Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
PLoS Biol ; 19(12): e3001498, 2021 12.
Article in English | MEDLINE | ID: mdl-34936658

ABSTRACT

The human gut symbiont Ruminococcus gnavus displays strain-specific repertoires of glycoside hydrolases (GHs) contributing to its spatial location in the gut. Sequence similarity network analysis identified strain-specific differences in blood-group endo-ß-1,4-galactosidase belonging to the GH98 family. We determined the substrate and linkage specificities of GH98 from R. gnavus ATCC 29149, RgGH98, against a range of defined oligosaccharides and glycoconjugates including mucin. We showed by HPAEC-PAD and LC-FD-MS/MS that RgGH98 is specific for blood group A tetrasaccharide type II (BgA II). Isothermal titration calorimetry (ITC) and saturation transfer difference (STD) NMR confirmed RgGH98 affinity for blood group A over blood group B and H antigens. The molecular basis of RgGH98 strict specificity was further investigated using a combination of glycan microarrays, site-directed mutagenesis, and X-ray crystallography. The crystal structures of RgGH98 in complex with BgA trisaccharide (BgAtri) and of RgGH98 E411A with BgA II revealed a dedicated hydrogen network of residues, which were shown by site-directed mutagenesis to be critical to the recognition of the BgA epitope. We demonstrated experimentally that RgGH98 is part of an operon of 10 genes that is overexpresssed in vitro when R. gnavus ATCC 29149 is grown on mucin as sole carbon source as shown by RNAseq analysis and RT-qPCR confirmed RgGH98 expression on BgA II growth. Using MALDI-ToF MS, we showed that RgGH98 releases BgAtri from mucin and that pretreatment of mucin with RgGH98 confered R. gnavus E1 the ability to grow, by enabling the E1 strain to metabolise BgAtri and access the underlying mucin glycan chain. These data further support that the GH repertoire of R. gnavus strains enable them to colonise different nutritional niches in the human gut and has potential applications in diagnostic and therapeutics against infection.


Subject(s)
Clostridiales/metabolism , Mucin-1/metabolism , ABO Blood-Group System/immunology , Blood Group Antigens/immunology , Clostridiales/genetics , Clostridiales/physiology , Gastrointestinal Microbiome , Gastrointestinal Tract , Glycoside Hydrolases/metabolism , Humans , Mucins/metabolism , Oligosaccharides/metabolism , Polysaccharides/metabolism , Ruminococcus/genetics , Ruminococcus/metabolism , Substrate Specificity , Tandem Mass Spectrometry/methods
2.
Biotechnol Lett ; 46(2): 201-211, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38280177

ABSTRACT

OBJECTIVES: Apiosidases are enzymes that cleave the glycosidic bond between the monosaccharides linked to apiose, a branched chain furanose found in the cell walls of vascular plants and aquatic monocots. There is biotechnological interest in this enzyme group because apiose is the flavor-active compound of grapes, fruit juice, and wine, and the monosaccharide is found to be a plant secondary metabolite with pharmaceutical properties. However, functional and structural studies of this enzyme family are scarce. Recently, a glycoside hydrolase family member GH140 was isolated from Bacteroides thetaiotaomicron and identified as an endo-apiosidase. RESULTS: The structural characterization and functional identification of a second GH140 family enzyme, termed MmApi, discovered through mangrove soil metagenomic approach, are described. Among the various substrates tested, MmApi exhibited activity on an apiose-containing oligosaccharide derived from the pectic polysaccharide rhamnogalacturonan-II. While the crystallographic model of MmApi was similar to the endo-apiosidase from Bacteroides thetaiotaomicron, differences in the shape of the binding sites indicated that MmApi could cleave apioses within oligosaccharides of different compositions. CONCLUSION: This enzyme represents a novel tool for researchers interested in studying the physiology and structure of plant cell walls and developing biocatalytic strategies for drug and flavor production.


