Inflammatory bowel diseases (IBD) are modern diseases, with incidence rising around the world. They are associated with perturbation of the intestinal microbiota, and with alteration and crossing of the mucus barrier by the commensal bacteria that feed on it. In the process of mucus catabolism and invasion by gut bacteria, carbohydrate-active enzymes (CAZymes) play a critical role since mucus is mainly made up by O- and N-glycans. Moreover, the occurrence of IBD seems to be associated with low-fiber diets. Conversely, supplementation with oligosaccharides, such as human milk oligosaccharides (HMOs), which are structurally similar to intestinal mucins and could thus compete with them towards bacterial mucus-degrading CAZymes, has been suggested to prevent inflammation. In this mini-review, we will establish the current state of knowledge regarding the identification and characterization of mucus-degrading enzymes from both cultured and uncultured species of gut commensals and enteropathogens, with a particular focus on the present technological opportunities available to further the discovery of mucus-degrading CAZymes within the entire gut microbiome, by coupling microfluidics with metagenomics and culturomics. Finally, we will discuss the challenges to overcome to better assess how CAZymes targeting specific functional oligosaccharides could be involved in the modulation of the mucus-driven cross-talk between gut bacteria and their host in the context of IBD.

Intestinal mucus is notably formed by glycoproteins, the O- and N-linked glycans which make up to 80% of their total mass. These structurally complex glycans influence the cohesiveness of the mucus network. Their structure and defects in their biosynthesis [1] may be differently modulated by environmental factors [2–4], in particular in inflammatory bowel diseases (IBD), such as Crohn’s disease (CD) and ulcerative colitis (UC). These diseases, which are more prevalent in the modern world, are associated with the Western diet with low gut microbiota-accessible carbohydrates, which is linked to a lack of microbial diversity and altered production of short-chain fatty acids [5]. IBD have become increasingly prevalent in Europe, North America and Australia since the 1950s, with their incidence rising beyond the western world over the past 30 years [6]. They are characterized by an imbalance in the composition of the intestinal microbiota [7–9], the so-called ‘dysbiosis’, and by an uncontrolled inflammatory response to luminal content [10]. In addition, the intestinal barrier function is severely impaired: significant changes in mucus secretion and mucin O-glycosylation occur, the inner mucus barrier is thinner and looser, and the space closest to the epithelium is invaded by commensal bacteria and/or enteropathogens [11–14]. Indeed, together with the dietary glycans (HMOs for infants, and then fibers of plant origin) and, to a minor extent, with microbial exopolysaccharides, the mucus glycans are the main carbon sources for gut bacteria. To deal with the huge structural diversity of glycans they feed on, and to break them down into metabolizable monosaccharides, intestinal bacteria produce a large panel of carbohydrate-active enzymes (CAZymes) with various specificities [15], and a battery of proteins to sense, bind and transport glycans into bacterial cells for their complete breakdown. These metabolic machineries are often encoded by multigenic clusters, defined as polysaccharide utilization loci (PUL) in the Gram-negative Bacteroidota (formerly Bacteroidetes [16]), one of the dominant phyla in the human gut. Bacteroidota include bacteria that are able to feed on mucins, such as the commensal species Bacteroides thetaiotamicron and Bacteroides fragilis, the abundance of which seems to be correlated to CD and UC [17,18]. CAZymes targeting mucins are also found in other Gram-negative mucin degraders, such as, for example, Akkermansia muciniphila [19], which is negatively associated with IBD [8], and in Gram-positive ones, such as Ruminococcus gnavus, which is, on the contrary, positively associated with these diseases, and CD in particular [20]. Given that mucin degraders can be both positively and negatively correlated to IBD, it is very difficult to determine whether mucin breakdown by gut bacterial CAZymes is involved in intestinal epithelium inflammation. CAZymes produced by mucin-degrading bacteria have been reviewed several times over the last decade [21–25]. Nevertheless, some recent functional metagenomics studies have highlighted the potential of CAZymes produced by uncultured gut bacteria, and appearing as IBD biomarkers, to degrade human O-glycans present in mucus [26,27] and in human milk oligosaccharides (HMOs). These oligosaccharides could indeed compete with the O-glycans of mucins lining the intestinal epithelium towards mucus-degrading CAZymes, since they are structurally similar to their extremities. HMOs seem to protect against IBD [28–30], although these data are still controversial [31], as for other functional foods that are metabolized thanks to CAZymes from the intestinal microbiome [32].

In this review, we will update the list of CAZymes that have been shown to target O-mucins, since mucins are much more sparsely N-glycosylated than O-glycosylated [33,34]. Here, we will discuss the advantages and limits of the methods that are currently available to identify them within the entire microbiome. Finally, we will explore the potential role that mucin-degrading CAZymes could play in the interrelationships between the host, diet and the microbiota in the context of gut inflammation.

Structure of the O-mucins

MUC2, the major colonic mucin is composed of ∼20% protein and ∼80% glycan. The protein sequence rich in Ser/Thr residues are primary targets for O-glycosylation with N-acetyl-d-galactosamine (GalNAc), creating the foundation upon which long oligosaccharide chains are built. While these side chains are composed of only five different monosaccharides (N-acetyl-d-glucosamine (GlcNac), GalNac, d-galactose, l-fucose and N-acetylneuraminic acid [also called sialic acid, Neu5Ac]), the order in which they can be assembled is hugely variable. In addition, the GlcNac and galactose residues can be sulfated, adding a supplemental level of complexity. The resulting O-glycans extend out from the mucin protein core in a ‘bottlebrush’ configuration and its heterogeneity provides some resistance to microbial degradation [33,35]. It should be emphasized that the mucin O-glycans vary enormously along the intestinal tract and between species (Table 1), compelling researchers in the area to take greater care when studying bacteria and mucin from the same niche.

Table 1
Main characteristics of substrates
Substrates categoriesRepresentative structures
Chromogenic /fluorogenic glycoside  
Origin: synthetic substrates
Commercial price: a few hundreds to a few thousand €/g
Applications: conventional screening; specificity characterization in microplates; droplet Microfluidics-based screening (fluorogenic substrates) 
X-GlcNAc, X-sulfate, X-Neu5Ac, …
pNP-Fuc, pNP-GlcNAc, …
Fluorescein-GlcNAc, … 
HMOs and blood antigens  
Origin: synthetic substrates
Commercial price: tens of thousands €/g
Applications: specificity characterization (HPAEC-PAD); droplet microfluidics-based screening and culturomics 

 
Pig gastric mucin (PGM)  
Origin: sampling on animals
Commercial price: €1 to €10/g
Applications: transcriptomics; characterization of synthetic cocktails of CAZmes 

 
Human colonic mucin  
Origin: surgical pieces, cell lines, organoids
Not commercial
Applications: ELISA assays; characterization of synthetic cocktails of CAZymes (though not exemplified yet) 

 
Substrates categoriesRepresentative structures
Chromogenic /fluorogenic glycoside  
Origin: synthetic substrates
Commercial price: a few hundreds to a few thousand €/g
Applications: conventional screening; specificity characterization in microplates; droplet Microfluidics-based screening (fluorogenic substrates) 
X-GlcNAc, X-sulfate, X-Neu5Ac, …
pNP-Fuc, pNP-GlcNAc, …
Fluorescein-GlcNAc, … 
HMOs and blood antigens  
Origin: synthetic substrates
Commercial price: tens of thousands €/g
Applications: specificity characterization (HPAEC-PAD); droplet microfluidics-based screening and culturomics 

 
Pig gastric mucin (PGM)  
Origin: sampling on animals
Commercial price: €1 to €10/g
Applications: transcriptomics; characterization of synthetic cocktails of CAZmes 

 
Human colonic mucin  
Origin: surgical pieces, cell lines, organoids
Not commercial
Applications: ELISA assays; characterization of synthetic cocktails of CAZymes (though not exemplified yet) 

 

Symbols: GlcNac , Gal , GalNAc , Fucose , Sialic acid , Glc , Sulfate S; (a) Representative, not exhaustive structures decorating PGM chains (mainly 4–6 units), leading to extreme diversity. (b) Schematic view of the most common, core 3-based branch (adapted from [36]). For the sake of clarity, the nature of the bonds between the units are not depicted.

Identification and characterization of O-mucin degrading CAZymes

Several comparative genomic analyses have attempted to determine the CAZyme mucin-degrading profiles of the human gut microbiota [37,38], and there have been efforts to couple these data with transcriptomic, proteomic, genetic and biochemical studies in order to understand mucin catabolic pathways. One of the earliest and most studied commensal bacteria is B. thetaiotaomicron, which is one of the richest human bacteria in glycan-degrading enzymes [15,39]. In 2008, Martens and his team performed whole-genome transcriptional profiling and genetic tests to define the mechanisms underlying host glycan foraging in vivo (mice) and in vitro [40]. Mucin O-glycans are the principal substrate foraged in vivo and 16 PULs were identified with probable specificities for O-glycans. Recently, two of these PULs were characterized highlighting the major role of GH16 enzymes [41] and sulfatases [42–44]. Growth assays on mucins combined with transcriptomic analyses were also performed on other Bacteroidota such as B. fragilis [45,46] and B. massiliensis [45]. Similar studies were performed on the mucin degraders A. muciniphila [47–50] (Verrucomicrobia), R. gnavus [51] (Firmicutes) and Bifidobacterium bifidum [52] (Actinobacteria). This list is destined to get longer as 23 representative gut microbes were shown to grow on porcine intestinal mucin as their sole carbon source [38].

The O-mucin-degrading CAZymes identified by genomic and transcriptomics analysis are usually biochemically characterized using porcine gastric and intestinal mucins, bovine submaxillary mucin, mouse crude mucin as well as HMOs, blood-group oligosaccharides and synthetic oligosaccharides. To date, 18 glycoside hydrolase (GH) families were shown to be involved in the degradation of O-mucin glycans (www.cazy.org). GH33 sialidases cleave terminal sialic acid residues while fucose residues are removed by GH29 or GH95. Subsequent degradation of the glycan chains involves lacto-N-biosidase (GH136), lacto-N-biose phosphorylases (GH112), N-acetyl-glucosaminidases (GH84, GH85, GH89, GH20), N-acetyl-galactosaminidases (GH31, GH101, GH109, GH129) and galactosidases (GH2, GH35, GH42, GH98). There are also endo-acting O-glycanases (GH16), which can cleave large glycan structures. Table 2 lists the characterized O-mucin-degrading CAZymes since 2015. It is an update of a previous review [22]. To provide an overview of the complete catabolic pathway of O-mucins, the proteases that have been shown to be specific to this substrate are listed in Table 3, and the sulfatases and the sialate O-acetylesterase are listed in Table 4.

Table 2
The list of biochemically characterized O-mucin degrading CAZymes
CAZy familyName or locus tagActivityOrganismAccession numbers
GH2
 
Amuc_0824 β-galactosidase (preference for galactose-β-1,3-N-acetyl-d-galactosamine) Akkermansia muciniphila ATCC BAA-835
 
ACD04657.1 [53
Amuc_1666 β-galactosidase (preference for galactose-β-1,4-N-acetyl-d-glucosamine) ACD05484.1 [53
GH16
 
 
 
 
 
 
 
 
BT2824 O-Glycan β-1,4-galactosidase with a requirement for a β1,3-linked
sugar at the -2 position
 
 
 
 
 
 
 
 
Bacteroides thetaiotaomicron VPI-5482 AAO77930.1 [41
BF4058 Bacteroides fragilis NCTC 9343
 
CAH09732.1 [41
BF4060 CAH09734.1 [41]
PDB 6T2S 
BACCAC_02679; CGC64_00695 Bacteroides caccae ATCC 43185
 
 
ASM64646.1 [41
BACCAC_02680; CGC64_00690 ASM64645.1 [41]
PDB 6T2Q, 6T2P, 2T2O 
BACCAC_03717; CGC64_17650 ASM67518.1 [41]
PDB 6T2R 
Amuc_0724 Akkermansia muciniphila ATCC BAA-835
 
 
ACD04561.1 [41]
PDB 6T2N 
Amuc_875 ACD04708.1 [41
Amuc_2108 ACD05917.1 [41
GH29
 
 
 
 
 
 
 
 
RiFuc29, ROSEINA2194_01891/01890 α-1,4-fucosidase Roseburia inulinivorans DSM 16841 C0FT24/C0FT23 (Uniprot) [54
E1_10125; RUGNEv3_10125 α-l-fucosidase (preference for α1,3/4 fucosylated linkages)
 
Ruminococcus gnavus E1
 
A0A6N3BKT0 [55]
PDB 6TR3, 6TR4 
E1_10180; RUGNEv3_10180 - [55
ATCC_03833; RUMGNA_03833 α-l-fucosidase (preference for α1,2 linkages) Ruminococcus gnavus ATCC 29149 - [55
Afc2; CPF_2130 α-l-fucosidase (preference for α1,3/4 fucosylated linkages) Clostridium perfringens ATCC 13124 ABG83106.1 [56
AmGH29A, Amuc_0010 α-l-fucosidase (preference for Fucα1,3GlcNAc)
 
Akkermansia muciniphila ATCC BAA‐835
 
 
 
ACD03857.1 [57
AmGH29B, Amuc_0146 ACD03990.1 [57
AmGH29C, Amuc_0392 α-l-fucosidase (preference for α1,3/4 fucosylated linkages)
 
ACD04231.1 [57
AmGH29D, Amuc_0846 ACD04679.1 [58
GH31
 
BpGH31 α-N-acetylgalactosaminidase
 
Phocaeicola plebeius (formerly Bacteroides plebeiusWP_007559952.1 [58
BcGH31 Bacteroides caccae WP_005682123.1 [58
GH33
 
 
 
RgNanH Intramolecular trans-sialidase (IT-sialidase) producing 2,7-anhydro-Neu5Ac selectively from α2-3-linked sialic acid substrates Ruminococcus gnavus ATCC 29 149 ETD19277.1 [59
NanH, BF1806 Sialidase Bacteroides fragilis NCTC 9343 WP_010992682.1 [60
AmGH33A, Amuc_0625 α2,3‐ and α2,6‐sialyl linkages
 
Akkermansia muciniphila ATCC BAA‐835
 
ACD04462.1 [57
AmGH33B, Amuc_1835 ACD05653.1 [57
GH35
 
Am0874; Amuc_0771 β-galactosidase (β1,3- and β1,6-linked galactose residues from various substrates, preference for galactose-β-1,3-N-acetyl-d-glucosamine) Akkermansia muciniphila ATCC BAA-835
 
ACD04606.1 [53,61
Amuc_1686 β-galactosidase (Galβ1-3GalNAc) ACD05504.1 [62
GH95
 
 
 
 
 
RiFuc95, ROSEINA2194_01889/01888/01887 α-1,2-fucosidase Roseburia inulinivorans DSM 16841 C0FT22/C0FT21/C0FT2 (Uniprot) [54
ATCC_00842; RUMGNA_00842 α-l-fucosidase (preference for α1,2 linkages) Ruminococcus gnavus ATCC 29149 [55
E1_10587; RUGNEv3_10587 α-l-fucosidase (low activity, not possible to assess its substrate specificity) Ruminococcus gnavus E1 [55
Afc3; CPF_2129 α-l-fucosidase (preference for α1,2 linkages) Clostridium perfringens ATCC 13124 ABG82552.1 [56
AmGH95A, Amuc_0186 α-l-fucosidase (preference for α1,2/3 fucosylated linkages)
 
Akkermansia muciniphila ATCC BAA‐835
 
ACD04030.1 [57
AmGH95B, Amuc_1120 ACD04946.1 [57
GH98 RiGH98, ROSEINA2194_02198 Blood-group endo-β-1,4-galactosidase Roseburia inulinivorans DSM 16841 C0FTX7 (Uniprot) [54
GH109 BvGH109_1 α-N-acetylgalactosaminidase Bacteroides vulgatus WP_005848863.1 [58
GH112
 
RhGLnbp112, RHOM_04120 β-1,3-galactosyl-N-acetylhexosamine phosphorylase
 
Roseburia hominis A2-183 AEN95946.1 [54
RiGLnbp112, ROSEINA2194_01885 Roseburia inulinivorans DSM 16841 EEG94248.1 [54
GH136
 
 
RhLnb136I, RHOM_04110 + RhLnb136II, RHOM_04115 Lacto-N-biosidase Roseburia hominis A2-183 G2T0V0 + G2T0V1 (Uniprot) [54
RiLea/bI136, ROSEINA2194_01899 + RiLea/bII136, ROSEINA2194_01898 Lacto-N-biosidase. Releases Lewis a triose or Lewis b tetraose from fucosylated HMOs including lacto-N-fucopentaose II, lacto-N-difucohexaose I and lacto-N-difucohexaose II Roseburia inulinivorans DSM 16841 C0FT31 + C0FT32 (Uniprot) [54
ErLnb136, HMPREF0373_02965 Lacto-N-biosidase Eubacterium ramulus ATCC 29099 ERK42296.1 [54]
PDB 6KQS, 6KQT 
New family to be assigned Amuc_1547 Exclusive specificity towards the sialyl‐T‐antigen but accepts α2,6‐sialyation or β1,6‐substitution of the GalNAc unit Akkermansia muciniphila ATCC BAA‐835 ACD05368.1 [57
CAZy familyName or locus tagActivityOrganismAccession numbers
GH2
 
Amuc_0824 β-galactosidase (preference for galactose-β-1,3-N-acetyl-d-galactosamine) Akkermansia muciniphila ATCC BAA-835
 
ACD04657.1 [53
Amuc_1666 β-galactosidase (preference for galactose-β-1,4-N-acetyl-d-glucosamine) ACD05484.1 [53
GH16
 
 
 
 
 
 
 
 
BT2824 O-Glycan β-1,4-galactosidase with a requirement for a β1,3-linked
sugar at the -2 position
 
 
 
 
 
 
 
 
Bacteroides thetaiotaomicron VPI-5482 AAO77930.1 [41
BF4058 Bacteroides fragilis NCTC 9343
 
CAH09732.1 [41
BF4060 CAH09734.1 [41]
PDB 6T2S 
BACCAC_02679; CGC64_00695 Bacteroides caccae ATCC 43185
 
 
ASM64646.1 [41
BACCAC_02680; CGC64_00690 ASM64645.1 [41]
PDB 6T2Q, 6T2P, 2T2O 
BACCAC_03717; CGC64_17650 ASM67518.1 [41]
PDB 6T2R 
Amuc_0724 Akkermansia muciniphila ATCC BAA-835
 
 
ACD04561.1 [41]
PDB 6T2N 
Amuc_875 ACD04708.1 [41
Amuc_2108 ACD05917.1 [41
GH29
 
 
 
 
 
 
 
 
RiFuc29, ROSEINA2194_01891/01890 α-1,4-fucosidase Roseburia inulinivorans DSM 16841 C0FT24/C0FT23 (Uniprot) [54
E1_10125; RUGNEv3_10125 α-l-fucosidase (preference for α1,3/4 fucosylated linkages)
 
Ruminococcus gnavus E1
 
A0A6N3BKT0 [55]
PDB 6TR3, 6TR4 
E1_10180; RUGNEv3_10180 - [55
ATCC_03833; RUMGNA_03833 α-l-fucosidase (preference for α1,2 linkages) Ruminococcus gnavus ATCC 29149 - [55
Afc2; CPF_2130 α-l-fucosidase (preference for α1,3/4 fucosylated linkages) Clostridium perfringens ATCC 13124 ABG83106.1 [56
AmGH29A, Amuc_0010 α-l-fucosidase (preference for Fucα1,3GlcNAc)
 
Akkermansia muciniphila ATCC BAA‐835
 
 
 
ACD03857.1 [57
AmGH29B, Amuc_0146 ACD03990.1 [57
AmGH29C, Amuc_0392 α-l-fucosidase (preference for α1,3/4 fucosylated linkages)
 
ACD04231.1 [57
AmGH29D, Amuc_0846 ACD04679.1 [58
GH31
 
BpGH31 α-N-acetylgalactosaminidase
 
Phocaeicola plebeius (formerly Bacteroides plebeiusWP_007559952.1 [58
BcGH31 Bacteroides caccae WP_005682123.1 [58
GH33
 
 
 
RgNanH Intramolecular trans-sialidase (IT-sialidase) producing 2,7-anhydro-Neu5Ac selectively from α2-3-linked sialic acid substrates Ruminococcus gnavus ATCC 29 149 ETD19277.1 [59
NanH, BF1806 Sialidase Bacteroides fragilis NCTC 9343 WP_010992682.1 [60
AmGH33A, Amuc_0625 α2,3‐ and α2,6‐sialyl linkages
 
Akkermansia muciniphila ATCC BAA‐835
 
ACD04462.1 [57
AmGH33B, Amuc_1835 ACD05653.1 [57
GH35
 
Am0874; Amuc_0771 β-galactosidase (β1,3- and β1,6-linked galactose residues from various substrates, preference for galactose-β-1,3-N-acetyl-d-glucosamine) Akkermansia muciniphila ATCC BAA-835
 
ACD04606.1 [53,61
Amuc_1686 β-galactosidase (Galβ1-3GalNAc) ACD05504.1 [62
GH95
 
 
 
 
 
RiFuc95, ROSEINA2194_01889/01888/01887 α-1,2-fucosidase Roseburia inulinivorans DSM 16841 C0FT22/C0FT21/C0FT2 (Uniprot) [54
ATCC_00842; RUMGNA_00842 α-l-fucosidase (preference for α1,2 linkages) Ruminococcus gnavus ATCC 29149 [55
E1_10587; RUGNEv3_10587 α-l-fucosidase (low activity, not possible to assess its substrate specificity) Ruminococcus gnavus E1 [55
Afc3; CPF_2129 α-l-fucosidase (preference for α1,2 linkages) Clostridium perfringens ATCC 13124 ABG82552.1 [56
AmGH95A, Amuc_0186 α-l-fucosidase (preference for α1,2/3 fucosylated linkages)
 
Akkermansia muciniphila ATCC BAA‐835
 
ACD04030.1 [57
AmGH95B, Amuc_1120 ACD04946.1 [57
GH98 RiGH98, ROSEINA2194_02198 Blood-group endo-β-1,4-galactosidase Roseburia inulinivorans DSM 16841 C0FTX7 (Uniprot) [54
GH109 BvGH109_1 α-N-acetylgalactosaminidase Bacteroides vulgatus WP_005848863.1 [58
GH112
 
RhGLnbp112, RHOM_04120 β-1,3-galactosyl-N-acetylhexosamine phosphorylase
 
Roseburia hominis A2-183 AEN95946.1 [54
RiGLnbp112, ROSEINA2194_01885 Roseburia inulinivorans DSM 16841 EEG94248.1 [54
GH136
 
 
RhLnb136I, RHOM_04110 + RhLnb136II, RHOM_04115 Lacto-N-biosidase Roseburia hominis A2-183 G2T0V0 + G2T0V1 (Uniprot) [54
RiLea/bI136, ROSEINA2194_01899 + RiLea/bII136, ROSEINA2194_01898 Lacto-N-biosidase. Releases Lewis a triose or Lewis b tetraose from fucosylated HMOs including lacto-N-fucopentaose II, lacto-N-difucohexaose I and lacto-N-difucohexaose II Roseburia inulinivorans DSM 16841 C0FT31 + C0FT32 (Uniprot) [54
ErLnb136, HMPREF0373_02965 Lacto-N-biosidase Eubacterium ramulus ATCC 29099 ERK42296.1 [54]
PDB 6KQS, 6KQT 
New family to be assigned Amuc_1547 Exclusive specificity towards the sialyl‐T‐antigen but accepts α2,6‐sialyation or β1,6‐substitution of the GalNAc unit Akkermansia muciniphila ATCC BAA‐835 ACD05368.1 [57
Table 3
The list of proteases that have been shown to be involved in O-mucin degradation
Protease familyName or locus tagActivityOrganismAccession numbers
Serine protease Pic (Protein involved in Intestinal Colonization) autotransporter Pic Active on bovine submaxillary mucin and mouse crude mucin. Similar cleavage pattern to BT4244 but no tolerance for the presence of sialic acid Escherichia coli (EAEC) 042 AF097644.1 [63
 Pic Active on bovine submaxillary mucin and mouse crude mucin Shigella flexneri 2457T Q54151 (Uniprot) [63
M60-like zinc metalloprotease BT4244 Cleaves peptides N-terminally to glycosylated Ser or Thr residues and preferred the modified residue to bear atruncated, tumor-associated glycan such as the Tn (O-GalNAc) or T (O-GalNAc-galactose) antigen. Sensitivity to sialic acid Bacteroides thetaiotaomicron VPI-5482 WP_008764444.1 [64–66]
PDB 5KD2, 5KD5, 5KD8 
 SslE Active on bovine submaxillary mucin Escherichia coli IHE3034 WP_001034562.1 [67
 YghJ Active on human MUC2 and MUC3 Escherichia coli H10407 ETEC E3PJ90 [68] (Uniprot) 
 AM0627 Cleaves between two O-glycosylated Ser or Thr residues bearing atruncated, tumor-associated glycan such as the Tn (O-GalNAc) or T (O-GalNAc-galactose) antigen.
Sensitivity to sialic acid 
Akkermansia muciniphila ATCC BAA-835D-5 B2UPI7 (Uniprot) [42,69,70]
PDB 7SCI, 7YX8 
 AM0908 Cleaves N-terminally to a glycosylated Ser or Thr with a marked preference for an adjacent Ser or Thr residue. Tolerate sialic acid  B2UQK5 (Uniprot) [66
 AM1514 Cleaves N-terminally to a glycosylated Ser or Thr with a marked preference for an adjacent Ser or Thr residue. Specific for O-GalNAc-galactose and O-GalNAc antigen-modified amino acids.  B2ULE8 (Uniprot) [66
 PA0572, IMPa Hydrolyze a minimally O-glycosylated substrate with only the Tn- or TAg-glycans. Highest affinity when this core modification is extended with a β-1,6-linked GlcNAc (GlcNAcβ1-6GalNAcα1-Thr). Pseudomonas aeruginosa (PAO1) Q9I5W4 (Uniprot) [65,71]
PDB 5KDV, 5KDW, 5KDX 
 ZmpB, CPF_1489 Absolute requirement for the α-2,6-Neu5Ac present on the peptide-linked GalNAc. Clostridium perfringens ATCC 13124 A0A0H2YN38 (Uniprot) [65,72]
PDB 5KDJ, 5KDN, 5KDS, 5KDU 
 ZmpC, CP4_3468 Action on similar protein than ZmpB 1 (75% sequence identity) Clostridium perfringens CP4 F8UNJ8 (Uniprot) [72]
PDB 6XSZ, 6XT1 
M66-like zinc metalloprotease StcE Reduce the inner mucus layer in human colonic mucosal biopsies and the MUC2 glycoprotein levels in mucin-producing LS174T colon carcinoma cells Escherichia coli (EHEC) O82882 (Uniprot) [73
Peptidase_M43 domain-containing protein OgpA, Amuc_1119, OpeRATOR® and GlycOCATCH® Active on Fetuin and IgA. Exclusively hydrolyzes the peptide bond N-terminal to serine or threonine residues substituted with an O-glycan Akkermansia muciniphila ATCC BAA‐835 ACD04945.1 [74]
PDB 6Z2D, 6Z2O 
Protease familyName or locus tagActivityOrganismAccession numbers
Serine protease Pic (Protein involved in Intestinal Colonization) autotransporter Pic Active on bovine submaxillary mucin and mouse crude mucin. Similar cleavage pattern to BT4244 but no tolerance for the presence of sialic acid Escherichia coli (EAEC) 042 AF097644.1 [63
 Pic Active on bovine submaxillary mucin and mouse crude mucin Shigella flexneri 2457T Q54151 (Uniprot) [63
M60-like zinc metalloprotease BT4244 Cleaves peptides N-terminally to glycosylated Ser or Thr residues and preferred the modified residue to bear atruncated, tumor-associated glycan such as the Tn (O-GalNAc) or T (O-GalNAc-galactose) antigen. Sensitivity to sialic acid Bacteroides thetaiotaomicron VPI-5482 WP_008764444.1 [64–66]
PDB 5KD2, 5KD5, 5KD8 
 SslE Active on bovine submaxillary mucin Escherichia coli IHE3034 WP_001034562.1 [67
 YghJ Active on human MUC2 and MUC3 Escherichia coli H10407 ETEC E3PJ90 [68] (Uniprot) 
 AM0627 Cleaves between two O-glycosylated Ser or Thr residues bearing atruncated, tumor-associated glycan such as the Tn (O-GalNAc) or T (O-GalNAc-galactose) antigen.
Sensitivity to sialic acid 
Akkermansia muciniphila ATCC BAA-835D-5 B2UPI7 (Uniprot) [42,69,70]
PDB 7SCI, 7YX8 
 AM0908 Cleaves N-terminally to a glycosylated Ser or Thr with a marked preference for an adjacent Ser or Thr residue. Tolerate sialic acid  B2UQK5 (Uniprot) [66
 AM1514 Cleaves N-terminally to a glycosylated Ser or Thr with a marked preference for an adjacent Ser or Thr residue. Specific for O-GalNAc-galactose and O-GalNAc antigen-modified amino acids.  B2ULE8 (Uniprot) [66
 PA0572, IMPa Hydrolyze a minimally O-glycosylated substrate with only the Tn- or TAg-glycans. Highest affinity when this core modification is extended with a β-1,6-linked GlcNAc (GlcNAcβ1-6GalNAcα1-Thr). Pseudomonas aeruginosa (PAO1) Q9I5W4 (Uniprot) [65,71]
PDB 5KDV, 5KDW, 5KDX 
 ZmpB, CPF_1489 Absolute requirement for the α-2,6-Neu5Ac present on the peptide-linked GalNAc. Clostridium perfringens ATCC 13124 A0A0H2YN38 (Uniprot) [65,72]
PDB 5KDJ, 5KDN, 5KDS, 5KDU 
 ZmpC, CP4_3468 Action on similar protein than ZmpB 1 (75% sequence identity) Clostridium perfringens CP4 F8UNJ8 (Uniprot) [72]
PDB 6XSZ, 6XT1 
M66-like zinc metalloprotease StcE Reduce the inner mucus layer in human colonic mucosal biopsies and the MUC2 glycoprotein levels in mucin-producing LS174T colon carcinoma cells Escherichia coli (EHEC) O82882 (Uniprot) [73
Peptidase_M43 domain-containing protein OgpA, Amuc_1119, OpeRATOR® and GlycOCATCH® Active on Fetuin and IgA. Exclusively hydrolyzes the peptide bond N-terminal to serine or threonine residues substituted with an O-glycan Akkermansia muciniphila ATCC BAA‐835 ACD04945.1 [74]
PDB 6Z2D, 6Z2O 
Table 4
The list of sulfatases and sialate O-acetylesterase that have been shown to be involved in O-mucin degradation
Sulfatase familyName or locus tagActivityOrganismAccession numbers
Sulfatase S1_4 BT3487 6S-Gal Bacteroides thetaiotaomicron VPI-5482 AAO78593.1 [43
 BT4683 3S-Gal  AAO79788.1 [43]
PDB 7ALL 
Sulfatase S1_11 BT1628 6S-GlcNAc Bacteroides thetaiotaomicron VPI-5482 AAO76735.1 [43
 BT3177   AAO78283.1 [43,44]
PDB 7P24 
Sulfatase S1_15 BT1624 6S-Gal/GalNAc Bacteroides thetaiotaomicron VPI-5482 AAO76731.1 [43,44]
PDB 7OZE 
 BT3109   AAO78215.1 [43,44]
PDB 7OZC 
 BT4631   AAO79736.1 [43,44]
PDB 7P26 
Sulfatase S1_16 BT3057 4S-Gal/4S-GalNAc Bacteroides thetaiotaomicron VPI-5482 AAO78163.1 [43,44]
PDB 7OZ9 
 BT3796   AAO78901.1 [43,44]
PDB 7OZA 
Sulfatase S1_20 BT1622 3S-Gal preferentially binding N-acetyl-d-galactosamine (GalNAc) and cleaving 3S-GalNAc Bacteroides thetaiotaomicron VPI-5482 AAO76729.1 [43]
PDB 7ANB, 7ANA 
 BT1636 3S-Gal required enzyme to access colonic mucin by Bth  AAO76743.1 [43]
PDB 7AN1, 7OQD 
Sulfatase S1_46 BT1918 3S-GlcNAc, using the N-acetyl group as an absolute specificity determinant Bacteroides thetaiotaomicron VPI-5482 AAO77025.1 [43,44]
PDB 7OZ8 
Sialate O-acetylesterase EstA Active on 9-O-acetylated sialoglycans Bacteroides fragilis NCTC 9343 WP_005794991.1 [60
Sulfatase familyName or locus tagActivityOrganismAccession numbers
Sulfatase S1_4 BT3487 6S-Gal Bacteroides thetaiotaomicron VPI-5482 AAO78593.1 [43
 BT4683 3S-Gal  AAO79788.1 [43]
PDB 7ALL 
Sulfatase S1_11 BT1628 6S-GlcNAc Bacteroides thetaiotaomicron VPI-5482 AAO76735.1 [43
 BT3177   AAO78283.1 [43,44]
PDB 7P24 
Sulfatase S1_15 BT1624 6S-Gal/GalNAc Bacteroides thetaiotaomicron VPI-5482 AAO76731.1 [43,44]
PDB 7OZE 
 BT3109   AAO78215.1 [43,44]
PDB 7OZC 
 BT4631   AAO79736.1 [43,44]
PDB 7P26 
Sulfatase S1_16 BT3057 4S-Gal/4S-GalNAc Bacteroides thetaiotaomicron VPI-5482 AAO78163.1 [43,44]
PDB 7OZ9 
 BT3796   AAO78901.1 [43,44]
PDB 7OZA 
Sulfatase S1_20 BT1622 3S-Gal preferentially binding N-acetyl-d-galactosamine (GalNAc) and cleaving 3S-GalNAc Bacteroides thetaiotaomicron VPI-5482 AAO76729.1 [43]
PDB 7ANB, 7ANA 
 BT1636 3S-Gal required enzyme to access colonic mucin by Bth  AAO76743.1 [43]
PDB 7AN1, 7OQD 
Sulfatase S1_46 BT1918 3S-GlcNAc, using the N-acetyl group as an absolute specificity determinant Bacteroides thetaiotaomicron VPI-5482 AAO77025.1 [43,44]
PDB 7OZ8 
Sialate O-acetylesterase EstA Active on 9-O-acetylated sialoglycans Bacteroides fragilis NCTC 9343 WP_005794991.1 [60

Overall, about a hundred mucin-degrading CAZymes have been biochemically characterized with substrates of defined structure, allowing the determination of their specificity towards the host and dietary glycans that could compete to feed gut bacteria. This limits the understanding, at the molecular level, of the relationships between the microbiota, its host and diet. In addition, these biochemical data are far from being exhaustive, especially when we consider that uncultured species dominate in the human gut microbiota [75]. Recently, activity-based metagenomics [76] have enabled the capture of the first CAZymes from uncultured gut bacteria (α-fucosidases, β-N-acetyl-galactosidases, β-galactosidases, β-N-acetyl-glucosidases, β-N-acetyl-neuraminidases, α- and β-mannosidases), which have proved to affect the structure of various O-, N-glycans, HMOs and human colonic mucus [26,27,77]. These studies also provided the first evidence of the over-representation of the genes coding for some of these enzymes in the human gut metagenome of patients suffering from IBD.

It is noteworthy that metagenome assemblies generated from the gut microbiota include a wide range of other organisms, apart from bacteria, such as archaea, eukaryotes and viruses that warrant a more thorough investigation [78]. For instance, gut commensal fungi are likely involved in IBD [79] but to our knowledge, none of their CAZymes has been characterized to date.

The combination of growth assays on mucins combined with transcriptomics, or proteomics [80] is currently the best strategy to highlight the CAZymes involved in mucin degradation by targeted species. These species tend to be either highly prevalent commensals or species that are associated with intestinal diseases [51].

In addition, this approach can be used to identify the enzymes from bacterial mini-consortia that act synergistically to break down the mucin network, or, on the contrary, that compete to forage on mucin glycans [81]. It is nevertheless restricted to the study of cultured species, and although ‘artificial’ consortia could be studied, they do not represent the diversity and the complexity of the bacterial and enzymatic machineries that are involved in mucus breakdown in the gut microbiome. Although those that exist are extremely interesting, there are currently very few metatranscriptomic and/or metaproteomic studies available [82,83] giving access to the battery of CAZyme-encoding genes expressed during mucin consumption by the entire microbiota, in particular in an IBD context [84].

Technically speaking, performing growth assays and transcriptomics or proteomics studies with biological and technical replicates requires grams of substrate, which is easy when such studies are performed on widely available natural substrates such as plant cell wall polysaccharides. When the focus of the studies is human mucin degradation, the challenge is very different because of the impossibility of sampling human intestinal mucus in large amounts. The solution currently used is to perform growth studies on porcine mucins, which share structural similarities with human mucin core proteins. Unfortunately, however, there are important differences in the relative abundance of core types, neutral, acidic or sulfated glycans [85] (Table 1), which are crucial in terms of enzymatic accessibility to the network of mucins. In brief, the main structural characteristics that differentiate pig gastric mucin (PGM) – one of the mostly used substrates for in vitro studies – from human colonic mucin are respectively [36,86–88]: MUC5 dominant versus MUC2 enriched mucin; cores 2 and 1 versus core 3 dominant; low degree of sialylation versus extensive sialylation (mono, di tri) and sulfate groups, leading to acidic human colonic mucus; extensive presence of blood type ABH and Lewis epitopes and i/I branches versus extensive presence of the Sda/Cad antigen (GalNAc(NeuAc-)Gal) and low abundance, mono-fucosylation, including blood group ABH antigens; galactose as the most common terminal residue, as well as presence of a sulfate group, approximately 50% of termini for both in PGM. In addition, using such complex polymeric structures does not allow for identification of the specificity of the expressed CAZymes towards a particular oligosaccharidic motif or linkage type, except using powerful but complex techniques combining high-performance anion-exchange chromatography with pulsed amperometric detection (HPAE-PAD), mass spectrometry and liquid chromatography-mass spectrometry (LC-MS) to analyze the structure of the oligosaccharides that are released during bacterial consumption of mucins or their in vitro enzymatic hydrolysis [41,89].

Screening and characterizing the substrate specificity of CAZymes can be easily performed using chromogenic glycosides mimicking the mucin chain extremities, such as pNP- or X-sugars. However, there are some mucin-acting CAZymes that do not act on these artificial substrates, leading to false negative results. It is the case, for example, of the AfcA GH95 α-fucosidase from Bifidobacterium longum JCM1254, which is inactive on pNP-fucose [90]. Other GH95 and GH29 α-fucosidases act more or less efficiently on this substrate, with catalytic efficiencies varying by up to four orders of magnitude between GH29 enzymes [55].

On the contrary, the rate of false positives might be high in screening campaigns using unpurified enzymes and chemically modified substrates, that can be recognized by the native glycosidases produced by the recombinant host. For example, when screening Escherichia coli recombinant libraries for β-galactosidase activity, the LacZ GH2 β-galactosidase is highly problematic. E. coli also produces other glycosidases, such as the GH3 AWY88947 β-N-acetylhexosaminidase, resulting in background activity that has to be systematically taken into account to get information on the ability, or not, of the target enzymes to hydrolyze the substrates under study [27].

This is of particular importance for screening genomic or metagenomic libraries for activities of mucin hydrolysis, using conventional solid-plates or micro-plate screens. Using a battery of chromogenic glycosides in primary screens to represent the structural diversity of mucin side chains, our group discovered several mucin-targeting PUL-like multigenic clusters from a metagenomic library constructed from the mucosal-associated ileal microbiota [26]. These metagenomic PULs, cloned in fosmids and expressed in E. coli, encode a cocktail of synergistic CAZymes that were proven, by ELISA assays using fluorescein-conjugated lectins, to affect the structure of human colon mucus sampled from surgical pieces. In order to circumvent the use of clinical samples, validation screening assays and substrate specificity profiling could also be performed using either mucus-secreting human cell lines [41,91], cell-based platforms displaying tunable structures and patterns of O-glycans [92], or even – although this has not been performed to date – organoids on a 2D culture system [93,94].

Regarding the rapid identification of mucin-degrading CAZymes, metagenomics has recently been paired with microfluidics, and a fluorogenic glycoside substrate was used in a novel droplet microfluidics workflow developed to directly access metagenomic PULs cloned in fosmids and expressed in E. coli [27]. Indeed, droplet microfluidics screening makes it possible to perform 106 assays per hour, with <1 mg of substrates, which is 1000 times faster and less expensive than conventional technologies. This technology is thus compatible with the fast exploration, at low cost, of large sequence spaces to identify the target enzymes. It is based on the detection and sorting of droplet hits in function of their fluorescent or absorbance levels [27,95], bearing in mind that absorbance detection is less sensitive than fluorescence detection by orders of magnitude.

Using such workflows requiring a chemically modified substrate to identify the droplet where the target enzymatic reaction occurs, validation of activity on non-chemically modified substrates (such as commercial oligosaccharides with defined structures, including HMOs and blood antigens, that are both mucin-like motifs) is crucial. In any case, profiling substrate specificity using such structurally defined oligosaccharides, is highly expensive. In primary screening, it is thus very tempting to use such mucin-like oligosaccharides with defined structures, which represent the structural complexity and diversity of physiological substrates. It would prevent screening bias and the costly and time-consuming validation steps on real substrates. Of course, screening large libraries with such oligosaccharides using solid-plates or micro-plate screens is not conceivable, given the price of these substrates. However, recently some ultra-miniaturized droplet-based microfluidics workflows have been developed, based on positive selection of bacteria (recombinant [96] or native bacteria [97]) on native, non-chemically modified oligosaccharides. Until now, these technologies have only been exemplified for the activities of metabolization of plant-derived dietary fibers. What is more, they have not so far led to the discovery of novel functional genes, since the proofs of concepts have been established with a mini-metagenomic library, or with a microbiota sample, for which the taxonomical marker genes have only been sequenced after selecting fiber-metabolizing bacteria. Nevertheless, the development of these workflows opens some very interesting prospects for ultra-high throughput functional genomics, metagenomics and culturomics studies (using single cells or bacterial consortia), targeting the activities of mucin degradation.

Several approaches have been used over the last decade to study the links between diet, gut bacteria and their enzymes, mucus structure/penetrability, and IBD [98]. In particular, key relationships have been established between the protease activities produced by both intestinal epithelium and opportunistic pathogens, mucosal biofilm structure, and IBD.

Regarding CAZymes from the intestinal microbiome, pioneer transcriptomics studies targeting B. thetaiotamicron (Bth) revealed the potential role of these enzymes in gut inflammation, in relationship with diet. Indeed, mucin-targeting Bth PULs were shown to be up-regulated in fiber-deprived diets, leading to an alteration of the mucus layer, which promoted the implantation of pathogens and/or commensal opportunists [5,99]. These PULs were also shown to be up-regulated with some particular prebiotics, such as β-galacto-oligosaccharides (GOS), and HMOs [80,100]. GOS are plant-derived oligosaccharides, but as O-mucins, they contain β-linked galactosyl units that could be recognized and hydrolyzed by some β-galactosidases or promiscous β-hexosidases or -hexosaminidases of the gut microbiome. GOS have been tested as prebiotics on experimental colitis in rats, but no impact on inflammation was observed [101]. Moreover, the effects of other prebiotics containing β-linked galactosyl units, such as lactulose, on bacterial translocation and inflammation are still unclear [32]. In contrast, consumption of HMOs, which are much more similar to mucins than GOS, seems to protect against IBD [28–30]. Due to their structural convergence, HMOs are indeed thought to act as excellent competitor substrates of mucins towards mucin-degrading CAZymes [102] (Figure 1). In addition, certain HMOs, such as 2′-fucosyllactose, can competitively inhibit the binding of enteric pathogens to epithelial cell membranes, thus modulating specific pro-inflammatory signaling molecules [30]. In healthy adults, 2′-fucosyllactose is well tolerated and promotes the growth of beneficial bifidobacteria [103]. Furthermore, infants fed with a formula containing 2′-fucosyllactose have lower inflammatory cytokine levels. In contrast, other HMOs, such as 3′-sialyllactose, have been shown to directly interact with the host, resulting in structural modification of glycans, or even in pro-inflammatory stimulation [31]. In conclusion, with such a diversity of structures and mechanisms of interaction with both the gut microbiota and the host [104], the effect of functional oligosaccharides like GOS prebiotics and HMOs in the triggering or in the prevention of inflammation is today unclear, and too few CAZymes targeting these motifs in the intestinal microbiome have been characterized to date.

Schematic representation of bowel intestinal epithelium and lumen in healthy (A), IBD condition (B) or IBD condition in presence of HMOs or GOS (C)

Figure 1
Schematic representation of bowel intestinal epithelium and lumen in healthy (A), IBD condition (B) or IBD condition in presence of HMOs or GOS (C)

In the healthy condition (A), both mucus layers (in dark, inner and sterile layer and light brown, outer layer) are present. Immune cells are present but not activated. In this condition, mucus secretion and mucus degradation by microbiota are balanced. In the IBD condition (B), mucus thickness is highly reduced and both mucus layers are invaded by microbiota, epithelial cell functions are impaired with loss of tight junctions. Immune cells are activated and dysbiosis is observed. Bacteria possibly infiltrate the epithelium. The origin of this unbalanced state could be defects in mucin O-glycosylation, resulting in lower density, higher penetrability and degradability of the inner mucus gel, the secretion of which is also affected [12,14]. With a diet supplemented with Human Milk Oligosaccharides (HMOs) and/or Galacto-Oligosaccharides (GMOs) in IBD condition (C), an improvement of the situation might be observed. Key: green bars and gray circles: commensal bacteria; dark bars: pathogenic bacteria.

Figure 1
Schematic representation of bowel intestinal epithelium and lumen in healthy (A), IBD condition (B) or IBD condition in presence of HMOs or GOS (C)

In the healthy condition (A), both mucus layers (in dark, inner and sterile layer and light brown, outer layer) are present. Immune cells are present but not activated. In this condition, mucus secretion and mucus degradation by microbiota are balanced. In the IBD condition (B), mucus thickness is highly reduced and both mucus layers are invaded by microbiota, epithelial cell functions are impaired with loss of tight junctions. Immune cells are activated and dysbiosis is observed. Bacteria possibly infiltrate the epithelium. The origin of this unbalanced state could be defects in mucin O-glycosylation, resulting in lower density, higher penetrability and degradability of the inner mucus gel, the secretion of which is also affected [12,14]. With a diet supplemented with Human Milk Oligosaccharides (HMOs) and/or Galacto-Oligosaccharides (GMOs) in IBD condition (C), an improvement of the situation might be observed. Key: green bars and gray circles: commensal bacteria; dark bars: pathogenic bacteria.

Close modal
  • There is a clear correlation between IBD, alteration of the colonic mucus and its barrier effect, and dysbiosis of the intestinal microbiota.

  • To date, a few hundred mucus-degrading CAZymes from about ten prevalent gut bacterial species have been biochemically characterized, and only a handful of CAZymes active on mucus or mucin-motifs have been identified from uncultured bacteria, demonstrating the lack of knowledge about mucin catabolic pathways in the gut microbiome.

  • The difficulties of mucus sampling, mucus interspecies structural diversity, and the cost and availability of oligosaccharides that properly mimic mucin structure, limit the identification and biochemical characterization of mucin-degrading CAZymes, and of their synergistic action to break down the mucus network.

  • Novel droplet microfluidics-based screening technologies developed recently are a great opportunity to explore, at ultra-high throughput and at low cost, the mucin-degrading functions of the gut microbiome, using either functional genomics, metagenomics or culturomics.

  • Some genes encoding mucin-degrading CAZymes are more prevalent in the microbiome of IBD patients compared with that of healthy individuals, although the role played by these biomarkers in inflammation has never been studied.

  • The action of mucin-targeting CAZymes on mucus invasion by gut bacteria and on the host response should be explored, using a combination of in vitro, ex vivo and in vivo approaches specifically designed to analyze mucus characteristics and inflammation. Also, the protective role of functional foods, more specifically that of prebiotics and HMOs competing with mucin glycans towards mucus-degrading CAZymes, should be assessed. Such holobiomics studies should help demonstrate the potential causality links between mucus degradation, its prevention through functional foods, and the control of the inflammatory response of the host.

The authors declare that there are no competing interests associated with the manuscript.

The work is funded by the ANR [grant number CAZIBD 20-CE14-0019-01].

A.L., G.P-V., C.D. and M.M-B. designed the draft of the manuscript. A.L. and G.P-V. wrote the manuscript. J-L.P. made Table 1. A.L. made Tables 2–4. S.L. made Figure 1.

CAZymes

carbohydrate-active enzymes

Gal

galactose

GalNAc

N-acetyl-d-galactosamine

Glc

glucose

GlcNac

N-acetyl-d-glucosamine

GH

glycoside hydrolase

GOS

β-galacto-oligosaccharides

HMO

human milk oligosaccharide

IBD

inflammatory bowel diseases

MUC

mucin

Neu5Ac

N-acetylneuraminic acid

1.
Burger-van Paassen
N.
,
Vincent
A.
,
Puiman
P.J.
,
van der Sluis
M.
,
Bouma
J.
,
Boehm
G.
et al.
(
2009
)
The regulation of intestinal mucin MUC2 expression by short-chain fatty acids: implications for epithelial protection
.
Biochem. J.
420
,
211
219
[PubMed]
2.
Gillois
K.
,
Lévêque
M.
,
Theodorou
V.
,
Robert
H.
and
Mercier-Bonin
M.
(
2018
)
Mucus: an underestimated gut target for environmental pollutants and food additives
.
Microorganisms
6
,
53
[PubMed]
3.
Talbot
P.
,
Radziwill-Bienkowska
J.M.
,
Kamphuis
J.B.J.
,
Steenkeste
K.
,
Bettini
S.
,
Robert
V.
et al.
(
2018
)
Food-grade TiO2 is trapped by intestinal mucus in vitro but does not impair mucin O-glycosylation and short-chain fatty acid synthesis in vivo: Implications for gut barrier protection
.
J. Nanobiotechnol.
16
,
53
[PubMed]
4.
da Silva
S.
,
Robbe-Masselot
C.
,
Ait-Belgnaoui
A.
,
Mancuso
A.
,
Mercade-Loubière
M.
,
Salvador-Cartier
C.
et al.
(
2014
)
Stress disrupts intestinal mucus barrier in rats via mucin O-glycosylation shift: prevention by a probiotic treatment
.
Am. J. Physiol. Gastrointest. Liver Physiol.
307
,
420
429
5.
Sonnenburg
E.D.
and
Sonnenburg
J.L.
(
2014
)
Starving our microbial self: The deleterious consequences of a diet deficient in microbiota-accessible carbohydrates
.
Cell Metab.
20
,
779
786
[PubMed]
6.
Coward
S.
and
Kaplan
G.G.
(
2017
)
IBD in the New World, Old World, and Your World
.
13
27
in
Inflammatory Bowel Disease. Clinical Gastroenterology.
,
Cohen
R.D.
(ed.)
Humana Press
,
Cham
.
7.
Frank
D.N.
,
Amand
A.L.S.
,
Feldman
R.A.
,
Boedeker
E.C.
,
Harpaz
N.
and
Pace
N.R.
(
2007
)
Molecular-phylogenetic characterization of microbial community imbalances in human inflammatory bowel diseases
.
PNAS
104
,
13780
13785
[PubMed]
8.
Png
C.W.
,
Lindén
S.K.
,
Gilshenan
K.S.
,
Zoetendal
E.G.
,
McSweeney
C.S.
,
Sly
L.I.
et al.
(
2010
)
Mucolytic bacteria with increased prevalence in IBD mucosa augment in vitro utilization of mucin by other bacteria
.
Am. J. Gastroenterol.
105
,
2420
2428
[PubMed]
9.
Joossens
M.
,
Huys
G.
,
Cnockaert
M.
,
de Preter
V.
,
Verbeke
K.
,
Rutgeerts
P.
et al.
(
2011
)
Dysbiosis of the faecal microbiota in patients with Crohn's disease and their unaffected relatives
.
Gut
60
,
631
637
[PubMed]
10.
Torres
J.
,
Mehandru
S.
,
Colombel
J.F.
and
Peyrin-Biroulet
L.
(
2017
)
Crohn’s disease
.
Lancet
389
,
1741
1755
[PubMed]
11.
Strugala
V.
,
Dettmar
P.W.
and
Pearson
J.P.
(
2008
)
Thickness and continuity of the adherent colonic mucus barrier in active and quiescent ulcerative colitis and Crohn's disease
.
Int. J. Clin. Pract.
62
,
762
769
[PubMed]
12.
Gersemann
M.
,
Becker
S.
,
Kübler
I.
,
Koslowski
M.
,
Wang
G.
,
Herrlinger
K.R.
et al.
(
2009
)
Differences in goblet cell differentiation between Crohn's disease and ulcerative colitis
.
Differentiation
77
,
84
94
[PubMed]
13.
Johansson
M.E.V.
,
Gustafsson
J.K.
,
Holmen-Larsson
J.
,
Jabbar
K.S.
,
Xia
L.
,
Xu
H.
et al.
(
2014
)
Bacteria penetrate the normally impenetrable inner colon mucus layer in both murine colitis models and patients with ulcerative colitis
.
Gut
63
,
281
291
[PubMed]
14.
Johansson
M.E.V.
(
2014
)
Mucus layers in inflammatory bowel disease
.
Inflamm. Bowel Dis.
20
,
2124
2131
[PubMed]
15.
El Kaoutari
A.
,
Armougom
F.
,
Gordon
J.I.
,
Raoult
D.
and
Henrissat
B.
(
2013
)
The abundance and variety of carbohydrate-active enzymes in the human gut microbiota
.
Nat. Rev. Microbiol.
11
,
497
504
[PubMed]
16.
Lapébie
P.
,
Lombard
V.
,
Drula
E.
,
Terrapon
N.
and
Henrissat
B.
(
2019
)
Bacteroidetes use thousands of enzyme combinations to break down glycans
.
Nat. Commun.
10
,
2043
[PubMed]
17.
Poxton
I.R.
,
Brown
R.
,
Sawyerr
A.
and
Ferguson
A.
(
1997
)
Mucosa-associated bacterial flora of the human colon
.
J. Med. Microbiol.
46
,
85
91
[PubMed]
18.
Swidsinski
A.
,
Weber
J.
,
Loening-Baucke
V.
,
Hale
L.P.
and
Lochs
H.
(
2005
)
Spatial organization and composition of the mucosal flora in patients with inflammatory bowel disease
.
J. Clin. Microbiol.
43
,
3380
3389
[PubMed]
19.
Derrien
M.
,
Vaughan
E.E.
,
Plugge
C.M.
and
de Vos
W.M.
(
2004
)
Akkermansia municiphila gen. nov., sp. nov., a human intestinal mucin-degrading bacterium
.
Int. J. Syst. Evol. Microbiol.
54
,
1469
1476
[PubMed]
20.
Hall
A.B.
,
Yassour
M.
,
Sauk
J.
,
Garner
A.
,
Jiang
X.
,
Arthur
T.
et al.
(
2017
)
A novel Ruminococcus gnavus clade enriched in inflammatory bowel disease patients
.
Genome Med.
9
,
103
[PubMed]
21.
Koropatkin
N.M.
,
Cameron
E.A.
and
Martens
E.C.
(
2012
)
How glycan metabolism shapes the human gut microbiota
.
Nat. Rev. Microbiol.
10
,
323
335
[PubMed]
22.
Tailford
L.E.
,
Crost
E.H.
,
Kavanaugh
D.
and
Juge
N.
(
2015
)
Mucin glycan foraging in the human gut microbiome
.
Front. Genet.
6
,
81
,
[PubMed]
23.
Cockburn
D.W.
and
Koropatkin
N.M.
(
2016
)
Polysaccharide degradation by the intestinal microbiota and its influence on human health and disease
.
J. Mol. Biol. Acad. Press
428
,
3230
3252
24.
Ndeh
D.
and
Gilbert
H.J.
(
2018
)
Biochemistry of complex glycan depolymerisation by the human gut microbiota
.
FEMS Microbiol. Rev.
42
,
146
164
25.
Brown
H.A.
and
Koropatkin
N.M.
(
2021
)
Host glycan utilization within the Bacteroidetes Sus-like paradigm
.
Glycobiology
31
,
697
706
26.
Laville
E.
,
Perrier
J.
,
Bejar
N.
,
Maresca
M.
,
Esque
J.
,
Tauzin
A.S.
et al.
(
2019
)
Investigating host microbiota relationships through functional metagenomics
.
Front. Microbiol.
10
,
1286
[PubMed]
27.
Tauzin
A.S.
,
Pereira
M.R.
,
van Vliet
L.D.
,
Colin
P.Y.
,
Laville
E.
,
Esque
J.
et al.
(
2020
)
Investigating host-microbiome interactions by droplet-based microfluidics
.
Microbiome
8
,
141
[PubMed]
28.
Bode
L.
(
2009
)
Human milk oligosaccharides: prebiotics and beyond
.
Nutr. Rev.
67
,
S183
191
[PubMed]
29.
Fuhrer
A.
,
Sprenger
N.
,
Kurakevich
E.
,
Borsig
L.
,
Chassard
C.
and
Hennet
T.
(
2010
)
Milk sialyllactose influences colitis in mice through selective intestinal bacterial colonization
.
J. Exp. Med.
207
,
2843
2854
[PubMed]
30.
He
Y.Y.
,
Liu
S.B.
,
Kling
D.E.
,
Leone
S.
,
Lawlor
N.T.
,
Huang
Y.
et al.
(
2016
)
The human milk oligosaccharide 2′-fucosyllactose modulates CD14 expression in human enterocytes, thereby attenuating LPS-induced inflammation
.
Gut
65
,
33
46
[PubMed]
31.
Kurakevich
E.
,
Hennet
T.
,
Hausmann
M.
,
Rogler
G.
and
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
[PubMed]
32.
Looijer-van Langen
M.A.C.
and
Dieleman
L.A.
(
2009
)
Prebiotics in chronic intestinal inflammation
.
Inflamm. Bowel Dis.
15
,
454
462
[PubMed]
33.
Bansil
R.
and
Turner
B.S.
(
2018
)
The biology of mucus: composition, synthesis and organization
.
Adv. Drug Deliv. Rev.
124
,
3
15
34.
Reily
C.
,
Stewart
T.J.
,
Renfrow
M.B.
and
Novak
J.
(
2019
)
Glycosylation in health and disease
.
Nat. Rev. Nephrol.
15
,
346
366
35.
Arike
L.
and
Hansson
G.C.
(
2016
)
The densely O-glycosylated MUC2 mucin protects the intestine and provides food for the commensal bacteria
.
J. Mol. Biol. Academic Press
428
,
3221
3229
[PubMed]
36.
Larsson
J.M.H.
,
Karlsson
H.
,
Sjövall
H.
and
Hansson
G.C.
(
2009
)
A complex, but uniform O-glycosylation of the human MUC2 mucin from colonic biopsies analyzed by nanoLC/MSn
.
Glycobiology
19
,
756
766
[PubMed]
37.
Ravcheev
D.A.
,
Godzik
A.
,
Osterman
A.L.
and
Rodionov
D.A.
(
2013
)
Polysaccharides utilization in human gut bacterium Bacteroides thetaiotaomicron: Comparative genomics reconstruction of metabolic and regulatory networks
.
BMC Genomics
14
,
873
,
[PubMed]
,
38.
Glover
J.S.
,
Ticer
T.D.
and
Engevik
M.A.
(
2022
)
Characterizing the mucin-degrading capacity of the human gut microbiota
.
Sci. Rep.
12
,
8456
[PubMed]
39.
Terrapon
N.
,
Lombard
V.
,
Drula
É.
,
Lapébie
P.
,
Al-Masaudi
S.
,
Gilbert
H.J.
et al.
(
2018
)
PULDB: The expanded database of Polysaccharide Utilization Loci
.
Nucleic Acids Res.
46
,
D677
D683
[PubMed]
40.
Martens
E.C.
,
Chiang
H.C.
and
Gordon
J.I.
(
2008
)
Mucosal glycan foraging enhances fitness and transmission of a saccharolytic human gut bacterial symbiont
.
Cell Host Microbe
4
,
447
457
[PubMed]
41.
Crouch
L.I.
,
Liberato
M.v.
,
Urbanowicz
P.A.
,
Baslé
A.
,
Lamb
C.A.
,
Stewart
C.J.
et al.
(
2020
)
Prominent members of the human gut microbiota express endo-acting O-glycanases to initiate mucin breakdown
.
Nat. Commun.
11
,
4017
[PubMed]
42.
Barbeyron
T.
,
Brillet-Guéguen
L.
,
Carré
W.
,
Carrière
C.
,
Caron
C.
,
Czjzek
M.
et al.
(
2016
)
Matching the diversity of sulfated biomolecules: Creation of a classification database for sulfatases reflecting their substrate specificity
.
PloS ONE
11
,
e0164846
[PubMed]
43.
Luis
A.S.
,
Jin
C.
,
Pereira
G.V.
,
Glowacki
R.W.P.
,
Gugel
S.R.
,
Singh
S.
et al.
(
2021
)
A single sulfatase is required to access colonic mucin by a gut bacterium
.
Nature
598
,
332
337
[PubMed]
44.
Luis
A.S.
,
Baslé
A.
,
Byrne
D.P.
,
Wright
G.S.A.
,
London
J.A.
,
Jin
C.
et al.
(
2022
)
Sulfated glycan recognition by carbohydrate sulfatases of the human gut microbiota
.
Nat. Chem. Biol.
18
,
841
849
[PubMed]
45.
Pudlo
N.A.
,
Urs
K.
,
Kumar
S.S.
,
German
J.B.
,
Mills
D.A.
and
Martens
E.C.
(
2015
)
Symbiotic human gut bacteria with variable metabolic priorities for host mucosal glycans
.
mBio
6
,
e01282
15
[PubMed]
46.
Donaldson
G.P.
,
Chou
W.C.
,
Manson
A.L.
,
Rogov
P.
,
Abeel
T.
,
Bochicchio
J.
et al.
(
2020
)
Spatially distinct physiology of Bacteroides fragilis within the proximal colon of gnotobiotic mice
.
Nat. Microbiol.
5
,
746
756
[PubMed]
47.
Ottman
N.
,
Davids
M.
,
Suarez-Diez
M.
,
Boeren
S.
,
Schaap
P.J.
,
dos Santos
V.A.P.M.
et al.
(
2017
)
Genome-scale model and omics analysis of metabolic capacities of Akkermansia muciniphila reveal a preferential mucin-degrading lifestyle
.
Appl. Environ. Microbiol.
83
,
e01014
17
[PubMed]
48.
Shin
J.
,
Noh
J.R.
,
Chang
D.H.
,
Kim
Y.H.
,
Kim
M.H.
,
Lee
E.S.
et al.
(
2019
)
Elucidation of Akkermansia muciniphila probiotic traits driven by mucin depletion
.
Front. Microbiol.
10
,
1137
[PubMed]
49.
Kostopoulos
I.
,
Aalvink
S.
,
Kovatcheva-Datchary
P.
,
Nijsse
B.
,
Bäckhed
F.
,
Knol
J.
et al.
(
2021
)
A Continuous Battle for Host-Derived Glycans Between a Mucus Specialist and a Glycan Generalist in vitro and in vivo
.
Front. Microbiol.
12
,
632454
,
[PubMed]
50.
Liu
X.
,
Zhao
F.
,
Liu
H.
,
Xie
Y.
,
Zhao
D.
and
Li
C.
(
2021
)
Transcriptomics and metabolomics reveal the adaption of Akkermansia muciniphila to high mucin by regulating energy homeostasis
.
Sci. Rep.
11
,
9073
[PubMed]
51.
Crost
E.H.
,
Tailford
L.E.
,
le Gall
G.
,
Fons
M.
,
Henrissat
B.
and
Juge
N.
(
2013
)
Utilisation of Mucin Glycans by the Human Gut Symbiont Ruminococcus gnavus Is Strain-Dependent
.
PloS ONE
8
,
e76341
[PubMed]
52.
Turroni
F.
,
Milani
C.
,
van Sinderen
D.
and
Ventura
M.
(
2011
)
Genetic strategies for mucin metabolism in Bifidobacterium bifidum PRL2010: An example of possible human-microbe co-evolution
.
Gut Microbes
2
,
183
189
[PubMed]
53.
Kosciow
K.
and
Deppenmeier
U.
(
2020
)
Characterization of three novel β-galactosidases from Akkermansia muciniphila involved in mucin degradation
.
Int. J. Biol. Macromol.
149
,
331
340
[PubMed]
54.
Pichler
M.J.
,
Yamada
C.
,
Shuoker
B.
,
Alvarez-Silva
C.
,
Gotoh
A.
,
Leth
M.L.
et al.
(
2020
)
Butyrate producing colonic Clostridiales metabolise human milk oligosaccharides and cross feed on mucin via conserved pathways
.
Nat. Commun.
11
,
3285
[PubMed]
55.
Wu
H.
,
Rebello
O.
,
Crost
E.H.
,
Owen
C.D.
,
Walpole
S.
,
Bennati-Granier
C.
et al.
(
2021
)
Fucosidases from the human gut symbiont Ruminococcus gnavus
.
Cell. Mol. Life Sci.
78
,
675
693
[PubMed]
56.
Fan
S.
,
Zhang
H.
,
Chen
X.
,
Lu
L.
,
Xu
L.
and
Xiao
M.
(
2016
)
Cloning, characterization, and production of three α-l-fucosidases from Clostridium perfringens ATCC 13124
.
J. Basic Microbiol.
56
,
347
357
[PubMed]
57.
Shuoker
B.
,
Pichler
M.J.
,
Jin
C.
,
Hiroka
S.
,
Wu
H.
,
Gascueña
A.M.
et al.
(
2022
)
Sialidases and Fucosidases of Akkermansia muciniphila are crucial for growth on mucin and nutrient sharing with mucus-associated gut bacteria
.
bioRxiv
58.
Rahfeld
P.
,
Wardman
J.F.
,
Mehr
K.
,
Huff
D.
,
Morgan-Lang
C.
,
Chen
H.M.
et al.
(
2019
)
Prospecting for microbial α-N-acetylgalactosaminidases yields a new class of GH31 O-glycanase
.
J. Biol. Chem.
294
,
16400
16415
[PubMed]
59.
Tailford
L.E.
,
Owen
C.D.
,
Walshaw
J.
,
Crost
E.H.
,
Hardy-Goddard
J.
,
le Gall
G.
et al.
(
2015
)
Discovery of intramolecular trans-sialidases in human gut microbiota suggests novel mechanisms of mucosal adaptation
.
Nat. Commun.
6
,
7624
,
[PubMed]
60.
Robinson
L.S.
,
Lewis
W.G.
and
Lewis
A.L.
(
2017
)
The sialate O-acetylesterase EstA from gut Bacteroidetes species enables sialidase-mediated cross-species foraging of 9-O-acetylated sialoglycans
.
J. Biol. Chem.
292
,
11861
11872
[PubMed]
61.
Guo
B.S.
,
Zheng
F.
,
Crouch
L.
,
Cai
Z.P.
,
Wang
M.
,
Bolam
D.N.
et al.
(
2018
)
Cloning, purification and biochemical characterisation of a GH35 beta-1,3/beta-1,6-galactosidase from the mucin-degrading gut bacterium Akkermansia muciniphila
.
Glycoconj. J.
35
,
255
263
[PubMed]
62.
Kosciow
K.
and
Deppenmeier
U.
(
2019
)
Characterization of a phospholipid-regulated β-galactosidase from Akkermansia muciniphila involved in mucin degradation
.
Microbiol. Open
8
,
e00796
[PubMed]
63.
Henderson
I.R.
,
Czeczulin
J.
,
Eslava
C.
,
Noriega
F.
and
Nataro
J.P.
(
1999
)
Characterization of pic, a secreted protease of Shigella flexneri and enteroaggregative Escherichia coli
.
Infect. Immun.
67
,
5587
5596
[PubMed]
64.
Nakjang
S.
,
Ndeh
D.A.
,
Wipat
A.
,
Bolam
D.N.
and
Hirt
R.P.
(
2012
)
A novel extracellular metallopeptidase domain shared by animal Host-Associated mutualistic and pathogenic microbes
.
PloS ONE
7
,
e30287
[PubMed]
65.
Noach
I.
,
Ficko-Blean
E.
,
Pluvinage
B.
,
Stuart
C.
,
Jenkins
M.L.
,
Brochu
D.
et al.
(
2017
)
Recognition of protein-linked glycans as a determinant of peptidase activity
.
Proc. Natl. Acad. Sci. U.S A.
114
,
E679
E688
[PubMed]
66.
Shon
D.J.
,
Malaker
S.A.
,
Pedram
K.
,
Yang
E.
,
Krishnan
V.
,
Dorigo
O.
et al.
(
2020
)
An enzymatic toolkit for selective proteolysis, detection, and visualization of mucin-domain glycoproteins
.
Proc. Natl. Acad. Sci. U. S. A.
117
,
21299
21307
[PubMed]
67.
Nesta
B.
,
Valeri
M.
,
Spagnuolo
A.
,
Rosini
R.
,
Mora
M.
,
Donato
P.
et al.
(
2014
)
SslE Elicits Functional Antibodies That Impair In Vitro Mucinase Activity and In Vivo Colonization by Both Intestinal and Extraintestinal Escherichia coli Strains
.
PLoS Pathog.
10
,
e1004124
[PubMed]
68.
Luo
Q.
,
Kumar
P.
,
Vickers
T.J.
,
Sheikh
A.
,
Lewis
W.G.
,
Rasko
D.A.
et al.
(
2014
)
Enterotoxigenic Escherichia coli secretes a highly conserved mucin-degrading metalloprotease to effectively engage intestinal epithelial cells
.
Infect. Immun.
82
,
509
521
[PubMed]
69.
Shon
D.J.
,
Fernandez
D.
,
Riley
N.M.
,
Ferracane
M.J.
and
Bertozzi
C.R.
(
2022
)
Structure-guided mutagenesis of a mucin-selective metalloprotease from Akkermansia muciniphila alters substrate preferences
.
J. Biol. Chem.
298
,
101917
[PubMed]
70.
Taleb
V.
,
Liao
Q.
,
Narimatsu
Y.
,
García-García
A.
,
Compañón
I.
,
Borges
R.J.
et al.
(
2022
)
Structural and mechanistic insights into the cleavage of clustered O-glycan patches-containing glycoproteins by mucinases of the human gut
.
Nat. Commun.
13
,
4324
[PubMed]
71.
Bardoel
B.W.
,
Hartsink
D.
,
Vughs
M.M.
,
de Haas
C.J.C.
,
van Strijp
J.A.G.
and
van Kessel
K.P.M.
(
2012
)
Identification of an immunomodulating metalloprotease of Pseudomonas aeruginosa (IMPa)
.
Cell. Microbiol.
14
,
902
913
[PubMed]
72.
Pluvinage
B.
,
Ficko-Blean
E.
,
Noach
I.
,
Stuart
C.
,
Thompson
N.
,
McClure
H.
et al.
(
2021
)
Architecturally complex O-glycopeptidases are customized for mucin recognition and hydrolysis
.
Proc. Natl. Acad. Sci. U.S.A.
118
,
e2019220118
[PubMed]
73.
Hews
C.L.
,
Tran
S.L.
,
Wegmann
U.
,
Brett
B.
,
Walsham
A.D.S.
,
Kavanaugh
D.
et al.
(
2017
)
The StcE metalloprotease of enterohaemorrhagic Escherichia coli reduces the inner mucus layer and promotes adherence to human colonic epithelium ex vivo
.
Cell. Microbiol.
19
,
e12717
[PubMed]
74.
Trastoy
B.
,
Naegeli
A.
,
Anso
I.
,
Sjögren
J.
and
Guerin
M.E.
(
2020
)
Structural basis of mammalian mucin processing by the human gut O-glycopeptidase OgpA from Akkermansia muciniphila
.
Nat. Commun.
11
,
4844
[PubMed]
75.
Almeida
A.
,
Nayfach
S.
,
Boland
M.
,
Strozzi
F.
,
Beracochea
M.
,
Shi
Z.J.
et al.
(
2021
)
A unified catalog of 204,938 reference genomes from the human gut microbiome
.
Nat. Biotechnol.
39
,
105
114
[PubMed]
76.
Tasse
L.
,
Bercovici
J.
,
Pizzut-Serin
S.
,
Robe
P.
,
Tap
J.
,
Klopp
C.
et al.
(
2010
)
Functional metagenomics to mine the human gut microbiome for dietary fiber catabolic enzymes
.
Genome Res.
20
,
1605
1612
[PubMed]
77.
Ladevéze
S.
,
Tarquis
L.
,
Cecchini
D.A.
,
Bercovici
J.
,
André
I.
,
Topham
C.M.
et al.
(
2013
)
Role of glycoside phosphorylases in mannose foraging by human gut bacteria
.
J. Biol. Chem.
288
,
32370
32383
[PubMed]
78.
Almeida
A.
,
Mitchell
A.L.
,
Boland
M.
,
Forster
S.C.
,
Gloor
G.B.
,
Tarkowska
A.
et al.
(
2019
)
A new genomic blueprint of the human gut microbiota
.
Nature
568
,
499
504
[PubMed]
79.
Pérez
J.C.
(
2021
)
Fungi of the human gut microbiota: roles and significance
.
Int. J. Med. Microbiol.
311
,
151490
,
[PubMed]
80.
van Bueren
A.L.
,
Mulder
M.
,
van Leeuwen
S.
and
Dijkhuizen
L.
(
2017
)
Prebiotic galacto-oligosaccharides activate mucin and pectic galactan utilization pathways in the human gut symbiont Bacteroides thetaiotaomicron
.
Sci. Rep.
7
,
40478
[PubMed]
81.
Chia
L.W.
,
Hornung
B.V.H.
,
Aalvink
S.
,
Schaap
P.J.
,
de Vos
W.M.
,
Knol
J.
et al.
(
2018
)
Deciphering the trophic interaction between Akkermansia muciniphila and the butyrogenic gut commensal Anaerostipes caccae using a metatranscriptomic approach
.
Antonie van Leeuwenhoek, Int. J. General Mol. Microbiol.
111
,
859
873
[PubMed]
82.
Li
L.
,
Zhang
X.
,
Ning
Z.
,
Mayne
J.
,
Moore
J.I.
,
Butcher
J.
et al.
(
2018
)
Evaluating in vitro culture medium of gut microbiome with orthogonal experimental design and a metaproteomics approach
.
J. Proteome Res.
17
,
154
163
[PubMed]
83.
Li
A.
,
Laville
E.
,
Tarquis
L.
,
Lombard
V.
,
Ropartz
D.
,
Terrapon
N.
et al.
(
2020
)
Analysis of the diversity of the glycoside hydrolase family 130 in mammal gut microbiomes reveals a novel mannoside-phosphorylase function
.
Microb. Genom.
6
,
mgen000404
[PubMed]
84.
Erickson
A.R.
,
Cantarel
B.L.
,
Lamendella
R.
,
Darzi
Y.
,
Mongodin
E.F.
,
Pan
C.
et al.
(
2012
)
Integrated metagenomics/metaproteomics reveals human host-microbiota signatures of Crohn's disease
.
PloS ONE
7
,
e49138
[PubMed]
85.
Nordman
H.
,
Davies
J.R.
,
Herrmann
A.
,
Karlsson
N.G.
,
Hansson
G.C.
and
Carlstedt
I.
(
1997
)
Mucus glycoproteins from pig gastric mucosa: identification of different mucin populations from the surface epithelium
.
Biochem. J.
326
,
903
910
[PubMed]
86.
Padra
M.
,
Adamczyk
B.
,
Benktander
J.
,
Flahou
B.
,
Skoog
E.C.
,
Padra
J.T.
et al.
(
2018
)
Helicobacter suis binding to carbohydrates on human and porcine gastric mucins and glycolipids occurs via two modes
.
Virulence
9
,
898
918
[PubMed]
87.
Robbe
C.
,
Capon
C.
,
Maes
E.
,
Rousset
M.
,
Zweibaum
A.
,
Zanetta
J.P.
et al.
(
2003
)
Evidence of regio-specific glycosylation in human intestinal mucins: Presence of an acidic gradient along the intestinal tract
.
J. Biol. Chem.
278
,
46337
46348
[PubMed]
88.
Jin
C.
,
Kenny
D.T.
,
Skoog
E.C.
,
Padra
M.
,
Adamczyk
B.
,
Vitizeva
V.
et al.
(
2017
)
Structural diversity of human gastric mucin glycans
.
Mol. Cell. Proteomics
16
,
743
758
[PubMed]
89.
Robbe
C.
,
Capon
C.
,
Coddeville
B.
and
Michalski
J.C.
(
2004
)
Structural diversity and specific distribution of O-glycans in normal human mucins along the intestinal tract
.
Biochem. J.
384
,
307
316
,
[PubMed]
90.
Katayama
T.
,
Fujita
K.
and
Yamamoto
K.
(
2005
)
Novel bifidobacterial glycosidases acting on sugar chains of mucin glycoproteins
.
J. Biosci. Bioeng.
99
,
457
465
[PubMed]
91.
Gagnon
M.
,
Zihler Berner
A.
,
Chervet
N.
,
Chassard
C.
and
Lacroix
C.
(
2013
)
Comparison of the Caco-2, HT-29 and the mucus-secreting HT29-MTX intestinal cell models to investigate Salmonella adhesion and invasion
.
J. Microbiol. Methods
94
,
274
279
[PubMed]
92.
Nason
R.
,
Büll
C.
,
Konstantinidi
A.
,
Sun
L.
,
Ye
Z.
,
Halim
A.
et al.
(
2021
)
Display of the human mucinome with defined O-glycans by gene engineered cells
.
Nat. Commun.
12
,
4070
[PubMed]
93.
Wang
Y.
,
Kim
R.
,
Sims
C.E.
and
Allbritton
N.L.
(
2019
)
Building a thick mucus hydrogel layer to improve the physiological relevance of in vitro primary colonic epithelial models
.
CMGH
8
,
653.e5
655.e5
[PubMed]
94.
Zhao
A.
,
Qin
H.
,
Sun
M.
,
Tang
M.
,
Mei
J.
,
Ma
K.
et al.
(
2021
)
Chemical conversion of human epidermal stem cells into intestinal goblet cells for modeling mucus-microbe interaction and therapy
.
Sci. Adv.
7
,
eabb2213
[PubMed]
95.
Gielen
F.
,
Hours
R.
,
Emond
S.
,
Fischlechner
M.
,
Schell
U.
and
Hollfelder
F.
(
2016
)
Ultra-high-throughput-directed enzyme evolution by absorbance-activated droplet sorting (AADS)
.
Proc. Natl. Acad. Sci. U. S. A.
113
,
E7383
E7389
[PubMed]
96.
Dagkesamanskaya
A.
,
Langer
K.
,
Tauzin
A.S.
,
Rouzeau
C.
,
Lestrade
D.
,
Potocki-Veronese
G.
et al.
(
2018
)
Use of photoswitchable fluorescent proteins for droplet-based microfluidic screening
.
J. Microbiol. Methods
147
,
59
65
[PubMed]
97.
Villa
M.M.
,
Bloom
R.J.
,
Silverman
J.D.
,
Durand
H.K.
,
Jiang
S.
,
Wu
A.
et al.
(
2020
)
Interindividual variation in dietary carbohydrate metabolism by gut bacteria revealed with droplet microfluidic culture
.
mSystems
5
,
e00864
19
[PubMed]
98.
Kamphuis
J.B.J.
,
Mercier-Bonin
M.
,
Eutamène
H.
and
Theodorou
V.
(
2017
)
Mucus organisation is shaped by colonic content; A new view
.
Sci. Rep.
7
,
8527
[PubMed]
99.
Desai
M.S.
,
Seekatz
A.M.
,
Koropatkin
N.M.
,
Kamada
N.
,
Hickey
C.A.
,
Wolter
M.
et al.
(
2016
)
A Dietary Fiber-Deprived Gut Microbiota Degrades the Colonic Mucus Barrier and Enhances Pathogen Susceptibility
.
Cell
167
,
1339.e21
1353.e21
[PubMed]
100.
Marcobal
A.
,
Barboza
M.
,
Sonnenburg
E.D.
,
Pudlo
N.
,
Martens
E.C.
,
Desai
P.
et al.
(
2011
)
Bacteroides in the infant gut consume milk oligosaccharides via mucus-utilization pathways
.
Cell Host Microbe
10
,
507
514
[PubMed]
101.
Holma
R.
,
Juvonen
P.
,
Asmawi
M.Z.
,
Vapaatalo
H.
and
Korpela
R.
(
2002
)
Galacto-oligosaccharides stimulate the growth of bifidobacteria but fail to attenuate inflammation in experimental colitis in rats
.
Scand. J. Gastroenterol.
37
,
1042
1047
[PubMed]
102.
Belzer
C.
(
2022
)
Nutritional strategies for mucosal health: the interplay between microbes and mucin glycans
.
Trends Microbiol.
30
,
13
21
,
[PubMed]
103.
Elison
E.
,
Vigsnaes
L.K.
,
Rindom Krogsgaard
L.
,
Rasmussen
J.
,
Sorensen
N.
,
McConnell
B.
et al.
(
2016
)
Oral supplementation of healthy adults with 2′-O-fucosyllactose and lacto-N-neotetraose is well tolerated and shifts the intestinal microbiota
.
Br. J. Nutr.
116
,
1356
1368
[PubMed]
104.
Angeloni
S.
,
Ridet
J.L.
,
Kusy
N.
,
Gao
H.
,
Crevoisier
F.
,
Guinchard
S.
et al.
(
2005
)
Glycoprofiling with micro-arrays of glycoconjugates and lectins
.
Glycobiology
15
,
31
41
[PubMed]
This is an open access article published by Portland Press Limited on behalf of the Biochemical Society and distributed under the Creative Commons Attribution License 4.0 (CC BY).