Abstract
Dysfunction of gut barrier is known as “leaky gut” or increased intestinal permeability. Numerous recent scientific evidences showed the association between gut dysfunction and multiple gastrointestinal tract (GI) and non‐GI diseases. Research also demonstrated that food plays a crucial role to cause or remedy gut dysfunction related to diseases. We reviewed recent articles from electronic databases, mainly PubMed. The data were based on animal models, cell models, and human research in vivo and in vitro models. In this comprehensive review, our aim focused on the relationship between dietary factors, intestinal permeability dysfunction, and related diseases. This review synthesizes currently available literature and is discussed in three parts: (a) the mechanism of gut barrier and function, (b) food and dietary supplements that may promote gut health, and food or medication that may alter gut function, and (c) a table that organizes the synthesized information by general mechanisms for diseases related to leaky gut/intestinal permeability and associated dietary influences. With future research, dietary intervention could be a new target for individualized disease prevention and management.
Keywords: diet, intestinal permeability, leaky gut, microbiota, tight junction
Short abstract
This review summarizes current literature, mainly from PubMed, on the mechanisms of gut barrier and function, including food and dietary supplements or medications that promote or alter gut health. It also provides a comprehensive and clear table about basic mechanisms for disease‐related leaky gut/increased intestinal permeability with associated dietary influences.
1. INTRODUCTION
The gut barrier is the most important defense system of human body. The surface of intestinal tract is approximately 7000–8000m2, the largest interface between human body and external environment (Bengmark, 2013; De Santis et al., 2015; Lopetuso et al., 2015; Suzuki, 2013). The gut barrier includes mucus layer, commensal microbiota, and single layer of intestinal epithelium. The 20‐μm‐thick layer of intestinal epithelium contains enterocytes, endocrine cells, microfold (M) cells, goblet cells, and Paneth cells in the small intestine (Farré et al., 2020; Kinashi & Hase, 2021). There are more goblet cells and no Paneth cells in the colon. Lamina propria and muscularis mucosa of intestinal mucosa support and articulate the epithelial layer. Under physiological circumstances, the epithelial cells are joined together by the tight junction (TJ) and the adherens junction (AJ), thus forming a contiguous and relatively impermeable membrane. TJ and AJ are protein complexes. TJ connects in the apex of adjacent cells to regulate ion, solute, and microbe diffusion across the gut barrier and maintain homeostasis (Seo et al., 2021). AJ is more basal than TJ. Besides the role to link adjacent cells, AJ does actin cytoskeleton regulation, cell signaling, and gene transcription (Guo et al., 2007; Seo et al., 2021).
Impaired integrity of intestinal barrier and structure of the TJ barrier may trigger local or systemic inflammation and diseases. It results in increasing intestinal permeability which allows antigens, endotoxins, pathogens, and other proinflammatory substances to pass through intestinal barrier into circulation (Fukui, 2016). Ongoing studies showed that dietary factor, extra stress, and lack of physical activities could change gut microbiota, impair epithelial cell TJ and AJ, cause gut dysbiosis, and increase intestinal permeability (Lopetuso et al., 2015; Plaza‐Díaz et al., 2020). Physical exercises regulate gut microbiota diversity; increase abundance of short‐chain fatty acids (SCFAs); and provide long‐term benefits to the gut barrier (Plaza‐Díaz et al., 2020). Unexpected or prolonged psychological and physiological stress threatens intestinal barrier homoeostasis which responds through gut–brain axis, hypothalamic–pituitary–adrenal axis and sympathetic nerve system (SNS), leading to autoimmune and other disorders (Bajaj et al., 2019; Camilleri, 2019; Foster et al., 2017; Ilchmann‐Diounou & Menard, 2020; Karl et al., 2017; Yue et al., 2017). Food provides nutrition to human body, and diet is also the most important factor to modify and regulate or influence diversity of gut microbiota. It may alter the intestinal barrier and impact gut homeostasis. Disruption of intestinal barrier and intestinal permeability may induce inflammatory states or diseases in multiple systems (Camilleri et al., 2019; Khoshbin & Camilleri, 2020).
Many measurement tools exist to measure intestinal permeability ex vivo and in vitro, such as Ussing chamber, assessing urinary excretion probes, like the lactulose:mannitol ratio (LAMA), sucralose, sucrose, PEG4000/400, and 51Cr‐EDTA; bacterial‐related tests, like assays of serum lipopolysaccharides (LPSs) and other bacteria toxins, using IgA/IgM responses to Gram‐negative bacteria, plasma D‐lactate, fecal Butyrate production, fecal Hemolysin test; tests for biomarkers like plasma citrulline, plasm FABP, plasma and urine α‐glutathione S‐transferase (αGST), serum zonulin enzyme‐linked immunosorbent assay (ELISA), urine claudin‐3, fecal calprotectin, and α1‐anti‐trypsin test; histological approaches like Western blot for TJ expression, Goblet cell analysis, and shedding of epithelium (Bischoff et al., 2014; Duerksen et al., 2010; Fasano, 2020; Odenwald & Turner, 2013); others like serum fluorescein isothiocyanate dextran (FITC‐d; Vuong et al., 2021), and endomysial (EMA) and tissue transglutaminase (TTG) antibodies specifically for celiac disease (Duerksen et al., 2010), and so on. Human gut biopsies, confocal endomicroscopy, and endoscopic mucosal impedance also are used for assessing intestinal permeability (Bischoff et al., 2014; Camilleri, 2019). However, there are no current gold standard tests for gut barrier function (Camilleri, 2019).
Given the current lack of comprehensive review on how dietary therapy approaches can play an important role in the prevention, management, and treatment of diseases, we summarize dietary factors in this review.
2. MECHANISMS OF INTESTINAL EPITHELIAL BARRIER
In a review of the current understanding, intestinal epithelium provides a dynamic permeable barrier to selectively absorb nutrients while simultaneously separating mucosal tissues from luminal commensal bacteria, pathogens, and dietary antigens. Epithelial barrier functions include functions of an apical TJ, subjacent junction AJ, and desmosome (Luissint et al., 2016). The imbalance of intestinal microbiota is known as gut dysbiosis, which causes increasing intestinal permeability or leaky gut syndrome (LGS). It directly or indirectly triggers immune system reaction. Functional food or non‐functional food may change the intestinal microbiota bidirectionally (Ferreira et al., 2020).
2.1. Physiological structure of epithelium and its function
A well‐structured network of intestinal epithelial and stromal cells provides efficient nutrients and physical barrier against potentially harmful entities, including microorganisms, dietary antigens, or other noxious agents. There are multi‐layer physicochemical barriers in the intestinal mucosa. The outer layer is the microbiota that competes and represses pathogens. The next is mucus layer, which contains antimicrobial peptides (AMPs) and secretory IgA. The mucus layer consists of gel‐forming mucins including water and glycosylated proteins. It prevents microbiota and large molecules contacting the epithelial cells. The mucus layer in the colon forms a double layer, an inner and an outer layer. The third layer is a single layer of columnar epithelial cells (Seo et al., 2021). Epithelial stem cells can differentiate into four cells: enterocytes, goblet cells, enteroendocrine cells, and Paneth cells (in small intestine) and the intestinal epithelium renews approximately every five days (Bischoff et al., 2014). The 80% of epithelial cells are enterocytes which form an effective barrier to protect the internal milieu. The epithelial cells absorb beneficial molecules which include broken down proteins, fats, sugars, as well as water, electrolytes, vitamins, and bile salts from the gut lumen and transport them into the body. Enterocytes also secrete hormones such as leptin. Paneth cells are mainly located at the crypt and produce AMPs that play an important role in host immunity. Goblet cells secrete mucus forming a mucous layer. The enteroendocrine cells produce GI hormones (Bischoff et al., 2014; Farré et al., 2020). M cells are in the epithelium covering mucosa‐associated lymphoid tissues, such as the Peyer's patches of small intestine. M cells actively transport luminal antigens to underlying lymphoid follicles to initiate an immune response (Allaire et al., 2018). The epithelial cells are interconnected tightly with TJ and AJ. The deep part of the gut barrier contains complex network of immune cells, known as gut‐associated lymphoid tissue (GALT). The underlying lamina propria contains dendritic cells (DCs), intraepithelial DCs, macrophages, intraepithelial lymphocytes (IEL), T regulatory cells (T Regs), TCD4+ lymphocytes, B lymphocytes, and plasma cells. Plasma cells release secretory immunoglobulin A (slgA; Rescigno, 2011; Schenk & Mueller, 2008).
2.2. Tight junction, adherens junction, and desmosomes
The essential role of intestinal epithelium is to separate intestinal luminal contents that leak to circulatory system (Odenwald & Turner, 2013). Intestinal epithelial cells are sealed by protein complexes of apical TJs, subjacent AJs and desmosomes (DMs). Epithelial barrier function is mediated by TJs, AJs, and DMs (Luissint et al., 2016). The TJ plays important role to maintain the intestinal permeability. The AJs along with desmosomes provide strong adhesive bonds between the epithelial cells, and also intercellular communication, but do not determine paracellular permeability (Suzuki, 2013). AJs and DMs mediate direct cell‐to‐cell contacts. The AJ's key transmembrane protein, E‐cadherin, mediates calcium‐dependent homotypic intercellular adhesions. Two proteins of AJ are nectin–afadin and cadherin–catenin complexes. The nectin–afadin are responsible for the maturation of AJ, and the E‐cadherin‐β‐catenin interacts with components of the cytoskeleton, and provides organization and maintenance of AJ (Cardoso‐Silva et al., 2019). DMs provide anchorage sites for intermediate filaments, which are important for the maintenance of tissue architecture, and provide mechanical strength to the epithelium (Luissint et al., 2016).
The TJ biochemical structure is as follows. The TJ consists of transmembrane proteins: claudin, occludin, tricellulin, junctional adhesion molecule‐A (JAM‐A), intracellular plaque proteins, such as zonula occludens (ZO), and cingulin (Lerner & Matthias, 2015; Suzuki, 2013, 2020). Claudins contain 27 tetraspan integral membrane proteins in humans. Claudins oligomerize in cis and trans, and provide diverse combinations and the complement in TJ strands influences cellular barrier function. Based on permeability, claudins have been grouped into barrier‐forming isoforms (claudin 1, 3, 4, 5, and 18), which provide barrier to macromolecules and ions, thereby increasing barrier tightness, and pore‐forming isoforms (claudin 2, 10, 12, 15), which select pores to ions and water, which increases paracellular permeability (Luissint et al., 2016; Suzuki, 2020). Claudins are responsible for TJ and epithelial barrier formation, constitution of TJ strands, and cytoskeleton organization (Cardoso‐Silva et al., 2019). Occludin belongs to TJ‐associate MARVEL domain‐containing proteins (Luissint et al., 2016). Occludin is important in TJ stability and barrier function (Saitoa et al., 2021). Claudin and occludin form homotypic complexes between cells. The tricellulin junctions contact three adjacent cells and regulate the permeability of small ions and solutes. Tricellulin and occludin maintain epithelial barrier integrity by helping to form and stabilize TJ strand branching points (Saitoa et al., 2021). ZO proteins include ZO‐1, ZO‐2, and ZO‐3. ZO proteins are scaffolding proteins which provide the structural basis for the assembly of multiprotein complexes at the cytoplasmic surface of intercellular junctions. ZO‐1, 2, and 3 connect occludin and claudin to actin cytoskeleton and maintain the TJ formation (Suzuki, 2013, 2020; Zihni et al., 2016). JAM‐A is an immunoglobulin superfamily (IgSF) – cell adhesion molecule (CAM). JAM‐A recruit protein scaffolds to specific sites of cell–cell adhesion and assemble signaling complexes (Steinbacher et al., 2018). JAM plays a role in barrier regulation and TJ maintenance (Cardoso‐Silva et al., 2019). Cingulin is an actin cytoskeleton‐associated protein, and one role is for TJ assembly (Suzuki, 2013).
Impairment of intestinal TJ barrier causes increasing intestinal permeability, thereby resulting in intestinal and systemic inflammation and disorders. Currently, there are no effective therapies available that specifically target the tightening of the intestinal TJ barrier or no FDA‐approved agents to treat the epithelial barrier dysfunction (Al‐Sadi et al., 2021; Odenwald & Turner, 2017). According to research, using nutrition and dietary factors could effectively maintain and protect TJ barrier dysfunction. This review summarizes food influences on TJ barrier.
2.3. Gut microbiota
Obrenovich et al. (Obrenovich et al., 2020) identified the novel name “holobiota” to describe all the microbiota that live within human's gut, including entire and diverse collection of microbes, mycobiota, commensals, pathogens, viruses, viroids, protozoan parasites, and their genomes, nucleic acid and so on. There are more than 100 trillion microorganisms which total 1–2 kg in mass in human's GI tract and each individual has a unique gut microbiota profile. The gut microbiota provides nutrient metabolism, participates in growth and immune regulation, eliminates pathogenic microorganisms, and maintains gut barrier integrity and normal homeostasis. Research shows that gut microbiota benefits the hosts by producing vitamins, preventing the growth of harmful bacteria, training the immune system, and fermenting unused food. The gut microbiota plays an important role in regulating the intestinal mucosa permeability and changing the microbial community impacts on gut mucosal barrier function. The bacterial population is currently the most well characterized in the literature. The more abundant and diverse the microbiota, the more powerful it is to fight external threats throughout the life for human being (Rinninella, Pauline Raoul, et al., 2019; Sung et al., 2016). The human gut virome mainly consists of bacteriophages, eukaryotic viruses, and other plant‐ and animal‐derived viruses along with food (Carding et al., 2017). Viruses could be pathogenic or beneficial for the hosts. GI microbiota of healthy hosts has been detected following eukaryotic viruses' families: Adenoviridae, Anelloviridae, Astroviridae, Caliciviridae, Circoviridae, Coronaviridae, Parvoviridae, Picobirnaviridae, Picornaviridae, Polyomaviridae, and Reoviridae (Julio‐Pieper et al., 2021).
The individualized microbiota is shaped in early life and its diversity increases with age until mature adult. The majority of GI tracts in stable adults are representatives of three main bacterial phyla: Firmicutes (Lachnospiraceae and Ruminococcaceae), Bacteroidetes (Bacteroidaceae, Prevotellaceae, and Rikenellaceae), and Actinobacteria (Bifidobacteriaceae and Coriobacteriaceae), which may be influenced by genetics, environment, diet, lifestyle, stress, and gut physiology (Maukonen & Saarela, 2015; Rea et al., 2020; Rinninella, Cintoni, et al., 2019; Tidjani Alou et al., 2016). In healthy human adults, the dominant five bacterial phyla were reported as Firmicutes, Bacteroidetes, Actinobacteria, Proteobacteria, and Verrucomicrobia. Among them, Firmicutes and Bacteroidetes account for more than 90% (Hansen & Sams, 2018). With the aging of the body, there are obvious changes in the diversity, structure ratio, and metabolism of gut microbes, leading to the imbalance of homeostasis, such as a decrease in anaerobic bacteria like Bifidobacterium spp. and an increase in Clostridium and Proteobacteria (Odamaki et al., 2016). Aside from age, there are other factors that affect the human gut microbiota, such as unbalanced diet, genetics, medications, and diseases (Maukonen & Saarela, 2015). The monozygotic twins have been reported with more similar microbiota than control group (Turnbaugh et al., 2010). Medication is an important external factor affecting human gut microbiota (Maukonen & Saarela, 2015). Daily exercises may increase resistance of the intestinal barrier through diversification of gut microbiota through a Firmicutes enrichment microbiota: Clostridiales, Roseburia, Lachnospiraceae, and Erysipelotrichaceae, producing SCFAs, with increased expression of TJ proteins (Bai, Hu, & Bruner, 2019; Monda et al., 2017).
A healthy gut microbiota is recognized as a relatively balanced state called normobiosis (Doré & G Corthier, 2010). Intestinal microbiota is significant for regulating the immune system during pathogenesis conditions, such as a decrease in bacterial diversity and an aberrant increase in some commensal bacteria (Al‐Sadi et al., 2021). The imbalance of microbial communities in or on the body is called dysbiosis. Dysbiosis can alter metabolite profile because metabolites are produced by the microbiota (Ferreira et al., 2020; Kim, 2018; Raza et al., 2019; Tang et al., 2019). Altered balance in the host–microbe relationship may impair the development of the immune system, potentially leading to diseases (Patterson et al., 2014). The recent research has shown that imbalance of gut microbiota can cause a range of different diseases through following axes: Brain–Gut axis/Gut–brain axis (Camara‐Lemarroy et al., 2018), Gut–brain‐muscle axis (van Krimpen et al., 2021), Gut–brain neuroendocrine metabolic (GBNM) axis (Obrenovich, 2018), Gut–joint axis (Yang et al., 2016), Gut–kidney axis (Ondrussek‐Sekac et al., 2021), Gut–liver axis (Plaza‐Díaz et al., 2020; Preveden et al., 2017; Sung et al., 2016), Gut–liver–brain axis (Woodhouse et al., 2018), Gut–liver–immune system axis (Woodhouse et al., 2018), Gut–liver–kidney axis (Amornphimoltham et al., 2019), Gut–lung axis (Trivedi & Barve, 2020), Gut–retina axis (Rinninella et al., 2018), Hypothalamic–pituitary–adrenal (HPA) axis (Chu et al., 2018), Hypothalamic–pituitary–thyroid (HPT) axis (Krysiak et al., 2019), Microbiota–Gut–Immune–Glia (MGIG) axis (Rudzki & Maes, 2020), Thyroid–gut axis (Cayres et al., 2021; Knezevic et al., 1769), and other routes, through metabolism, inflammation, and immunity. Human enteric nervous system (ENS) is located within the gut wall and is connected with the enteroendocrine, GI immune system, the peripheral nervous system (PND), the central nervous system (CNS), and gut microbiota to regulate the intestinal epithelial barrier permeability and keep dynamic balance (Niesler et al., 2021; Neunlist et al., 2003). Diet may restore dysbiosis to normobiosis through immune regulation (Baraniya et al., 2020; Beliaeva et al., 2013).
2.4. Immunological function of intestinal barrier
During gut microbiota–host interaction, gut releases cytokines, chemokines, neurotransmitters, neuropeptides, endocrine messages, and microbial byproducts including SCFAs, LPS, peptidoglycans, among others (Rea et al., 2020). Gut microbiota produce metabolites including carbohydrate metabolites, amino acid metabolites, and bile acid metabolites, which regulate host immune system through histone deacetylases (HDACs), G‐protein coupled receptors (GPCRs), Aryl hydrocarbon receptor (AhR), and so on, to regulate neutrophil chemotaxin, macrophages, DCs, T‐Cells, IgA and IgG, and cytokines. The results can be barrier function and tolerance or inflammation (Kim, 2018).
In the human colon, anaerobic fermentation of undigested nutrients, such as dietary fiber, resistant starch, and complex polysaccharides can be recognized by GPCRs and metabolized by the microbiota, resulting in the synthesis of SCFAs. SCFAs are the major metabolic products of coliform bacteria. SCFAs contain <6 carbon chains, including acetate (60%), propionate (25%), and butyrate (15%), which consider the major energy substrate for colonocytes to maintain intestinal barrier integrity and homeostasis. SCFAs fuel and fortify epithelial cells, regulate macrophages and DCs through GPCRs and HDACs, and also fuel B cells and promote their differentiation into IgA/IgG‐producing plasma B‐cells. SCFAs may influence GI and systemic health through immunomodulatory and anti‐inflammatory effects. It can induce immune tolerance via Treg cells. Recent studies have shown that SCFAs can maintain normal colorectal function by regulating the metabolism of colon cells, thereby preventing the occurrence of diseases (Ferreira et al., 2020; Kim, 2018; Rinninella, Cintoni, et al., 2019; Tang et al., 2019).
LPS, peptidoglycans, muramyl‐dipeptides, bacterial DNA, etc. are produced by bacteria and translocated from the gut to mesenteric lymph nodes. Gram‐negative bacteria have outer membranes, in part comprised of LPS, also known as a bacterial endotoxin. LPS can increase immune permeability, cross immune barrier, and result in inflammation. LPS can alter signaling of Toll‐like receptors (TLRs), related to leaky gut syndrome (Obrenovich et al., 2020; Plaza‐Díaz et al., 2020; Sperandeo et al., 2017).
Cytokines are produced by lymphokines, monokines, chemokines, interleukins, interferons, colony‐stimulating factors, and growth factors. Cytokines involve regulation of immune system pathways (Ramani et al., 2015). The main proinflammatory cytokines are interleukin (IL)‐1β, IL‐6, IL 12, tumor necrosis factor‐α (TNFα), interferon‐ γ (IFN‐ γ), and nuclear factor kappa‐activated B cells (NF‐κB) and the main anti‐inflammatory cytokines are IL 4, 10, 11, 13, and transforming growth factor‐β (TGF‐β). Pro‐inflammatory cytokines cause a disturbance in intestinal TJ barrier, allowing increased intestinal permeability (Bamias et al., 2012; Fang, 2018; Opal & DePalo, 2000; Ramani et al., 2015; Seo et al., 2021).
The host innate immune system detects microorganisms and responds to their stimuli mainly through recognition of TLRs. TLRs recognize invading microbes and activate immune cell response. TLRs are crucial in the innate response to pathogens, in that they recognize and respond to pathogen‐associated molecular patterns (PAMPs) and damage‐associated molecular patterns (DAMPs), which lead to activation of intracellular signaling pathways and altered gene expression. Myeloid differentiation factor 88 (MyD88) is the key adaptor protein for intracellular signaling that mediates multiple TLRs. The host innate immune system detects microorganisms and responds to their stimuli mainly through recognition of TLRs (Kawasaki & Kawai, 2014; Mukherjee et al., 2016).
Intestinal epithelial mucus contains AMPs such as α‐ and β‐defensins secreted by Paneth cells and sIgA at mucosal surfaces which can protect from invasion of pathogens like coating bacteria, fungi, and enveloped virus, and neutralize bacterial toxins (Camilleri et al., 2019; Ouellette, 2010; Pabst, 2012).
Many factors can alter intestinal permeability such as dietary factors, epithelial damage, mucus layer dysfunction, and gut microbiota modification (Bischoff et al., 2014). Environmental factors like diet or drugs may cause dysbiosis, which impairs epithelial barrier function and elicits proinflammatory cytokines. This process damages TJ integrity and increase gut permeability, leading to leaky gut syndrome (Lopetuso et al., 2015). Compromised intestinal barrier function has been associated with intestinal and systemic diseases (Odenwald & Turner, 2017).
3. FOOD/FOOD COMPONENTS AND DIETARY SUPPLEMENTS MAY PROMOTE GUT HEALTH AND FUNCTION
Diet or food‐derived compounds and their metabolites may have broad effects for regulating gut microbial composition and intestinal permeability directly or indirectly; however, those effects are not thoroughly understood. The diversity of gut microbiota has been implicated in the synthesis of vitamins, secondary bile acids, neurotransmitters, and metabolism and degradation of dietary components (Camilleri et al., 2019; Peron et al., 2020).
3.1. Diets
3.1.1. Mediterranean diet
Mediterranean diet (MD) was first described by the nutritionist Ancel Keys in 1945 and people who live in Spain, Greece, Southern Italy, and other countries facing the Mediterranean basin have MD as a typical dietary habit. MD includes high intake of fruits, vegetables, legumes, nuts, and minimally processed cereals, moderate high consumption of fish and olive oil, low‐to‐moderate intake of wine during meals, low‐to‐moderate amount of yogurt and cheese, and low consumption of saturated fats, red meat, and meat products. MD contains high level of antioxidants, polyunsaturated fatty acids (PUFAs), monounsaturated fatty acid (MUFA), and polyphenols. The main fat of MD is from extra‐virgin olive oil (EVO). The EVO is the key component of MD which can interfere with arachidonic acid and NF‐κB signaling pathways and ameliorate intestinal permeability. The EVO may decrease IL‐1β, TGFβ, IL‐6 and decrease chronic inflammation. Research demonstrated that MD increases SCFAs, microbiota diversity and stability, such as increasing Bifidobacteria, Lactobacillus, Lachnospiraceae, and Bacteroidetes and decreasing Clostridium and Enterobacteria. MD increases IL‐10 and IL 22, and decreases IL‐17, and therefore improves immune function. Studies showed that MD is beneficial for obesity, type 2 diabetes, inflammatory diseases, cardiovascular diseases, stroke, cancer, Parkinson's disease, and Alzheimer's disease (Braga et al., 2013; Cariello et al., 2020; Filippis et al., 2016; Laura Soldati et al., 2018; Lourida et al., 2013; Psaltopoulou et al., 2013; Rinninella, Cintoni, et al., 2019; Sofi et al., 2008).
3.1.2. Ketogenic diet
Ketogenic diet (KD) was established in the 1920s and continues to be used worldwide for the treatment of drug‐resistant epilepsy, which is about one‐third of epilepsy patients (Fan et al., 2019), especially for pediatric epilepsy patients (Arulsamy et al., 2020). KD is a high‐fat, sufficient protein and very low‐carbohydrate diet and the fat to protein and carbohydrate ratio is about 4:1 (Dahlin & Prast‐Nielsen, 2019). Studies showed that KD made a significant difference in gut microbiota diversity: decreasing Bifidobacteria, Eubacterium rectale, Dialister, Proteobacteria, and Firmicutes, and increasing Enterobacteria, Desulfovibrio spp, Parabacteroides, Bacteroidetes, Actinobacteria, and Akkermansia (Arulsamy et al., 2020; Rinninella, Cintoni, et al., 2019). However, it may decrease total bacteria abundance and decrease SCFAs (Rinninella, Cintoni, et al., 2019). Epilepsy could be triggered by commensal microbiota via elevating IL‐6 and IL‐1β and driving Th17 responses. KD has been shown to improve epilepsy frequency or duration (Arulsamy et al., 2020). Very low‐carbohydrate KD can decrease serum levels of glucose and insulin growth factor (IGF) and delay cancer progression. Gut microbiota influences the brain through endocrine, immune, and metabolic systems via MGB/GMB axis. In addition to epilepsy, KD is beneficial for Glucose transporter 1 (GLUT1) Deficiency Syndrome, autism spectrum disorder, obesity, cancer, migraine, glaucoma, multiple sclerosis, Parkinson's disease, and Alzheimer's disease. Unfortunately, long‐term use of KD may have some adverse effects, including constipation, hypercholesterolemia, etc. It has been recommended to add probiotics, vitamins, and minerals to KD (Ersoz Alan & Gulerman, 2019; Fan et al., 2019; Laura Soldati et al., 2018; Olson et al., 2018; Rinninella, Cintoni, et al., 2019; Tagliabue et al., 2017).
3.1.3. Low‐FODMAP diet
Low fermentable oligosaccharide, disaccharide, monosaccharides, and polyols (FOMAP) diet was created by Monash University in 2004 (Rinninella, Cintoni, et al., 2019). FODMAPs are a group of carbohydrates from fructose, fructans, lactose, polyols and galactooligosaccharides that are poorly absorbed in small intestine and subsequently fermented in the small or large intestine which cause GI discomfort and symptoms. High‐FODMAP diet includes grains: wheat‐, rye‐, barley‐based grains, semolina, etc.; fruits: ripe banana, apples, pears, cherries, watermelon, peaches, mango, dry fruits, etc.; vegetables: artichokes, onion, garlic, leeks, asparagus, cauliflower, beetroot, peas, etc.; dairy: cow's milk, ice cream, soy milk, yogurt, soft cheese, etc.; nuts, seeds, and legumes: baked beans, butter beans, kidney beans, soybeans, lentils, etc. Low‐FODMAP diet includes grains: gluten‐free products, quinoa, amaranth, brown rice, bulgur, etc.; fruits: small firm banana, avocado, blueberries, strawberries, raspberries, kiwi, lemon, oranges, papaya, pineapple, etc.; vegetables: broccoli, carrots, arugula, kale, lettuce, spinach, tomatoes, zucchini, etc.; dairy and alternatives: lactose free, almond, coconut or rice‐based milk, hard or aged cheese, etc.; nuts, seeds and legumes: almonds, hazelnuts, macadamia nuts, pecans, pine nuts, walnuts, chia seeds, pumpkin seeds, sesame seeds, sunflower seeds, etc. Peeled apples and pears are included in the low‐FODMAP Diet. Excessive intake of FODMAP causes bacteria overgrowth in small intestine which increase intestinal permeability and induce colon epithelial irritation or injury. Low‐FODMAP diet decreases total bacteria abundance, such as decreasing Bifidobacteria, Ruminococcus gnavus, Clostridium, F. prausnitzii, and Akkermansia. Low‐FODMAP diet has been used to treat inflammatory bowel disease (IBD) and irritable bowel syndrome (IBS; Guagnozzi et al., 2012; Martín et al., 2022; Mazzawi et al., 2013; Nanayakkara et al., 2016; Ostgaard et al., 2012; Rinninella, Cintoni, et al., 2019).
3.2. Probiotics & prebiotics
3.2.1. Probiotics
According to the FAO/WHO, probiotics are defined as “live microorganisms that, when administered in adequate amounts, confer a health benefit on the host” (FAO/WHO, 2002). The main currently used probiotic blend contains the most commonly lactic acid‐producing Bifidobacterium and Lactobacillus spp. (Maukonen & Saarela, 2015). The action of probiotics includes increasing microbial diversity, competing with pathogens, producing bacteriocin, producing vitamins and SCFAs, enhancing immunity such as increasing IgA and C‐creative protein (CRP) and increasing anti‐inflammatory cytokine production like IL‐10 reducing intestinal permeability and oxidative stress, improving the mucosal integrity and barrier function, and producing neurotransmitters (Angelisa et al., 2019; Ballan et al., 2020; Kim, Keogh, & Clifton, 2018; Singh et al., 2017). Probiotics are also effective for small intestinal bacterial overgrowth (SIBO) decontamination and symptom relief (Judkins et al., 2020). Research demonstrated evidence of positive effects by using probiotics for gut microbiota restoration and modulation in gut dysbiosis (Hills Jr. et al., 2019; Marasco et al., 2020; Sharma & Singh, 2016; Singh et al., 2017).
