Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Nature ; 598(7880): 332-337, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34616040

RESUMEN

Humans have co-evolved with a dense community of microbial symbionts that inhabit the lower intestine. In the colon, secreted mucus creates a barrier that separates these microorganisms from the intestinal epithelium1. Some gut bacteria are able to utilize mucin glycoproteins, the main mucus component, as a nutrient source. However, it remains unclear which bacterial enzymes initiate degradation of the complex O-glycans found in mucins. In the distal colon, these glycans are heavily sulfated, but specific sulfatases that are active on colonic mucins have not been identified. Here we show that sulfatases are essential to the utilization of distal colonic mucin O-glycans by the human gut symbiont Bacteroides thetaiotaomicron. We characterized the activity of 12 different sulfatases produced by this species, showing that they are collectively active on all known sulfate linkages in O-glycans. Crystal structures of three enzymes provide mechanistic insight into the molecular basis of substrate specificity. Unexpectedly, we found that a single sulfatase is essential for utilization of sulfated O-glycans in vitro and also has a major role in vivo. Our results provide insight into the mechanisms of mucin degradation by a prominent group of gut bacteria, an important process for both normal microbial gut colonization2 and diseases such as inflammatory bowel disease3.


Asunto(s)
Bacteroides/enzimología , Colon/metabolismo , Colon/microbiología , Microbioma Gastrointestinal , Mucinas/metabolismo , Sulfatasas/metabolismo , Acetilgalactosamina/química , Acetilgalactosamina/metabolismo , Animales , Colon/química , Cristalografía por Rayos X , Femenino , Galactosa/metabolismo , Humanos , Masculino , Ratones , Modelos Moleculares , Especificidad por Sustrato , Sulfatasas/química
2.
Nat Chem Biol ; 18(8): 841-849, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35710619

RESUMEN

Sulfated glycans are ubiquitous nutrient sources for microbial communities that have coevolved with eukaryotic hosts. Bacteria metabolize sulfated glycans by deploying carbohydrate sulfatases that remove sulfate esters. Despite the biological importance of sulfatases, the mechanisms underlying their ability to recognize their glycan substrate remain poorly understood. Here, we use structural biology to determine how sulfatases from the human gut microbiota recognize sulfated glycans. We reveal seven new carbohydrate sulfatase structures spanning four S1 sulfatase subfamilies. Structures of S1_16 and S1_46 represent novel structures of these subfamilies. Structures of S1_11 and S1_15 demonstrate how non-conserved regions of the protein drive specificity toward related but distinct glycan targets. Collectively, these data reveal that carbohydrate sulfatases are highly selective for the glycan component of their substrate. These data provide new approaches for probing sulfated glycan metabolism while revealing the roles carbohydrate sulfatases play in host glycan catabolism.


Asunto(s)
Microbioma Gastrointestinal , Sulfatasas , Bacterias/metabolismo , Humanos , Polisacáridos/química , Sulfatasas/química , Sulfatos/química
4.
Nature ; 544(7648): 65-70, 2017 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-28329766

RESUMEN

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.


Asunto(s)
Bacteroides thetaiotaomicron/enzimología , Bacteroides thetaiotaomicron/metabolismo , Biocatálisis , Tracto Gastrointestinal/microbiología , Glicósido Hidrolasas/metabolismo , Pectinas/química , Pectinas/metabolismo , Bacteroides thetaiotaomicron/crecimiento & desarrollo , Boratos/química , Boratos/metabolismo , Dominio Catalítico , Microbioma Gastrointestinal , Glicósido Hidrolasas/química , Glicósido Hidrolasas/clasificación , Humanos , Modelos Moleculares , Especificidad por Sustrato
5.
Biochem J ; 478(4): 735-748, 2021 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-33480417

RESUMEN

Sulfated carbohydrate metabolism is a fundamental process, which occurs in all domains of life. Carbohydrate sulfatases are enzymes that remove sulfate groups from carbohydrates and are essential to the depolymerisation of complex polysaccharides. Despite their biological importance, carbohydrate sulfatases are poorly studied and challenges remain in accurately assessing the enzymatic activity, specificity and kinetic parameters. Most notably, the separation of desulfated products from sulfated substrates is currently a time-consuming process. In this paper, we describe the development of rapid capillary electrophoresis coupled to substrate fluorescence detection as a high-throughput and facile means of analysing carbohydrate sulfatase activity. The approach has utility for the determination of both kinetic and inhibition parameters and is based on existing microfluidic technology coupled to a new synthetic fluorescent 6S-GlcNAc carbohydrate substrate. Furthermore, we compare this technique, in terms of both time and resources, to high-performance anion exchange chromatography and NMR-based methods, which are the two current 'gold standards' for enzymatic carbohydrate sulfation analysis. Our study clearly demonstrates the advantages of mobility shift assays for the quantification of near real-time carbohydrate desulfation by purified sulfatases, and will support the search for small molecule inhibitors of these disease-associated enzymes.


Asunto(s)
Electroforesis Capilar/métodos , Ensayo de Cambio de Movilidad Electroforética/métodos , Fluorometría/métodos , Ensayos Analíticos de Alto Rendimiento/métodos , Técnicas Analíticas Microfluídicas/métodos , Sulfotransferasas/análisis , Proteínas Bacterianas/análisis , Proteínas Bacterianas/antagonistas & inhibidores , Bacteroides thetaiotaomicron/enzimología , Compuestos de Boro/análisis , Conformación de Carbohidratos , Cromatografía Líquida de Alta Presión , Cromatografía por Intercambio Iónico , Sistemas de Computación , Colorantes Fluorescentes/análisis , Glicosaminoglicanos/metabolismo , Cinética , Resonancia Magnética Nuclear Biomolecular , Proteínas Recombinantes/análisis , Especificidad por Sustrato , Sulfotransferasas/antagonistas & inhibidores
6.
Nature ; 517(7533): 165-169, 2015 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-25567280

RESUMEN

Yeasts, which have been a component of the human diet for at least 7,000 years, possess an elaborate cell wall α-mannan. The influence of yeast mannan on the ecology of the human microbiota is unknown. Here we show that yeast α-mannan is a viable food source for the Gram-negative bacterium Bacteroides thetaiotaomicron, a dominant member of the microbiota. Detailed biochemical analysis and targeted gene disruption studies support a model whereby limited cleavage of α-mannan on the surface generates large oligosaccharides that are subsequently depolymerized to mannose by the action of periplasmic enzymes. Co-culturing studies showed that metabolism of yeast mannan by B. thetaiotaomicron presents a 'selfish' model for the catabolism of this difficult to breakdown polysaccharide. Genomic comparison with B. thetaiotaomicron in conjunction with cell culture studies show that a cohort of highly successful members of the microbiota has evolved to consume sterically-restricted yeast glycans, an adaptation that may reflect the incorporation of eukaryotic microorganisms into the human diet.


Asunto(s)
Bacteroidetes/metabolismo , Tracto Gastrointestinal/microbiología , Mananos/metabolismo , Modelos Biológicos , Levaduras/química , Animales , Bacteroidetes/citología , Bacteroidetes/enzimología , Bacteroidetes/genética , Evolución Biológica , Conformación de Carbohidratos , Dieta , Enzimas/genética , Enzimas/metabolismo , Femenino , Sitios Genéticos/genética , Vida Libre de Gérmenes , Glicoproteínas/química , Glicoproteínas/metabolismo , Humanos , Masculino , Mananos/química , Manosa/metabolismo , Ratones , Modelos Moleculares , Oligosacáridos/química , Oligosacáridos/metabolismo , Periplasma/enzimología
7.
J Biol Chem ; 294(19): 7711-7721, 2019 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-30877196

RESUMEN

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.


Asunto(s)
Proteínas Bacterianas/química , Bacteroides thetaiotaomicron/enzimología , Glicósido Hidrolasas/química , Ácidos Hexurónicos/química , Proteínas Bacterianas/genética , Bacteroides thetaiotaomicron/genética , Dominio Catalítico , Cristalografía por Rayos X , Glicósido Hidrolasas/genética , Relación Estructura-Actividad , Especificidad por Sustrato
8.
Proc Natl Acad Sci U S A ; 114(19): 4936-4941, 2017 05 09.
Artículo en Inglés | MEDLINE | ID: mdl-28396425

RESUMEN

The human gut microbiota use complex carbohydrates as major nutrients. The requirement for an efficient glycan degrading systems exerts a major selection pressure on this microbial community. Thus, we propose that these bacteria represent a substantial resource for discovering novel carbohydrate active enzymes. To test this hypothesis, we focused on enzymes that hydrolyze rhamnosidic bonds, as cleavage of these linkages is chemically challenging and there is a paucity of information on l-rhamnosidases. Here we screened the activity of enzymes derived from the human gut microbiota bacterium Bacteroides thetaiotaomicron, which are up-regulated in response to rhamnose-containing glycans. We identified an α-l-rhamnosidase, BT3686, which is the founding member of a glycoside hydrolase (GH) family, GH145. In contrast to other rhamnosidases, BT3686 cleaved l-Rha-α1,4-d-GlcA linkages through a retaining double-displacement mechanism. The crystal structure of BT3686 showed that the enzyme displayed a type A seven-bladed ß-propeller fold. Mutagenesis and crystallographic studies, including the structure of BT3686 in complex with the reaction product GlcA, revealed a location for the active site among ß-propeller enzymes cited on the posterior surface of the rhamnosidase. In contrast to the vast majority of GH, the catalytic apparatus of BT3686 does not comprise a pair of carboxylic acid residues but, uniquely, a single histidine functions as the only discernable catalytic amino acid. Intriguingly, the histidine, His48, is not invariant in GH145; however, when engineered into structural homologs lacking the imidazole residue, α-l-rhamnosidase activity was established. The potential contribution of His48 to the catalytic activity of BT3686 is discussed.


Asunto(s)
Proteínas Bacterianas/química , Bacteroides thetaiotaomicron/enzimología , Glicósido Hidrolasas/química , Proteínas Bacterianas/genética , Bacteroides thetaiotaomicron/genética , Cristalografía por Rayos X , Glicósido Hidrolasas/genética , Humanos , Mutagénesis
9.
Proc Natl Acad Sci U S A ; 114(27): 7037-7042, 2017 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-28630303

RESUMEN

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.


Asunto(s)
Bacteroides/enzimología , Microbioma Gastrointestinal , Glicosaminoglicanos/química , Bacteroides/genética , Calorimetría , Carbohidratos/química , Catálisis , Cristalografía por Rayos X , Citoplasma/enzimología , Carbohidratos de la Dieta , Heparina/química , Heparitina Sulfato/química , Humanos , Microscopía Fluorescente , Mutación , Oligosacáridos/química , Polisacárido Liasas/química , Polisacáridos/química , Sulfatasas/química , Azufre/química
10.
J Biol Chem ; 293(46): 17906-17916, 2018 11 16.
Artículo en Inglés | MEDLINE | ID: mdl-30262663

RESUMEN

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.


Asunto(s)
Proteínas Bacterianas/metabolismo , Bacteroides thetaiotaomicron/enzimología , Sulfatos de Condroitina/metabolismo , Dermatán Sulfato/metabolismo , Ácido Hialurónico/metabolismo , Polisacárido Liasas/metabolismo , Proteínas Bacterianas/química , Concentración de Iones de Hidrógeno , Metales Pesados/química , Polisacárido Liasas/química , Temperatura
12.
J Biol Chem ; 292(32): 13271-13283, 2017 08 11.
Artículo en Inglés | MEDLINE | ID: mdl-28637865

RESUMEN

The human gut microbiota utilizes complex carbohydrates as major nutrients. The requirement for efficient glycan degrading systems exerts a major selection pressure on this microbial community. Thus, we propose that this microbial ecosystem represents a substantial resource for discovering novel carbohydrate active enzymes. To test this hypothesis we screened the potential enzymatic functions of hypothetical proteins encoded by genes of Bacteroides thetaiotaomicron that were up-regulated by arabinogalactan proteins or AGPs. Although AGPs are ubiquitous in plants, there is a paucity of information on their detailed structure, the function of these glycans in planta, and the mechanisms by which they are depolymerized in microbial ecosystems. Here we have discovered a new polysaccharide lyase family that is specific for the l-rhamnose-α1,4-d-glucuronic acid linkage that caps the side chains of complex AGPs. The reaction product generated by the lyase, Δ4,5-unsaturated uronic acid, is removed from AGP by a glycoside hydrolase located in family GH105, producing the final product 4-deoxy-ß-l-threo-hex-4-enepyranosyl-uronic acid. The crystal structure of a member of the novel lyase family revealed a catalytic domain that displays an (α/α)6 barrel-fold. In the center of the barrel is a deep pocket, which, based on mutagenesis data and amino acid conservation, comprises the active site of the lyase. A tyrosine is the proposed catalytic base in the ß-elimination reaction. This study illustrates how highly complex glycans can be used as a scaffold to discover new enzyme families within microbial ecosystems where carbohydrate metabolism is a major evolutionary driver.


Asunto(s)
Proteínas Bacterianas/metabolismo , Bacteroides thetaiotaomicron/enzimología , Sitios Genéticos , Modelos Moleculares , Mucoproteínas/metabolismo , Polisacárido Liasas/metabolismo , Ramnosa/metabolismo , Secuencia de Aminoácidos , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Biocatálisis , Dominio Catalítico , Secuencia Conservada , Cristalografía por Rayos X , Bases de Datos de Proteínas , Hidrólisis , Isoenzimas , Cinética , Filogenia , Proteínas de Plantas/metabolismo , Polisacárido Liasas/química , Polisacárido Liasas/genética , Conformación Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Estereoisomerismo , Especificidad por Sustrato , Tirosina
13.
Blood ; 127(26): 3360-8, 2016 06 30.
Artículo en Inglés | MEDLINE | ID: mdl-27207788

RESUMEN

Carfilzomib, a proteasome inhibitor, is approved in the United States as a single agent, and in combination with dexamethasone or lenalidomide/dexamethasone (KRd) for relapsed or refractory multiple myeloma (MM). Under the single-agent and KRd approvals, carfilzomib is administered as a 10-minute IV infusion on days 1, 2, 8, 9, 15, and 16 of 28-day cycles (20 mg/m(2) [cycle 1, days 1-2]; 27 mg/m(2) thereafter). This multicenter, single-arm, phase 1/2 study, Community Harmonized Assessment of Myeloma Patients via an Integrated Oncology Network-1 (CHAMPION-1), evaluated once-weekly carfilzomib with dexamethasone in relapsed, or relapsed and refractory MM (1-3 prior therapies). Patients received carfilzomib (30-minute IV infusion) on days 1, 8, and 15 of 28-day cycles. The phase 1 portion used a 3 + 3 dose-escalation scheme to determine the maximum tolerated dose (MTD) of carfilzomib. During phase 2, patients received carfilzomib on the same schedule at the MTD. Patients received dexamethasone (40 mg) on days 1, 8, 15, and 22; dexamethasone was omitted on day 22 for cycles 9+. A total of 116 patients were enrolled. The MTD was 70 mg/m(2), and 104 patients (phase 1/2) received carfilzomib 70 mg/m(2) At 70 mg/m(2), the median number of prior regimens was 1; and 52% were bortezomib-refractory. At 70 mg/m(2), the most common grade ≥3 adverse events were fatigue (11%) and hypertension (7%). Overall response rate at 70 mg/m(2) was 77%. Median progression-free survival was 12.6 months. These findings merit additional evaluation of the once-weekly dosing regimen. This trial was registered at www.clinicaltrials.gov as #NCT01677858.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/mortalidad , Adulto , Anciano , Anciano de 80 o más Años , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Dexametasona/administración & dosificación , Dexametasona/efectos adversos , Supervivencia sin Enfermedad , Femenino , Humanos , Infusiones Intravenosas , Masculino , Persona de Mediana Edad , Oligopéptidos/administración & dosificación , Oligopéptidos/efectos adversos , Recurrencia , Tasa de Supervivencia
14.
Am J Hematol ; 92(5): 460-466, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28213943

RESUMEN

Elotuzumab, an immunostimulatory SLAMF7-targeting monoclonal antibody, induces myeloma cell death with minimal effects on normal tissue. In a previous phase 3 study in patients with relapsed/refractory multiple myeloma (RRMM), elotuzumab (10 mg/kg, ∼3-h infusion), combined with lenalidomide and dexamethasone, demonstrated durable efficacy and acceptable safety; 10% (33/321) of patients had infusion reactions (IRs; Grade 1/2: 29; Grade 3: 4). This phase 2 study (NCT02159365) investigated an accelerated infusion schedule in 70 patients with newly diagnosed multiple myeloma or RRMM. The primary endpoint was cumulative incidence of Grade 3/4 IRs by completion of treatment Cycle 2. Dosing comprised elotuzumab 10 mg/kg intravenously (weekly, Cycles 1-2; biweekly, Cycles 3+), lenalidomide 25 mg (daily, Days 1-21), and dexamethasone (28 mg orally and 8 mg intravenously, weekly, Cycles 1-2; 40 mg orally, weekly, Cycles 3+), in 28-day cycles. Premedication with diphenhydramine, acetaminophen, and ranitidine (or their equivalents) was given as in previous studies. If no IRs occurred, infusion rate was increased in Cycle 1 from 0.5 to 2 mL/min during dose 1 (∼2 h 50 min duration) to 5 mL/min for the entire infusion by dose 3 and also during all subsequent infusions (∼1-h duration). Median number of treatment cycles was six. No Grade 3/4 IRs occurred; only one Grade 1 and one Grade 2 IR occurred, both during the first infusion. These data support the safety of a faster infusion of elotuzumab administered over ∼1 h by the third dose, providing a more convenient alternative dosing option for patients.


Asunto(s)
Anticuerpos Monoclonales Humanizados/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Mieloma Múltiple/tratamiento farmacológico , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Dexametasona/administración & dosificación , Esquema de Medicación , Humanos , Lenalidomida , Seguridad del Paciente , Premedicación/métodos , Talidomida/administración & dosificación , Talidomida/análogos & derivados
17.
Angew Chem Int Ed Engl ; 54(18): 5378-82, 2015 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-25772148

RESUMEN

α-Mannosidases and α-mannanases have attracted attention for the insight they provide into nucleophilic substitution at the hindered anomeric center of α-mannosides, and the potential of mannosidase inhibitors as cellular probes and therapeutic agents. We report the conformational itinerary of the family GH76 α-mannanases studied through structural analysis of the Michaelis complex and synthesis and evaluation of novel aza/imino sugar inhibitors. A Michaelis complex in an (O) S2 conformation, coupled with distortion of an azasugar in an inhibitor complex to a high energy B2,5 conformation are rationalized through ab initio QM/MM metadynamics that show how the enzyme surface restricts the conformational landscape of the substrate, rendering the B2,5 conformation the most energetically stable on-enzyme. We conclude that GH76 enzymes perform catalysis using an itinerary that passes through (O) S2 and B2,5 (≠) conformations, information that should inspire the development of new antifungal agents.


Asunto(s)
Bacillus/enzimología , Proteínas Bacterianas/metabolismo , Candida albicans/enzimología , Inhibidores Enzimáticos/síntesis química , Proteínas Fúngicas/metabolismo , Manosidasas/antagonistas & inhibidores , Compuestos Aza/síntesis química , Compuestos Aza/química , Compuestos Aza/farmacología , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Iminoazúcares/síntesis química , Iminoazúcares/química , Iminoazúcares/farmacología , Manosidasas/química , Modelos Moleculares , Conformación Proteica
18.
Proc Natl Acad Sci U S A ; 108(13): 5237-42, 2011 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-21393568

RESUMEN

Clostridium thermocellum is a well-characterized cellulose-degrading microorganism. The genome sequence of C. thermocellum encodes a number of proteins that contain type I dockerin domains, which implies that they are components of the cellulose-degrading apparatus, but display no significant sequence similarity to known plant cell wall-degrading enzymes. Here, we report the biochemical properties and crystal structure of one of these proteins, designated CtCel124. The protein was shown to be an endo-acting cellulase that displays a single displacement mechanism and acts in synergy with Cel48S, the major cellulosomal exo-cellulase. The crystal structure of CtCel124 in complex with two cellotriose molecules, determined to 1.5 Å, displays a superhelical fold in which a constellation of α-helices encircle a central helix that houses the catalytic apparatus. The catalytic acid, Glu96, is located at the C-terminus of the central helix, but there is no candidate catalytic base. The substrate-binding cleft can be divided into two discrete topographical domains in which the bound cellotriose molecules display twisted and linear conformations, respectively, suggesting that the enzyme may target the interface between crystalline and disordered regions of cellulose.


Asunto(s)
Celulasa/química , Celulasa/metabolismo , Estructura Secundaria de Proteína , Secuencia de Carbohidratos , Dominio Catalítico , Celulasa/genética , Celulosa/metabolismo , Clostridium thermocellum/enzimología , Clostridium thermocellum/genética , Cristalografía por Rayos X , Modelos Moleculares , Datos de Secuencia Molecular , Estructura Molecular , Oligosacáridos/química , Oligosacáridos/metabolismo , Estructura Terciaria de Proteína
19.
Essays Biochem ; 67(3): 429-442, 2023 04 18.
Artículo en Inglés | MEDLINE | ID: mdl-36562177

RESUMEN

Sulfated host glycans (mucin O-glycans and glycosaminoglycans [GAGs]) are critical nutrient sources and colonisation factors for Bacteroidetes of the human gut microbiota (HGM); a complex ecosystem comprising essential microorganisms that coevolved with humans to serve important roles in pathogen protection, immune signalling, and host nutrition. Carbohydrate sulfatases are essential enzymes to access sulfated host glycans and are capable of exquisite regio- and stereo-selective substrate recognition. In these enzymes, the common recognition features of each subfamily are correlated with their genomic and environmental context. The exo-acting carbohydrate sulfatases are attractive drug targets amenable to small-molecule screening and subsequent engineering, and their high specificity will help elucidate the role of glycan sulfation in health and disease. Inhibition of carbohydrate sulfatases provides potential routes to control Bacteroidetes growth and to explore the influence of host glycan metabolism by Bacteroidetes on the HGM ecosystem. The roles of carbohydrate sulfatases from the HGM organism Bacteroides thetaiotaomicron and the soil isolated Pedobacter heparinus (P. heparinus) in sulfated host glycan metabolism are examined and contrasted, and the structural features underpinning glycan recognition and specificity explored.


Asunto(s)
Ecosistema , Sulfatasas , Humanos , Sulfatasas/metabolismo , Polisacáridos/metabolismo , Carbohidratos , Glicosaminoglicanos/química , Glicosaminoglicanos/metabolismo , Bacterias/metabolismo
20.
Nat Commun ; 14(1): 2233, 2023 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-37076525

RESUMEN

Bacterial cell growth and division require the coordinated action of enzymes that synthesize and degrade cell wall polymers. Here, we identify enzymes that cleave the D-arabinan core of arabinogalactan, an unusual component of the cell wall of Mycobacterium tuberculosis and other mycobacteria. We screened 14 human gut-derived Bacteroidetes for arabinogalactan-degrading activities and identified four families of glycoside hydrolases with activity against the D-arabinan or D-galactan components of arabinogalactan. Using one of these isolates with exo-D-galactofuranosidase activity, we generated enriched D-arabinan and used it to identify a strain of Dysgonomonas gadei as a D-arabinan degrader. This enabled the discovery of endo- and exo-acting enzymes that cleave D-arabinan, including members of the DUF2961 family (GH172) and a family of glycoside hydrolases (DUF4185/GH183) that display endo-D-arabinofuranase activity and are conserved in mycobacteria and other microbes. Mycobacterial genomes encode two conserved endo-D-arabinanases with different preferences for the D-arabinan-containing cell wall components arabinogalactan and lipoarabinomannan, suggesting they are important for cell wall modification and/or degradation. The discovery of these enzymes will support future studies into the structure and function of the mycobacterial cell wall.


Asunto(s)
Mycobacterium tuberculosis , Polisacáridos , Humanos , Polisacáridos/metabolismo , Mycobacterium tuberculosis/metabolismo , Glicósido Hidrolasas/metabolismo , Pared Celular/metabolismo
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda