Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 76
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nature ; 625(7993): 157-165, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38093016

ABSTRACT

Evidence is accumulating that perturbed postnatal development of the gut microbiome contributes to childhood malnutrition1-4. Here we analyse biospecimens from a randomized, controlled trial of a microbiome-directed complementary food (MDCF-2) that produced superior rates of weight gain compared with a calorically more dense conventional ready-to-use supplementary food in 12-18-month-old Bangladeshi children with moderate acute malnutrition4. We reconstructed 1,000 bacterial genomes (metagenome-assembled genomes (MAGs)) from the faecal microbiomes of trial participants, identified 75 MAGs of which the abundances were positively associated with ponderal growth (change in weight-for-length Z score (WLZ)), characterized changes in MAG gene expression as a function of treatment type and WLZ response, and quantified carbohydrate structures in MDCF-2 and faeces. The results reveal that two Prevotella copri MAGs that are positively associated with WLZ are the principal contributors to MDCF-2-induced expression of metabolic pathways involved in utilizing the component glycans of MDCF-2. The predicted specificities of carbohydrate-active enzymes expressed by their polysaccharide-utilization loci are correlated with (1) the in vitro growth of Bangladeshi P. copri strains, possessing varying degrees of polysaccharide-utilization loci and genomic conservation with these MAGs, in defined medium containing different purified glycans representative of those in MDCF-2, and (2) the levels of faecal carbohydrate structures in the trial participants. These associations suggest that identifying bioactive glycan structures in MDCFs metabolized by growth-associated bacterial taxa will help to guide recommendations about their use in children with acute malnutrition and enable the development of additional formulations.


Subject(s)
Food , Gastrointestinal Microbiome , Malnutrition , Polysaccharides , Humans , Infant , Bacteria/genetics , Bangladesh , Body Weight/genetics , Feces/microbiology , Gastrointestinal Microbiome/physiology , Genome, Bacterial/genetics , Malnutrition/microbiology , Metagenome/genetics , Polysaccharides/metabolism , Weight Gain
2.
Cell ; 159(2): 253-66, 2014 Oct 09.
Article in English | MEDLINE | ID: mdl-25284151

ABSTRACT

To study how microbes establish themselves in a mammalian gut environment, we colonized germ-free mice with microbial communities from human, zebrafish, and termite guts, human skin and tongue, soil, and estuarine microbial mats. Bacteria from these foreign environments colonized and persisted in the mouse gut; their capacity to metabolize dietary and host carbohydrates and bile acids correlated with colonization success. Cohousing mice harboring these xenomicrobiota or a mouse cecal microbiota, along with germ-free "bystanders," revealed the success of particular bacterial taxa in invading guts with established communities and empty gut habitats. Unanticipated patterns of ecological succession were observed; for example, a soil-derived bacterium dominated even in the presence of bacteria from other gut communities (zebrafish and termite), and human-derived bacteria colonized germ-free bystander mice before mouse-derived organisms. This approach can be generalized to address a variety of mechanistic questions about succession, including succession in the context of microbiota-directed therapeutics.


Subject(s)
Bacteria/classification , Bacteria/growth & development , Gastrointestinal Tract/microbiology , Mice/microbiology , Animals , Bacteria/metabolism , Ecosystem , Estuaries , Germ-Free Life , Humans , Isoptera/microbiology , Microbial Interactions , Skin/microbiology , Soil Microbiology , Symbiosis , Tongue/microbiology , Zebrafish/microbiology
3.
Nature ; 595(7865): 91-95, 2021 07.
Article in English | MEDLINE | ID: mdl-34163075

ABSTRACT

Changing food preferences brought about by westernization that have deleterious health effects1,2-combined with myriad forces that are contributing to increased food insecurity-are catalysing efforts to identify more nutritious and affordable foods3. Consumption of dietary fibre can help to prevent cardiovascular disease, type 2 diabetes and obesity4-6. A substantial number of reports have explored the effects of dietary fibre on the gut microbial community7-9. However, the microbiome is complex, dynamic and exhibits considerable intra- and interpersonal variation in its composition and functions. The large number of potential interactions between the components of the microbiome makes it challenging to define the mechanisms by which food ingredients affect community properties. Here we address the question of how foods containing different fibre preparations can be designed to alter functions associated with specific components of the microbiome. Because a marked increase in snack consumption is associated with westernization, we formulated snack prototypes using plant fibres from different sustainable sources that targeted distinct features of the gut microbiomes of individuals with obesity when transplanted into gnotobiotic mice. We used these snacks to supplement controlled diets that were consumed by adult individuals with obesity or who were overweight. Fibre-specific changes in their microbiomes were linked to changes in their plasma proteomes indicative of an altered physiological state.


Subject(s)
Dietary Fiber/pharmacology , Feces/microbiology , Gastrointestinal Microbiome/drug effects , Germ-Free Life , Snacks , Adolescent , Adult , Animals , Bacteroides/drug effects , Bacteroides/isolation & purification , Blood Proteins/analysis , Female , Humans , Male , Mice , Mice, Inbred C57BL , Middle Aged , Obesity/microbiology , Overweight/microbiology , Proteome/analysis , Proteome/drug effects , Young Adult
4.
Proc Natl Acad Sci U S A ; 119(20): e2123411119, 2022 05 17.
Article in English | MEDLINE | ID: mdl-35533274

ABSTRACT

Increases in snack consumption associated with Westernized lifestyles provide an opportunity to introduce nutritious foods into poor diets. We describe two 10-wk-long open label, single group assignment human studies that measured the effects of two snack prototypes containing fiber preparations from two sustainable and scalable sources; the byproducts remaining after isolation of protein from the endosperm of peas and the vesicular pulp remaining after processing oranges for the manufacture of juices. The normal diets of study participants were supplemented with either a pea- or orange fiber-containing snack. We focused our analysis on quantifying the abundances of genes encoding carbohydrate-active enzymes (CAZymes) (glycoside hydrolases and polysaccharide lyases) in the fecal microbiome, mass spectrometric measurements of glycan structures (glycosidic linkages) in feces, plus aptamer-based assessment of levels of 1,300 plasma proteins reflecting a broad range of physiological functions. Computational methods for feature selection identified treatment-discriminatory changes in CAZyme genes that correlated with alterations in levels of fiber-associated glycosidic linkages; these changes in turn correlated with levels of plasma proteins representing diverse biological functions, including transforming growth factor type ß/bone morphogenetic protein-mediated fibrosis, vascular endothelial growth factor-related angiogenesis, P38/MAPK-associated immune cell signaling, and obesity-associated hormonal regulators. The approach used represents a way to connect changes in consumer microbiomes produced by specific fiber types with host responses in the context of varying background diets.


Subject(s)
Gastrointestinal Microbiome , Microbiota , Dietary Fiber/metabolism , Gastrointestinal Microbiome/physiology , Humans , Polysaccharides/metabolism , Proteome
5.
New Phytol ; 242(4): 1676-1690, 2024 May.
Article in English | MEDLINE | ID: mdl-38148573

ABSTRACT

Soil fungi belonging to different functional guilds, such as saprotrophs, pathogens, and mycorrhizal symbionts, play key roles in forest ecosystems. To date, no study has compared the actual gene expression of these guilds in different forest soils. We used metatranscriptomics to study the competition for organic resources by these fungal groups in boreal, temperate, and Mediterranean forest soils. Using a dedicated mRNA annotation pipeline combined with the JGI MycoCosm database, we compared the transcripts of these three fungal guilds, targeting enzymes involved in C- and N mobilization from plant and microbial cell walls. Genes encoding enzymes involved in the degradation of plant cell walls were expressed at a higher level in saprotrophic fungi than in ectomycorrhizal and pathogenic fungi. However, ectomycorrhizal and saprotrophic fungi showed similarly high expression levels of genes encoding enzymes involved in fungal cell wall degradation. Transcripts for N-related transporters were more highly expressed in ectomycorrhizal fungi than in other groups. We showed that ectomycorrhizal and saprotrophic fungi compete for N in soil organic matter, suggesting that their interactions could decelerate C cycling. Metatranscriptomics provides a unique tool to test controversial ecological hypotheses and to better understand the underlying ecological processes involved in soil functioning and carbon stabilization.


Subject(s)
Forests , Fungi , Soil Microbiology , Transcriptome , Fungi/genetics , Fungi/physiology , Transcriptome/genetics , Mycorrhizae/physiology , Mycorrhizae/genetics , Gene Expression Profiling , Gene Expression Regulation, Fungal , Nitrogen/metabolism , Soil/chemistry , Ecosystem , RNA, Messenger/genetics , RNA, Messenger/metabolism
6.
Nucleic Acids Res ; 50(D1): D571-D577, 2022 01 07.
Article in English | MEDLINE | ID: mdl-34850161

ABSTRACT

Thirty years have elapsed since the emergence of the classification of carbohydrate-active enzymes in sequence-based families that became the CAZy database over 20 years ago, freely available for browsing and download at www.cazy.org. In the era of large scale sequencing and high-throughput Biology, it is important to examine the position of this specialist database that is deeply rooted in human curation. The three primary tasks of the CAZy curators are (i) to maintain and update the family classification of this class of enzymes, (ii) to classify sequences newly released by GenBank and the Protein Data Bank and (iii) to capture and present functional information for each family. The CAZy website is updated once a month. Here we briefly summarize the increase in novel families and the annotations conducted during the last 8 years. We present several important changes that facilitate taxonomic navigation, and allow to download the entirety of the annotations. Most importantly we highlight the considerable amount of work that accompanies the analysis and report of biochemical data from the literature.


Subject(s)
Carbohydrates/chemistry , Databases, Nucleic Acid , Databases, Protein , Enzymes/chemistry , Carbohydrates/classification , Enzyme Activation/genetics , Enzymes/classification , Humans
7.
Proc Natl Acad Sci U S A ; 118(21)2021 05 25.
Article in English | MEDLINE | ID: mdl-34001614

ABSTRACT

The concept that gut microbiome-expressed functions regulate ponderal growth has important implications for infant and child health, as well as animal health. Using an intergenerational pig model of diet restriction (DR) that produces reduced weight gain, we developed a feature-selection algorithm to identify representative characteristics distinguishing DR fecal microbiomes from those of full-fed (FF) pigs as both groups consumed a common sequence of diets during their growth cycle. Gnotobiotic mice were then colonized with DR and FF microbiomes and subjected to controlled feeding with a pig diet. DR microbiomes have reduced representation of genes that degrade dominant components of late growth-phase diets, exhibit reduced production of butyrate, a key host-accessible energy source, and are causally linked to reduced hepatic fatty acid metabolism (ß-oxidation) and the selection of alternative energy substrates. The approach described could aid in the development of guidelines for microbiome stewardship in diverse species, including farm animals, in order to support their healthy growth.


Subject(s)
Butyrates/metabolism , Gastrointestinal Microbiome/physiology , Lipid Metabolism/physiology , Malnutrition/metabolism , Phosphoric Monoester Hydrolases/metabolism , alpha-Glucosidases/metabolism , Algorithms , Animals , Body Weight , Diet/methods , Diet Therapy/methods , Disease Models, Animal , Feces/microbiology , Germ-Free Life , Liver/metabolism , Male , Malnutrition/physiopathology , Mice , Mice, Inbred C57BL , Starch/metabolism , Sucrose/metabolism , Swine , Taurocholic Acid/metabolism
8.
Proc Natl Acad Sci U S A ; 116(13): 6063-6068, 2019 03 26.
Article in English | MEDLINE | ID: mdl-30850540

ABSTRACT

Over the last two decades, the number of gene/protein sequences gleaned from sequencing projects of individual genomes and environmental DNA has grown exponentially. Only a tiny fraction of these predicted proteins has been experimentally characterized, and the function of most proteins remains hypothetical or only predicted based on sequence similarity. Despite the development of postgenomic methods, such as transcriptomics, proteomics, and metabolomics, the assignment of function to protein sequences remains one of the main challenges in modern biology. As in all classes of proteins, the growing number of predicted carbohydrate-active enzymes (CAZymes) has not been accompanied by a systematic and accurate attribution of function. Taking advantage of the CAZy database, which groups CAZymes into families and subfamilies based on amino acid similarities, we recombinantly produced 564 proteins selected from subfamilies without any biochemically characterized representatives, from distant relatives of characterized enzymes and from nonclassified proteins that show little similarity with known CAZymes. Screening these proteins for activity on a wide collection of carbohydrate substrates led to the discovery of 13 CAZyme families (two of which were also discovered by others during the course of our work), revealed three previously unknown substrate specificities, and assigned a function to 25 subfamilies.


Subject(s)
Carbohydrate Metabolism , Enzymes/genetics , Sequence Analysis, Protein , Amino Acid Sequence , Animals , Carbohydrate Metabolism/genetics , Enzymes/metabolism , Genomics/methods , Humans , Polysaccharides/metabolism , Sequence Analysis, DNA , Structure-Activity Relationship
9.
J Biol Chem ; 294(44): 15973-15986, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31501245

ABSTRACT

Glycoside hydrolase family (GH) 16 comprises a large and taxonomically diverse family of glycosidases and transglycosidases that adopt a common ß-jelly-roll fold and are active on a range of terrestrial and marine polysaccharides. Presently, broadly insightful sequence-function correlations in GH16 are hindered by a lack of a systematic subfamily structure. To fill this gap, we have used a highly scalable protein sequence similarity network analysis to delineate nearly 23,000 GH16 sequences into 23 robust subfamilies, which are strongly supported by hidden Markov model and maximum likelihood molecular phylogenetic analyses. Subsequent evaluation of over 40 experimental three-dimensional structures has highlighted key tertiary structural differences, predominantly manifested in active-site loops, that dictate substrate specificity across the GH16 evolutionary landscape. As for other large GH families (i.e. GH5, GH13, and GH43), this new subfamily classification provides a roadmap for functional glycogenomics that will guide future bioinformatics and experimental structure-function analyses. The GH16 subfamily classification is publicly available in the CAZy database. The sequence similarity network workflow used here, SSNpipe, is freely available from GitHub.


Subject(s)
Bacterial Proteins/chemistry , Fungal Proteins/chemistry , Glycoside Hydrolases/genetics , Phylogeny , Sequence Analysis, Protein/methods , Algorithms , Bacterial Proteins/classification , Bacterial Proteins/genetics , Catalytic Domain , Evolution, Molecular , Fungal Proteins/classification , Fungal Proteins/genetics , Glycomics/methods , Glycoside Hydrolases/chemistry , Glycoside Hydrolases/classification
10.
Nucleic Acids Res ; 46(D1): D677-D683, 2018 01 04.
Article in English | MEDLINE | ID: mdl-29088389

ABSTRACT

The Polysaccharide Utilization Loci (PUL) database was launched in 2015 to present PUL predictions in ∼70 Bacteroidetes species isolated from the human gastrointestinal tract, as well as PULs derived from the experimental data reported in the literature. In 2018 PULDB offers access to 820 genomes, sampled from various environments and covering a much wider taxonomical range. A Krona dynamic chart was set up to facilitate browsing through taxonomy. Literature surveys now allows the presentation of the most recent (i) PUL repertoires deduced from RNAseq large-scale experiments, (ii) PULs that have been subjected to in-depth biochemical analysis and (iii) new Carbohydrate-Active enzyme (CAZyme) families that contributed to the refinement of PUL predictions. To improve PUL visualization and genome browsing, the previous annotation of genes encoding CAZymes, regulators, integrases and SusCD has now been expanded to include functionally relevant protein families whose genes are significantly found in the vicinity of PULs: sulfatases, proteases, ROK repressors, epimerases and ATP-Binding Cassette and Major Facilitator Superfamily transporters. To cope with cases where susCD may be absent due to incomplete assemblies/split PULs, we present 'CAZyme cluster' predictions. Finally, a PUL alignment tool, operating on the tagged families instead of amino-acid sequences, was integrated to retrieve PULs similar to a query of interest. The updated PULDB website is accessible at www.cazy.org/PULDB_new/.


Subject(s)
Bacterial Proteins/metabolism , Bacteroidetes/metabolism , Databases, Chemical , Databases, Genetic , Genes, Bacterial , Operon/genetics , Polysaccharides/metabolism , Bacterial Proteins/genetics , Bacteroidetes/classification , Bacteroidetes/genetics , Biological Transport/genetics , Carrier Proteins/genetics , Carrier Proteins/metabolism , Chlorobi/classification , Chlorobi/genetics , Chlorobi/metabolism , Energy Metabolism/genetics , Enzymes/genetics , Enzymes/metabolism , Evolution, Molecular , Fibrobacteres/classification , Fibrobacteres/genetics , Fibrobacteres/metabolism , Gene Expression Regulation, Bacterial , Molecular Sequence Annotation , Multigene Family , RNA, Bacterial/genetics , Sequence Alignment , Species Specificity
11.
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
12.
Appl Environ Microbiol ; 85(24)2019 12 15.
Article in English | MEDLINE | ID: mdl-31604773

ABSTRACT

Pyrroloquinoline quinone (PQQ) is an ortho-quinone cofactor of several prokaryotic oxidases. Widely available in the diet and necessary for the correct growth of mice, PQQ has been suspected to be a vitamin for eukaryotes. However, no PQQ-dependent eukaryotic enzyme had been identified to use the PQQ until 2014, when a basidiomycete enzyme catalyzing saccharide dehydrogenation using PQQ as a cofactor was characterized and served to define auxiliary activity family 12 (AA12). Here we report the biochemical characterization of the AA12 enzyme encoded by the genome of the ascomycete Trichoderma reesei (TrAA12). Surprisingly, only weak activity against uncommon carbohydrates like l-fucose or d-arabinose was measured. The three-dimensional structure of TrAA12 reveals important similarities with bacterial soluble glucose dehydrogenases (sGDH). The enzymatic characterization and the structure solved in the presence of calcium confirm the importance of this ion in catalysis, as observed for sGDH. The structural characterization of TrAA12 was completed by modeling PQQ and l-fucose in the enzyme active site. Based on these results, the AA12 family of enzymes is likely to have a catalytic mechanism close to that of bacterial sGDH.IMPORTANCE Pyrroloquinoline quinone (PQQ) is an important cofactor synthesized by prokaryotes and involved in enzymatic alcohol and sugar oxidation. In eukaryotes, the benefit of PQQ as a vitamin has been suggested but never proved. Recently, the first eukaryotic enzyme using PQQ was characterized in the basidiomycete Coprinopsis cinerea, demonstrating that fungi are able to use PQQ as an enzyme cofactor. This discovery led to the classification of the fungal PQQ-dependent enzymes in auxiliary activity family 12 (AA12) of the Carbohydrate-Active Enzymes (CAZy) database (www.cazy.org) classification. In the present paper, we report on the characterization of the ascomycete AA12 enzyme from Trichoderma reesei (TrAA12). Our enzymatic and phylogenetic results show divergence with the only other member of the family characterized, that from the basidiomycete Coprinopsis cinerea The crystallographic structure of TrAA12 shows similarities to the global active-site architecture of bacterial glucose dehydrogenases, suggesting a common evolution between the two families.


Subject(s)
Glucose Dehydrogenases/metabolism , Oxidoreductases/metabolism , PQQ Cofactor/metabolism , Trichoderma/enzymology , Trichoderma/metabolism , Amino Acid Sequence , Arabinose/metabolism , Basidiomycota/enzymology , Carbohydrates , Catalysis , Fucose/metabolism , Oxidation-Reduction , Phylogeny , Protein Conformation
13.
Int Microbiol ; 22(4): 429-435, 2019 Dec.
Article in English | MEDLINE | ID: mdl-30875036

ABSTRACT

Studies of the digestive microbiota of ruminant animals most often focus on the bacterial diversity in the rumen or the feces of the animals, but little is known about the diversity and functions of their distal intestine. Here, the bacterial microbiota of the distal intestinal tract of two goats and two camels was investigated by metagenomics techniques. The bacterial taxonomic diversity and carbohydrate-active enzyme profile were estimated for samples taken from the small intestine, the large intestine, and the rectum of each animal. The bacterial diversity and abundance in the small intestine were lower than in the rectal and large intestinal samples. Analysis of the carbohydrate-active enzyme profiles at each site revealed a comparatively low abundance of enzymes targeting xylan and cellulose in all animals examined, similar to what has been reported earlier for sheep and therefore suggesting that plant cell wall digestion probably takes place elsewhere, such as in the rumen.


Subject(s)
Bacteria/enzymology , Bacterial Proteins/genetics , Camelus/microbiology , Carbohydrate Metabolism , Gastrointestinal Microbiome , Goats/microbiology , Animals , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , Bacterial Proteins/metabolism , Camelus/metabolism , Goats/metabolism , Intestines/microbiology , Metagenomics , Rumen/metabolism , Rumen/microbiology , Sheep
14.
Environ Microbiol ; 19(1): 185-197, 2017 01.
Article in English | MEDLINE | ID: mdl-27712009

ABSTRACT

The cellulosome is an extracellular multi-enzyme complex that is considered one of the most efficient plant cell wall-degrading strategies devised by nature. Its unique modular architecture, achieved by high affinity and specific interaction between protein modules (cohesins and dockerins) enables formation of various enzyme combinations. Extensive research has been dedicated to the mechanistic nature of the cellulosome complex. Nevertheless, little is known regarding its distribution and abundance among microbes in natural plant fibre-rich environments. Here, we explored these questions in bovine rumen microbial communities, specialized in efficient degradation of lignocellulosic plant material. We bioinformatically screened for cellulosomal modules in this complex environment using a previously published ultra-deep fibre-adherent rumen metagenome. Intriguingly, a large portion of the functions of the dockerin-containing proteins were related to alternative biological processes, and not necessarily to the classic fibre degradation function. Our analysis was experimentally validated by characterizing specific interactions between selected cohesins and dockerins and revealed that cellulosome is a more generalized strategy used by diverse bacteria, some of which were not previously associated with cellulosome production. Remarkably, our results provide additional proof of similarity among rumen microbial communities worldwide. This study suggests a broader and widespread role for the cellulosomal machinery in nature.


Subject(s)
Bacteria/isolation & purification , Cellulosomes/enzymology , Gastrointestinal Microbiome , Phylogeny , Rumen/microbiology , Animals , Bacteria/classification , Bacteria/genetics , Bacteria/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cattle , Cellulosomes/genetics , Metagenome , Multienzyme Complexes/genetics , Multienzyme Complexes/metabolism
15.
Proc Natl Acad Sci U S A ; 111(27): 9923-8, 2014 Jul 08.
Article in English | MEDLINE | ID: mdl-24958869

ABSTRACT

Basidiomycota (basidiomycetes) make up 32% of the described fungi and include most wood-decaying species, as well as pathogens and mutualistic symbionts. Wood-decaying basidiomycetes have typically been classified as either white rot or brown rot, based on the ability (in white rot only) to degrade lignin along with cellulose and hemicellulose. Prior genomic comparisons suggested that the two decay modes can be distinguished based on the presence or absence of ligninolytic class II peroxidases (PODs), as well as the abundance of enzymes acting directly on crystalline cellulose (reduced in brown rot). To assess the generality of the white-rot/brown-rot classification paradigm, we compared the genomes of 33 basidiomycetes, including four newly sequenced wood decayers, and performed phylogenetically informed principal-components analysis (PCA) of a broad range of gene families encoding plant biomass-degrading enzymes. The newly sequenced Botryobasidium botryosum and Jaapia argillacea genomes lack PODs but possess diverse enzymes acting on crystalline cellulose, and they group close to the model white-rot species Phanerochaete chrysosporium in the PCA. Furthermore, laboratory assays showed that both B. botryosum and J. argillacea can degrade all polymeric components of woody plant cell walls, a characteristic of white rot. We also found expansions in reducing polyketide synthase genes specific to the brown-rot fungi. Our results suggest a continuum rather than a dichotomy between the white-rot and brown-rot modes of wood decay. A more nuanced categorization of rot types is needed, based on an improved understanding of the genomics and biochemistry of wood decay.


Subject(s)
Basidiomycota/genetics , Basidiomycota/metabolism , Genome, Fungal , Wood , Basidiomycota/classification , Lignin/metabolism , Molecular Sequence Data , Phylogeny
16.
BMC Genomics ; 17: 671, 2016 08 23.
Article in English | MEDLINE | ID: mdl-27552843

ABSTRACT

BACKGROUND: Microorganisms constitute a reservoir of enzymes involved in environmental carbon cycling and degradation of plant polysaccharides through their production of a vast variety of Glycoside Hydrolases (GH). The CAZyChip was developed to allow a rapid characterization at transcriptomic level of these GHs and to identify enzymes acting on hydrolysis of polysaccharides or glycans. RESULTS: This DNA biochip contains the signature of 55,220 bacterial GHs available in the CAZy database. Probes were designed using two softwares, and microarrays were directly synthesized using the in situ ink-jet technology. CAZyChip specificity and reproducibility was validated by hybridization of known GHs RNA extracted from recombinant E. coli strains, which were previously identified by a functional metagenomic approach. The GHs arsenal was also studied in bioprocess conditions using rumen derived microbiota. CONCLUSIONS: The CAZyChip appears to be a user friendly tool for profiling the expression of a large variety of GHs. It can be used to study temporal variations of functional diversity, thereby facilitating the identification of new efficient candidates for enzymatic conversions from various ecosystems.


Subject(s)
Glycoside Hydrolases/genetics , Metagenome , Oligonucleotide Array Sequence Analysis/methods , Sequence Analysis, RNA/methods , Bacterial Proteins/genetics , Cell Wall/metabolism , Databases, Genetic , Escherichia coli/genetics , Gene Expression Regulation, Bacterial , Plants/metabolism , Polysaccharides/metabolism
17.
Bioinformatics ; 31(5): 647-55, 2015 Mar 01.
Article in English | MEDLINE | ID: mdl-25355788

ABSTRACT

MOTIVATION: A bacterial polysaccharide utilization locus (PUL) is a set of physically linked genes that orchestrate the breakdown of a specific glycan. PULs are prevalent in the Bacteroidetes phylum and are key to the digestion of complex carbohydrates, notably by the human gut microbiota. A given Bacteroidetes genome can encode dozens of different PULs whose boundaries and precise gene content are difficult to predict. RESULTS: Here, we present a fully automated approach for PUL prediction using genomic context and domain annotation alone. By combining the detection of a pair of marker genes with operon prediction using intergenic distances, and queries to the carbohydrate-active enzymes database (www.cazy.org), our predictor achieved above 86% accuracy in two Bacteroides species with extensive experimental PUL characterization. AVAILABILITY AND IMPLEMENTATION: PUL predictions in 67 Bacteroidetes genomes from the human gut microbiota and two additional species, from the canine oral sphere and from the environment, are presented in our database accessible at www.cazy.org/PULDB/index.php.


Subject(s)
Automation/methods , Bacteroidetes/genetics , Bacteroidetes/metabolism , Gastrointestinal Tract/microbiology , Genetic Loci , Genome, Bacterial/genetics , Microbiota/physiology , Polysaccharides/metabolism , Animals , Bacteroidetes/growth & development , Dogs , Humans , Symbiosis
18.
Appl Environ Microbiol ; 82(6): 1686-1692, 2016 Jan 04.
Article in English | MEDLINE | ID: mdl-26729713

ABSTRACT

The rapid rise in DNA sequencing has led to an expansion in the number of glycoside hydrolase (GH) families. The GH43 family currently contains α-l-arabinofuranosidase, ß-d-xylosidase, α-l-arabinanase, and ß-d-galactosidase enzymes for the debranching and degradation of hemicellulose and pectin polymers. Many studies have revealed finer details about members of GH43 that necessitate the division of GH43 into subfamilies, as was done previously for the GH5 and GH13 families. The work presented here is a robust subfamily classification that assigns over 91% of all complete GH43 domains into 37 subfamilies that correlate with conserved sequence residues and results of biochemical assays and structural studies. Furthermore, cooccurrence analysis of these subfamilies and other functional modules revealed strong associations between some GH43 subfamilies and CBM6 and CBM13 domains. Cooccurrence analysis also revealed the presence of proteins containing up to three GH43 domains and belonging to different subfamilies, suggesting significant functional differences for each subfamily. Overall, the subfamily analysis suggests that the GH43 enzymes probably display a hitherto underestimated variety of subtle specificity features that are not apparent when the enzymes are assayed with simple synthetic substrates, such as pNP-glycosides.


Subject(s)
Glycoside Hydrolases/classification , Glycoside Hydrolases/chemistry , Glycoside Hydrolases/genetics , Glycoside Hydrolases/metabolism , Hydrolysis , Pectins/metabolism , Polysaccharides/metabolism , Protein Domains
19.
Nucleic Acids Res ; 42(Database issue): D490-5, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24270786

ABSTRACT

The Carbohydrate-Active Enzymes database (CAZy; http://www.cazy.org) provides online and continuously updated access to a sequence-based family classification linking the sequence to the specificity and 3D structure of the enzymes that assemble, modify and breakdown oligo- and polysaccharides. Functional and 3D structural information is added and curated on a regular basis based on the available literature. In addition to the use of the database by enzymologists seeking curated information on CAZymes, the dissemination of a stable nomenclature for these enzymes is probably a major contribution of CAZy. The past few years have seen the expansion of the CAZy classification scheme to new families, the development of subfamilies in several families and the power of CAZy for the analysis of genomes and metagenomes. This article outlines the changes that have occurred in CAZy during the past 5 years and presents our novel effort to display the resolution and the carbohydrate ligands in crystallographic complexes of CAZymes.


Subject(s)
Carbohydrate Metabolism , Databases, Protein , Enzymes/chemistry , Carbohydrates/chemistry , Enzymes/classification , Enzymes/genetics , Genome , Internet , Ligands , Protein Conformation , Sequence Analysis, Protein , Substrate Specificity
20.
Glycobiology ; 25(3): 319-30, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25344445

ABSTRACT

Members of the GH73 glycosidase family cleave the ß-1,4-glycosidic bond between the N-acetylglucosaminyl (GlcNAc) and N-acetylmuramyl (MurNAc) moieties in bacterial peptidoglycan. A catalytic mechanism has been proposed for members FlgJ, Auto, AcmA and Atl(WM) and the structural analysis of FlgJ and Auto revealed a conserved α/ß fold reminiscent of the distantly related GH23 lysozyme. Comparison of the active site residues reveals variability in the nature of the catalytic general base suggesting two distinct catalytic mechanisms: an inverting mechanism involving two distant glutamate residues and a substrate-assisted mechanism involving anchimeric assistance by the C2-acetamido group of the GlcNAc moiety. Herein, we present the biochemical characterization and crystal structure of TM0633 from the hyperthermophilic bacterium Thermotoga maritima. TM0633 adopts the α/ß fold of the family and displays ß-N-acetylglucosaminidase activity on intact peptidoglycan sacculi. Site-directed mutagenesis identifies Glu34, Glu65 and Tyr118 as important residues for catalysis. A thorough bioinformatic analysis of the GH73 sequences identified five phylogenetic clusters. TM0633, FlgJ and Auto belong to a group of three clusters that conserve two carboxylate residues involved in a classical inverting acid-base mechanism. Members of the other two clusters lack a conserved catalytic general base supporting a substrate-assisted mechanism. Molecular modeling of representative members from each cluster suggests that variability in length of the ß-hairpin region above the active site confers ligand-binding specificity and modulates the catalytic mechanisms within the GH73 family.


Subject(s)
Acetylglucosaminidase/chemistry , Bacterial Proteins/chemistry , Thermotoga maritima/enzymology , Acetylglucosaminidase/genetics , Acetylglucosaminidase/metabolism , Amino Acid Sequence , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Catalytic Domain , Molecular Sequence Data , Phylogeny , Thermotoga maritima/genetics
SELECTION OF CITATIONS
SEARCH DETAIL