Next Article in Journal
Physicochemical and In Vitro Starch Residual Digestion Structures of Extruded Maize and Sorghum Starches Added with Sodium Stearoyl Lactylate
Previous Article in Journal
Antioxidant Activities of Dihydromyricetin Derivatives with Different Acyl Donor Chain Lengths Synthetized by Lipozyme TL IM
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Bidirectional Link between Nutritional Factors and Inflammatory Bowel Diseases: Dietary Deficits, Habits, and Recommended Interventions—A Narrative Review

by
Ilaria Maria Saracino
1,
Enzo Spisni
2,*,
Veronica Imbesi
3,
Chiara Ricci
4,
Nikolas Konstantine Dussias
5,
Patrizia Alvisi
6,
Paolo Gionchetti
5,
Fernando Rizzello
5 and
Maria Chiara Valerii
2
1
Microbiology Unit, IRCCS, Azienda Ospedaliero-Universitaria di Bologna, University of Bologna, Via Massarenti 9, 40138 Bologna, Italy
2
Department of Biological, Geological and Environmental Sciences, University of Bologna, Via Selmi 3, 40126 Bologna, Italy
3
Department of Medical and Surgical and Sciences, University of Bologna, Via Massarenti 9, 40138 Bologna, Italy
4
Gastroenterology Unit, ASST Spedali Civili di Brescia, University of Brescia, Piazza del Mercato 15, 25121 Brescia, Italy
5
IBD Unit, IRCCS, Azienda Ospedaliero-Universitaria di Bologna, University of Bologna, Via Massarenti 9, 40138 Bologna, Italy
6
Pediatric Unit, Maggiore Hospital, Largo Bartolo Nigrisoli, 2, 40133 Bologna, Italy
*
Author to whom correspondence should be addressed.
Foods 2023, 12(10), 1987; https://doi.org/10.3390/foods12101987
Submission received: 16 March 2023 / Revised: 28 April 2023 / Accepted: 8 May 2023 / Published: 14 May 2023
(This article belongs to the Section Food Nutrition)

Abstract

:
Inflammatory bowel diseases comprise Crohn’s disease and ulcerative colitis, two chronic inflammatory disorders of the digestive tract that develop in adolescence and early adulthood and show a rising pattern in industrialized societies, as well as in developing countries, being strongly influenced by environmental pressures such as nutrition, pollution and lifestyle behaviors. Here, we provide a narrative review of the bidirectional link between nutritional factors and IBD, of dietary deficits observed in IBD patients due to both the disease itself and dietary habits, and of the suggested nutritional interventions. Research of the literature was conducted. Clinical and basic research studies consistently demonstrate that diet could alter the risk of developing IBD in predisposed individuals. On the other hand, dietary interventions represent a valid tool in support of conventional therapies to control IBD symptoms, rebalance states of malnutrition, promote/maintain clinical remission and improve patients’ quality of life. Although there are no official dietary guidelines for patients with IBD, they should receive nutritional advice and undergo oral, enteral, or parenteral nutritional supplementation if needed. However, the dietary management of malnutrition in IBD patients is complex; future clinical studies are required to standardize its management.

1. Introduction

Epidemiologically we are now in the age of noncommunicable degenerative and human-influenced diseases that arise in industrialized and urbanized societies and are strongly influenced by environmental pressures such as nutrition, pollution and lifestyle behaviors. Inflammatory bowel diseases (IBD) are the epitome of these types of pathologies. IBD comprise Crohn’s disease (CD) and ulcerative colitis (UC), two chronic, inflammatory disorders of the digestive tract that develop in adolescence and early adulthood and affect about 1.5 million Americans and 2.2 million European patients [1]. IBD are characterized by an uncontrolled immune-mediated inflammatory response in genetically predisposed individuals exposed to environmental factors that include diet, medications, nicotine, infectious agents, stress, pollution and lifestyle, which together contribute to the trigger of the gut chronic inflammatory loop [2]. The disease burden is high, with prevalence surpassing 0.3% in North America, Oceania, and most countries in Europe. By contrast, countries in Africa, Asia, and South America, whose societies are becoming increasingly westernised and urbanised, are mirroring the progression of IBD in the Western world during the 1900s [3]. In an interesting perspective published in 2021 by Kaplan and collaborators [4], authors described the epidemiological stages of IBD in three main areas of the world: “developing countries”, “newly industrialized countries” and “Western world” (as defined by the United Nations) [5]. In the same paper, authors described the evolution of IBD across three distinct epidemiological stages: emergence, acceleration in incidence and compounding prevalence. Emergence is represented by sporadic cases of IBD; acceleration in incidence indicates a steadily increase in incidence, with a low prevalence [6]; compounding prevalence reflects a steady increase in IBD prevalence, despite a lowering or stabilization in incidence [7]. These stages reflect the nature of IBD: a chronic disease that can occur in individuals of any age, but is most commonly diagnosed in adolescents and young adults (ages 18–35 years) who have a long life expectancy [6,8]. The first burden in the world evolution of IBD occurred in North America and Western Europe following the advent of the industrial revolution [4], and by the 1950s, the Western world was shifting to the second epidemiological stage that spanned the second half of the 20th century [6]. Most countries of the Western world transitioned into the third stage in the evolution of IBD by the end of the 20th century, with a concomitant increase in pediatric IBD [9]. In the same period, newly industrialized countries in Asia and Latin America entered the second stage; if these regions will follow the epidemiological patterns of the Western world, they are likely to transit into the third stage over the next three decades. Developing countries are currently in the first stage of evolution and can expect to enter in the second stage if their societies westernize. Analyzing epidemiological data and making appropriate projections, it has been estimated that over the next decade, the prevalence of IBD in the Western world will be very close to 1% [4]. Several reviews have compared the environmental determinants of IBD in the Western world with those of newly industrialized countries [10,11,12]. For example, over the past 30 years, China experienced a massive population shift from rural communities to megacities, and inhabitants in these urban environments have been exposed to the westernization of their society including diets, pollution and lifestyle behaviors [13]. As a result of that, some regions in China have already reached IBD incidence values that are close to those of the Western world. Moreover, many studies have concluded that people emigrating from low-prevalence regions such as Asia to high-prevalence countries such as northern Europe are at greater risk of developing IBD. This is especially true for first-generation immigrant children who show similar IBD incidence and prevalence patterns to the indigenous population [14].
The rising pattern of IBD has provided a unique opportunity for researchers to focus on identifying the environmental risk factors contributing to its pathogenesis. Among them, until now, the most consistent results have been obtained for the correlation between diet and IBD prevalence, although it cannot be excluded that environmental pollutants will be identified as major determinants in the future.

1.1. Diets: The Good and the Bad

Mediterranean, Indian, Japanese and Southeast Asian traditional diets are all considered as healthy diets. In the 1960s, the term “Mediterranean diet” was introduced by the physiologist Ancel Keys, defining the diet followed by populations living along the Mediterranean Basin. These populations showed a lower mortality rate and incidence of cardiovascular disease than other populations. The Mediterranean diet (Figure 1) has been proven to enrich beneficial gut bacteria, support gut barrier function and reduce inflammation, thus acting as a preventive factor for the onset of IBD [15,16,17]. Traditional Indian [18], Japanese [19] and Southeast Asian [20] diets are basically similar to the Mediterranean diet, relying on seafood and plant foods such as vegetables, legumes, fruits, herbs and spices, as well as on a low consumption of meat, sugar and highly processed foods. These diets are naturally rich in minerals (e.g., calcium, potassium, iron, magnesium), vitamins, fiber and long-chain omega-3 fatty acids. Spices not only enhance the flavor and taste of foods, but are also considered as functional foods carrying biologically active phytochemicals such as curcumin, gingerols and essential oils, all known for their anti-inflammatory, antibacterial and anticancer properties [21].
A Western-style dietary habit (Figure 1) is instead considered an important risk factor for IBD [17]. The so-called Western diet is characterized by the consumption of high amounts of animal proteins, saturated fats and processed foods (rich in additives, emulsifiers and food preservatives) and low amounts of vegetables, unsaturated fats, fibers and fruits. The Western diet is also characterized by an unbalanced ratio of polyunsaturated fatty acids (n-3/n-6 PUFAs). Linoleic acid (a n-6 PUFA) consumption has markedly increased during the 20th century [22] and is associated with an increased risk of UC, whereas oleic acid (n-9 monounsaturated fatty acid or MUFA) or n-3 PUFAs consumption is beneficial. The low intake of fruit and vegetables is detrimental too, since it is well known that a long-term high intake of dietary fiber reduces CD risk [23,24,25,26].

1.2. FAO Survey on the “Nutrition Transition”

In the survey of food consumption, FAO addressed the “nutrition transition” in the world, mainly in developing countries, stating that “it is contributing to the causal factors underlying noncommunicable diseases even in the poorest countries”. The adverse dietary changes include shifts in the structure of the diet towards a higher energy density food with a greater role for fat and added sugars, greater saturated fat intake (mostly from animal sources), reduced intakes of whole grains and dietary fibers, and reduced fruit and vegetable intakes. Additionally, the increase in urbanization increased the distances of people from primary food production, leading to the use of processed long-life foods. This nutritional shift has taken place in South America (especially Brazil), East Asia (especially China) and the Near East/North Africa region, while some parts of Southeast Asia and Sub-Saharan Africa are still basically tied to traditional low-calorie, low-fat diets [27,28].
All these observations are consistent with the IBD epidemiological map (Figure 2).
Here, we provide a narrative review of the bidirectional link between nutritional factors and IBD, of dietary deficits observed in IBD patients due to both the disease itself and dietary habits, and of the desirable nutritional interventions. Because there are still no validated tools for screening malnutrition in patients with IBD and no standardized nutritional regimen for these patients, it is important to summarize the available studies and their results in order to increasingly refine these aspects of IBD patient management.

2. Materials and Methods

In order to provide an overview of research focused on diet, malnutrition and nutritional interventions in IBD patients, literature research was conducted on PubMed Central with “advanced” and “MeSH” tools, and links for articles were accessed with University of Bologna login.
Term queries: (((inflammatory bowel disease) AND (diet*ther* OR nutri*inter*)) OR ((inflammatory disease AND (nutri*deficienc* OR malnutrition))); (((inflammatory bowel disease) AND ((diet) OR (western diet) OR (nutri*))). Inclusion criteria were as follows: clinical trial, observational study (for the food habits-related causes of malnutrition), basic research (only for the latter term query), English language, full text available, published in the last 10 years (2012–2022). Exclusion criteria were as follows: clinical case studies, letters or editorials of magazines, abstracts to conferences, book chapters. The Grading of Recommendations Assessment, Development and Evaluation (GRADE) approach was not used in the study selection process. The final search included 155 articles, from which 83 abstracts were selected. From these abstracts, the 57 most relevant studies were selected for the complete text reading and analyzed in this review. The results are exposed in three main areas: (1) food-related pathogenetic molecular mechanisms—dietary risk factors, dietary protective factors; (2) malnutrition in IBD—macro and micronutrient deficiency prevalence in IBD; disease-related causes of malnutrition in IBD, food-habits-related causes of malnutrition in IBD; (3) nutritional interventions in IBD. PRISMA flow diagram is reported in Figure 3.

3. Results

3.1. Food-Related Pathogenetic Molecular Mechanisms of IBD

In IBD, altered gut microbial signatures have been consistently reported, and there is little doubt that industrialization goes along with alteration of the intestinal microbial ecology in humans. For example, metagenomic studies in stool specimens of IBD patients clearly indicate a reduction in ecological diversity with a lower proportion of Firmicutes and an increased abundance of Proteobacteria and Bacteroidetes [29]. Dietary habits may result in an altered susceptibility to IBD in a variety of ways; it has been proven that food components can modify intestinal permeability (e.g., short chain fatty acids—SCFAs, vitamin D, vitamin A, zinc, cysteine, methionine, glutamine, tryptophan and arginine decrease intestinal permeability, while gluten, glucose, fructose, fats, ethanol and emulsifiers increase it) and can serve as ligands for various receptors expressed in enterocytes (e.g., farnesoid X receptor, aryl hydrocarbon receptor, pregnane X receptor, and specific G protein-coupled receptors) [30,31]. A family of intracellular danger-sensing molecules called NLRs (NOD-like receptors) has been implicated in the pathogenesis of IBD. Within this family are the NOD (nucleotide-binding oligomerization domain) and NLRP (pyrin domain-containing NLR) subfamilies. The link between NOD2 mutation and CD has been well established, and it is the most common genetic variant associated with CD [32]. In addition, alterations in another family of bacterial sensor genes, the Toll-like receptors (TLRs), have been well described in IBD. It is noteworthy that the Western diet increases both NOD-2 and TLR-5 mRNA levels in experimental CD models [33].
The role of diet in shaping gut microbiota is well established. Thus, interactions between intestinal microbes and food can in turn influence IBD pathogenesis. Defective innate immunity could contribute to IBD pathogenesis by enabling a tolerance breakdown to a normal intestinal microbiota in genetically predisposed individuals. These findings demonstrate the innate immunity control of the adaptive immune response and the crucial role of the gene–environment interplay in IBD induction. The mucosal surface of the intestine plays a key role in the control of the host immune response by acting as a barrier against bacterial and dietary antigens, and it is strongly influenced by nutrients [34] (Figure 4).

3.1.1. Dietary Risk Factors

The Western diet is enriched with sugars, fat (e.g., saturated, polyunsaturated n-6 fatty acids and cholesterol) and food additives (e.g., emulsifiers, colourants, preservatives, flavoring and processed carbohydrates). These latter compounds may act as xenobiotic into the gut and directly induce compositional and functional alterations of the gut microbioma, which impairs epithelial functions in the gut, that is, perturbs Paneth cells and the gut barrier homeostasis [35]. An excessive intake of refined sugars promotes dysbiosis and gut inflammation, but their role of in the development or progression of IBD has been poorly explored yet [29]. Emerging evidence indicates that the consumption of food additives perturbs microbial composition and promotes experimental gut inflammation; for example, some artificial sweeteners promote dysbiosis (with increased Bacteroides and reduced Lactobacillus spp.) [36]; likewise, emulsifiers perturb gut microbial community structure and promote susceptibility to gut inflammation. Maltodextrins are common dietary polysaccharides used as emulsifiers or energy source, and their role in the impairment of defense mechanisms and in the overgrowth of harmful bacteria (i.e., E. coli and Salmonella spp.) has been demonstrated in both in vitro and in vivo studies [37]. Furthermore, the administration of emulsifiers at a low concentration leads to low-grade inflammation and obesity/metabolic syndrome in wild-type mice, while it promotes colitis in IL-10−/− mice [38]. Finally, food colourants Red40 (E129) and Yellow 6 (E110) metabolites (produced by commensals) promote colitis in mouse models [39].
A high red meat consumption is also a hallmark of the Western dietary pattern associated with an increased IBD risk [40]. The relative abundance of bile-tolerant anaerobes such as Bacteroides, Alistipes and Bilophila increased following consumption of animal-based protein, especially red and processed meat [41]. Dietary animal proteins enhance the sensitivity to experimentally induced gut inflammation, possibly by expansion of inflammatory genera such as Escherichia, Streptococcus and Enterococcus. Total fat and saturated fat suppressed richness and diversity of the gut microbiota, increasing anaerobic abundance [42]. For this reason, the high fat content of Western diet is believed to be a key factor in causing intestinal dysbiosis, [43] which promotes gut inflammation. It is noteworthy that obese patients have a more severe IBD course [44], which confirms the link between diet, obesity, activation of the innate immune system, inflammation, and organ dysfunction. Unsaturated fatty acids are essential components of the intestinal inflammatory response; indeed, they modulate cell membrane composition with a subsequent modification of the activation of receptors, for example, TLR-4 activation is facilitated by saturated fatty acids and inhibited by n-3 PUFAs (such as docosahexaenoic acid) [45]. Unsaturated fatty acids can also act as signaling molecules, targeting numerous membrane and nuclear receptors, and finally altering cellular gene expression in adipocytes, tissue macrophages and endothelial cells [46]. In conclusion, a Western dietary pattern may trigger a proinflammatory environment into the gut of susceptible individuals through alterations of the gut microbiome (leading to an upregulation of the TLR- and NOD-pathways) and the consequent impairment of the intestinal epithelial barrier functions. [18,47].

3.1.2. Protective Dietary Factors

Unrefined complex carbohydrates rich in fibers (typically whole grains), and their bacterial metabolites exert a protective effect on the gut. Bacterial short chain fatty acids and especially butyrate maintain gut homeostasis by protecting intestinal barrier integrity and host immune responses. SCFAs indeed stabilize HIF-1 a transcription factor coordinating barrier protection [48], and probiotic supplementation of butyrate-producing bacteria improves epithelial barrier integrity in CD [49]. Moreover, butyrate exerts anti-inflammatory effects in the gut mucosa by inhibition of histone deacetylases and activation of G protein-coupled receptors present in gut epithelium and mucosal immune cells [50]. Glutamine and arginine are non-essential amino acids, but they are considered essential in stress situations, and both demonstrated anti-inflammatory properties in IBD models [51,52]. Glutamine is the preferential substrate for enterocytes, and its effect has been evaluated in numerous experimental models of gut barrier dysfunction. For instance, Zhang and colleagues [53] showed that glutamine supplementation was able to improve the growth performance of LPS-challenged broilers, alleviate the inflammatory response, reverse the deleterious effects of LPS on intestinal permeability and the integrity of intestinal mucosa barrier, downregulating the expression of molecules involved in TLR4/FAK/MyD88 signaling pathways [53]. Intraperitoneal injection of alanyl-glutamine suppressed Th17 cytokine production, reduced luminal chemokine secretion, and decreased macrophage infiltration to the peritoneal cavity in DSS colitis models [54]. Treatment with arginine or isoleucine also upregulated in vitro the secretion of human beta-defensin-1 [hBD1], an antimicrobial peptide capable of counteracting the microbial overgrowth of pathobiont species [55]. The amino acid tryptophan has also been involved in host–microbiome interactions. Tryptophan and its metabolites regulate intestinal homeostasis; for example, it has been observed that Lactobacillus spp. are able to switch their fuel from sugar to tryptophan to produce indoles [56]. The aryl hydrocarbon receptor is expressed on intestinal dendritic cells and lymphocytes and helps to maintain epithelial integrity via IL-22 secretion; this receptor has many ligands, some of which are present in foods. These include indole or tryptophan metabolites (derived from cruciferous vegetables), stilbenes (e.g., resveratrol in grape), carotenoids (present in yellow/orange/red vegetables) and flavonoids [56,57,58]. In fact, some phytochemicals are known to positively influence intestinal physiology, e.g., Curcumin (Curcuma longa) has shown anti-inflammatory properties in different experimental models of IBD [59] and in human clinical trials [60]. One of the proposed mechanisms for different phytochemicals is the inhibition of TLR-4 activation [61]. It has been demonstrated that a ginger compound, 1-dehydro- 10-gingerdione, acts on TLR-4 expression in macrophages, inhibits LPS binding to a TLR-4 co-receptor and downregulates TLR-4–mediated expression of NF-κB and the expression of inflammatory cytokines in LPS-treated macrophage cell lines [62].
Studying the effects of natural compounds on murine models of colitis and on mice subjected to high-fat diet (HFD), our team focused on the effects of geraniol, an acyclic monoterpene present in high percentages in citronella, rose and palmarosa essential oils. In mice with DSS-induced colitis, geraniol intake resulted in a strong reduction in weight loss, an improvement in stool consistency and stool blood content. Geraniol administered orally halved weight loss in mice and reduced the colitis activity index; geraniol administered by enema improved colitis symptoms by preserving the integrity of the colonic mucosa. These clinical observations were further supported by a significant reduction in mRNA expression of the pro-inflammatory enzyme COX-2 in the colic mucosa of geraniol-treated mice [63]. In HFD-fed mice, a food supplement based on D-Limonene (a monoterpene commonly found in the essential oils of Citrus plants), counteracted the metabolic negative effects of the diet by positively modulating the gut microbiota [64]. So, it can be stated with reasonable confidence that many different specific factors in food have direct effects on mucosal integrity and immune function, and these effects may overall be enclosed in the concept of nutrient signaling. In addition to these mechanisms, that may explain the effects of different nutrition patterns on IBD, diet is a major determinant for the intestinal microbiome and metabolome. It is not clear whether microbiome changes are the cause or an effect of IBD, but it is a matter of facts that the features of a Western diet pattern including higher caloric intake and consumption of sugar-sweetened beverages are negatively associated with microbiome diversity. In contrast, the features of a Mediterranean style diet with a higher consumption of fruits, vegetables and phytochemicals have been associated with an increased gut microbial diversity. The results of these studies have been relatively consistent, pointing to a lower risk of IBD among people who consume more fruits and vegetables, and a higher risk in people who consume more animal fats and refined sugars [65,66,67].

3.2. Malnutrition in IBD

There are two main definitions of malnutrition: the European Society for Clinical Nutrition and Metabolism defines it as a state resulting from a lack of nutrient intake or absorption, leading to altered body composition (decreased body fat mass and cellular mass) resulting in decreased physical and mental function and impaired clinical outcomes of disease [68]; the World Health Organization (WHO) defines malnutrition as a deficiency, excess or imbalance in a person’s energy/nutrient intake [69].
The prevalence of malnutrition among patients with IBD can vary from study to study, depending on the definition adopted and on the different screening tests used [70] and ranged from 20% to 85%. Malnutrition is more present in CD than in UC, probably because CD can affect any part of the gastrointestinal tract including the small bowel where most of the nutrient absorption occurs [71,72]. Malnutrition has been correlated with poor clinical outcome and quality of life in IBD patients and has also been documented in patients in clinical remission [73]. The main micronutrient deficiencies in patients with IBD are shown in Table 1. The mechanisms responsible for nutritional deficiencies are not always clear and may be related to the disease itself, which causes malabsorption, but also to increased metabolic demand (related to the active inflammatory process), and/or dietary habits of IBD patients, which influence their nutrient intake [74,75,76].

3.2.1. Disease-Related Causes of Malnutrition

The nutritional status of patients with IBD can be altered due to the high catabolic load that can occur during the acute inflammatory phases of the disease. Inflammatory cytokines stimulate the release of glucagon, catecholamines and cortisol. This condition, without an adequate supply of nutrients, can lead to loss of weight, due to decreased subcutaneous adipose tissue and muscle mass [77].
Proinflammatory cytokines can also act as anorectic agents at the level of the intestine, where IL-1β is able to stimulate cholecystokinin release and potentiate its action [78], or in hypothalamic areas where TNF-α modulates glucose-sensitive neurons [79,80] affecting eating behavior [79,81,82].
These effects are consistent with the decreased appetite often experienced by IBD patients during the acute phase [83,84]. In addition to mechanisms directly stimulated by the inflammatory response, other common causes of malnutrition in IBD are gastrointestinal ulcerations and/or fistulas that lead to a functional or anatomical reduction in the gut absorbent surface [85]. Diarrhoea may cause a depletion of potassium, magnesium, phosphorus and zinc [86,87], while intestinal hemorrhages may cause iron deficiency [72,88]. Drugs used for the management of IBD such as sulfasalazine and methotrexate can cause a folic acid deficiency [89]. While in UC nutritional deficiencies are mainly related to diarrhoea and bleeding, in CD, surgeries, when the terminal ileum is extensively resected, can cause a significant reduction in the intestinal absorption surface area affecting nutrient assimilation including fats and fat-soluble vitamins (A, D, E, K) [90,91,92,93,94]. In CD patients, the reabsorption of bile salts could be impaired [95], and small intestinal bacterial overgrowth (SIBO) is a condition that often develops [96]. SIBO contributes to fat and vitamin B12 malabsorption due to bile acid deconjugation by luminal bacteria [97] and vitamin B12 consumption by bacterial metabolism [98].

3.2.2. Food Habits-Related Causes of Malnutrition

The adoption of unbalanced diets by IBD patients is mainly attributable both to the Western diet pattern and to specific patient choices in the belief that certain food restrictions can prevent disease exacerbations or improve symptoms in the acute phase. Based on epidemiological studies, 48 to 51% of IBD patients believe that diet can initiate the disease [83,99,100], while 62 to 89% of patients self-impose dietary restrictions to try to avoid relapse or control symptoms [83,84,99,100]. Spices, fibers (fruit and vegetables), alcohol, carbonate beverages, milk and dairy products are the most commonly avoided foods by IBD patients, especially in the acute phase [83,84,99,100]. Furthermore, in the study conducted by de Vries and colleagues in 2019 [99], 30% ca. of IBD patients stated that they had never received any nutritional advice and that they preferred to eliminate certain elements rather than eat beneficial foods or follow a specific diet; moreover, among the patients who had received nutritional advice from professionals, 81% still based their restrictions on their own experience.
In a study conducted by Crooks and colleagues, one third of the patients interviewed reported having tried a specific exclusion diet such as a lactose-free, gluten-free, low FODMAPS (Fermentable Oligo-, Di-, and Monosaccharides and Polyols) diet, a specific carbohydrate exclusion diet, an anti-inflammatory diet and paleo diet [100]. The tendency of patients to self-manage their diet in the context of an extremely debilitating disease may have a negative impact on the development of eating disorders and certainly may worsen their nutritional status.
In a review published by Peters and collaborators in 2022 [101], authors found that the prevalence of eating disorders in patients suffering from gastrointestinal diseases ranged from 13 to 55%. Factors associated with these disorders included sex (female), younger age, gastrointestinal symptom severity, anxiety, depression and lower quality of life. In particular, the EAT−26 (eating attitude test-26) and BES (binge eating scale) scores were both higher in IBD vs. healthy subjects (20% and 25% above clinical threshold vs. 4% and 2%, respectively). Two studies have assessed the nutritional status of IBD patients undergoing food restriction: the first, published in 2013, was performed on 59 UC patients (56 of whom were in remission) and showed an inadequate intake of fiber, fat-soluble vitamins (vitamin A and E), vitamin C and minerals (calcium and magnesium), linked to the low-residue diet that is often followed during the acute phase, but also maintained in the remission phase [102]; the second study published in 2019 considered 104 IBD patients with both active disease and in remission, documenting severe malnutrition in 12.2% of patients on self-prescribed exclusion diets vs. 5.5% of patients on free diets [103]. Self-prescribed dietary restrictions were also associated with an inadequate calcium intake and low bone mineral density (in 80% and 51% of IBD patients, respectively) in a study performed on 90 patients [104].
Another study conducted on 54 CD patients with active disease and 30 healthy subjects reported that the former had lower intakes of fiber, vitamins (A, E, C, B6, folic acid) and β-carotene, as well as calcium, potassium, phosphorus, iron, magnesium, copper, and iodine. These patients also showed a lower dietary oxygen radical absorbance capacity (ORAC) linked to low consumption of fruits and vegetables, and a higher renal potential load (PRAL) linked to high consumption of red meat. In addition, blood concentrations of total cholesterol, potassium, iron and amino acids were significantly lower in CD patients compared to healthy subjects [105]. A similar study in pediatric CD patients in clinical remission showed lower fiber and vitamin A intake, along with higher animal protein consumption; also in this study, ORAC was lower, while PRAL was higher in CD patients compared to healthy subjects [106]. These data are consistent with epidemiological studies showing that parents of pediatric IBD patients pay particular attention to highly processed foods, fast foods, spicy foods and dairy products, but little attention to meat consumption, with a tendency to believe that fruits and vegetables may have a negative impact on the disease course [107,108].

3.3. Nutritional Interventions

In recent years, increasing attention has been paid to the study and validation of nutritional protocols aimed at inducing remission or symptom control in pediatric IBD patients in an attempt to avoid or delay the use of drug therapies such as corticosteroids or biologics. Specific carbohydrate diets (SCDs), Mediterranean (MD) or Mediterranean-like (MD-like) diets, exclusion diets (EDs) and low FODMAPS diets have been tested in clinical controlled trials on IBD patients, with promising results (Table 2).
The SCD was introduced way back in 1920 [109] for the management of celiac disease. It is based on the exclusion of refined sugars and the full restriction of complex carbohydrates including gluten-containing ones. To date, this diet is considered effective not only for the management of celiac disease, but also for IBD [110]. Small, controlled studies in pediatric IBD patients have shown encouraging results: two prospective studies evaluated the SCD in pediatric IBD patients with active disease, achieving clinical remission [111] and the decrease in both Harvey–Bradshaw Index (HBI) and Pediatric Crohn’s Disease Activity Index (PCDAI) [112].
A randomized controlled trial compared the efficacy of SCD, a modified version of SCD (MSCD, which includes consumption of oats and rice) and of the Whole Food diet (WFD, which excludes wheat, corn, sugar, milk and food additives) in achieving the clinical remission [113]. All three diets have been shown to induce clinical remission. Additionally, two retrospective studies, in which mainly CD patients with active disease were enrolled, showed clinical remission [114] or strong improvement in disease activity indices induced by the experimental diets (including MSCD and WFD) in almost all enrolled pediatric patients [115]. Different studies were performed on the effects of the Mediterranean diet (MD) in the management of IBD. A randomized trial on 194 IBD patients compared the SCD with the Mediterranean diet, showing very similar remission rates between the two diets [116]. Another study was conducted on the effects of MD on 184 adult patients (84 CD and 58 UC) with mild to moderate disease or in remission, monitored for six months; in addition to improvements in anthropometric and metabolic parameters, out of the 14 patients with active disease and stable therapy, 10 achieved remission and 4 achieved a significant decrease in disease severity, which shifted to a milder form [117]. In other studies, nutritional interventions were based on modified MD diets, characterized by the typical features of MD (MD-like diets); in particular, a randomized trial carried out on 7 CD patients with an active disease compared a high-fiber MD-like diet (HF, in which specific portions of whole grains were prescribed to patients) with an MD-like exclusion diet characterized by the elimination of whole grains, dairy products and spicy foods. The HF group showed better performances, with a decrease in Harvey–Bradshaw index score maintained after 4 weeks of follow-up [118]. In 17 patients with UC in remission, a MD-like low-fat, high-fiber diet (LFHF) was compared with an improved standard American diet characterized by a higher intake of vegetables, fruits, fish and dietary fiber. Results demonstrated that only the LFHF group showed a reduction in inflammatory markers after 4 weeks of the diet [119]. In a broader study, conducted on 214 subjects with CD in remission, patients were randomized to high consumption of red/processed meat (2 servings per week of red or processed meat) or a low meat MD-like diet (no more than 1 serving per month) and were monitored for 49 weeks; the disease relapse rate was 62% in the high meat consumption group versus 42% in the low meat consumption group [120]. Two types of exclusion diets have been tested on IBD patients: the IgG4-based exclusion diet and the Crohn’s disease exclusion diet (CDED). The IgG4 exclusion diet is based on the exclusion of four foods depending on IgG4 titration and is patient-specific; this diet was tested on CD patients with an active disease and controlled with a sham diet. After 4 weeks, there was an overall improvement in Crohn’s disease activity index in the group eating the IgG4-driven diet [121]. However, IgG4 positivity in blood seems to correlate with food exposure. Following an IgG4-driven diet primarily means changing eating habits, with a tendency towards a sharp reduction in the foods most frequently consumed.
CDED is a diet based on the elimination of specific foods considered “proinflammatory” and the introduction of mandatory or recommended “anti-inflammatory” foods. CDED was tested alone in 32 CD adult patients, showing an 82.1% efficacy in inducing clinical remission after 12 weeks [122], or combined with partial enteral nutrition (PEN) in a randomized trial comparing CDED vs. CDED + PEN, with results showing similar efficacy [123]. CDED+PEN showed a similar efficacy if compared to the exclusive enteral nutrition in a six-week study also on pediatric patients [124], but a superior efficacy was detected in a 12-week study carried out by Levine and colleagues [125]. Enteral nutrition is based on elemental (amino acid-based), semi-elemental (oligopeptides) and polymeric (whole protein-based) formulas that can be administered to IBD patients as a complete meal replacement (exclusive enteral nutrition) or in addition to standard meals (partial enteral nutrition). Its efficacy in inducing and maintaining remission has been supported by clinical trials and meta-analyses, despite the heterogeneity of treatment protocols [126]. One explanation for enteral nutrition efficacy is that the polymeric formulation could minimize antigen exposure in the gut resulting from food ingestion [127]; however, whether administered via nasogastric tube or ingested directly, enteral nutrition diets are often poorly tolerated by patients and can be used only for short periods of time. The combination of CDED and PEN, however, appears to be better accepted by patients [125].
The low oligo-, di-, monosaccharides and fermentable polyols (FODMAP) diet is characterized by the exclusion of short-chain fermentable carbohydrates, which are poorly digested by gut enzymes and thus predominantly fermented at the level of the intestinal microbiota. FODMAP content is high in fruits, vegetables, legumes, dairy products and wheat. FODMAP diet has been used with some success in patients with irritable bowel syndrome, a functional disorder characterized by abdominal pain, bloating and altered bowel habits [128]. Low FODMAP diet has been tested in several clinical trials in patients with IBD in remission or with a mild disease meeting the criteria for functional gastrointestinal disorders. In almost all studies, patients with CD or UC showed a general improvement in gastrointestinal symptoms [129,130,131,132,133], but there are insufficient data to reach conclusions about its efficacy in IBD management. Despite a rather large number of diets tested on rather limited numbers of IBD patients, these different diet regimens share some common features such as the exclusion of red and processed meats; reduction in saturated fats, sugars and processed foods; and an increase in the consumption of fiber, fruits and whole raw foods. Overall, these data provide clear guidance on the type of diet that should be recommended for patients with IBD, even when they are not inserted into specific dietary plans.
Table 2. Interventional studies on IBD patients.
Table 2. Interventional studies on IBD patients.
Low FODMAPS Diet
AuthorsStudy
Design
Dietary
Intervention
Patient
Number
Clinical ActivityDays
of Treatment
Results
Cox et al., 2017 [129].Randomized, double blind, placebo controlled, crossover.LFD + 4 reintroduction challenge of fructans or galacto-oligosaccharides or sorbitol or placebo, each challenge preceded by 4 days of wash out.12 CD
17 UC
Remission. All patients also met criteria for FGS.3 days challenge.Fructane reintroduction induced a worsening of FGS.
Bodini et al., 201. [130].Randomized.LFD
vs.
Free diet
35 CD
20 UC
Remission/mild disease. All patients also met criteria for FGS.6 weeksDecrease in HBI index and calprotectin but not in Mayo score in LFD group.
Pedersen et al., 2017 [132].Randomized.LFD
vs.
Free diet
61 UC
28 CD
Remission/mild disease. All patients also met criteria for FGS.6 weeksDecrease in IBS-SSS score in LFD, decrease in SCCAI in UC LFD Group, no change in HBI score.
Cox et al., 2020 [131].Randomized.LFD
vs.
Free diet
26 UC
26 CD
Remission. All patients also met criteria for FGS.4 weeksDecrease in IBS-SSS score in UC patients but not in CD patients.
Melgaard et al., 2022 [133].Randomized, blinded placebo controlled.LFD
+
reintroduction challenge
16 UCRemission. All patients also met criteria for FGS.8 weeks LFD + 2 weeks Low FODMAPs or PlaceboNo effect on IBS-SSS score.
Enteral Nutrition
AuthorsStudy designDietary
Intervention
Patient numberClinical activityDays
of treatment
Results
Guo et al., 2013 [134].Pilot study.EEN13 CDActive.4 weeksClinical remission in 86% of patients.
Pigneur et al., 2019 [135].Randomized.EEN
vs.
steroids
19 CD, pediatricsActive.8 weeksSame efficacy in inducing remission, 89% of mucosal healing in EEN arm, 17% of mucosal healing steroids arm.
Brückner et al., 2020 [136].Open labelPEN
vs.
free Diet
41 CD, pediatrics.Remission/mild.12 monthsGrowth improvement.
Moriczi et al., 2020 [137].Retrospective.EEN235 CD, pediatrics.Active.8 weeks83% clinical remission.
Specific Carbohydrate Diet (SCD)
AuthorsStudy designDietary
Intervention
Patient numberClinical activityDays
of treatment
Results
Lewis et al., 2021 [116].Randomized.SCD
vs.
MD
194 CDMild/
moderate.
6 weeksClinical Remission: 46.5% SCD, 43.5% MD.
Suskind et al., 2020 [113].Randomized.SCD
vs.
Modified SCD
vs.
WFD
18 CD, pediatrics.Mild/
moderate.
12 weeksClinical remission in all arms.
Braly et al., 2017 [111].Prospective, open label, non-controlled.SCD9 CD/UC, pediatrics.Mild/
moderate.
12 weeksClinical remission.
Suskind et al., 2014 [114].Retrospective.SCD7 CD, pediatrics.Active.3 monthsClinical remission.
Obih et al., 2016 [115].Retrospective.SCD20 CD
6 UC
pediatrics
Active.6 monthsClinical remission in CD patients, decrease in PUCAI in UC patients.
Cohen et al., 2014 [112].Prospective,
non controlled.
SCD10 CD, pediatricsActive.12 weeksDecrease in HBI and PCDAI.
Mediterranean Diet/Mediterranean-like
AuthorsStudy
Design
Dietary
Intervention
Patient numberClinical
activity
Days
of treatment
Results
Chicco et al., 2021 [117].Prospective,
non controlled.
MD84 UC,
58 CD
Mild/moderate and remission.6 monthsRemission in patients with active disease.
Brotherton et al., 2014 [118].Randomized, controlled, single blind.HF
vs.
Exclusion Diet
7 CDActive.4 weeksDecrease in HBI score in HF group.
Albenberg et al., 2019 [120].Randomized.HM
vs.
LM
214 CDRemission.49 weeksRelapse in 62% HM group, 42% LM group.
Fritsch et al., 2021 [119].Randomized, crossover.LFHF
vs.
iSAD
17 UCRemission/mild.4 weeksLFHF decreased markers of inflammation.
Exclusion Diets
AuthorsStudy
Design
Dietary
Intervention
Patient numberClinical activityDays
of treatment
Results
Guasekeera et al., 2016 [121].Randomized, controlled.IgG4-guided diet
vs.
sham diet
98 CDActive.4 weeksImprovement of CDAI in the IgG4-guided diet.
Szczubełek et al., 2021 [122].Prospective, non-controlled.CDED32 CDActive.12 weeksClinical remission in 82.1% of cases.
Yanai et al., 2022 [123].Randomized, open label.CDED + PEN
vs.
CDED
44 CDMild/
moderate.
24 weeksClinical remission in 68% of patients undergoing CDED+ PEN, and in 57% of patients undergoing CDED.
Levine et al., 2019 [125].Randomized.PEN+CDED
vs.
EEN followed by PEN
78 CD, pediatric.Mild/
moderate.
12 weeksRemission: 75,6% in CDED +PEN arm, 45.1% EEN+ PEN arm.
Sigall et al., 2021 [124].Randomized.EEN
vs.
PEN + CDED
73 CD, pediatric.Mild/
Moderate.
6 weeksRemission: 61.5% in CDED+PEN arm, 64.7% in EEN arm (at week 3).
LFD: Low FODMAPs Diet; EEN: Exclusive Enteral Nutrition; PEN: Partial Enteral Nutrition; SCD: Specific carbohydrate diet; MD: Mediterranean diet; WFD: Whole Food Diet; HF: High Fiber, HM: High in take in red Meat; LFHF: Low Fat High Fiber, iSAD: improved Standard American, CDED: Crohn’s Disease Exclusion Diet. FGS: Functional Gastrointestinal Disorders. HBI: Harvey-Bradshaw Index; IBS-SSS: Irritable Bowel Syndrome -Severity Scoring System; SCCAI: Simple Clinical Colitis Activity Index; PUCDAI: Pediatric Ulcerative Colitis Activity Index; PCDAI: Pediatric Crohn’s Disease Activity Index; CDAI: Crohn’s Disease Activity Index.
The main features of nutritional interventions that have been implemented in IBD patients are shown in Table 3.

4. Conclusions

The rapidly increasing incidence of IBD and other immune-mediated diseases points to a role of environmental factors in their pathogenesis; in particular, there is growing evidence that diet plays a key role in the development of IBD. The complexity of the human diet makes it very difficult to identify single foods as risk factors; indeed, it is more likely that complex nutritional patterns may have an impact on the pathogenesis and clinical course of IBD. In evolutionary terms, people in industrialized countries can be described as nutritionally maladapted and thus more susceptible to many diet-related and gut-related noncommunicable diseases, including IBD.
Once the disease has developed, IBD patients with active disease tend to adopt unbalanced diets, probably as an indirect response to their symptoms, that contribute to a reduced intake of certain nutrients with antioxidant, anti-dysbiotic and anti-inflammatory effects. Nevertheless, most of their nutrient deficiencies depend on impaired intestinal absorption caused by the disease itself. However, the role that diet can play in supporting conventional therapies for IBD is too often underestimated since it may help to induce and maintain clinical remission, but also improve patients’ quality of life. As a matter of fact, there are clear indications that diet can significantly modulate disease onset and activity, but unfortunately, there are no specific dietary guidelines for patients with IBD. This leaves the choice to the patients for choosing do-it-yourself diets which tend, in general, to reduce protective foods such as those of vegetable origin in favor of meat, especially red ones. IBD patients should be referred to a nutritionist for a counseling program soon after diagnosis to avoid do-it-yourself attempts to improve their nutrition. The diet model to be preferred for IBD patients should be a Mediterranean or Mediterranean-like diet including the traditional Asian ones, with a strong presence of raw foods of vegetable origin, fish and a reduced intake of meats (especially red and processed) and industrialized processed foods. In patients in whom the response to this type of diet is not satisfactory, an approach based on CDED + PEN or CDED alone can be adopted, certainly more complex to implement, but which seems to have greater efficacy in inducing remission or in maintaining it.
IBD patients, being at significant risk for malnutrition, should be screened for nutrient deficiencies using validated tools, and should receive nutritional advice and undergo oral, enteral or parenteral nutritional supplementation. However, the management of malnutrition in IBD is complex, and future clinical studies are needed to provide guidance to standardize the diagnosis and management of malnutrition in these patients.

Author Contributions

Conceptualization, E.S., M.C.V., I.M.S. and C.R.; methodology, M.C.V., I.M.S. and V.I.; validation, E.S., F.R. and P.G., data curation, M.C.V., I.M.S. and E.S.; writing—original draft preparation, M.C.V., I.M.S. and E.S.; writing—review and editing, M.C.V., I.M.S., E.S., V.I., N.K.D., P.A., C.R., F.R. and P.G.; supervision, E.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Data Availability Statement

Data available in a publicly accessible repository. The data presented in this study are openly available in the Pub Med database.

Acknowledgments

The authors received no financial support for the research or publication of this article.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

IBD: inflammatory bowel diseases, CD: Crohn’s disease; UC: ulcerative colitis; PUFAs: polyunsaturated fatty acids; MUFAs: Monounsaturated fatty acids; FAO: Food and Agriculture Organization of the United Nations; SCFAs: short chain fatty acids; NLRs: NOD-like receptors; NOD: nucleotide-binding oligomerization domain; NLRP: pyrin domain containing NLR; TLRs: Toll-like receptors; LPS: Lipopolysaccharide; DSS: Dextran sulfate sodium; hBD1: human beta-defensin- 1; IL: interelukine; NF-κB: nuclear factor kappa-light-chain-enhancer of activated B cells; HFD: High Fat Diet; COX-2: Cyclooxygenase-2; WHO: World Health Organization; GI: gastrointestinal; FODMAPs: Fermentable Oligo-, Di-, and Monosaccharides and Polyols; EAT−26: Eating Attitude Test-26; BES: Binge Eating Scale; ORAC: Oxygen Radical Absorbance Capacity; PRAL: renal potential load; LFD: Low FODMAPs Diet; EEN: Exclusive Enteral Nutrition; PEN: Partial Enteral Nutrition; SCD: Specific carbohydrate diet; MD: Mediterranean diet; WFD: Whole Food Diet; HF: High Fiber, HM: High in take in red Meat; LFHF: Low Fat High Fiber; iSAD: improved Standard American; CDED: Crohn’s Disease Exclusion Diet; FGS: Functional Gastrointestinal Disorders; HBI: Har-vey-Bradshaw Index; IBS-SSS: Irritable Bowel Syndrome -Severity Scoring System; SCCAI: Simple Clinical Colitis Activity Index; PUCDAI: Pediatric Ulcerative Colitis Activity Index; PCDAI: Pedi-atric Crohn’s Disease Activity Index; CDAI: Crohn’s Disease Activity Index.

References

  1. Ananthakrishnan, A.N. Environmental risk factors for inflammatory bowel diseases: A review. Dig. Dis. Sci. 2015, 60, 290–298. [Google Scholar] [CrossRef] [PubMed]
  2. Malik, T.A. Inflammatory bowel disease: Historical perspective, epidemiology, and risk factors. Surg. Clin. N. Am. 2015, 95, 1105–1122. [Google Scholar] [CrossRef] [PubMed]
  3. Ng, S.C.; Shi, H.Y.; Hamidi, N.; Underwood, F.E.; Tang, W.; Benchimol, E.I.; Panaccione, R.; Ghosh, S.; Wu, J.C.Y.; Chan, F.K.L.; et al. Worldwide incidence and prevalence of inflammatory bowel disease in the 21st century: A systematic review of population-based studies. Lancet 2017, 390, 2769–2778. [Google Scholar] [CrossRef]
  4. Kaplan, G.G.; Windsor, J.W. The four epidemiological stages in the global evolution of inflammatory bowel disease. Nat. Rev. Gastroenterol. Hepatol. 2021, 18, 56–66. [Google Scholar] [CrossRef] [PubMed]
  5. United Nations. United Nations World Economic Situation and Prospects. Available online: https://www.un.org/development/desa/dpad/publication/world-economic-situation-and-prospects-2022 (accessed on 16 January 2023).
  6. Molodecky, N.A.; Soon, I.S.; Rabi, D.M.; Ghali, W.A.; Ferris, M.; Chernoff, G.; Benchimol, E.I.; Panaccione, R.; Ghosh, S.; Barkema, H.W.; et al. Increasing incidence and prevalence of the inflammatory bowel diseases with time, based on systematic review. Gastroenterology 2012, 142, 46–54.e42. [Google Scholar] [CrossRef]
  7. Coward, S.; Clement, F.; Benchimol, E.I.; Bernstein, C.N.; Avina-Zubieta, J.A.; Bitton, A.; Carroll, M.W.; Hazlewood, G.; Jacobson, K.; Jelinski, S.; et al. Past and Future Burden of Inflammatory Bowel Diseases Based on Modeling of Population-Based Data. Gastroenterology 2019, 156, 1345–1353.e4. [Google Scholar] [CrossRef]
  8. Bitton, A.; Vutcovici, M.; Sewitch, M.; Suissa, S.; Brassard, P. Mortality trends in Crohn’s disease and ulcerative colitis: A population- based study in Quebec, Canada. Inflamm. Bowel Dis. 2016, 22, 416–423. [Google Scholar] [CrossRef] [PubMed]
  9. Roberts, S.E.; Thorne, K.; Thapar, N.; Broekaert, I.; Benninga, M.A.; Dolinsek, J.; Mas, E.; Miele, E.; Orel, R.; Pienar, C.; et al. A Systematic Review and Meta-analysis of Paediatric Inflammatory Bowel Disease Incidence and Prevalence Across Europe. J. Crohns Colitis. 2020, 14, 1119–1148. [Google Scholar] [CrossRef]
  10. Kaplan, G.G.; Ng, S.C. Understanding and preventing the global increase of inflammatory bowel disease. Gastroenterology 2017, 152, 313–321. [Google Scholar] [CrossRef]
  11. Ananthakrishnan, A.N. Epidemiology and risk factors for IBD. Nat. Rev. Gastroenterol. Hepatol. 2015, 12, 205–217. [Google Scholar] [CrossRef]
  12. Mak, W.Y.; Zhao, M.; Ng, S.C.; Burisch, J. The epidemiology of inflammatory bowel disease: East meets west. J. Gastroenterol. Hepatol. 2020, 35, 380–389. [Google Scholar] [CrossRef] [PubMed]
  13. Ng, S.C.; Tang, W.; Leong, R.W.; Chen, M.; Ko, Y.; Studd, C.; Niewiadomski, O.; Bell, S.; Kamm, M.A.; de Silva, H.J.; et al. Asia-Pacific Crohn's and Colitis Epidemiology Study ACCESS Group. Environmental risk factors in inflammatory bowel disease: A population-based case-control study in Asia-Pacific. Gut 2015, 64, 1063–1071. [Google Scholar] [CrossRef] [PubMed]
  14. Molodecky, N.A.; Panaccione, R.; Ghosh, S.; Barkema, H.W.; Kaplan, G.G. Alberta Inflammatory Bowel Disease Consortium. Challenges associated with identifying the environmental determinants of the inflammatory bowel diseases. Inflamm. Bowel Dis. 2011, 17, 1792–1799. [Google Scholar] [CrossRef]
  15. Ratajczak, A.E.; Festa, S.; Aratari, A.; Papi, C.; Dobrowolska, A.; Krela-Kaźmierczak, I. Should the Mediterranean diet be recommended for inflammatory bowel diseases patients? A narrative review. Front Nutr. 2023, 9, 1088693. [Google Scholar] [CrossRef] [PubMed]
  16. Bach-Faig, A.; Berry, E.M.; Lairon, D.; Reguant, J.; Trichopoulou, A.; Dernini, S.; Medina, F.X.; Battino, M.; Belahsen, R.; Miranda, G.; et al. Mediterranean diet pyramid today. Science and cultural updates. Public Health Nutr. 2011, 14, 2274–2284. [Google Scholar] [CrossRef]
  17. Rizzello, F.; Spisni, E.; Giovanardi, E.; Imbesi, V.; Salice, M.; Alvisi, P.; Valerii, M.C.; Gionchetti, P. Implications of the westernized diet in the onset and progression of IBD. Nutrients 2019, 11, 1033. [Google Scholar] [CrossRef]
  18. Sharma, M.; Kishore, A.; Roy, D.; Joshi, K. A comparison of the Indian diet with the EAT-Lancet reference diet. BMC Public Health. 2020, 20, 812. [Google Scholar] [CrossRef]
  19. The Traditional Japanese Diet May Have Lessons for Modern Life. Available online: https://www.nature.com/articles/d42859-021-00062-7 (accessed on 16 January 2023).
  20. Ooraikul, B.; Sirichote, A.; Siripongvutikorn, S. Southeast Asian Diets and Health Promotion. In Wild-Type Food in Health Promotion and Disease Prevention; De Meester, F., Watson, R.R., Eds.; Humana Press: Totowa, NJ, USA, 2008. [Google Scholar]
  21. Schaffer, M.; Schaffer, P.M.; Zidan, J.; Bar Sela, G. Curcuma as a functional food in the control of cancer and inflammation. Curr. Opin. Clin. Nutr Metab. Care 2011, 14, 588–597. [Google Scholar] [CrossRef]
  22. Blasbalg, T.L.; Hibbeln, J.R.; Ramsden, C.E.; Majchrzak, S.F.; Rawlings, R.R. Changes in consumption of omega-3 and omega-6 fatty acids in the United States during the 20th century. Am. J. Clin. Nutr. 2011, 93, 950–962. [Google Scholar] [CrossRef] [PubMed]
  23. Tjonneland, A.; Overvad, K.; Bergmann, M.M.; Nagel, G.; Linseisen, J.; Hallmans, G.; Palmqvist, R.; Sjodin, H.; Hagglund, G.; Berglund, G.; et al. Linoleic acid, a dietary n-6 polyunsaturated fatty acid, and the aetiology of ulcerative colitis: A nested case–control study within a European prospective cohort study. Gut 2009, 58, 1606–1611. [Google Scholar] [CrossRef] [PubMed]
  24. Ananthakrishnan, A.N.; Khalili, H.; Konijeti, G.G.; Higuchi, L.M.; de Silva, P.; Fuchs, C.S.; Willett, W.C.; Richter, J.M.; Chan, A.T. Long-term intake of dietary fat and risk of ulcerative colitis and Crohn’s disease. Gut 2014, 63, 776–784. [Google Scholar] [CrossRef]
  25. de Silva, P.S.; Luben, R.; Shrestha, S.S.; Khaw, K.T.; Hart, A.R. Dietary arachidonic and oleic acid intake in ulcerative colitis etiology: A prospective cohort study using 7-day food diaries. Eur. J. Gastroenterol. Hepatol. 2014, 26, 11–18. [Google Scholar] [CrossRef]
  26. John, S.A.; Luben, R.B.; Shrestha, S.S.B.; Welch, A.C.; Khaw, K.-T.D.; Hart, A.R. Dietary n-3 polyunsaturated fatty acids and the aetiology of ulcerative colitis: A UK prospective cohort study. Eur. J. Gastroenterol. Hepatol. 2010, 22, 602–606. [Google Scholar] [CrossRef] [PubMed]
  27. Global and Regional Food Consumption Patterns and Trends. Available online: https://www.fao.org/3/ac911e/ac911e05.htm (accessed on 16 January 2023).
  28. The State of Food Security and Nutrition in the World 2022. Available online: https://www.fao.org/documents/card/en/c/cc0639en (accessed on 16 January 2023).
  29. Adolph, T.E.; Zhang, J. Diet fuelling inflammatory bowel diseases: Preclinical and clinical concepts. Gut 2022, 71, 2574–2586. [Google Scholar] [CrossRef] [PubMed]
  30. Khoshbin, K.; Camilleri, M. Effects of dietary components on intestinal permeability in health and disease. Am. J. Physiol. Gastrointest. Liver Physiol. 2020, 319, G589–G608. [Google Scholar] [CrossRef] [PubMed]
  31. Lewis, J.D.; Abreu, M.T. Diet as a Trigger or Therapy for Inflammatory Bowel Diseases. Gastroenterology 2017, 152, 398–414.e6. [Google Scholar] [CrossRef]
  32. Hugot, J.P.; Chamaillard, M.; Zouali, H.; Lesage, S.; Cézard, J.P.; Belaiche, J.; Almer, S.; Tysk, C.; O´Morain, C.A.; Gassull, M.; et al. Association of NOD2 leucine-rich repeat variants with susceptibility to Crohn’s disease. Nature 2001, 411, 599–603. [Google Scholar] [CrossRef]
  33. Martinez-Medina, M.; Denizot, J.; Dreux, N.; Robin, F.; Billard, E.; Bonnet, R.; Darfeuille-Michaud, A.; Barnich, N. Western diet induces dysbiosis with increased E coli in CEABAC10 mice, alters host barrier function favouring AIEC colonisation. Gut 2014, 63, 116–124. [Google Scholar] [CrossRef]
  34. Lee, D.; Albenberg, L.; Compher, C.; Baldassano, R.; Piccoli, D.; Lewis, J.D.; Wu, G.D. Diet in the pathogenesis and treatment of inflammatory bowel diseases. Gastroenterology 2015, 148, 1087–1106. [Google Scholar] [CrossRef]
  35. Liu, T.C.; Kern, J.T.; Jain, U.; Sonnek, N.M.; Xiong, S.; Simpson, K.F.; VanDussen, K.L.; Winkler, E.S.; Haritunians, T.; Malique, A.; et al. Western diet induces Paneth cell defects through microbiome alterations and farnesoid X receptor and type I interferon activation. Cell Host Microbe. 2021, 29, 988–1001. [Google Scholar] [CrossRef]
  36. Suez, J.; Korem, T.; Zeevi, D.; Zilberman-Schapira, G.; Thaiss, C.A.; Maza, O.; Israeli, D.; Zmora, N.; Gilad, S.; Weinberger, A.; et al. Artificial sweeteners induce glucose intolerance by altering the gut microbiota. Nature 2014, 514, 181–186. [Google Scholar] [CrossRef] [PubMed]
  37. Nickerson, K.P.; McDonald, C. Crohn’s disease-associated adherent-invasive Escherichia coli adhesion is enhanced by exposure to the ubiquitous dietary polysaccharide maltodextrin. PLoS ONE 2012, 7, e52132. [Google Scholar] [CrossRef] [PubMed]
  38. Chassaing, B.; Koren, O.; Goodrich, J.K.; Poole, A.C.; Srinivasan, S.; Ley, R.E.; Gewirtz, A.T. Dietary emulsifiers impact the mouse gut microbiota promoting colitis and metabolic syndrome. Nature 2015, 519, 92–96. [Google Scholar] [CrossRef]
  39. He, Z.; Chen, L.; Catalan-Dibene, J.; Bongers, G.; Faith, J.J.; Suebsuwong, C.; DeVita, R.J.; Shen, Z.; Fox, J.G.; Lafaille, J.J.; et al. Food colorants metabolized by commensal bacteria promote colitis in mice with dysregulated expression of interleukin-23. Cell Metab. 2021, 33, 1358–1371.e5. [Google Scholar] [CrossRef]
  40. Zhou, X.L.; Zhao, Q.Q.; Li, X.F.; Li, Z.; Zhao, S.X.; Li, Y.M. Protein intake and risk of inflammatory bowel disease: A meta-analysis. Asia Pac. J. Clin. Nutr. 2022, 31, 443–449. [Google Scholar] [CrossRef]
  41. David, L.A.; Maurice, C.F.; Carmody, R.N.; Gootenberg, D.B.; Button, J.E.; Wolfe, B.E.; Ling, A.V.; Devlin, A.S.; Varma, Y.; Fischbach, M.A.; et al. Diet rapidly and reproducibly alters the human gut microbiome. Nature 2014, 505, 559–563. [Google Scholar] [CrossRef]
  42. Fava, F.; Gitau, R.; Griffin, B.A.; Gibso, G.R.; Tuohy, K.M.; Lovegrove, J.A. The type and quantity of dietary fat and carbohydrate alter faecal microbiome and short-chain fatty acid excretion in a metabolic syndrome ’at-risk’ population. Int. J. Obes. 2013, 37, 216–223. [Google Scholar] [CrossRef]
  43. Zinöcker, M.K.; Lindseth, I.A. The Western Diet-Microbiome-Host interaction and its role in metabolic disease. Nutrients 2018, 10, 365. [Google Scholar] [CrossRef]
  44. Moran, G.W.; Dubeau, M.-F.; Kaplan, G.G.; Panaccione, R.; Ghosh, S. The increasing weight of Crohn’s disease subjects in clinical trials: A hypothesis- generatings time-trend analysis. Inflamm. Bowel Dis. 2013, 19, 2949–2956. [Google Scholar] [CrossRef]
  45. Rocha, D.M.; Caldas, A.P.; Oliveira, L.L.; Bressan, J.; Hermsdorff, H.H. Saturated fatty acids trigger TLR4-mediated inflammatory response. Atherosclerosis 2016, 244, 211–215. [Google Scholar] [CrossRef] [PubMed]
  46. Alatshan, A.; Benkő, S. Nuclear Receptors as Multiple Regulators of NLRP3 Inflammasome Function. Front. Immunol. 2021, 12, 630569. [Google Scholar] [CrossRef] [PubMed]
  47. Khalili, H.; Chan, S.S.M.; Lochhead, P.; Ananthakrishnan, A.N.; Hart, A.R.; Chan, A.T. The role of diet in the aetiopathogenesis of inflammatory bowel disease. Nat. Rev. Gastroenterol. Hepatol. 2018, 15, 525–535. [Google Scholar] [CrossRef]
  48. Bach Knudsen, K.E.; Lærke, H.N.; Hedemann, M.S.; Nielsen, T.S.; Ingerslev, A.K.; Gundelund Nielsen, D.S.; Theil, P.K.; Purup, S.; Hald, S.; Schioldan, A.G.; et al. Impact of Diet-Modulated Butyrate Production on Intestinal Barrier Function and Inflammation. Nutrients 2018, 10, 1499. [Google Scholar] [CrossRef]
  49. Geirnaert, A.; Calatayud, M.; Grootaert, C.; Laukens, D.; Devriese, S.; Smagghe, G.; De Vos, M.; Boon, N.; Van de Wiele, T. Butyrate-producing bacteria supplemented in vitro to Crohn’s disease patient microbiota increased butyrate production and enhanced intestinal epithelial barrier integrity. Sci. Rep. 2017, 7, 11450. [Google Scholar] [CrossRef]
  50. Chang, P.V.; Hao, L.; Offermanns, S.; Medzhitov, R. The microbial metabolite butyrate regulates intestinal macrophage function via histone deacetylase inhibition. Proc. Natl. Acad. Sci. USA 2014, 111, 2247–2252. [Google Scholar] [CrossRef]
  51. Severo, J.S.; da Silva Barros, V.J.; Alves da Silva, A.C.; Luz Parente, J.M.; Lima, M.M.; Moreira Lima, A.Â.; Dos Santos, A.A.; Matos Neto, E.M.; Tolentino, M. Effects of glutamine supplementation on inflammatory bowel disease: A systematic review of clinical trials. Clin. Nutr. ESPEN 2021, 42, 53–60. [Google Scholar] [CrossRef]
  52. Li, J.Y.; Guo, Y.C.; Zhou, H.F.; Yue, T.T.; Wang, F.X.; Sun, F.; Wang, W.Z. Arginine metabolism regulates the pathogenesis of inflammatory bowel disease. Nutr. Rev. 2022, 30, nuac070. [Google Scholar] [CrossRef] [PubMed]
  53. Zhang, B.; Zhong, Q.; Liu, N.; Song, P.; Zhu, P.; Zhang, C.; Sun, Z. Dietary Glutamine Supplementation Alleviated Inflammation Responses and Improved Intestinal Mucosa Barrier of LPS-Challenged Broilers. Animals 2022, 12, 1729. [Google Scholar] [CrossRef] [PubMed]
  54. Hou, Y.C.; Liu, J.J.; Pai, M.H.; Tsou, S.S.; Yeh, S.L. Alanyl-glutamine administration suppresses Th17 and reduces inflammatory reaction in dextran sulfate sodium–induced acute colitis. Int. Immunopharmacol. 2013, 17, 1–8. [Google Scholar] [CrossRef]
  55. Ren, W.; Chen, S.; Yin, J.; Duan, J.; Li, T.; Liu, G.; Feng, Z.; Tan, B.; Yin, Y.; Wu, G. Dietary Arginine Supplementation of Mice Alters the Microbial Population and Activates Intestinal Innate Immunity. J. Nutr. 2014, 144, 988–995. [Google Scholar] [CrossRef]
  56. Zelante, T.; Iannitti, R.G.; Cunha, C.; De Luca, A.; Giovannini, G.; Pieraccini, G.; Zecchi, R.; D’Angelo, C.; Massi-Benedetti, C.; Fallarino, F.; et al. Tryptophan catabolites from microbiota engage aryl hydrocarbon receptor and balance mucosal reactivity via interleukin-22. Immunity 2013, 39, 372–385. [Google Scholar] [CrossRef] [PubMed]
  57. Jaeger, C.; Tischkau, S.A. Role of aryl hydrocarbon receptor in circadian clock disruption and metabolic dysfunction. Environ. Health Insights 2016, 10, 133–141. [Google Scholar] [CrossRef] [PubMed]
  58. Chng, S.H.; Kundu, P.; Dominguez-Brauer, C.; Teo, W.L.; Kawajiri, K.; Fujii-Kuriyama, Y.; Mak, T.W.; Pettersson, S. Ablating the aryl hydrocarbon receptor (AhR) in CD11cþ cells perturbs intestinal epithelium development and intestinal immunity. Sci. Rep. 2016, 6, 23820. [Google Scholar] [CrossRef] [PubMed]
  59. Fu, Z.; Chen, X.; Guan, S.; Yan, Y.; Lin, H.; Hua, Z. Curcumin inhibits angiogenesis and improves defective hematopoiesis induced by tumor-derived VEGF in tumor model through modulating VEGF-VEGFR2 signaling pathway. Oncotarget 2015, 6, 19469–19482. [Google Scholar] [CrossRef]
  60. Lang, A.; Salomon, N.; Wu, J.C.; Kopylov, U.; Lahat, A.; Har-Noy, O.; Ching, J.Y.; Cheong, P.K.; Avidan, B.; Gamus, D.; et al. Curcumin in combination with mesalamine induces remission in patients with mild-to-moderate ulcerative colitis in a randomized controlled trial. Clin. Gastroenterol. Hepatol. 2015, 13, 1444–1449. [Google Scholar] [CrossRef]
  61. Lin, Y.; Liu, H.; Bu, L.; Chen, C.; Ye, X. Review of the Effects and Mechanism of Curcumin in the Treatment of Inflammatory Bowel Disease. Front. Pharmacol. 2022, 13, 908077. [Google Scholar] [CrossRef]
  62. Park, S.H.; Kyeong, M.S.; Hwang, Y.; Ryu, S.Y.; Han, S.B.; Kim, Y. Inhibition of LPS binding to MD-2 co-receptor for suppressing TLR4-mediated expression of inflammatory cytokine by 1-dehydro-10-gingerdione from dietary ginger. Biochem. Biophys. Res. Commun. 2012, 419, 735–740. [Google Scholar] [CrossRef]
  63. De Fazio, L.; Spisni, E.; Cavazza, E.; Strillacci, A.; Candela, M.; Centanni, M.; Ricci, C.; Rizzello, F.; Campieri, M.; Valerii, M.C. Dietary Geraniol by Oral or Enema Administration Strongly Reduces Dysbiosis and Systemic Inflammation in Dextran Sulfate Sodium-Treated Mice. Front. Pharmacol. 2016, 3, 38. [Google Scholar] [CrossRef]
  64. Valerii, M.C.; Turroni, S.; Ferreri, C.; Zaro, M.; Sansone, A.; Dalpiaz, A.; Botti, G.; Ferraro, L.; Spigarelli, R.; Bellocchio, I.; et al. Effect of a Fiber D-Limonene-Enriched Food Supplement on Intestinal Microbiota and Metabolic Parameters of Mice on a High-Fat Diet. Pharmaceutics 2021, 21, 1753. [Google Scholar] [CrossRef]
  65. Ananthakrishnan, A.N.; Khalili, H.; Konijeti, G.G.; Higuchi, L.M.; de Silva, P.; Korzenik, J.R.; Fuchs, C.S.; Willett, W.C.; Richter, J.M.; Chan, A.T. A prospective study of long-term intake of dietary fiber and risk of Crohn’s disease and ulcerative colitis. Gastroenterology 2013, 145, 970–997. [Google Scholar] [CrossRef]
  66. Racine, A.; Carbonnel, F.; Chan, S.S.; Hart, A.R.; Bueno-de-Mesquita, H.B.; Oldenburg, B.; van Schaik, F.D.; Tjønneland, A.; Olsen, A.; Dahm, C.C.; et al. Dietary patterns and risk of inflammatory bowel disease in Europe: Results from the EPIC study. Inflamm. Bowel Dis. 2016, 22, 345–354. [Google Scholar] [CrossRef] [PubMed]
  67. Nitescu, M.; Istratescu, D.; Preda, C.M.; Manuc, T.E.; Louis, E.; Manuc, M.; Stroie, T.; Catrinoiu, M.; Tieranu, C.G.; Badea, L.E.; et al. Role of an Exclusion Diet (Reduced Disaccharides, Saturated Fats, Emulsifiers, Red and Ultraprocessed Meats) in Maintaining the Remission of Chronic Inflammatory Bowel Diseases in Adults. Medicina 2023, 59, 329. [Google Scholar] [CrossRef]
  68. Cederholm, T.; Barazzoni, R.; Austin, P.; Ballmer, P.; Biolo, G.; Bischoff, S.C.; Compher, C.; Correia, I.; Higashiguchi, T.; Holst, M.; et al. ESPEN guidelines on definitions and terminology of clinical nutrition. Clin. Nutr. 2017, 36, 49–64. [Google Scholar] [CrossRef] [PubMed]
  69. Fact Sheets–Malnutrition. Available online: https://www.who.int/news-room/factsheets/detail/malnutrition (accessed on 16 January 2023).
  70. Singh, A.; Wall, C.; Levine, A.; Midha, V.; Mahajan, R.; Sood, A. Nutritional screening and assessment in inflammatory bowel disease. Indian J. Gastroenterol. 2022, 41, 5–22. [Google Scholar] [CrossRef] [PubMed]
  71. Donnellan, C.F.; Yann, L.H.; Lal, S. Nutritional management of Crohn’s disease. Therap. Adv. Gastroenterol. 2013, 6, 231–242. [Google Scholar] [CrossRef] [PubMed]
  72. Balestrieri, P.; Ribolsi, M.; Guarino, M.P.L.; Emerenziani, S.; Altomare, A.; Cicala, M. Nutritional Aspects in Inflammatory Bowel Diseases. Nutrients 2020, 12, 372. [Google Scholar] [CrossRef]
  73. Ünal, N.G.; Oruç, N.; Tomey, O.; Ömer Özütemiz, A. Malnutrition and sarcopenia are prevalent among inflammatory bowel disease patients with clinical remission. Eur. J. Gastroenterol. Hepatol. 2021, 33, 1367–1375. [Google Scholar] [CrossRef]
  74. Massironi, S.; Rossi, R.E.; Cavalcoli, F.A.; Della Valle, S.; Fraquelli, M.; Conte, D. Nutritional deficiencies in inflammatory bowel disease: Therapeutic approaches. Clin. Nutr. 2013, 32, 904–910. [Google Scholar] [CrossRef]
  75. Weisshof, R.; Chermesh, I. Micronutrient deficiencies in inflammatory bowel disease. Curr. Opin. Clin. Nutr. Metab. Care 2015, 18, 576–581. [Google Scholar] [CrossRef]
  76. Vitamin and Mineral Deficiencies in Inflammatory Bowel Disease. Available online: https://www.uptodate.com/contents/vitamin-and-mineral-deficiencies-in-inflammatory-bowel-disease (accessed on 16 January 2023).
  77. Sharma, K.; Mogensen, K.M.; Robinson, M.K. Pathophysiology of Critical Illness and Role of Nutrition. Nutr. Clin. Pract. 2019, 34, 12–22. [Google Scholar] [CrossRef]
  78. Daun, J.M.; McCarthy, D.O. The role of cholecystokinin in interleukin-1-induced anorexia. Physiol. Behav. 1993, 54, 237–241. [Google Scholar] [CrossRef] [PubMed]
  79. Plata-Salaman, C.R. Brain cytokine production and action in anorexia and cachexia. Cytokine 2001, 15, 1–3. [Google Scholar] [CrossRef] [PubMed]
  80. Plata-Salamán, C.R.; Oomura, Y.; Kai, Y. Tumor necrosis factor and interleukin-1 beta: Suppression of food intake by direct action in the central nervous system. Brain Res. 1988, 448, 106–114. [Google Scholar] [CrossRef] [PubMed]
  81. Kuriyama, K.; Hori, T.; Mori, T.; Nakashima, T. Actions of interferonα and interleukin-1β on the glucose-responsive neurons in the ventromedial hypothalamus. Brain Res. Bull 1990, 24, 803–810. [Google Scholar] [CrossRef] [PubMed]
  82. Suto, G.; Kiraly, A.; Tache, Y. Interleukin-1 beta inhibits gastric emptying in rats: Mediation through prostaglandin and corticotropin- releasing factor. Gastroenterology 1994, 106, 1568–1575. [Google Scholar] [CrossRef] [PubMed]
  83. Limdi, J.K.; Aggarwal, D.; McLaughlin, J.T. Dietary Practices and Beliefs in Patients with Inflammatory Bowel Disease. Inflamm. Bowel Dis. 2016, 22, 164–170. [Google Scholar] [CrossRef]
  84. Murtagh, A.; Cooney, L.; Higginbotham, C.; Heavey, P. Dietary practices, beliefs and behaviours of adults with inflammatory bowel disease: A cross-sectional study. Ir. J. Med. Sci. 2022, 1971, 1–10. [Google Scholar] [CrossRef]
  85. Capristo, E.; Addolorato, G.; Mingrone, G.; Greco, A.V.; Gasbarrini, G. Effect of disease localization on the anthropometric and metabolic features of Crohn’s disease. Am. J. Gastroenterol. 1998, 93, 2411–2419. [Google Scholar] [CrossRef]
  86. Barkas, F.; Liberopoulos, E.; Kei, A.; Elisaf, M. Electrolyte and acid-base disorders in inflammatory bowel disease. Ann. Gastroenterol. 2013, 26, 23–28. [Google Scholar]
  87. Zupo, R.; Sila, A.; Castellana, F.; Bringiotti, R.; Curlo, M.; De Pergola, G.; De Nucci, S.; Giannelli, G.; Mastronardi, M.; Sardone, R. Prevalence of Zinc Deficiency in Inflammatory Bowel Disease: A Systematic Review and Meta-Analysis. Nutrients 2022, 14, 4052. [Google Scholar] [CrossRef]
  88. Song, H.K.; Buzby, G.P. Nutritional support for Crohn´s disease. Surg. Clin. N. Am. 2001, 81, 103–115. [Google Scholar] [CrossRef] [PubMed]
  89. Bermejo, F.; Algaba, A.; Guerra, I.; Gisbert, J.P. Response to letter: Folate deficiency in Crohn’s disease. Scand. J. Gastroenterol. 2014, 49, 255–256. [Google Scholar] [CrossRef]
  90. Vitek, L. Bile acid malabsorption in inflammatory bowel disease. Inflamm. Bowel Dis. 2015, 21, 476–483. [Google Scholar] [CrossRef] [PubMed]
  91. Torki, M.; Gholamrezaei, A.; Mirbagher, L.; Danesh, M.; Kheiri, S.; Emami, M.H. Vitamin D deficiency associated with disease activity in patients with inflammatory bowel diseases. Dig. Dis. Sci. 2015, 60, 3085–3091. [Google Scholar] [CrossRef] [PubMed]
  92. Fabisiak, N.; Fabisiak, A.; Watala, C.; Fichna, J. Fat-soluble Vitamin Deficiencies and Inflammatory Bowel Disease: Systematic Review and Meta-Analysis. J. Clin. Gastroenterol. 2017, 51, 878–889. [Google Scholar] [CrossRef]
  93. Lai, Y.; Masatoshi, H.; Ma, Y.; Guo, Y.; Zhang, B. Role of Vitamin K in Intestinal Health. Front Immunol. 2022, 12, 791565. [Google Scholar] [CrossRef]
  94. Schoon, E.J.; Müller, M.C.; Vermeer, C.; Schurgers, L.J.; Brummer, R.J.; Stockbrügger, R.W. Low serum and bone vitamin K status in patients with longstanding Crohn’s disease: Another pathogenetic factor of osteoporosis in Crohn’s disease? Gut 2001, 48, 473–477. [Google Scholar] [CrossRef]
  95. Adike, A.; DiBaise, J.K. Small Intestinal Bacterial Overgrowth: Nutritional Implications, Diagnosis, and Management. Gastroenterol. Clin. N. Am. 2018, 47, 193–208. [Google Scholar] [CrossRef]
  96. Rana, S.V.; Sharma, S.; Malik, A.; Kaur, J.; Prasad, K.K.; Sinha, S.K.; Singh, K. Small intestinal bacterial overgrowth and orocecal transit time in patients of inflammatory bowel disease. Dig. Dis. Sci. 2013, 58, 2594–2598. [Google Scholar] [CrossRef]
  97. Tabaqchali, S.; Hatzioannou, J.; Booth, C.C. Bile-salt deconjugation and steatorrhoea in patients with the stagnant-loop syndrome. Lancet 1968, 2, 12–16. [Google Scholar] [CrossRef]
  98. Welkos, S.L.; Toskes, P.P.; Baer, H. Importance of anaerobic bacteria in the cobalamin malabsorption of the experimental rat blind loop syndrome. Gastroenterology 1981, 80, 313–320. [Google Scholar] [CrossRef] [PubMed]
  99. de Vries, J.H.M.; Dijkhuizen, M.; Tap, P.; Witteman, B.J.M. Patient’s Dietary Beliefs and Behaviours in Inflammatory Bowel Disease. Dig. Dis. 2019, 37, 131–139. [Google Scholar] [CrossRef] [PubMed]
  100. Crooks, B.; Misra, R.; Arebi, N.; Kok, K.; Brookes, M.J.; McLaughlin, J.; Limdi, J.K. The dietary practices and beliefs of British South Asian people living with inflammatory bowel disease: A multicenter study from the United Kingdom. Intest. Res. 2022, 20, 53–63. [Google Scholar] [CrossRef]
  101. Peters, J.E.; Basnayake, C.; Hebbard, G.S.; Salzberg, M.R.; Kamm, M.A. Prevalence of disordered eating in adults with gastrointestinal disorders: A systematic review. Neurogastroenterol. Motil. 2022, 34, e14278. [Google Scholar] [CrossRef] [PubMed]
  102. Urbano, A.P.; Sassaki, L.Y.; Dorna, M.S.; Carvalhaes, M.A.; Martini, L.A.; Ferreira, A.L. Nutritional intake according to injury extent in ulcerative colitis patients. J. Hum. Nutr. Diet. 2013, 26, 445–451. [Google Scholar] [CrossRef] [PubMed]
  103. Lim, H.S.; Kim, S.K.; Hong, S.J. Food Elimination Diet and Nutritional Deficiency in Patients with Inflammatory Bowel Disease. Clin. Nutr. Res. 2018, 7, 48–55. [Google Scholar] [CrossRef]
  104. Larussa, T.; Suraci, E.; Marasco, R.; Imeneo, M.; Abenavoli, L.; Luzza, F. Self-Prescribed Dietary Restrictions are Common in Inflammatory Bowel Disease Patients and Are Associated with Low Bone Mineralization. Medicina 2019, 20, 07. [Google Scholar] [CrossRef]
  105. Rizzello, F.; Gionchetti, P.; Spisni, E.; Saracino, I.M.; Bellocchio, I.; Spigarelli, R.; Collini, N.; Imbesi, V.; Dervieux, T.; Alvisi, P.; et al. Dietary Habits and Nutrient Deficiencies in a Cohort of European Crohn’s Disease Adult Patients. Int. J. Mol. Sci. 2023, 24, 1494. [Google Scholar] [CrossRef]
  106. Labriola, F.; Marcato, C.; Zarbo, C.; Betti, L.; Catelli, A.; Valerii, M.C.; Spisni, E.; Alvisi, P. Dietary Habits of a Group of Children with Crohn’s Disease Compared to Healthy Subjects: Assessment of Risk of Nutritional Deficiencies through a Bromatological Analysis. Nutrients 2022, 14, 499. [Google Scholar] [CrossRef]
  107. Pituch-Zdanowska, A.; Kowalska-Duplaga, K.; Jarocka-Cyrta, E.; Stawicka, A.; Dziekiewicz, M.; Banaszkiewicz, A. Dietary Beliefs and Behaviors Among Parents of Children with Inflammatory Bowel Disease. J. Med. Food. 2019, 22, 817–822. [Google Scholar] [CrossRef]
  108. Bramuzzo, M.; Grazian, F.; Grigoletto, V.; Daidone, A.; Martelossi, S.; Mario, F.; Maurel, E.; Lega, S.; Giudici, F.; Di Leo, G.; et al. Dietary Beliefs in Children and Adolescents with Inflammatory Bowel Disease and their Parents. J. Pediatr. Gastroenterol. Nutr. 2022, 75, e43–e48. [Google Scholar] [CrossRef] [PubMed]
  109. Haas, S.V.; Haas, M.P. The treatment of celiac disease with the specific carbohydrate diet; report on 191 additional cases. Am. J. Gastroenterol. 1955, 23, 344–360. [Google Scholar] [PubMed]
  110. Breaking The Vicious Cycle, Intestinal Health Through Diet. Available online: http://www.breakingtheviciouscycle.info (accessed on 16 January 2023).
  111. Braly, K.; Williamson, N.; Shaffer, M.L.; Lee, D.; Wahbeh, G.; Klein, J.; Giefer, M.; Suskind, D.L. Nutritional Adequacy of the Specific Carbohydrate Diet in Pediatric Inflammatory Bowel Disease. J. Pediatr. Gastroenterol. Nutr. 2017, 65, 533–538. [Google Scholar] [CrossRef] [PubMed]
  112. Cohen, S.A.; Gold, B.D.; Oliva, S.; Lewis, J.; Stallworth, A.; Koch, B.; Eshee, L.; Mason, D. Clinical and mucosal improvement with specific carbohydrate diet in pediatric Crohn disease. J. Pediatr. Gastroenterol. Nutr. 2014, 59, 516–521. [Google Scholar] [CrossRef] [PubMed]
  113. Suskind, D.L.; Lee, D.; Kim, Y.M.; Wahbeh, G.; Singh, N.; Braly, K.; Nuding, M.; Nicora, C.D.; Purvine, S.O.; Lipton, M.S.; et al. The Specific Carbohydrate Diet and Diet Modification as Induction Therapy for Pediatric Crohn’s Disease: A Randomized Diet Controlled Trial. Nutrients 2020, 12, 3749. [Google Scholar] [CrossRef] [PubMed]
  114. Suskind, D.L.; Wahbeh, G.; Gregory, N.; Vendettuoli, H.; Christie, D. Nutritional therapy in pediatric Crohn disease: The specific carbohydrate diet. J. Pediatr. Gastroenterol. Nutr. 2014, 58, 87–91. [Google Scholar] [CrossRef]
  115. Obih, C.; Wahbeh, G.; Lee, D.; Braly, K.; Giefer, M.; Shaffer, M.L.; Nielson, H.; Suskind, D.L. Specific carbohydrate diet for pediatric inflammatory bowel disease in clinical practice within an academic IBD center. Nutrition 2016, 32, 418–425. [Google Scholar] [CrossRef]
  116. Lewis, J.D.; Sandler, R.S.; Brotherton, C.; Brensinger, C.; Li, H.; Kappelman, M.D.; Daniel, S.G.; Bittinger, K.; Albenberg, L.; Valentine, J.F.; et al. A Randomized Trial Comparing the Specific Carbohydrate Diet to a Mediterranean Diet in Adults with Crohn’s Disease. Gastroenterology 2021, 161, 837–852.e9. [Google Scholar] [CrossRef]
  117. Chicco, F.; Magrì, S.; Cingolani, A.; Paduano, D.; Pesenti, M.; Zara, F.; Tumbarello, F.; Urru, E.; Melis, A.; Casula, L.; et al. Multidimensional Impact of Mediterranean Diet on IBD Patients. Inflamm. Bowel Dis. 2021, 27, 1–9. [Google Scholar] [CrossRef]
  118. Brotherton, C.S.; Taylor, A.G.; Bourguignon, C.; Anderson, J.G. A high-fiber diet may improve bowel function and health-related quality of life in patients with Crohn disease. Gastroenterol. Nurs. 2014, 37, 206–216. [Google Scholar] [CrossRef]
  119. Fritsch, J.; Garces, L.; Quintero, M.A.; Pignac-Kobinger, J.; Santander, A.M.; Fernández, I.; Ban, Y.J.; Kwon, D.; Phillips, M.C.; Knight, K.; et al. Low-Fat, High-Fiber Diet Reduces Markers of Inflammation and Dysbiosis and Improves Quality of Life in Patients with Ulcerative Colitis. Clin. Gastroenterol. Hepatol. 2021, 19, 1189–1199.e30. [Google Scholar] [CrossRef] [PubMed]
  120. Albenberg, L.; Brensinger, C.M.; Wu, Q.; Gilroy, E.; Kappelman, M.D.; Sandler, R.S.; Lewis, J.D. A Diet Low in Red and Processed Meat Does Not Reduce Rate of Crohn’s Disease Flares. Gastroenterology 2019, 157, 128–136.e5. [Google Scholar] [CrossRef] [PubMed]
  121. Gunasekeera, V.; Mendall, M.A.; Chan, D.; Kumar, D. Treatment of Crohn's Disease with an IgG4-Guided Exclusion Diet: A Randomized Controlled Trial. Dig. Dis. Sci. 2016, 61, 1148–1157. [Google Scholar] [CrossRef] [PubMed]
  122. Szczubełek, M.; Pomorska, K.; Korólczyk-Kowalczyk, M.; Lewandowski, K.; Kaniewska, M.; Rydzewska, G. Effectiveness of Crohn’s Disease Exclusion Diet for Induction of Remission in Crohn’s Disease Adult Patients. Nutrients 2021, 13, 4112. [Google Scholar] [CrossRef] [PubMed]
  123. Yanai, H.; Levine, A.; Hirsch, A.; Boneh, R.S.; Kopylov, U.; Eran, H.B.; Cohen, N.A.; Ron, Y.; Goren, I.; Leibovitzh, H.; et al. The Crohn’s disease exclusion diet for induction and maintenance of remission in adults with mild-to-moderate Crohn's disease (CDED-AD): An open-label, pilot, randomised trial. Lancet Gastroenterol. Hepatol. 2022, 7, 49–59. [Google Scholar] [CrossRef]
  124. Sigall Boneh, R.; Van Limbergen, J.; Wine, E.; Assa, A.; Shaoul, R.; Milman, P.; Cohen, S.; Kori, M.; Peleg, S.; On, A.; et al. Dietary Therapies Induce Rapid Response and Remission in Pediatric Patients with Active Crohn's Disease. Clin. Gastroenterol. Hepatol. 2021, 19, 752–759. [Google Scholar] [CrossRef]
  125. Levine, A.; Wine, E.; Assa, A.; Sigall Boneh, R.; Shaoul, R.; Kori, M.; Cohen, S.; Peleg, S.; Shamaly, H.; On, A.; et al. Crohn’s Disease Exclusion Diet Plus Partial Enteral Nutrition Induces Sustained Remission in a Randomized Controlled Trial. Gastroenterology 2019, 157, 440–450.e8. [Google Scholar] [CrossRef]
  126. Luo, J.; Xie, Y.M.; Wu, M.; Zhao, J.G.; Hu, L.L. Global attitudes on and the status of enteral nutrition therapy for pediatric inflammatory bowel disease. Front. Med. 2022, 9, 1036793. [Google Scholar] [CrossRef]
  127. Damas, O.M.; Garces, L.; Abreu, M.T. Diet as Adjunctive Treatment for Inflammatory Bowel Disease: Review and Update of the Latest Literature. Curr. Treat. Options Gastroenterol. 2019, 17, 313–325. [Google Scholar] [CrossRef]
  128. Algera, J.P.; Demir, D.; Törnblom, H.; Nybacka, S.; Simrén, M.; Störsrud, S. Low FODMAP diet reduces gastrointestinal symptoms in irritable bowel syndrome and clinical response could be predicted by symptom severity: A randomized crossover trial. Clin. Nutr. 2022, 41, 2792–2800. [Google Scholar] [CrossRef]
  129. Cox, S.R.; Prince, A.C.; Myers, C.E.; Irving, P.M.; Lindsay, J.O.; Lomer, M.C.; Whelan, K. Fermentable Carbohydrates [FODMAPs] Exacerbate Functional Gastrointestinal Symptoms in Patients with Inflammatory Bowel Disease: A Randomised, Double-blind, Placebo-controlled, Cross-over, Re-challenge Trial. J. Crohns Colitis. 2017, 11, 1420–1429. [Google Scholar] [CrossRef] [PubMed]
  130. Bodini, G.; Zanella, C.; Crespi, M.; Lo Pumo, S.; Demarzo, M.G.; Savarino, E.; Savarino, V.; Giannini, E.G. A randomized, 6-wk trial of a low FODMAP diet in patients with inflammatory bowel disease. Nutrition 2019, 67–68, 110542. [Google Scholar] [CrossRef] [PubMed]
  131. Cox, S.R.; Lindsay, J.O.; Fromentin, S.; Stagg, A.J.; McCarthy, N.E.; Galleron, N.; Ibraim, S.B.; Roume, H.; Levenez, F.; Pons, N.; et al. Effects of Low FODMAP Diet on Symptoms, Fecal Microbiome, and Markers of Inflammation in Patients with Quiescent Inflammatory Bowel Disease in a Randomized Trial. Gastroenterology 2020, 158, 176–188.e7. [Google Scholar] [CrossRef] [PubMed]
  132. Pedersen, N.; Ankersen, D.V.; Felding, M.; Wachmann, H.; Végh, Z.; Molzen, L.; Burisch, J.; Andersen, J.R.; Munkholm, P. Low-FODMAP diet reduces irritable bowel symptoms in patients with inflammatory bowel disease. World J. Gastroenterol. 2017, 23, 3356–3366. [Google Scholar] [CrossRef] [PubMed]
  133. Melgaard, D.; Sørensen, J.; Riis, J.; Ovesen, T.S.; Leutscher, P.; Sørensen, S.; Knudsen, J.K.; Bundgaard-Nielsen, C.; Ejstrup, J.; Jensen, A.-M.; et al. Efficacy of FODMAP Elimination and Subsequent Blinded Placebo-Controlled Provocations in a Randomised Controlled Study in Patients with Ulcerative Colitis in Remission and Symptoms of Irritable Bowel Syndrome: A Feasibility Study. Nutrients 2022, 14, 1296. [Google Scholar] [CrossRef]
  134. Guo, Z.; Wu, R.; Zhu, W.; Gong, J.; Zhang, W.; Li, Y.; Gu, L.; Li, N.; Li, J. Effect of exclusive enteral nutrition on health-related quality of life for adults with active Crohn's disease. Nutr. Clin. Pract. 2013, 28, 499–505. [Google Scholar] [CrossRef]
  135. Pigneur, B.; Ruemmele, F.M. Nutritional interventions for the treatment of IBD: Current evidence and controversies. Therap. Adv. Gastroenterol. 2019, 12, 1756284819890534. [Google Scholar] [CrossRef]
  136. Brückner, A.; Werkstetter, K.J.; Frivolt, K.; Shokry, E.; Ahmed, M.; Metwaly, A.; Marques, J.G.; Uhl, O.; Krohn, K.; Hajji, M.; et al. Partial enteral nutrition has no benefit on bone health but improves growth in paediatric patients with quiescent or mild Crohn’s disease. Clin. Nutr. 2020, 39, 3786–3796. [Google Scholar] [CrossRef]
  137. Moriczi, M.; Pujol-Muncunill, G.; Martín-Masot, R.; Jiménez Treviño, S.; Segarra Cantón, O.; Ochoa Sangrador, C.; Peña Quintana, L.; González Santana, D.; Rodríguez Martínez, A.; Rosell Camps, A.; et al. Predictors of Response to Exclusive Enteral Nutrition in Newly Diagnosed Crohn´s Disease in Children: PRESENCE Study from SEGHNP. Nutrients 2020, 12, 1012. [Google Scholar] [CrossRef]
Figure 1. Comparison between the Mediterranean diet (upper) and the Western-style diet (lower) pyramids [17].
Figure 1. Comparison between the Mediterranean diet (upper) and the Western-style diet (lower) pyramids [17].
Foods 12 01987 g001
Figure 2. Epidemiological stages of IBD in the world. Stage 1: emergence. Stage 2: acceleration of incidence. Stage 3: compounding prevalence [4]. Developed economies: United States, Canada, Japan, Australia and New Zealand, Europe (except Southeastern Europe). Economies in transition: The Commonwealth of Independent States and Georgia, Southeastern Europe. Developing economies: Africa, Asia (except Japan), Latin America and the Caribbean (modified from [4]).
Figure 2. Epidemiological stages of IBD in the world. Stage 1: emergence. Stage 2: acceleration of incidence. Stage 3: compounding prevalence [4]. Developed economies: United States, Canada, Japan, Australia and New Zealand, Europe (except Southeastern Europe). Economies in transition: The Commonwealth of Independent States and Georgia, Southeastern Europe. Developing economies: Africa, Asia (except Japan), Latin America and the Caribbean (modified from [4]).
Foods 12 01987 g002
Figure 3. PRISMA Flow Diagram.
Figure 3. PRISMA Flow Diagram.
Foods 12 01987 g003
Figure 4. Interactions between food, environmental factors, intestinal microbes and immune system in IBD pathogenesis.
Figure 4. Interactions between food, environmental factors, intestinal microbes and immune system in IBD pathogenesis.
Foods 12 01987 g004
Table 1. Prevalence (%), causes and effects of micronutrient deficiency in IBD patients.
Table 1. Prevalence (%), causes and effects of micronutrient deficiency in IBD patients.
Micronutrient Deficiency Prevalence (%)
UCCDCauses of Deficiencies *Main Effects
Folic acid3554–67GI resections; use of sulfasalazine, methotrexate, cholestyramine; achlorhydria; small intestinal bacterial overgrowth (SIBO).Cause and worsen anemia, hyperhomocysteinemia (causes thromboembolic events), risk of carcinogenesis.
Vitamin B12548Terminal ileitis, resection if the terminal ileum > 20 cm, small intestinal bacterial overgrowth (SIBO).Exacerbate anemia, hyperhomocysteinemia (causes thromboembolic events).
Vitamin C1624Malabsorption, polymorphisms in genes encoding vitamin C transporters. TNF-α also downregulates transcription of transporters necessary for vitamin C uptake.Impaired uptake and utilization of iron, wound healing deficit, bleeding.
Vitamin A **26–9311–50Terminal ileum disease or ileal resection (malabsorption).Associated with severity of the disease, wound healing deficit.
Vitamin D **4070Terminal ileum disease or ileal resection (malabsorption), small intestinal bacterial overgrowth (SIBO)Calcium deficiency, bone mineral density loss, compromised mucosal barrier functions, associated with disease activity, risk of carcinogenesis.
Vitamin E **55Terminal ileum disease or ileal resection (malabsorption).Lipid peroxidation, reduced wound healing.
Vitamin K **4454Terminal ileum disease or ileal resection (malabsorption).Bone mineral density loss, bleeding.
Iron8139Disease in/resection of the proximal small bowel (limited intestinal absorption), rectal bleeding, achlorhydria, small intestinal bacterial overgrowth (SIBO).Anemia, fatigue, abnormal growth and inadequate cognitive development in children and adolescents.
Potassium NA6–20Colectomy/pouch, prednisone, diarrhoea, vomiting, mucosal inflammation (increased secretion).Muscle weakness/cramps, cardiac arrhythmia.
Calcium1013Disease in/resection of the proximal small bowel (limited intestinal absorption), corticosteroid therapy, disturbed metabolism of vitamin D.Bone mineral density loss, osteoporosis, risk of carcinogenesis.
MagnesiumNA 14–88Chronic or severe acute diarrhoea, short gut (reduced intestinal absorption), disturbed metabolism of vitamin D.Bone mineral density loss.
Zinc38-4540-50Diarrhoea, ostomies, high-exit fistulas, chronic malabsorption state due to intestinal inflammation.Oxidative cellular damage, wound healing deficit, dysfunctional epithelial barrier, altered mucosal immunity, increased pro-inflammatory cytokines.
SeleniumNA35–40Impaired intestinal absorption (duodenum, caecum).Inflammation, risk of carcinogenesis.
* Besides the reduced intake, ** fat soluble vitamins. NA: not available.
Table 3. Types of nutritional interventions in IBD patients.
Table 3. Types of nutritional interventions in IBD patients.
Low Fermentable Oligosaccharides,
Disaccharides, Monosaccharides, and Polyols (FODMAP) Diet
Allowed foodsEggs and meat.
Certain cheeses such as brie, camembert, cheddar and feta.
Almond milk, rice, quinoa and oats.
Vegetables such as eggplant, potatoes, tomatoes, cucumbers and zucchini.
Fruits such as grapes, oranges, strawberries, blueberries and pineapple.
Restricted foodsDairy-based products, wheat-based, beans and lentils.
Vegetables, such as artichokes, asparagus, onions and garlic.
Fruits such as apples, cherries, pears and peaches.
Other DetailsThis diet consists of three steps: (1) elimination of all FODMAPs for 4–6 weeks, (2) gradual reintroduction aimed to individuate foods related to symptomatology, (3) personalization of diet.
Enteral Nutrition
Allowed foodsElemental formulas (amino acids), semi-elemental formulas (oligopeptides), or polymeric formulas (whole proteins).
Restricted foodsAll other foods.
Other DetailsEnteral nutrition can be administered both by nasogastric tube or ingested. Enteral nutrition can be “exclusive” when formulas completely substitute meals, or “partial” when it is administered together with meals. Partial enteral mutrition is often prescribed in association to Crohn’s disease exclusion diet.
Specific Carbohydrate Diet (SCD)
Allowed foodsAdditive-free meats, poultry, fish and shellfish.
Additive-free and sugar-free oils, white vinegar, cider and mustard.
Additive-free and sugar-free coffee, tea and fruit juice.
All-natural, sugar-free peanut butter.
Cheeses such as sharp cheddar, colby, swiss and dry curd cottage cheese.
Fresh, frozen, raw or cooked vegetables, including string beans.
Fresh fruits or frozen, cooked or dried fruits without added sugar.
Eggs.
Homemade yogurt that ferments for at least 24 h.
Honey.
Legumes such as dried navy beans, lentils, peas, split peas and lima beans. Also, unroasted cashews and unroasted peanuts in the shell.
Nuts, peanuts and nut flours.
Restricted foodsGrains such as barley, corn, oats, quinoa, rice and wheat.
Grain products such as bread, cereal and pasta.
Candy, chocolates and other products made with sugar, high fructose corn syrup, or fructo-oligosaccharides (FOS). Canned or processed meats.
Canned vegetables with additives.
Certain legumes such as soybeans, chickpeas and bean sprouts.
Dairy products high in lactose such as mild cheddar, store-bought yogurt, milk, cream, ice cream and sour cream.
Powdered spices such as curry, garlic and onion.
Seaweed.
Starches such as potatoes, sweet potatoes and turnips.
Sugars including molasses, corn syrup, maple syrup, fructose, sucrose and other processed sugars.
Mediterranean Diet
Allowed foodsVegetables, fruits, cereals, nuts, legumes, unsaturated fat such as olive oil.
Medium intake of fish, dairy products, wine.
Restricted foodsSaturated fat, meat, processed foods, processed meat and sweets
Crohn’s Disease Exclusion Diets
Allowed foodsPhase I
Mandatory daily foods: fresh chicken breast 150–200 g, 2 eggs, 2 bananas, 1 fresh apple, 2 potatoes (potatoes must be cooked and refrigerated before use).
Allowed daily foods: fresh strawberries, fresh melon (1 slice), rice flour, white rice and rice noodles (unlimited), 2 tomatoes (additional allowed for cooking), 2 cucumbers (2 medium size), 2 avocado halves, 1 arrot, spinach 1 cup uncooked leaves, lettuce (3 leaves), onion, fresh green herbs (e.g., basil, parsley, coriander, rosemary, thyme, mint, dill), 1 glass freshly squeezed orange juice from fresh oranges (not from cartons or bottles), water, sparkling water, salt, pepper, paprika, cinnamon, cumin, turmeric, 3 tablespoons of honey, 4 teaspoons of sugar, fresh ginger and garlic cloves, lemons and limes.
Foods allowed only once a week: fresh lean fish (not deep fried, dietitian guidance required).
Phase II
Mandatory daily foods: fresh chicken breast 150–200 g, 2 eggs, 2 bananas, 1 fresh apple, 2 potatoes (potatoes must be cooked and refrigerated before use).
Allowed daily foods: fresh strawberries, fresh melon (1 slice), Rice flour, White rice and rice noodles (unlimited), 2 Tomatoes (additional allowed for cooking), 2 Cucumbers (2 medium size), 2 Avocado halves, 1 Carrot, 1 cup uncooked spinach leaves, lettuce (3 leaves), onion, fresh green herbs (e.g., basil, parsley, coriander, rosemary, thyme, mint, dill), 1 glass freshly squeezed orange juice from fresh oranges (not from cartons or bottles), water, sparkling water, salt, pepper, paprika, cinnamon, cumin, turmeric. 3 tablespoons of honey, 4 teaspoons of sugar, fresh ginger and garlic cloves, one slice whole grain bread daily, quinoa, 3 tablespoons of cooked lentils or peas, 6 almonds or walnut halves (unprocessed), baking soda.
Foods allowed only once a week: fresh lean fish (not deep fried, dietitian guidance required), 200 gr Sirloin or fillet steak (maximum), 1 slice whole grain bread (maximum), 1 can of tuna (in olive or canola oil) drained, ½ cup of oatmeal or cut oats.
Additional daily foods from week 7: broccoli, cauliflower 2 florets daily, 4 fresh mushrooms (not canned), ½ red bell pepper, 1 zucchini or slice squash, 1 pear or kiwi or ripe nectarine.
Additional daily foods from week 10: most vegetables (restricted amounts with dietitian guidance), most fruits (restricted amounts with dietitian guidance), quinoa, 3–4 tablespoons of cooked lentils or peas.
Restricted foodsDairy, animal fat, wheat, emulsifiers, artificial sweeteners, other cuts or parts of chicken, other sources animal or soy protein, carrageenan, maltodextrins (and sucralose), sulfite containing foods, xanthan gum, packaged/canned/frozen precooked foods, doughs, baked goods, frozen, canned fruits and vegetables, oral iron supplements, soy or gluten-free products, ready to use sauces, syrups, spreads dressings, margarine, butter, vinegar, soy sauce, ketchup, mayonnaise, alcoholic beverages, soft drinks, juices, deep-fried or oily foods.
Other DetailsEach phase has a duration of 6 weeks. CDED is often associated with polymeric formulas which, during phase 1, represent 50% of the caloric intake. Fruit and vegetables are progressively reintroduced during phase II.
IgG 4-guided diet
Allowed foodsFoods not associated with IgG4 reactivity.
Restricted foodsFoods associated with IgG4 reactivity.
Other DetailsIgG4-guided exclusion diet is a personalized approach excluding and replacing in each patient 4 food types with the highest IgG4 titration. Screening is performed on 16 food types: milk, peanuts, soya, shrimp, egg, tomato, pork, beef, cod fish, potato, wheat, yeast, cheddar cheese, chicken, lamb and rice.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Saracino, I.M.; Spisni, E.; Imbesi, V.; Ricci, C.; Dussias, N.K.; Alvisi, P.; Gionchetti, P.; Rizzello, F.; Valerii, M.C. The Bidirectional Link between Nutritional Factors and Inflammatory Bowel Diseases: Dietary Deficits, Habits, and Recommended Interventions—A Narrative Review. Foods 2023, 12, 1987. https://doi.org/10.3390/foods12101987

AMA Style

Saracino IM, Spisni E, Imbesi V, Ricci C, Dussias NK, Alvisi P, Gionchetti P, Rizzello F, Valerii MC. The Bidirectional Link between Nutritional Factors and Inflammatory Bowel Diseases: Dietary Deficits, Habits, and Recommended Interventions—A Narrative Review. Foods. 2023; 12(10):1987. https://doi.org/10.3390/foods12101987

Chicago/Turabian Style

Saracino, Ilaria Maria, Enzo Spisni, Veronica Imbesi, Chiara Ricci, Nikolas Konstantine Dussias, Patrizia Alvisi, Paolo Gionchetti, Fernando Rizzello, and Maria Chiara Valerii. 2023. "The Bidirectional Link between Nutritional Factors and Inflammatory Bowel Diseases: Dietary Deficits, Habits, and Recommended Interventions—A Narrative Review" Foods 12, no. 10: 1987. https://doi.org/10.3390/foods12101987

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop