Journal of Nutrition & Food Sciences

Journal of Nutrition & Food Sciences
Open Access

ISSN: 2155-9600

+32 25889658

Research Article - (2018) Volume 8, Issue 6

Recent Advances In Nutritional Sciences: An Overview Of Glycans And MiRNAs

Marcello Menapace*
M & Ms Consulting Ltd., Borehamwood, Hertfordshire, United Kingdom
*Corresponding Author: Marcello Menapace, Managing Director, M & Ms Consulting Ltd., Devonshire House Manor Way, Borehamwood, Hertfordshire WD6 1QQ, United Kingdom, Tel: +44-7423466123

Abstract

There are many nutritional substances that humans consume on a daily basis: water, carbohydrates lipids and proteins are main biochemical components of food. But others in a smaller amount are vitamins minerals and enzymes. At a possibly lesser quantity are glycans and miRNAs. The presence of oligoglycans in all food sources has been an established fact for many years. These special carbohydrates are present as glycoconjugates (glycoproteins or glycolipids) in and on the surface of all the cells (glycocalyx) of all organisms that we eat and remain intact through the GI tract as we lack the enzymatic repertoire of the human body to unbind their particular β-linkages. Glycans bind to naturally present human lectins (through protein-carbohydrate interactions), but also with other human glycans (through carbohydrate-carbohydrate interactions, or CCI). Moreover, these glycans, like fibres, are digested by the gut microbiota that resides within the intestine. As our biochemistry shapes the composition of the microbiome, so does the composition of glycans and foods that we consume, triggering biological responses. miRNAs are small, single-stranded, 19- to 23- nucleotide-long RNA molecules and affect the stability of messenger RNA (mRNA) influencing protein synthesis. miRNAs are also present in foods and act on both the microbial composition in our gut and may be absorbed by the walls of the GI tract, demonstrating resistance to food processing and enzymatic attack. Though still a topic of controversy, these small, noncoding RNAs that control gene expression may directly enter into the circulating miRNA population of dietary exogenous miRNAs. It can hence be possible to identify a relationship between glycans and miRNAs in food on one side, microbiota composition on the other and the resultant health status of the host (immune system), on the third side.

Keywords: Glycan; miRNA; Microbiome; Glycotopes; Food antigens; Glycobiology; ABO blood group

Introduction

Modern science has amply discussed and explained the nutritional requirement of macronutrients and micronutrients [1].

The diet generally provides three macronutrients [2], carbohydrates, proteins and fats [3], and three micronutrients (vitamins, minerals and water) [4], which are essential for life [5]. These are absorbed in the gastrointestinal tract (GIT) after being processed by human and eventually microbic enzymes [6].

There are, nevertheless, other classes of bioactive compounds that have been recently studied, namely glycans and microRNA (miRNA or miR). In reality, glycans are carbohydrates (specifically, oligosaccharides, another group of carbohydrates), which have been studied for their special antioxidant and immunostimulating properties [7]. But, almost unbeknown to the nutritional research community is that oligosaccharides are ubiquitous [8,9], especially in the form of glycoconjugates [10]: either protein-bound [11] or lipid-bound [12]. In a similar manner, miRNA, a group of small non-coding RNA molecules that regulate genes at the post-transcriptional level [13], are found in animal and plant based foods [14]. Although mixed results have been obtained from studies on cross-kingdom transfer of exogenous miRNAs (exomiRs) in humans [15,16], many reports suggest exomiRs pass through the gastrointestinal tract and are found in human serum regulating the expression of endogenous mRNA [17].

These two particular bioactive compounds, because of their diverse structure and functions, can have enormous effects on health. A thorough review of current and past scientific literature has been performed on the advances of both fields and the results are presented, along with recommendations for future research.

Glycans and miRNAs

Glycans are not just fibers

According to the National Institute of Health [18], dietary fiber is defined as nondigestible carbohydrates while functional fibers are isolated, nondigestible carbohydrates with shown beneficial physiological effects in humans. Total fiber is the sum of dietary fiber and functional fiber [19]. In general, fibers and/or some types of resistant starches (RS) cannot be digested enzymatically by humans [20].

Dietary fibers are classified in a variety of ways, depending on the origin of the fiber, the molecular composition and the functional effects (viscosity, fermentability and solubility) [21].

Most beneficial effects reflect their laxative effect [22]. As just mentioned, all fibers are not hydrolized by the human digestive enzymes [23]. We, humans, do not have the enzymatic capability to degrade complex nonstarch polysaccharides - dietary fiber [6,24-26]. Our digestive capabilities are extremely reduced and are essentially limited to starch, sucrose, and lactose [27-29].

Undigested dietary carbohydrates include polysaccharides (including cellulose, hemicellulose, xylan, and pectin [30]) as well as some undigested starch [31]. And hence they are not absorbed in the small intestine (upper GIT), unlike most starches, having the function of increasing stools bulk [26,32], whereby the regularity effect.

RS are essentially long glucose chains linked with α-D-(1-4) and α-D-(1-6) glycosidic linkages, but are also hardly accessible to amylases [33,34]. In this sense, RS are similar to normal glucose based starches: α-D-(1-4) glycosidic bonds for amylose, a linear polysaccharide, and both α-D-(1-4) and α-D-(1-6) for branched amylopectin [35]. RS interspersed within amylose and amylopectin, rendering them more inaccessible to human hydrolases [36]. In the end, RS exhibit a level of slow digestibility (up to the large intestine), which manifests as a slow release of glucose [34].

Dietary fibers are thus very important for health [6,26].

Moreover, dietary fibers have other beneficial effects such as appetite control and energy intake through fermentation and production of short chain fatty acids (SCFA) by gut bacteria [37].

Less fermentable fibers tend to increase fecal weight to a greater degree than more fermentable fibers [38].

But, functional fibers are also composed of small oligosaccharides [32]. Nevertheless, no nutritional study has reviewed those carbohydrates that resist digestion in the small intestine (fibers, according to the 2009 CA [39] and are present in all food sources. Glycans, are one of four principal components of the cell [40].

These carbohydrates have different structural composition with respect to standard insoluble [41], soluble [42], prebiotic homopolysaccharides [43], which constitute most of the known fibers. Chemically, backbone of these indigestible glycans is composed of sugar residues linked together by β-1,4-glycosidic bonds [30].

Figure 1 shows the difference between starch, fibres and widespread glycan antigens (mostly, branched N-linked [44-46] and O-linked oligosaccharides [47] from glycoconjugates [48].

nutrition-food-sciences-fibers-glycans

Note: The first structure represents a branched starch (glucose) with its α1-4 and 1-6 glycosidic linkages. The second represents indigestible cellulose (glucose) with its β1-4 glycosidic bond. The third structure is a cartoon representation of a simple (linear) glycan with many linkage types between several different saccharide residues. On the right, a simplified version of the explanation of which monosaccharides the cartoon geometric figures represent.

Figure 1: Structural differences between starches, fibers and glycans.

For an in-depth review of glycoconjugates and glycans, reference is made to an entire journal series, Glycoconjugates, and to arguably the most important textbook on the topic: Essentials of Glycobiology [49]. This extensive analysis of the scientific literature included the multiplicity of functions, the intrinsic diversity (microheterogeneity [50]) and ubiquitousness (including protein site occupancy [51] with O- and N-glycosylation [52]) of these sugar antigens and will be briefly and partially introduced.

Glycans are ubiquitous

Glycans are ubiquitous in nature [53,54]. The cell surface is literally coated with carbohydrates in the form of glycoproteins, with oligosaccharides (sugar residues), proteoglycans [43,55-57], polysaccharides, and glycolipids (as glycoconjugates) [58-60]. This little-known part of the cell is called the glycocalyx [61-63]. The glycocalyx is responsible for a vast number of biological functions from ion exchange, to cellular recognition, cell adhesion and development [64,65], from protein folding and activity [66], to regulation of signalling, or immune regulation [67], self or non-self-recognition [68], and homeostasis [69,70].

The cell surface interacts with the extracellular matrix (ECM [56,71,72]), also made up of glycans [73-77], apart from proteins.

Glycoconjugates are widely present in animal [78], as well as plant [79-83] and microbial [84-87] structural and functional systems [88]. Many of the plant glycan antigens are on the cell wall [89,90]. So is the bacterial cell wall [84,91], as it is surrounded by peptidoglycans [92] and lipopolysaccharides [93].

More than 50% of all proteins in eukaryotes are known to be glycosylated [46]. Nearly all human secreted and cell surface proteins are co- and post-translationally modified by glycans [40,65]. The different protein glycoforms depend on glycan site-specific microheterogeneity [94] and influence the physical, rheological [95], chemical and biological [96,97] properties of proteins, though having similar molecular weights.

Asparagine (N)-linked glycosylation is the most widespread form of modification found in secreted glycoproteins of all eukaryotes and some prokaryotes [45]. It is closely followed by O-glycosylation as a major form of protein glycosylation at the serine/threonine residues [98].

Given the diverse and complicated structures of the oligosaccharides (multiple branched chains of monosaccharides between 3 and 20 sugar units in length) [99], a standard symbolic representation system has been devised [49].

The presence and importance of glycans in human milk cannot be underestimated [100]. Human milk oligosaccharides (HMO) [101] are a family of unconjugated, structurally diverse and complex glycans that are very abundant in human milk [102-105].

Thus, conjugates of carbohydrates are ubiquitous in nature [78]. Glycans are reactive chemical species Glycans are recognized by glycan-binding proteins (GBPs) [53,106], or lectins, which play a pivotal role in many different aspects of the physiology [40], including the immune defence [107]. Thus, glycans react with lectins and bind to them.

Lectins are carbohydrate-specific reagents and biological recognition molecules [108,109]. Lectins, too, are ubiquitous (in nature) [110,111], as carbohydrate binding proteins [60]. They are found abundantly in both plants [112] and animals [113], and in bacteria [114]. Lectins have specific carbohydrate-binding sites (or domains, CBD) [115], and that preferentially recognize with low affinity (protein-carbohydrate interactions, or PCI, usually in the mM range) [113,116]. Lectins are thus perfectly suitable as decoders for carbohydrate-encoded information [117].

Lectin-glycan interactions are involved in a myriad different biological processes [118,119], including intracellular signalling pathways that regulate the immune response [120,121]. Glycans are so reactive that lectins will also bind to carbohydrate chains on glycolipids and proteoglycans or interact with polysaccharides and free sugars [122].

Many classification systems for lectins are based on morphology and interaction [123]. Given the heterogenicity of structures and specificities of GBPs [119], recently, the number of lectin families has jumped from 10 by the end of the 20th century to 50 [124].

There are many lectins that have specific immune functions [125], namely

1. Galectins [126,127] also called S-type lectins,

2. C-type lectins (like DC-SIGNs and Dectin 1 [128-130]),

3. Siglecs [131,132], a major subfamily of I-type lectins [133]

4. I-type lectins [134] (among which intercellular adhesion molecule-1 (ICAM-1) [135-137], neural cell adhesion molecules (NCAM) [138,139], and some clusters of differentiation (CDs) [140], are the major examples), and

5. Selectins [141-143].

The complementarity between glycans and lectins has been defined as the third alphabet of life [138,144]. An alphabet capable of transmitting maximum information in a minimum of space [145].

Moreover, glycans are known to allow proteins to reorganize (or self-associate) themselves spatially on the cell surface through PCI or carbohydrate-carbohydrate interactions (CCI) [64,83] to form ‘lipid rafts’ [58,59,146,147]. These microdomains, or clusters of glycoproteins (lectins) [148] and glycolipids, once formed, regulate several biological functions [149-152], including immune reactions [116,153].

Figure 2 is a schematic representation of the formation of lipid rafts on the glycocalyx, as special microdomains [154].

nutrition-food-sciences-Lipid-raft

Note: The two sections of a cell membrane represent the before and during the lipid raft event. Glycans are shown as small blue hexagons on the top of proteins or lipids (the phospholipids of the bilayer membrane). Depending on the type of glycans (ABO) the creation of the lipid raft will change slightly in architecture [155], allowing certain proteins to become closer and thus be able to recognize foreign matter differentially.

Figure 2: Lipid raft enacted by ABO glycans.

The biologically significant CCI have lower affinities with respect to PCI [156] but are still powerful enough to participate in cellular processes [157-159]. This occurs thanks to simultaneous multivalent noncovalent interactions with the appropriate saccharide or amino acid moieties [54,157].

A hypothesis that the carbohydrate structures are another potential source of antigenicity (food antigens) was proposed in the 1980s [160,161]. The glycans on different plant and animal protein allergens were then found to possess properties of immunological cross-reactivity [162]. Many food glycans (especially from plants) were isolated and structurally identified [163,164]. These glycans were called cross-reactive carbohydrate determinants (CCD) [165,166].

Many CCDs have been characterised as N-glycans [167,168], having weak allergic [169] or non-allergenic immunomodulatory properties (Table 1) [170].

Function/Effect Glycans1 MIR
Immune modulatory HMO with PRRs (lectins) In milk or foods
Self/non-self recognition glycans from microbe or diet with human receptors (lipid rafts) -
Regulation of signalling On free and cell glycoconjugates (lipid rafts) Microbiome-GIT interaction
Protein folding On cell surface proteins (lipid rafts) -
Homeostasis glycans from microbe or diet with human receptors Food MIR modify epigenetics
Cross-communication with ECM On free and cell glycoconjugates  
Cell to cell communication On cell surface glycoconjugates (lipid rafts) MIR from one cell acts on another

1Glycans react with lectins through PCI/CCI.

Table 1: Interaction of glycans and MIRs with health.

miRNA are ubiquitously small and reactive

MicroRNA (miRNA or miR) are small, single-stranded, 19- to 23- nucleotide-long RNA molecules and affect the stability of messenger RNA (mRNA) thus influencing protein synthesis [171]. miRNA are small non-coding RNA (ncRNA) molecules that are not translated into proteins [172]. Various proteins are involved in the process of reduction of ncRNAs to miRNAs [173]. miRNAs regulate gene expression by silencing genes through translational repression and/ or messenger RNA (mRNA) destabilization [174,175]. Since binding to mRNA is not homologous [176], a single miRNA could potentially regulate hundreds of genes [177]. Although small RNA molecules are classified into two other types, small interfering RNAs (siRNAs) and Piwi-interacting RNAs (piRNAs) based on different characteristics and origins [178,179], we shall refer to miR for all of them, for simplicity.

Several studies have demonstrated that bioactive food components can affect miRNA expression and transport through the bloodstream through exosomes, and thus their pathways, in many diseases, including cancer [180]. Exosomes are 40-100 nano-sized vescicles present in various biologic fluids and are known to contain miRNAs [181].

It has recently been discovered that small RNAs are present in human gut and regulate homeostasis by targeting chromatin and transcripts [182].

miRNA, as a class of small, noncoding RNAs that control gene expression of almost every cell process, by targeting mRNA, have been shown to be not only modulated by these dietary bioactive compounds [183], but also be present in the diet. miR have been detected and classified in cow’s milk and colostrum [184], mammalian milk [185,186], human breast milk [187]. miR are present in plants [188,189].

There are several recent studies suggesting that plant miRNA may be absorbed by cells of the mammalian epithelial wall from food and are packaged into appropriate microvescicles where they are protected from degradation and delivered into the bloodstream [190].

Small RNA molecules can be attacked and broken down by special enzymatic machineries called nucleases/nucleotidases [191-193]. Nonetheless, miRNAs are able to withstand RNAse and other extreme conditions [194] indeed, small strands of RNA (siRNA and miRNA) are able to be absorbed by the walls of the intestinal barrier intact, demonstrating resistance to food processing and to enzymatic attacks [195,196].

The absorption of exomiRs in plant food has been a topic of controversy in past years [197,198]. The dietary delivery of miRNAs in humans is at least feasible [199], and several researchers recently found plant exogenous miRs in human samples [16,200]. Indeed, orally delivered therapeutic RNA strands, called short interfering RNA (siRNA) has been demonstrated in vivo [201].

miR, as small endogenous RNAs that pair to sites in mRNAs to direct post-transcriptional repression, are predicted to regulate up to 60% of human protein-coding genes [202,203]. The mechanism of action involves Being similar to methylation, miR are involved in epigenetic regulation of protein synthesis [172]. miR are involved in homeostasis as well as disease progression [204].

Health Impact

The relationship between diet, the gut microbiota, microbial activity, and gut physiology is quite complex [39]. Many studies have been completed to model the impact of the microbiota on human health [205-207] and homeostasis [208]. Other studies have linked interactions between the immune system and the gut to several metabolic diseases [206,209,210]. A pattern seems to emerge in which several environmental factors are linked together and trigger disease or health [211]. A recent relationship has emerged between diet, microbiota and health status [212].

All these factors and triggers must be explained somehow by fundamental interactions between the gut, the diet and the microbiome. This can be aided by considering the impact of glycans and miRNAs. Table 1 shows this interaction which will be explained in the next subsections.

The Gut and the immune system

The gastrointestinal tract is the body’s foremost tissue boundary, interacting with nutrients, exogenous compounds and gut microflora [213]. The gut-associated lymphoid tissue (GALT) is expectedly the largest immune organ in the body [214].

The intestinal surface is covered by a single cell lining of intestinal epithelial cells (IEC) [215,216], and a mucus layer [98,217]. The gut secretes intestinal mucins [182], like MUC2 [98], the major colonic mucin, which is a large protein extensively O-glycosylated [211].

Many other cell types make up the GI mucosa [191,218-224], among these are dendritic cells (DCs) [225], that sample commensals and antigens and interact with lymphocytes: B and T cells [226,227].

The innate immune system is the first line of host defence of both humans and animals and is present in the gut [224]. The function of the innate immune is to recognize pathogen-associated molecular patterns (PAMPs) or microbial-associated molecular patterns (MAMPs) [228], using a variety of host receptors called pattern recognition receptors (PRRs) [229,230]. Interestingly, many of the PRRs are lectins [120,122] and interact with foreign or endogenous glycans.

PRRs are involved in antigen capture, presentation, and activation of immune [208,231] and inflammatory responses [230,232-234].

After the mucus [235], the IECs have a significant role in limiting exposure to commensal microbes by producing antimicrobial molecules [236]. Many of these are lectins [228,237-241].

Most of these immune-related effects are triggered by the commensal microbiota [242] to maintain a state of equilibrium between defence against pathogens and tolerance to resident microbes [243].

Gut microbiota

In the distal part of our digestive tract resides the human intestinal microbiome, a complex and dense ecosystem of bacteria [6,241]. The gut microbiota is, in fact, a vast mutualistic and symbiotic collection of diverse microbes [24,208,215,216,236,244-246], providing the host with a broad range of functions in exchange for a stable environment [24]. This mutualism [205], or cooperative partnership [241,247] and centred around diet [248]. This strategic symbiotic/commensal relationship of the human holobiont [249] is enacted in two ways [250].

Firstly, the microbiome can degrade otherwise indigestible components (glycans) of our diet [251-253] and transform these plant and animal-derived glycans into SCFA [26,250,254-258], which serve as multifaceted nutrients for colonocytes with health benefits [24,252,259-263].

Secondly, the gut provides a plethora of glycan substrates (glycoconjugates like chondroitin or heparin or mucin) that are utilized by the microbes for growth [250,264-268].

The microbiota shifts rapidly in response to dietary changes in fiber (glycan specificity or glycan preference [269]) or macronutrient content [23].

Moreover, it is known that the gut microbiota shapes aspects of host metabolism and immune function [24,270-274], is implicated in several disease states [252,275-277], and influences myriad of other biological activities [278], such as host-energy balance [206,208,254,279] and colonization resistance [211,246].

Many studies have evaluated the effects of different compositions of bacteria [280-285], causing dysbiosis [276], a disruption in the host’s nutrition [286] and immunity [287] (or inflammation [173,232] and other diseases [276]), as opposed to normobiosis [288].

The innate immune system also shapes the commensal microbial communities [271], through multiple lectins, PRRs, [208,289,290], as regulators of intestinal microbiota for homeostasis [291]. The resident microbiota utilizes molecular mimicry [292] as a tolerance mechanism, whereby bacteria display surface molecules (glycans) that resemble those of the host’s surface to render them immunologically inert [293]. Glycoconjugates like lipopolysaccharides, peptidoglycans [279], lipoteicoic acid [294,295], and other microbial polysaccharides have been identified cell-to-cell contact [205,274,296,297], mediated by both PCI and CCI [298].

By their part, the commensal microbes have an immunomodulatory effect on the host [242,243,299]. The capsular polysaccharide (CP) as shown in Figure 3, have both protective functions for the bacteria [300,301] and immunologic potential (contributing to virulence) [302,303]. Other CPs or immunomodulatory glycans [304] activate yet further molecular pathways for host immune responses [305-309].

nutrition-food-sciences-membrane-microbes

Figure 3: Glycoconjugates of external capsule/membrane of microbes.

Overall, these mechanisms allow a complex interplay between the bacterial flora and the host, mediated by glycans.

Gut-microbiome-diet interaction

It is known that imbalances in gut microbial populations can be managed through diet [310,311]. Diet impacts microbial ecology [311], through differences in glycan metabolic activities [312,313]. This depends on the microbiome’s differential expression of a number of carbohydrate-active enzymes (CAZymes) [27,28,314], capable of targeting a wide range of complex glycans [28,263,315-318]. Glycans are thus differentially consumed by the gut microbiota [94,263,319].

Many glycan substrates in foods differ in quantity and quality [252] and can greatly impact microbial composition [253,257,283,314,320- 332] and activity [31,311,323,329,330,333-337].

Although these depend on the initial genetic composition of an individual [287,335,338].

Host chemical features, which are genetically encoded (e.g., mucin chemotypes), are primary determinants of the normal microbiota [313]. Such factors are also nutrient sources for the microbiome [26,256,257,339].

Several studies have demonstrated this interaction [340-342]. The two types are: GIT glycans-microbe lectins and gut lectins-microbe glycans.

The first type of interaction occurs between receptors of microbes (lectins) and glycans of the gut. Microorganisms have been known to be susceptible to histo-blood group antigens (HBGA) [343-347] and ABO determinants (glycans) [348-351] since the 1980s [352,353], and these are present along the entire GI tract [354]. Microbes include H. pylori [265,355-357], cholera [358-360] and rotavirus [361,362].

The second type of interaction between microbe glycans (foremostly, HBGA) and human lectins has also been recognized [130,293,363-368].

Irrespective as to which type of interaction, micro-organisms have been conclusively shown to bind differentially to glycans on host cells or mucins [40,63,358,369-371], including ABO [298,320,372,373].

The ABO blood group is one of the genetically determined host factors modulating the composition [374,375] (differences in proportion and profiles [375]) and function of the human intestinal microbiota, through glycan-glycan cross-talks [69].

One example of food item to consider is human milk. This food contains HMOs [376-378], which are essentially sialylated and α1–2, α1–3, α1–4 fucosylated glycans [105,379,380]. The presence of β1-3, β1-4 and β1-6 glycosidic linkages [377] make them a selective food source for both the microbiome and the host [102,381-383].

HMOs have been shown to cross the epithelial barrier [384,385], and to circulate in the blood [379,386], before being excreted in the urines [387], or to pass directly through the GIT into the faeces [388]. HMOs can bind to cell surface receptors of IECs and leucocytes and thus modulate neonatal immunity [103]. Thus, HMOs, similarly to other food-derived oligosaccharides [389], seem to have direct and indirect effects on the mucosal and systemic immune function of the consumer [381,390-403].

Milk also contains high expression levels of miR [404]. Whether these miRNAs can affect the human commensal microbes may be suspected but is currently unknown. But, biologically meaningful amounts of miRNA, present in cow’s milk, are absorbed during consumption [405] and circulate in the blood [196]. Quite a few studies have found miR present in cow’s milk and human milk [406]. These milk miRs have been linked to various immunoregulatory roles on target genes among mammals [404,407].

Studies have linked microbes with differential expression of caecal miR signature and thereby with regulation of the intestinal barrier function [173] and immune response [172,177,408,409]. As an epigenetic factor of regulation, miR may be controlled by microbiota through SCFA production [410].

Finally, human endogenous miR excreted by IECs, and called faecal miRNA, are known to shape intestinal commensal microbes [411,412]. It has been shown that IEC’s miRNAs are absorbed by the microbiota [413] and modulate the gene transcripts affecting bacterial growth [414]. This is part of the complex regulatory networks of host pathogen interaction [202]. Exosomal miRNAs are released for the regulation of bacterial gene expression [415].

Impact of food on health: the triple relationship

An interplay between diet, microbes and the gut as herewith briefly outlined has already been noticed [416]. This is not a unique intuition, as it copies very closely what has already been described [271]. The novelty is the outlook given to glycans and MIRs.

Figure 4 summarizes the previous sections.

nutrition-food-sciences-microbiome-gut

Note: Three-way interaction of food, GI microbial communities and GI tract. Firstly, food shapes the commensal microbiota in the gut by defining which microorganism will thrive and which won’t. Secondly, human gut will modulate the composition of the intestinal microbiota. Finally, glycans and miRNAs from both food and commensal bacteria can be pass through the GI tract walls and interact with the immune system.

Figure 4: Interaction among food, microbiome and gut.

Briefly, Figure 4 explains how

1. Food interacts with both the commensal microbiome and the gut to control immune responses and consequently health

2. Food modulates the composition of the microbiome (3.3), which affects immunity (3.2)

3. Food directly influences immune responses through glycans (2.2) and miR (2.3), by acting on the gut linen and on the GALT

4. The gut also influences the microbiome through glycans and miR (3.3), and

5. Commensal bacteria modulate gut development and homeostasis and immunity (3.1).

So, since the gut microbiota can be altered upon feeding of specific glycans and MIRs (from food) that promote the selective growth of one or more microbial genus (era)/species depending on the glycanic alignment with the gut, this confers health benefits to the host [288,416]. But, food (glycans) must be aligned with the genetic makeup (glycans) of an individual for the benefits to be maximised, otherwise there may be an opposite effect: an immune or inflammatory reaction. The afore-represented relationship could be called the health triangle, as diet can modulate health maintenance and disease prevention [417], through the various known mechanisms [214,228,418-423] with the help of glycans and MIRs.

Therefore, environmental factors within the intestines (e.g., glycans and MIRs from diet and microbiota) may influence innate immunity to cause chronic inflammation [211,342,424,425].

A holistic perspective

One last variable to input in therefore mentioned equation is the typology of glycans that constellate the glycocalyx of practically every human cell. Without exception, the most important glycotope system of the human body is the HBGA system (ABO and Lewis), given its ubiquitous tissue distribution in humans [175,426-434], free or bound to the ECM [116]. HBGA (Lewis rather than ABO) are also present on oligosaccharides abundant in human milk [101,379].

Particularly, the ABO epitopes have been shown to be fundamental in the characterization of the different possible biochemical cascades originating in self or non-self (inflammatory and/or immunologic) reactions [62]. Moreover, since glycans on proteins and lipids can slightly modify the glycoconjugate’s shape and functionality [70,155], HBGA can contribute to special spatial plasma membrane architecture [435], allowing for peculiar cellular properties [436]. These special clusters, or lipid rafts, have been shown to form differentially in the presence of ABO antigens [155,437]. Figure 2 shows schematically an such event where the terminal of glycans can be substituted with HBGAs.

Hence, it is noticeable how HBGAs contribute to each individual glycophenotype [99], with subsequent dissimilar events following consumption of the same food items as outlined in section 3.4.

In this context, the advent of Blood-Type Diets (BTD) [438], after it was first proposed by P. J. D’Adamo in 1996 [439], becomes now theoretically admissible. Clinically, other practitioners followed the nutritional indications of the BTD [440-442], such as Italian physician P. Mozzi [443], with success. Although little academic attention was given to BTD due to lack of both suitable experimental clinical data [444,445], an initial primary mechanism was proposed involving dietary lectins [446], and their potential biological activity [112,447].

Since HBGA-like glycans have been found in food items [448], and because microbes bear HBGA-like substances (outside the cell surface) [449], recognition events triggered by PCI or CCI with endogenous lectins are the norm. This cascades in an alignment of food items and microbiota with the genetic constitution of an individual (gut linen) based on an ABO glycophenotype. The mechanistic explanation of BTDs (through PCI or CCI) becomes therefore straightforward and highly probable. This is one genetic way in which microbial communities in the gut can be highly individual and influence health [306].

Conclusions and Future Research

As has been amply documented, there is a multifaceted and intricate relationship between food, microbiota and the gut [450]. This relationship is heralded by glycans and miRs. We are currently just starting to appreciate the complexities of these relationships on a totally new level: that of glycans and small ncRNAs.

This is not to say that proteins, fatty acids and DNA or other substances present in food should not also be included in this health triangle. On the contrary, these macronutrients should indeed be included to gain a full picture of nutritional epigenomics [451] and nutritional immunology. The main scope of this work was to underline two outliers, which have too often been neglected.

As newer technologies become available to analyse these two different molecules, many new questions can be answered. Chiefly important are profiling of glycans and exomiR in food items. Due to their size and difficulties in the isolation process, this has evaded for so long a time.

Once the glycans and exomiRs are identified, their kinetics should be closely studied. Independent analysis of each glycan or miR should follow the principles of pharmacokinetics. Quali-quantitative absorption of the substance from food items is the first step in the study of its bioavailability. Tissue distribution and relative metabolism or interaction is the next step of the study. The endpoint is to discover how these molecules influence health. A task requiring an in-depth description of the biochemical pathways that involve cross-talks between these and other macro- and micro-nutrients.

In ultimate analysis, the complexity of human glycans (especially HBGA), their presence in the animal, vegetable and microbial world and their relative reactivity with the ubiquitous lectins cannot be set aside any longer. As we are gaining access to new strategies for study of glycomics, the primary differentiation in responses to foods between distinct ABO blood individuals will become easier to understand. Chief among these strategies is the possibility of full structural characterization of oligosaccharides in their unique glycoforms and their absolute quantitation in foods. As we have just scratched the surface of glycans and miRs, the next step is to appraise their vast networks of biological functionalities.

A task that, in by itself, may well take several decades to complete.

References

  1. Wang LL, Wang Q, Hong Y, Ojo O, Jiang Q, et al. (2018) The effect of low-carbohydrate diet on glycemic control in patients with type 2 diabetes mellitus. Nutrients 10: E661
  2. Staubo SC, Aakre JA, Vemuri P, Syrjanen JA, Mielke MM, et al. (2017) Mediterranean diet, micro- and macronutrients, and MRI measures of cortical thickness. Alzheimers Dement 13: 168-177
  3. Shondelmyer K, Knight R, Sanivarapu A, Ogino S, Vanamala JKP (2018) Ancient Thali Diet: Gut Microbiota, Immunity, and health. Yale J Biol Med 91: 177-184
  4. Whitney EN, Rolfes SR (2018) Understanding nutrition (15th Edn). Cengage Learning; US, p: 848
  5. Tasse L, Bercovici J, Pizzut-Serin S, Robe P, Tap J, et al. (2010) Functional metagenomics to mine the human gut microbiome for dietary fiber catabolic enzymes. Genome Res 20: 1605-1612
  6. Hamed I, Özogul F, Özogul Y, Regenstein JM (2015) Marine bioactive compounds and their health benefits: a review. Compr Rev Food Sci Food Saf 14: 446-465
  7. Roychoudhury R, Pohl NL (2013) Light fluorous-tag-assisted synthesis of oligosaccharides. Modern Synthetic Methods in Carbohydrate Chemistry: From Monosaccharides to Complex Glycoconjugates. 8: 221-239
  8. Dabrowski J, Kozar T, Grosskurth H, Nifant'ev NE (1995) Conformational mobility of oligosaccharides: experimental evidence for the existence of an "Anti" conformer of the Gal.beta.1-3Glc.beta.1-OMe disaccharide. J Am Chem Soc 117: 5534-5539
  9. Varki A (1993) Biological roles of oligosaccharides: all of the theories are correct. Glycobiology 3: 97-130
  10. Patel HV, Vyas AA, Vyas KA, Liu Y-S, Chiang C-M, et al. (1997) Heparin and heparan sulfate bind to snake cardiotoxin. Sulfated oligosaccharides as a potential target for cardiotoxin action. J Biol Chem 272: 1484-1492
  11. Tarasenko O, Scott A, Soderberg L, Alusta P (2012) Glycoconjugates prevent B. anthracis toxin-induced cell death through binding while activating macrophages. Glycoconj J 29: 25-33
  12. Zaiou M, Rihn B, Bakillah A (2018) Epigenetic regulation of genes involved in the reverse cholesterol transport through interaction with miRNAs. Front Biosci (Landmark Ed) 23: 2090-2105
  13. Otsuka K, Yamamoto Y, Matsuoka R, Ochiya T (2018) Maintaining good miRNAs in the body keeps the doctor away?: Perspectives on the relationship between food‐derived natural products and microRNAs in relation to exosomes/extracellular vesicles. Mol Nutr Food Res 62: 1700080
  14. Huang H, Davis CD, Wang TT (2018) Extensive degradation and low bioavailability of orally consumed corn miRNAs in mice. Nutrients 10: 215
  15. Zhao Q, Mao Q, Zhao Z, Dou T, Wang Z, et al. (2018) Predict plant-derived xenomiRs from plant miRNA sequences using random forest and one-dimensional convolutional neural network models. bioRxiv 2018: 345249
  16. Shukla N, Shukla V, Saxena S (2018) Plant miRNAs and phytomolecules as anticancer therapeutics. Anticancer Plants: Mechanisms and Molecular Interactions. Springer, pp: 27-41
  17. Trumbo P, Schlicker S, Yates AA, Poos M, Food and Nutrition Board of the Institute of Medicine, et al. (2002) Dietary reference intakes for energy, carbohydrate, fiber, fat, fatty acids, cholesterol, protein, and amino acids. J Am Diet Assoc 102: 1621-1630
  18. Slavin J (2003) Impact of the proposed definition of dietary fiber on nutrient databases. J Food Compos Anal 16: 287-291
  19. Sharma P, Bhandari C, Kumar S, Sharma B, Bhadwal P, et al. (2018) Dietary fibers: a way to a healthy microbiome. Diet, Microbiome and Health. Elsevier 11: 299-345
  20. Brouns F, Delzenne N, Gibson G (2017) The dietary fibers–FODMAPs controversy. Cereal Food World 62: 98-103
  21. McRorie JW Jr, McKeown NM (2017) Understanding the physics of functional fibers in the gastrointestinal tract: an evidence-based approach to resolving enduring misconceptions about insoluble and soluble fiber. J Acad Nutr Diet 117: 251-264
  22. Dahl WJ, Stewart ML (2015) Position of the Academy of Nutrition and Dietetics: health implications of dietary fiber. J Acad Nutr Diet 115: 1861-1870
  23. Koh A, De Vadder F, Kovatcheva-Datchary P, Bäckhed F (2016) From dietary fiber to host physiology: short-chain fatty acids as key bacterial metabolites. Cell 165: 1332-1345
  24. Marcobal A, Southwick AM, Earle KA, Sonnenburg JL (2013) A refined palate: bacterial consumption of host glycans in the gut. Glycobiology 23: 1038-1046
  25. Desai MS, Seekatz AM, Koropatkin NM, Kamada N, Hickey CA, et al. (2016) A dietary fiber-deprived gut microbiota degrades the colonic mucus barrier and enhances pathogen susceptibility. Cell 167: 1339-1353
  26. Cantarel BL, Lombard V, Henrissat B (2012) Complex carbohydrate utilization by the healthy human microbiome. PloS one 7: e28742
  27. El Kaoutari A, Armougom F, Gordon JI, Raoult D, Henrissat B (2013) The abundance and variety of carbohydrate-active enzymes in the human gut microbiota. Nat Rev Microbiol 11: 497
  28. Al-Masaudi S, El Kaoutari A, Drula E, Al-Mehdar H, Redwan EM, et al. (2017) A metagenomics investigation of carbohydrate-active enzymes along the gastrointestinal tract of Saudi sheep. Front Microbiol 8: 666
  29. Busch A, Kunert G, Heckel DG, Pauchet Y (2017) Evolution and functional characterization of CAZymes belonging to subfamily 10 of glycoside hydrolase family 5 (GH5_10) in two species of phytophagous beetles. PloS one 12: e0184305
  30. Jain A, Gupta Y, Jain SK (2007) Perspectives of biodegradable natural polysaccharides for site-specific drug delivery to the colon. J Pharm Pharm Sci 10: 86-128
  31. Jones JR, Lineback DM, Levine MJ (2006) Dietary reference intakes: implications for fiber labeling and consumption: a summary of the International Life Sciences Institute North America Fiber Workshop, June 1-2, 2004, Washington, DC. Nutr Rev 64: 31-38
  32. British Nutrition Foundation (1990) Complex carbohydrates in food: the report of the British nutrition foundation's task force. (1st Edn). Chapman and Hall; New York, p: 164
  33. Haralampu S (2000) Resistant starch—a review of the physical properties and biological impact of RS3. Carbohydr Polym 41: 285-292
  34. Nugent AP (2005) Health properties of resistant starch. Nutr Bull 30: 27-54
  35. Sajilata MG, Singhal RS, Kulkarni PR (2006) Resistant starch–a review. Compr Rev Food Sci Food Saf 5: 1-17
  36. Poutanen KS, Dussort P, Erkner A, Fiszman S, Karnik K, et al. (2017) A review of the characteristics of dietary fibers relevant to appetite and energy intake outcomes in human intervention trials. Am J Clin Nutr 106: 747-754
  37. de Vries J, Birkett A, Hulshof T, Verbeke K, Gibes K (2016) Effects of cereal, fruit and vegetable fibers on human fecal weight and transit time: a comprehensive review of intervention trials. Nutrients 8: 130
  38. Sawicki CM, Livingston KA, Obin M, Roberts SB, Chung M, et al. (2017) Dietary fiber and the human gut microbiota: application of evidence mapping methodology. Nutrients 9: 125
  39. Lauc G, Pezer M, Rudan I, Campbell H (2016) Mechanisms of disease: the human N-glycome. Biochim Biophys Acta 1860: 1574-1582
  40. Chassard C, Delmas E, Robert C, Bernalier-Donadille A (2010) The cellulose-degrading microbial community of the human gut varies according to the presence or absence of methanogens. FEMS Microbiol Ecol 74: 205-213
  41. Miyazato S, Kishimoto Y, Takahashi K, Kaminogawa S, Hosono A (2016) Continuous intake of resistant maltodextrin enhanced intestinal immune response through changes in the intestinal environment in mice. Biosci Microbiota Food Health 35: 1-7
  42. Belitz H, Grosch W, Schieberle P (2009) Food Chemistry: 4th revised and extendended Edition. Germany: Springer-Verlag Berlin Heidelberg 53: 377-385
  43. Hamm M, Wang Y, Rustandi RR (2013) Characterization of N-linked glycosylation in a monoclonal antibody produced in NS0 cells using capillary electrophoresis with laser-induced fluorescence detection. Pharmaceuticals 6: 393-406
  44. Jalaludin I, Sudin AH, Said IM, Azizan KA, Baharum SN, et al. (2017) Fluorescence and evaporative light scattering hplc profiling of intracellular asparagine (N)-linked oligosaccharides from Saccharomyces cerevisiae using the alg8 mutant. Malay J Anal Sci 21: 1210-1218
  45. Loziuk PL, Hecht ES, Muddiman DC (2017) N-linked glycosite profiling and use of Skyline as a platform for characterization and relative quantification of glycans in differentiating xylem of Populus trichocarpa. Anal Bioanal Chem 409: 487-497
  46. Kattla J, Struwe W, Doherty M, Adamczyk B, Saldova R, et al. (2011) Protein glycosylation. In: Comprehensive Biotechnology (Murray MY, 2nd Edn). Elsevier, p: 467-486
  47. Lehninger A, Nelson D, Cox M (2008) Lehninger principles of biochemistry (5th Edn). New York: WH Freeman, p: 1158
  48. Varki A, Cummings RD, Esko JD (2017) Essentials of glycobiology (3rd Edn). Cold Spring Harbor Laboratory Press; p: 823
  49. Chandler KB, Pompach P, Goldman R, Edwards N (2013) Exploring site-specific N-glycosylation microheterogeneity of haptoglobin using glycopeptide CID tandem mass spectra and glycan database search. J Proteome Res 12: 3652-3666
  50. Chandler KB, Brnakova Z, Sanda M, Wang S, Stalnaker SH, et al. (2014) Site-specific glycan microheterogeneity of inter-alpha-trypsin inhibitor heavy chain H4. J Proteome Res 13: 3314-3329.
  51. Takahashi K, Wall SB, Suzuki H, Smith AD, Hall S, et al. (2010) Clustered O-glycans of IgA1: defining macro-and micro-heterogeneity by use of electron capture/transfer dissociation. Mol Cell Proteomics 9: 2545-2557.
  52. Briard JG, Jiang H, Moremen KW, Macauley MS, Wu P (2018) Cell-based glycan arrays for probing glycan-glycan binding protein interactions. Nat Commun 9: 880.
  53. Brito A, Reis RL, Pires RA, Pashkuleva I (2018) Sweet building blocks for self-assembling biomaterials with molecular recognition. Self-assembling Biomaterials (1st Edn). Elsevier, pp: 79-94.
  54. Tkachenko E, Rhodes JM, Simons M (2005) Syndecans: new kids on the signaling block. Circ Res 96: 488-500.
  55. Kang I, Chang MY, Wight TN, Frevert CW (2018) Proteoglycans as immunomodulators of the innate immune response to lung infection. J Histochem Cytochem 66: 241-259.
  56. Rédini F (2012) Proteoglycans: methods and protocols (1st Edn). Humana Press, p: 359.
  57. Bolsover SR, Shephard EA, White HA, Hyams JS (2011) Cell biology: a short course (3rd Edn). John Wiley & Sons Ltd, p: 432
  58. Alberts B, Bray D, Hopkin K, Johnson A, Lewis J, et al. (2013) Essential cell biology (4th Edn). Garland Science , p: 723.
  59. Sato S (2018) Cytosolic galectins and their release and roles as carbohydrate-binding proteins in host–pathogen interaction. Trends Glycosci Glycotechnol 30: SE199-SE209.
  60. Hart GW, Copeland RJ (2010) Glycomics hits the big time. Cell 143: 672-676.
  61. Heggelund JE, Varrot A, Imberty A, Krengel U (2017) Histo-blood group antigens as mediators of infections. Curr Opin Struct Biol 44: 190-200.
  62. Lai CH, Hütter J, Hsu CW, Tanaka H, Varela-Aramburu S, et al. (2015) Analysis of carbohydrate–carbohydrate interactions using sugar-functionalized silicon nanoparticles for cell imaging. Nano Lett 16: 807-811.
  63. Moremen KW, Tiemeyer M, Nairn AV (2012) Vertebrate protein glycosylation: diversity, synthesis and function. Nat Rev Mol Cell Biol 13: 448-462.
  64. Crespo HJ, Lau JT, Videira PA (2013) Dendritic cells: a spot on sialic acid. Front Immunol 4: 491.
  65. Van Slambrouck S, Groux-Degroote S, Krzewinski-Recchi MA, Cazet A, Delannoy P, et al. (2014) Carbohydrate-to-carbohydrate interactions between α2, 3-linked sialic acids on α2 integrin subunits and asialo-GM1 underlie the bone metastatic behaviour of LNCAP-derivative C4-2B prostate cancer cells. Biosci Rep 34: e00138.
  66. Coombe DR, Parish CR (2015) Carbohydrates: the yet to be tasted sweet spot of immunity. Front Immunol 6: 314.
  67. Vasta GR (2009) Roles of galectins in infection. Nat Rev Microbiol 7: 424.
  68. Dam TK, Brewer CF (2010) Lectins as pattern recognition molecules: the effects of epitope density in innate immunity. Glycobiology 20: 270-279.
  69. Rienks M, Papageorgiou AP, Frangogiannis NG, Heymans S (2014) Myocardial extracellular matrix: an ever-changing and diverse entity. Circ Res 114: 872-888.
  70. Hynes RO (2009) The extracellular matrix: not just pretty fibrils. Science 326: 1216-1219.
  71. Rhiner C, Hengartner MO (2006) Sugar antennae for guidance signals: syndecans and glypicans integrate directional cues for navigating neurons. Scientific World Journal 6: 1024-1036.
  72. Ghatak S, Maytin EV, Mack JA, Hascall VC, Atanelishvili I, et al. (2015) Roles of proteoglycans and glycosaminoglycans in wound healing and fibrosis. Int J Cell Biol 2015: 20.
  73. Hynes RO, Naba A (2012) Overview of the matrisome-an inventory of extracellular matrix constituents and functions. Cold Spring Harb Perspect Biol 4: a004903.
  74. Bansal MB, Friedman SL (2018) Hepatic fibrogenesis. Sherlock's Diseases of the Liver and Biliary System (13th Edn). Wiley-Blackwell, pp: 82-92.
  75. Módis L (2018) Organization of the extracellular matrix: a polarization microscopic approach (1st Edn). CRC Press, p: 295.
  76. Gabius HJ (2018) The sugar code: why glycans are so important. Biosystems 164: 102-111.
  77. Kračun SK, Fangel JU, Rydahl MG, Pedersen HL, Vidal-Melgosa S, et al. (2017) Carbohydrate microarray technology applied to high-throughput mapping of plant cell wall glycans using comprehensive microarray polymer profiling (CoMPP). Methods Mol Biol 1503: 147-165.
  78. Fanuel M, Garajova S, Ropartz D, McGregor N, Brumer H, et al. (2017) The Podospora anserina lytic polysaccharide monooxygenase Pa LPMO9H catalyzes oxidative cleavage of diverse plant cell wall matrix glycans. Biotechnol Biofuels 10: 63.
  79. Horiuchi R, Nakajima Y, Kashiwada S, Miyanishi N (2018) Effects of silver nanocolloids on plant complex type N-glycans in Oryza sativa roots. Sci Rep 8: 1000.
  80. Voiniciuc C, Pauly M, Usadel B (2018) Monitoring polysaccharide dynamics in the plant cell wall. Plant physiol 176: 2590-2600.
  81. Yeats TH, Bacic A, Johnson KL (2018) Plant glycosylphosphatidylinositol (GPI) anchored proteins at the plasma membrane‐cell wall nexus. J Integr Plant Biol 60: 649-669.
  82. Turner RD, Mesnage S, Hobbs JK, Foster SJ (2018) Molecular imaging of glycan chains couples cell-wall polysaccharide architecture to bacterial cell morphology. Nat Commun 9: 1263.
  83. Radkov AD, Hsu YP, Booher G, VanNieuwenhze MS (2018) Imaging bacterial cell wall biosynthesis. Annu Rev Biochem 87: 991-1014.
  84. Primo ED, Otero LH, Ruiz F, Klinke S, Giordano W (2018) The disruptive effect of lysozyme on the bacterial cell wall explored by an in‐silico structural outlook. Biochem Mol Biol Educ 46: 83-90.
  85. Schaefer K, Owens TW, Kahne D, Walker S (2018) Substrate preferences establish the order of cell wall assembly in Staphylococcus aureus. J Am Chem Soc 140: 2442-2445.
  86. Chaliha C, Rugen MD, Field RA, Kalita E (2018) Glycans as modulators of plant defense against filamentous pathogens. Front Plant Sci 9: 928.
  87. Bacete L, Mélida H, Miedes E, Molina A (2018) Plant cell wall‐mediated immunity: cell wall changes trigger disease resistance responses. Plant J 93: 614-636.
  88. Rydahl MG, Hansen AR, Kračun SK, Mravec J (2018) Report on the current inventory of the toolbox for plant cell wall analysis: proteinaceous and small molecular probes. Front Plant Sci 9: 581.
  89. Hussain S, Wivagg CN, Szwedziak P, Wong F, Schaefer K, et al. (2018) MreB filaments align along greatest principal membrane curvature to orient cell wall synthesis. Elife 7: e32471.
  90. Rohs PA, Buss J, Sim S, Squyres G, Srisuknimit V, et al. (2018) An activation pathway governs cell wall polymerization by a bacterial morphogenic machine. bioRxiv: 359208.
  91. Lee J, Georgiou G (2018) High-affinity IgA against microbial glycans. Nat Immunol 19: 514-515.
  92. Gilar M, Yu YQ, Ahn J, Xie H, Han H, et al. (2011) Characterization of glycoprotein digests with hydrophilic interaction chromatography and mass spectrometry. Anal Biochem 417: 80-88.
  93. Dawson M, Wirtz D, Hanes J (2003) Enhanced viscoelasticity of human cystic fibrotic sputum correlates with increasing microheterogeneity in particle transport. J Biol Chem 278: 50393-50401.
  94. Pritchard LK, Spencer DI, Royle L, Vasiljevic S, Krumm SA, et al. (2015) Glycan microheterogeneity at the PGT135 antibody recognition site on HIV-1 gp120 reveals a molecular mechanism for neutralization resistance. J Virol 89: 6952-6959.
  95. Khatri K, Klein JA, White MR, Grant OC, Leymarie N, et al. (2016) Integrated omics and computational glycobiology reveal structural basis for influenza A virus glycan microheterogeneity and host interactions. Mol Cell Proteomics 15: 1895-1912.
  96. Bergstrom KSB, Xia L (2013) Mucin-type O-glycans and their roles in intestinal homeostasis. Glycobiology 23: 1026-1037.
  97. Gabius HJ, Kayser K (2014) Introduction to glycopathology: the concept, the tools and the perspectives. Diagn Pathol 9: 4.
  98. Ayechu-Muruzabal V, Stigt A, Mank M, Willemsen L, Stahl B, et al. (2018) Diversity of Human Milk Oligosaccharides in Early Life Immune Development. Front Pediatr 6: 239.
  99. Krammer EM, Bouckaert JMJ (2018) Norovirus devours human milk oligosaccharides rich in α-fucose. J Biol Chem 293: 11966-11967
  100. Bode L (2018) Human milk oligosaccharides at the interface of maternal–infant health. Breastfeed Med 13: S7-S8
  101. Triantis V, Bode L, Van Neerven R (2018) Immunological effects of human milk oligosaccharides. Front Pediat 6: 190
  102. Kunz C, Meyer C, Collado MC, Geiger L, García-Mantrana I, et al. (2017) Influence of gestational age, secretor, and lewis blood group status on the oligosaccharide content of human milk. J Pediatr Gastroenterol Nutr 64: 789-798
  103. Martin-Sosa S, Martín MJ, García-Pardo LA, Hueso P (2003) Sialyloligosaccharides in human and bovine milk and in infant formulas: variations with the progression of lactation. J Dairy Sci 86: 52-59
  104. Schnaar RL (2015) Glycans and glycan-binding proteins in immune regulation: a concise introduction to glycobiology for the allergist. J Allergy Clin Immunol 135: 609-615
  105. Van Breedam W, Pöhlmann S, Favoreel HW, de Groot RJ, Nauwynck HJ (2014) Bitter-sweet symphony: glycan–lectin interactions in virus biology. FEMS Microbiol Rev 38: 598-632
  106. Rhodes JM, Milton JD (1998) Lectin methods and protocols. Springer, Humana Press, p: 616
  107. Sharon N (2007) Lectins: carbohydrate-specific reagents and biological recognition molecules. J Biol Chem 282: 2753-2764
  108. Slifkin M, Doyle R (1990) Lectins and their application to clinical microbiology. Clin Microbiol Rev 3: 197-218
  109. Jiang QL, Zhang S, Tian M, Zhang SY, Xie T, et al. (2015) Plant lectins, from ancient sugar‐binding proteins to emerging anti‐cancer drugs in apoptosis and autophagy. Cell Prolif 48: 17-28
  110. Sharon N, Lis H (2004) History of lectins: from hemagglutinins to biological recognition molecules. Glycobiology 14: 53R-62R
  111. Gupta GS (2012) Animal lectins: form, function and clinical applications. Springer Science & Business Media; p: 823
  112. Cockburn DW, Suh C, Medina KP, Duvall RM, Wawrzak Z, et al. (2018) Novel carbohydrate binding modules in the surface anchored α-amylase of Eubacterium rectale provide a molecular rationale for the range of starches used by this organism in the human gut. Mol Microbiol 107: 249-264
  113. Sharon N, Lis H (2007) Lectins (2nd Edn). Dordrecht, NL: Springer; p: 454
  114. Vasta GR, Ahmed H, Nita-Lazar M, Banerjee A, Pasek M, et al. (2012) Galectins as self/non-self recognition receptors in innate and adaptive immunity: an unresolved paradox. Front Immunol 3: 199
  115. Ambrosi M, Cameron NR, Davis BG (2005) Lectins: tools for the molecular understanding of the glycocode. Org Biomol Chem 3: 1593-1608
  116. Yau T, Dan X, Ng CC, Ng TB (2015) Lectins with potential for anti-cancer therapy. Molecules 20: 3791-3810
  117. Anderson K, Evers D, Rice KG (2008) Structure and function of mammalian carbohydrate-lectin interactions. Glycoscience. Springer; p. 2445-2482
  118. Yan H, Kamiya T, Suabjakyong P, Tsuji NM (2015) Targeting C-type lectin receptors for cancer immunity. Front Immunol 6: 408
  119. O’Sullivan JA, Carroll DJ, Bochner BS (2017) Glycobiology of eosinophilic inflammation: contributions of siglecs, glycans, and other glycan-binding proteins. Front Med 4: 116
  120. De Schutter K, Van Damme EJ (2015) Protein-carbohydrate interactions as part of plant defense and animal immunity. Molecules 20: 9029-9053
  121. Kapoor C, Vaidya S, Kaur H, Jain A (2014) Role of lectins in clinical settings. Clin Cancer Investig J 3: 472-477
  122. Iwaki J, Hirabayashi J (2018) Carbohydrate-binding specificity of human galectins: an overview by frontal affinity chromatography. Trends Glycosci Glycotechnol 30: SE137-SE153
  123. Geijtenbeek TB, Gringhuis SI (2009) Signalling through C-type lectin receptors: shaping immune responses. Nat Rev Immunol 9: 465-479
  124. Leffler H, Carlsson S, Hedlund M, Qian Y, Poirier F (2002) Introduction to galectins. Glycoconj J 19: 433-440
  125. Rapoport E, Kurmyshkina O, Bovin N (2008) Mammalian galectins: structure, carbohydrate specificity, and functions. Biochemistry (Mosc) 73: 393-405
  126. Geijtenbeek TB, Torensma R, van Vliet SJ, van Duijnhoven GC, Adema GJ, et al. (2000) Identification of DC-SIGN, a novel dendritic cell–specific ICAM-3 receptor that supports primary immune responses. Cell 100: 575-585
  127. Lugo-Villarino G, Troegeler A, Balboa L, Lastrucci C, Duval C, et al. (2018) The C-type lectin receptor DC-SIGN has an anti-inflammatory role in human M (IL-4) macrophages in response to Mycobacterium tuberculosis. Front Immunol 9: 1123
  128. Kaisar MM, Ritter M, del Fresno C, Jónasdóttir HS, van der Ham AJ, et al. (2018) Dectin-1/2–induced autocrine PGE2 signaling licenses dendritic cells to prime Th2 responses. PLoS Biol 16: e2005504
  129. Crocker PR (2002) Siglecs: sialic-acid-binding immunoglobulin-like lectins in cell–cell interactions and signalling. Curr Opin Struct Biol 12: 609-615
  130. Macauley MS, Crocker PR, Paulson JC (2014) Siglec-mediated regulation of immune cell function in disease. Nat Rev Immunol 14: 653-666
  131. Varki A, Angata T (2005) Siglecs—the major subfamily of I-type lectins. Glycobiology 16: 1R-27R
  132. Angata T, Brinkman-Van der Linden E (2002) I-type lectins. Biochim Biophys Acta 1572: 294-316
  133. Woodfin A, Beyrau M, Voisin MB, Ma B, Whiteford JR, et al. (2016) ICAM-1 expressing neutrophils exhibit enhanced effector functions in murine models of endotoxemia. Blood 127: 898-907
  134. Comrie WA, Li S, Boyle S, Burkhardt JK (2015) The dendritic cell cytoskeleton promotes T cell adhesion and activation by constraining ICAM-1 mobility. J Cell Biol 208: 457-473
  135. Roy R, Murphy PV, Gabius HJ (2016) Multivalent carbohydrate-lectin interactions: How synthetic chemistry enables insights into nanometric recognition. Molecules 21: E629
  136. Linnartz-Gerlach B, Mathews M, Neumann H (2014) Sensing the neuronal glycocalyx by glial sialic acid binding immunoglobulin-like lectins. Neuroscience 275: 113-124
  137. Gabius HJ, Kaltner H, Kopitz J, André S (2015) The glycobiology of the CD system: a dictionary for translating marker designations into glycan/lectin structure and function. Trends Biochem Sci 40: 360-376
  138. Lasky LA (1992) Selectins: interpreters of cell-specific carbohydrate information during inflammation. Science 258: 964-969
  139. Ye ZS, Huang RC (2018) Selectins modify dendritic cells during atherosclerosis. Chron Dis Transl Med 4: In press
  140. Gabius HJ, Manning JC, Kopitz J, André S, Kaltner H (2016) Sweet complementarity: the functional pairing of glycans with lectins. Cell Mol Life Sci 73: 1989-2016
  141. Ledeen RW, Kopitz J, Abad-Rodríguez J, Gabius HJ (2018) Chapter Ten- Glycan chains of gangliosides: functional ligands for tissue lectins (Siglecs/Galectins). In: Progress in Molecular Biology and Translational Science (Schnaar RL, Lopez PH Edn). Academic Press, 156: 289-324
  142. Toledo A, Huang Z, Coleman JL, London E, Benach JL (2018) Lipid rafts can form in the inner and outer membranes of Borrelia burgdorferi and have different properties and associated proteins. Mol Microbiol 108: 63-76
  143. Baynes J, Dominiczak M (2018) Carbohydrates and lipids. Medical Biochemistry (5th Edn). Elsevier 3: 25
  144. Rabinovich GA, Toscano MA, Jackson SS, Vasta GR (2007) Functions of cell surface galectin-glycoprotein lattices. Curr Opin Struct Biol 17: 513-520
  145. Head BP, Patel HH, Insel PA (2014) Interaction of membrane/lipid rafts with the cytoskeleton: impact on signaling and function: membrane/lipid rafts, mediators of cytoskeletal arrangement and cell signaling. Biochim Biophys Acta 1838: 532-545
  146. Hakomori SI (2008) Structure and function of glycosphingolipids and sphingolipids: recollections and future trends. Biochim Biophys Acta 1780: 325-346
  147. Jordan S, Rodgers W (2003) T cell glycolipid-enriched membrane domains are constitutively assembled as membrane patches that translocate to immune synapses. J Immunol 171: 78-87
  148. Hsu DK, Chernyavsky AI, Chen HY, Yu L, Grando SA, et al. (2009) Endogenous galectin-3 is localized in membrane lipid rafts and regulates migration of dendritic cells. J Invest Dermatol 129: 573-583
  149. Sonnino S, Aureli M, Loberto N, Chigorno V, Prinetti A (2010) Fine tuning of cell functions through remodeling of glycosphingolipids by plasma membrane-associated glycohydrolases. FEBS Lett 584: 1914-1922
  150. Cohen M, Varki A (2014) Modulation of glycan recognition by clustered saccharide patches. Int Rev Cell Mol Biol 308: 75-125
  151. Won S, Hindmarsh S, Gibson MI (2018) Triggerable multivalent glyconanoparticles for probing carbohydrate–carbohydrate interactions. ACS Macro Lett 7: 178-183
  152. Sletmoen M, Gerken TA, Stokke BT, Burchell J, Brewer CF (2018) Tn and STn are members of a family of carbohydrate tumor antigens that possess carbohydrate–carbohydrate interactions. Glycobiology 28: 437-442
  153. Xiong Y, Li M, Wang H, Qing G, Sun T (2018) Sialic acid-triggered macroscopic properties switching on a smart polymer surface. Appl Surf Sci 427: 1152-1164
  154. Qi W, Zhang Y, Wang J, Tao G, Wu L, et al. (2018) Deprotection-induced morphology transition and immuno-activation of glyco-vesicles: a strategy of smart delivery polymersomes. J Am Chem Soc 140: 8851-8857
  155. Van Ree R, Cabanes-Macheteau M, Akkerdaas J, Milazzo JP, Loutelier-Bourhis C, et al. (2000) β(1,2)-xylose and α(1,3)-fucose residues have a strong contribution in IgE binding to plant glycoallergens. J Biol Chem 275: 11451-11458
  156. Aalberse RC, Crameri R (2011) IgE‐binding epitopes: a reappraisal. Allergy 66: 1261-1274
  157. Aalberse RC, Van Ree R (1997) Crossreactive carbohydrate determinants. Clin Rev Allergy Immunol 15: 375
  158. Wilson IBH, Harthill JE, Mullin NP, Ashford DA, Altmann F (1998) Core α1,3-fucose is a key part of the epitope recognized by antibodies reacting against plant N-linked oligosaccharides and is present in a wide variety of plant extracts. Glycobiology 8: 651-661
  159. Wilson IBH, Zeleny R, Kolarich D, Staudacher E, Stroop CJM, et al. (2001) Analysis of Asn-linked glycans from vegetable foodstuffs: widespread occurrence of Lewis a, core α1,3-linked fucose and xylose substitutions. Glycobiology 11: 261-274
  160. Mari A, Iacovacci P, Afferni C, Barletta B, Tinghino R, et al. (1999) Specific IgE to cross-reactive carbohydrate determinants strongly affect the in vitro diagnosis of allergic diseases. J Allergy Clin Immunol 103: 1005-1011
  161. Jin C, Hantusch B, Hemmer W, Stadlmann J, Altmann F (2008) Affinity of IgE and IgG against cross-reactive carbohydrate determinants on plant and insect glycoproteins. J Allergy Clin Immunol 121: 185-190
  162. Wilson IBH, Altmann F (1998) Structural analysis of N-glycans from allergenic grass, ragweed and tree pollens: core α1,3-linked fucose and xylose present in all pollens examined. Glycoconj J 15: 1055-1070
  163. Ebo DG, Hagendorens MM, Bridts CH, De Clerck LS, Stevens WJ (2004) Sensitization to cross‐reactive carbohydrate determinants and the ubiquitous protein profilin: mimickers of allergy. Clin Exp Allergy 34: 137-144
  164. Soh JY, Huang CH, Lee BW (2015) Carbohydrates as food allergens. Asia Pac Allergy 5: 17-24
  165. Le Parc A, Lee H, Chen K, Barile D (2014) Rapid quantification of functional carbohydrates in food products. Food Nutr Sci 5: 71
  166. Weber JA, Baxter DH, Zhang S, Huang DY, Huang KH, et al. (2010) The microRNA spectrum in 12 body fluids. Clin Chem 56: 1733-1741
  167. Cenit MC, Olivares M, Codoñer-Franch P, Sanz Y (2015) Intestinal microbiota and celiac disease: cause, consequence or co-evolution? Nutrients 7: 6900-6923
  168. Nakata K, Sugi Y, Narabayashi H, Kobayakawa T, Nakanishi Y, et al. (2017) Commensal microbiota-induced microRNA modulates intestinal epithelial permeability through a small GTPase ARF4. J Biol Chem 292: 15426-15433
  169. Djuranovic S, Nahvi A, Green R (2012) miRNA-mediated gene silencing by translational repression followed by mRNA deadenylation and decay. Science 336: 237-240
  170. Jing Q, Huang S, Guth S, Zarubin T, Motoyama A, et al. (2005) Involvement of microRNA in AU-rich element-mediated mRNA instability. Cell 120: 623-634
  171. Taganov KD, Boldin MP, Baltimore D (2007) MicroRNAs and immunity: tiny players in a big field. Immunity 26: 133-137
  172. Dalmasso G, Nguyen HT, Yan Y, Laroui H, Charania MA, et al. (2011) Microbiota modulate host gene expression via microRNAs. PLoS One 6: e19293
  173. Ambros V, Chen X (2007) The regulation of genes and genomes by small RNAs. Development 134: 1635-1641
  174. Kim VN, Han J, Siomi MC (2009) Biogenesis of small RNAs in animals. Nat Rev Mol Cell Biol 10: 126-139
  175. Gavrilas LI, Ionescu C, Tudoran O, Lisencu C, Balacescu O, et al. (2016) The role of bioactive dietary components in modulating miRNA expression in colorectal cancer. Nutrients 8: E590
  176. Zhang J, Li S, Li L, Li M, Guo C, et al. (2015) Exosome and exosomal microRNA: trafficking, sorting, and function. Genomics Proteomics Bioinformatics 13: 17-24
  177. Lee J, Park EJ, Kiyono H (2016) MicroRNA-orchestrated pathophysiologic control in gut homeostasis and inflammation. BMB Rep 49: 263
  178. Carotenuto F, Albertini MC, Coletti D, Vilmercati A, Campanella L, et al. (2016) How diet intervention via modulation of DNA damage response through microRNAs may have an effect on cancer prevention and aging, an in silico study. Int J Mol Sci 17: 752
  179. Izumi H, Kosaka N, Shimizu T, Sekine K, Ochiya T, et al. (2012) Bovine milk contains microRNA and messenger RNA that are stable under degradative conditions. J Dairy Sci 95: 4831-4841
  180. Modepalli V, Kumar A, Hinds LA, Sharp JA, Nicholas KR, et al. (2014) Differential temporal expression of milk miRNA during the lactation cycle of the marsupial tammar wallaby (Macropus eugenii). BMC Genomics 15: 1012
  181. Melnik BC, Kakulas F, Geddes DT, Hartmann PE, John SM, et al. (2016) Milk miRNAs: simple nutrients or systemic functional regulators? Nutr Metab 13: 42
  182. Zhou Q, Li M, Wang X, Li Q, Wang T, et al. (2012) Immune-related microRNAs are abundant in breast milk exosomes. Int J Biol Sci 8: 118-123
  183. Yu B, Yang Z, Li J, Minakhina S, Yang M, et al. (2005) Methylation as a crucial step in plant microRNA biogenesis. Science 307: 932-935
  184. Li J, Yang Z, Yu B, Liu J, Chen X (2005) Methylation protects miRNAs and siRNAs from a 3'-end uridylation activity in Arabidopsis. Curr Biol 15: 1501-1507
  185. Chen X, Dai GH, Ren ZM, Tong YL, Yang F, et al. (2016) Identification of dietetically absorbed rapeseed (Brassica campestris L.) bee pollen microRNAs in serum of mice. Biomed Res Int 2016: 5413849
  186. Guyton AC, Hall JE (2006) Guyton and Hall textbook of medical physiology (11th Edn). Philadelphia, US: Elsevier Saunders; p: 1116
  187. Dane S, Hänninen O (2009) Enzymes of digestion. Physiology and Maintenance-Volume II: Enzymes: The Biological Catalysts of Life, Nutrition and Digestion. EOLSS Publications, pp: 45-60
  188. Shaw W (2002) Abnormalities of the digestive system: gluten and casein, peptides, secretin, CCK, and pancreatic atrophy. Biological treatments for autism and PDD. The Great Plains Laboratory, Inc. 6: 74
  189. Ji L, Chen X (2012) Regulation of small RNA stability: methylation and beyond. Cell Res 22: 624-636
  190. Witwer KW (2012) XenomiRs and miRNA homeostasis in health and disease: evidence that diet and dietary miRNAs directly and indirectly influence circulating miRNA profiles. RNA Biol 9: 1147-1154
  191. Baier SR, Nguyen C, Xie F, Wood JR, Zempleni J (2014) MicroRNAs are absorbed in biologically meaningful amounts from nutritionally relevant doses of cow milk and affect gene expression in peripheral blood mononuclear cells, HEK-293 kidney cell cultures, and mouse livers. J Nutr 144: 1495-1500
  192. Dickinson B, Zhang Y, Petrick JS, Heck G, Ivashuta S, et al. (2013) Lack of detectable oral bioavailability of plant microRNAs after feeding in mice. Nat Biotechnol 31: 965-967
  193. Kang W, Bang-Berthelsen CH, Holm A, Houben AJS, Müller AH, et al. (2017) Survey of 800+ data sets from human tissue and body fluid reveals xenomiRs are likely artifacts. RNA 23: 433-445
  194. Yang J, Hirschi KD, Farmer LM (2015) Dietary RNAs: new stories regarding oral delivery. Nutrients 7: 3184-3199
  195. Zhao Q, Liu Y, Zhang N, Hu M, Zhang H, et al. (2018) Evidence for plant-derived xenomiRs based on a large-scale analysis of public small RNA sequencing data from human samples. PloS One 13: e0187519
  196. Aouadi M, Tesz GJ, Nicoloro SM, Wang M, Chouinard M, et al. (2009) Orally delivered siRNA targeting macrophage Map4k4 suppresses systemic inflammation. Nature 458: 1180
  197. Duval M, Cossart P, Lebreton A (2017) Mammalian microRNAs and long noncoding RNAs in the host-bacterial pathogen crosstalk. Semin Cell Dev Biol 65: 11-19
  198. Friedman RC, Farh KK, Burge CB, Bartel DP (2009) Most mammalian mRNAs are conserved targets of microRNAs. Genome Res 19: 92-105
  199. Santosh B, Varshney A, Yadava P (2015) Non-coding RNAs: Biological functions and applications. Cell Biochem Funct 33: 14-22
  200. Round JL, Mazmanian SK (2009) The gut microbiota shapes intestinal immune responses during health and disease. Nat Rev Immunol 9: 313-323
  201. Org E, Parks BW, Joo JW, Emert B, Schwartzman W, et al. (2015) Genetic and environmental control of host-gut microbiota interactions. Genome Res 25: 1558-1569
  202. Cho I, Blaser MJ (2012) The human microbiome: at the interface of health and disease. Nat Rev Genet 13: 260-270
  203. Frosali S, Pagliari D, Gambassi G, Landolfi R, Pandolfi F, et al. (2015) How the intricate interaction among toll-like receptors, microbiota, and intestinal immunity can influence gastrointestinal pathology. J Immunol Res 2015: 489821
  204. Giongo A, Gano KA, Crabb DB, Mukherjee N, Novelo LL, et al. (2011) Toward defining the autoimmune microbiome for type 1 diabetes. ISME J 5: 82-91
  205. McKenna P, Hoffmann C, Minkah N, Aye PP, Lackner A, et al. (2008) The macaque gut microbiome in health, lentiviral infection, and chronic enterocolitis. PLoS Pathog 4: e20
  206. Stecher B (2015) The roles of inflammation, nutrient availability and the commensal microbiota in enteric pathogen infection. Microbiol Spectr 3: 3
  207. Claesson MJ, Jeffery IB, Conde S, Power SE, O’connor EM, et al. (2012) Gut microbiota composition correlates with diet and health in the elderly. Nature 488: 178-184
  208. Roberts MA, Mutch DM, German JB (2001) Genomics: food and nutrition. Curr Opin Biotechnol 12: 516-522
  209. O'Hara AM, Shanahan F (2006) The gut flora as a forgotten organ. EMBO Rep 7: 688-693
  210. Leppkes M, Siegmund B, Becker C (2018) Editorial: Immune-epithelial crosstalk in inflammatory bowel diseases and mucosal wound healing. Front Immunol 9: 1171
  211. Ubeda C, Djukovic A, Isaac S (2017) Roles of the intestinal microbiota in pathogen protection. Clin Transl Immunol 6: e128
  212. Cruchley AT, Bergmeier LA (2018) Structure and functions of the oral mucosa. In: Oral Mucosa in Health and Disease. Springer; pp: 1-18
  213. Banerjee A, McKinley ET, von Moltke J, Coffey RJ, Lau KS (2018) Interpreting heterogeneity in intestinal tuft cell structure and function. J Clin Invest 128: 1711-1719
  214. Neurath MF (2018) Intestinal inflammation and its relevance for the liver. Liver-Gut-Microbiome Interactions Workshop
  215. Allen A (2018) The structure and function of gastrointestinal mucus. In: Attachment of organisms to the gut mucosa (Boedeker EC Edn). CRC Press; pp: 3-12
  216. Oteiza P, Fraga C, Mills D, Taft D (2018) Flavonoids and the gastrointestinal tract: local and systemic effects. Mol Aspects Med 61: 41-49
  217. Kumar V, Abbas A, Aster J (2013) Robbins basic pathology (9th Edn). Philadelphia, PA: Elsevier/Saunders; p: 910
  218. Montalban-Arques A, Chaparro M, Gisbert JP, Bernardo D (2018) The innate immune system in the gastrointestinal tract: role of intraepithelial lymphocytes and lamina propria innate lymphoid cells in intestinal inflammation. Inflamm Bowel Dis 24: 1649-1659
  219. Schiavi E, Smolinska S, O’Mahony L (2015) Intestinal dendritic cells. Curr Opin Gastroenterol 31: 98-103
  220. Kelsall B (2008) Recent progress in understanding the phenotype and function of intestinal dendritic cells and macrophages. Mucosal Immunol 1: 460-469
  221. Macpherson AJ, Uhr T (2004) Induction of protective IgA by intestinal dendritic cells carrying commensal bacteria. Science 303: 1662-1665
  222. Belkaid Y, Hand TW (2014) Role of the microbiota in immunity and inflammation. Cell 157: 121-141
  223. Tian R, Chen M, Chai S, Rong X, Chen B, et al. (2018) Divergent selection of pattern recognition receptors in mammals with different ecological characteristics. J Mol Evol 86: 138-149
  224. Hardison SE, Brown GD (2012) C-type lectin receptors orchestrate antifungal immunity. Nat Immunol 13: 817-822
  225. Zeng M, Inohara N, Nuñez G (2017) Mechanisms of inflammation-driven bacterial dysbiosis in the gut. Mucosal Immunol 10: 18-26
  226. Martinon F, Mayor A, Tschopp J (2009) The inflammasomes: guardians of the body. Ann Rev Immunol 27: 229-265
  227. Latz E, Xiao TS, Stutz A (2013) Activation and regulation of the inflammasomes. Nat Rev Immunol 13: 397-411
  228. McGuckin MA, Lindén SK, Sutton P, Florin TH (2011) Mucin dynamics and enteric pathogens. Nat Rev Microbiol 9: 265-278
  229. Hooper LV, Macpherson AJ (2010) Immune adaptations that maintain homeostasis with the intestinal microbiota. Nat Rev Immunol 10: 159-169
  230. Cash HL, Whitham CV, Behrendt CL, Hooper LV (2006) Symbiotic bacteria direct expression of an intestinal bactericidal lectin. Science 313: 1126-1130
  231. Brandl K, Plitas G, Schnabl B, DeMatteo RP, Pamer EG (2007) MyD88-mediated signals induce the bactericidal lectin RegIIIγ and protect mice against intestinal Listeria monocytogenes infection. J Exp Med 204: 1891-1900
  232. Ismail AS, Severson KM, Vaishnava S, Behrendt CL, Yu X, et al. (2011) γδ intraepithelial lymphocytes are essential mediators of host–microbial homeostasis at the intestinal mucosal surface. Proc Natl Acad Sci U S A 108: 8743-8748
  233. Mukherjee S, Partch CL, Lehotzky RE, Whitham CV, Chu H, et al. (2009) Regulation of C-type lectin antimicrobial activity by a flexible N-terminal prosegment. J Biol Chem 284: 4881-4888
  234. Vaishnava S, Yamamoto M, Severson KM, Ruhn KA, Yu X, et al. (2011) The antibacterial lectin RegIIIγ promotes the spatial segregation of microbiota and host in the intestine. Science 334: 255-258
  235. Ivanov II (2017) Microbe hunting hits home. Cell Host Microbe 21: 282-285
  236. Abraham C, Medzhitov R (2011) Interactions between the host innate immune system and microbes in inflammatory bowel disease. Gastroenterology 140: 1729-1737
  237. Earle KA, Billings G, Sigal M, Lichtman JS, Hansson GC, et al. (2015) Quantitative imaging of gut microbiota spatial organization. Cell Host Microbe 18: 478-488
  238. Fletcher CM, Coyne MJ, Villa OF, Chatzidaki-Livanis M, Comstock LE (2009) A general O-glycosylation system important to the physiology of a major human intestinal symbiont. Cell 137: 321-331
  239. Sekirov I, Finlay BB (2009) The role of the intestinal microbiota in enteric infection. J Physiol 587: 4159-4167
  240. Werner GD, Cornelissen JH, Cornwell WK, Soudzilovskaia NA, Kattge J, et al. (2018) Symbiont switching and alternative resource acquisition strategies drive mutualism breakdown. Proc Natl Acad Sci U S A 115: 5229-5234
  241. Makki K, Deehan EC, Walter J, Bäckhed F (2018) The impact of dietary fiber on gut microbiota in host health and disease. Cell Host Microbe 23: 705-715
  242. Zilber-Rosenberg I, Rosenberg E (2008) Role of microorganisms in the evolution of animals and plants: the hologenome theory of evolution. FEMS Microbiol Rev 32: 723-735
  243. Hooper LV, Midtvedt T, Gordon JI (2002) How host-microbial interactions shape the nutrient environment of the mammalian intestine. Ann Rev Nutr 22: 283-307
  244. Brunton LL, Chabner B, Knollmann BC (2011) Goodman & Gilman's the pharmacological basis of therapeutics (12th Edn). Mcgraw-Hill Education – Europe; p: 1808
  245. Koropatkin NM, Cameron EA, Martens EC (2012) How glycan metabolism shapes the human gut microbiota. Nat Rev Microbiol 10: 323-335
  246. Turnbaugh PJ, Ley RE, Mahowald MA, Magrini V, Mardis ER, et al. (2006) An obesity-associated gut microbiome with increased capacity for energy harvest. Nature 444: 1027-1031
  247. Samuel BS, Gordon JI (2006) A humanized gnotobiotic mouse model of host–archaeal–bacterial mutualism. Proc Natl Acad Sci U S A 103: 10011-10016
  248. Abuajah CI, Ogbonna AC, Osuji CM (2015) Functional components and medicinal properties of food: a review. J Food Sci Technol 52: 2522-2529
  249. Ndeh D, Gilbert HJ (2018) Biochemistry of complex glycan depolymerisation by the human gut microbiota. FEMS Microbiol Rev 42: 146-164
  250. Martens EC, Lowe EC, Chiang H, Pudlo NA, Wu M, et al. (2011) Recognition and degradation of plant cell wall polysaccharides by two human gut symbionts. PLoS Biol 9: e1001221
  251. Sheridan PO, Martin JC, Lawley TD, Browne HP, Harris HM, et al. (2016) Polysaccharide utilization loci and nutritional specialization in a dominant group of butyrate-producing human colonic Firmicutes. Microb Genom 2: e000043
  252. Soverini M, Turroni S, Biagi E, Quercia S, Brigidi P, et al. (2017) Variation of carbohydrate-active enzyme patterns in the gut microbiota of Italian healthy subjects and type 2 diabetes patients. Front Microbiol 8: 2079
  253. Barratt MJ, Lebrilla C, Shapiro HY, Gordon JI (2017) The gut microbiota, food science, and human nutrition: a timely marriage. Cell Host Microbe 22: 134-141
  254. Monge EC, Tuveng TR, Vaaje-Kolstad G, Eijsink VGH, Gardner JG (2018) Systems analysis of the family Glycoside Hydrolase family 18 enzymes from Cellvibrio japonicus characterizes essential chitin degradation functions. J Biol Chem 293: 3849-3859
  255. Little DJ, Pfoh R, Le Mauff F, Bamford NC, Notte C, et al. (2018) PgaB orthologues contain a glycoside hydrolase domain that cleaves deacetylated poly-β(1,6)-N-acetylglucosamine and can disrupt bacterial biofilms. PLoS Pathog 14: e1006998
  256. Valdes AM, Walter J, Segal E, Spector TD (2018) Role of the gut microbiota in nutrition and health. BMJ 361: k2179
  257. Rodríguez-Díaz J, García-Mantrana I, Vila-Vicent S, Gozalbo-Rovira R, Buesa J, et al. (2017) Relevance of secretor status genotype and microbiota composition in susceptibility to rotavirus and norovirus infections in humans. Sci Rep 7: 45559
  258. Martens EC, Chiang HC, Gordon JI (2008) Mucosal glycan foraging enhances fitness and transmission of a saccharolytic human gut bacterial symbiont. Cell Host Microbe 4: 447-457
  259. Salyers AA, O'Brien M, Kotarski SF (1982) Utilization of chondroitin sulfate by Bacteroides thetaiotaomicron growing in carbohydrate-limited continuous culture. J Bacteriol 150: 1008-1015
  260. Salyers A, Vercellotti J, West S, Wilkins T (1977) Fermentation of mucin and plant polysaccharides by strains of Bacteroides from the human colon. Appl Environ Microbiol 33: 319-322
  261. Cheng Q, Salyers AA (1995) Use of suppressor analysis to find genes involved in the colonization deficiency of a Bacteroides thetaiotaomicron mutant unable to grow on the host-derived mucopolysaccharides chondroitin sulfate and heparin. Appl Environ Microbiol 61: 734-740
  262. Sonnenburg ED, Zheng H, Joglekar P, Higginbottom SK, Firbank SJ, et al. (2010) Specificity of polysaccharide use in intestinal bacteroides species determines diet-induced microbiota alterations. Cell 141: 1241-1252
  263. Laugerette F, Vors C, Peretti N, Michalski MC (2011) Complex links between dietary lipids, endogenous endotoxins and metabolic inflammation. Biochimie 93: 39-45
  264. Jin C, Flavell RA (2013) Innate sensors of pathogen and stress: linking inflammation to obesity. J Allergy Clin Immunol 132: 287-294
  265. Garrett WS, Gordon JI, Glimcher LH (2010) Homeostasis and inflammation in the intestine. Cell 140: 859-870
  266. Hooper LV, Stappenbeck TS, Hong CV, Gordon JI (2003) Angiogenins: a new class of microbicidal proteins involved in innate immunity. Nature Immunol 4: 269-273
  267. Mazmanian SK, Liu CH, Tzianabos AO, Kasper DL (2005) An immunomodulatory molecule of symbiotic bacteria directs maturation of the host immune system. Cell 122: 107-118
  268. Imhann F, Vila AV, Bonder MJ, Fu J, Gevers D, et al. (2018) Interplay of host genetics and gut microbiota underlying the onset and clinical presentation of inflammatory bowel disease. Gut 67: 108-119
  269. Kriss M, Hazleton KZ, Nusbacher NM, Martin CG, Lozupone CA (2018) Low diversity gut microbiota dysbiosis: drivers, functional implications and recovery. Curr Opin Microbiol 44: 34-40
  270. Gilbert JA, Blaser MJ, Caporaso JG, Jansson JK, Lynch SV, et al. (2018) Current understanding of the human microbiome. Nature Med 24: 392-400
  271. Goodman AL, McNulty NP, Zhao Y, Leip D, Mitra RD, et al. (2009) Identifying genetic determinants needed to establish a human gut symbiont in its habitat. Cell Host Microbe 6: 279-289
  272. Tremaroli V, Bäckhed F (2012) Functional interactions between the gut microbiota and host metabolism. Nature 489: 242-249
  273. Dicksved J, Halfvarson J, Rosenquist M, Järnerot G, Tysk C, et al. (2008) Molecular analysis of the gut microbiota of identical twins with Crohn's disease. ISME J 2: 716-727
  274. Willing BP, Dicksved J, Halfvarson J, Andersson AF, Lucio M, et al. (2010) A pyrosequencing study in twins shows that gastrointestinal microbial profiles vary with inflammatory bowel disease phenotypes. Gastroenterology 139: 1844-1854
  275. Lepage P, Häsler R, Spehlmann ME, Rehman A, Zvirbliene A, et al. (2011) Twin study indicates loss of interaction between microbiota and mucosa of patients with ulcerative colitis. Gastroenterology 141: 227-236
  276. Tims S, Derom C, Jonkers DM, Vlietinck R, Saris WH, et al. (2013) Microbiota conservation and BMI signatures in adult monozygotic twins. ISME J 7: 707-717
  277. Kassinen A, Krogius-Kurikka L, Mäkivuokko H, Rinttilä T, Paulin L, et al. (2007) The fecal microbiota of irritable bowel syndrome patients differs significantly from that of healthy subjects. Gastroenterology 133: 24-33
  278. Biagi E, Nylund L, Candela M, Ostan R, Bucci L, et al. (2010) Through ageing, and beyond: gut microbiota and inflammatory status in seniors and centenarians. PloS One 5: e10667
  279. Pickard JM, Zeng MY, Caruso R, Núñez G (2017) Gut microbiota: role in pathogen colonization, immune responses, and inflammatory disease. Immunol Rev 279: 70-89
  280. Bolnick DI, Snowberg LK, Hirsch PE, Lauber CL, Org E, et al. (2014) Individual diet has sex-dependent effects on vertebrate gut microbiota. Nat Commun 5: 4500
  281. Roberfroid M, Gibson GR, Hoyles L, McCartney AL, Rastall R, et al. (2010) Prebiotic effects: metabolic and health benefits. Br J Nutr 104: S1-S63
  282. Hooper LV, Littman DR, Macpherson AJ (2012) Interactions between the microbiota and the immune system. Science 336: 1268-1273
  283. Maynard CL, Elson CO, Hatton RD, Weaver CT (2012) Reciprocal interactions of the intestinal microbiota and immune system. Nature 489: 231-241
  284. Petnicki-Ocwieja T, Hrncir T, Liu Y-J, Biswas A, Hudcovic T, et al. (2009) Nod2 is required for the regulation of commensal microbiota in the intestine. Proc Natl Acad Sci U S A 106: 15813-15818
  285. Bishop JR, Gagneux P (2007) Evolution of carbohydrate antigens—microbial forces shaping host glycomes? Glycobiology 17: 23R-34R
  286. Stowell SR, Arthur CM, Dias-Baruffi M, Rodrigues LC, Gourdine JP, et al. (2010) Innate immune lectins kill bacteria expressing blood group antigen. Nature Med 16: 295-301
  287. Cleveland MG, Gorham JD, Murphy TL, Tuomanen E, Murphy KM (1996) Lipoteichoic acid preparations of gram-positive bacteria induce interleukin-12 through a CD14-dependent pathway. Infect Immun 64: 1906-1912
  288. Koyama N, Inokoshi J, Tomoda H (2013) Anti-infectious agents against MRSA. Molecules 18: 204-224
  289. Shen Y, Torchia Maria Letizia G, Lawson Gregory W, Karp Christopher L, Ashwell Jonathan D, et al. (2012) Outer membrane vesicles of a human commensal mediate immune regulation and disease protection. Cell Host Microbe 12: 509-520
  290. Schwandner R, Dziarski R, Wesche H, Rothe M, Kirschning CJ (1999) Peptidoglycan-and lipoteichoic acid-induced cell activation is mediated by toll-like receptor 2. J Biol Chem 274: 17406-1709
  291. Day CJ, Tran EN, Semchenko EA, Tram G, Hartley-Tassell LE, et al. (2015) Glycan: glycan interactions: high affinity biomolecular interactions that can mediate binding of pathogenic bacteria to host cells. Proc Natl Acad Sci U S A 112: E7266-E7275
  292. Lee YK, Mazmanian SK (2014) Microbial learning lessons: SFB educate the immune system. Immunity 40: 457-459
  293. Wang PG, Guo H, Yi W, Song JK (2008) Current understanding on biosynthesis of microbial polysaccharides. Curr Top Med Chem 8: 141-151
  294. Taylor CM, Roberts IS (2005) Capsular polysaccharides and their role in virulence. Contrib Microbiol 12: 55-66
  295. Comstock LE, Kasper DL (2006) Bacterial glycans: key mediators of diverse host immune responses. Cell 126: 847-850
  296. Mazmanian SK, Kasper DL (2006) The love–hate relationship between bacterial polysaccharides and the host immune system. Nat Rev Immunol 6: 849-858
  297. Comstock LE (2009) Importance of glycans to the host-bacteroides mutualism in the mammalian intestine. Cell Host Microbe 5: 522-526
  298. Geurtsen J, Chedammi S, Mesters J, Cot M, Driessen NN, et al. (2009) Identification of mycobacterial α-glucan as a novel ligand for DC-SIGN: involvement of mycobacterial capsular polysaccharides in host immune modulation. J Immunol 183: 5221-5231
  299. Donaldson GP, Ladinsky MS, Yu KB, Sanders JG, Yoo BB, et al. (2018) Gut microbiota utilize immunoglobulin A for mucosal colonization. Science 360: 795-800
  300. Lebeer S, Vanderleyden J, De Keersmaecker SC (2010) Host interactions of probiotic bacterial surface molecules: comparison with commensals and pathogens. Nat Rev Microbiol 8: 171-184
  301. Surana NK, Kasper DL (2011) The yin yang of bacterial polysaccharides: lessons learned from B. fragilis PSA. Immunol Rev 245: 13-26
  302. Neff CP, Rhodes ME, Arnolds KL, Collins CB, Donnelly J, et al. (2016) Diverse Intestinal Bacteria Contain Putative Zwitterionic Capsular Polysaccharides with Anti-inflammatory Properties. Cell Host Microbe 20: 535-547
  303. Kashyap PC, Marcobal A, Ursell LK, Larauche M, Duboc H, et al. (2013) Complex interactions among diet, gastrointestinal transit, and gut microbiota in humanized mice. Gastroenterology 144: 967-977
  304. Garcia-Mantrana I, Selma-Royo M, Alcantara C, Collado MC (2018) Shifts on gut microbiota associated to Mediterranean diet adherence and specific dietary intakes on general adult population. Front Microbiol 9: 890
  305. Rakoff-Nahoum S, Coyne MJ, Comstock LE (2014) An ecological network of polysaccharide utilization among human intestinal symbionts. Curr Biol 24: 40-49
  306. Gaskins HR, Croix JA, Nakamura N, Nava GM (2008) Impact of the intestinal microbiota on the development of mucosal defense. Clin Infect Dis 46: S80-S86
  307. McNulty NP, Wu M, Erickson AR, Pan C, Erickson BK, et al. (2013) Effects of diet on resource utilization by a model human gut microbiota containing Bacteroides cellulosilyticus WH2, a symbiont with an extensive glycobiome. PLoS Biol 11: e1001637
  308. Martens EC, Koropatkin NM, Smith TJ, Gordon JI (2009) Complex glycan catabolism by the human gut microbiota: the Bacteroidetes Sus-like paradigm. J Biol Chem 284: 24673-24677
  309. Bjursell MK, Martens EC, Gordon JI (2006) Functional genomic and metabolic studies of the adaptations of a prominent adult human gut symbiont, bacteroides thet aiotaomicron, to the suckling period. J Biol Chem 281: 36269-3679
  310. Hugenholtz F, de Vos WM (2018) Mouse models for human intestinal microbiota research: a critical evaluation. Cell Mol Life Sci 75: 149-160
  311. Gill SR, Pop M, DeBoy RT, Eckburg PB, Turnbaugh PJ, et al. (2006) Metagenomic analysis of the human distal gut microbiome. Science. 312: 1355-1359
  312. Lee FJ, Miller KI, McKinlay JB, Newton IL (2018) Differential carbohydrate utilization and organic acid production by honey bee symbionts. FEMS Microbiol Ecol 94: 113
  313. Wacklin P, Tuimala J, Nikkilä J, Tims S, Mäkivuokko H, et al. (2014) Faecal microbiota composition in adults is associated with the FUT2 gene determining the secretor status. PloS one 9: e94863
  314. Ley RE, Bäckhed F, Turnbaugh P, Lozupone CA, Knight RD, et al. (2005) Obesity alters gut microbial ecology. Proc Natl Acad Sci U S A 102: 11070-11075
  315. Ley RE, Turnbaugh PJ, Klein S, Gordon JI (2006) Microbial ecology: human gut microbes associated with obesity. Nature 444: 1022-1023
  316. Duncan SH, Belenguer A, Holtrop G, Johnstone AM, Flint HJ, et al. (2007) Reduced dietary intake of carbohydrates by obese subjects results in decreased concentrations of butyrate and butyrate-producing bacteria in feces. Appl Environ Microbiol 73: 1073-1078
  317. Schwiertz A, Taras D, Schäfer K, Beijer S, Bos NA, et al. (2010) Microbiota and SCFA in lean and overweight healthy subjects. Obesity 18: 190-195
  318. Hildebrandt MA, Hoffmann C, Sherrill–Mix SA, Keilbaugh SA, Hamady M, et al. (2009) High-fat diet determines the composition of the murine gut microbiome independently of obesity. Gastroenterology 137: 1716-1724.
  319. Burcelin R, Crivelli V, Dacosta A, Roy-Tirelli A, Thorens B (2002) Heterogeneous metabolic adaptation of C57BL/6J mice to high-fat diet. Am J Physiol Endocrinol Metab 282: E834-E842
  320. Serino M, Luche E, Gres S, Baylac A, Bergé M, et al. (2011) Metabolic adaptation to a high-fat diet is associated with a change in the gut microbiota. Gut 61: 543-553
  321. Sonnenburg ED, Smits SA, Tikhonov M, Higginbottom SK, Wingreen NS, et al. (2016) Diet-induced extinctions in the gut microbiota compound over generations. Nature 529: 212-215
  322. De Filippo C, Cavalieri D, Di Paola M, Ramazzotti M, Poullet JB, et al. (2010) Impact of diet in shaping gut microbiota revealed by a comparative study in children from Europe and rural Africa. Proc Natl Acad Sci U S A 107: 14691-14696
  323. Wu GD, Chen J, Hoffmann C, Bittinger K, Chen YY, et al. (2011) Linking long-term dietary patterns with gut microbial enterotypes. Science 334: 105-108
  324. David LA, Maurice CF, Carmody RN, Gootenberg DB, Button JE, et al. (2014) Diet rapidly and reproducibly alters the human gut microbiome. Nature 505: 559-563
  325. Thursby E, Juge N (2017) Introduction to the human gut microbiota. Biochem J 474: 1823-1836
  326. Koutsos A, Tuohy KM, Lovegrove JA (2015) Apples and cardiovascular health—is the gut microbiota a core consideration? Nutrients 7: 3959-3998
  327. Glick-Bauer M, Yeh MC (2014) The health advantage of a vegan diet: exploring the gut microbiota connection. Nutrients 6: 4822-4838
  328. Sela DA, Mills DA (2014) The marriage of nutrigenomics with the microbiome: the case of infant-associated bifidobacteria and milk. Am J Clin Nutr 99: 697S-703S
  329. Garrido D, Barile D, Mills DA (2012) A molecular basis for bifidobacterial enrichment in the infant gastrointestinal tract. Adv Nutr 3: 415S-421S
  330. Walker AW, Ince J, Duncan SH, Webster LM, Holtrop G, et al. (2011) Dominant and diet-responsive groups of bacteria within the human colonic microbiota. ISME J 5: 220-230
  331. Turroni F, Bottacini F, Foroni E, Mulder I, Kim JH, et al. (2010) Genome analysis of Bifidobacterium bifidum PRL2010 reveals metabolic pathways for host-derived glycan foraging. Proc Natl Acad Sci U S A 107: 19514-19519
  332. Zoetendal EG, Cheng B, Koike S, Mackie RI (2004) Molecular microbial ecology of the gastrointestinal tract: from phylogeny to function. Curr Issues Intest Microbiol 5: 31-48
  333. Stewart JA, Chadwick VS, Murray A (2005) Investigations into the influence of host genetics on the predominant eubacteria in the faecal microflora of children. J Med Microbiol 54: 1239-1242
  334. Delzenne NM, Neyrinck AM, Cani PD (2013) Gut microbiota and metabolic disorders: how prebiotic can work? Br J Nutr 109: S81-S85
  335. Miura T, Sano D, Suenaga A, Yoshimura T, Fuzawa M, et al. (2013) Histo-blood group antigen-like substances of human enteric bacteria as specific adsorbents for human noroviruses. J Virol 87: 9441-9451
  336. Shirato H, Ogawa S, Ito H, Sato T, Kameyama A, et al. (2008) Noroviruses distinguish between type 1 and type 2 histo-blood group antigens for binding. J Virol 82: 10756-10767
  337. Glass RI, Parashar UD, Estes MK (2009) Norovirus gastroenteritis. N Engl J Med 361: 1776-1785
  338. Böhm R, Fleming FE, Maggioni A, Dang VT, Holloway G, et al. (2015) Revisiting the role of histo-blood group antigens in rotavirus host-cell invasion. Nature Commun 6: 5907
  339. Shanker S, Choi JM, Sankaran B, Atmar RL, Estes MK, et al. (2011) Structural analysis of HBGA binding specificity in a norovirus GII. 4 epidemic variant: implications for epochal evolution. J Virol 85: 8635-8645
  340. Nordgren J, Nitiema LW, Ouermi D, Simpore J, Svensson L (2013) Host genetic factors affect susceptibility to norovirus infections in Burkina Faso. PloS One 8: e69557
  341. Kindberg E, Åkerlind B, Johnsen C, Knudsen JD, Heltberg O, et al. (2007) Host genetic resistance to symptomatic norovirus (GGII. 4) infections in Denmark. J Clin Microbiol 45: 2720-2722
  342. Singh BK, Leuthold MM, Hansman GS (2015) Human noroviruses' fondness for histo-blood group antigens. J Virol 89: 2024-2040
  343. Shanker S, Czako R, Sankaran B, Atmar RL, Estes MK, et al. (2014) Structural analysis of determinants to HBGA binding specificity in GI noroviruses. J Virol 88: 6168-6180
  344. Kinane DF, Blackwell CC, Brettle RP, Weir DM, Winstanley FP, et al. (1982 )ABO blood group, secretor state, and susceptibility to recurrent urinary tract infection in women. Br Med J 285: 7-9
  345. Kinane DF, Blackwell CC, Brettle RP, Weir DM, Winstanley FP, et al. (1983) ABO Blood Group, Secretor State, and Susceptibility to Recurrent Urinary Tract Infection in Women. J Urol 129: 673
  346. Ravn V, Dabelsteen E (2000) Tissue distribution of histo‐blood group antigens. APMIS 108: 1-28.
  347. Watanabe M, Kinoshita H, Nitta M, Yukishita R, Kawai Y, et al. (2010) Identification of a new adhesin‐like protein from Lactobacillus mucosae ME‐340 with specific affinity to the human blood group A and B antigens. J Appl Microbiol 109: 927-935
  348. Ilver D, Arnqvist A, Ögren J, Frick IM, Kersulyte D, et al. (1998) Helicobacter pylori adhesin binding fucosylated histo-blood group antigens revealed by retagging. Science 279: 373-377
  349. Hutson AM, Atmar RL, Graham DY, Estes MK (2002) Norwalk virus infection and disease is associated with ABO histo-blood group type. J Infect Dis 185: 1335-1337
  350. Levine MM, Browne RMR (2012) Factors that explain excretion of enteric pathogens by persons without diarrhea. Clin Infect Dis 55: S303-S311
  351. Harris JB, Khan AI, LaRocque RC, Dorer DJ, Chowdhury F, et al. (2005) Blood group, immunity, and risk of infection with Vibrio cholerae in an area of endemicity. Infect Immun 73: 7422-7427
  352. Tacket CO, Cohen MB, Wasserman SS, Losonsky G, Livio S, et al. (1999) Randomized, double-blind, placebo-controlled, multicentered trial of the efficacy of a single dose of live oral cholera vaccine CVD 103-HgR in preventing cholera following challenge with Vibrio cholerae O1 El tor inaba three months after vaccination. Infect Immun 67: 6341-6345
  353. Tan M, Jiang X (2014) Histo-blood group antigens: a common niche for norovirus and rotavirus. Expert Rev Mol Med 16
  354. Hu L, Crawford SE, Czako R, Penfield NWC, Smith DF, et al. (2012) Cell attachment protein VP8* of a human rotavirus specifically interacts with A-type histo-blood group antigen. Nature 485: 256-259
  355. Jacques M (1996) Role of lipo-oligosaccharides and lipopolysaccharides in bacterial adherence. Trends in microbiology 4: 408-410
  356. Mandrell RE, Apicella MA (1993) Lipo-oligosaccharides (LOS) of mucosal pathogens: molecular mimicry and host-modification of LOS. Immunobiology 187: 382-402
  357. Monteiro MA, Zheng PY, Ho B, Yokota SI, Amano KI, et al. (2000) Expression of histo-blood group antigens by lipopolysaccharides of Helicobacter pylori strains from Asian hosts: the propensity to express type 1 blood-group antigens. Glycobiology 10: 701-713
  358. Li D, Breiman A, Le Pendu J, Uyttendaele M (2015) Binding to histo-blood group antigen-expressing bacteria protects human norovirus from acute heat stress. Front Microbiol 6: 659
  359. Jones MK, Watanabe M, Zhu S, Graves CL, Keyes LR, et al. (2014) Enteric bacteria promote human and mouse norovirus infection of B cells. Science 346: 755-759
  360. Chang CJ, Lin CS, Martel J, Ojcius DM, Lai WF, et al. (2016) Modulation of host immune response by Bacteroides fragilis polysaccharides: a review of recent observations. J Biomed Lab Sci 28: 1
  361. Cohen PS, Arruda J, Williams TJ, Laux D (1985) Adhesion of a human fecal Escherichia coli strain to mouse colonic mucus. Infect Immun 48: 139-145
  362. Collins BE, Paulson JC (2004) Cell surface biology mediated by low affinity multivalent protein–glycan interactions. Curr Opin Chem Biol 8: 617-625
  363. Nothaft H, Szymanski CM (2010) Protein glycosylation in bacteria: sweeter than ever. Nat Rev Microbiol 8: 765-778.
  364. Rausch P, Rehman A, Künzel S, Häsler R, Ott SJ, et al. (2011) Colonic mucosa-associated microbiota is influenced by an interaction of Crohn disease and FUT2 (Secretor) genotype. Proc Natl Acad Sci USA 108: 19030-19035.
  365. Uchida H, Kawai Y, Kinoshita H, Kitazawa H, Miura K, et al. (2006) Lactic acid bacteria (LAB) bind to human B-or H-antigens expressed on intestinal mucosa. Biosci Biotechnol Biochem 70: 3073-3076.
  366. Gampa A, Engen PA, Shobar R, Mutlu EA (2017) Relationships between gastrointestinal microbiota and blood group antigens. Physiol Genomics 49: 473-483.
  367. Mäkivuokko H, Lahtinen SJ, Wacklin P, Tuovinen E, Tenkanen H, et al. (2012) Association between the ABO blood group and the human intestinal microbiota composition. BMC Microbiol 12: 94.
  368. Bode L, Rudloff S, Kunz C, Strobel S, Klein N, et al. (2004) Human milk oligosaccharides reduce platelet-neutrophil complex formation leading to a decrease in neutrophil beta 2 integrin expression. J Leukoc Biol 76: 820-826.
  369. Smilowitz JT, Lebrilla CB, Mills DA, German JB, Freeman SL, et al. (2014) Breast milk oligosaccharides: structure-function relationships in the neonate. Annu Rev Nutr 34: 143-169.
  370. Gabrielli O, Zampini L, Galeazzi T, Padella L, Santoro L, et al. (2011) Preterm milk oligosaccharides during the first month of lactation. Pediatrics 128: e1520-e1531.
  371. Rudloff S, Kunz C (2012) Milk oligosaccharides and metabolism in infants. Adv Nutr 3: 398S-405S.
  372. Le Pendu J (2004) Histo-blood group antigen and human milk oligosaccharides: genetic polymorphism and risk of infectious diseases. Adv Exp Med Biol 554: 135-143.
  373. Morrin ST, Irwin JA, Hickey RM (2018) The Role of Milk Oligosaccharides in Host–Microbial Interactions and Their Defensive Function in the Gut. Diet, Microbiome and Health: Elsevier 8: 199-236.
  374. Katayama T (2016) Host-derived glycans serve as selected nutrients for the gut microbe: human milk oligosaccharides and bifidobacteria. Biosci Biotechnol Biochem 80: 621-632.
  375. McGuire M (2018) The human milk microbiome – A Paradigm shift in infant nutrition. Breastfeed Med 13: S-9.
  376. Gnoth MJ, Rudloff S, Kunz C, Kinne RK (2001) Investigations on the in vitro transport of human milk oligosaccharides by a Caco-2 monolayer using a novel HPLC-MS technique. J Biol Chem 276: 34363-34370.
  377. Dotz V, Rudloff S, Meyer C, Lochnit G, Kunz C, et al. (2015) Metabolic fate of neutral human milk oligosaccharides in exclusively breast‐fed infants. Mol Nutr Food Res 59: 355-364.
  378. Bode L (2009) Human milk oligosaccharides: prebiotics and beyond. ‎Nutr Rev 67: S183-S91.
  379. Rudloff S, Pohlentz G, Diekmann L, Egge H, Kunz C, et al. (1996) Urinary excretion of lactose and oligosaccharides in preterm infants fed human milk or infant formula. Acta Paediatr 85: 598-603.
  380. Albrecht S, Schols HA, van den Heuvel EG, Voragen AG, Gruppen H, et al. (2011) Occurrence of oligosaccharides in feces of breast-fed babies in their first six months of life and the corresponding breast milk. Carbohydr Res 346: 2540-2550.
  381. Rijnierse A, Jeurink PV, van Esch BCAM, Garssen J, Knippels LMJ, et al. (2011) Food-derived oligosaccharides exhibit pharmaceutical properties. Eur J Pharmacol 668: S117-S123.
  382. Donovan SM, Comstock SS (2016) Human Milk Oligosaccharides Influence Neonatal Mucosal and Systemic Immunity. Ann Nutr Metab 69: 42-51.
  383. Newburg DS, He Y (2015) Neonatal gut microbiota and human milk glycans cooperate to attenuate infection and inflammation. Clin Obstet Gynecol 58: 814-826.
  384. Kulinich A, Liu L (2016) Human milk oligosaccharides: The role in the fine-tuning of innate immune responses. Carbohydr Res 432: 62-70.
  385. Jantscher-Krenn E, Bode L (2012) Human milk oligosaccharides and their potential benefits for the breast-fed neonate. Minerva Pediatr 64: 83-99.
  386. Noll AJ, Yu Y, Lasanajak Y, Duska-McEwen G, Buck R, et al. (2016) Human DC-SIGN binds specific human milk glycans. Biochem J 473: 1343-1353.
  387. Naarding MA, Ludwig IS, Groot F, Berkhout B, Geijtenbeek TB, et al. (2005) Lewis X component in human milk binds DC-SIGN and inhibits HIV-1 transfer to CD4+ T lymphocytes. J Clin Invest 115: 3256-3264.
  388. Hirabayashi J, Hashidate T, Arata Y, Nishi N, Nakamura T, et al. (2002) Oligosaccharide specificity of galectins: a search by frontal affinity chromatography. Biochimica et Biophysica Acta (BBA)-General Subjects 1572: 232-254.
  389. Shams-Ud-Doha K, Kitova EN, Kitov PI, St-Pierre Y, Klassen JS (2017) Human Milk Oligosaccharide Specificities of Human Galectins. Comparison of Electrospray Ionization Mass Spectrometry and Glycan Microarray Screening Results. Anal chem 89: 4914-4921.
  390. Koliwer-Brandl H, Siegert N, Umnus K, Kelm A, Tolkach A, et al. (2011) Lectin inhibition assays for the analysis of bioactive milk sialoglycoconjugates. Int dairy J 21: 413-420.
  391. Chichlowski M, German JB, Lebrilla CB, Mills DA (2011) The influence of milk oligosaccharides on microbiota of infants: opportunities for formulas. Annu Rev Food Sci Technol 2: 331-351.
  392. Schumacher G, Bendas G, Stahl B, Beermann C (2006) Human milk oligosaccharides affect P-selectin binding capacities: in vitro investigation. Nutrition 22: 620-627.
  393. Bode L, Kunz C, Muhly-Reinholz M, Mayer K, Seeger W, et al. (2004) Inhibition of monocyte, lymphocyte, and neutrophil adhesion to endothelial cells by human milk oligosaccharides. Thromb Haemost 92: 1402-1410.
  394. Kurakevich E, Hennet T, Hausmann M, Rogler G, Borsig L (2013) Milk oligosaccharide sialyl (α2, 3) lactose activates intestinal CD11c+ cells through TLR4. Proc Natl Acad Sci U S A 110: 17444-17449.
  395. Thomas PG, Carter MR, Atochina O, Da’Dara AA, Piskorska D, et al. (2003) Maturation of dendritic cell 2 phenotype by a helminth glycan uses a Toll-like receptor 4-dependent mechanism. J Immunol 171: 5837-1541.
  396. Kosaka N, Izumi H, Sekine K, Ochiya T (2010) microRNA as a new immune-regulatory agent in breast milk. Silence 1: 7.
  397. Izumi H, Tsuda M, Sato Y, Kosaka N, Ochiya T, et al. (2015) Bovine milk exosomes contain microRNA and mRNA and are taken up by human macrophages. J Dairy Sci 98: 2920-2933.
  398. Fromm B, Tosar JP, Yu L, Halushka MK, Witwer KW (2018) miR-21-5p and miR-30a-5p are identical in human and bovine, have similar isomiR distribution, and cannot be used to identify xenomiR uptake from cow milk. bioRxiv 275834.
  399. Chokeshaiusaha K, Sananmuang T, Puthier D, Nguyen C (2018) An innovative approach to predict immune-associated genes mutually targeted by cow and human milk microRNAs (miRNAs) expression profiles. Vet World 11: 1203-1209.
  400. Singh N, Shirdel EA, Waldron L, Zhang RH, Jurisica I, et al. (2012) The murine caecal microRNA signature depends on the presence of the endogenous microbiota. Int J Biol Sci 8: 171-186.
  401. Masotti A (2012) Interplays between gut microbiota and gene expression regulation by miRNAs. Front Cell Infect Microbiol 2: 137.
  402. Sanchez HN, Taylor JR, Munoz K, Zan H, Casali P (2016) Short-chain fatty acid (SCFA) histone deacetylase inhibitors produced by gut microbiota regulate selected microRNAs to modulate local and systemic antibody responses. Am Assoc Immnol 10: 946-956
  403. Viennois E, Chassaing B, Pujada A, Tahsin A, Merlin D (2018) P039 fecal mirnas shape the intestinal microbiota. Gastroenterology 154: S20.
  404. Liu S, da Cunha AP, Rezende RM, Cialic R, Wei Z, et al. (2016) The host shapes the gut microbiota via fecal microRNA. Cell Host Microbe 19: 32-43.
  405. Zhao L, Zhou X, Cai W, Shi R, Yang G (2017) Host intestinal epithelium derived mirnas shape the microbiota and its implication in cardiovascular diseases. J Am Coll Cardiol 69: 1075.
  406. Chang L-C, Yu Y-L (2016) Dietary components as epigenetic-regulating agents against cancer. Biomedicine 6: 1.
  407. Celluzzi A, Masotti A (2016) Interplays between gut microbiota and gene expression regulation by miRNAs: towards a symbiotic vision of host and guest. Non-coding RNAs and Inter-kingdom Communication. Springer pp: 53-65.
  408. Bäckhed F (2012) Host responses to the human microbiome. Nutr Rev 70: S14-S7.
  409. Martemucci G, D’Alessandro AG (2012) Fat content, energy value and fatty acid profile of donkey milk during lactation and implications for human nutrition. Lipids Health Dis 11: 113.
  410. Gaboriau-Routhiau V, Rakotobe S, Lécuyer E, Mulder I, Lan A, et al. (2009) The key role of segmented filamentous bacteria in the coordinated maturation of gut helper T cell responses. Immunity 31: 677-689.
  411. Ivanov II, de Llanos Frutos R, Manel N, Yoshinaga K, Rifkin DB, et al. (2008) Specific microbiota direct the differentiation of IL-17-producing T-helper cells in the mucosa of the small intestine. Cell Host Microbe 4: 337-349.
  412. Farkas AM, Ivanov II (2015) Escaping negative selection: ILC you in the gut. Immunity 43: 12-14.
  413. Mowat AM (2003) Anatomical basis of tolerance and immunity to intestinal antigens. Nat Rev Immunol 3: 331.
  414. Lehner MD, Morath S, Michelsen KS, Schumann RR, Hartung T (2001) Induction of cross-tolerance by lipopolysaccharide and highly purified lipoteichoic acid via different Toll-like receptors independent of paracrine mediators. J Immunol 166: 5161-5167.
  415. Worbs T, Bode U, Yan S, Hoffmann MW, Hintzen G, et al. (2006) Oral tolerance originates in the intestinal immune system and relies on antigen carriage by dendritic cells. J Exp Med 203: 519-527.
  416. Cani PD, Amar J, Iglesias MA, Poggi M, Knauf C, et al. (2007) Metabolic endotoxemia initiates obesity and insulin resistance. Diabetes 56: 1761-1772.
  417. Cani PD, Osto M, Geurts L, Everard A (2012 Involvement of gut microbiota in the development of low-grade inflammation and type 2 diabetes associated with obesity. Gut Microbes 3: 279-288.
  418. Le Pendu J, Marionneau S, Cailleau‐Thomas A, Rocher J, Le Moullac‐Vaidye B, et al. (2001) ABH and Lewis histo‐blood group antigens in cancer. Apmis 109: 9-26
  419. Hirneisen KA, Kniel KE (2013) Norovirus Attachment. Food Prot Trends 3: 290-299.
  420. Marionneau S, Ruvoën N, Le Moullac–Vaidye B, Clement M, Cailleau–Thomas A, et al. (2002) Norwalk virus binds to histo-blood group antigens present on gastroduodenal epithelial cells of secretor individuals. Gastroenterology 122: 1967-1977.
  421. Marionneau S, Cailleau-Thomas A, Rocher J, Le Moullac-Vaidye B, Ruvoën N, et al. (2001) ABH and Lewis histo-blood group antigens, a model for the meaning of oligosaccharide diversity in the face of a changing world. Biochimie 83: 565-573.
  422. Wolpin BM, Chan AT, Hartge P, Chanock SJ, Kraft P, et al. (2009) ABO blood group and the risk of pancreatic cancer. J Natl Cancer Inst 101: 424-431.
  423. Storry J, Olsson ML (2009) The ABO blood group system revisited: a review and update. Immunohematology 25: 48.
  424. Li Y, Huang X, Wang C, Li Y, Luan M (2015) Ganglioside GM3 exerts opposite effects on motility via epidermal growth factor receptor and hepatocyte growth factor receptor-mediated migration signaling. Mol Med Rep 11: 2959-2966.
  425. Reid ME, Lomas-Francis C, Olsson ML (2012) The blood group antigen factsbook (3rd Edn). Academic Press; p: 758
  426. Imberty A, Varrot A (2008) Microbial recognition of human cell surface glycoconjugates. Curr Opin Struct Biol 18: 567-576.
  427. Cohen M, Varki A (2010) The sialome-far more than the sum of its parts. OMICS 14: 455-464
  428. Hall M, Weidner D, Zhu Y, Dayal S, Whitman A, et al. (2016) Predominant expression of hybrid N-glycans has distinct cellular roles relative to complex and oligomannose N-glycans. Int J mol sci 17: 925
  429. Cohen M (2015) Notable aspects of glycan-protein interactions. Biomolecules 5: 2056-2072
  430. Wang J, García-Bailo B, Nielsen D, El-Sohemy A (2014) ABO genotype,‘blood-type’diet and cardiometabolic risk factors. PloS one 9: e84749
  431. Freed D (1999) Eat Right 4 Your Type: The Individualized Diet Solution to Staying Healthy, Living Longer and Achieving Your Ideal Weight. J Nutr Environ Med 9: 168
  432. Poleszynski D (2001) Blodtypediettens vitenskapelige grunnlag. Tidsskrift-Norske Laegeforening 121: 1838-1839
  433. Meltzer H, Haugen M, Haavardsholm K, Hagen K, Heier H, et al. (2002) Blodtypedietten-visjonaer vitenskap eller tull? Tidsskrift-Norske Laegeforening 122: 1402-1405
  434. Power L (2007) Biotype Diets System®: Blood types and food allergies. Journal of Nutritional & Environmental Medicine 16: 125-135
  435. Mozzi P (2012) La dieta del dottor Mozzi (5th Edn). Piacenza (IT): Mogliazze
  436. Moen T (2001) "Blood type diet"--science or fantasy? Tidsskrift for den Norske laegeforening: tidsskrift for praktisk medicin, ny raekke 121: 355-358
  437. Cusack L, De Buck E, Compernolle V, Vandekerckhove P (2013) Blood type diets lack supporting evidence: a systematic review. Am J Clin Nutr 98: 99-104
  438. d'Adamo P, Whitney C (1996) Eat right 4 your type. NY: GP Putnam’s Sons
  439. André S, Kaltner H, Manning J, Murphy P, Gabius HJ, et al. (2015) Lectins: getting familiar with translators of the sugar code. Molecules 20: 1788-1823
  440. Gao X, Esseili M, Lu Z, Saif L, Wang Q, et al. (2016) Recognizing HBGA-like carbohydrates in lettuce by human GII. 4 norovirus. Appl Environ Microbiol p: 04096
  441. Springer G, Williamson P, Brandes W (1961) Blood group activity of gram-negative bacteria. J Exp Med 113: 1077-1093
  442. Tamura K, Hemsworth G, Déjean G, Rogers T, Pudlo N, et al. (2017) Molecular mechanism by which prominent human gut bacteroidetes utilize mixed-linkage beta-glucans, major health-promoting cereal polysaccharides. Cell Rep 21: 417-430
  443. Choi S-W, Claycombe K, Martinez J, Friso S, Schalinske K, et al. (2013) Nutritional epigenomics: a portal to disease prevention. Adv Nutr 4: 530-532
Citation: Menapace M (2018) Recent Advances in Nutritional Sciences: An Overview of Glycans and miRNAs. J Nutr Food Sci 8: 734.

Copyright: © 2018 Menapace M. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Top