Fermented food contains probiotics, prebiotics, and biogenics (functional metabolites). Fermented foods can produce vitamins, lacto‐try‐peptides, bacteriocins, and immunopotentiators during process of fermentation (Aslam et al., 2020). Fermented food shows a significant positive improvement in balancing intestinal permeability and barrier function (Bell et al., 2018). Main probiotic foods are yogurt, kefir, sauerkraut, tempeh, kimchi, miso, kombucha, pickles, traditional butter milk, soy sauce, natto, and cheese (Aslam et al., 2020; Marco et al., 2017)
Yogurt
Research has shown that yogurt can reduce counts of the enteropathogens E. coli and Helicobacter pylori (Liu et al., 2010; Yang & Sheu, 2012). Yogurt increases St. thermophilus, L. delbrueckii ssp. and Bulgaricus (Marco et al., 2017). Yogurt can reduce Caco‐2 transepithelial electrical resistance (TEER), modulate intestinal barrier function by improving occludin and ZO‐1 at TJ and preventing trinitrobenzene sulfonic acid‐induced intestinal inflammation, and reduce TLR4 (Putt et al., 2017). Yogurt can improve fasting blood glucose and antioxidant status for type 2 diabetes (T2DM) patients (Ejtahed et al., 2012).
Kimchi
Kimchi is one of most popular Korean side dishes made from fermenting vegetables, and various ingredients like red pepper powder, garlic, ginger, green onion, fermented seafood, and salt, which contain beneficial microbiota (Kim, Keogh, & Clifton, 2018; Shin et al., 2016). Kimchi has been reported by Health Magazine in 2006 as one of the five world's healthiest foods (Dharaneedharan & Heo, 2016; Patra et al., 2016). Research showed that the fermented kimchi is healthier than fresh kimchi (An et al., 2013). Fermented kimchi showed positive effects including improvement of metabolic function, lipid metabolism, insulin resistance/sensitivity, systolic and diastolic blood pressure, and enhancement of immune response (Han et al., 2015; Kim et al., 2011; Shin et al., 2016). Kimchi has anti‐inflammatory, antibacterial, antioxidant, anticancer, antiobesity, probiotic properties, cholesterol reduction, and antiaging properties (Patra et al., 2016). Kimchi increases Lu. mesenteroides, L. plantarum, and L. brevis (Marco et al., 2017).
3.2.2. Prebiotics
Prebiotics have been defined as “selective fermented ingredients that allow specific changes, both in the composition and/or activity in the gastrointestinal microflora that confer benefits upon host well‐being and health” (Gibson et al., 2004) and selectively promote beneficial bacteria, mainly Bifidobacterium, Lactobacillus, and Faecalibacterium prausnitzii (Marasco et al., 2020; Maukonen & Saarela, 2015; Scott et al., 2013). Those beneficial microbes have a function of anti‐inflammation, improvement of gut barrier and gut health. Prebiotics increase SCFAs and IL‐10 and decrease LPS and IL‐6 (Singh et al., 2017). The important dietary fibers, such as fructo‐oligosaccharides (FOS), galacto‐oligosaccharides (GOS), isomalto‐oligosaccharides (IMO), xylo‐oligosaccharides (XOS), arabino‐xylo‐oligosaccharides (AXOS), pectic‐oligosaccharides (POS) and resistant starch (RS) pass undigested through the GI tract until reaching the colon to be fermented by gut microbiota. Prebiotics include FOS, GOS, XOS, IMO, lactulose, and polysaccharides like inulin, RS, cellulose, etc. FOS, XOS, and AXOS improve the metabolic activity of intestinal microbiota, and decrease the synthesis of protein‐derived toxic compounds, such as pcresol, phenol, biogenic amines (Angelisa et al., 2019; Hills Jr. et al., 2019; Maukonen & Saarela, 2015; Yan et al., 2018; Yao et al., 2016). Inulin is a soluble fiber and fructans, which can normalize glucose tolerance or lipid profile (Kim, Keogh, & Clifton, 2018). Prebiotics enriched foods are artichokes, onions, garlic, leeks, soybeans, chicory roots, honey, banana, seeds, and selective fibers (Kim, Keogh, & Clifton, 2018; Maukonen & Saarela, 2015).
Synbiotics
Synbiotics are mixtures of probiotics and prebiotics. Synbiotics have shown effects for celiac disease, T2DM, nonalcoholic fatty liver disease (NAFLD), liver cirrhosis and steatosis, muscle wasting and many other diseases (Kim, Keogh, & Clifton, 2018; Marasco et al., 2020; van Krimpen et al., 2021; Woodhouse et al., 2018). Probiotics, prebiotics, and synbiotics have been shown to restore immunity, correct altered intestinal microbiota, balance the equilibrium between TREG and Th17 cells, and maintain human normal health state (Amati et al., 2010; Angelisa et al., 2019).
3.3. Fruits and vegetables
High dietary intake of fruits and vegetables can reduce risks of diseases by multiple factors (Amasheh et al., 2009). Fruits, vegetables, and other plant‐derived food like tea and juice contain phytochemicals including major compounds such as polyphenols including flavonoids like anthocyanins (ACNs) and quercetin, resveratrol, epigallocatechin gallate (ECGC), and curcumin, carotenoid, organosulfur compounds, phytoestrogens, etc. Phytochemicals can reduce intestinal permeability and improve intestinal function through reducing proinflammatory cytokines and increasing anti‐inflammatory cytokines, increasing beneficial bacteria, and reducing pathogenic bacteria. Phytochemical can also reduce NF‐κB and signal transducer, activate transcription (STAT), myosin light chain kinase (MLCK), and mitogen‐activated protein kinase expression, and increase antioxidant activity and improve disease activity index (DAI) score (Hossen et al., 2020). Dietary phytochemicals can regulate miRNA and play a tumor suppressor role (Budisan et al., 2017).
Polyphenols are well‐known antioxidants created by forming stable phenoxyl radicals and disrupting chain oxidation reactions in cellular components. The main sources of polyphenols are fruits, tea, and vegetables. Polyphenols may modulate the gut microbiota like stimulating Lactobacillus spp., Bifidobacterium spp., Akkermansia muciniphila, and Faecalibacterium prausnitzii and inhibiting Helicobacter pylori, Staphylococcus aureus, and Listeria monocytogenes (Manach et al., 2005; Septembre‐Malaterre et al., 2018; Kumar Singh et al., 2019). Polyphenols can prevent or limit oxidative damage to cell components, DNA, protein, and lipids accumulation that are related to aging‐associated diseases: CVDs, degenerative diseases, osteoporosis, and cancer (Parkar et al., 2008; Scalbert et al., 2005). Polyphenols have antioxidant and anti‐inflammatory properties because polyphenols can reduce oxidative stress, reactive oxygen species (ROS), endothelial nitric oxide synthase (eNOS), nitric oxide (NO), and decrease intercellular cell adhesion molecule‐1 (ICAM‐1), vascular endothelial adhesion molecule‐1 (VCAM‐1), monocyte chemoattractant protein‐1(MCP‐1), TNFα, IFN‐ γ, IL‐6, IL‐8, and NF‐κB (Khurana et al., 2013). ROS is responsible for developing DM, CADs, and other age‐related diseases such as Parkinson's and Alzheimer's disease (Selvaraju et al., 2012). The colors of fruits and vegetables are from ACNs. ACN‐rich fruits and vegetables like strawberries, blueberries, blackberries, cherries, plums, grapes, red apples, red beets possess antidiabetic, anticancer, anti‐inflammatory, antimicrobial, and antiobesity effects and preventing CVDs. Anthocyanin‐rich bilberry extract can decrease inducible nitric oxide synthase (iNOS), cyclooxygenase‐2 (COX‐2), and pro‐inflammatory cytokines like TNFα, IFN‐ γ, and increase anti‐inflammatory cytokines like IL‐10, IL‐22, and Th17 (Ghosh & Konishi, 2007; Lee et al., 2017; Lin et al., 2017; Manolescu et al., 2019; O'Donovan et al., 2019; Roth et al., 2016; Silva et al., 2016; Tsuda, 2016). Resveratrol is a stilbene compound and a phytoalexin found in plants such as red grapes, blueberries, red wine, ports, sherries, peanuts, itadori tea, hops, and pistachios (Burns et al., 2002). Resveratrol contributes a wide range of healing and preventive functions like cardiopreventive, neuropreventive, antitumor, and antioxidant roles (Gatuillat et al., 2010). Quercetin showed decreasing intestinal permeability through enhancing TJ function with claudin‐1, claudin‐4, ZO‐2, and occludin in human intestinal CaCo‐2 cells (Suzuki & Hara, 2009). A polyphenol‐rich beverage, green tea, has been reported to alleviate inflammatory and hypersensitive disorders like metabolic syndrome and IBD. The green tea's major components are EGCG and catechins which may neutralize proinflammatory cytokines, diminish oxidative stress to epithelial cells, and decrease intestinal permeability (Peron et al., 2020; Van Buiten et al., 2018; Watson et al., 2004).
3.4. Omega‐3 fatty acids
Diets rich in PUFAs, especially ω−3FAs have positive health benefits. Unsaturated fatty acids (UFAs) include MUFA with one double‐bond like omega‐9 fatty acids (ω−9FAs). PUFAs have more than one double‐bond such as ω−3FAs and ω−6FAs, and cis‐ and trans‐fats (Simopoulos, 2002). ω−3FAs are mainly available in eicosapentaenoic acid (EPA), docosahexanoic acid (DHA), and docosapentaenoic acid (DPA). ω−3FAs have positive effects or prevent DM, IBD, CADs like MI and acute coronary syndrome, central nervous system diseases like dementia and depression through gut–brain axis, autoimmune diseases like systemic lupus erythematosus, rheumatoid arthritis and psoriasis, carcinogenesis, and infections (Ballan et al., 2020; Merendino et al., 2013; Noriega et al., 2016; Watanabe & Tatsuno, 2017). A high ω−3FAs diet has the potential of increasing Bifidobacterium, Lactobacillus population, and SCFAs release (Robertson et al., 2017). Consumption of ω−3FAs has been reported to decrease serum triglycerides (TGs) and increase high‐density lipoprotein cholesterol (HDL‐C). ω−3FAs have anti‐inflammatory properties such as suppressing TNFα, IL‐1β, IL‐6, and producing anti‐inflammatory lipid mediators (E‐series resolvins, D‐series resolvins, protectins, and maresins). However, more research is needed to fully elucidate (Kaliannan et al., 2015; Miles & Calder, 2012; Watanabe & Tatsuno, 2017).
3.5. Some other dietary supplements
Vitamins and minerals play a very important role in the regulation of GI homeostasis, especially vitamin A and vitamin D. Research showed that Vitamin A inhibited LPS and enhanced TEER, so preventing inflammation. Vitamin A strengthened ZO‐1, Occludin, and claudin‐1, while vitamin D increased ZO‐1, claudin‐1, claudin‐2, and E‐cadherin, thereby enhancing TJ and intestinal permeability. Vitamin D may have long‐term implications for immune system modulation (Bischoff et al., 2014; Farré et al., 2020; Kong et al., 2008; Rinninella, Cintoni, et al., 2019). Minerals are involved in numerous bacterial physiological processes that impact the gut microbiota. Zinc is necessary to maintain expression of ZO‐1, occludin and intestinal barrier function. Zinc deprivation increases intestinal permeability by reducing TER and altering TJ and AJ with delocalization of ZO‐1, occludin, β‐catenin, E‐cadherin, and F‐actin. Its depletion may also cause infectious diseases. Zinc is an important component of DNA polymerase and enzymes involved in cell replication and differentiation (Amasheh et al., 2009; Finamore et al., 2008).
4. FOOD/FOOD COMPONENTS OR MEDICATION MAY ALTER GUT FUNCTION
4.1. Western diet
Western diet is rich in saturated fatty acids, high glycemic‐index sugars, especially fructose, refined carbohydrate, high salt, poor in fiber, and also includes processed food. The negative effects include high energy load, inadequate energy balance, and changing gut microbiota (Bischoff et al., 2014; Manzel et al., 2014; Riccio & Rossano, 2015). Saturated fats (SFAs) include very long‐chain fatty acids (VLCFAs) like arachidic acid (C20), long‐chain fatty acids (LCFAs): [like stearic acid (C18), palmitic acid (C16), myristic acid (C14), and lauric acid (C12)], and medium‐chain fatty acids (MCFAs) like caproic acid (C6) and SCFAs. Main dietary fats in high‐fat diet (HFD) are LCFAs (Rohr et al., 2020). HFD changes gut microbiota, promotes LPS, stimulates TNFα, IL‐1β, IL‐6, IFN‐ γ, and decrease IL‐10, IL17, and IL‐22 directly or indirectly. Additionally, HFD increases susceptibility to food allergy. Overall, diets rich in SFAs are associated with negative health impacts (Bischoff et al., 2014; Ke et al., 2020; Rohr et al., 2020; Shi et al., 2019). The major route of high sugar consumption is through the apical transient insertion of glucose transporter type 2 (GLUT2)‐mediated sugar absorption (Kellett et al., 2008). Fructose, glucose, and sucrose, especially fructose, increase intestinal permeability through altering TJ proteins like occludin, ZO‐1, claudin 1 and 4, and interacting with cytochrome P450‐2E1 (CYP2E1; Binienda et al., 2020). High fructose or glucose intake correlates with detrimental health outcomes, such as obesity, metabolic disorders, and hepatic steatosis (Do et al., 2018). Western diet decreases microbiota diversity, such as decreasing Bifidobacteria, Roseburia, Eubacterium rectale, Ruminococcus bromii, Lactobacillus, and Prevotella and increasing Ruminococcus torques, Enterobacteria, Bilophila, Alistipes, Bacteroides, and Akkermansia. Western diet increases LPS and proinflammatory cytokines, such as IL‐17, TNFα, and IFN‐ γ, and decreases Treg/Th17 ratio and SFCAs, and therefore causes chronic inflammation and autoimmune diseases (Riccio & Rossano, 2015; Rinninella, Cintoni, et al., 2019). Individuals consuming Western diet showed depleted metabolic fuels, dysbiosis, and leading to high levels of endotoxin in plasma (endotoxemia). It increases gram‐negative bacteria‐related LPS and induces inflammation via TLR2 and TLR4 (Bengmark, 2013). Western diet contains excessive ω−6FAs and deficient ω−3FAs. It increases ω−6FAs/ω−3FAs ratio from 1:1 to 4:1 (normal) to 10:1 to 50:1 and thus promotes the pathogenesis of many chronic diseases like CADs, cancer, inflammatory, and autoimmune diseases (Kang, 2011; Simopoulos, 2008). It can increase translocation of bacteria and bacteria products in the intestine, and then enhance endotoxin in the portal vein, leading to low‐grade liver inflammation (Bischoff et al., 2014).
Processed foods/industrial foods are detrimental to human health (Zinöcker & Lindseth, 2018). The thermal process of mixing amino acids and sugar generates advanced glycation endproducts (AGEs). AGEs can pass through GI tract to colon and alter gut microbial metabolism (Aljahdali & Carbonero, 2019; Hellwig & Henle, 2014). Almost all processed foods contain artificial sweeteners to improve the taste and texture, which can alter gut microbiota (Spencer et al., 2016; Suez et al., 2014). Chronic consumption of processed foods can compromise TJ protein, increase serum LPS, and impact intestinal permeability, causing multiple diseases, such as CADs, microvascular diseases like chronic kidney disease (CKD), obesity, DM, and cancer (Fiolet et al., 2018; Rico‐Campà et al., 2019; Snelson et al., 2021).
Dietary additive emulsifiers are widely used in bakery, confectionary, dairy, ice cream, sauces, butter, gum, beverages, chocolate, and convenient food industries. The major food additive emulsifiers include lecithin, mono‐and diglycerides of FA, guar gum, xanthan gum, carrageenan, celluloses, and polysorbates (Cox et al., 2021; Partridge et al., 2019). Dietary emulsifiers decreased gut microbiota diversity, decreasing anti‐inflammatory genera Akkermansia and Lupinus, promoting proinflammatory genera Escherichia, Roseburia, Bradyrhizobium, and Turicibacter, and developing dysbiosis (Jiang et al., 2018). Emulsifiers stimulate various inflammatory pathways, such as activating LPS‐induced inflammation through Bcell lymphoma/leukaemia‐10 (Bcl‐10) pathway, and triggering NF‐κB through TLR4 pathway, and increasing TNFα, IL‐1β, IL‐6, IL‐8 pathway (Bancil et al., 2021). Emulsifiers can drop hydrophobicity of the mucus layer, reduce mucus thickness, and alter ZO‐1 to allow bacteria translocation, which is associated with increasing intestinal permeability (Csáki, 2011).
4.2. Gluten
Gluten is a main storage protein in wheat and is also contained in non‐wheat cereals like barley, rye, and oats. Wheat contains nonprotein components like FODMAPs as well. Gluten proteins can be divided into alcohol‐soluble gliadins and insoluble glutenins (Khoshbin & Camilleri, 2020; Vader et al., 2003; Wieser, 2007). The adverse effects of gluten through human gut post‐translational modification of proteins (PTMP) and dysbiota mechanisms include the following: increasing systemic inflammation, oxidative stress, impacting systemic epigenetics, altering TJ and impacting microbiome, decreasing cellular viability, cell differentiation, and synthesis of DNA, RNA, and glycoprotein. Research also demonstrates that gluten increased apoptosis in cellular effects, increased immunogenicity and cytotoxicity, Th17 activity, neutrophil migration, natural killer group 2D (NKG2D) costimulatory molecule, TLR4 signaling pathway, changing innate and adaptive immune system functions, and Treg functions in immune effects (Fasano, 2020). Gluten impairs function of the TJ, AJ, DMs, and the colon mucosa, translocates bacteria, and elevates intestinal permeability (Menta et al., 2019). Dietary wheat germ agglutinin (WGA) induces the production of TNFα, IL‐1β, IL‐12, and IFN‐ γ, and can cross intestinal barrier or increase intestinal permeability (Pusztai et al., 1993; Sodhi & Kesherwani, 2007). Wheat proteins are also responsible for celiac disease (CD) and other gluten‐/wheat‐sensitive or intolerance conditions like IBD and IBS, and are associated with many other diseases due to chronic inflammation, such as CADs, metabolic syndrome, cancer, autoimmune diseases, neurological diseases like multiple sclerosis, dementia, schizophrenia, and depression. It is also responsible for baker's asthma, rhinitis, and contact urticaria though IgE‐mediated allergic reactions (Barnes & Adcock, 2009; Cardoso‐Silva et al., 2019; Fasano, 2020; Jonnalagadda et al., 2011; Tatham & Shewry, 2008). Gluten is clearly implicated damaging gut barrier in CD, and consequently, a gluten‐free diet (GFD) is the only available treatment for CD (Al‐Toma et al., 2019; Khoshbin & Camilleri, 2020). GFD increases Bifidobacteria, Victivallaceae, and Enterobacteria, and decreases Coriobacteriaceae, Veillonellaceae, Ruminococcus bromii, Roseburia, Lactobacillus, Clostridium lituseburense, and F. prausnitzii, and regulates the immunomodulatory role via cytokine induction of IL‐10, TNFα, and IFN‐γ (Rinninella, Cintoni, et al., 2019). However, even with a GFD, the abundance of gut bacterial population is different in normal individuals, CD patients, and nonceliac gluten‐/wheat‐sensitive patients (Caio et al., 2020).
4.3. Medications
Oral antibiotics can cause acute or long‐term effects due to eliminating antiproteolytic bacteria and increasing proteolytic activities in the colon (Yoon et al., 2018). In general, antibiotics change gut microbiota, implicate dysbiosis, impair gut barrier, and cause intestinal inflammation like Crohn's disease (Grigg & Sonnenberg, 2017; Hills Jr. et al., 2019; Yoon et al., 2018). The central mechanism of nonsteroidal anti‐inflammatory drugs (NSAIDs) suggests induced intestinal hyperpermeability and causes small bowel disease (Bjarnason & Takeuchi, 2009; Ganda Mall et al., 2020). NSAIDs damage enterocytes through biochemical processes. Then luminal substances like bile acids, bacterial degradation products, acid, and pepsin have access to mucosa, resulting in inflammation due to increased intestinal permeability. With long‐term use of NSAIDs, the inflammation may progress to GI ulcer, bleeding, or perforation (Bjarnason & Takeuchi, 2009). Proton pump inhibitors (PPIs), antipsychotic medications, opioids, and statins influence the gut microbiome and are detrimental to gut barrier and function (Imhann et al., 2016; Le Bastard et al., 2018).
4.4. Lectins, aquaporins, heated vegetable oils, and food antigens
A lot of common foods like tomatoes, potatoes, eggplants, bell peppers, legumes, etc. are known as healthy foods. However, research shows some contradictory evidence. Legumes and nightshades (tomatoes, white potatoes, eggplant, peppers, and seed species) contain lectin. Lectins can increase intestinal permeability and increase innate immune cell activation, which may be associated with rheumatoid arthritis (RA) symptoms (Cordain et al., 2000). Soybean meal‐based diets alter TJ protein mRNAs and decrease protein absorption, induce intestinal neutrophil turnover, and increase intestinal permeability in zebrafish (Solis et al., 2020). Dietary aquaporins (AQPs; corn, soybean, spinach leaf, and tomato AQP) may be mistaken for human aquaporin (AQP4) and the antibodies can cross‐react with brain astrocytic endfeet, breaking blood–brain barrier (BBB), leading to neuro‐autoimmunity and neurodegeneration, and cause neurological disorders, such as Alzheimer's disease, multiple sclerosis, and autism spectrum disorders. Individuals who lose their immune tolerance can remove the food AQPs and may improve the clinical conditions (Lambert et al., 2018). Soybean oil is the major oil in Western diet and contains more than 85% of ω−6FAs (Liu et al., 2014). The prolonged consumption of heated soy oil, and palm oil, or heated vegetable oils can cause hypertension and promote oxidative stress due to biochemical and vascular mechanisms such as impaired endothelium‐dependent and endothelium‐independent vasorelaxation as well as increased vasoconstriction responses (Leong et al., 2010; Siti et al., 2019). Vegetable oils have high ω−6/ω−3FAs radio which is a risk factor for health. Long‐term ingestion of heated vegetable oil including fried food can lead to obesity, T2DM, and CVD or hypertension and other lifestyle‐related diseases (Okuyama et al., 2016; Sayon‐Orea et al., 2015). Food antigens such as bovine milk proteins and collagen, egg allergen, and wheat gluten cause food sensitivities that lead to intestinal and systemic inflammation through IgA, IgE, IgG‐mediated transport (Li et al., 1999; Ma et al., 2021; Ménard et al., 2010; Severance et al., 2016).
5. POTENTIAL MECHANISMS OF DISEASES RELATED TO GUT FUNCTION AND DIETARY INFLUENCES
The goal of this review was to synthesize the current literature related to gut barrier dysfunction, associated diseases, and food influences. We summarized related diseases, possible mechanism, and dietary influences in the table below. The disease classifications are based on the International Classification of Disease, Eleventh Revision (ICD‐11). Dietary influences are the most important factor to gut microbiota. However, fecal microbiota transplantation has been reported to be effective to most of diseases due to modification of gut microbiota. It is not included below as this table is mainly concerned with dietary intervention (Table 1).
TABLE 1.
Diseases | Basic mechanism | Potential dietary components/Supplements that influence diseases | References |
---|---|---|---|
Dermatological disease | |||
General |
Dietary allergy & environmental factors ➔ gut microbiome sensitivity ➔ dysbiosis ➔ LGS ➔ disturb skin ecosystem ➔ cause multiple skin conditions: Acne (two major bacteria: Propionibacterium acnes & Staphylococcus epidermidis), aging skin (also exposure to UV), psoriasis, atopic dermatitis, eczema, seborrheic dermatitis, vitiligo, epidermolysis bullosa, rosacea (long‐term treating by antibiotics), blepharitis, malaria and attractiveness to mosquitoes, & skin cancer |
(+) Probiotics, Prebiotics (−) Process food (emulsifiers), Gluten, some skin care and cosmetic products (may alter skin microbiome) |
Ellebrecht et al. (2016), Kong et al. (2012), Maguire and Maguire (2017) |
Eczema |
Food allergens ➔ ↑ IgE & IgG4, IL‐10, CLA positive T‐cell & TNFα ➔ intestinal mucosal damage & increase intestine permeability ➔ skin reaction (erythematous rash, urticaria, & angioedema) & GI, respiratory, & cardiovascular symptoms of anaphylaxis; More children than adults |
(+) Dietary restriction with allergic food (−) Common allergic food: Cow's milk, egg, wheat (gluten), soy, peanut, fish, cashew (varies with individuals) |
Hauk (2010), Jaervinen et al. (2003), McAllister et al. (2019), Pike et al. (1986), Sicherer and Sampson (1999) |
Psoriasis |
Disturbs of the Firmicutes/Bacteroidetes Ratio ➔ microbiota dysbiosis, ➔↑ IL 1,2,6,8,12,17,23, TNFα, INF‐γ, ↓SCFAs, ↑TMAO; Patients presents with positive antigliadin antibodies; Associates with metabolic syndrome, DM, Crohn's disease, ulcerative colitis, cardiovascular disease & cancer |
(+) Probiotics, Fatty fish, Vitamin D (−) Western diet, Gluten, chronic consumption of alcohol |
Codoñer et al. (2018), Hidalgo‐Cantabrana et al. (2019), Lerner et al. (2017), Polak et al. (2021), Scher et al. (2015), Shi et al. (2020), Watanabe and Tatsuno (2017), Yu et al. (2019) |
Diseases of circulatory system (cardiovascular disease – CVD) | |||
General |
Obesity/Western diet ➔ gut dysbiosis, disrupt TJ; choline/carnitine ‐ ↑TMA ➔ ↑gut permeability ➔ ↑LPS, activation of TLR4, signal transduction including NF‐κB ➔ metabolic endotoxemia, TMAO ➔ inflammation, platelet aggregation, cholesterol‐laden macrophage foam cell formation ➔ CAD Ketogenic diets ➔ ↓total cholesterol, LDL‐cholesterol, triglycerides, HbA1C, & ↑HDL‐cholesterol in human research; however, there were some conflicts with rodent model research |
(+) Probiotics, Prebiotics, Dietary fish oil and oily fish (ω−3FAs), Ketogenic diets, Food rich in polyphenols such as resveratrol, EGCG, & curcumin, quercetin, Fruit polyphenol including berries (−) Western diet, Processed food, Gluten, Heated vegetable oil |
Amar (2018), Khurana et al. (2013), Kosinski and Jornayvaz (2017), Lourida et al. (2013), Merendino et al. (2013), Miles and Calder (2012), Mitchell et al. (2015), Moludi et al. (2020), Okuyama et al. (2016), Snelson et al. (2021), Tang et al. (2019), Villa‐Rodriguez et al. (2019), Watanabe and Tatsuno (2017) |
Atherosclerosis |
Obesity/HFD ➔ gut dysbiosis, disrupt TJ; choline/carnitine ‐ ↑TMA ➔ ↑gut permeability ➔ ↑LPS ➔ TMAO ‐ oxidative stress, ↑platelet reactivity & thrombus potential, macrophage foam cell activation, endothelial cell activation, plaque localization, & vascular inflammation, & ↓reverse cholesterol transport ➔ Atherosclerosis Animal‐based diet ➔ ↑bile‐tolerant microorganism such as Alistipes, Bilophila, Bacteroides & ↓Firmicutes |
(+) Probiotics, Prebiotics, Plant‐based diet (↑SCFAs), Food rich in polyphenols, EVO (‐) Western diet, Processed food |
Karlsson et al. (2012), Khurana et al. (2013), Miyake and Yamamoto (2013), Moludi et al. (2020), Tang et al. (2019), Woodhouse et al. (2018) |
Heart failure (HF) | Disrupt gut dysbiosis & disrupt TJ; choline/carnitine ‐ ↑TMA ➔ ↑gut permeability ➔ ↑LPS, ↑ TNFα, IL‐6 & CRP ➔ TMAO ‐ oxidative stress, ↑platelet reactivity & macrophage foam cell activation, endothelial cell activation, plaque localization, & vascular inflammation, & ↓reverse cholesterol transport oxidative stress, plaque localization, platelet activation ➔ HF, MI & stroke | (+) Probiotics, Prebiotics | Fukui (2016), Nagatomo and Tang (2015), Tang et al. (2019) |
Hypertension (HTN) |
HFD/high salt diets & other dietary factors ➔ disrupt gut microbiota ➔ ↑gut permeability ➔ ↑I‐FABP, LPS, TH17, IL22; ↑TMA – TMAO ➔ dysfunctional sympathetic‐gut communication ➔ HTN High‐salt diet ➔ change gut microbiota through an effect on TH17 lymphocytes pathway EVO decrease systolic BP |
(+) Mediterranean diet, High fiber food, Fruit, Oily fish, Paleolithic type diet, Low‐gluten diet, Food rich in polyphenols (fruits, tea, vegetables) (‐) Animal fat, Processed meat, High‐salt diet, High‐gluten diet, Heated vegetable oil |
Amar (2018), de Punder and Pruimboom (2013), Hul et al. (2018), Jaworska et al. (2017), Kaličanin et al. (2020), Khurana et al. (2013), Kim, Ruby Goel, et al. (2018), Leong et al. (2010), Li et al. (2017), Okuyama et al. (2016), Santisteban et al. (2017), Sayon‐Orea et al. (2015), Siti et al. (2019), Tang et al. (2019) |
Myocardial Infarction (MI) | Obesity/high‐fat diet ➔ gut dysbiosis, disrupt TJ; choline/carnitine ‐ ↑TMA ➔ ↑gut permeability ➔ ↑LPS ➔ TMAO ‐ oxidative stress, ↑platelet reactivity & thrombus potential, macrophage foam cell activation, endothelial cell activation, plaque localization, & vascular inflammation, & ↓reverse cholesterol transport oxidative stress, plaque localization, platelet activation ➔ Atherosclerosis ➔ MI, heart failure & stroke |
(+) Probiotics, Prebiotics, Plant‐based diet (↑SCFAs), Food rich in polyphenols, EVO (‐) Western diet, Processed food |
Carrera‐Bastos et al. (2018), Khurana et al. (2013), Miyake and Yamamoto (2013), Moludi et al. (2020), Tang et al. (2019), Woodhouse et al. (2018) |
Diseases of digestive system | |||
Acute Pancreatitis (AP) | Disruption of gut barrier & gut permeation in early stage of AP ➔ lower level of occludin and ZO‐1 expression &↑inflammatory cytokines ➔ intestinal dysbiosis: ↑Enterococcus & ↓ Bifidobacterium ➔ infection & pancreatic necrosis ➔ systemic inflammation & complications | (+) Probiotics | Capurso et al. (2012), DeMeo et al. (2002), Fukui (2016) |
Alcoholic liver disease (ALD) |
Alcohol ➔ gut dysbiosis/SIBO ‐ ↑Escherichia coli, Enterobacteria, Alcaligenes, Meagsphaera, & ↓Lactobacillus & Bifidobacterium ➔ ↑intestinal permeability ➔ LPS endotoxin and other PAMPs ➔ activate Kupffer cells ➔ ↑TNFα, IL‐6, IL‐1β, TLR4, MPO ➔ hepatocyte damage ➔ ↑AST & ALT ➔ ALD (through Gut‐liver axis) Probiotics ➔ ↑Lactobacillus, Bifidobacterium & ↓ Escherichia coli, Enterobacteria, Alcaligenes, & Actinobacteria; ↑Claudin‐1, Occludin, ZO‐1, Symplerin, p130, & HIF‐2α ➔ ↓intestinal permeability |
(+) Probiotics, Prebiotics, Zinc, Niacin, Appropriate nutritional intake, Dietary fatty acids | Fukui (2016), Hong et al. (2019), Sung et al. (2016), Zhou and Zhong (2017) |
Autoimmune hepatitis |
Genetics, food antigens, food‐borne pathogens & environmental cues disturb diversity of microbiota ➔ ↑plasma LPS, ↓duodenal TJ proteins ➔ imbalanced bidirectional gut‐liver axis ➔ dysbiosis ‐ ↑aerobic bacteria & ↓anaerobic bacteria ➔ autoimmunity ➔ autoimmune hepatitis Autoimmune hepatitis is associated with CD, MS, & RA |
(+) Probiotics, High‐fiber diet (‐) Western diet, Gluten, Long time alcohol consumption (↑gram negative bacteria), Excessive food intake |
Iqbal et al. (2017), Lerner et al. (2017), Lopetuso et al. (2015), McAllister et al. (2019), Plaza‐Díaz et al. (2020) |
Celiac Disease (CD) |
90% of CD patients genetically associate with HLA DQ2/DQ8 haplotypes; tTG2 involved; CD triggered by gliadin fraction of wheat gluten & prolamines of barley & rye; Gluten releases toxic peptides in stomach ➔ ↑plasma IgA, IgG, IgM ➔ against gluten peptides, & antibodies against TG2 & EMA ➔ TJ disassembly by upregulating zonulin pathway, ➔ gut dysbiosis ‐ ↓Bifidobacteria, Firmicutes, Lactobacilli & Streptococceae & ↑Bacteroides, Bacterioidetes, Bacteroides fragilis, Prevotella, E. Coli, Proteobacteria, Haemophilus, Serratia, Klebsiella ➔↑intestinal permeability ➔ gut & systemic inflammation 1% population with this gluten‐induced, cell‐mediated disorder; CD associates with multiple autoimmune diseases & psychological diseases such as atopic dermatitis, autoimmune hepatitis, anxiety, MS, depression, schizophrenia & panic disorder, dermatitis herpetiformis, HT, RA, Sjogren's syndrome, T1D |
(+) Lifelong Gluten Free Diet – the only available treatment for CD (Allowed grains: Corn, Millet, Rice Buckwheat, Sorghum, Wild rice, Teff), Probiotics (‐) Prohibited grains: Wheat, Rye, Barley, Kamut, Triticale (wheat‐rye hybrid), Spelt, Oats, FODMAP diet |
Cardoso‐Silva et al. (2019), de Punder and Pruimboom (2013), DeMeo et al. (2002), Drago et al. (2006), Fasano (2011), Ferrari et al. (2021), Caio et al. (2020), Leffler et al. (2015), McAllister et al. (2019), Neunlist et al. (2003), Rallabhandi (2012), Severance et al. (2016), Valitutti and Fasano (2019), Weaver and Herfarth (2021) |
Cirrhosis |
Gut microbiota disrupted ➔ altering bile acid homeostasis, ↓beneficial taxa like Lachnospiraceae, Bacteroidetes, Roseburia, Blautia, & Ruminococcaceae, ↑Proteobacteria, Fusobacterium spp., Veillonellaceae, Streptococcaceae & ↓ SCFA ➔ dysbiosis & bacterial overgrowth ➔ disrupt TJ & ↑gut permeability ➔ release both LPS & FGF 19 (produce by bile acid bind to FXR) to portal circulation ➔ ↑proinflammatory cytokines ➔ portal hypertension & cirrhosis (through gut–liver–immune system axis & Gut–liver axis); Gut dysbiosis ➔ ↑ammonia, & ↑glutamine in astrocytes ➔ astrocyte swelling ➔ oxidative stress ➔ systemic inflammation through Gut‐liver‐brain axis Fructose drinks ➔↓TJ & AJ proteins ➔ ↑plasma bacterial endotoxin levels ➔ endotoxemia. Dietary choline may remove fat from hepatocytes. High‐protein diet may ↑Prevotella & Oscillospira. Physical exercises can regulate intestinal permeability & improve microbiota diversity |
(+) Probiotics, Prebiotics, Synbiotics, High‐protein diet ‐ egg, meat, Cruciferous vegetable (due to rich in dietary choline) (‐) High fat & high‐sugar diet, Unlimited fat diet, Choline‐deficient diet, Alcohol ingestion, Excess fructose intake like fructose drinks |
Arab et al. (2018), Cho et al. (2021), Fukui (2016), Miyake and Yamamoto (2013), Norman et al. (2012), Plaza‐Díaz et al. (2020), Sharma and Singh (2016), Wang et al. (2019), Woodhouse et al. (2018) |
Fatty liver disease |
Poor diet habit, obesity/metabolic syndrome, type 2 DM, alcohol/drugs ➔ alteration of gut microbiota and bacteria overgrowth ➔ increase intestinal permeability ➔ ↑LPS, activation of TLR4 & TLR9 ➔ ↑Serum TNF‐α, IL‐1β, IL‐6, CCL2, CCL5, CXCL8, & ↑ROS ➔ Oxidative stress, fibrosis, inflammation ➔ FLD (through Gut‐liver axis); Oxidative stress ➔ neurodegenerative disease; LPS ➔ oxidative stress, plaque localization, platelet activation ➔ MI, CVD, peripheral artery disease |
(+) Mediterranean diet, especially EVO; Probiotics, Prebiotics, Synbiotics, Vitamin E (‐) Diet rich in fat, milk, dairy products, Excessive alcohol intake, Excessive fructose intake |
Ferro et al. (2020), Hossen et al. (2020), Li et al. (2013), Miyake and Yamamoto (2013), Ohlsson et al. (2017), Safari and Gérard (2019), Woodhouse et al. (2018) |
Inflammatory bowel disease (IBD) Types of IBD: Ulcerative Colitis (UC) Crohn's disease (Crohn's) |
Genetic (some with HLA‐B27 ‐ HLA‐DQ2/8 positive) & environmental factors (dietary patterns/components, malnutrition, medication & food allergy, for instance, excessive intake of FODMAP diet) ➔ SIBO ➔ ↑ intestinal permeability (↓Paneth cell antimicrobial function in Crohn's & ↓goblet cells & altered mucus function in UC) ➔ luminal changes in colon ➔ intestinal inflammation ➔ IBD Gut dysbiosis in IBD: IBD showed reduced common commensals – Bifidobacterial & Bacteroides fragilis & ↑Proteobacteria & Actinobacteria. Yersinia & Pseudomonas triggers Crohn's, & Samonella, Campylobacter jejuni, Clostridium difficile, Adenovirus, & Mycoplasma associated with Crohn's relapsing. Fusobacterium varium in colon may cause UC, & E. coli may maintain UC Dietary emulsifiers ➔ ↓β‐diversity & cause gut dysbiosis Phytochemicals suppress pro‐inflammatory cytokines like TNFα, IL‐1β, IL‐6 & IFN‐ γ. Ginger can maintain Caco‐2 and HT‐29/B6 cells and prevent pro‐inflammatory TNFα and TJ dysfunction T‐cells response to enteric bacteria ➔ ↑mucosal 5‐HT & dopamine, & ↓ tissue 5‐HT & NE ➔ related to CNS disorder |
(+) Low FODMAP diet, Mediterranean diet, Gluten‐free diet (especially for HLA‐DQ2/8 positive IBD patients), Green tea, Probiotics, Prebiotics, Vitamin D, folic acid, Zinc, Glutamine, Dietary fibers (to produce SCFAs), Psyllium (↓severity of UC), Ginger (especially for Crohn's), Red wine extract (‐) High FODMAP diet, Gluten (mainly HLA‐DQ2/8‐positive Crohn's patients), Processed food including dietary emulsifiers, Food rich in ω−6FA, NSAIDs (Crohn's), Antibiotics (Crohn's), Smoking (Crohn's) |
Arrieta et al. (2009), Bancil et al. (2021), Beguin et al. (2013), Cordain et al. (2000), DeMeo et al. (2002), Fukui (2016), Guagnozzi et al. (2012), Hossen et al. (2020), Kikuchi et al. (2019), Kim, Keogh, and Clifton (2018), Lacerda et al. (2021), Li et al. (2019), Llewellyn et al. (2018), Luettig et al. (2016), Magnuson et al. (2016), Malíčková et al. (2017), McAllister et al. (2019), Michielan and D'Incà (2015), Nanayakkara et al. (2016), Neish (2009), Nunes et al. (2019), Sasson et al. (2021), Scaldaferri et al. (2013), Shin and Lim (2020), Van Buiten et al. (2018), Vazquez‐Roque et al. (2013), Weaver and Herfarth (2021), Yeoh et al. (2013), Yoon et al. (2018) |
Irritable Bowel Syndrome (IBS) |
Genetic (some with HLA) & diet (Food triggers or aggravates 2/3 of IBS patients.) ➔ ↑serum Zonulin level ➔ ↑Serum TNF‐α, IL‐1β, IL‐6 ➔↑epithelial permeability ➔ low‐grade intestinal inflammation. IBS patients present visceral hypersensitivity due to neuronal dysfunction of ENS. |
(+) Low FODMAP diet, Probiotics (‐) High FODMAP diet, Gluten |
Barbaro et al. (2020), Camilleri and Gorman (2007), Guagnozzi et al. (2012), Martín et al. (2022), Nanayakkara et al. (2016), Niesler et al. (2021), Rinninella, Cintoni, et al. (2019), Wu et al. (2021) |
Liver steatosis |
Bidirectional communication between the gut & liver through Gut–liver axis. Primary bile acid from liver & Ig A ➔ Eubiosis. Dietary habits, age, ↓physical activities, & other causes (such as NAFLD, MAFLD, ALD, drugs, food contaminants of environment origin, etc.) ➔ gut dysbiosis ➔ ↑intestinal permeability ➔ secondary bile acids metabolites ➔ portal vein ➔ filtration of damaging agents like MAMPs or PAMPs ➔ ↑ cytokines (TNFα, IL‐1β, IL‐6, IL‐12, IL 18), chemokines (CXCL1, CXCL2, CCL2, CCL5, CCL3, CCL4), NO & ROS ➔ systemic inflammation ➔ multiple liver diseases including liver steatosis & cirrhosis. Physical exercises can modify insulin sensitivity & glucose metabolism, and ↑Bacteriodetes/Firmicutes ratio, ↑SCFAs, ↑T‐reg cells ➔ improve microbiota diversity, regulate intestinal permeability & anti‐inflammatory cytokines – positive effects on immune system. |
(+) Probiotics, Synbiotics, Mediterranean diet, High‐protein diet: egg, meat, Cruciferous vegetable (due to rich in dietary choline) (‐) Western‐style diet (high fat & high sugar diet), Low adherence to Mediterranean diet, High salt intake, Unlimited fat diet, Choline‐deficient diet, Alcohol ingestion, Excessive fructose intake, Excessive caloric intake |
Chen, Zhang, et al. (2020), Di Ciaula et al. (2020), Di Palo et al. (2020), Kim, Ruby Goel, et al. (2018), Plaza‐Díaz et al. (2020), Woodhouse et al. (2018) |
Necrotizing enterocolitis |
LPS induces the intestinal barrier dysfunction, impaired TJ protein, ➔ activates IL‐1β, IL‐6, IL‐8 & TNFα ➔ systemic inflammation |
(‐) Corn gluten meal | Bai, Gu, et al. (2019), Ling et al. (2016) |
Disease of genitourinary system | |||
Chronic Kidney disease (CKD) |
Chronic intake of Western diet including heat‐treated food (leaking albumin to urine), ↓intake dietary fibers (↓potassium), prolonged GI transit time due to lacking of physical activities, dietary restrictions & comorbidities (mainly DM & CVD), change intestinal microbiota diversity (↑proteolytic microbes) ➔ gut dysbiosis, SIBO ➔ ↑intestinal permeability ➔↑ blood ammonia & uremic toxicity ➔ renal injury, fibrosis & systemic inflammation through Gut‐kidney axis |
(+) Dietary fiber like fruits & vegetables (for potassium), Mediterranean diet, Probiotics, Prebiotics, Low‐protein diet (‐) Western diet, Processed food, Gluten |
Briskey et al. (2017), Fukui (2016), Hobby et al. (2019), Ondrussek‐Sekac et al. (2021), Sabatino et al. (2017), Snelson et al. (2021), Yang et al. (2019) |
Diseases of musculoskeletal system | |||
Ankylosing Spondylitis (AS) |
Immunity through Gut–Joint Axis – IL‐23/Th17 ➔ chronic inflammation; Oral microbiota dysfunction ➔ periodontitis; intestinal dysbiosis (↑Lachnospiraceae, Prevotellaceae, Rikenellaceae, Porphyromonadaceae, Bacteroidaceae & decreasing Ruminococcaceae families) ➔ immune response (↑total IgA) ➔ decreasing zonulin ➔ Leaky gut; 90‐95% of AS patients present major risk gene ‐ HLA‐B27 |
(+) Dietary fish oil & oily fish, Probiotics (with fermented food), Prebiotics; Low starch intake (‐) Smoking, Western diet, Gluten |
Chen et al. (2015), Ciccia et al. (2014, 2017), Costello et al. (2015), de Punder and Pruimboom (2013), Fasano (2012), Obrenovich (2018), Rath et al. (1999), Tito et al. (2017), Welsby and Goriely (2016), Yang et al. (2016), Yeoh et al. (2013), Yu et al. (2015) |
Muscle wasting |
Gut–brain–muscle crosstalk: Lactobacillus species pluralis (spp.) ➔ reduce gut permeability ➔ ameliorate muscle wasting |
(+) Probiotics (Fermented food like Kimchi), Prebiotics Synbiotics | van Krimpen et al. (2021) |
Rheumatoid Arthritis (RA) |
Oral microbiota dysfunction: dental caries & periodontitis ➔ develop ACPA; Gut dysbiosis: food antigens, food‐borne pathogens disturb immune homeostasis & cause T‐lymphocytes/antibody reaction ➔ reducing common commensals – Bifidobacterial & Bacteroides fragilis ➔ impaired TJ ➔ systemic inflammation ➔ RA Adley (Coix lachrymal‐jobi L., an annual crop) has anti‐inflammatory and immunological activities, such as ↓TNFα and IL8 and intestinal TJ permeability. Legumes (Lectin) and nightshades (lectin & alkaloids) ➔↑intestinal permeability & innate immune cell activation ➔ RA symptoms. |
(+) Dietary fish oil & oily fish, Probiotics, Ginger, Adley (Coix lachrymal‐jobi L.) (‐) Nightshades, Legumes, Western diet, Gluten |
Cordain et al. (2000), Farshchi et al. (2017), Lerner et al. (2017), Lopetuso et al. (2015), Simopoulos (2008), Vojdani (2015) |
Disease of respiratory system | |||
Asthma |
Environmental allergens activate immune system – TH2 ➔ Gut dysbiosis, ↓ratio of Bacteroidetes/Firmicutes, ↓Bifidobacterium, Lactobacillus, ↑Escherichia coli, Streptococcus & Staphylococcus ➔ ↑intestinal permeability ➔ ↑TNF & IL‐6, LPS, lower expression of TLR4 ➔ inflammation through Gut–lung axis ➔ asthma Dietary fiber regulates SCFA & ↓gastric acid to protect duodenal mucosa |
(+) Probiotics, Prebiotics, Synbiotics, Dietary fiber, Plant gums, Plantain family seeds (‐) Allergic cascade intake, Western diet |
Farshchi et al. (2017), Fasano (2012), Hijazi et al. (2004), Sahagu´n et al. (2015), Simopoulos (2008), Trivedi and Barve (2020), Trompette et al. (2014) |
Endocrine or metabolic diseases | |||
Diabetes Mellitus (DM) Type I DM (T1D) Type II DM (T2D) |
T1D: Factors influence gut microbiota composition: delivery mode ‐ cesarean section (CS) ↑risk of childhood onset T1D than virginal delivery due to ↓ gut microbiota diversity (mainly ↓Bacteroidetes phylum) from mother; Starting cereal diet early (cDNA clones of diverse wheat storage globulins ➔ islet damage); Geographic difference: highest in Sardinia & Finland, & lowest in China & Venezuela due to genetic (HLA class II genes) & environmental effects; Immunological status; Using antibiotics – induce the proliferation of pathogen like Clostridium difficile & S. enterica; Viral infection Dietary related antigens ➔ ↑zonulin ➔ microbiota dysbiosis ➔ ↓TJ ➔↑APC ➔ ↑IgA, LPS, T cell activation ➔ PLN immune response ➔ β cell autoimmunity ➔ insulin deficiency T2D: T2D associated with visceral obesity; Lower microbial diversity ➔ dysbiosis ➔↑LPS, ↑zonulin, ↑intestinal permeability ➔ ↓saccharolytic microbes ➔↓SCFA (SCFA suppress proinflammatory mediators like TNFα, IL‐1β, IL‐6, & NO) ➔ endotoxemia, insulin resistance & chronical inflammation |
T1D: (+) Hydrolyzed formula, Gluten‐free diet or delayed introduce gluten, ω−3FAs, Probiotics (‐) Early diet: Cereal, Cow milk T2D: (+) Dietary fish oil and oily fish (ω−3 FAs), Probiotics, Prebiotics, Synbiotics, Mediterranean diet, Fermented food like Kimchi, yogurt, Food rich in polyphenols like cinnamon & grapes (‐) Western diet, Processed food, Gluten, Heated vegetable oil |
T1D: Bekkering et al. (2013), Cardwell et al. (2008), de Kort et al. (2011), Fasano (2020), Gianchecchi and Fierabracci (2017), Li and Atkinson (2015), Petersa and Wekerle (2019), Sapone et al. (2006), Simpson et al. (2009) T2D: An et al. (2013), Bekkering et al. (2013), de Kort et al. (2011), Ejtahed et al. (2012), Fasano (2020), Jayashree et al. (2014), Lopetuso et al. (2015), Lourida et al. (2013), Merendino et al. (2013), Miles and Calder (2012), Moreno‐Navarrete et al. (2012), Okuyama et al. (2016), Patra et al. (2016), Sabatino et al. (2017), Snelson et al. (2021), Watanabe and Tatsuno (2017), Zhang et al. (2014) |
Hashimoto Thyroiditis (HT) |
Thyroid–Gut Axis: HT, Graves' disease & thyroid carcinoma share similar mechanism, all cause dysbiosis ➔ stimulate GALT & TLR ➔ change TSH & T3 level; HPT Axis ➔ ATTA. ↓selenium & zinc ➔ decrease T4 ➔ T3; ↓iron & iodine ➔ impaired thyroid hormone synthesis. Gluten‐free diet decrease thyroid antibody titers. HT related to Hepatitis C, Helicobacter pylori, Yersinia enterolitica & Borrelia burgdorferi infection HT often presents CD. |
(+) Selenium, Zinc, Vitamin D, Iron, Iodine, Copper, Probiotics, Mediterranean diet, Oily fish, Gluten‐free diet (‐) Gluten, Processed meat |
Bodinham et al. (2014), Cayres et al. (2021), Fasano (2020), Kaličanin et al. (2020), Fenneman et al. (2020), Knezevic et al. (1769), Krysiak et al. (2019)) |
Metabolic endotoxemia |
HFD ➔ gut dysbiosis, SIBO, ↓IAP activity, ↑CM & bile ➔ ↑intestinal permeability ➔ LPS & TLR4 ➔ ↑TNFα, IL‐1β, IL‐6 ➔ chronic low‐grade inflammation ➔ metabolic endotoxaemia ➔ many chronic diseases such as obesity, DM, atherosclerosis |
(+) Probiotics, Prebiotics, Flavonoids, ω−3FAs (‐) HFD, Western diet |
Kaliannan et al. (2015), Moreira et al. (2012)) |
Obesity |
HFD, excessive gluten diet ➔ change gut microbiota, ↑Lactobacillus, Staphylococcus aureus, Enterobacter cloacae strain B29, ↓Faecalibacterium prausnitzii ➔ ↑LPS ➔ disrupt TJ & ↑gut permeability ➔ ↑TLR4 & MLCK activation, ↑release TNFα, IL‐1β, IL‐6, IL‐8 ➔ endotoxaemia ➔ systemic inflammation ➔ obesity. Obesity associated with metabolic disorders including T2DM, insulin resistance, liver diseases and diseases in multiple systems. |
(+) Food rich in polyphenols Probiotics, Prebiotics, Flavonoids (‐) HFD, Gluten, Heated vegetable oil |
Bekkering et al. (2013), Casselbran et al. (2015), Di Palo et al. (2020), Frazier et al. (2011), Gerard (2016), Gil‐Cardoso et al. (2016), Hul et al. (2018), Kim and Ko (2018), Guercio Nuzio et al. (2017), Okuyama et al. (2016), Shi et al. (2021), Silva et al. (2020), Stenman et al. (2016), Zak‐Gołąb et al. (2013)) |
Immune disease | |||
Systemic Lupus Erythematosus (SLE) |
Dysbiosis in gut microbiome (↓Firmicutes/Bacteroidetes ratio) ➔ Leaky Gut ➔ ↑ LPS (↑soluble CD 14) & proinflammatory cytokines (IL17, IL 21‐23 & IFNα) ➔ systematic inflammation, cell apoptosis, anti‐dsDNA Ig production; Fc gamma receptor IIb dysfunction ➔ ↓immune response; Related to RA, T1D, MS, & IBD |
(+) Dietary fish oil and oily fish (ω−3FAs), Probiotics, Prebiotics (‐) Western diet, Nightshades |
Abdelhamid and Luo (2018), Lopetuso et al. (2015), Mu et al. (2017), Thim‐uam et al. (2020), van der Meulen et al. (2016), Watanabe and Tatsuno (2017) |
Neoplasms | |||
General |
Disruption of microbiota ➔ gut dysbiosis ➔ MAMPS ➔ ↑ LPS, activate TLRs, & release proinflammatory cytokines ➔ carcinogenesis Bacteria associated with carcinogenesis such as Helicobacter pylori, Fusobacterium nucleatum, Streptococcus gallolyticus, Bacteroides fragilis, Salmonella typhi, Chlamydia pneumonia, Mycoplasma sp., Prevotella sp., Bacillus sp. ➔ genotoxicity, release of ROS, RNS ➔ carcinogenesis Very low‐carbohydrate ketogenic diets ➔ ↓serum glucose & insulin growth factor ➔ delay cancer progression Intestinal mucosa release SCFAs to protect carcinogens. Glutenous diets ➔ ↑risk of lymphoma (usually non‐Hodgkin's lymphoma) due to associate with CD especially silent & latent CD |
(+) Probiotics, Prebiotics, Mediterranean diet, Vegetarian diet, Japanese diet, very low‐carbohydrate Ketogenic diets, Dietary fish oil and oily fish (ω−3FAs), Dietary phytochemicals, Resveratrol, Vitamins, Essential minerals (‐) Western diet, Processed food, Gluten |
de Punder and Pruimboom (2013), Ferrari et al. (2021), Hoggan (1997), Klein et al. (2012), Laura Soldati et al. (2018), Merendino et al. (2013), Miles and Calder (2012), Raza et al. (2019), Shabbir et al. (2021), Snelson et al. (2021), Watanabe and Tatsuno (2017) |
Gliomas |
↑zonulin expression ➔ altered gut TJ ➔ altered BBB ➔ glioma C6 conditioned media ‐ ↑HGF, VEGF, zonulin, PGE2, & ↓EGF ➔ IL‐1β, IL‐6, IL‐8, TNFα, CXL13 ➔ gliomas |
(+) Ketogenic diet (‐) High‐calorie diet |
Díaz‐Coránguez et al. (2013), Jonathan et al. (2020), Skardelly et al. (2009)) |
Neurological and psychological diseases | |||
General |
Leaky gut ➔ leaky brain: through MGB axis, GBNM axis, HPA axis & multiple pathways like neuroendocrine immune, ANS, & ENS. The BBB includes endothelial cells, TJ, pericytes, basement membrane & astrocyte end‐feet ensheathing the capillary wall. Brain presents similar junctional proteins as gut TJ proteins. Gastro‐intestinal‐derived hormonal secretion, commensal bacteria produce neurotransmitters & neuromodulators: Lactobacillus, Bifidobacterium (produces GABA), Escherichia, Bacillus & Saccharomyces (produces NE), Bacillus (produces dopamine), Lactobacillus (produces acetylcholine), Candida, Streptococcus, Escheridia, Enterococcus spp. (produces serotonin) & Lactobacillus plantarum (may stimulate BDNF). Gut bacterial composition manipulates behavior. Dysbiosis ➔ altering TJ at BBB ➔ dysregulation of MGB axis ➔ dysfunction of GABA, serotonin & BDNF ➔ releasing pro‐inflammatory cytokine: IL‐1, IL‐6, IL‐18 (bacterial cell wall LPS induced), TNF, IFN; inhibiting IL‐10, IL‐12, IL‐15 & T‐cells; & chemically related to 5HT, 5HIAA, & HVA ➔ effect on brain & behavior. Chronic stresses in adult effects gut microbiota ➔ IL‐1 & IL‐6 & ↑cortisol release through HPA axis. Stress ↓gluten tolerance ➔ “hyper‐excitable celiac brain”. Dietary aquaporin (corn, soybean, spinach leaf, tomato aquaporins) may cross‐react with brain astrocytic endfeet ➔ broken BBB ➔ neuroautoimmunity ➔ neurological disorders. Dietary fiber produces SCFAs. SCFAs – the key signaling metabolite (Eubiosis) – regulate & maintain the BBB. |
(+) Probiotics, Prebiotics, Ketogenic diet, Yogurt, Dietary fiber (produces SCFA) (‐) Gluten (for some diseases); Antibiotic use; Western diet; Dietary aquaporins ‐ corn, soybean, spinach leaf, tomato aquaporins (related to demyelinating diseases, Gluten ataxia, Guillain Barre syndrome, Miller Fisher syndrome, MS, motor neuron disease, myasthenia gravis, etc.) |
Braniste et al. (2014), Cryan et al. (2020), Dinan and Cryan (2017), Galland (2014), Kuwahara et al. (2020), Lambert et al. (2018), Mittal et al. (2017), Obrenovich (2018), Osadchiy et al. (2019), Rahman et al. (2018), Rea et al. (2020), Rieder et al. (2017), Rutsch et al. (2020), Seguella et al. (2019), Slyepchenko et al. (2017) |
Aging |
Human keeps stable & diversity of gut microbiota till 40 years. Changing diet, using antibiotics & medications, ↓exercises, & ↑stress ➔ ↓ microbiota diversity ➔ low grade, chronic inflammation, ↑IL‐6 & TNF with changing Bacteroidaceae & Erysipelotrichaeae; thinner mucus & ↓mucin with changing Clostidiaceae, Akkermansiaceae, Bifidobacteriaceae, & Bacteroidaceae; ↓immune tolerance/immune senescence with changing Clostidiaceae, Bifidobacteriaceae, Lachnospiraceae, & Coriobacteriaceae; & ↓ SCFA ➔ aging & frailty. Bland food ➔ ↓microbiota diversity. If ↑ microbiota diversity ➔ less aging. |
(+) Probiotics, Prebiotics, Mediterranean diet, High‐fiber diet, Food rich in polyphenols such as resveratrol, EGCG, curcumin & quercetin (‐) Bland & low‐fiber food (often in nursing homes), Antibiotics & medications, High saturation fat diet, High‐sugar diet |
Cryan et al. (2020), DeJong et al. (2020), Khurana et al. (2013) |
Alzheimer's disease (AD) |
AD is the most common form of dementia. Aging ➔ ↓microbial diversity & naïve T cells ➔ ↑risk of AD. Obesity, HTN & T2DM also disrupt gut & BBB permeability ➔ ↑risk of AD. APOE gene – risk of AD APOE gene, aging & unbalanced diet ➔ ↑Verrucomicrobia, Escerchia/Shigella, Proteobacteria, & Pseudomonas aeruginosa, & ↓Eubacterium hallii, Bacillus fragilis, Bacteroides fragilis, Eubacterium rectale, & Faecalibacterium prausnitzii ➔ disturb intestinal permeability & BBB permeability through MGB axis, communicating with vagal & spinal nerve & HPA axis ➔ dysbiosis of microbiota (gut, mouth, nose) ➔ ↑ secretion of β‐amyloid (Aβ) & LPS ➔ ↑IL‐1β, IL‐6, IL‐18, TNFα & IFN‐γ in neurons, microglia, & astrocytes; imbalance of GABA, serotonin, histamine, & dopamine, &↓SCFA ➔ Aβ plaques & neurofibrillary tangles ➔ neuroinflammation, cognitive deficits & neurodegeneration ➔ AD Western diet ➔ ↑Flavobacterium, Runella, & Flectobacillus ➔ ↑ IL‐1β & NF‐κB ➔ disrupt GB axis ➔ inflammation AD patients showed ↓melatonin level. Using melatonin may prevent AD or slow down AD progression |
(+) Mediterranean diet, Ketogenic diet, Probiotics, Prebiotics, Vitamins (B compound), ω−3FAs, High fiber diet, Polyphenol ‐ high consumption of vegetables & fruits, Antioxidants, Melatonin (‐) Western diet, Antibiotics, Dietary aquaporin |
De‐Paula et al. (2018), Fan et al. (2019), Fox et al. (2019), Kesika et al. (2021), Khurana et al. (2013), La Rosa et al. (2018), Lambert et al. (2018), Liu et al. (2020), Lourida et al. (2013), Megur et al. (2020), Moreno‐Navarrete et al. (2012), Obrenovich (2018), Rutsch et al. (2020), Saji et al. (2020), Shabbir et al. (2021), Sochocka et al. (2019), Vogt et al. (2017) |
Amyotrophic Lateral Sclerosis (ALS) |
Through MGB axis: intrinsic or extrinsic factors ➔ change gut microbiota diversity ➔ disturbance of TJ proteins & intestinal barrier ➔ ↑LPS, ↑IL‐1β, IL‐6, TNFα, IFN‐γ ➔ Disruption of BBB & BSCB (endothelial cells, TJ, pericytes, basal lamina & astrocytic foot processes) ➔ neuropathy of ENS (multi‐functional RNA‐binding protein TDP43 linking between ENS & CNS) ➔ ↓GABA, glial cell loss, proteostasis‐associated defects, glutamate excitotoxicity; dysfunction of mitochondrial, oxidative stress, SOD‐1, TCP‐43, C9orf72, jettisoned, organelles; cytoskeletal, axonal, & transport vesicle defects ➔ ALS Use Probiotics ‐ “BUGS as DRUGs” (Obrenovich et al., 2020) for neurodegenerative diseases including ALS |
(+) Probiotics, Prebiotics, Symbiotics, Polyphenols, Flavonoids, Caffeine | Blacher et al. (2019), Fang (2018), Niesler et al. (2021), Obrenovich (2018), Obrenovich et al. (2020) |
Anorexia nervosa (AN) |
Poor diet or insufficient food intake like protein‐deficient diet, plus stress ➔ alter gut‐brain interaction through MGB axis, HPA axis, & vagus nerve ➔ gut dysbiosis ➔ ↓SCFA, serotonin, & BDNF at hippocampus area & ↑cortisol & cytokines ➔ neuroinflammation, cognitive dysfunction & mood changes. AN associate with Crohn's disease & IBS. |
(+) Probiotics, Prebiotics, Vitamins & minerals, Diet with high fiber & low saturated fat (‐) Western diet, Processed food |
Herpertz‐Dahlmann et al. (2017) |
Anxiety disorder |
Gut dysbiosis plus chronic stress through MGB axis ➔ ↓SCFA‐producing bacteria & ↑Escherichia‐Shigella, Fusobacterium, & Ruminococcus gnavus ➔ Brain chemical imbalance (↓serotonin, GABA & dopamine & ↑cortisol & NE), glia abnormalities ➔ central & peripheral persistent low‐grade immune inflammation through HPA axis & ANS ➔Anxiety. Anxiety related to IBD, CD. Fermented food ↑ SCFAs, GABA, & ↓LPS, IL‐1β, IL‐6, TNFα, & NF‐κB and improve TJ and gut barrier function. |
(+) Vitamin B2, 3, 6, & 12; Probiotics, Prebiotics, Fermented food (‐) Gluten, HFD |
Aslam et al. (2020), Dinan and Cryan (2017), Jianga et al. (2018), McAllister et al. (2019), Plaza‐Díaz et al. (2020), Rudzki and Maes (2020) |
Attention deficit hyperactivity disorder (ADHD) |
Genetic, epigenetic, & dietary factors ➔ imbalanced gut microbiomes, ↑Bacteroides uniformis, Bacteroides ovatus, & Sutterella stercoricanis ➔ ↑zonulin ➔ ↑permeability in GI system & BBB through MGB axis ➔ glia abnormalities ➔ ADHD (inattention, hyperactivity, impulsivity & impairment of social function) Comorbidity with ASD |
(+) Probiotics, Prebiotics, Synbiotics, Vitamins (‐) High fat intake, High sugar intake, High refined grains intake, Antibiotics |
Chou et al. (2018), Wang et al. (2020), Özyurt et al. (2018), Partty et al. (2015), Proctor et al. (2017), Rudzki and Maes (2020) |
Autism spectrum disorder (ASD) |
Prenatal stress (maternal infection, inflammation & antibiotics, & maternal high fat diet), diet ➔ ↓gut microbiota diversity, alter microbiota, ↑Firmicutes/Bacteroidetes ratio, ↑Acidobacteria, Enterobacteriaceae, Pseudomonadaceae, Veillonellaceae, & Megamonas; ↑Lactobacillus spp. & Desulfovibrio spp., & ↑GI Candida albicans ➔ ↑zonulin & LPS ➔ ↑permeability in GI system & disrupt BBB through MGB axis, HPA axis, ANS (vagus nerve) ➔ ↑IL‐6, IL‐1β, IL‐8, TNFα & IFN‐γ; alteration of serotonin, GABA, dopamine, glutamate, SCFAs & Treg cells; accumulation of AGEs in the brain through AGE‐RAGE axis ➔ ASD. Ketogenic diet regulates mitochondrial function & improve mood balance of the brain, but may have adverse effects, such as constipation, hypercholesterolemia, etc. Gluten‐ and casein‐free diet – prevent immune response to gluten (antibodies against gluten in ASD is different with celiac disease) & casein, & ↓urinary peptide and improve behavior. ASD related to IBD (UC & Crohn's Disease). |
(+) Ketogenic diet, Probiotics, Prebiotics, Mother's milk (↑SCFA), Gluten & casein‐free diet (‐) Gluten, Casein (cow & goat's milk, ice cream, butter & cheese), Gluten, Dietary aquaporin |
Esnafoglu et al. (2017), Fattorusso et al. (2019), Fowlie et al. (2018), Galland (2014), Hughes et al. (2018), Józefczuk et al. (2018), Karakuła‐Juchnowicz et al. (2017), Lambert et al. (2018), Mayer et al. (2014), Obrenovich (2018), Özyurt et al. (2018), Tagliabue et al. (2017) |
Bipolar disorder (BD) |
Through Gut–brain axis: Stress & other factors ➔ ↑IgM/IgA response to LPS & other Gram‐negative bacteria ➔ gut dysbiosis ➔ intestinal permeability ➔ ↑proinflammatory cytokines: IL‐2, IL‐6, IL‐1β, TNFα & ↓anti‐inflammatory cytokines: IL‐4 ➔ disrupt TJ protein of BBB ➔ ↑BBB permeability ➔ imbalance of neurotransmitters: 5‐HT, tryptophan, kynurenine, picolinic acid, quinolinic acid ➔ immune system dysfunction ➔ BD. BD is associated with metabolic & cardiovascular diseases |
(+) Probiotics (‐) Smoking |
Chen, Park, et al. (2020), Doney et al. (2021), Painold et al. (2019), Simeonova et al. (2020) |
Dementia |
Change in microbiota ➔↑Enterotype I & III bacteria & ↑serum triglyceride & serum C‐reactive protein, ↓Clostridia & its phylum Firmicutes & ↓SCFAs & indole‐3‐pyruvic acid ➔ gut dysbiosis & disrupt BBB through MGB axis cause glia abnormalities ➔ ↓memory, & other cognitive deficits |
(+) Mediterranean diet, Low fat diet with calorie restriction, ω−3FAs (‐) Western diet with high fat & sugar, Gluten, Alcohol |
Wang et al. (2020), Lourida et al. (2013), Merendino et al. (2013), Obrenovich (2018), Rudzki and Maes (2020), Shabbir et al. (2021), Watanabe and Tatsuno (2017) |
Epilepsy |
Through MGB axis or GMB axis: Genetic & environmental factors ➔ disrupt gut microbiota diversity ➔ disrupt TJ protein ZO‐1, claudin‐5, & occludin ➔ TJ disassembly ➔ ↑ intestinal permeability ➔ ↑IL‐17A, IL6, IL1β, IFN‐γ ➔ breakdown BBB ➔ Epilepsy Approximately 1/3 patients with epilepsy have drug resistance. Ketogenic diet is used for drug resistant epileptic patients. After Ketogenic diet therapy, epilepsy patients showed ↑ in gut Bacteroidetes, Actinobacteria phylum & ↓Proteobacteria Genetic disorder ‐ GLUT1 DS often associates with epilepsy |
(+) Ketogenic diet, Probiotics, Prebiotics, Vitamins, Minerals | Arulsamy et al. (2020), Dahlin and Prast‐Nielsen (2019), Fan et al. (2019), Rahman et al. (2018), Rinninella, Cintoni, et al. (2019) |
Major depressive disorder (MDD) |
Food antigens plus physiological & psychological stress ➔ altered microbiota (such as ↑Actinomycineae, Coriobacterineae, Lactobacillaceae, Streptococcaceae, Clostridiales, Eubacteriaceae, Lachnospiraceae, Ruminococcaceae, Erysipelotrichaceae; ↓Bacteroidaceae, Rikenellaceae, Lachnospiraceae, Acidaminococcaceae, Veillonellaceae, Sutterellaceae) through MGB axis, more specifically MGIG axis. Dysbiosis, SIBO, parasitic & fungal infection ➔ ↑BBB permeability & autoimmunity ➔ ↑TNFα, IL‐1β, IL‐6, IFN‐γ, NF‐κB, O&NS, & hsCRP, ↓IL‐4, also ↑ serum IgA & IgM to LPS of G‐ enterobacteria, ↑I‐FABP ➔ brain chemical imbalance: ↓serotonin, NE, dopamine, GABA, BDNF & ↑cortisol ➔ glia abnormalities ➔ central & peripheral persistent low‐grade immune inflammation through HPA axis & ANS ➔ MDD; Gluten sensitivity ➔ ↑ serum IgG ➔ effect to MDD; HFD ➔ ↑the ratio of Firmicutes to Bacteroidetes & Proteobacteria (G‐ LPS contained bacteria); Fermented foods can increase lactic acid bacteria such as Lactobacillus, Streptococcus, Enterococcus, Lactococcus, Bifidobacterium, & Leuconostoc and ↑SCFAs, GABA, & ↓LPS, IL‐1β, IL‐6, TNFα, & NF‐κB, improve gut barrier function and anti‐inflammation. MDD is associated with IBS, obesity, T2DM & chronic fatigue syndrome, & they share similar mechanisms. |
(+) Mediterranean diet, Low fat diet with calorie restriction, Cabbage (AHR ligands – anti‐inflammatory properties), Probiotics, Prebiotics, Fermented foods, Vitamin B2, 3, 6, & 12 (‐) Western diet with high fat & sugar, Gluten, Alcohol, Pesticides, Drugs (NSAIDs, antibiotics) |
Alvarez‐Mon et al. (2019), Aslam et al. (2020), de Punder and Pruimboom (2013), Doney et al. (2021), Fukui (2016), Fung et al. (2017), Karakuła‐Juchnowicz et al. (2017), Liśkiewicz et al. (2021), McAllister et al. (2019), Obrenovich (2018), Plaza‐Díaz et al. (2020), Rudzki and Maes (2020), Slyepchenko et al. (2017) |
Migraine headache (MH) |
Disrupt gut microbiota ➔ ↑intestinal permeability, ↑LPS, IL‐1β, IL‐6, TNFα, & ↓SCFA, ↓5‐HT & serotonin ➔ BBB dysfunction ➔ inflammation through gut–brain axis ➔ may act on trigeminovascular system ➔ MH Food allergy ➔ ↑ serum IgG ➔ MH MH is associated with CD, IBD, anxiety & depression. |
(+) Probiotics, prebiotics, Ketogenic diet, Ginger (‐) Food allergy |
Dai et al. (2017), Fan et al. (2019), Karakuła‐Juchnowicz et al. (2017), Luettig et al. (2016), Nouri et al. (2014), Parohan et al. (2020), Roos et al. (2017) |
Multiple Sclerosis (MS) |
Through gut–brain axis & HPA axis: one of the factors is dietary factor (mainly Western diet) ➔ ↑lactulose/mannitol ratio, disturb commensal gut microbiota (such as ↑Methanobrevibacter, Akkermansia; ↓Butyricimonas) ➔↑serum zonulin ➔ Gut dysbiosis ➔ ↓Treg/Th17 ratio at intestinal mucosa, ↑LPS ‐ damage intestinal barrier ➔ NF‐κB & AP‐1 ➔ exdotoxemia & ↑TNFα, IFN‐γ, IL1β, 2, 6, 18, hs‐CRP, RANKL, ROS ➔ gut inflammation (similar to IBD) & chronic systemic inflammation ➔ breakdown BBB ➔ damage nerve fibers and myelin sheath at brain, spinal cord & optical nerve. Hypercaloric diets ➔ postprandial inflammation; Antigens ➔ ↓ SCFAs; Food contains lectin & alkaloids may influence nerve, brain, muscle & GI function negatively. Two special diets for MS: 1. Swank Diet: A low‐fat diet eliminated all red meat and dairy; 2. Wahls Elimination Diet: modified Paleo diet, exclude all grains, dairy, legumes & nightshades and emphasized fruits and vegetables, meat and fish. |
(+) Two special diets for MS: 1. Swank Diet, 2. Wahls Elimination Diet; Mediterranean Diet (Gluten limits to whole grain); Ketogenic diet, Probiotics; Vegetables, Seafood & fish oil; Seaweed & Algae; Fermented food; Multivitamins/minerals (‐) Western diet; Nightshades; Gluten; Cow's milk (bovine milk casein); Sweeteners; Dietary aquaporins; Smoking |
Berer et al. (2017), Buscarinu et al. (2018, 2019), Camara‐Lemarroy et al. (2018, 2020), Cantarel et al. (2015), Chen et al. (2018), Chu et al. (2018), de Punder and Pruimboom (2013), Esposito et al. (2018), Jangi et al. (2016), Lambert et al. (2018), Lerner et al. (2017), Lopetuso et al. (2015), Norman et al. (2012), Nouri et al. (2014), Ochoa‐Repáraz et al. (2018), Pellizoni et al. (2021), Rahman et al. (2018), Raza et al. (2019), Reynders et al. (2020), Riccio and Rossano (2015), Rutsch et al. (2020), Shahi et al. (2017), Wahls et al. (2019) |
Parkinson's Disease (PD) |
Unbalanced nutrition, aging, ↑ toxins, infection, & use of antibiotics ➔ microbiota dysbiosis (such as ↑Blautia, Coprococcus, Roseburia, Proteobacteria; ↓Faecalibacterium, Prevotellaceae) ↓SCFA, ↑LPS, & accumulation of α‐syn in ENS (↑oxidative stress & localized inflammation) ➔ ↑gut permeability & BBB permeability ➔ ↑enteric glial‐related proinflammatory markers such as GFAP, Sox‐10, IL‐6, IL‐1β, TNFα ➔ bidirectional communication between the brain & gut through gut–brain axis, MGB axis, GBNM axis, ENS, Vagus nerve & innate immunity (via TLR signaling) ➔ accumulation of α‐syn & Lewy bodies in CNS & widespread in other nervous systems ➔ neurodegeneration & neuroinflammation ➔ PD Antibiotic use is still a controversial topic for PD patients in research articles (e.g., using antibiotics is a negative effect for microbiota; Koutzoumis et al., 2020), but antibiotics such as Rifaximin is effective to treat SIBO (Aho et al., 2021) |
(+) Mediterranean diet, Ketogenic diet, Food contains PUFA, ω−3FA, High fiber diet, Probiotics, Prebiotics, Synbiotics (such as fermented milk), Polyphenol ‐ high consumption of vegetables & fruits, Caffeine (‐) Western diet (high intake of animal protein, saturated fat, refined grain, sugar, alcohol, high salt diet, high fructose corn syrup & low intake of fruits & vegetables) |
Aho et al. (2021), Cryan et al. (2020), Fan et al. (2019), Fang (2018), Fung et al. (2017), Gatta and Scarpignato (2017), Koutzoumis et al. (2020), O'Donovan et al. (2019), Obrenovich (2018), Rutsch et al. (2020), Uyar and Yildiran (2019) |
Schizophrenia |
Multiple risk factors such as infection, inflammation, antibiotics use, stress, toxins, genes (HLA), food antigens ➔ breakdown TJ, AJ, & BBB ➔ ↑intestinal & BBB permeability through gut‐brain axis ➔ ↑C1q, cytokines, MHC, pentraxin, sCD14, TLRs ➔ autoimmunity & glia abnormalities ➔ Schizophrenia Schizophrenia presents a non‐celiac IgG sensitivity. tTG & deamidation modify toxic peptides from broken‐down gluten proteins may be attacked by T‐cell immune response. Schizophrenia is associated with CD, metabolic & cardiovascular diseases. |
(+) Gluten & casein‐free diet, Probiotics, Prebiotics, High in fruits, vegetable & fiber (‐) Gluten, Bovine milk casein, Western diet, Smoking |
Barber et al. (2019), McAllister et al. (2019), Rudzki and Maes (2020), Severance et al. (2016), Simeonova et al. (2020), Turner (2009), Wahls et al. (2019)) |
Stroke |
Disruption of microbiota diversity, ↑proteobacteria & ↓Bacteroides, Prevotella, & Faecalibaterium, ↓blood trimethylamine N‐oxide ➔ disrupt TJ protein ZO‐1, claudin‐5, & occludin ➔ TJ disassembly ➔ ↑ intestinal permeability & breakdown BBB ➔ ↑D‐Lactate & IFABP (markers of intestinal barrier integrity); ↑IL‐17A, IL‐17 γδ T cells, IFN‐γ & TMA ➔ TMAO ➔ ischemic stroke After stroke ➔ ↑gut permeability ➔ systemic inflammation |
(+) Mediterranean diet, ACN rich diet or ACN supplements (‐) Western diet (red meat, processed meat & saturated fat) |
Camara‐Lemarroy et al. (2021), Cryan et al. (2020), Lourida et al. (2013), Manolescu et al. (2019), Psaltopoulou et al. (2013), Rahman et al. (2018), Tang et al. (2019) |
Ophthalmological diseases | |||
Age related macular degeneration (AMD) | The pathogenesis is through gut–retina axis. Western diet interrupts ecosystem. Reduce gut microbiota – mainly Bacteroidetes & Firmicutes ➔ ↓TJ protein – ZO1 & Occludin ➔ ↑ intestinal permeability ➔ low‐grade chronic systemic inflammation |
(+) ω−3FAs rich food: dietary fish oil and oily fish, wild plants, eggs, nuts, & berries; Low fat, Low glucose/fructose diet; Vitamin C, E, D, Zinc, β‐carotene, Lutein, Zeaxanthin (‐) Western diet, Smoking |
Fasano (2012), Merendino et al. (2013), Miles and Calder (2012), Rinninella et al. (2018), Schleicher et al. (2013), Simopoulos (2008), Watanabe and Tatsuno (2017) |
Dry eye syndrome | Similar as AMD. Increased inflammatory cytokines (IL‐1, IL‐6, & TNFα) in tears | Similar as AMD | Simopoulos (2008), Solomon et al. (2001) |
Abbreviations: (‐), Negative effect/Risk factors; (+), Positive effect/beneficial factors; 5HIAA, 5‐hydroxyindoleacetic acid; 5HT, 5‐hydroxytryptamine; ACN, anthocyanin; ACPA, anti‐citrullinated protein antibodies; ADEs, advanced glycation end products; AHR, aryl hydrocarbon receptor; ALD, alcoholic liver disease; ANS autonomic system; AP‐1, Activator Protein‐1; APC, antigen‐presenting cell; ATTA, anti‐tissue transglutaminase antibodies; BBB, blood brain barrier; BDNF, brain derived neurotrophic factor; BSCB, brain‐spinal cord barrier; C1q, complement component 1q; CLA, cutaneous lymphocyte antigen; CM, chylomicrons; CRP, C‐reactive protein; EGF, epidermal growth factor; FGF 19, fibroblast growth factor 19; FXR, farnesoid X receptor; GABA, gamma‐aminobutyric acid; GALT, gut‐associated lymphatic tissue; GBNM, gut–brain neuroendocrine metabolic; GLUT1 DS, Glucose Transporter 1 Deficiency Syndrome; HGF, hepatocyte growth factor; HLA, human leukocyte antigen; hs‐CRP, high‐sensitivity C‐reactive protein; HVA, homovanillic acid; IAP, intestinal alkaline phosphatase; IFABP, intestinal fatty acid‐binding protein; LBP, LPS‐binding protein; MAFLD, metabolic‐dysfunction‐associated fatty liver disease; MAMPs, microbial‐associated molecular patterns; MGB, microbiota‐gut‐brain; MHC, major histocompatibility complex; NE, norepinephrine; O&NS, oxidative and nitrosative stress; PLN, perihepatic lymph node; RAGE, Receptors for AGEs; RANKL, receptor activator of nuclear factor kappa beta; ROS, reactive oxygen species; sCD, soluble cluster of differentiation; spp, plural of species; Th, T helper cell; TMAO, proatherogenic trimethylamine‐N‐oxide; TSH, thyroid‐stimulating hormone; tTG, tissue Transglutaminase; tTG2, Auto‐antigen‐tissue transglutaminase 2; VEGF, vascular endothelial growth factor; α‐syn, alpha‐synucleinopathy.
6. CONCLUSIONS
Based upon the current literature, there is strong evidence that dietary changes might offer therapeutic strategies to address gut barrier dysfunction. Research is underway to study and examine the relationship between food, gut function, and disease management. Nevertheless, “leaky gut syndrome” is not completely substantiated and there are still conflicts in multiple areas. However, the identification of offending foods can play an important role in the prevention and mitigation of different conditions. Personalized abstaining from harmful food and diet may prevent and treat multiple diseases. Diet could be a controllable lifestyle choice in both positive and negative effects. Diet management could be future personalized medicine.
CONFLICT OF INTEREST STATEMENT
The authors declare no conflict of interest.
ACKNOWLEDGMENTS
None.
Liang, L. , Saunders, C. , & Sanossian, N. (2023). Food, gut barrier dysfunction, and related diseases: A new target for future individualized disease prevention and management. Food Science & Nutrition, 11, 1671–1704. 10.1002/fsn3.3229
DATA AVAILABILITY STATEMENT
Data sharing is not applicable to this article as no datasets were generated or analyzed during the current review.
REFERENCES
- Abdelhamid, L. , & Luo, X. M. (2018). Retinoic acid, leaky gut, and autoimmune diseases. Nutrients, 10, 1016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Aho, V. T. E. , Houser, M. C. , Pereira, P. A. B. , Chang, J. , Rudi, K. , Paulin, L. , Hertzberg, V. , Auvinen, P. , Tansey, M. G. , & Scheperjans, F. (2021). Relationships of gut microbiota, short‐chain fatty acids, inflammation, and the gut barrier in Parkinson's disease. Molecular Neurodegeneration, 16(1), 6. 10.1186/s13024-021-00427-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Aljahdali, N. , & Carbonero, F. (2019). Impact of Maillard reaction products on nutrition and health: Current knowledge and need to understand their fate in the human digestive system. Critical Reviews in Food Science and Nutrition, 59, 474–487. [DOI] [PubMed] [Google Scholar]
- Allaire, J. M. , Crowley, S. M. , Law, H. T. , Chang, S. Y. , Ko, H. J. , & Vallance, B. A. (2018). The intestinal epithelium: Central coordinator of mucosal immunity. Trends in Immunology, 39, 677–696. [DOI] [PubMed] [Google Scholar]
- Al‐Sadi, R. , Nighot, P. , Nighot, M. , Haque, M. , Rawat, M. , & Ma, T. Y. (2021). Lactobacillus acidophilus induces a strain‐specific and toll‐like receptor 2‐dependent enhancement of intestinal epithelial tight junction barrier and protection against intestinal inflammation. The American Journal of Pathology, 191, 872–884. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Al‐Toma, A. , Volta, U. , Auricchio, R. , Castillejo, G. , Sanders, D. S. , Cellier, C. , Mulder, C. J. , & Lundin, K. E. A. (2019). European society for the study of coeliac disease (ESsCD) guideline for coeliac disease and other gluten‐related disorders. United European Gastroenterology Journal, 7, 583–613. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Alvarez‐Mon, M. A. , Gómez, A. M. , Orozco, A. , Lahera, G. , Sosa, M. D. , Diaz, D. , Auba, E. , Albillos, A. , Monserrat, J. , & Alvarez‐Mon, M. (2019). Abnormal distribution and function of circulating monocytes and enhanced bacterial translocation in major depressive disorder. Frontiers in Psychiatry, 10, 812. 10.3389/fpsyt.2019.00812 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Amar, J. (2018). Microbiota–Host crosstalk: A bridge between cardiovascular risk factors, diet, and cardiovascular disease. American Journal of Hypertension, 31(8), 941–944. [DOI] [PubMed] [Google Scholar]
- Amasheh, M. , Andres, S. , Amasheh, S. , Fromm, M. , & Schulzke, J. D. (2009). Barrier effects of nutritional factors. Annals of the New York Academy of Sciences, 1165, 267–273. [DOI] [PubMed] [Google Scholar]
- Amati, L. , Marzulli, G. , Martulli, M. , Pugliese, V. , Caruso, C. , Candore, G. , Vasto, S. , & Jirillo, E. (2010). Administration of a synbiotic to free‐living elderly and evaluation of cytokines. A pilot study. Current Pharmaceutical Design, 16, 854–858. [DOI] [PubMed] [Google Scholar]
- Amornphimoltham, P. , Yuen, P. S. T. , Star, R. A. , & Leelahavanichkul, A. (2019). Gut leakage of fungal‐derived inflammatory mediators: Part of a gut‐liver‐kidney axis in bacterial sepsis. Digestive Diseases and Sciences, 64, 2416–2428. [DOI] [PubMed] [Google Scholar]
- An, S. , Lee, M. S. , Jeon, J. Y. , Ha, E. S. , Kim, T. H. , Yoon, J. Y. , Ok, C. O. , Lee, H. K. , Hwang, W. S. , Choe, S. J. , Han, S. J. , Kim, H. J. , Kim, D. J. , & Lee, K. W. (2013). Beneficial effects of fresh and fermented kimchi in prediabetic individuals. Annals of Nutrition & Metabolism, 63, 111–119. [DOI] [PubMed] [Google Scholar]
- Arab, J. P. , Martin‐Mateos, R. M. , & Shah, V. H. (2018). Gut–liver axis, cirrhosis and portal hypertension: The chicken and the egg. Hepatology International, 12, S24–S33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Arrieta, M. C. , Madsen, K. , Doyle, J. , & Meddings, J. (2009). Reducing small intestinal permeability attenuates colitis in the IL10 gene‐deficient mouse. Gut, 58(1), 41–48. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Arulsamy, A. , Tan, Q. Y. , Balasubramaniam, V. , O'Brien, T. J. , & Shaikh, M. F. (2020). Gut microbiota and epilepsy: A systematic review on their relationship and possible therapeutics. ACS Chemical Neuroscience, 11(21), 3488–3498. 10.1021/acschemneuro.0c00431 [DOI] [PubMed] [Google Scholar]
- Aslam, H. , Green, J. , Jacka, F. N. , Collier, F. , Berk, M. , Pasco, J. , & Dawson, S. L. (2020). Fermented foods, the gut and mental health: A mechanistic overview with implications for depression and anxiety. Nutritional Neuroscience, 23, 659–671. [DOI] [PubMed] [Google Scholar]
- Bai, J. , Hu, Y. , & Bruner, D. W. (2019). Composition of gut microbiota and its association with body mass index and lifestyle factors in a cohort of 7–18 years old children from the American Gut Project. Pediatric Obesity, 14(4), e12480. [DOI] [PubMed] [Google Scholar]
- Bai, N. , Gu, M. , Liu, M. , Jia, Q. , Pan, S. , & Zhang, Z. (2019). Corn gluten meal induces enteritis and decreases intestinal immunity and antioxidant capacity in turbot (Scophthalmus maximus) at high supplementation levels. PLoS One, 14(3), e0213867. 10.1371/journal.pone.0213867 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bajaj, J. S. , Sikaroodi, M. , Fagan, A. , Heuman, D. , Gilles, H. , Gavis, E. A. , Fuchs, M. , Gonzalez‐Maeso, J. , Nizam, S. , Gillevet, P. M. , & Wade, J. B. (2019). Posttraumatic stress disorder is associated with altered gut microbiota that modulates cognitive performance in veterans with cirrhosis. American Journal of Physiology. Gastrointestinal and Liver Physiology, 317, G661–G669. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ballan, R. , Battistini, C. , Xavier‐Santos, D. , & Saad, S. M. I. (2020). Interactions of probiotics and prebiotics with the gut microbiota. Progress in Molecular Biology and Translational Science, 171, 265–300. [DOI] [PubMed] [Google Scholar]
- Bamias, G. , Corridoni, D. , Pizarro, T. T. , & Cominelli, F. (2012). New insights into the dichotomous role of innate cytokines in gut homeostasis and inflammation. Cytokine, 59, 451–459. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bancil, A. S. , Sandall, A. M. , Rossi, M. , Chassaing, B. , Lindsay, J. O. , & Whelan, K. (2021). Food additive emulsifiers and their impact on gut microbiome, permeability, and inflammation: Mechanistic insights in inflammatory bowel disease. Journal of Crohn's & Colitis, 15, 1068–1079. [DOI] [PubMed] [Google Scholar]
- Baraniya, D. , Naginyte, M. , Chen, T. , Albandar, J. M. , Chialastri, S. M. , Devine, D. A. , Marsh, P. D. , & al‐Hebshi, N. N. (2020). Modeling normal and dysbiotic subgingival microbiomes: Effect of nutrients. Journal of Dental Research, 99(6), 695–702. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Barbaro, M. R. , Cremon, C. , Morselli‐Labate, A. M. , di Sabatino, A. , Giuffrida, P. , Corazza, G. R. , di Stefano, M. , Caio, G. , Latella, G. , Ciacci, C. , Fuschi, D. , Mastroroberto, M. , Bellacosa, L. , Stanghellini, V. , Volta, U. , & Barbara, G. (2020). Serum zonulin and its diagnostic performance in non‐coeliac gluten sensitivity. Gut, 69, 1966–1974. [DOI] [PubMed] [Google Scholar]
- Barber, G. S. , Sturgeon, C. , Fasano, A. , Cascella, N. G. , Eaton, W. W. , McMahon, R. , & Kelly, D. L. (2019). Elevated zonulin, a measure of tight‐junction permeability, may be implicated in schizophrenia. Schizophrenia Research, 211, 111–112. [DOI] [PubMed] [Google Scholar]
- Barnes, P. J. , & Adcock, I. M. (2009). Glucocorticoid resistance in inflammatory diseases. Lancet, 373, 1905–1917. [DOI] [PubMed] [Google Scholar]
- Beguin, P. , Errachid, A. , Larondelle, Y. , & Schneider, Y. J. (2013). Effect of polyunsaturated fatty acids on tight junctions in a model of the human intestinal epithelium under normal and inflammatory conditions. Food & Function, 4, 923–931. [DOI] [PubMed] [Google Scholar]
- Bekkering, P. , Jafri, I. , van Overveld, F. J. , & Rijkers, G. T. (2013). The intricate association between gut microbiota and development of type 1, type 2 and type 3 diabetes. Expert Review of Clinical Immunology, 9(11), 1031–1041. 10.1586/1744666X.2013.848793 [DOI] [PubMed] [Google Scholar]
- Beliaeva, E. A. , Chervinets, V. M. , Chervinets, I. V. , Samoukina, A. M. , Mikhaĭlova, E. S. , Piatova, A. I. , & Mironov, A. I. (2013). The disbiotic changes of intestines microflora in healthy people. Klinicheskaia Laboratornaia Diagnostika, (3), 45–47. [PubMed] [Google Scholar]
- Bell, V. , Ferrão, J. , Pimentel, L. , Pintado, M. , & Fernandes, T. (2018). One health, fermented foods, and gut microbiota. Foods (Basel, Switzerland), 7(12), 195. 10.3390/foods7120195 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bengmark, S. (2013). Gut microbiota, immune development and function. Pharmacological Research, 69, 87–113. [DOI] [PubMed] [Google Scholar]
- Berer, K. , Gerdes, L. A. , Cekanaviciute, E. , Jia, X. , Xiao, L. , Xia, Z. , Liu, C. , Klotz, L. , Stauffer, U. , Baranzini, S. E. , Kümpfel, T. , Hohlfeld, R. , Krishnamoorthy, G. , & Wekerle, H. (2017). Gut microbiota from multiple sclerosis patients enables spontaneous autoimmune encephalomyelitis in mice. Proceedings of the National Academy of Sciences, USA, 114(40), 10719–10724. 10.1073/pnas.1711233114 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Binienda, A. , Twardowska, A. , Makaro, A. , & Salaga, M. (2020). Dietary carbohydrates and lipids in the pathogenesis of leaky gut syndrome: An overview. International Journal of Molecular Sciences, 21(21), 8368. 10.3390/ijms21218368 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bischoff, S. C. , Barbara, G. , Buurman, W. , Ockhuizen, T. , Schulzke, J. D. , Serino, M. , Tilg, H. , Watson, A. , & Wells, J. M. (2014). Intestinal permeability – a new target for disease prevention and therapy. BMC Gastroenterology, 14, 189. 10.1186/s12876-014-0189-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bjarnason, I. , & Takeuchi, K. (2009). Intestinal permeability in the pathogenesis of NSAID‐induced enteropathy. Journal of Gastroenterology, 44(19), 23–29. [DOI] [PubMed] [Google Scholar]
- Blacher, E. , Bashiardes, S. , Shapiro, H. , Mor, U. , Dori‐Bachash, M. , Kleimeyer, C. , Moresi, C. , Harnik, Y. , Zur, M. , Zabari, M. , Brik, R. B. , Kviatcovsky, D. , Zmora, N. , Cohen, Y. , Bar, N. , Levi, I. , Amar, N. , Mehlman, T. , Brandis, A. , … Elinav, E. (2019). Potential roles of gut microbiome and metabolites in modulating ALS in mice. Nature, 572, 474–480. [DOI] [PubMed] [Google Scholar]
- Bodinham, C. L. , Smith, L. , Thomas, E. L. , Bell, J. D. , Swann, J. R. , Costabile, A. , Russell‐Jones, D. , Umpleby, A. M. , & Robertson, M. D. (2014). Efficacy of increased resistant starch consumption in human type 2 diabetes. Endocrine Connections, 3(2), 75–84. 10.1530/EC-14-0036 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Braga, M. , Wischmeyer, P. E. , Drover, J. , & Heyland, D. K. (2013). Clinical evidence for pharmaconutrition in major elective surgery. JPEN Journal of Parenteral and Enteral Nutrition, 37, 66Se72S. [DOI] [PubMed] [Google Scholar]
- Braniste, V. , Al‐Asmakh, M. , Kowal, C. , Anuar, F. , Abbaspour, A. , Tóth, M. , Korecka, A. , Bakocevic, N. , Ng, L. G. , Kundu, P. , Gulyás, B. , Halldin, C. , Hultenby, K. , Nilsson, H. , Hebert, H. , Volpe, B. T. , Diamond, B. , & Pettersson, S. (2014). The gut microbiota influences blood‐brain barrier permeability in mice. Science Translational Medicine, 6(263), 263ra158. 10.1126/scitranslmed.3009759 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Briskey, D. , Tucker, P. , Johnson, D. W. , & Coombes, J. S. (2017). The role of the gastrointestinal tract and microbiota on uremic toxins and chronic kidney disease development. Clinical and Experimental Nephrology, 21, 7–15. [DOI] [PubMed] [Google Scholar]
- Budisan, L. , Gulei, D. , Zanoaga, O. M. , Irimie, A. I. , Sergiu, C. , Braicu, C. , Gherman, C. D. , & Berindan‐Neagoe, I. (2017). Dietary intervention by phytochemicals and their role in modulating coding and non‐coding genes in cancer. International Journal of Molecular Sciences, 18(6), 1178. 10.3390/ijms18061178 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Burns, J. , Yokota, T. , Ashihara, H. , Lean, M. E. , & Crozier, A. (2002). Plant foods and herbal sources of resveratrol. Journal of Agricultural and Food Chemistry, 50, 3337–3340. [DOI] [PubMed] [Google Scholar]
- Buscarinu, M. C. , Fornasiero, A. , Romano, S. , Ferraldeschi, M. , Mechelli, R. , Reniè, R. , Morena, E. , Romano, C. , Pellicciari, G. , Landi, A. C. , Salvetti, M. , & Ristori, G. (2019). The contribution of gut barrier changes to multiple sclerosis pathophysiology. Frontiers in Immunology, 10, 1916. 10.3389/fimmu.2019.01916 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Buscarinu, M. C. , Romano, S. , Mechelli, R. , Pizzolato Umeton, R. , Ferraldeschi, M. , Fornasiero, A. , Reniè, R. , Cerasoli, B. , Morena, E. , Romano, C. , Loizzo, N. D. , Umeton, R. , Salvetti, M. , & Ristori, G. (2018). Intestinal permeability in relapsing‐remitting multiple sclerosis. Neurotherapeutics, 15, 68–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Caio, G. , Lungaro, L. , Segata, N. , Guarino, M. , Zoli, G. , Volta, U. , & De Giorgio, R. (2020). Effect of gluten‐free diet on gut microbiota composition in patients with celiac disease and non‐celiac gluten/wheat sensitivity. Nutrients, 12(6), 1832. 10.3390/nu12061832 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Camara‐Lemarroy, C. R. , Escobedo‐Zúñiga, N. , Guzmán‐de la Garza, F. , Castro‐Garza, J. , Vargas‐Villarreal, J. , & Góngora‐Rivera, F. (2021). D‐Lactate and intestinal fatty acid‐binding protein are elevated in serum in patients with acute ischemic stroke. Acta Neurologica Belgica, 121, 87–93. [DOI] [PubMed] [Google Scholar]
- Camara‐Lemarroy, C. R. , Metz, L. M. , & Yong, V. W. (2018). Focus on the gut‐brain axis: Multiple sclerosis, the intestinal barrier and the microbiome. World Journal of Gastroenterology, 24(37), 4217–4223. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Camara‐Lemarroy, C. R. , Silva, C. , Greenfield, J. , Liu, W. Q. , Metz, L. M. , & Yong, V. W. (2020). Biomarkers of intestinal barrier function in multiple sclerosis are associated with disease activity. Multiple Sclerosis (Houndmills, Basingstoke, England), 26(11), 1340–1350. 10.1177/1352458519863133 [DOI] [PubMed] [Google Scholar]
- Camilleri, M. (2019). Leaky gut: Mechanisms, measurement and clinical implications in humans. Gut, 68, 1516–1526. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Camilleri, M. , & Gorman, H. (2007). Intestinal permeability and irritable bowel syndrome. Neurogastroenterology and Motility, 19(7), 545–552. [DOI] [PubMed] [Google Scholar]
- Camilleri, M. , Lyle, B. J. , Madsen, K. L. , Sonnenburg, J. , Verbeke, K. , & Wu, G. D. (2019). Role for diet in normal gut barrier function: Developing guidance within the framework of food‐labeling regulations. American Journal of Physiology. Gastrointestinal and Liver Physiology, 317, G17–G39. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cantarel, B. L. , Waubant, E. , Chehoud, C. , Kuczynski, J. , DeSantis, T. , Warrington, J. , Venkatesan, A. , Fraser, C. M. , & Mowry, E. M. (2015). Gut microbiota in multiple sclerosis: Possible influence of immunomodulators. Journal of Investigative Medicine, 63, 729–734. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Capurso, G. , Zerboni, G. , Signoretti, M. , Valente, R. , Stigliano, S. , Piciucchi, M. , & Fave, G. D. (2012). Role of the gut barrier in acute pancreatitis. Journal of Clinical Gastroenterology, 46, S46–S51. [DOI] [PubMed] [Google Scholar]
- Carding, S. R. , Davis, N. , & Hoyles, L. (2017). Review article: The human intestinal virome in health and disease. Alimentary Pharmacology & Therapeutics, 46, 800–815. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cardoso‐Silva, D. , Delbue, D. , Itzlinger, A. , Moerkens, R. , Withoff, S. , Branchi, F. , & Schumann, M. (2019). Intestinal barrier function in gluten‐related disorders. Nutrients, 11(10), 2325. 10.3390/nu11102325 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cardwell, C. R. , Stene, L. C. , Joner, G. , Cinek, O. , Svensson, J. , Goldacre, M. J. , Parslow, R. C. , Pozzilli, P. , Brigis, G. , Stoyanov, D. , Urbonaite, B. , Sipetić, S. , Schober, E. , Ionescu‐Tirgoviste, C. , Devoti, G. , de Beaufort, C. E. , Buschard, K. , & Patterson, C. C. (2008). Caesarean section is associated with an increased risk of childhood‐onset type 1 diabetes mellitus: A meta‐analysis of observational studies. Diabetologia, 51, 726–735. [DOI] [PubMed] [Google Scholar]
- Cariello, M. , Contursi, A. , Gadaleta, R. M. , Piccinin, E. , De Santis, S. , Piglionica, M. , Spaziante, A. F. , Sabbà, C. , Villani, G. , & Moschetta, A. (2020). Extra‐virgin olive oil from Apulian cultivars and intestinal inflammation. Nutrients, 12(4), 1084. 10.3390/nu12041084 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Carrera‐Bastos, P. , Picazo, O. , Fontes‐Villalba, M. , Pareja‐Galeano, H. , Lindeberg, S. , Martínez‐Selles, M. , Lucia, A. , & Emanuele, E. (2018). Serum zonulin and endotoxin levels in exceptional longevity versus precocious myocardial infarction. Aging and Disease, 9(2), 317–321. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Casselbran, A. , Elias, E. , Fändriks, L. , & Wallenius, V. (2015). Expression of tight‐junction proteins in human proximal small intestinal mucosa before and after Roux‐en‐Y gastric bypass surgery. Surgery for Obesity and Related Diseases, 11, 45–53. [DOI] [PubMed] [Google Scholar]
- Cayres, L. C. F. , de Salis, L. V. V. , Rodrigues, G. S. P. , Lengert, A. V. H. , Biondi, A. P. C. , Sargentini, L. D. B. , Brisotti, J. L. , Gomes, E. , & de Oliveira, G. L. V. (2021). Detection of alterations in the gut microbiota and intestinal permeability in patients with Hashimoto thyroiditis. Frontiers in Immunology, 12, 579140. 10.3389/fimmu.2021.579140 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen, A. , Park, T. , Li, K. , & DeLisi, L. (2020). Antipsychotics and the microbiota. Current Opinion in Psychiatry, 33, 225–230. [DOI] [PubMed] [Google Scholar]
- Chen, J. , Chia, N. , Kalari, K. R. , Yao, J. Z. , Novotna, M. , Paz Soldan, M. M. , Luckey, D. H. , Marietta, E. V. , Jeraldo, P. R. , Chen, X. , Weinshenker, B. G. , Rodriguez, M. , Kantarci, O. H. , Nelson, H. , Murray, J. A. , & Mangalam, A. K. (2018). Multiple sclerosis patients have a distinct gut microbiota compared to healthy controls. Microorganisms, 6, 28484. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen, L. , Zou, Y. , Peng, J. , Lu, F. , Yin, Y. , Li, F. , & Yang, J. (2015). Lactobacillus acidophilus suppresses colitis‐associated activation of the IL‐23/Th17 axis. Journal of Immunology Research, 2015, 909514. 10.1155/2015/909514 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen, X. , Zhang, Z. , Cui, B. , Jiang, A. , Tao, H. , Cheng, S. , & Liu, Y. (2020). Combination of chronic alcohol consumption and high‐salt intake elicits gut microbial alterations and liver steatosis in mice. Journal of Agricultural and Food Chemistry, 68, 1750–1759. [DOI] [PubMed] [Google Scholar]
- Cho, Y.‐E. , Kim, D.‐K. , Seo, W. , Gao, B. , Yoo, S. H. , & Song, B. J. (2021). Fructose promotes leaky gut, endotoxemia, and liver fibrosis through ethanol‐inducible cytochrome P450‐2E1–mediated oxidative and nitrative stress. Hepatology, 73, 2180–2195. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chou, W. J. , Lee, M. F. , Hou, M. L. , Hsiao, L. S. , Lee, M. J. , Chou, M. C. , & Wang, L. J. (2018). Dietary and nutrient status of children with attentiondefcit/hyperactivity disorder: A case‐control study. Asia Pacific Journal of Clinical Nutrition, 27(6), 1325–1331. [DOI] [PubMed] [Google Scholar]
- Chu, F. , Shi, M. , Lang, Y. , Shen, D. , Jin, T. , Zhu, J. , & Cui, L. (2018). Gut microbiota in multiple sclerosis and experimental autoimmune encephalomyelitis: Current applications and future perspectives. Mediators of Inflammation, 2018, 8168717. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ciccia, F. , Accardo‐Palumbo, A. , Rizzo, A. , Guggino, G. , Raimondo, S. , Giardina, A. , Cannizzaro, A. , Colbert, R. A. , Alessandro, R. , & Triolo, G. (2014). Evidence that autophagy, but not the unfolded protein response, regulates the expression of IL‐23 in the gut of patients with ankylosing spondylitis and subclinical gut inflammation. Annals of the Rheumatic Diseases, 73, 1566–1574. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ciccia, F. , Guggino, G. , Rizzo, A. , Alessandro, R. , Luchetti, M. M. , Milling, S. , Saieva, L. , Cypers, H. , Stampone, T. , di Benedetto, P. , Gabrielli, A. , Fasano, A. , Elewaut, D. , & Triolo, G. (2017). Dysbiosis and zonulin upregulation alter gut epithelial and vascular barriers in patients with ankylosing spondylitis. Annals of the Rheumatic Diseases, 76(6), 1123–1132. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Codoñer, F. M. , Ramírez‐Bosca, A. , Climent, E. , Carrión‐Gutierrez, M. , Guerrero, M. , Pérez‐Orquín, J. M. , Horga de la Parte, J. , Genovés, S. , Ramón, D. , Navarro‐López, V. , & Chenoll, E. (2018). Gut microbial composition in patients with psoriasis. Scientific Reports, 8(1), 3812. 10.1038/s41598-018-22125-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cordain, L. , Toohey, L. , Smith, M. J. , & Hickey, M. S. (2000). Modulation of immune function by dietary lectins in rheumatoid arthritis. British Journal of Nutrition, 83, 207–217. [DOI] [PubMed] [Google Scholar]
- Costello, M. E. , Ciccia, F. , Willner, D. , Warrington, N. , Robinson, P. C. , Gardiner, B. , Marshall, M. , Kenna, T. J. , Triolo, G. , & Brown, M. A. (2015). Brief report: Intestinal dysbiosis in ankylosing spondylitis. Arthritis & Rheumatology (Hoboken, N.J.), 67(3), 686–691. 10.1002/art.38967 [DOI] [PubMed] [Google Scholar]
- Cox, S. , Sandall, A. , Smith, L. , Rossi, M. , & Whelan, K. (2021). Food additive emulsifiers: A review of their role in foods, legislation and classifications, presence in food supply, dietary exposure, and safety assessment. Nutrition Reviews, 79(6), 726–741. [DOI] [PubMed] [Google Scholar]
- Cryan, J. F. , O'Riordan, K. J. , Sandhu, K. , Peterson, V. , & Dinan, T. G. (2020). The gut microbiome in neurological disorders. Lancet Neurology, 19, 179–194. [DOI] [PubMed] [Google Scholar]
- Csáki, K. F. (2011). Synthetic surfactant food additives can cause intestinal barrier dysfunction. Medical Hypotheses, 76, 676–681. [DOI] [PubMed] [Google Scholar]
- Dahlin, M. , & Prast‐Nielsen, S. (2019). The gut microbiome and epilepsy. eBioMedicine, 44, 741–746. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dai, Y. , Wang, H. , Wang, X. , Kaye, A. D. , & Sun, Y. H. (2017). Potential beneficial effects of probiotics on human migraine headache: A literature review. Pain Physician, 20, E251–E255. [PubMed] [Google Scholar]
- De Angelis, M. , Garruti, G. , Minervini, F. , Bonfrate, L. , Portincasa, P. , & Gobbetti, M. (2019). The food‐gut human axis: The effects of diet on gut microbiota and metabolome. Current Medicinal Chemistry, 26(19), 3567–3583. 10.2174/0929867324666170428103848 [DOI] [PubMed] [Google Scholar]
- de Kort, S. , Keszthelyi, D. , & Masclee, A. A. M. (2011). Leaky gut and diabetes mellitus: What is the link? Obesity Reviews, 12, 449–458. [DOI] [PubMed] [Google Scholar]
- de Punder, K. , & Pruimboom, L. (2013). The dietary intake of wheat and other cereal grains and their role in inflammation. Nutrients, 5, 771–787. [DOI] [PMC free article] [PubMed] [Google Scholar]
- De Santis, S. , Cavalcanti, E. , Mastronardi, M. , Jirillo, E. , & Chieppa, M. (2015). Nutritional keys for intestinal barrier modulation. Frontiers in Immunology, 6, 612. 10.3389/fimmu.2015.00612 [DOI] [PMC free article] [PubMed] [Google Scholar]
- DeJong, E. N. , Surette, M. G. , & Bowdish, D. M. E. (2020). The gut microbiota and unhealthy aging: Disentangling cause from consequence. Cell Host & Microbe, 28, 2,180–2,189. [DOI] [PubMed] [Google Scholar]
- DeMeo, M. T. , Mutlu, E. A. , Keshavarzian, A. , & Tobin, M. C. (2002). Intestinal permeation and gastrointestinal disease. Journal of Clinical Gastroenterology, 34(4), 385–396. [DOI] [PubMed] [Google Scholar]
- De‐Paula, V. J. R. , Forlenza, A. S. , & Forlenza, O. V. (2018). Relevance of gut microbiota in cognition, behaviour and Alzheimer's disease. Pharmacological Research, 136, 29–34. [DOI] [PubMed] [Google Scholar]
- Dharaneedharan, S. , & Heo, M. S. (2016). Korean Traditional fermented foods – A potential resource of beneficial microorganisms and their applications. Journal of Life Sciences, 26, 496–502. [Google Scholar]
- Di Ciaula, A. , Baj, J. , Garruti, G. , Celano, G. , De Angelis, M. , Wang, H. H. , Di Palo, D. M. , Bonfrate, L. , Wang, D. Q. , & Portincasa, P. (2020). Liver steatosis, gut‐liver axis, microbiome and environmental factors. A never‐ending bidirectional cross‐talk. Journal of Clinical Medicine, 9, 2648. 10.3390/jcm9082648 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Di Palo, D. M. , Garruti, G. , Di Ciaula, A. , Molina‐Molina, E. , Shanmugam, H. , De Angelis, M. , & Portincasa, P. (2020). Increased colonic permeability and lifestyles as contributing factors to obesity and liver steatosis. Nutrients, 12(2), 564. 10.3390/nu12020564 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Díaz‐Coránguez, M. , Segovia, J. , López‐Ornelas, A. , Puerta‐Guardo, H. , Ludert, J. , Chávez, B. , Meraz‐Cruz, N. , & González‐Mariscal, L. (2013). Transmigration of neural stem cells across the blood brain barrier induced by glioma cells. PLoS One, 8(4), e60655. 10.1371/journal.pone.0060655 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dinan, T. G. , & Cryan, J. F. (2017). The microbiome‐gut‐brain axis in health and disease. Gastroenterology Clinics of North America, 46, 77–89. [DOI] [PubMed] [Google Scholar]
- Do, M. H. , Lee, E. , Oh, M. J. , Kim, Y. , & Park, H. Y. (2018). High‐glucose or ‐fructose diet cause changes of the gut microbiota and metabolic disorders in mice without body weight change. Nutrients, 10(6), 761. 10.3390/nu10060761 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Doney, E. , Cadoret, A. , Dion‐Albert, L. , Lebel, M. , & Menard, C. (2021). Inflammation‐driven brain and gut barrier dysfunction in stress and mood disorders. The European Journal of Neuroscience, 55, 2851–2894. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Doré, J. , & G Corthier, G. (2010). The human intestinal microbiota. Gastroentérologie Clinique et Biologique, 34, S7–S15. [DOI] [PubMed] [Google Scholar]
- Drago, S. , El Asmar, R. , Di Pierro, M. , Grazia Clemente, M. , Tripathi, A. , Sapone, A. , Thakar, M. , Iacono, G. , Carroccio, A. , D'Agate, C. , Not, T. , Zampini, L. , Catassi, C. , & Fasano, A. (2006). Gliadin, zonulin and gut permeability: Effects on celiac and non‐celiac intestinal mucosa and intestinal cell lines. Scandinavian Journal of Gastroenterology, 41(4), 408–419. 10.1080/00365520500235334 [DOI] [PubMed] [Google Scholar]
- Duerksen, D. R. , Wilhelm‐Boyles, C. , Veitch, R. , Kryszak, D. , & Parry, D. M. (2010). A Comparison of antibody testing, permeability testing, and zonulin levels with small‐bowel biopsy in celiac disease patients on a gluten‐free diet. Digestive Diseases and Sciences, 55, 1026–1031. [DOI] [PubMed] [Google Scholar]
- Ejtahed, H. S. , Mohtadi‐Nia, J. , Homayouni‐Rad, A. , Niafar, M. , Asghari‐Jafarabadi, M. , & Mofid, V. (2012). Probiotic yogurt improves antioxidant status in type 2 diabetic patients. Nutrition, 28, 539–543. [DOI] [PubMed] [Google Scholar]
- Ellebrecht, C. T. , Srinivas, G. , Bieber, K. , Banczyk, D. , Kalies, K. , Künzel, S. , Hammers, C. M. , Baines, J. F. , Zillikens, D. , Ludwig, R. J. , & Westermann, J. (2016). Skin microbiota‐associated inflammation precedes autoantibody induced tissue damage in experimental epidermolysis bullosa acquisita. Journal of Autoimmunity, 68, 14–22. [DOI] [PubMed] [Google Scholar]
- Ersoz Alan, B. , & Gulerman, F. (2019). The role of gut microbiota in autism spectrum disorder. Türk Psikiyatri Dergisi, 30(3), u23560. [PubMed] [Google Scholar]
- Esnafoglu, E. , Cırrık, S. , Ayyıldız, S. N. , Erdil, A. , Ertürk, E. Y. , Daglı, A. , & Noyan, T. (2017). Increased serum zonulin levels as an intestinal permeability marker in autistic subjects. The Journal of Pediatrics, 188, 240–244. [DOI] [PubMed] [Google Scholar]
- Esposito, S. , Bonavita, S. , Sparaco, M. , Gallo, A. , & Tedeschi, G. (2018). The role of diet in multiple sclerosis: A review. Nutritional Neuroscience, 21, 377–390. [DOI] [PubMed] [Google Scholar]
- Fan, Y. , Wang, H. , Liu, X. , Zhang, J. , & Liu, G. (2019). Crosstalk between the ketogenic diet and epilepsy: From the perspective of gut microbiota. Mediators of Inflammation, 2019, 8373060. 10.1155/2019/8373060 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fang, X. (2018). Impaired tissue barriers as potential therapeutic targets for Parkinson's disease and amyotrophic lateral sclerosis. Metabolic Brain Disease, 33, 1031–1043. [DOI] [PubMed] [Google Scholar]
- FAO/WHO . (2002). Guidelines for the evaluation of probiotics in food. Food and Agriculture Organization of the United Nations and World Health Organization Working Group Report. [Google Scholar]
- Farré, R. , Fiorani, M. , Rahiman, S. A. , & Matteoli, G. (2020). Intestinal permeability, inflammation and the role of nutrients. Nutrients, 12, 1185. 10.3390/nu12041185 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Farshchi, M. K. , Azad, F. J. , Salari, R. , Mirsadraee, M. , & Anushiravani, M. (2017). A viewpoint on the leaky gut syndrome to treat allergic asthma: A novel opinion. Journal of Evidence‐Based Complementary and Alternative Medicine, 22, 378–380. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fasano, A. (2011). Zonulin and its regulation of intestinal barrier function: The biological door to inflammation, autoimmunity, and cancer. Physiological Reviews, 91, 151–175. [DOI] [PubMed] [Google Scholar]
- Fasano, A. (2012). Leaky gut and autoimmune diseases. Clinical Reviews in Allergy and Immunology, 42, 71–78. [DOI] [PubMed] [Google Scholar]
- Fasano, A. (2020). All disease begins in the (leaky) gut: Role of zonulin‐mediated gut permeability in the pathogenesis of some chronic inflammatory diseases. F1000Research, 9, 69. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fattorusso, A. , Di Genova, L. , Dell'Isola, G. B. , Mencaroni, E. , & Esposito, S. (2019). Autism spectrum disorders and the gut microbiota. Nutrients, 11(3), 521. 10.3390/nu11030521 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fenneman, A. C. , Rampanelli, E. , Yin, Y. S. , Ames, J. , Blaser, M. J. , Fliers, E. , & Nieuwdorp, M. (2020). Gut microbiota and metabolites in the pathogenesis of endocrine disease. Biochemical Society Transactions, 48, 915–931. [DOI] [PubMed] [Google Scholar]
- Ferrari, E. , Monzani, R. , Saverio, V. , Gagliardi, M. , Pańczyszyn, E. , Raia, V. , Villella, V. R. , Bona, G. , Pane, M. , Amoruso, A. , & Corazzari, M. (2021). Probiotics supplements reduce ER stress and gut inflammation associated with gliadin intake in a mouse model of gluten sensitivity. Nutrients, 13(4), 1221. 10.3390/nu13041221 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ferreira, R. D. S. , Mendonça, L. A. B. M. , Ribeiro, C. F. A. , Calças, N. C. , Guimarães, R. C. A. , Nascimento, V. A. D. , Gielow, K. C. F. , Carvalho, C. M. E. , Castro, A. P. , & Franco, O. L. (2022). Relationship between intestinal microbiota, diet and biological systems: An integrated view. Critical Reviews in Food Science and Nutrition, 62(5), 1166–1186. 10.1080/10408398.2020.1836605 [DOI] [PubMed] [Google Scholar]
- Ferro, D. , Baratta, F. , Pastori, D. , Cocomello, N. , Colantoni, A. , Angelico, F. , & Del Ben, M. (2020). New insights into the pathogenesis of non‐alcoholic fatty liver disease: Gut‐derived lipopolysaccharides and oxidative stress. Nutrients, 12(9), 2762. 10.3390/nu12092762 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Filippis, F. , Pellegrini, N. , Vannini, L. , Jeffery, I. B. , la Storia, A. , Laghi, L. , Serrazanetti, D. I. , di Cagno, R. , Ferrocino, I. , Lazzi, C. , Turroni, S. , Cocolin, L. , Brigidi, P. , Neviani, E. , Gobbetti, M. , O'Toole, P. W. , & Ercolini, D. (2016). High‐level adherence to a Mediterranean diet beneficially impacts the gut microbiota and associated metabolome. Gut, 65, 1812–1821. [DOI] [PubMed] [Google Scholar]
- Finamore, A. M. , Massimi, L. , Conti Devirgiliis, L. , & Mengheri, E. (2008). Zinc deficiency induces membrane barrier damage and increases neutrophil transmigration in Caco‐2 cells. The Journal of Nutrition, 138, 1664–1670. [DOI] [PubMed] [Google Scholar]
- Fiolet, T. , Srour, B. , Sellem, L. , Kesse‐Guyot, E. , Allès, B. , Méjean, C. , Deschasaux, M. , Fassier, P. , Latino‐Martel, P. , Beslay, M. , Hercberg, S. , Lavalette, C. , Monteiro, C. A. , Julia, C. , & Touvier, M. (2018). Consumption of ultra‐processed foods and cancer risk: results from NutriNet‐Santé prospective cohort. BMJ (Clinical research ed.), 360, k322. 10.1136/bmj.k322 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Foster, J. A. , Rinaman, L. , & Cryan, J. F. (2017). Stress & the gut‐brain axis: Regulation by the microbiome. Neurobiology Stress, 7, 124–136. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fowlie, G. , Cohen, N. , & Ming, X. (2018). The perturbance of microbiome and gut‐brain axis in autism spectrum disorders. International Journal of Molecular Sciences, 19, 2251–2255. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fox, M. , Knorr, D. A. , & Haptonstall, K. M. (2019). Alzheimer's disease and symbiotic microbiota: An evolutionary medicine perspective. The New York Academy of Sciences, 1449(3), 24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Frazier, T. H. , DiBaise, J. K. , & McClain, C. J. (2011). Gut microbiota, intestinal permeability, obesity‐induced inflammation, and liver injury. Journal of Parenteral and Enteral Nutrition, 35, 14S–20S. [DOI] [PubMed] [Google Scholar]
- Fukui, H. (2016). Increased intestinal permeability and decreased barrier function: Does it really influence the risk of inflammation? Inflammatory Intestinal Diseases, 1, 135–145. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fung, T. C. , Olson, C. A. , & Hsiao, E. Y. (2017). Interactions between the microbiota, immune and nervous systems in health and disease. Nature Neuroscience, 20(2), 145–155. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Galland, L. (2014). The gut microbiome and the brain. Journal of Medicinal Food, 17, 1261–1272. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ganda Mall, J. P. , Fart, F. , Sabet, J. A. , Lindqvist, C. M. , Nestestog, R. , Hegge, F. T. , Keita, Å. V. , Brummer, R. J. , & Schoultz, I. (2020). Effects of dietary fibres on acute indomethacin‐induced intestinal hyperpermeability in the elderly: A randomised placebo controlled parallel clinical trial. Nutrients, 12(7), 1954. 10.3390/nu12071954 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gatta, L. , & Scarpignato, C. (2017). Systematic review with meta‐analysis: Rifaximin is effective and safe for the treatment of small intestine bacterial overgrowth. Alimentary Pharmacology & Therapeutics, 45(5), 604–616. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gatouillat, G. , Balasse, E. , Joseph‐Pietras, D. , Morjani, H. , & Madoulet, C. (2010). Resveratrol induces cell‐cycle disruption and apoptosis in chemoresistant B16 melanoma. Journal of Cellular Biochemistry, 110(4), 893–902. 10.1002/jcb.22601 [DOI] [PubMed] [Google Scholar]
- Gerard, P. (2016). Gut microbiota and obesity. Cellular and Molecular Life Sciences, 73, 147–161. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ghosh, D. , & Konishi, T. (2007). Anthocyanins and anthocyanin‐rich extracts: Role in diabetes and eye function. Asia Pacific Journal of Clinical Nutrition, 16, 200. [PubMed] [Google Scholar]
- Gianchecchi, E. , & Fierabracci, A. (2017). On the pathogenesis of insulin‐dependent diabetes mellitus: the role of microbiota. Immunologic Research, 65(1), 242–256. 10.1007/s12026-016-8832-8 [DOI] [PubMed] [Google Scholar]
- Gibson, G. R. , Probert, H. M. , Loo, J. V. , Rastall, R. A. , & Roberfroid, M. B. (2004). Dietary modulation of the human colonic microbiota: Updating the concept of prebiotics. Nutrition Research Reviews, 17, 259–275. [DOI] [PubMed] [Google Scholar]
- Gil‐Cardoso, K. , Ginés, I. , Pinent, M. , Ardévol, A. , Blay, M. , & Terra, X. (2016). Effects of flavonoids on intestinal inflammation, barrier integrity and changes in gut microbiota during diet‐induced obesity. Nutrition Research Reviews, 29(2), 234–248. [DOI] [PubMed] [Google Scholar]
- Grigg, J. B. , & Sonnenberg, G. F. (2017). Host‐microbiota interactions shape local and systemic inflammatory diseases. Journal of Immunology, 198, 564–571. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Guagnozzi, D. , González‐Castillo, S. , Olveira, A. , & Lucendo, A. J. (2012). Nutritional treatment in inflammatory bowel disease. An update. Revista Española de Enfermedades Digestivas, 104, 479–488. [DOI] [PubMed] [Google Scholar]
- Guercio Nuzio, S. , Di Stasi, M. , Pierri, L. , Troisi, J. , Poeta, M. , Bisogno, A. , Belmonte, F. , Tripodi, M. , Di Salvio, D. , Massa, G. , Savastano, R. , Cavallo, P. , Boffardi, M. , Ziegenhardt, D. , Bergheim, I. , Mandato, C. , & Vajro, P. (2017). Multiple gut‐liver axis abnormalities in children with obesity with and without hepatic involvement. Pediatric Obesity, 12(6), 446–452. 10.1111/ijpo.12164 [DOI] [PubMed] [Google Scholar]
- Guo, R. , Sakamoto, H. , Sugiura, S. , & Ogawa, M. (2007). Endothelial cell motility is compatible with junctional integrity. Journal of Cellular Physiology, 211(2), 327–335. [DOI] [PubMed] [Google Scholar]
- Han, K. , Bose, S. , Wang, J. H. , Kim, B. S. , Kim, M. J. , Kim, E. J. , & Kim, H. (2015). Contrasting effects of fresh and fermented kimchi consumption on gut microbiota composition and gene expression related to metabolic syndrome in obese Korean women. Molecular Nutrition & Food Research, 59, 1004–1008. [DOI] [PubMed] [Google Scholar]
- Hansen, N. W. , & Sams, A. (2018). The microbiotic highway to health—new perspective on food structure, gut microbiota, and host inflammation. Nutrients, 10, 1590. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hauk, P. J. (2010). The role of food allergy in atopic dermatitis. Allergy and Asthma Proceedings, 31, 392–397. [DOI] [PubMed] [Google Scholar]
- Hellwig, M. , & Henle, T. (2014). Baking, ageing, diabetes: A short history of the Maillard reaction. Angewandte Chemie (International Ed. in English), 53, 10316–10329. [DOI] [PubMed] [Google Scholar]
- Herpertz‐Dahlmann, B. , Seitz, J. , & Baines, J. (2017). Food matters: How the microbiome and gut–brain interaction might impact the development and course of anorexia nervosa. European Child & Adolescent Psychiatry, 26, 1031–1041. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hidalgo‐Cantabrana, C. , Gómez, J. , Delgado, S. , Requena‐López, S. , Queiro‐Silva, R. , Margolles, A. , Coto, E. , Sánchez, B. , & Coto‐Segura, P. (2019). Gut microbiota dysbiosis in a cohort of patients with psoriasis. The British Journal of Dermatology, 181, 1287–1295. [DOI] [PubMed] [Google Scholar]
- Hijazi, Z. , Molla, A. M. , al‐Habashi, H. , Muawad, W. M. , Molla, A. M. , & Sharma, P. N. (2004). Intestinal permeability is increased in bronchial asthma. Archives of Disease in Childhood, 89, 227–229. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hills, R. D., Jr. , Pontefract, B. A. , Mishcon, H. R. , Black, C. A. , Sutton, S. C. , & Theberge, C. R. (2019). Gut microbiome: Profound implications for diet and disease. Nutrients, 11, 1613. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hobby, G. P. , Karaduta, O. , Dusio, G. F. , Singh, M. , Zybailov, B. L. , & Arthur, J. M. (2019). Chronic kidney disease and the gut microbiome. American Journal of Physiology, Renal Physiology, 316(6), F1211–F1217. 10.1152/ajprenal.00298.2018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hoggan, R. (1997). Considering wheat, rye, and barley proteins as aids to carcinogens. Medical Hypotheses, 49, 285–288. [DOI] [PubMed] [Google Scholar]
- Hong, M. , Han, D. H. , Hong, J. , Kim, D. J. , & Suk, K. T. (2019). Are probiotics effective in targeting alcoholic liver diseases? Probiotics and Antimicrobial Proteins, 11, 335–347. [DOI] [PubMed] [Google Scholar]
- Hossen, I. , Hua, W. , Ting, L. , Mehmood, A. , Jingyi, S. , Duoxia, X. , Yanping, C. , Hongqing, W. , Zhipeng, G. , Kaiqi, Z. , Fang, Y. , & Junsong, X. (2020). Phytochemicals and inflammatory bowel disease: A review. Critical Reviews in Food Science and Nutrition, 60(8), 1321–1345. [DOI] [PubMed] [Google Scholar]
- Hughes, H. K. , Rose, D. , & Ashwood, P. (2018). The gut microbiota and dysbiosis in autism spectrum disorders. Current Neurology and Neuroscience Reports, 18, 81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hul, M. , Geurts, L. , Plovier, H. , Druart, C. , Everard, A. , Ståhlman, M. , Rhimi, M. , Chira, K. , Teissedre, P. L. , Delzenne, N. M. , Maguin, E. , Guilbot, A. , Brochot, A. , Gérard, P. , Bäckhed, F. , & Cani, P. D. (2018). Reduced obesity, diabetes, and steatosis upon cinnamon and grape pomace are associated with changes in gut microbiota and markers of gut barrier. American Journal of Physiology. Endocrinology and Metabolism, 314(4), E334–E352. [DOI] [PubMed] [Google Scholar]
- Ilchmann‐Diounou, H. , & Menard, S. (2020). Psychological stress, intestinal barrier dysfunctions, and autoimmune disorders: An overview. Frontiers in Immunology, 11, 1823. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Imhann, F. , Bonder, M. J. , Vich Vila, A. , Fu, J. , Mujagic, Z. , Vork, L. , Tigchelaar, E. F. , Jankipersadsing, S. A. , Cenit, M. C. , Harmsen, H. J. , Dijkstra, G. , Franke, L. , Xavier, R. J. , Jonkers, D. , Wijmenga, C. , Weersma, R. K. , & Zhernakova, A. (2016). Proton pump inhibitors affect the gut microbiome. Gut, 65, 740–748. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Iqbal, U. , Chaudhary, A. , Karim, M. A. , Siddiqui, M. A. , Anwar, H. , & Merrell, N. (2017). Association of autoimmune hepatitis and celiac disease: Role of gluten‐free diet in reversing liver dysfunction. Journal of Investigative Medicine High Impact Case Reports, 5, 1–3. 10.1177/2324709617705679 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jaervinen, K. M. , Turpeinen, M. , & Suomalainen, H. (2003). Concurrent cereal allergy in children with cow's milk allergy manifested with atopic dermatitis. Clinical and Experimental Allergy, 33, 1060–1066. [DOI] [PubMed] [Google Scholar]
- Jangi, S. , Gandhi, R. , Cox, L. M. , Li, N. , von Glehn, F. , Yan, R. , Patel, B. , Mazzola, M. A. , Liu, S. , Glanz, B. L. , Cook, S. , Tankou, S. , Stuart, F. , Melo, K. , Nejad, P. , Smith, K. , Topçuolu, B. D. , Holden, J. , Kivisäkk, P. , … Weiner, H. L. (2016). Alterations of the human gut microbiome in multiple sclerosis. Nature Communications, 7, 12015. 10.1038/ncomms12015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jaworska, K. , Huc, T. , Samborowska, E. , Dobrowolski, L. , Bielinska, K. , Gawlak, M. , & Ufnal, M. (2017). Hypertension in rats is associated with an increased permeability of the colon to TMA, a gut bacteria metabolite. PLoS One, 12(12), e0189310. 10.1371/journal.pone.0189310 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jayashree, B. , Bibin, Y. S. , Prabhu, D. , Shanthirani, C. S. , Gokulakrishnan, K. , Lakshmi, B. S. , Mohan, V. , & Balasubramanyam, M. (2014). Increased circulatory levels of lipopolysaccharide (LPS) and zonulin signify novel biomarkers of proinflammation in patients with type 2 diabetes. Molecular and Cellular Biochemistry, 388, 203–210. [DOI] [PubMed] [Google Scholar]
- Jiang, Z. , Zhao, M. , Zhang, H. , Li, Y. , Liu, M. , & Feng, F. (2018). Antimicrobial emulsifier‐glycerol monolaurate induces metabolic syndrome, gut microbiota dysbiosis, and systemic low‐grade inflammation in low‐fat diet fed mice. Molecular Nutrition & Food Research, 62(3). 10.1002/mnfr.201700547 [DOI] [PubMed] [Google Scholar]
- Jiang, H. Y. , Zhang, X. , Yu, Z. H. , Zhang, Z. , Deng, M. , Zhao, J. H. , & Ruan, B. (2018). Altered gut microbiota profile in patients with generalized anxiety disorder. Journal of Psychiatric Research, 104, 130–136. 10.1016/j.jpsychires.2018.07.007 [DOI] [PubMed] [Google Scholar]
- Jonathan, G. , Thomas, J. G. , & Veznedaroglu, E. (2020). Ketogenic diet for malignant gliomas: A review. Current Nutrition Reports, 9, 258–263. [DOI] [PubMed] [Google Scholar]
- Jonnalagadda, S. S. , Harnack, L. , Liu, R. H. , McKeown, N. , Seal, C. , Liu, S. , & Fahey, G. C. (2011). Putting the whole grain puzzle together: Health benefits associated with whole grains—summary of American Society for Nutrition 2010 Satellite Symposium. The Journal of Nutrition, 141, 1011S–1022S. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Józefczuk, J. , Konopka, E. , Bierła, J. B. , Trojanowska, I. , Sowińska, A. , Czarnecki, R. , Sobol, L. , Józefczuk, P. , Surdy, W. , & Cukrowska, B. (2018). The occurrence of antibodies against gluten in children with autism spectrum disorders does not correlate with serological markers of impaired intestinal permeability. Journal of Medicinal Food, 21(2), 181–187. [DOI] [PubMed] [Google Scholar]
- Judkins, T. C. , Archer, D. L. , Kramer, D. C. , & Solch, R. J. (2020). Probiotics, nutrition, and the small intestine. Current Gastroenterology Reports, 22(1), 2. 10.1007/s11894-019-0740-3 [DOI] [PubMed] [Google Scholar]
- Julio‐Pieper, M. , López‐Aguilera, A. , Eyzaguirre‐Velásquez, J. , Olavarría‐Ramírez, L. , Ibacache‐Quiroga, C. , Bravo, J. A. , & Cruz, G. (2021). Gut susceptibility to viral invasion: contributing roles of diet, microbiota and enteric nervous system to mucosal barrier preservation. International Journal of Molecular Sciences, 22(9), 4734. 10.3390/ijms22094734 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kaliannan, K. , Wang, B. , Li, X. Y. , Kim, K. J. , & Kang, J. X. (2015). A host‐microbiome interaction mediates the opposing effects of omega‐6 and omega‐3 fatty acids on metabolic endotoxemia. Scientific Reports, 5, 11276. 10.1038/srep11276 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kaličanin, D. , Brčić, L. , Ljubetić, K. , Barić, A. , Gračan, S. , Brekalo, M. , Torlak Lovrić, V. , Kolčić, I. , Polašek, O. , Zemunik, T. , Punda, A. , & Boraska Perica, V. (2020). Differences in food consumption between patients with Hashimoto's thyroiditis and healthy individuals. Scientific Reports, 10(1), 10670. 10.1038/s41598-020-67719-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kang, J. X. (2011). The omega‐6/omega‐3 fatty acid ratio in chronic diseases: Animal models and molecular aspects. World Review of Nutrition and Dietetics, 102, 22–29. [DOI] [PubMed] [Google Scholar]
- Karakuła‐Juchnowicz, H. , Szachta, P. , Opolska, A. , Morylowska‐Topolska, J. , Gałęcka, M. , Juchnowicz, D. , Krukow, P. , & Lasik, Z. (2017). The role of IgG hypersensitivity in the pathogenesis and therapy of depressive disorders. Nutritional Neuroscience, 20(2), 110–118. [DOI] [PubMed] [Google Scholar]
- Karl, J. P. , Margolis, L. M. , Madslien, E. H. , Castellani, J. W. , Gundersen, Y. , Hoke, A. V. , Levangie, M. W. , Kumar, R. , Chakraborty, N. , Gautam, A. , Hammamieh, R. , Martini, S. , Montain, S. J. , Pasiakos, S. M. , & Pasiakos, S. M. (2017). Changes in intestinal microbiota composition and metabolism coincide with increased intestinal permeability in young adults under prolonged physiological stress. American Journal of Physiology. Gastrointestinal and Liver Physiology, 312, G559–G571. [DOI] [PubMed] [Google Scholar]
- Karlsson, F. H. , Fåk, F. , Nookaew, I. , Tremaroli, V. , Fagerberg, B. , Petranovic, D. , Bäckhed, F. , & Nielsen, J. (2012). Symptomatic atherosclerosis is associated with an altered gut metagenome. Nature Communications, 3, 1245. 10.1038/ncomms2266 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kawasaki, T. , & Kawai, T. (2014). Toll‐like receptor signaling pathways. Frontiers in Immunology, 5, 461. 10.3389/fmmu.2014.00461 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ke, W. , Bonilla‐Rosso, G. , Engel, P. , Wang, P. , Chen, F. , & Hu, X. (2020). Suppression of high‐fat diet–induced obesity by platycodon grandiflorus in mice is linked to changes in the gut microbiota. The Journal of Nutrition, 150, 2364–2374. [DOI] [PubMed] [Google Scholar]
- Kellett, G. L. , Brot‐Laroche, E. , Mace, O. J. , & Leturque, A. (2008). Sugar absorption in the Intestine: The role of GLUT2. Annual Review of Nutrition, 28, 35–54. [DOI] [PubMed] [Google Scholar]
- Kesika, P. , Suganthy, N. , Sivamaruthi, B. S. , & Chaiyasut, C. (2021). Role of gut‐brain axis, gut microbial composition, and probiotic intervention in Alzheimer's disease. Life Sciences, 264, 118627. 10.1016/j.lfs.2020.118627 [DOI] [PubMed] [Google Scholar]
- Khoshbin, K. , & Camilleri, M. (2020). Effects of dietary components on intestinal permeability in health and disease. American Journal of Physiology. Gastrointestinal and Liver Physiology, 319, G589–G608. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Khurana, S. , Venkataraman, K. , Hollingsworth, A. , Piche, M. , & Tai, T. C. (2013). Polyphenols: Benefits to the cardiovascular system in health and in aging. Nutrients, 5, 3779–3827. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kikuchi, Y. , Matsuo, M. , Nabeta, C. , Akahoshi, A. , & Minami, H. (2019). Elemental diet regulates intestinal permeability and antibody production in indomethacin‐induced intestinal injury rats. Journal of Nutritional Science and Vitaminology, 65, 31–37. [DOI] [PubMed] [Google Scholar]
- Kim, A. S. , & Ko, H. J. (2018). Plasma concentrations of zonulin are elevated in obese men with fatty liver disease. Diabetes, Metabolic Syndrome and Obesity, 11, 149–157. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kim, C. H. (2018). Immune regulation by microbiome metabolites. Clinical & Translational Immunology, 154, 220–229. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kim, S. , Goel, R. , Kumar, A. , Qi, Y. , Lobaton, G. , Hosaka, K. , Mohammed, M. , Handberg, E. M. , Richards, E. M. , Pepine, C. J. , & Raizada, M. K. (2018). Imbalance of gut microbiome and intestinal epithelial barrier dysfunction in patients with high blood pressure. Clinical Science, 132, 701–718. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kim, Y. A. , Keogh, J. B. , & Clifton, P. M. (2018). Probiotics, prebiotics, synbiotics and insulin sensitivity. Nutrition Research Reviews, 31, 35–51. [DOI] [PubMed] [Google Scholar]
- Kim, E. K. , An, S. Y. , Lee, M. S. , Kim, T. H. , Lee, H. K. , Hwang, W. S. , Choe, S. J. , Kim, T. Y. , Han, S. J. , Kim, H. J. , Kim, D. J. , & Lee, K. W. (2011). Fermented kimchi reduces body weight and improves metabolic parameters in overweight and obese patients. Nutrition research (New York, N.Y.), 31, 436–443. 10.1016/j.nutres.2011.05.011 [DOI] [PubMed] [Google Scholar]
- Kinashi, Y. , & Hase, K. (2021). Partners in leaky gut syndrome: Intestinal dysbiosis and autoimmunity. Frontiers in Immunology, 12, 673708. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Klein, G. L. , Petschow, B. W. , Shaw, A. L. , & Weaver, E. (2013). Gut barrier dysfunction and microbial translocation in cancer cachexia: A new therapeutic target. Current Opinion in Supportive and Palliative Care, 7(4), 361–367. 10.1097/SPC.0000000000000017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Knezevic, J. , Starchl, C. , Berisha, A. T. , & Amrein, K. (1769). Thyroid‐gut‐axis: How does the microbiota influence thyroid function? Nutrients, 2020, 12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kong, H. H. , Oh, J. , Deming, C. , Conlan, S. , Grice, E. A. , Beatson, M. A. , Nomicos, E. , Polley, E. C. , Komarow, H. D. , NISC Comparative Sequence Program , Murray, P. R. , Turner, M. L. , & Segre, J. A. (2012). Temporal shifts in the skin microbiome associated with disease flares and treatment in children with atopic dermatitis. Genome Research, 22, 850–859. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kong, J. , Zhang, Z. , Musch, M. W. , Ning, G. , Sun, J. , Hart, J. , Bissonnette, M. , & Li, Y. C. (2008). Novel role of the vitamin D receptor in maintaining the integrity of the intestinal mucosal barrier. American Journal of Physiology. Gastrointestinal and Liver Physiology, 294, G208–G216. [DOI] [PubMed] [Google Scholar]
- Kosinski, C. , & Jornayvaz, F. R. (2017). Effects of Ketogenic diets on cardiovascular risk factors: Evidence from animal and human studies. Nutrients, 9, 517. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Koutzoumis, D. N. , Vergara, M. , Pino, J. , Buddendorff, J. , Khoshbouei, H. , Mandel, R. J. , & Torres, G. E. (2020). Alterations of the gut microbiota with antibiotics protects dopamine neuron loss and improve motor deficits in a pharmacological rodent model of Parkinson's disease. Experimental Neurology, 325, 113159. 10.1016/j.expneurol.2019.113159 [DOI] [PubMed] [Google Scholar]
- Krysiak, R. , Szkróbka, W. , & Okopień, B. (2019). The effect of gluten‐free diet on thyroid autoimmunity in drug‐naïve women with Hashimoto's Thyroiditis: A pilot study. Experimental and Clinical Endocrinology & Diabetes, 127, 417–422. [DOI] [PubMed] [Google Scholar]
- Kuwahara, A. , Matsuda, K. , Kuwahara, Y. , Asano, S. , Inui, T. , & Marunaka, Y. (2020). Microbiota‐gut‐brain axis: Enteroendocrine cells and the enteric nervous system form an interface between the microbiota and the central nervous system. Biomedical Research, 41, 199–216. [DOI] [PubMed] [Google Scholar]
- Kumar Singh, A. , Cabral, C. , Kumar, R. , Ganguly, R. , Kumar Rana, H. , Gupta, A. , Rosaria Lauro, M. , Carbone, C. , Reis, F. , & Pandey, A. K. (2019). Beneficial effects of dietary polyphenols on gut microbiota and strategies to improve delivery efficiency. Nutrients, 11(9), 2216. 10.3390/nu11092216 [DOI] [PMC free article] [PubMed] [Google Scholar]
- La Rosa, F. , Clerici, M. , Ratto, D. , Occhinegro, A. , Licito, A. , Romeo, M. , Iorio, C. D. , & Rossi, P. (2018). The gut‐brain axis in Alzheimer's disease and omega‐3. A critical overview of clinical trials. Nutrients, 10(9), 1267. 10.3390/nu10091267 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lacerda, J. F. , Lagos, A. C. , Carolino, E. , Silva‐Herdade, A. S. , Silva, M. , & Sousa Guerreiro, C. (2021). Functional food components, intestinal permeability and inflammatory markers in patients with inflammatory bowel disease. Nutrients, 13(2), 642. 10.3390/nu13020642 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lambert, J. , Mejia, S. , & Vojdani, A. (2018). Plant and human aquaporins: Pathogenesis from gut to brain. Immunologic Research, 67, 2–20. [DOI] [PubMed] [Google Scholar]
- Le Bastard, Q. , al‐Ghalith, G. A. , Grégoire, M. , Chapelet, G. , Javaudin, F. , Dailly, E. , Batard, E. , Knights, D. , & Montassier, E. (2018). Systematic review: Human gut dysbiosis induced by non‐antibiotic prescription medications. Alimentary Pharmacology & Therapeutics, 47, 332–345. [DOI] [PubMed] [Google Scholar]
- Lee, Y. M. , Yoon, Y. , Yoon, H. , Park, H. M. , Song, S. , & Yeum, K. J. (2017). Dietary Anthocyanins against obesity and inflammation. Nutrients, 9(10), 1089. 10.3390/nu9101089 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Leffler, D. A. , Kelly, C. P. , Green, P. H. , Fedorak, R. N. , DiMarino, A. , Perrow, W. , Rasmussen, H. , Wang, C. , Bercik, P. , Bachir, N. M. , & Murray, J. A. (2015). Larazotide acetate for persistent symptoms of celiac disease despite a gluten‐free diet: a randomized controlled trial. Gastroenterology, 148(7), 1311–1319 e6. 10.1053/j.gastro.2015.02.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Leong, X. F. , Mustafa, M. R. , Das, S. , & Jaarin, K. (2010). Association of elevated blood pressure and impaired vasorelaxation in experimental Sprague‐Dawley rats fed with heated vegetable oil. Lipids in Health and Disease, 9, 66. 10.1186/1476-511X-9-66 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lerner, A. , & Matthias, T. (2015). Changes in intestinal tight junction permeability associated with industrial food additives explain the rising incidence of autoimmune disease. Autoimmunity Reviews, 14, 479–489. [DOI] [PubMed] [Google Scholar]
- Lerner, A. , Shoenfeld, Y. , & Matthias, T. (2017). Adverse effects of gluten ingestion and advantages of gluten withdrawal in nonceliac autoimmune disease. Nutrition Reviews, 75(12), 1046–1058. [DOI] [PubMed] [Google Scholar]
- Li, D. , Yang, M. , Edwards, S. , & Ye, S. (2013). Nonalcoholic fatty liver disease: For better or worse, blame the gut microbiota? JPEN Journal of Parenteral and Enteral Nutrition, 37(6), 787–793. [DOI] [PubMed] [Google Scholar]
- Li, J. , Zhao, F. , Wang, Y. , Chen, J. , Tao, J. , Tian, G. , Wu, S. , Liu, W. , Cui, Q. , Geng, B. , Zhang, W. , Weldon, R. , Auguste, K. , Yang, L. , Liu, X. , Chen, L. , Yang, X. , Zhu, B. , & Cai, J. (2017). Gut microbiota dysbiosis contributes to the development of hypertension. Microbiome, 5(1), 14. 10.1186/s40168-016-0222-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li, X. , & Atkinson, M. A. (2015). The role for gut permeability in the pathogenesis of type 1 diabetes – A solid or leaky concept? Pediatric Diabetes, 16, 485–492. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li, X. , Schofield, B. H. , Huang, C. , Kleiner, G. I. , & Sampson, H. A. (1999). A murine model of IgE‐mediated cow's milk hypersensitivity. The Journal of Allergy and Clinical Immunology, 103(2), 206–214. [DOI] [PubMed] [Google Scholar]
- Li, Y. , Tian, X. , Li, S. , Chang, L. , Sun, P. , Lu, Y. , Yu, X. , Chen, S. , Wu, Z. , Xu, Z. , & Kang, W. (2019). Total polysaccharides of adlay bran (Coix lachryma‐jobi L.) improve TNF‐α induced epithelial barrier dysfunction in Caco‐2 cells via inhibition of the inflammatory response. Food & Function, 10(5), 2906–2913. 10.1039/c9fo00590k [DOI] [PubMed] [Google Scholar]
- Lin, B. W. , Gong, C. C. , Song, H. F. , & Cui, Y. Y. (2017). Effects of anthocyanins on the prevention and treatment of cancer. British Journal of Pharmacology, 174(11), 1226–1243. 10.1111/bph.13627 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ling, X. , Linglong, P. , Weixia, D. , & Hong, W. (2016). Protective effects of Bifidobacterium on intestinal barrier function in LPS‐induced enterocyte barrier injury of Caco‐2 monolayers and in a rat NEC model. PLoS One, 11(8), e0161635. 10.1371/journal.pone.0161635 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liśkiewicz, P. , Kaczmarczyk, M. , Misiak, B. , Wroński, M. , Bąba‐Kubiś, A. , Skonieczna‐Żydecka, K. , Marlicz, W. , Bieńkowski, P. , Misera, A. , Pełka‐Wysiecka, J. , Kucharska‐Mazur, J. , Konopka, A. , Łoniewski, I. , & Samochowiec, J. (2021). Analysis of gut microbiota and intestinal integrity markers of inpatients with major depressive disorder. Progress in Neuro‐Psychopharmacology & Biological Psychiatry, 106, 110076. 10.1016/j.pnpbp.2020.110076 [DOI] [PubMed] [Google Scholar]
- Liu, J. E. , Zhang, Y. , Zhang, J. , Dong, P. L. , Chen, M. , & Duan, Z. P. (2010). Probiotic yogurt effects on intestinal fora of patients with chronic liver disease. Nursing Research, 59, 426–432. [DOI] [PubMed] [Google Scholar]
- Liu, P. , Kerr, B. J. , Weber, T. E. , Chen, C. , Johnston, L. J. , & Shurson, G. C. (2014). Influence of thermally oxidized vegetable oils and animal fats on intestinal barrier function and immune variables in young pigs. Journal of Animal Science, 92, 2971–2979. [DOI] [PubMed] [Google Scholar]
- Liu, S. , Gao, J. , Zhu, M. , Liu, K. , & Zhang, H.‐L. (2020). Gut microbiota and dysbiosis in Alzheimer's disease: Implications for pathogenesis and treatment. Molecular Neurobiology, 57, 5026–5043. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Llewellyn, S. R. , Britton, G. J. , Contijoch, E. J. , Vennaro, O. H. , Mortha, A. , Colombel, J. F. , Grinspan, A. , Clemente, J. C. , Merad, M. , & Faith, J. J. (2018). Interactions between diet and the intestinal microbiota alter intestinal permeability and colitis severity in mice. Gastroenterology, 154(4), 1037–1046 e2. 10.1053/j.gastro.2017.11.030 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lopetuso, L. R. , Scaldaferri, F. , Bruno, G. , Petito, V. , Franceschi, F. , & Gasbarrini, A. (2015). The therapeutic management of gut barrier leaking: The emerging role for mucosal barrier protectors. European Review for Medical and Pharmacological Sciences, 19, 1068–1076. [PubMed] [Google Scholar]
- Lourida, I. , Soni, M. , Thompson‐Coon, J. , Purandare, N. , Lang, I. A. , Ukoumunne, O. C. , & Llewellyn, D. J. (2013). Mediterranean diet, cognitive function, and dementia A systematic review. Epidemiology, 24(4), 479–489. [DOI] [PubMed] [Google Scholar]
- Luettig, J. , Rosenthal, R. , Lee, I. M. , Krug, S. M. , & Schulzke, J. D. (2016). The ginger component 6‐shogaol prevents TNF‐α‐induced barrier loss via inhibition of PI3K/Akt and NF‐κB signaling. Molecular Nutrition & Food Research, 60(12), 2576–2586. 10.1002/mnfr.201600274 [DOI] [PubMed] [Google Scholar]
- Luissint, A. , Parkos, C. A. , & Nusrat, A. (2016). Inflammation and the intestinal barrier: Leukocyte–epithelial cell interactions, cell junction remodeling, and mucosal repair. Gastroenterology, 151, 616–632. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ma, Y. , Yin, Z. , Li, L. , Chen, B. , Dai, H. , Wu, D. , Cong, J. , Ye, L. , Liao, C. , Li, L. , Ye, Z. , & Huang, Z. (2021). Food antigens exacerbate intestinal damage and inflammation following the disruption of the mucosal barrier. International Immunopharmacology, 96, 107670. 10.1016/j.intimp.2021.107670 [DOI] [PubMed] [Google Scholar]
- Magnuson, B. A. , Carakostas, M. C. , Moore, N. H. , Poulos, S. P. , & Renwick, A. G. (2016). Biological fate of low‐calorie sweeteners. Nutrition Reviews, 74(11), 670–689. [DOI] [PubMed] [Google Scholar]
- Maguire, M. , & Maguire, G. (2017). The role of microbiota, and probiotics and prebiotics in skin health. Archives of Dermatological Research, 309, 411–421. [DOI] [PubMed] [Google Scholar]
- Malíčková, K. , Francová, I. , Lukáš, M. , Kolář, M. , Králíková, E. , Bortlík, M. , Ďuricová, D. , Štěpánková, L. , Zvolská, K. , Pánková, A. , & Zima, T. (2017). Fecal zonulin is elevated in Crohn's disease and in cigarette smokers. Practical Laboratory Medicine, 9, 39–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Manach, C. , Williamson, G. , Morand, C. , Scalbert, A. , & Rémésy, C. (2005). Bioavailability and bioefficacy of polyphenols in humans. I. Review of 97 bioavailability studies. The American Journal of Clinical Nutrition, 81(1 Suppl), 230S–242S. 10.1093/ajcn/81.1.230S [DOI] [PubMed] [Google Scholar]
- Manolescu, B. N. , Oprea, E. , Mititelu, M. , Ruta, L. L. , & Farcasanu, I. C. (2019). Dietary anthocyanins and stroke: A review of pharmacokinetic and pharmacodynamic studies. Nutrients, 11(7), 1479. 10.3390/nu11071479 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Manzel, A. , Muller, D. N. , Hafler, D. A. , Erdman, S. E. , Linker, R. A. , & Kleinewietfeld, M. (2014). Role of "Western diet" in inflammatory autoimmune diseases. Current Allergy and Asthma Reports, 14(1), 404. 10.1007/s11882-013-0404-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Marasco, G. , Cirota, G. G. , Rossini, B. , Lungaro, L. , Di Biase, A. R. , Colecchia, A. , Volta, U. , De Giorgio, R. , Festi, D. , & Caio, G. (2020). Probiotics, prebiotics and other dietary supplements for gut microbiota modulation in celiac disease patients. Nutrients, 12(9), 2674. 10.3390/nu12092674 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Marco, M. L. , Heeney, D. , Binda, S. , Cifelli, C. J. , Cotter, P. D. , Foligné, B. , Gänzle, M. , Kort, R. , Pasin, G. , Pihlanto, A. , Smid, E. J. , & Hutkins, R. (2017). Health benefits of fermented foods: microbiota and beyond. Current Opinion in Biotechnology, 44, 94–102. 10.1016/j.copbio.2016.11.010 [DOI] [PubMed] [Google Scholar]
- Martín, I. S. M. , Vilar, E. G. , Oliva, S. L. , & Rojo, S. S. (2022). Existing differences between available lists of FODMAP containing foods. Revista Española de Enfermedades Digestivas. 10.17235/reed.2022.8463/2021 [DOI] [PubMed] [Google Scholar]
- Maukonen, J. , & Saarela, M. (2015). Human gut microbiota: Does diet matter? The Proceedings of the Nutrition Society, 74, 23–36. [DOI] [PubMed] [Google Scholar]
- Mayer, E. A. , Padua, D. , & Tillisch, K. (2014). Altered brain‐gut axis in autism: Comorbidity or causative mechanisms? BioEssays, 36, 933–939. [DOI] [PubMed] [Google Scholar]
- Mazzawi, T. , Hausken, T. , Gundersen, D. , & el‐Salhy, M. (2013). Effects of dietary guidance on the symptoms, quality of life and habitual dietary intake of patients with irritable bowel syndrome. Molecular Medicine Reports, 8(3), 845–852. [DOI] [PubMed] [Google Scholar]
- McAllister, B. P. , Williams, E. , & Clarke, K. A. (2019). Comprehensive review of celiac disease/gluten‐sensitive enteropathies. Clinical Reviews in Allergy and Immunology, 57, 226–243. [DOI] [PubMed] [Google Scholar]
- Megur, A. , Baltriukienė, D. , Bukelskienė, V. , & Burokas, A. (2020). The microbiota‐gut‐brain axis and Alzheimer's disease: Neuroinflammation is to blame? Nutrients, 13(1), 37. 10.3390/nu13010037 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ménard, S. , Cerf‐Bensussan, N. , & Heyman, M. (2010). Multiple facets of intestinal permeability and epithelial handling of dietary antigens. Mucosal Immunology, 3(3), 247–259. [DOI] [PubMed] [Google Scholar]
- Menta, P. L. R. , Andrade, M. E. R. , Leocádio, P. C. L. , Fraga, J. R. , Dias, M. T. S. , Cara, D. C. , Cardoso, V. N. , Borges, L. F. , Capettini, L. S. A. , Aguilar, E. C. , & Alvarez‐Leite, J. I. (2019). Wheat gluten intake increases the severity of experimental colitis and bacterial translocation by weakening of the proteins of the junctional complex. The British Journal of Nutrition, 121, 361–373. [DOI] [PubMed] [Google Scholar]
- Merendino, N. , Costantini, L. , Manzi, L. , Molinari, R. , D'Eliseo, D. , & Velotti, F. (2013). Dietary omega‐3 polyunsaturated fatty acid DHA: A potential adjuvant in the treatment of cancer. BioMed Research International, 2013, 310186. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Michielan, A. , & D'Incà, R. (2015). Intestinal permeability in inflammatory bowel disease: Pathogenesis, clinical evaluation, and therapy of leaky gut. Mediators of Inflammation, 2015, 628157. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Miles, E. A. , & Calder, P. C. (2012). Influence of marine n‐3 polyunsaturated fatty acids on immune function and a systematic review of their effects on clinical outcomes in rheumatoid arthritis. The British Journal of Nutrition, 107, S171–S184. [DOI] [PubMed] [Google Scholar]
- Mitchell, C. M. , Davy, B. M. , Halliday, T. M. , Hulver, M. W. , Neilson, A. P. , Ponder, M. A. , & Davy, K. P. (2015). The effect of prebiotic supplementation with inulin on cardiometabolic health: Rationale, design, and methods of a controlled feeding efficacy trial in adults at risk of type 2 diabetes. Contemporary Clinical Trials, 45, 328–337. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mittal, R. , Debs, L. H. , Patel, A. P. , Nguyen, D. , Patel, K. , O'Connor, G. , Grati, M. , Mittal, J. , Yan, D. , Eshraghi, A. A. , Deo, S. K. , Daunert, S. , & Liu, X. Z. (2017). Neurotransmitters: The critical modulators regulating gut–brain axis. Journal of Cellular Physiology, 232, 2359–2372. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Miyake, Y. , & Yamamoto, K. (2013). Role of gut microbiota in liver diseases. Hepatology Research, 43, 139–146. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Moludi, J. , Maleki, V. , Jafari‐Vayghyan, H. , Vaghef‐Mehrabany, E. , & Alizadeh, M. (2020). Metabolic endotoxemia and cardiovascular disease: A systematic review about potential roles of prebiotics and probiotics. Clinical and Experimental Pharmacology & Physiology, 47, 927–939. [DOI] [PubMed] [Google Scholar]
- Monda, V. , Villano, I. , Messina, A. , Valenzano, A. , Esposito, T. , Moscatelli, F. , Viggiano, A. , Cibelli, G. , Chieffi, S. , Monda, M. , & Messina, G. (2017). Exercise modifies the gut microbiota with positive health effects. Oxidative Medicine and Cellular Longevity, 2017, 3831972. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Moreira, A. P. B. , Texeira, T. F. S. , Ferreira, A. B. , Peluzio Mdo, C. , & Alfenas Rde, C. (2012). Influence of a high‐fat diet on gut microbiota, intestinal permeability and metabolic endotoxaemia. The British Journal of Nutrition, 108, 801–809. [DOI] [PubMed] [Google Scholar]
- Moreno‐Navarrete, J. M. , Sabater, M. , Ortega, F. , Ricart, W. , & Fernández‐Real, J. M. (2012). Circulating zonulin, a marker of intestinal permeability, is increased in association with obesity‐associated insulin resistance. PLoS One, 7(5), e37160. 10.1371/journal.pone.0037160 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mu, Q. , Kirby, J. , Reilly, C. M. , & Luo, X. M. (2017). Leaky gut as a danger signal for autoimmune diseases. Frontiers in Immunology, 8, 598. 10.3389/fimmu.2017.00598 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mukherjee, S. , Karmakar, S. , & Babu, S. P. (2016). TLR2 and TLR4 mediated host immune responses in major infectious diseases: A review. The Brazilian Journal of Infectious Diseases, 20(2), 193–204. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nagatomo, Y. , & Tang, W. H. W. (2015). Intersections between microbiome and heart failure: Revisiting the gut hypothesis. Journal of Cardiac Failure, 21(12), 973–980. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nanayakkara, W. S. , Skidmore, P. M. L. , O'Brien, L. , Wilkinson, T. J. , & Gearry, R. B. (2016). Efficacy of the low FODMAP diet for treating irritable bowel syndrome: The evidence to date. Clinical and Experimental Gastroenterology, 9, 131–142. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Neish, A. S. (2009). Reviews in basic and clinical gastroenterology. Gastroenterology, 136, 65–80.19026645 [Google Scholar]
- Neunlist, M. , Toumi, F. , Oreschkova, T. , Denis, M. , Leborgne, J. , Laboisse, C. L. , Galmiche, J. P. , & Jarry, A. (2003). Human ENS regulates the intestinal epithelial barrier permeability and a tight junction‐associated protein ZO‐1 via VIPergic pathways. American Journal of Physiology. Gastrointestinal and Liver Physiology, 285, G1028–G1036. [DOI] [PubMed] [Google Scholar]
- Niesler, B. , Stefanie Kuerten, S. , Ekin Demir, E. , & Schäfer, K. (2021). Disorders of the enteric nervous system – A holistic view. Nature Reviews. Gastroenterology & Hepatology, 18, 393–410. [DOI] [PubMed] [Google Scholar]
- Noriega, B. S. , Sanchez‐Gonzalez, M. A. , Salyakina, D. , & Coffman, J. (2016). Understanding the impact of omega‐3 rich diet on the gut microbiota. Clinical Medicine. Case Reports, 2016, 3089303. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Norman, K. , Pirlich, M. , Schulzke, J. D. , Smoliner, C. , Lochs, H. , Valentini, L. , & Bühner, S. (2012). Increased intestinal permeability in malnourished patients with liver cirrhosis. European Journal of Clinical Nutrition, 66, 1116–1119. [DOI] [PubMed] [Google Scholar]
- Nouri, M. , Bredberg, A. , Weström, B. , & Lavasani, S. (2014). Intestinal barrier dysfunction develops at the onset of experimental autoimmune encephalomyelitis, and can be induced by adoptive transfer of auto‐reactive T cells. PLoS One, 9(9), e106335. 10.1371/journal.pone.0106335 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nunes, C. , Freitas, V. , Almeida, L. , & Laranjinha, J. (2019). Red wine extract preserves tight junctions in intestinal epithelial cells under inflammatory conditions: implications for intestinal inflammation. Food & Function, 10(3), 1364–1374. 10.1039/c8fo02469c [DOI] [PubMed] [Google Scholar]
- O'Donovan, S. M. , Crowley, E. K. , Brown, J. R. , O'Sullivan, O. , O'Leary, O. F. , Timmons, S. , Nolan, Y. M. , Clarke, D. J. , Hyland, N. P. , Joyce, S. A. , Sullivan, A. M. , & O'Neill, C. (2020). Nigral overexpression of α‐synuclein in a rat Parkinson's disease model indicates alterations in the enteric nervous system and the gut microbiome. Neurogastroenterology and Motility, 32(1), e13726. 10.1111/nmo.13726 [DOI] [PubMed] [Google Scholar]
- Obrenovich, M. , Jaworski, H. , Tadimalla, T. , Mistry, A. , Sykes, L. , Perry, G. , & Bonomo, R. A. (2020). The role of the microbiota‐gut‐brain axis and antibiotics in ALS and neurodegenerative diseases. Microorganisms, 8(5), 784. 10.3390/microorganisms8050784 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Obrenovich, M. E. M. (2018). Leaky gut, leaky brain? Microorganisms, 6(107), 6040107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ochoa‐Repáraz, J. , Kirby, T. O. , & Kasper, L. H. (2018). The gut microbiome and multiple sclerosis. Cold Spring Harbor Perspectives in Medicine, 8, a029017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Odamaki, T. , Kato, K. , Sugahara, H. , Hashikura, N. , Takahashi, S. , Xiao, J. Z. , Abe, F. , & Osawa, R. (2016). Age‐related changes in gut microbiota composition from newborn to centenarian: a cross‐sectional study. BMC Microbiology, 16, 90. 10.1186/s12866-016-0708-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Odenwald, M. A. , & Turner, J. R. (2013). Intestinal permeability defects: Is it time to treat? Clinical Gastroenterology and Hepatology, 11, 1075–1083. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Odenwald, M. A. , & Turner, J. R. (2017). The intestinal epithelial barrier: A therapeutic target? Nature Reviews. Gastroenterology & Hepatology, 14(1), 9–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ohlsson, B. , Orho‐Melander, M. , & Nilsson, P. (2017). Higher levels of serum zonulin may rather be associated with increased risk of obesity and hyperlipidemia, than with gastrointestinal symptoms or disease manifestations. International Journal of Molecular Sciences, 18(3), 582. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Okuyama, H. , Langsjoen, P. H. , Ohara, N. , Hashimoto, Y. , Hamazaki, T. , Yoshida, S. , Kobayashi, T. , & Langsjoen, A. M. (2016). Medicines and vegetable oils as hidden causes of cardiovascular disease and diabetes. Pharmacology, 98, 134–170. [DOI] [PubMed] [Google Scholar]
- Olson, C. A. , Vuong, H. E. , Yano, J. M. , Liang, Q. Y. , Nusbaum, D. J. , & Hsiao, E. Y. (2018). The gut microbiota mediates the anti‐seizure effects of the ketogenic diet. Cell, 173(7), 1728–1741 e13. 10.1016/j.cell.2018.04.027 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ondrussek‐Sekac, M. , Navas‐Carrillo, D. , & Orenes‐Piñero, E. (2021). Intestinal microbiota alterations in chronic kidney disease and the influence of dietary components. Critical Reviews in Food Science and Nutrition, 61(9), 1490–1502. [DOI] [PubMed] [Google Scholar]
- Opal, S. M. , & DePalo, V. A. (2000). Anti‐inflammatory cytokines. Chest, 117, 1162–1172. [DOI] [PubMed] [Google Scholar]
- Osadchiy, V. , Martin, C. R. , & Mayer, E. A. (2019). The gut‐ brain axis and the microbiome: Mechanisms and clinical implications. Clinical Gastroenterology and Hepatology, 17, 322–332. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ostgaard, H. , Hausken, T. , Gundersen, D. , & El‐Salhy, M. (2012). Diet and effects of diet management on quality of life and symptoms in patients with irritable bowel syndrome. Molecular Medicine Reports, 5(6), 1382–1390. [DOI] [PubMed] [Google Scholar]
- Ouellette, A. J. (2010). Paneth cells and innate mucosal immunity. Current Opinion in Gastroenterology, 26, 547–553. [DOI] [PubMed] [Google Scholar]
- Özyurt, G. , Öztürk, Y. , Appak, Y. Ç. , Arslan, F. D. , Baran, M. , Karakoyun, İ. , Tufan, A. E. , & Pekcanlar, A. A. (2018). Increased zonulin is associated with hyperactivity and social dysfunctions in children with attention deficit hyperactivity disorder. Comprehensive Psychiatry, 87, 138–142. [DOI] [PubMed] [Google Scholar]
- Pabst, O. (2012). New concepts in the generation and functions of IgA. Nature Reviews. Immunology, 12, 821–832. [DOI] [PubMed] [Google Scholar]
- Painold, A. , Mörkl, S. , Kashofer, K. , Halwachs, B. , Dalkner, N. , Bengesser, S. , Birner, A. , Fellendorf, F. , Platzer, M. , Queissner, R. , Schütze, G. , Schwarz, M. J. , Moll, N. , Holzer, P. , Holl, A. K. , Kapfhammer, H. P. , Gorkiewicz, G. , & Reininghaus, E. Z. (2019). A step ahead: Exploring the gut microbiota in inpatients with bipolar disorder during a depressive episode. Bipolar Disorders, 21, 40–49. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Parkar, S. G. , Stevenson, D. E. , & Skinner, M. A. (2008). The potential influence of fruit polyphenols on colonic microflora and human gut health. International Journal of Food Microbiology, 124, 295–298. [DOI] [PubMed] [Google Scholar]
- Parohan, M. , Djalali, M. , Sarraf, P. , Yaghoubi, S. , Seraj, A. , Foroushani, A. R. , Ranji‐Burachaloo, S. , & Javanbakht, M. H. (2020). Effect of probiotic supplementation on migraine prophylaxis: A systematic review and meta‐analysis of randomized controlled trials. Nutritional Neuroscience, 25, 511–518. 10.1080/1028415X.2020.1764292 [DOI] [PubMed] [Google Scholar]
- Partridge, D. , Lloyd, K. A. , Rhodes, J. M. , Walker, A. W. , Johnstone, A. M. , & Campbell, B. J. (2019). Food additives: Assessing the impact of exposure to permitted emulsifiers on bowel and metabolic health – introducing the FADiets study. Nutrition Bulletin, 44, 329–349. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pärtty, A. , Kalliomäki, M. , Wacklin, P. , Salminen, S. , & Isolauri, E. (2015). A possible link between early probiotic intervention and the risk of neuropsychiatric disorders later in childhood: A randomized trial. Pediatric Research, 77(6), 823–828. [DOI] [PubMed] [Google Scholar]
- Patra, J. K. , Das, G. , Paramithiotis, S. , & Shin, H. (2016). Kimchi and other widely consumed traditional fermented foods of Korea: A review. Frontiers in Microbiology, 7, 1493. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Patterson, E. , Cryan, J. F. , Fitzgerald, G. F. , Ross, R. P. , Dinan, T. G. , & Stanton, C. (2014). Gut microbiota, the pharmabiotics they produce and host health. The Proceedings of the Nutrition Society, 73, 477–489. [DOI] [PubMed] [Google Scholar]
- Pellizoni, F. P. , Leite, A. Z. , Rodrigues, N. C. , Ubaiz, M. J. , Gonzaga, M. I. , Takaoka, N. N. C. , Mariano, V. S. , Omori, W. P. , Pinheiro, D. G. , Matheucci Junior, E. , Gomes, E. , & de Oliveira, G. L. V. (2021). Detection of dysbiosis and increased intestinal permeability in Brazilian patients with relapsing‐remitting multiple sclerosis. International Journal of Environmental Research and Public Health, 18(9), 4621. 10.3390/ijerph18094621 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Peron, G. , Hidalgo‐Liberona, N. , González‐Domínguez, R. , Garcia‐Aloy, M. , Guglielmetti, S. , Bernardi, S. , Kirkup, B. , Kroon, P. A. , Cherubini, A. , Riso, P. , & Andrés‐Lacueva, C. (2020). Exploring the molecular pathways behind the effects of nutrients and dietary polyphenols on gut microbiota and intestinal permeability: A perspective on the potential of metabolomics and future clinical applications. Journal of Agricultural and Food Chemistry, 68, 1780–1789. [DOI] [PubMed] [Google Scholar]
- Petersa, A. , & Wekerle, H. (2019). Autoimmune diabetes mellitus and the leaky gut. Proceedings of the National Academy of Sciences, 116(30), 14788–14790. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pike, M. G. , Heddle, R. J. , Boulton, P. , Turner, M. W. , & Atherton, D. J. (1986). Increased intestinal permeability in atopic eczema. The Journal of Investigative Dermatology, 86(2), 101–104. 10.1111/1523-1747.ep12284035 [DOI] [PubMed] [Google Scholar]
- Plaza‐Díaz, J. , Solís‐Urra, P. , Rodríguez‐Rodríguez, F. , Olivares‐Arancibia, J. , Navarro‐Oliveros, M. , Abadía‐Molina, F. , & Álvarez‐Mercado, A. I. (2020). The gut barrier, intestinal microbiota, and liver disease: Molecular mechanisms and strategies to manage. International Journal of Molecular Sciences, 21(21), 8351. 10.3390/ijms21218351 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Polak, K. , Bergler‐Czop, B. , Szczepanek, M. , Wojciechowska, K. , Frątczak, A. , & Kiss, N. (2021). Psoriasis and gut microbiome‐current state of art. International Journal of Molecular Sciences, 22(9), 4529. 10.3390/ijms22094529 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Preveden, T. , Scarpellini, E. , Milić, N. , Luzza, F. , & Abenavoli, L. (2017). Gut microbiota changes and chronic hepatitis C virus infection. Expert Review of Gastroenterology & Hepatology, 11(9), 813–819. 10.1080/17474124.2017.1343663 [DOI] [PubMed] [Google Scholar]
- Proctor, C. , Thiennimitr, P. , Chattipakorn, N. , & Chattipakorn, S. C. (2017). Diet, gut microbiota and cognition. Metabolic Brain Disease, 32(1), 1–17. [DOI] [PubMed] [Google Scholar]
- Psaltopoulou, T. , Sergentanis, T. N. , Panagiotakos, D. B. , Sergentanis, I. N. , Kosti, R. , & Scarmeas, N. (2013). Mediterranean diet, stroke, cognitive impairment, and depression: A meta‐analysis. Annals of Neurology, 74, 580–591. [DOI] [PubMed] [Google Scholar]
- Pusztai, A. , Ewen, S. W. , Grant, G. , Brown, D. S. , Stewart, J. C. , Peumans, W. J. , van Damme, E. , & Bardocz, S. (1993). Antinutritive effects of wheat‐germ agglutinin and other N‐acetylglucosamine‐specific lectins. The British Journal of Nutrition, 70, 313–321. [DOI] [PubMed] [Google Scholar]
- Putt, K. K. , Pei, R. , White, H. M. , & Bolling, B. W. (2017). Yogurt inhibits intestinal barrier dysfunction in Caco‐2 cells by increasing tight junctions. Food & Function, 8, 406. [DOI] [PubMed] [Google Scholar]
- Rahman, M. T. , Ghosh, C. , Hossain, M. , Linfield, D. , Rezaee, F. , Janigro, D. , Marchi, N. , & van Boxel‐Dezaire, A. (2018). IFN‐g, IL‐17A, or Zonulin rapidly increase the permeability of the blood‐brain and small intestinal epithelial barriers: Relevance for neuro‐inflammatory diseases. Biochemical and Biophysical Research Communications, 507, 274–279. [DOI] [PubMed] [Google Scholar]
- Rallabhandi, P. (2012). Gluten and celiac disease‐An immunological perspective. Journal of AOAC International, 95, 349–355. [DOI] [PubMed] [Google Scholar]
- Ramani, T. , Auletta, C. S. , Weinstock, D. , Mounho‐Zamora, B. , Ryan, P. C. , Salcedo, T. W. , & Bannish, G. (2015). Cytokines: The good, the bad, and the deadly. International Journal of Toxicology, 34(4), 355–365. 10.1177/1091581815584918 [DOI] [PubMed] [Google Scholar]
- Rath, H. C. , Wilson, K. H. , & Sartor, R. B. (1999). Differential induction of colitis and gastritis in HLA‐B27 transgenic rats selectively colonized with Bacteroides vulgatus or Escherichia coli . Infection and Immunity, 67, 2969–2974. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Raza, M. H. , Gul, K. , Arshad, A. , Riaz, N. , Waheed, U. , Rauf, A. , Aldakheel, F. , Alduraywish, S. , Rehman, M. U. , Abdullah, M. , & Arshad, M. (2019). Microbiota in cancer development and treatment. Journal of Cancer Research and Clinical Oncology, 145, 49–63. [DOI] [PubMed] [Google Scholar]
- Rea, K. , Dinana, T. G. , & Cryan, J. F. (2020). Gut microbiota: A perspective for psychiatrists. Neuropsychobiology, 79, 50–62. [DOI] [PubMed] [Google Scholar]
- Rescigno, M. (2011). The intestinal epithelial barrier in the control of homeostasis and immunity. Trends in Immunology, 32, 256–264. [DOI] [PubMed] [Google Scholar]
- Reynders, T. , Devolder, L. , Valles‐Colomer, M. , van Remoortel, A. , Joossens, M. , de Keyser, J. , Nagels, G. , D'hooghe, M. , & Raes, J. (2020). Gut microbiome variation is associated to multiple sclerosis phenotypic subtypes. Annals of Clinical Translational Neurology, 7, 406–419. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Riccio, P. , & Rossano, R. (2015). Nutrition facts in multiple sclerosis. ASN Neuro, 7(1), 25694551. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rico‐Campà, A. , Martínez‐González, M. A. , Alvarez‐Alvarez, I. , Mendonça, R. D. , de la Fuente‐Arrillaga, C. , Gómez‐Donoso, C. , & Bes‐Rastrollo, M. (2019). Association between consumption of ultra‐processed foods and all cause mortality: SUN prospective cohort study. BMJ (Clinical research ed.), 365, l1949. 10.1136/bmj.l1949 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rieder, R. , Wisniewski, P. J. , Alderman, B. L. , & Campbell, S. C. (2017). Microbes and mental health: A review. Brain, Behavior, and Immunity, 66, 9–17. [DOI] [PubMed] [Google Scholar]
- Rinninella, E. , Cintoni, M. , Raoul, P. , Lopetuso, L. R. , Scaldaferri, F. , Pulcini, G. , Miggiano, G. A. D. , Gasbarrini, A. , & Mele, M. C. (2019). Food components and dietary habits: Keys for a healthy gut microbiota composition. Nutrients, 11(10), 2393. 10.3390/nu11102393 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rinninella, E. , Mele, M. C. , Merendino, N. , Cintoni, M. , Anselmi, G. , Caporossi, A. , Gasbarrini, A. , & Minnella, A. M. (2018). The role of diet, micronutrients and the gut microbiota in age‐related macular degeneration: new perspectives from the gut–retina axis. Nutrients, 10(11), 1677. 10.3390/nu10111677 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rinninella, E. , Raoul, P. , Cintoni, M. , Franceschi, F. , Miggiano, G. A. D. , Gasbarrini, A. , & Mele, M. C. (2019). What is the healthy gut microbiota composition? A changing ecosystem across age, environment, diet, and diseases. Microorganisms, 7, 14. 10.3390/microorganisms7010014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Robertson, R. C. , Seira Oriach, C. , Murphy, K. , Moloney, G. M. , Cryan, J. F. , Dinan, T. G. , Paul Ross, R. , & Stanton, C. (2017). Omega‐3 polyunsaturated fatty acids critically regulate behaviour and gut microbiota development in adolescence and adulthood. Brain, Behavior, and Immunity, 59, 21–37. [DOI] [PubMed] [Google Scholar]
- Rohr, M. W. , Narasimhulu, C. A. , Rudeski‐Rohr, T. A. , & Parthasarathy, S. (2020). Negative effects of a high‐fat diet on intestinal permeability: A review. Advances in Nutrition, 11, 77–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Roos, N. M. , van Hemert, S. , Rovers, J. M. P. , Smits, M. G. , & Witteman, B. J. M. (2017). The effects of a multispecies probiotic on migraine and markers of intestinal permeability–results of a randomized placebo‐controlled study. European Journal of Clinical Nutrition, 71, 1455–1462. [DOI] [PubMed] [Google Scholar]
- Roth, S. , Spalinger, M. R. , Gottier, C. , Biedermann, L. , Zeitz, J. , Lang, S. , Weber, A. , Rogler, G. , & Scharl, M. (2016). Bilberry‐derived anthocyanins modulate cytokine expression in the intestine of patients with ulcerative colitis. PLoS One, 11(5), e0154817. 10.1371/journal.pone.0154817 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rudzki, L. , & Maes, M. (2020). The microbiota‐gut‐immune‐glia (MGIG) axis in major depression. Molecular Neurobiology, 57, 4269–4295. [DOI] [PubMed] [Google Scholar]
- Rutsch, A. , Kantsjö, J. B. , & Ronchi, F. (2020). The gut‐brain axis: How microbiota and host inflammasome influence brain physiology and pathology. Frontiers in Immunology, 11, 604179. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sabatino, A. , Regolisti, G. , Cosola, C. , Gesualdo, L. , & Fiaccadori, E. (2017). Intestinal microbiota in type 2 diabetes and chronic kidney disease. Current Diabetes Reports, 17(3), 16. 10.1007/s11892-017-0841-z [DOI] [PubMed] [Google Scholar]
- Safari, Z. , & Gérard, P. (2019). The links between the gut microbiome and non‐alcoholic fatty liver disease (NAFLD). Cellular and Molecular Life Sciences, 76, 1541–1558. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sahagún, A. M. , Vaquera, J. , García, J. J. , Calle, Á. P. , Diez, M. J. , Fernández, N. , Loro, J. F. , Portilla, H. O. , & Sierra, M. (2015). Study of the protective effect on intestinal mucosa of the hydrosoluble fiber Plantago ovata husk. BMC Complementary and Alternative Medicine, 15, 298. 10.1186/s12906-015-0827-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Saito, A. C. , Higashi, T. , Fukazawa, Y. , Otani, T. , Tauchi, M. , Higashi, A. Y. , Furuse, M. , & Chiba, H. (2021). Occludin and tricellulin facilitate formation of anastomosing tight‐junction strand network to improve barrier function. Molecular Biology of the Cell, 32(8), 722–738. 10.1091/mbc.E20-07-0464 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Saji, N. , Murotani, K. , Hisada, T. , Kunihiro, T. , Tsuduki, T. , Sugimoto, T. , Kimura, A. , Niida, S. , Toba, K. , & Sakurai, T. (2020). Relationship between dementia and gut microbiome‐associated metabolites: a cross‐sectional study in Japan. Scientific Reports, 10(1), 8088. 10.1038/s41598-020-65196-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Santisteban, M. M. , Qi, Y. , Zubcevic, J. , Kim, S. , Yang, T. , Shenoy, V. , Cole‐Jeffrey, C. T. , Lobaton, G. O. , Stewart, D. C. , Rubiano, A. , Simmons, C. S. , Garcia‐Pereira, F. , Johnson, R. D. , Pepine, C. J. , & Raizada, M. K. (2017). Hypertension‐linked pathophysiological alterations in the gut. Circulation Research, 120, 312–323. 10.1161/CIRCRESAHA.116.309006 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sapone, A. , de Magistris, L. , Pietzak, M. , Clemente, M. G. , Tripathi, A. , Cucca, F. , Lampis, R. , Kryszak, D. , Cartenì, M. , Generoso, M. , Iafusco, D. , Prisco, F. , Laghi, F. , Riegler, G. , Carratu, R. , Counts, D. , & Fasano, A. (2006). Zonulin upregulation is associated with increased gut permeability in subjects with type 1 diabetes and their relatives. Diabetes, 55(5), 1443–1449. [DOI] [PubMed] [Google Scholar]
- Sasson, A. N. , Ananthakrishnan, A. N. , & Raman, M. (2021). Diet in treatment of inflammatory bowel diseases. Clinical Gastroenterology and Hepatology, 19, 425–435. [DOI] [PubMed] [Google Scholar]
- Sayon‐Orea, C. , Carlos, S. , & Martı´nez‐Gonzalez, M. A. (2015). Does cooking with vegetable oils increase the risk of chronic diseases?: A systematic review. The British Journal of Nutrition, 113, S36–S48. [DOI] [PubMed] [Google Scholar]
- Scalbert, A. , Manach, C. , Morand, C. , Rémésy, C. , & Jiménez, L. (2005). Dietary polyphenols and the prevention of diseases. Critical Reviews in Food Science and Nutrition, 45, 287–306. [DOI] [PubMed] [Google Scholar]
- Scaldaferri, F. , Gerardi, V. , Lopetuso, L. R. , Del Zompo, F. , Mangiola, F. , Boškoski, I. , Bruno, G. , Petito, V. , Laterza, L. , Cammarota, G. , Gaetani, E. , Sgambato, A. , & Gasbarrini, A. (2013). Gut microbial flora, prebiotics, and probiotics in IBD: their current usage and utility. BioMed Research International, 2013, 435268. 10.1155/2013/435268 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schenk, M. , & Mueller, C. (2008). The mucosal immune system at the gastrointestinal barrier. Best Practice & Research. Clinical Gastroenterology, 22, 391–409. [DOI] [PubMed] [Google Scholar]
- Scher, J. U. , Ubeda, C. , Artacho, A. , Attur, M. , Isaac, S. , Reddy, S. M. , Marmon, S. , Neimann, A. , Brusca, S. , Patel, T. , Manasson, J. , Pamer, E. G. , Littman, D. R. , & Abramson, S. B. (2015). Decreased bacterial diversity characterizes the altered gut microbiota in patients with psoriatic arthritis, resembling dysbiosis in inflammatory bowel disease. Arthritis & Rheumatology (Hoboken, N.J.), 67(1), 128–139. 10.1002/art.38892 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schleicher, M. , Weikel, K. , Garber, C. , & Taylor, A. (2013). Diminishing risk for age‐related macular degeneration with nutrition: A current view. Nutrients, 5, 2405–2456. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Scott, K. P. , Gratz, S. W. , Sheridan, P. O. , Flint, H. J. , & Duncan, S. H. (2013). The influence of diet on the gut microbiota. Pharmacological Research, 69, 52–60. [DOI] [PubMed] [Google Scholar]
- Seguella, L. , Capuano, R. , Sarnelli, G. , & Esposito, G. (2019). Play in advance against neurodegeneration: Exploring enteric glial cells in gut‐brain axis during neurodegenerative diseases. Expert Review of Clinical Pharmacology, 12, 555–564. [DOI] [PubMed] [Google Scholar]
- Selvaraju, V. , Joshi, M. , Suresh, S. , Sanchez, J. A. , Maulik, N. , & Maulik, G. (2012). Diabetes, oxidative stress, molecular mechanism, and cardiovascular disease—An overview. Toxicology Mechanisms and Methods, 22, 330–335. [DOI] [PubMed] [Google Scholar]
- Seo, K. , Seo, J. , Yeun, J. , Choi, H. , Kim, Y. I. , & Chang, S. Y. (2021). The role of mucosal barriers in human gut health. Archives of Pharmacal Research, 44, 325–341. [DOI] [PubMed] [Google Scholar]
- Septembre‐Malaterre, A. , Remize, F. , & Poucheret, P. (2018). Fruits and vegetables, as a source of nutritional compounds and phytochemicals: Changes in bioactive compounds during lactic fermentation. Food Research International, 104, 86–99. [DOI] [PubMed] [Google Scholar]
- Severance, E. G. , Yolken, R. H. , & Eaton, W. W. (2016). Autoimmune diseases, gastrointestinal disorders and the microbiome in schizophrenia: More than a gut feeling. Schizophrenia Research, 176, 23–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shabbir, U. , Arshad, M. S. , Sameen, A. , & Oh, D. H. (2021). Crosstalk between gut and brain in Alzheimer's disease: The role of gut microbiota modulation strategies. Nutrients, 13(2), 690. 10.3390/nu13020690 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shahi, S. K. , Freedman, S. N. , & Mangalam, A. K. (2017). Gut microbiome in multiple sclerosis: The players involved and the roles they play. Gut Microbes, 8, 607–615. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sharma, B. C. , & Singh, J. (2016). Probiotics in management of hepatic encephalopathy. Metabolic Brain Disease, 31, 1295–1301. [DOI] [PubMed] [Google Scholar]
- Shi, C. , Li, H. , Qu, X. , Huang, L. , Kong, C. , Qin, H. , Sun, Z. , & Yan, X. (2019). High fat diet exacerbates intestinal barrier dysfunction and changes gut microbiota in intestinal‐specific ACF7 knockout mice. Biomedicine & Pharmacotherapy, 110, 537–545. [DOI] [PubMed] [Google Scholar]
- Shi, Z. , Fang, Z. , Gao, X.‐X. , Yu, H. , Zhu, Y. W. , Ouyang, H. L. , Song, Y. X. , du, X. L. , Wang, Z. , Li, X. W. , & Liu, G. W. (2021). Nuciferine improves high‐fat diet‐induced obesity via reducing intestinal permeability by increasing autophagy and remodeling the gut microbiota. Food & Function, 12, 5850–5861. [DOI] [PubMed] [Google Scholar]
- Shi, Z. , Wu, X. , Yu, S. , Huynh, M. , Jena, P. K. , Nguyen, M. , Wan, Y. Y. , & Hwang, S. T. (2020). Short‐term exposure to a western diet induces psoriasiform dermatitis by promoting accumulation of IL‐17A‐Producing γδ T Cells. Journal of Investigative Dermatology, 140, 1815–1823. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shin, D. W. , & Lim, B. O. (2020). Nutritional interventions using functional foods and nutraceuticals to improve inflammatory bowel disease. Journal of Medicinal Food, 23(11), 1136–1145. [DOI] [PubMed] [Google Scholar]
- Shin, G. H. , Kang, B. , & Jang, D. J. (2016). Metabolic pathways associated with kimchi, a traditional Korean food, based on in silico modeling of published data. Genomics and Informatics, 14(4), 222–229. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sicherer, S. H. , & Sampson, H. A. (1999). Food hypersensitivity and atopic dermatitis: Pathophysiology, epidemiology, diagnosis, and management. The Journal of Allergy and Clinical Immunology, 104, S114–S122. [DOI] [PubMed] [Google Scholar]
- Silva, R. B. , Rodrigues, E. , Coelho, B. S. , Andrade, K. , Fonseca, L. , Fernandes‐Braga, W. , Ferreira, A. , Shivappa, N. , Hébert, J. R. , Silvestre, S. C. , Fasano, A. , Freire, R. H. , & Alvarez‐Leite, J. I. (2020). Inconsistent effects of gluten on obesity: Is there a role for the haptoglobin isoforms? Clinical Nutrition ESPEN, 40, 269–276. [DOI] [PubMed] [Google Scholar]
- Silva, S. , Costa, E. M. , Mendes, M. , Morais, R. M. , Calhau, C. , & Pintado, M. M. (2016). Antimicrobial, antiadhesive and antibiofilm activity of an ethanolic, anthocyanin‐rich blueberry extract purified by solid phase extraction. Journal of Applied Microbiology, 121(3), 693–703. 10.1111/jam.13215 [DOI] [PubMed] [Google Scholar]
- Simeonova, D. , Stoyanov, D. , Leunis, J. , Carvalho, A. F. , Kubera, M. , Murdjeva, M. , & Maes, M. (2020). Increased serum immunoglobulin responses to gut commensal gram‐negative bacteria in unipolar major depression and bipolar disorder type 1, especially when melancholia is present. Neurotoxicity Research, 37, 338–348. [DOI] [PubMed] [Google Scholar]
- Simopoulos, A. P. (2002). Omega‐3 fatty acids in inflammation and autoimmune diseases. Journal of the American College of Nutrition, 21, 495–505. [DOI] [PubMed] [Google Scholar]
- Simopoulos, A. P. (2008). The importance of the omega‐6/omega‐3 fatty acid ratio in cardiovascular disease and other chronic diseases. Experimental Biology and Medicine, 233, 674–688. [DOI] [PubMed] [Google Scholar]
- Simpson, M. , Mojibian, M. , Barriga, K. , Scott, F. W. , Fasano, A. , Rewers, M. , & Norris, J. M. (2009). An exploration of Glo‐3A antibody levels in children at increased risk for type 1 diabetes mellitus. Pediatric Diabetes, 10(8), 563–572. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Singh, R. K. , Chang, H. W. , Yan, D. , Lee, K. M. , Ucmak, D. , Wong, K. , Abrouk, M. , Farahnik, B. , Nakamura, M. , Zhu, T. H. , Bhutani, T. , & Liao, W. (2017). Influence of diet on the gut microbiome and implications for human health. Journal of Translational Medicine, 15(1), 73. 10.1186/s12967-017-1175-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- Siti, H. N. , Kamisah, Y. , Mohamed, S. , & Jaarin, K. (2019). Effects of citrus leaf extract on aortic vascular reactivity in hypertensive rats fed repeatedly heated vegetable oil. Applied physiology, Nutrition, and Metabolism, 44(4), 373–380. 10.1139/apnm-2018-0175 [DOI] [PubMed] [Google Scholar]
- Skardelly, M. , Armbruster, F. P. , Meixensberger, J. , & Hilbig, H. (2009). Expression of zonulin, c‐kit, and glial fibrillary acidic protein in human gliomas. Translational Oncology, 2(3), 117–120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Slyepchenko, A. , Maes, M. , Jacka, F. N. , Köhler, C. A. , Barichello, T. , McIntyre, R. , Berk, M. , Grande, I. , Foster, J. A. , Vieta, E. , & Carvalho, A. F. (2017). Gut Microbiota, bacterial translocation, and interactions with diet: Pathophysiological links between major depressive disorder and non‐communicable medical comorbidities. Psychotherapy and Psychosomatics, 86, 31–46. [DOI] [PubMed] [Google Scholar]
- Snelson, M. , Tan, S. M. , Clarke, R. E. , de Pasquale, C. , Thallas‐Bonke, V. , Nguyen, T.‐V. , Penfold, S. A. , Harcourt, B. E. , Sourris, K. C. , Lindblom, R. S. , Ziemann, M. , Steer, D. , El‐Osta, A. , Davies, M. J. , Donnellan, L. , Deo, P. , Kellow, N. J. , Cooper, M. E. , Woodruff, T. M. , … Coughlan, M. T. (2021). Processed foods drive intestinal barrier permeability and microvascular diseases. Science Advances, 7(14), eabe4841. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sochocka, M. , Donskow‐Łysoniewska, K. , Diniz, B. S. , Kurpas, D. , Brzozowska, E. , & Leszek, J. (2019). The gut microbiome alterations and inflammation‐driven pathogenesis of Alzheimer's disease—A critical review. Molecular Neurobiology, 56, 1841–1851. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sodhi, A. , & Kesherwani, V. (2007). Production of TNF‐alpha, IL‐1beta, IL‐12 and IFN‐gamma in murine peritoneal macrophages on treatment with wheat germ agglutinin in vitro: Involvement of tyrosine kinase pathways. Glycoconjugate Journal, 24, 573–582. [DOI] [PubMed] [Google Scholar]
- Sofi, F. , Cesari, F. , Abbate, R. , Gensini, G. F. , & Casini, A. (2008). Adherence to Mediterranean diet and health status: meta‐analysis. BMJ (Clinical research ed.), 337, a1344. 10.1136/bmj.a1344 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Soldati, L. , Di Renzo, L. , Jirillo, E. , Ascierto, P. A. , Marincola, F. M. , & De Lorenzo, A. (2018). The influence of diet on anti‐cancer immune responsiveness. Journal of Translational Medicine, 16(1), 75. 10.1186/s12967-018-1448-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Solis, C. J. , Hamilton, M. K. , Caruffo, M. , Garcia‐Lopez, J. P. , Navarrete, P. , Guillemin, K. , & Feijoo, C. G. (2020). Intestinal inflammation induced by soybean meal ingestion increases intestinal permeability and neutrophil turnover independently of microbiota in zebrafish. Frontiers in Immunology, 11, 1330. 10.3389/fimmu.2020.01330 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Solomon, A. , Dursun, D. , Liu, Z. , Xie, Y. , Macri, A. , & Pflugfelder, S. C. (2001). Pro‐ and anti‐inflammatory forms of interleukin‐1 in the tear fluid and conjunctiva of patients with dry‐eye disease. Investigative Ophthalmology & Visual Science, 42, 2283–2292. [PubMed] [Google Scholar]
- Spencer, M. , Gupta, A. , Dam, L. V. , Shannon, C. , Menees, S. , & Chey, W. D. (2016). Artificial sweeteners: A systematic review and primer for gastroenterologists. Journal of Neurogastroenterology and Motility, 22, 168–180. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sperandeo, P. , Martorana, A. M. , & Polissi, A. (2017). Lipopolysaccharide biogenesis and transport at the outer membrane of Gram‐negative bacteria. Biochimica et Biophysica Acta ‐ Molecular and Cell Biology of Lipids, 1862(11), 1451–1460. [DOI] [PubMed] [Google Scholar]
- Steinbacher, T. , Kummer, D. , & Ebnet, K. (2018). Junctional adhesion molecule‐A: Functional diversity through molecular promiscuity. Cellular and Molecular Life Sciences, 75, 1393–1409. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Stenman, L. K. , Lehtinen, M. J. , Meland, N. , Christensen, J. E. , Yeung, N. , Saarinen, M. T. , Courtney, M. , Burcelin, R. , Lähdeaho, M. L. , Linros, J. , Apter, D. , Scheinin, M. , Kloster Smerud, H. , Rissanen, A. , & Lahtinen, S. (2016). Probiotic with or without fiber controls body fat mass, associated with serum zonulin, in overweight and obese adults—Randomized controlled trial. eBioMedicine, 13, 190–200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Suez, J. , Korem, T. , Zeevi, D. , Zilberman‐Schapira, G. , Thaiss, C. A. , Maza, O. , Israeli, D. , Zmora, N. , Gilad, S. , Weinberger, A. , Kuperman, Y. , Harmelin, A. , Kolodkin‐Gal, I. , Shapiro, H. , Halpern, Z. , Segal, E. , & Elinav, E. (2014). Artificial sweeteners induce glucose intolerance by altering the gut microbiota. Nature, 514(7521), 181–186. 10.1038/nature13793 [DOI] [PubMed] [Google Scholar]
- Sung, H. , Kim, S. W. , Hong, M. , & Suk, K. T. (2016). Microbiota‐based treatments in alcoholic liver disease. World Journal of Gastroenterology, 22(29), 6673–6682. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Suzuki, T. (2013). Regulation of intestinal epithelial permeability by tight junctions. Cellular and Molecular Life Sciences, 70, 631–659. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Suzuki, T. (2020). Regulation of the intestinal barrier by nutrients: The role of tight junctions. Animal Science Journal, 91, e13357. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Suzuki, T. , & Hara, H. (2009). Quercetin enhances intestinal barrier function through the assembly of zonula occludens‐2, occludin, and claudin‐1 and the expression of claudin‐4 in Caco‐2 cells. The Journal of Nutrition, 139, 965–974. [DOI] [PubMed] [Google Scholar]
- Tagliabue, A. , Ferraris, C. , Uggeri, F. , Trentani, C. , Bertoli, S. , de Giorgis, V. , Veggiotti, P. , & Elli, M. (2017). Short‐term impact of a classical ketogenic diet on gut microbiota in GLUT1 Deficiency Syndrome: A 3‐month prospective observational study. Clinical Nutrition ESPEN, 17, 33–37. [DOI] [PubMed] [Google Scholar]
- Tang, W. H. W. , Bäckhed, F. , Landmesser, U. , & Hazen, S. L. (2019). Intestinal microbiota in cardiovascular health and disease. Journal of the American College of Cardiology, 73(16), 2089–2105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tatham, A. S. , & Shewry, P. R. (2008). Allergens to wheat and related cereals. Clinical and Experimental Allergy, 38, 1712–1726. [DOI] [PubMed] [Google Scholar]
- Thim‐Uam, A. , Surawut, S. , Issara‐Amphorn, J. , Jaroonwitchawan, T. , Hiengrach, P. , Chatthanathon, P. , Wilantho, A. , Somboonna, N. , Palaga, T. , Pisitkun, P. , & Leelahavanichkul, A. (2020). Leaky‐gut enhanced lupus progression in the Fc gamma receptor‐IIb deficient and pristane‐induced mouse models of lupus. Scientific Reports, 10(1), 777. 10.1038/s41598-019-57275-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tidjani Alou, M. , Lagier, J. C. , & Raoult, D. (2016). Diet influence on the gut microbiota and dysbiosis related to nutritional disorders. Human Microbiome Journal, 1, 3–11. [Google Scholar]
- Tito, R. Y. , Cypers, H. , Joossens, M. , Varkas, G. , Van Praet, L. , Glorieus, E. , Van den Bosch, F. , De Vos, M. , Raes, J. , & Elewaut, D. (2017). Brief Report: Dialister as a microbial marker of disease activity in spondyloarthritis. Arthritis & Rheumatology (Hoboken, N.J.), 69(1), 114–121. 10.1002/art.39802 [DOI] [PubMed] [Google Scholar]
- Trivedi, R. , & Barve, K. (2020). Gut microbiome a promising target for management of respiratory diseases. The Biochemist, 477, 2679–2696. [DOI] [PubMed] [Google Scholar]
- Trompette, A. , Gollwitzer, E. S. , Yadava, K. , Sichelstiel, A. K. , Sprenger, N. , Ngom‐Bru, C. , Blanchard, C. , Junt, T. , Nicod, L. P. , Harris, N. L. , & Marsland, B. J. (2014). Gut microbiota metabolism of dietary fiber influences allergic airway disease and hematopoiesis. Nature Medicine, 20, 159–166. [DOI] [PubMed] [Google Scholar]
- Tsuda, T. (2016). Recent progress in anti‐obesity and anti‐diabetes effect of berries. Antioxidants, 5, 13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Turnbaugh, P. J. , Quince, C. , Faith, J. J. , McHardy, A. , Yatsunenko, T. , Niazi, F. , Affourtit, J. , Egholm, M. , Henrissat, B. , Knight, R. , & Gordon, J. I. (2010). Organismal, genetic, and transcriptional variation in the deeply sequenced gut microbiomes of identical twins. Proceedings of the National Academy of Sciences of the United States of America, 107, 7503–7508. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Turner, J. R. (2009). Intestinal mucosal barrier function in health and disease. Nature Reviews. Immunology, 9(11), 799–809. [DOI] [PubMed] [Google Scholar]
- Uyar, G. O. , & Yildiran, H. (2019). A nutritional approach to microbiota in Parkinson's disease. Bioscience of Microbiota, Food and Health, 38, 115–127. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vader, L. W. , Stepniak, D. T. , Bunnik, E. M. , Kooy, Y. M. , de Haan, W. , Drijfhout, J. W. , van Veelen, P. , & Koning, F. (2003). Characterization of cereal toxicity for celiac disease patients based on protein homology in grains. Gastroenterology, 125, 1105–1113. [DOI] [PubMed] [Google Scholar]
- Valitutti, F. , & Fasano, A. (2019). Breaking down barriers: How understanding celiac disease pathogenesis informed the development of novel treatments. Digestive Diseases and Sciences, 64, 1748–1758. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Van Buiten, C. B. , Lambert, J. D. , & Elias, R. J. (2018). Green tea polyphenols mitigate gliadin‐mediated inflammation and permeability in vitro. Molecular Nutrition & Food Research, 62, 1700879. [DOI] [PubMed] [Google Scholar]
- van der Meulen, T. A. , Harmsen, H. J. M. , Bootsma, H. , Spijkervet, F. , Kroese, F. , & Vissink, A. (2016). The microbiome–systemic diseases connection. Oral Diseases, 22, 719–734. [DOI] [PubMed] [Google Scholar]
- van Krimpen, S. J. , Jansen, F. A. C. , Ottenheim, V. L. , Belzer, C. , van der Ende, M. , & van Norren, K. (2021). The effects of pro‐, pre‐, and synbiotics on muscle wasting, a systematic review‐gut permeability as potential treatment target. Nutrients, 13(4), 1115. 10.3390/nu13041115 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vazquez‐Roque, M. I. , Camilleri, M. , Smyrk, T. , Murray, J. A. , Marietta, E. , O'Neill, J. , Carlson, P. , Lamsam, J. , Janzow, D. , Eckert, D. , Burton, D. , & Zinsmeister, A. R. (2013). A controlled trial of gluten‐free diet in patients with irritable bowel syndrome‐diarrhea: effects on bowel frequency and intestinal function. Gastroenterology, 144(5), 903–911 e3. 10.1053/j.gastro.2013.01.049 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Villa‐Rodriguez, J. A. , Ifie, I. , Gonzalez‐Aguilar, G. A. , & Roopchand, D. E. (2019). The gastrointestinal tract as prime site for cardiometabolic protection by dietary polyphenols. Advances in Nutrition, 10, 999–1011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vogt, N. M. , Kerby, R. L. , Dill‐McFarland, K. A. , Harding, S. J. , Merluzzi, A. P. , Johnson, S. C. , Carlsson, C. M. , Asthana, S. , Zetterberg, H. , Blennow, K. , Bendlin, B. B. , & Rey, F. E. (2017). Gut microbiome alterations in Alzheimer's disease. Scientific Reports, 7(1), 13537. 10.1038/s41598-017-13601-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vojdani, A. (2015). Lectins, agglutinins, and their roles in autoimmune reactivities. Alternative Therapies in Health and Medicine, 21, 46–51. [PubMed] [Google Scholar]
- Vuong, C. N. , Mullenix, G. J. , Kidd, M. T. , Bottje, W. G. , Hargis, B. M. , & Tellez‐Isaias, G. (2021). Research Note: Modified serum fluorescein isothiocyanate dextran (FITC‐d) assay procedure to determine intestinal permeability in poultry fed diets high in natural or synthetic pigments. Poultry Science, 100(6), 101138. 10.1016/j.psj.2021.101138 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wahls, T. L. , Chenard, C. A. , & Snetselaar, L. G. (2019). Review of two popular eating plans within the multiple sclerosis community: Low saturated fat and modified paleolithic. Nutrients, 11, 352. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang, X. , Li, M. , Niu, Y. , Zhang, X. , Yin, J.‐B. , Zhao, C.‐J. , & Wang, R.‐T. (2019). Serum zonulin in HBV‐associated chronic hepatitis, liver cirrhosis, and hepatocellular carcinoma. Disease Markers, 2019, 5945721. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang, L. J. , Yang, C. Y. , Chou, W. J. , Lee, M. J. , Chou, M. C. , Kuo, H. C. , Yeh, Y. M. , Lee, S. Y. , Huang, L. H. , & Li, S. C. (2020). Gut microbiota and dietary patterns in children with attention‐deficit/hyperactivity disorder. European Child & Adolescent Psychiatry, 29(3), 287–297. 10.1007/s00787-019-01352-2 [DOI] [PubMed] [Google Scholar]
- Watanabe, Y. , & Tatsuno, I. (2017). Omega‐3 polyunsaturated fatty acids for cardiovascular diseases: Present, past and future. Expert Review of Clinical Pharmacology, 10, 865–873. [DOI] [PubMed] [Google Scholar]
- Watson, J. L. , Ansari, S. , Cameron, H. , Wang, A. , Akhtar, M. , & McKay, D. (2004). Green tea polyphenol (‐)‐epigallocatechin gallate blocks epithelial barrier dysfunction provoked by IFN‐gamma but not by IL‐4. American Journal of Physiology. Gastrointestinal and Liver Physiology, 287, G954–G961. [DOI] [PubMed] [Google Scholar]
- Weaver, K. N. , & Herfarth, H. (2021). Gluten‐free diet in IBD: Time for a recommendation? Molecular nutrition and food research. Hepatology, 73, 2180–2195. [DOI] [PubMed] [Google Scholar]
- Welsby, I. , & Goriely, S. (2016). Regulation of interleukin‐23 expression in health and disease. Advances in Experimental Medicine and Biology, 941, 167–189. [DOI] [PubMed] [Google Scholar]
- Wieser, H. (2007). Chemistry of gluten proteins. Food Microbiology, 24, 115–119. [DOI] [PubMed] [Google Scholar]
- Woodhouse, C. A. , Patel, V. C. , Singanayagam, A. , & Shawcross, D. L. (2018). Review article: The gut microbiome as a therapeutic target in the pathogenesis and treatment of chronic liver disease. Alimentary Pharmacology & Therapeutics, 47, 192–202. [DOI] [PubMed] [Google Scholar]
- Wu, R. L. , Vazquez‐Roque, M. I. , Carlson, P. , Burton, D. , Grover, M. , Camilleri, M. , & Turner, J. R. (2017). Gluten‐induced symptoms in diarrhea‐predominant irritable bowel syndrome are associated with increased myosin light chain kinase activity and claudin‐15 expression. Laboratory Investigation, 97(1), 14–23. 10.1038/labinvest.2016.118 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yan, Y. L. , Hu, Y. , & Ganzle, M. G. (2018). Prebiotics, FODMAPs and dietary fiber—Conflicting concepts in development of functional food products? Current Opinion in Food Science, 20, 30–37. [Google Scholar]
- Yang, J. , Lim, S. Y. , Ko, Y. S. , Lee, H. Y. , Oh, S. W. , Kim, M. G. , Cho, W. Y. , & Jo, S. K. (2019). Intestinal barrier disruption and dysregulated mucosal immunity contribute to kidney fibrosis in chronic kidney disease. Nephrology, Dialysis, Transplantation, 34(3), 419–428. 10.1093/ndt/gfy172 [DOI] [PubMed] [Google Scholar]
- Yang, L. , Wang, L. , Wang, X. , et al. (2016). Possible role of intestinal microbiota in the pathogenesis of ankylosing spondylitis. International Journal of Molecular Sciences, 17, 2126. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yang, L. , Wang, L. , Wang, X. , Xian, C. J. , & Lu, H. (2016). A possible role of intestinal microbiota in the pathogenesis of ankylosing spondylitis. International Journal of Molecular Sciences, 17(12), 2126. 10.3390/ijms17122126 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yao, C. K. , Muir, J. G. , & Gibson, P. R. (2016). Review article: Insights into colonic protein fermentation, its modulation and potential health implications. Alimentary Pharmacology & Therapeutics, 43(2), 181–196. [DOI] [PubMed] [Google Scholar]
- Yeoh, N. , Burton, J. P. , Suppiah, P. , Reid, G. , & Stebbings, S. (2013). The role of the microbiome in rheumatic diseases. Current Rheumatology Reports, 15(3), 314. 10.1007/s11926-012-0314-y [DOI] [PubMed] [Google Scholar]
- Yoon, H. , Schaubeck, M. , Lagkouvardos, I. , et al. (2018). Increased pancreatic protease activity in response to antibiotics impairs gut barrier and triggers colitis. Cellular and Molecular Gastroenterology and Hepatology, 3, 370–388. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yu, H. C. , Lu, M. C. , Huang, K. Y. , Huang, H. L. , Liu, S. Q. , Huang, H. B. , & Lai, N. S. (2015). Sulfasalazine treatment suppresses the formation of HLA‐B27 heavy chain homodimer in patients with ankylosing spondylitis. International Journal of Molecular Sciences, 17(1), 46. 10.3390/ijms17010046 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yu, S. , Wu, X. , Zhou, Y. , Sheng, L. , Jena, P. K. , Han, D. , Wan, Y. J. Y. , & Hwang, S. T. (2019). A Western diet, but not a high‐fat and low‐sugar diet, predisposes mice to enhanced susceptibility to imiquimod‐induced psoriasiform dermatitis. Journal of Investigative Dermatology, 139, 1404–1407. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yue, Y. , Guo, Y. , & Yang, Y. (2017). Effects of dietary l‐tryptophan supplementation on intestinal response to chronic unpredictable stress in broilers. Amino Acids, 49, 1227–1236. [DOI] [PubMed] [Google Scholar]
- Zak‐Gołąb, A. , Kocełak, P. , Aptekorz, M. , Zientara, M. , Juszczyk, L. , Martirosian, G. , Chudek, J. , & Olszanecka‐Glinianowicz, M. (2013). Gut microbiota, microinflammation, metabolic profile, and zonulin concentration in obese and normal weight subjects. International Journal of Endocrinology, 2013, 674106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang, D. , Zhang, L. , Zheng, Y. , Yue, F. , Russell, R. D. , & Zeng, Y. (2014). Circulating zonulin levels in newly diagnosed Chinese type 2 diabetes patients. Diabetes Research and Clinical Practice, 106(2), 312–318. [DOI] [PubMed] [Google Scholar]
- Zhou, Z. , & Zhong, W. (2017). Targeting the gut barrier for the treatment of alcoholic liver disease. Liver Research, 1, 197–207. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zihni, C. , Mills, C. , Matter, K. , & Balda, M. S. (2016). Tight junctions: From simple barriers to multifunctional molecular gates. Nature Reviews. Molecular Cell Biology, 17(9), 564–580. [DOI] [PubMed] [Google Scholar]
- Zinöcker, M. K. , & Lindseth, I. A. (2018). The Western diet–microbiome‐host interaction and its role in metabolic disease. Nutrients, 10, 365. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
Data sharing is not applicable to this article as no datasets were generated or analyzed during the current review.