Subject(s)
Microbiota , Polysaccharides , Oligosaccharides/chemistry , Glycoside Hydrolases/genetics , Glycoside Hydrolases/chemistry , Monosaccharides
4.
Nature ; 544(7648): 65-70, 2017 04 06.
Article in English | MEDLINE | ID: mdl-28329766

ABSTRACT

The metabolism of carbohydrate polymers drives microbial diversity in the human gut microbiota. It is unclear, however, whether bacterial consortia or single organisms are required to depolymerize highly complex glycans. Here we show that the gut bacterium Bacteroides thetaiotaomicron uses the most structurally complex glycan known: the plant pectic polysaccharide rhamnogalacturonan-II, cleaving all but 1 of its 21 distinct glycosidic linkages. The deconstruction of rhamnogalacturonan-II side chains and backbone are coordinated to overcome steric constraints, and the degradation involves previously undiscovered enzyme families and catalytic activities. The degradation system informs revision of the current structural model of rhamnogalacturonan-II and highlights how individual gut bacteria orchestrate manifold enzymes to metabolize the most challenging glycan in the human diet.


Subject(s)
Bacteroides thetaiotaomicron/enzymology , Bacteroides thetaiotaomicron/metabolism , Biocatalysis , Gastrointestinal Tract/microbiology , Glycoside Hydrolases/metabolism , Pectins/chemistry , Pectins/metabolism , Bacteroides thetaiotaomicron/growth & development , Borates/chemistry , Borates/metabolism , Catalytic Domain , Gastrointestinal Microbiome , Glycoside Hydrolases/chemistry , Glycoside Hydrolases/classification , Humans , Models, Molecular , Substrate Specificity
5.
Cell Mol Life Sci ; 78(2): 675-693, 2021 Jan.
Article in English | MEDLINE | ID: mdl-32333083

ABSTRACT

The availability and repartition of fucosylated glycans within the gastrointestinal tract contributes to the adaptation of gut bacteria species to ecological niches. To access this source of nutrients, gut bacteria encode α-L-fucosidases (fucosidases) which catalyze the hydrolysis of terminal α-L-fucosidic linkages. We determined the substrate and linkage specificities of fucosidases from the human gut symbiont Ruminococcus gnavus. Sequence similarity network identified strain-specific fucosidases in R. gnavus ATCC 29149 and E1 strains that were further validated enzymatically against a range of defined oligosaccharides and glycoconjugates. Using a combination of glycan microarrays, mass spectrometry, isothermal titration calorimetry, crystallographic and saturation transfer difference NMR approaches, we identified a fucosidase with the capacity to recognize sialic acid-terminated fucosylated glycans (sialyl Lewis X/A epitopes) and hydrolyze α1-3/4 fucosyl linkages in these substrates without the need to remove sialic acid. Molecular dynamics simulation and docking showed that 3'-Sialyl Lewis X (sLeX) could be accommodated within the binding site of the enzyme. This specificity may contribute to the adaptation of R. gnavus strains to the infant and adult gut and has potential applications in diagnostic glycomic assays for diabetes and certain cancers.


Subject(s)
Bacterial Proteins/metabolism , Clostridiales/metabolism , Gastrointestinal Microbiome , alpha-L-Fucosidase/metabolism , Bacterial Proteins/chemistry , Clostridiales/chemistry , Clostridiales/enzymology , Gastrointestinal Tract/microbiology , Glycoconjugates/metabolism , Humans , Oligosaccharides/metabolism , Polysaccharides/metabolism , Substrate Specificity , alpha-L-Fucosidase/chemistry
6.
J Biol Chem ; 295(52): 18625-18637, 2020 12 25.
Article in English | MEDLINE | ID: mdl-33097594

ABSTRACT

Pectins are a major dietary nutrient source for the human gut microbiota. The prominent gut microbe Bacteroides thetaiotaomicron was recently shown to encode the founding member (BT1017) of a new family of pectin methylesterases essential for the metabolism of the complex pectin rhamnogalacturonan-II (RG-II). However, biochemical and structural knowledge of this family is lacking. Here, we showed that BT1017 is critical for the metabolism of an RG-II-derived oligosaccharide ΔBT1017oligoB generated by a BT1017 deletion mutant (ΔBT1017) during growth on carbohydrate extract from apple juice. Structural analyses of ΔBT1017oligoB using a combination of enzymatic, mass spectrometric, and NMR approaches revealed that it is a bimethylated nonaoligosaccharide (GlcA-ß1,4-(2-O-Me-Xyl-α1,3)-Fuc-α1,4-(GalA-ß1,3)-Rha-α1,3-Api-ß1,2-(Araf-α1,3)-(GalA-α1,4)-GalA) containing components of the RG-II backbone and its side chains. We showed that the catalytic module of BT1017 adopts an α/ß-hydrolase fold, consisting of a central twisted 10-stranded ß-sheet sandwiched by several α-helices. This constitutes a new fold for pectin methylesterases, which are predominantly right-handed ß-helical proteins. Bioinformatic analyses revealed that the family is dominated by sequences from prominent genera of the human gut microbiota, including Bacteroides and Prevotella Our re-sults not only highlight the critical role played by this family of enzymes in pectin metabolism but also provide new insights into the molecular basis of the adaptation of B. thetaiotaomicron to the human gut.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Bacteroides thetaiotaomicron/enzymology , Carboxylic Ester Hydrolases/chemistry , Carboxylic Ester Hydrolases/metabolism , Gastrointestinal Microbiome , Oligosaccharides/metabolism , Bacteroides thetaiotaomicron/growth & development , Crystallography, X-Ray , Humans , Models, Molecular , Phylogeny , Protein Conformation
7.
J Biol Chem ; 294(19): 7711-7721, 2019 05 10.
Article in English | MEDLINE | ID: mdl-30877196

ABSTRACT

The metabolism of carbohydrate polymers drives microbial diversity in the human gut microbiome. The selection pressures in this environment have spurred the evolution of a complex reservoir of microbial genes encoding carbohydrate-active enzymes (CAZymes). Previously, we have shown that the human gut bacterium Bacteroides thetaiotaomicron (Bt) can depolymerize the most structurally complex glycan, the plant pectin rhamnogalacturonan II (RGII), commonly found in the human diet. Previous investigation of the RGII-degrading apparatus in Bt identified BT0997 as a new CAZyme family, classified as glycoside hydrolase 138 (GH138). The mechanism of substrate recognition by GH138, however, remains unclear. Here, using synthetic substrates and biochemical assays, we show that BT0997 targets the d-galacturonic acid-α-1,2-l-rhamnose linkage in chain A of RGII and that it absolutely requires the presence of a second d-galacturonic acid side chain (linked ß-1,3 to l-rhamnose) for activity. NMR analysis revealed that BT0997 operates through a double displacement retaining mechanism. We also report the crystal structure of a BT0997 homolog, BPA0997 from Bacteroides paurosaccharolyticus, in complex with ligands at 1.6 Å resolution. The structure disclosed that the enzyme comprises four domains, including a catalytic TIM (α/ß)8 barrel. Characterization of several BT0997 variants identified Glu-294 and Glu-361 as the catalytic acid/base and nucleophile, respectively, and we observed a chloride ion close to the active site. The three-dimensional structure and bioinformatic analysis revealed that two arginines, Arg-332 and Arg-521, are key specificity determinants of BT0997 in targeting d-galacturonic acid residues. In summary, our study reports the first structural and mechanistic analyses of GH138 enzymes.


Subject(s)
Bacterial Proteins/chemistry , Bacteroides thetaiotaomicron/enzymology , Glycoside Hydrolases/chemistry , Hexuronic Acids/chemistry , Bacterial Proteins/genetics , Bacteroides thetaiotaomicron/genetics , Catalytic Domain , Crystallography, X-Ray , Glycoside Hydrolases/genetics , Structure-Activity Relationship , Substrate Specificity
8.
Proc Natl Acad Sci U S A ; 114(27): 7037-7042, 2017 07 03.
Article in English | MEDLINE | ID: mdl-28630303

ABSTRACT

The human microbiota, which plays an important role in health and disease, uses complex carbohydrates as a major source of nutrients. Utilization hierarchy indicates that the host glycosaminoglycans heparin (Hep) and heparan sulfate (HS) are high-priority carbohydrates for Bacteroides thetaiotaomicron, a prominent member of the human microbiota. The sulfation patterns of these glycosaminoglycans are highly variable, which presents a significant enzymatic challenge to the polysaccharide lyases and sulfatases that mediate degradation. It is possible that the bacterium recruits lyases with highly plastic specificities and expresses a repertoire of enzymes that target substructures of the glycosaminoglycans with variable sulfation or that the glycans are desulfated before cleavage by the lyases. To distinguish between these mechanisms, the components of the B. thetaiotaomicron Hep/HS degrading apparatus were analyzed. The data showed that the bacterium expressed a single-surface endo-acting lyase that cleaved HS, reflecting its higher molecular weight compared with Hep. Both Hep and HS oligosaccharides imported into the periplasm were degraded by a repertoire of lyases, with each enzyme displaying specificity for substructures within these glycosaminoglycans that display a different degree of sulfation. Furthermore, the crystal structures of a key surface glycan binding protein, which is able to bind both Hep and HS, and periplasmic sulfatases reveal the major specificity determinants for these proteins. The locus described here is highly conserved within the human gut Bacteroides, indicating that the model developed is of generic relevance to this important microbial community.


Subject(s)
Bacteroides/enzymology , Gastrointestinal Microbiome , Glycosaminoglycans/chemistry , Bacteroides/genetics , Calorimetry , Carbohydrates/chemistry , Catalysis , Crystallography, X-Ray , Cytoplasm/enzymology , Dietary Carbohydrates , Heparin/chemistry , Heparitin Sulfate/chemistry , Humans , Microscopy, Fluorescence , Mutation , Oligosaccharides/chemistry , Polysaccharide-Lyases/chemistry , Polysaccharides/chemistry , Sulfatases/chemistry , Sulfur/chemistry
9.
J Biol Chem ; 293(46): 17906-17916, 2018 11 16.
Article in English | MEDLINE | ID: mdl-30262663

ABSTRACT

Glycosaminoglycans (GAGs) and GAG-degrading enzymes have wide-ranging applications in the medical and biotechnological industries. The former are also an important nutrient source for select species of the human gut microbiota (HGM), a key player in host-microbial interactions. How GAGs are metabolized by the HGM is therefore of interest and has been extensively investigated in the model human gut microbe Bacteroides thetaiotaomicron. The presence of as-yet uncharacterized GAG-inducible genes in its genome and of related species, however, is testament to our incomplete understanding of this process. Nevertheless, it presents a potential opportunity for the discovery of additional GAG-degrading enzymes. Here, we investigated a gene of unknown function (BT_3328) from the chondroitin sulfate (CS) utilization locus of B. thetaiotaomicron NMR and UV spectroscopic assays revealed that it encodes a novel polysaccharide lyase (PL), hereafter referred to as BtCDH, reflecting its source (B. thetaiotaomicron (Bt)) and its ability to degrade the GAGs CS, dermatan sulfate (DS), and hyaluronic acid (HA). When incubated with HA, BtCDH generated a series of unsaturated HA sugars, including Δ4,5UA-GlcNAc, Δ4,5UA-GlcNAc-GlcA-GlcNac, Δ4,5UA-[GlcNAc-GlcA]2-GlcNac, and Δ4,5UA-[GlcNAc-GlcA]3-GlcNac, as end products and hence was classed as endo-acting. A combination of genetic and biochemical assays revealed that BtCDH localizes to the cell surface of B. thetaiotaomicron where it enables extracellular GAG degradation. BtCDH homologs were also detected in several other HGM species, and we therefore propose that it represents the founding member of a new polysaccharide lyase family (PL29). The current discovery also contributes new insights into CS metabolism by the HGM.


Subject(s)
Bacterial Proteins/metabolism , Bacteroides thetaiotaomicron/enzymology , Chondroitin Sulfates/metabolism , Dermatan Sulfate/metabolism , Hyaluronic Acid/metabolism , Polysaccharide-Lyases/metabolism , Bacterial Proteins/chemistry , Hydrogen-Ion Concentration , Metals, Heavy/chemistry , Polysaccharide-Lyases/chemistry , Temperature
11.
Commun Chem ; 7(1): 137, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38890439

ABSTRACT

Microbial α-L-fucosidases catalyse the hydrolysis of terminal α-L-fucosidic linkages and can perform transglycosylation reactions. Based on sequence identity, α-L-fucosidases are classified in glycoside hydrolases (GHs) families of the carbohydrate-active enzyme database. Here we explored the sequence-function space of GH29 fucosidases. Based on sequence similarity network (SSN) analyses, 15 GH29 α-L-fucosidases were selected for functional characterisation. HPAEC-PAD and LC-FD-MS/MS analyses revealed substrate and linkage specificities for α1,2, α1,3, α1,4 and α1,6 linked fucosylated oligosaccharides and glycoconjugates, consistent with their SSN clustering. The structural basis for the substrate specificity of GH29 fucosidase from Bifidobacterium asteroides towards α1,6 linkages and FA2G2 N-glycan was determined by X-ray crystallography and STD NMR. The capacity of GH29 fucosidases to carry out transfucosylation reactions with GlcNAc and 3FN as acceptors was evaluated by TLC combined with ESI-MS and NMR. These experimental data supported the use of SSN to further explore the GH29 sequence-function space through machine-learning models. Our lightweight protein language models could accurately allocate test sequences in their respective SSN clusters and assign 34,258 non-redundant GH29 sequences into SSN clusters. It is expected that the combination of these computational approaches will be used in the future for the identification of novel GHs with desired specificities.

12.
mBio ; 12(4): e0136821, 2021 08 31.
Article in English | MEDLINE | ID: mdl-34340552

ABSTRACT

The human gut microbiota (HGM) contributes to the physiology and health of its host. The health benefits provided by dietary manipulation of the HGM require knowledge of how glycans, the major nutrients available to this ecosystem, are metabolized. Arabinogalactan proteins (AGPs) are a ubiquitous feature of plant polysaccharides available to the HGM. Although the galactan backbone and galactooligosaccharide side chains of AGPs are conserved, the decorations of these structures are highly variable. Here, we tested the hypothesis that these variations in arabinogalactan decoration provide a selection mechanism for specific Bacteroides species within the HGM. The data showed that only a single bacterium, B. plebeius, grew on red wine AGP (Wi-AGP) and seaweed AGP (SW-AGP) in mono- or mixed culture. Wi-AGP thus acts as a privileged nutrient for a Bacteroides species within the HGM that utilizes marine and terrestrial plant glycans. The B. plebeius polysaccharide utilization loci (PULs) upregulated by AGPs encoded a polysaccharide lyase, located in the enzyme family GH145, which hydrolyzed Rha-Glc linkages in Wi-AGP. Further analysis of GH145 identified an enzyme with two active sites that displayed glycoside hydrolase and lyase activities, respectively, which conferred substrate flexibility for different AGPs. The AGP-degrading apparatus of B. plebeius also contained a sulfatase, BpS1_8, active on SW-AGP and Wi-AGP, which played a pivotal role in the utilization of these glycans by the bacterium. BpS1_8 enabled other Bacteroides species to access the sulfated AGPs, providing a route to introducing privileged nutrient utilization into probiotic and commensal organisms that could improve human health. IMPORTANCE Dietary manipulation of the HGM requires knowledge of how glycans available to this ecosystem are metabolized. The variable structures that decorate the core component of plant AGPs may influence their utilization by specific organisms within the HGM. Here, we evaluated the ability of Bacteroides species to utilize a marine and terrestrial AGP. The data showed that a single bacterium, B. plebeius, grew on Wi-AGP and SW-AGP in mono- or mixed culture. Wi-AGP is thus a privileged nutrient for a Bacteroides species that utilizes marine and terrestrial plant glycans. A key component of the AGP-degrading apparatus of B. plebeius is a sulfatase that conferred the ability of the bacterium to utilize these glycans. The enzyme enabled other Bacteroides species to access the sulfated AGPs, providing a route to introducing privileged nutrient utilization into probiotic and commensal organisms that could improve human health.


Subject(s)
Bacteroides/metabolism , Mucoproteins/metabolism , Nutrients/metabolism , Sulfatases/metabolism , Bacteroides/enzymology , Bacteroides/genetics , Bacteroides/growth & development , Dietary Carbohydrates/metabolism , Gastrointestinal Microbiome/physiology , Plant Proteins/metabolism , Polysaccharide-Lyases/genetics , Polysaccharide-Lyases/metabolism , Polysaccharides/metabolism
13.
Nat Commun ; 11(1): 4396, 2020 08 28.
Article in English | MEDLINE | ID: mdl-32859948

ABSTRACT

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

14.
Nat Commun ; 11(1): 646, 2020 01 31.
Article in English | MEDLINE | ID: mdl-32005816

ABSTRACT

The human gut microbiota (HGM), which is critical to human health, utilises complex glycans as its major carbon source. Glycosaminoglycans represent an important, high priority, nutrient source for the HGM. Pathways for the metabolism of various glycosaminoglycan substrates remain ill-defined. Here we perform a biochemical, genetic and structural dissection of the genetic loci that orchestrates glycosaminoglycan metabolism in the organism Bacteroides thetaiotaomicron. Here, we report: the discovery of two previously unknown surface glycan binding proteins which facilitate glycosaminoglycan import into the periplasm; distinct kinetic and genetic specificities of various periplasmic lyases which dictate glycosaminoglycan metabolic pathways; understanding of endo sulfatase activity questioning the paradigm of how the 'sulfation problem' is handled by the HGM; and 3D crystal structures of the polysaccharide utilisation loci encoded sulfatases. Together with comparative genomic studies, our study fills major gaps in our knowledge of glycosaminoglycan metabolism by the HGM.


Subject(s)
Bacterial Proteins/genetics , Bacteroides thetaiotaomicron/genetics , Bacteroides thetaiotaomicron/metabolism , Glycosaminoglycans/metabolism , Bacterial Proteins/metabolism , Gastrointestinal Microbiome , Genetic Loci , Humans , Polysaccharides/metabolism , Sulfatases/genetics , Sulfatases/metabolism
15.
ISME J ; 13(7): 1883-1889, 2019 07.
Article in English | MEDLINE | ID: mdl-30936421

ABSTRACT

Microbes in the intestines of mammals degrade dietary glycans for energy and growth. The pathways required for polysaccharide utilization are functionally diverse; moreover, they are unequally dispersed between bacterial genomes. Hence, assigning metabolic phenotypes to genotypes remains a challenge in microbiome research. Here we demonstrate that glycan uptake in gut bacteria can be visualized with fluorescent glycan conjugates (FGCs) using epifluorescence microscopy. Yeast α-mannan and rhamnogalacturonan-II, two structurally distinct glycans from the cell walls of yeast and plants, respectively, were fluorescently labeled and fed to Bacteroides thetaiotaomicron VPI-5482. Wild-type cells rapidly consumed the FGCs and became fluorescent; whereas, strains that had deleted pathways for glycan degradation and transport were non-fluorescent. Uptake of FGCs, therefore, is direct evidence of genetic function and provides a direct method to assess specific glycan metabolism in intestinal bacteria at the single cell level.


Subject(s)
Bacteroides thetaiotaomicron/metabolism , Carbohydrate Metabolism , Dietary Carbohydrates/metabolism , Gastrointestinal Microbiome , Genome, Bacterial/genetics , Polysaccharides/metabolism , Bacteroides thetaiotaomicron/genetics , Cell Wall/chemistry , Fluorescence , Intestines/microbiology , Pectins/metabolism
16.
Nat Microbiol ; 4(9): 1571-1581, 2019 09.
Article in English | MEDLINE | ID: mdl-31160824

ABSTRACT

Glycans are the major carbon sources available to the human colonic microbiota. Numerous N-glycosylated proteins are found in the human gut, from both dietary and host sources, including immunoglobulins such as IgA that are secreted into the intestine at high levels. Here, we show that many mutualistic gut Bacteroides spp. have the capacity to utilize complex N-glycans (CNGs) as nutrients, including those from immunoglobulins. Detailed mechanistic studies using transcriptomic, biochemical, structural and genetic techniques reveal the pathway employed by Bacteroides thetaiotaomicron (Bt) for CNG degradation. The breakdown process involves an extensive enzymatic apparatus encoded by multiple non-adjacent loci and comprises 19 different carbohydrate-active enzymes from different families, including a CNG-specific endo-glycosidase activity. Furthermore, CNG degradation involves the activity of carbohydrate-active enzymes that have previously been implicated in the degradation of other classes of glycan. This complex and diverse apparatus provides Bt with the capacity to access the myriad different structural variants of CNGs likely to be found in the intestinal niche.


Subject(s)
Bacteroides/enzymology , Bacteroides/genetics , Gene Expression Regulation, Bacterial , Intestines/microbiology , Polysaccharides/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacteroides/growth & development , Crystallography, X-Ray , Gene Expression Profiling , Genetic Loci/genetics , Glycoproteins/chemistry , Glycoproteins/metabolism , Humans , Polysaccharides/chemistry , Symbiosis
18.
FEMS Microbiol Rev ; 42(2): 146-164, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29325042

ABSTRACT

The human gut microbiota (HGM) makes an important contribution to health and disease. It is a complex microbial community of trillions of microbes with a majority of its members represented within two phyla, the Bacteroidetes and Firmicutes, although it also contains species of Actinobacteria and Proteobacteria. Reflecting its importance, the HGM is sometimes referred to as an 'organ' as it performs functions analogous to systemic tissues within the human host. The major nutrients available to the HGM are host and dietary complex carbohydrates. To utilise these nutrient sources, the HGM has developed elaborate, variable and sophisticated systems for the sensing, capture and utilisation of these glycans. Understanding nutrient acquisition by the HGM can thus provide mechanistic insights into the dynamics of this ecosystem, and how it impacts human health. Dietary nutrient sources include a wide variety of simple and complex plant and animal-derived glycans most of which are not degraded by enzymes in the digestive tract of the host. Here we review how various adaptive mechanisms that operate across the major phyla of the HGM contribute to glycan utilisation, focusing on the most complex carbohydrates presented to this ecosystem.


Subject(s)
Gastrointestinal Microbiome/physiology , Polysaccharides/metabolism , Bacteria/enzymology , Bacteria/metabolism , Diet , Humans
19.
Nat Microbiol ; 3(11): 1314-1326, 2018 11.
Article in English | MEDLINE | ID: mdl-30349080

ABSTRACT

Glycans are major nutrients for the human gut microbiota (HGM). Arabinogalactan proteins (AGPs) comprise a heterogenous group of plant glycans in which a ß1,3-galactan backbone and ß1,6-galactan side chains are conserved. Diversity is provided by the variable nature of the sugars that decorate the galactans. The mechanisms by which nutritionally relevant AGPs are degraded in the HGM are poorly understood. Here we explore how the HGM organism Bacteroides thetaiotaomicron metabolizes AGPs. We propose a sequential degradative model in which exo-acting glycoside hydrolase (GH) family 43 ß1,3-galactanases release the side chains. These oligosaccharide side chains are depolymerized by the synergistic action of exo-acting enzymes in which catalytic interactions are dependent on whether degradation is initiated by a lyase or GH. We identified two GHs that establish two previously undiscovered GH families. The crystal structures of the exo-ß1,3-galactanases identified a key specificity determinant and departure from the canonical catalytic apparatus of GH43 enzymes. Growth studies of Bacteroidetes spp. on complex AGP revealed 3 keystone organisms that facilitated utilization of the glycan by 17 recipient bacteria, which included B. thetaiotaomicron. A surface endo-ß1,3-galactanase, when engineered into B. thetaiotaomicron, enabled the bacterium to utilize complex AGPs and act as a keystone organism.


Subject(s)
Bacterial Proteins/metabolism , Bacteroides thetaiotaomicron/enzymology , Glycoside Hydrolases/metabolism , Mucoproteins/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacteroides thetaiotaomicron/classification , Bacteroides thetaiotaomicron/growth & development , Bacteroides thetaiotaomicron/metabolism , Crystallography, X-Ray , Gastrointestinal Microbiome/physiology , Glycoside Hydrolases/chemistry , Glycoside Hydrolases/genetics , Humans , Oligosaccharides/metabolism , Plant Proteins/metabolism , Substrate Specificity
20.
Nat Microbiol ; 3(2): 210-219, 2018 02.
Article in English | MEDLINE | ID: mdl-29255254

ABSTRACT

The major nutrients available to human colonic Bacteroides species are glycans, exemplified by pectins, a network of covalently linked plant cell wall polysaccharides containing galacturonic acid (GalA). Metabolism of complex carbohydrates by the Bacteroides genus is orchestrated by polysaccharide utilization loci (PULs). In Bacteroides thetaiotaomicron, a human colonic bacterium, the PULs activated by different pectin domains have been identified; however, the mechanism by which these loci contribute to the degradation of these GalA-containing polysaccharides is poorly understood. Here we show that each PUL orchestrates the metabolism of specific pectin molecules, recruiting enzymes from two previously unknown glycoside hydrolase families. The apparatus that depolymerizes the backbone of rhamnogalacturonan-I is particularly complex. This system contains several glycoside hydrolases that trim the remnants of other pectin domains attached to rhamnogalacturonan-I, and nine enzymes that contribute to the degradation of the backbone that makes up a rhamnose-GalA repeating unit. The catalytic properties of the pectin-degrading enzymes are optimized to protect the glycan cues that activate the specific PULs ensuring a continuous supply of inducing molecules throughout growth. The contribution of Bacteroides spp. to metabolism of the pectic network is illustrated by cross-feeding between organisms.


Subject(s)
Bacteroides/metabolism , Colon/microbiology , Diet , Pectins/metabolism , Polysaccharides/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacteroides/enzymology , Bacteroides/genetics , Bacteroides/growth & development , Genes, Bacterial/genetics , Glycoside Hydrolases , Hexuronic Acids , Humans , Mutagenesis, Site-Directed , Plant Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL