysis of Crohn Dedicated immunosensing of the mouse intestinal epithelium facilitated by
disease in Japan: increased dietary intake of n-6 polyunsaturated fatty acids a pair of genetically coupled lectin-like receptors. Mucosal Immunol (2015)
and animal protein relates to the increased incidence of Crohn disease in 8:232–42. doi:10.1038/mi.2014.60
Japan. Am J Clin Nutr (1996) 63:741–5. 144. Schmitz F, Kooy-Winkelaar Y, Wiekmeijer AS, Brugman MH, Mearin
125. Maconi G, Ardizzone S, Cucino C, Bezzio C, Russo AG, Bianchi Porro G. ML, Mulder C, et al. The composition and differentiation potential of
Pre-illness changes in dietary habits and diet as a risk factor for inflam- the duodenal intraepithelial innate lymphocyte compartment is altered
matory bowel disease: a case-control study. World J Gastroenterol (2010) in coeliac disease. Gut (2016) 65:1269–78. doi:10.1136/gutjnl-2014-
16:4297–304. doi:10.3748/wjg.v16.i34.4297 308153
126. Devkota S, Wang Y, Musch MW, Leone V, Fehlner-Peach H, 145. Grimstad T, Berge RK, Bohov P, Skorve J, Goransson L, Omdal R, et al.
Nadimpalli A, et al. Dietary-fat-induced taurocholic acid promotes pathobiont Salmon diet in patients with active ulcerative colitis reduced the simple
expansion and colitis in Il10-/- mice. Nature (2012) 487:104–8. doi:10.1038/ clinical colitis activity index and increased the anti-inflammatory fatty acid
nature11225 index – a pilot study. Scand J Clin Lab Invest (2011) 71:68–73. doi:10.3109/0
127. Wright R, Truelove SC. A controlled therapeutic trial of various diets in 0365513.2010.542484
ulcerative colitis. Br Med J (1965) 2:138–41. doi:10.1136/bmj.2.4724.138 146. Yang H, Xun P, He K. Fish and fish oil intake in relation to risk of asthma:
128. Campbell BE, Lodge CJ, Lowe AJ, Burgess JA, Matheson MC, a systematic review and meta-analysis. PLoS One (2013) 8:e80048.
Dharmage SC. Exposure to ’farming’ and objective markers of atopy: a systematic doi:10.1371/journal.pone.0080048
147. Vannice G, Rasmussen H. Position of the academy of nutrition and dietet- Leukot Essent Fatty Acids (2009) 81:187–91. doi:10.1016/j.plefa.2009.
ics: dietary fatty acids for healthy adults. J Acad Nutr Diet 114(1):136–53. 05.010
doi:10.1016/j.jand.2013.11.001 166. Schmitz G, Ecker J. The opposing effects of n-3 and n-6 fatty acids. Prog Lipid
148. Huang S, Rutkowsky JM, Snodgrass RG, Ono-Moore KD, Schneider DA, Res (2008) 47:147–55. doi:10.1016/j.plipres.2007.12.004
Newman JW, et al. Saturated fatty acids activate TLR-mediated proinflam- 167. Stenson WF. The universe of arachidonic acid metabolites in inflammatory
matory signaling pathways. J Lipid Res (2012) 53:2002–13. doi:10.1194/jlr. bowel disease: can we tell the good from the bad? Curr Opin Gastroenterol
D029546 (2014) 30:347–51. doi:10.1097/MOG.0000000000000075
149. Yang X, Haghiac M, Glazebrook P, Minium J, Catalano PM, Hauguel- 168. Lee JY, Plakidas A, Lee WH, Heikkinen A, Chanmugam P, Bray G, et al.
de Mouzon S. Saturated fatty acids enhance TLR4 immune pathways in Differential modulation of toll-like receptors by fatty acids: preferential
human trophoblasts. Hum Reprod (2015) 30:2152–9. doi:10.1093/humrep/ inhibition by n-3 polyunsaturated fatty acids. J Lipid Res (2003) 44:479–86.
dev173 doi:10.1194/jlr.M200361-JLR200
150. Lee JY, Ye J, Gao Z, Youn HS, Lee WH, Zhao L, et al. Reciprocal modulation 169. Oh DY, Talukdar S, Bae EJ, Imamura T, Morinaga H, Fan W, et al. GPR120
of toll-like receptor-4 signaling pathways involving MyD88 and phospha- is an omega-3 fatty acid receptor mediating potent anti-inflammatory
tidylinositol 3-kinase/AKT by saturated and polyunsaturated fatty acids. and insulin-sensitizing effects. Cell (2010) 142:687–98. doi:10.1016/j.cell.
J Biol Chem (2003) 278:37041–51. doi:10.1074/jbc.M305213200 2010.07.041
151. Moreira AP, Texeira TF, Ferreira AB, Peluzio Mdo C, Alfenas Rde C. Influence 170. Barbalho SM, Goulart Rde A, Quesada K, Bechara MD, de Carvalho Ade C.
of a high-fat diet on gut microbiota, intestinal permeability and metabolic Inflammatory bowel disease: can omega-3 fatty acids really help? Ann
endotoxaemia. Br J Nutr (2012) 108:801–9. doi:10.1017/S0007114512001213 Gastroenterol (2016) 29:37–43.
152. Fernandez ML, West KL. Mechanisms by which dietary fatty acids modulate 171. Tabbaa M, Golubic M, Roizen MF, Bernstein AM. Docosahexaenoic acid,
plasma lipids. J Nutr (2005) 135:2075–8. inflammation, and bacterial dysbiosis in relation to periodontal disease,
153. Stewart CR, Stuart LM, Wilkinson K, van Gils JM, Deng J, Halle A, et al. inflammatory bowel disease, and the metabolic syndrome. Nutrients (2013)
CD36 ligands promote sterile inflammation through assembly of a toll-like 5:3299–310. doi:10.3390/nu5083299
receptor 4 and 6 heterodimer. Nat Immunol (2010) 11:155–61. doi:10.1038/ 172. Liu Y, Chen F, Odle J, Lin X, Jacobi SK, Zhu H, et al. Fish oil enhances
ni.1836 intestinal integrity and inhibits TLR4 and NOD2 signaling pathways in
154. Gruber L, Kisling S, Lichti P, Martin FP, May S, Klingenspor M, et al. High weaned pigs after LPS challenge. J Nutr (2012) 142:2017–24. doi:10.3945/jn.
fat diet accelerates pathogenesis of murine Crohn’s disease-like ileitis 112.164947
independently of obesity. PLoS One (2013) 8:e71661. doi:10.1371/journal. 173. Emelyanov A, Fedoseev G, Krasnoschekova O, Abulimity A, Trendeleva T,
pone.0071661 Barnes PJ. Treatment of asthma with lipid extract of New Zealand green-
155. Paik J, Fierce Y, Treuting PM, Brabb T, Maggio-Price L. High-fat diet-induced lipped mussel: a randomised clinical trial. Eur Respir J (2002) 20:596–600.
obesity exacerbates inflammatory bowel disease in genetically susceptible doi:10.1183/09031936.02.02632001
Mdr1a-/- male mice. J Nutr (2013) 143:1240–7. doi:10.3945/jn.113.174615 174. Mickleborough TD, Lindley MR, Ionescu AA, Fly AD. Protective effect
156. Reddy KV, Naidu KA. Oleic acid, hydroxytyrosol and n-3 fatty acids collec- of fish oil supplementation on exercise-induced bronchoconstriction in
tively modulate colitis through reduction of oxidative stress and IL-8 syn- asthma. Chest (2006) 129:39–49. doi:10.1378/chest.129.1.39
thesis; in vitro and in vivo studies. Int Immunopharmacol (2016) 35:29–42. 175. Hardy MS, Kekic A, Graybill NL, Lancaster ZR. A systematic review of
doi:10.1016/j.intimp.2016.03.019 the association between fish oil supplementation and the development
157. Jenkins DJ, Chiavaroli L, Wong JM, Kendall C, Lewis GF, Vidgen E, et al. of asthma exacerbations. SAGE Open Med (2016) 4:2050312116666216.
Adding monounsaturated fatty acids to a dietary portfolio of cholesterol-low- doi:10.1177/2050312116666216
ering foods in hypercholesterolemia. CMAJ (2010) 182:1961–7. doi:10.1503/ 176. Blasbalg TL, Hibbeln JR, Ramsden CE, Majchrzak SF, Rawlings RR. Changes
cmaj.092128 in consumption of omega-3 and omega-6 fatty acids in the United States
158. Chan KL, Pillon NJ, Sivaloganathan DM, Costford SR, Liu Z, Theret M, during the 20th century. Am J Clin Nutr (2011) 93:950–62. doi:10.3945/
et al. Palmitoleate reverses high fat-induced proinflammatory macrophage ajcn.110.006643
polarization via AMP-activated protein kinase (AMPK). J Biol Chem (2015) 177. Simopoulos AP. The importance of the omega-6/omega-3 fatty acid ratio in
290:16979–88. doi:10.1074/jbc.M115.646992 cardiovascular disease and other chronic diseases. Exp Biol Med (Maywood)
159. Sawai T, Drongowski RA, Lampman RW, Coran AG, Harmon CM. The effect (2008) 233:674–88. doi:10.3181/0711-MR-311
of phospholipids and fatty acids on tight-junction permeability and bacterial 178. Pearl DS, Masoodi M, Eiden M, Brummer J, Gullick D, McKeever TM, et al.
translocation. Pediatr Surg Int (2001) 17:269–74. doi:10.1007/s003830100592 Altered colonic mucosal availability of n-3 and n-6 polyunsaturated fatty
160. Emmanouil E, Manios Y, Grammatikaki E, Kondaki K, Oikonomou E, acids in ulcerative colitis and the relationship to disease activity. J Crohns
Papadopoulos N, et al. Association of nutrient intake and wheeze or asthma Colitis (2014) 8:70–9. doi:10.1016/j.crohns.2013.03.013
in a Greek pre-school population. Pediatr Allergy Immunol (2010) 21:90–5. 179. Kompauer I, Demmelmair H, Koletzko B, Bolte G, Linseisen J,
doi:10.1111/j.1399-3038.2009.00876.x Heinrich J. n6/n3 hypothesis and allergies: biologically plausible, but not
161. DeCoffe D, Quin C, Gill SK, Tasnim N, Brown K, Godovannyi A, et al. confirmed. Eur J Med Res (2004) 9:378–82.
Dietary lipid type, rather than total number of calories, alters outcomes of 180. Sonnenburg ED, Sonnenburg JL. Starving our microbial self: the deleterious
enteric infection in mice. J Infect Dis (2016) 213:1846–56. doi:10.1093/infdis/ consequences of a diet deficient in microbiota-accessible carbohydrates. Cell
jiw084 Metab (2014) 20:779–86. doi:10.1016/j.cmet.2014.07.003
162. Estaki M, DeCoffe D, Gibson DL. Interplay between intestinal alkaline 181. Cummings JH, Macfarlane GT. Role of intestinal bacteria in nutrient metab-
phosphatase, diet, gut microbes and immunity. World J Gastroenterol (2014) olism. JPEN J Parenter Enteral Nutr (1997) 21:357–65. doi:10.1177/0148607
20:15650–6. doi:10.3748/wjg.v20.i42.15650 197021006357
163. Sanchez-Fidalgo S, Cardeno A, Sanchez-Hidalgo M, Aparicio-Soto M, 182. Maslowski KM, Mackay CR. Diet, gut microbiota and immune responses.
Villegas I, Rosillo MA, et al. Dietary unsaponifiable fraction from extra Nat Immunol (2011) 12:5–9. doi:10.1038/ni0111-5
virgin olive oil supplementation attenuates acute ulcerative colitis in mice. 183. Hamer HM, Jonkers D, Venema K, Vanhoutvin S, Troost FJ,
Eur J Pharm Sci (2013) 48:572–81. doi:10.1016/j.ejps.2012.12.004 Brummer RJ. Review article: the role of butyrate on colonic function. Aliment
164. Cardeno A, Magnusson MK, Strid H, Alarcon de La Lastra C, Sanchez- Pharmacol Ther (2008) 27:104–19. doi:10.1111/j.1365-2036.2007.03562.x
Hidalgo M, Ohman L. The unsaponifiable fraction of extra virgin olive oil 184. Arpaia N, Campbell C, Fan X, Dikiy S, van der Veeken J, deRoos P, et al.
promotes apoptosis and attenuates activation and homing properties of Metabolites produced by commensal bacteria promote peripheral regulatory
T cells from patients with inflammatory bowel disease. Food Chem (2014) T-cell generation. Nature (2013) 504:451–5. doi:10.1038/nature12726
161:353–60. doi:10.1016/j.foodchem.2014.04.016 185. Josefowicz SZ, Niec RE, Kim HY, Treuting P, Chinen T, Zheng Y, et al.
165. Chapkin RS, Kim W, Lupton JR, McMurray DN. Dietary docosahexaenoic and Extrathymically generated regulatory T cells control mucosal TH2 inflam-
eicosapentaenoic acid: emerging mediators of inflammation. Prostaglandins mation. Nature (2012) 482:395–9. doi:10.1038/nature10772
186. Millard AL, Mertes PM, Ittelet D, Villard F, Jeannesson P, Bernard J. Butyrate 207. Rossi M, Amaretti A, Raimondi S. Folate production by probiotic bacteria.
affects differentiation, maturation and function of human monocyte-derived Nutrients (2011) 3:118–34. doi:10.3390/nu3010118
dendritic cells and macrophages. Clin Exp Immunol (2002) 130:245–55. 208. Mora JR, Iwata M, Eksteen B, Song SY, Junt T, Senman B, et al. Generation of
doi:10.1046/j.0009-9104.2002.01977.x gut-homing IgA-secreting B cells by intestinal dendritic cells. Science (2006)
187. Wang B, Morinobu A, Horiuchi M, Liu J, Kumagai S. Butyrate inhibits 314:1157–60. doi:10.1126/science.1132742
functional differentiation of human monocyte-derived dendritic cells. Cell 209. Goverse G, Labao-Almeida C, Ferreira M, Molenaar R, Wahlen S, Konijn T,
Immunol (2008) 253:54–8. doi:10.1016/j.cellimm.2008.04.016 et al. Vitamin A controls the presence of RORgamma+ innate lymphoid cells
188. Koh A, De Vadder F, Kovatcheva-Datchary P, Backhed F. From dietary fiber and lymphoid tissue in the small intestine. J Immunol (2016) 196:5148–55.
to host physiology: short-chain fatty acids as key bacterial metabolites. Cell doi:10.4049/jimmunol.1501106
(2016) 165:1332–45. doi:10.1016/j.cell.2016.05.041 210. Seo GY, Jang YS, Kim HA, Lee MR, Park MH, Park SR, et al. Retinoic acid,
189. Annese V, Rogai F, Settesoldi A, Bagnoli S. PPARgamma in inflammatory acting as a highly specific IgA isotype switch factor, cooperates with TGF-
bowel disease. PPAR Res (2012) 2012:620839. doi:10.1155/2012/620839 beta1 to enhance the overall IgA response. J Leukoc Biol (2013) 94:325–35.
190. Frank DN, St Amand AL, Feldman RA, Boedeker EC, Harpaz N, doi:10.1189/jlb.0313128
Pace NR. Molecular-phylogenetic characterization of microbial community 211. Iwata M, Hirakiyama A, Eshima Y, Kagechika H, Kato C, Song SY. Retinoic
imbalances in human inflammatory bowel diseases. Proc Natl Acad Sci U S A acid imprints gut-homing specificity on T cells. Immunity (2004) 21:527–38.
(2007) 104:13780–5. doi:10.1073/pnas.0706625104 doi:10.1016/j.immuni.2004.08.011
191. Machiels K, Joossens M, Sabino J, De Preter V, Arijs I, Eeckhaut V, et al. 212. Ruiter B, Patil SU, Shreffler WG. Vitamins A and D have antagonistic effects
A decrease of the butyrate-producing species Roseburia hominis and on expression of effector cytokines and gut-homing integrin in human innate
Faecalibacterium prausnitzii defines dysbiosis in patients with ulcerative lymphoid cells. Clin Exp Allergy (2015) 45:1214–25. doi:10.1111/cea.12568
colitis. Gut (2014) 63:1275–83. doi:10.1136/gutjnl-2013-304833 213. Sirisinha S. The pleiotropic role of vitamin A in regulating mucosal immu-
192. Zhang T, Ding C, Zhao M, Dai X, Yang J, Li Y, et al. Sodium butyrate reduces nity. Asian Pac J Allergy Immunol (2015) 33:71–89.
colitogenic immunoglobulin A-coated bacteria and modifies the composi- 214. Tejon G, Manriquez V, De Calisto J, Flores-Santibanez F, Hidalgo Y,
tion of microbiota in IL-10 deficient mice. Nutrients (2016) 8. doi:10.3390/ Crisostomo N, et al. Vitamin A impairs the reprogramming of Tregs into
nu8120728 IL-17-producing cells during intestinal inflammation. Biomed Res Int (2015)
193. Gollwitzer ES, Saglani S, Trompette A, Yadava K, Sherburn R, McCoy KD, 2015:137893. doi:10.1155/2015/137893
et al. Lung microbiota promotes tolerance to allergens in neonates via PD-L1. 215. Fransen K, Franzen P, Magnuson A, Elmabsout AA, Nyhlin N, Wickbom A,
Nat Med (2014) 20:642–7. doi:10.1038/nm.3568 et al. Polymorphism in the retinoic acid metabolizing enzyme CYP26B1
194. Halnes I, Baines KJ, Berthon BS, MacDonald-Wicks LK, Gibson PG, and the development of Crohn’s disease. PLoS One (2013) 8:e72739.
Wood LG. Soluble fibre meal challenge reduces airway inflammation doi:10.1371/journal.pone.0072739
and expression of GPR43 and GPR41 in asthma. Nutrients (2017) 9(1). 216. Bai A, Lu N, Guo Y, Liu Z, Chen J, Peng Z. All-trans retinoic acid down-reg-
doi:10.3390/nu9010057 ulates inflammatory responses by shifting the Treg/Th17 profile in human
195. Zhang Z, Shi L, Pang W, Liu W, Li J, Wang H, et al. Dietary fiber intake reg- ulcerative and murine colitis. J Leukoc Biol (2009) 86:959–69. doi:10.1189/
ulates intestinal microflora and inhibits ovalbumin-induced allergic airway jlb.0109006
inflammation in a mouse model. PLoS One (2016) 11:e0147778. doi:10.1371/ 217. Conway TF, Hammer L, Furtado S, Mathiowitz E, Nicoletti F,
journal.pone.0147778 Mangano K, et al. Oral delivery of particulate transforming growth factor
196. Jantchou P, Morois S, Clavel-Chapelon F, Boutron-Ruault MC, beta 1 and all-trans retinoic acid reduces gut inflammation in murine models
Carbonnel F. Animal protein intake and risk of inflammatory bowel dis- of inflammatory bowel disease. J Crohns Colitis (2015) 9:647–58. doi:10.1093/
ease: the E3N prospective study. Am J Gastroenterol (2010) 105:2195–201. ecco-jcc/jjv089
doi:10.1038/ajg.2010.192 218. Penny HL, Prestwood TR, Bhattacharya N, Sun F, Kenkel JA,
197. Bremer J. Carnitine – metabolism and functions. Physiol Rev (1983) Davidson MG, et al. Restoring retinoic acid attenuates intestinal inflamma-
63:1420–80. tion and tumorigenesis in APCMin/+ mice. Cancer Immunol Res (2016)
198. Koeth RA, Wang Z, Levison BS, Buffa JA, Org E, Sheehy BT, et al. Intestinal 4:917–26. doi:10.1158/2326-6066.CIR-15-0038
microbiota metabolism of l-carnitine, a nutrient in red meat, promotes 219. McDaniel KL, Restori KH, Dodds JW, Kennett MJ, Ross AC, Cantorna MT.
atherosclerosis. Nat Med (2013) 19:576–85. doi:10.1038/nm.3145 Vitamin A-deficient hosts become nonsymptomatic reservoirs of Escherichia
199. Wilson A, Teft WA, Morse BL, Choi YH, Woolsey S, DeGorter MK, et al. coli-like enteric infections. Infect Immun (2015) 83:2984–91. doi:10.1128/
Trimethylamine-N-oxide: a novel biomarker for the identification of IAI.00201-15
inflammatory bowel disease. Dig Dis Sci (2015) 60:3620–30. doi:10.1007/ 220. Ruane D, Chorny A, Lee H, Faith J, Pandey G, Shan M, et al. Microbiota
s10620-015-3797-3 regulate the ability of lung dendritic cells to induce IgA class-switch recom-
200. Hodge L, Salome CM, Peat JK, Haby MM, Xuan W, Woolcock AJ. bination and generate protective gastrointestinal immune responses. J Exp
Consumption of oily fish and childhood asthma risk. Med J Aust (1996) 164: Med (2016) 213:53–73. doi:10.1084/jem.20150567
137–40. 221. Barbachano A, Fernandez-Barral A, Ferrer-Mayorga G, Costales-Carrera A,
201. Gupta NK, Thaker AI, Kanuri N, Riehl TE, Rowley CW, Stenson WF, Larriba MJ, Munoz A. The endocrine vitamin D system in the gut. Mol Cell
et al. Serum analysis of tryptophan catabolism pathway: correlation with Endocrinol (2016). doi:10.1016/j.mce.2016.11.028
Crohn’s disease activity. Inflamm Bowel Dis (2012) 18:1214–20. doi:10.1002/ 222. Christakos S, Dhawan P, Verstuyf A, Verlinden L, Carmeliet G. Vitamin D:
ibd.21849 metabolism, molecular mechanism of action, and pleiotropic effects. Physiol
202. Kim CJ, Kovacs-Nolan JA, Yang C, Archbold T, Fan MZ, Mine Y. l- Rev (2016) 96:365–408. doi:10.1152/physrev.00014.2015
Tryptophan exhibits therapeutic function in a porcine model of dextran 223. Yamamoto T, Nakahigashi M, Saniabadi AR. Review article: diet and inflam-
sodium sulfate (DSS)-induced colitis. J Nutr Biochem (2010) 21:468–75. matory bowel disease–epidemiology and treatment. Aliment Pharmacol Ther
doi:10.1016/j.jnutbio.2009.01.019 (2009) 30(2):99–112.
203. Thurnham DI. Micronutrients and immune function: some recent develop- 224. Simmons JD, Mullighan C, Welsh KI, Jewell DP. Vitamin D receptor gene
ments. J Clin Pathol (1997) 50:887–91. doi:10.1136/jcp.50.11.887 polymorphism: association with Crohn’s disease susceptibility. Gut (2000)
204. Weisshof R, Chermesh I. Micronutrient deficiencies in inflammatory bowel 47:211–4. doi:10.1136/gut.47.2.211
disease. Curr Opin Clin Nutr Metab Care (2015) 18:576–81. doi:10.1097/ 225. Nurmatov U, Devereux G, Sheikh A. Nutrients and foods for the primary pre-
MCO.0000000000000226 vention of asthma and allergy: systematic review and meta-analysis. J Allergy
205. Allen S, Britton JR, Leonardi-Bee JA. Association between antioxidant vita- Clin Immunol (2011) 127:724–33.e721–30. doi:10.1016/j.jaci.2010.11.001
mins and asthma outcome measures: systematic review and meta-analysis. 226. Kim SH, Pei QM, Jiang P, Yang M, Qian XJ, Liu JB. Effect of active vitamin D3
Thorax (2009) 64:610–9. doi:10.1136/thx.2008.101469 on VEGF-induced ADAM33 expression and proliferation in human airway
206. Burkholder PR, McVeigh I. Synthesis of vitamins by intestinal bacteria. Proc smooth muscle cells: implications for asthma treatment. Respir Res (2017)
Natl Acad Sci U S A (1942) 28:285–9. doi:10.1073/pnas.28.7.285 18:7. doi:10.1186/s12931-016-0490-9
227. Margolis RN, Christakos S. The nuclear receptor superfamily of steroid 246. Wang T, Zhang HP, Zhang X, Liang ZA, Ji YL, Wang G. Is folate status a risk
hormones and vitamin D gene regulation. An update. Ann N Y Acad Sci factor for asthma or other allergic diseases? Allergy Asthma Immunol Res
(2010) 1192:208–14. doi:10.1111/j.1749-6632.2009.05227.x (2015) 7:538–46. doi:10.4168/aair.2015.7.6.538
228. Liu W, Chen Y, Golan MA, Annunziata ML, Du J, Dougherty U, et al. 247. Beulens JW, Booth SL, van den Heuvel EG, Stoecklin E, Baka A, Vermeer C.
Intestinal epithelial vitamin D receptor signaling inhibits experimental The role of menaquinones (vitamin K(2)) in human health. Br J Nutr (2013)
colitis. J Clin Invest (2013) 123:3983–96. doi:10.1172/JCI65842 110:1357–68. doi:10.1017/S0007114513001013
229. Ordonez-Moran P, Larriba MJ, Palmer HG, Valero RA, Barbachano A, 248. Hill MJ. Intestinal flora and endogenous vitamin synthesis. Eur J Cancer Prev
Dunach M, et al. RhoA-ROCK and p38MAPK-MSK1 mediate vitamin D (1997) 6(Suppl 1):S43–5. doi:10.1097/00008469-199703001-00009
effects on gene expression, phenotype, and Wnt pathway in colon cancer 249. Nowak JK, Grzybowska-Chlebowczyk U, Landowski P, Szaflarska-
cells. J Cell Biol (2008) 183:697–710. doi:10.1083/jcb.200803020 Poplawska A, Klincewicz B, Adamczak D, et al. Prevalence and correlates of
230. Golan MA, Liu W, Shi Y, Chen L, Wang J, Liu T, et al. Transgenic expres- vitamin K deficiency in children with inflammatory bowel disease. Sci Rep
sion of vitamin D receptor in gut epithelial cells ameliorates spontaneous (2014) 4:4768. doi:10.1038/srep04768
colitis caused by interleukin-10 deficiency. Dig Dis Sci (2015) 60:1941–7. 250. Shiraishi E, Iijima H, Shinzaki S, Nakajima S, Inoue T, Hiyama S, et al. Vitamin
doi:10.1007/s10620-015-3634-8 K deficiency leads to exacerbation of murine dextran sulfate sodium-induced
231. Ooi JH, Li Y, Rogers CJ, Cantorna MT. Vitamin D regulates the gut microbi- colitis. J Gastroenterol (2016) 51:346–56. doi:10.1007/s00535-015-1112-x
ome and protects mice from dextran sodium sulfate-induced colitis. J Nutr 251. Kaitha S, Bashir M, Ali T. Iron deficiency anemia in inflammatory bowel
(2013) 143:1679–86. doi:10.3945/jn.113.180794 disease. World J Gastrointest Pathophysiol (2015) 6:62–72. doi:10.4291/wjgp.
232. Ryz NR, Patterson SJ, Zhang Y, Ma C, Huang T, Bhinder G, et al. Active v6.i3.62
vitamin D (1,25-dihydroxyvitamin D3) increases host susceptibility 252. Kulnigg S, Gasche C. Systematic review: managing anaemia in Crohn’s
to Citrobacter rodentium by suppressing mucosal Th17 responses. Am disease. Aliment Pharmacol Ther (2006) 24:1507–23. doi:10.1111/j.
J Physiol Gastrointest Liver Physiol (2012) 303:G1299–311. doi:10.1152/ 1365-2036.2006.03146.x
ajpgi.00320.2012 253. Stein J, Dignass AU. Management of iron deficiency anemia in inflammatory
233. Assa A, Vong L, Pinnell LJ, Rautava J, Avitzur N, Johnson-Henry KC, et al. bowel disease – a practical approach. Ann Gastroenterol (2013) 26:104–13.
Vitamin D deficiency predisposes to adherent-invasive Escherichia coli-in- 254. Werner T, Wagner SJ, Martinez I, Walter J, Chang JS, Clavel T, et al. Depletion
duced barrier dysfunction and experimental colonic injury. Inflamm Bowel of luminal iron alters the gut microbiota and prevents Crohn’s disease-like
Dis (2015) 21:297–306. doi:10.1097/MIB.0000000000000282 ileitis. Gut (2011) 60:325–33. doi:10.1136/gut.2010.216929
234. Su D, Nie Y, Zhu A, Chen Z, Wu P, Zhang L, et al. Vitamin D signaling 255. Carrier JC, Aghdassi E, Jeejeebhoy K, Allard JP. Exacerbation of dextran
through induction of Paneth cell defensins maintains gut microbiota and sulfate sodium-induced colitis by dietary iron supplementation: role of
improves metabolic disorders and hepatic steatosis in animal models. Front NF-kappaB. Int J Colorectal Dis (2006) 21:381–7. doi:10.1007/s00384-005-
Physiol (2016) 7:498. doi:10.3389/fphys.2016.00498 0011-7
235. Hoffman R. Thiamine deficiency in the Western diet and dementia risk. 256. Dostal A, Lacroix C, Pham VT, Zimmermann MB, Del’homme C, Bernalier-
Br J Nutr (2016) 116:188–9. doi:10.1017/S000711451600177X Donadille A, et al. Iron supplementation promotes gut microbiota metabolic
236. Kiela PR, Ghishan FK. Physiology of intestinal absorption and secretion. activity but not colitis markers in human gut microbiota-associated rats.
Best Pract Res Clin Gastroenterol (2016) 30:145–59. doi:10.1016/j.bpg. Br J Nutr (2014) 111:2135–45. doi:10.1017/S000711451400021X
2016.02.007 257. Barrett CW, Short SP, Williams CS. Selenoproteins and oxidative stress-
237. Knecht C, Fretter C, Rosenstiel P, Krawczak M, Hutt MT. Distinct met- induced inflammatory tumorigenesis in the gut. Cell Mol Life Sci (2017)
abolic network states manifest in the gene expression profiles of pediatric 74:607–16. doi:10.1007/s00018-016-2339-2
inflammatory bowel disease patients and controls. Sci Rep (2016) 6:32584. 258. Ojuawo A, Keith L. The serum concentrations of zinc, copper and sele-
doi:10.1038/srep32584 nium in children with inflammatory bowel disease. Cent Afr J Med (2002)
238. Quince C, Ijaz UZ, Loman N, Eren AM, Saulnier D, Russell J, et al. Extensive 48:116–9.
modulation of the fecal metagenome in Children with Crohn’s disease 259. Kaushal N, Kudva AK, Patterson AD, Chiaro C, Kennett MJ, Desai D, et al.
during exclusive enteral nutrition. Am J Gastroenterol (2015) 110:1718–29; Crucial role of macrophage selenoproteins in experimental colitis. J Immunol
quiz 1730. doi:10.1038/ajg.2015.357 (2014) 193:3683–92. doi:10.4049/jimmunol.1400347
239. Mooney S, Leuendorf JE, Hendrickson C, Hellmann H. Vitamin B6: a long 260. Norton RL, Hoffmann PR. Selenium and asthma. Mol Aspects Med (2012)
known compound of surprising complexity. Molecules (2009) 14:329–51. 33:98–106. doi:10.1016/j.mam.2011.10.003
doi:10.3390/molecules14010329 261. Michielan A, D’Inca R. Intestinal permeability in inflammatory bowel dis-
240. Selhub J, Byun A, Liu Z, Mason JB, Bronson RT, Crott JW. Dietary vitamin ease: pathogenesis, clinical evaluation, and therapy of leaky gut. Mediators
B6 intake modulates colonic inflammation in the IL10-/- model of inflam- Inflamm (2015) 2015:628157. doi:10.1155/2015/628157
matory bowel disease. J Nutr Biochem (2013) 24:2138–43. doi:10.1016/j. 262. Ariaee N, Farid R, Shabestari F, Shabestari M, Jabbari Azad F. Trace elements
jnutbio.2013.08.005 status in sera of patients with allergic asthma. Rep Biochem Mol Biol (2016)
241. Fleischman NM, Das D, Kumar A, Xu Q, Chiu HJ, Jaroszewski L, et al. 5:20–5.
Molecular characterization of novel pyridoxal-5’-phosphate-dependent 263. Reeves PG, Nielsen FH, Fahey GC Jr. AIN-93 purified diets for laboratory
enzymes from the human microbiome. Protein Sci (2014) 23:1060–76. rodents: final report of the American Institute of Nutrition ad hoc writing
doi:10.1002/pro.2493 committee on the reformulation of the AIN-76A rodent diet. J Nutr (1993)
242. D’Aimmo MR, Mattarelli P, Biavati B, Carlsson NG, Andlid T. The poten- 123:1939–51.
tial of bifidobacteria as a source of natural folate. J Appl Microbiol (2012)
112:975–84. doi:10.1111/j.1365-2672.2012.05261.x
Conflict of Interest Statement: The authors declare that the research was con-
243. de Crecy-Lagard V. Identification of genes encoding tRNA modification
ducted in the absence of any commercial or financial relationships that could be
enzymes by comparative genomics. Methods Enzymol (2007) 425:153–83.
construed as a potential conflict of interest.
doi:10.1016/S0076-6879(07)25007-4
244. Battat R, Kopylov U, Szilagyi A, Saxena A, Rosenblatt DS, Warner M, et al.
Vitamin B12 deficiency in inflammatory bowel disease: prevalence, risk Copyright © 2017 Statovci, Aguilera, MacSharry and Melgar. This is an open-access
factors, evaluation, and management. Inflamm Bowel Dis (2014) 20:1120–8. article distributed under the terms of the Creative Commons Attribution License (CC
doi:10.1097/MIB.0000000000000024 BY). The use, distribution or reproduction in other forums is permitted, provided the
245. Yakut M, Ustun Y, Kabacam G, Soykan I. Serum vitamin B12 and folate original author(s) or licensor are credited and that the original publication in this
status in patients with inflammatory bowel diseases. Eur J Intern Med (2010) journal is cited, in accordance with accepted academic practice. No use, distribution
21:320–3. doi:10.1016/j.ejim.2010.05.007 or reproduction is permitted which does not comply with these terms.