Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Biol Chem ; 296: 100415, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33587952

RESUMO

Complex glycans that evade our digestive system are major nutrients that feed the human gut microbiota (HGM). The prevalence of Bacteroidetes in the HGM of populations worldwide is engendered by the evolution of polysaccharide utilization loci (PULs), which encode concerted protein systems to utilize the myriad complex glycans in our diets. Despite their crucial roles in glycan recognition and transport, cell-surface glycan-binding proteins (SGBPs) remained understudied cogs in the PUL machinery. Here, we report the structural and biochemical characterization of a suite of SGBP-A and SGBP-B structures from three syntenic ß(1,3)-glucan utilization loci (1,3GULs) from Bacteroides thetaiotaomicron (Bt), Bacteroides uniformis (Bu), and B. fluxus (Bf), which have varying specificities for distinct ß-glucans. Ligand complexes provide definitive insight into ß(1,3)-glucan selectivity in the HGM, including structural features enabling dual ß(1,3)-glucan/mixed-linkage ß(1,3)/ß(1,4)-glucan-binding capability in some orthologs. The tertiary structural conservation of SusD-like SGBPs-A is juxtaposed with the diverse architectures and binding modes of the SGBPs-B. Specifically, the structures of the trimodular BtSGBP-B and BuSGBP-B revealed a tandem repeat of carbohydrate-binding module-like domains connected by long linkers. In contrast, BfSGBP-B comprises a bimodular architecture with a distinct ß-barrel domain at the C terminus that bears a shallow binding canyon. The molecular insights obtained here contribute to our fundamental understanding of HGM function, which in turn may inform tailored microbial intervention therapies.


Assuntos
Microbioma Gastrointestinal/fisiologia , beta-Glucanas/metabolismo , Proteínas de Bactérias/metabolismo , Bacteroides/metabolismo , Bacteroides thetaiotaomicron/metabolismo , Microbioma Gastrointestinal/genética , Trato Gastrointestinal/metabolismo , Glucanos/metabolismo , Glicosídeo Hidrolases/metabolismo , Humanos , Proteínas de Membrana/metabolismo , Polissacarídeos/metabolismo , Especificidade da Espécie
2.
Biochem J ; 478(16): 3063-3078, 2021 08 27.
Artigo em Inglês | MEDLINE | ID: mdl-34338284

RESUMO

Plant cell walls are highly dynamic structures that are composed predominately of polysaccharides. As such, endogenous carbohydrate active enzymes (CAZymes) are central to the synthesis and subsequent modification of plant cells during morphogenesis. The endo-glucanase 16 (EG16) members constitute a distinct group of plant CAZymes, angiosperm orthologs of which were recently shown to have dual ß-glucan/xyloglucan hydrolase activity. Molecular phylogeny indicates that EG16 members comprise a sister clade with a deep evolutionary relationship to the widely studied apoplastic xyloglucan endo-transglycosylases/hydrolases (XTH). A cross-genome survey indicated that EG16 members occur as a single ortholog across species and are widespread in early diverging plants, including the non-vascular bryophytes, for which functional data were previously lacking. Remarkably, enzymological characterization of an EG16 ortholog from the model moss Physcomitrella patens (PpEG16) revealed that EG16 activity and sequence/structure are highly conserved across 500 million years of plant evolution, vis-à-vis orthologs from grapevine and poplar. Ex vivo biomechanical assays demonstrated that the application of EG16 gene products caused abrupt breakage of etiolated hypocotyls rather than slow extension, thereby indicating a mode-of-action distinct from endogenous expansins and microbial endo-glucanases. The biochemical data presented here will inform future genomic, genetic, and physiological studies of EG16 enzymes.


Assuntos
Bryopsida/genética , Celulase/genética , Proteínas de Plantas/genética , Plantas/genética , Sequência de Aminoácidos , Biocatálise , Bryopsida/enzimologia , Celulase/química , Celulase/metabolismo , Evolução Molecular , Glucanos/metabolismo , Cinética , Modelos Moleculares , Filogenia , Proteínas de Plantas/química , Proteínas de Plantas/metabolismo , Plantas/classificação , Plantas/enzimologia , Conformação Proteica , Homologia de Sequência de Aminoácidos , Especificidade por Substrato , Xilanos/metabolismo , beta-Glucanas/metabolismo
3.
Cell Mol Life Sci ; 76(21): 4319-4340, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31062073

RESUMO

The human gut microbiota, which underpins nutrition and systemic health, is compositionally sensitive to the availability of complex carbohydrates in the diet. The Bacteroidetes comprise a dominant phylum in the human gut microbiota whose members thrive on dietary and endogenous glycans by employing a diversity of highly specific, multi-gene polysaccharide utilization loci (PUL), which encode a variety of carbohydrases, transporters, and sensor/regulators. PULs invariably also encode surface glycan-binding proteins (SGBPs) that play a central role in saccharide capture at the outer membrane. Here, we present combined biophysical, structural, and in vivo characterization of the two SGBPs encoded by the Bacteroides ovatus mixed-linkage ß-glucan utilization locus (MLGUL), thereby elucidating their key roles in the metabolism of this ubiquitous dietary cereal polysaccharide. In particular, molecular insight gained through several crystallographic complexes of SGBP-A and SGBP-B with oligosaccharides reveals that unique shape complementarity of binding platforms underpins specificity for the kinked MLG backbone vis-à-vis linear ß-glucans. Reverse-genetic analysis revealed that both the presence and binding ability of the SusD homolog BoSGBPMLG-A are essential for growth on MLG, whereas the divergent, multi-domain BoSGBPMLG-B is dispensable but may assist in oligosaccharide scavenging from the environment. The synthesis of these data illuminates the critical role SGBPs play in concert with other MLGUL components, reveals new structure-function relationships among SGBPs, and provides fundamental knowledge to inform future (meta)genomic, biochemical, and microbiological analyses of the human gut microbiota.


Assuntos
Bacteroides/fisiologia , Grão Comestível/metabolismo , Trato Gastrointestinal/metabolismo , Trato Gastrointestinal/microbiologia , Proteínas de Membrana/fisiologia , Polissacarídeos/metabolismo , beta-Glucanas/metabolismo , Bacteroides/genética , Bacteroides/metabolismo , Metabolismo dos Carboidratos/fisiologia , Sequência de Carboidratos , Fibras na Dieta/metabolismo , Microbioma Gastrointestinal/fisiologia , Regulação Bacteriana da Expressão Gênica , Loci Gênicos , Glicosídeo Hidrolases/genética , Glicosídeo Hidrolases/metabolismo , Humanos , Proteínas de Membrana/metabolismo
4.
J Bacteriol ; 199(15)2017 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-28138099

RESUMO

The complex carbohydrates of terrestrial and marine biomass represent a rich nutrient source for free-living and mutualistic microbes alike. The enzymatic saccharification of these diverse substrates is of critical importance for fueling a variety of complex microbial communities, including marine, soil, ruminant, and monogastric microbiota. Consequently, highly specific carbohydrate-active enzymes, recognition proteins, and transporters are enriched in the genomes of certain species and are of critical importance in competitive environments. In Bacteroidetes bacteria, these systems are organized as polysaccharide utilization loci (PULs), which are strictly regulated, colocalized gene clusters that encode enzyme and protein ensembles required for the saccharification of complex carbohydrates. This review provides historical perspectives and summarizes key findings in the study of these systems, highlighting a critical shift from sequence-based PUL discovery to systems-based analyses combining reverse genetics, biochemistry, enzymology, and structural biology to precisely illuminate the molecular mechanisms underpinning PUL function. The ecological implications of dynamic PUL deployment by key species in the human gastrointestinal tract are explored, as well as the wider distribution of these systems in other gut, terrestrial, and marine environments.


Assuntos
Bacteroidetes/genética , Bacteroidetes/metabolismo , Metabolismo Energético , Regulação Bacteriana da Expressão Gênica , Família Multigênica , Polissacarídeos/metabolismo , Hidrólise
5.
Curr Opin Struct Biol ; 68: 26-40, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33285501

RESUMO

The complex glycans comprising 'dietary fiber' evade the limited repertoire of human digestive enzymes and hence feed the vast community of microbes in the lower gastrointestinal tract. As such, complex glycans drive the composition of the human gut microbiota and, in turn, influence diverse facets of our nutrition and health. To access these otherwise recalcitrant carbohydrates, gut bacteria produce coordinated, substrate-specific arsenals of carbohydrate-active enzymes, glycan-binding proteins, oligosaccharide transporters, and transcriptional regulators. A recent explosion of biochemical and enzymological studies of these systems has led to the discovery of manifold new carbohydrate-active enzyme (CAZyme) families. Crucially underpinned by structural biology, these studies have also provided unprecedented molecular insight into the exquisite specificity of glycan recognition in the diverse CAZymes and non-catalytic proteins from the HGM. The revelation of a multitude of new three-dimensional structures and substrate complexes constitutes a 'gold rush' in the structural biology of the human gut microbiota.


Assuntos
Microbioma Gastrointestinal , Polissacarídeos/metabolismo , Bactérias , Proteínas de Transporte , Humanos
6.
ACS Chem Biol ; 16(11): 2087-2102, 2021 11 19.
Artigo em Inglês | MEDLINE | ID: mdl-34709792

RESUMO

The ß-glucans are a disparate group of structurally diverse polysaccharides, whose members are widespread in human diets as components of the cell walls of plants, algae, and fungi (including yeasts), and as bacterial exopolysaccharides. Individual ß-glucans from these sources have long been associated with positive effects on human health through metabolic and immunological effects. Remarkably, the ß-configured glucosidic linkages that define these polysaccharides render them inaccessible to the limited repertoire of digestive enzymes encoded by the human genome. As a result, the various ß-glucans become fodder for the human gut microbiota (HGM) in the lower gastrointestinal tract, where they influence community composition and metabolic output, including fermentation to short chain fatty acids (SCFAs). Only recently, however, have the specific molecular systems that enable the utilization of ß-glucans by select members of the HGM been fully elucidated by combined genetic, biochemical, and structural biological approaches. In the context of ß-glucan structures and their effects on human nutrition and health, we summarize here the functional characterization of individual polysaccharide utilization loci (PULs) responsible for the saccharification of mixed-linkage ß(1→3)/ß(1→4)-glucans, ß(1→6)-glucans, ß(1→3)-glucans, ß(1→2)-glucans, and xyloglucans in symbiotic human gut bacteria. These exemplar PULs serve as well-defined biomarkers for the prediction of ß-glucan metabolic capability in individual bacterial taxa and across the global human population.


Assuntos
Microbioma Gastrointestinal/fisiologia , beta-Glucanas/metabolismo , Bactérias/enzimologia , Bactérias/genética , Bactérias/metabolismo , Fermentação , Glicosídeo Hidrolases/química , Glicosídeo Hidrolases/genética , Glicosídeo Hidrolases/metabolismo , Humanos , beta-Glucanas/química
7.
ACS Chem Biol ; 16(10): 1968-1984, 2021 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-33988963

RESUMO

Small molecule irreversible inhibitors are valuable tools for determining catalytically important active-site residues and revealing key details of the specificity, structure, and function of glycoside hydrolases (GHs). ß-glucans that contain backbone ß(1,3) linkages are widespread in nature, e.g., mixed-linkage ß(1,3)/ß(1,4)-glucans in the cell walls of higher plants and ß(1,3)glucans in yeasts and algae. Commensurate with this ubiquity, a large diversity of mixed-linkage endoglucanases (MLGases, EC 3.2.1.73) and endo-ß(1,3)-glucanases (laminarinases, EC 3.2.1.39 and EC 3.2.1.6) have evolved to specifically hydrolyze these polysaccharides, respectively, in environmental niches including the human gut. To facilitate biochemical and structural analysis of these GHs, with a focus on MLGases, we present here the facile chemo-enzymatic synthesis of a library of active-site-directed enzyme inhibitors based on mixed-linkage oligosaccharide scaffolds and N-bromoacetylglycosylamine or 2-fluoro-2-deoxyglycoside warheads. The effectiveness and irreversibility of these inhibitors were tested with exemplar MLGases and an endo-ß(1,3)-glucanase. Notably, determination of inhibitor-bound crystal structures of a human-gut microbial MLGase from Glycoside Hydrolase Family 16 revealed the orthogonal labeling of the nucleophile and catalytic acid/base residues with homologous 2-fluoro-2-deoxyglycoside and N-bromoacetylglycosylamine inhibitors, respectively. We anticipate that the selectivity of these inhibitors will continue to enable the structural and mechanistic analyses of ß-glucanases from diverse sources and protein families.


Assuntos
Celulases/antagonistas & inibidores , Inibidores Enzimáticos/química , Oligossacarídeos/química , Proteínas de Bactérias/antagonistas & inibidores , Proteínas de Bactérias/química , Bacteroides/enzimologia , Domínio Catalítico/efeitos dos fármacos , Celulases/química , Cristalografia por Raios X , Ensaios Enzimáticos , Inibidores Enzimáticos/síntese química , Cinética , Oligossacarídeos/síntese química , Proteínas de Plantas/antagonistas & inibidores , Proteínas de Plantas/química , Vitis/enzimologia
8.
mBio ; 11(2)2020 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-32265336

RESUMO

The human gut microbiota (HGM) has far-reaching impacts on human health and nutrition, which are fueled primarily by the metabolism of otherwise indigestible complex carbohydrates commonly known as dietary fiber. However, the molecular basis of the ability of individual taxa of the HGM to address specific dietary glycan structures remains largely unclear. In particular, the utilization of ß(1,3)-glucans, which are widespread in the human diet as yeast, seaweed, and plant cell walls, had not previously been resolved. Through a systems-based approach, here we show that the symbiont Bacteroides uniformis deploys a single, exemplar polysaccharide utilization locus (PUL) to access yeast ß(1,3)-glucan, brown seaweed ß(1,3)-glucan (laminarin), and cereal mixed-linkage ß(1,3)/ß(1,4)-glucan. Combined biochemical, enzymatic, and structural analysis of PUL-encoded glycoside hydrolases (GHs) and surface glycan-binding proteins (SGBPs) illuminates a concerted molecular system by which B. uniformis recognizes and saccharifies these distinct ß-glucans. Strikingly, the functional characterization of homologous ß(1,3)-glucan utilization loci (1,3GUL) in other Bacteroides further demonstrated that the ability of individual taxa to utilize ß(1,3)-glucan variants and/or ß(1,3)/ß(1,4)-glucans arises combinatorially from the individual specificities of SGBPs and GHs at the cell surface, which feed corresponding signals to periplasmic hybrid two-component sensors (HTCSs) via TonB-dependent transporters (TBDTs). These data reveal the importance of cooperativity in the adaptive evolution of GH and SGBP cohorts to address individual polysaccharide structures. We anticipate that this fine-grained knowledge of PUL function will inform metabolic network analysis and proactive manipulation of the HGM. Indeed, a survey of 2,441 public human metagenomes revealed the international, yet individual-specific, distribution of each 1,3GUL.IMPORTANCEBacteroidetes are a dominant phylum of the human gut microbiota (HGM) that target otherwise indigestible dietary fiber with an arsenal of polysaccharide utilization loci (PULs), each of which is dedicated to the utilization of a specific complex carbohydrate. Here, we provide novel insight into this paradigm through functional characterization of homologous PULs from three autochthonous Bacteroides species, which target the family of dietary ß(1,3)-glucans. Through detailed biochemical and protein structural analysis, we observed an unexpected diversity in the substrate specificity of PUL glycosidases and glycan-binding proteins with regard to ß(1,3)-glucan linkage and branching patterns. In combination, these individual enzyme and protein specificities support taxon-specific growth on individual ß(1,3)-glucans. This detailed metabolic insight, together with a comprehensive survey of individual 1,3GULs across human populations, further expands the fundamental roadmap of the HGM, with potential application to the future development of microbial intervention therapies.


Assuntos
Bacteroides/enzimologia , Proteínas de Transporte/metabolismo , Fibras na Dieta , Trato Gastrointestinal/microbiologia , Glicosídeo Hidrolases/metabolismo , beta-Glucanas/metabolismo , Proteínas de Bactérias/metabolismo , Membrana Celular/enzimologia , Estudos de Coortes , Cristalografia por Raios X , Microbioma Gastrointestinal , Humanos , Especificidade por Substrato
9.
Cell Rep ; 21(2): 417-430, 2017 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-29020628

RESUMO

Microbial utilization of complex polysaccharides is a major driving force in shaping the composition of the human gut microbiota. There is a growing appreciation that finely tuned polysaccharide utilization loci enable ubiquitous gut Bacteroidetes to thrive on the plethora of complex polysaccharides that constitute "dietary fiber." Mixed-linkage ß(1,3)/ß(1,4)-glucans (MLGs) are a key family of plant cell wall polysaccharides with recognized health benefits but whose mechanism of utilization has remained unclear. Here, we provide molecular insight into the function of an archetypal MLG utilization locus (MLGUL) through a combination of biochemistry, enzymology, structural biology, and microbiology. Comparative genomics coupled with growth studies demonstrated further that syntenic MLGULs serve as genetic markers for MLG catabolism across commensal gut bacteria. In turn, we surveyed human gut metagenomes to reveal that MLGULs are ubiquitous in human populations globally, which underscores the importance of gut microbial metabolism of MLG as a common cereal polysaccharide.


Assuntos
Bacteroides/metabolismo , Microbioma Gastrointestinal , Genes Bacterianos , beta-Glucanas/metabolismo , Bacteroides/genética , Grão Comestível/química , Humanos , Metabolismo , Metagenoma
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA