Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
J Biol Chem ; 300(3): 105774, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38382672

RESUMO

Gum arabic (GA) is widely used as an emulsion stabilizer and edible coating and consists of a complex carbohydrate moiety with a rhamnosyl-glucuronate group capping the non-reducing ends. Enzymes that can specifically cleave the glycosidic chains of GA and modify their properties are valuable for structural analysis and industrial application. Cryogenic X-ray crystal structure of GA-specific L-rhamnose-α-1,4-D-glucuronate lyase from Fusarium oxysporum (FoRham1), belonging to the polysaccharide lyase (PL) family 42, has been previously reported. To determine the specific reaction mechanism based on its hydrogen-containing enzyme structure, we performed joint X-ray/neutron crystallography of FoRham1. Large crystals were grown in the presence of L-rhamnose (a reaction product), and neutron and X-ray diffraction datasets were collected at room temperature at 1.80 and 1.25 Å resolutions, respectively. The active site contained L-rhamnose and acetate, the latter being a partial analog of glucuronate. Incomplete H/D exchange between Arg166 and acetate suggested that a strong salt-bridge interaction was maintained. Doubly deuterated His105 and deuterated Tyr150 supported the interaction between Arg166 and the acetate. The unique hydrogen-rich environment functions as a charge neutralizer for glucuronate and stabilizes the oxyanion intermediate. The NE2 atom of His85 was deprotonated and formed a hydrogen bond with the deuterated O1 hydroxy of L-rhamnose, indicating the function of His85 as the base/acid catalyst for bond cleavage via ß-elimination. Asp83 functions as a pivot between the two catalytic histidine residues by bridging them. This His-His-Asp structural motif is conserved in the PL 24, 25, and 42 families.


Assuntos
Fusarium , Polissacarídeo-Liases , Humanos , Acetatos , Cristalografia por Raios X , Ácido Glucurônico/química , Hidrogênio , Liases , Polissacarídeo-Liases/química , Ramnose/química , Fusarium/enzimologia
2.
J Biol Chem ; : 107596, 2024 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-39032652

RESUMO

Alginate is a polysaccharide consumed by humans in edible seaweed and different foods where it is applied as a texturizing hydrocolloid or in encapsulations of drugs and probiotics. While gut bacteria are found to utilize and ferment alginate to health beneficial short chain fatty acids, knowledge on details of the molecular reactions is sparse. Alginates are composed of mannuronic acid (M) and its C-5 epimer guluronic acid (G). An alginate related polysaccharide utilization locus (PUL) has been identified in the gut bacterium Bacteroides eggerthii DSM 20697. The PUL encodes two polysaccharide lyases (PLs) from the PL6 (BePL6) and PL17 (BePL17) families as well as a KdgF-like metalloprotein (BeKdgF) known to catalyze ring-opening of 4,5-unsaturated monouronates yielding 4-deoxy-l-erythro-5-hexoseulose uronate (DEH). B. eggerthii DSM 20697 does not grow on alginate, but readily proliferates with a lag phase of a few hours in the presence of an endo-acting alginate lyase A1-I from the marine bacterium Sphingomonas sp. A1. The B. eggerthii lyases are both exo-acting and while BePL6 is strictly G-block specific, BePL17 prefers M-blocks. BeKdgF retained 10-27% activity in the presence of 0.1-1 mM EDTA. X-ray crystallography was used to investigate the three-dimensional structure of BeKdgF, based on which a catalytic mechanism was proposed to involve Asp102, acting as acid/base having pKa of 5.9 as determined by NMR pH titration. BePL6 and BePL17 cooperate in alginate degradation with BeKdgF linearizing produced 4,5-unsaturated monouronates. Their efficiency of alginate degradation was much enhanced by addition of the A1-I alginate lyase.

3.
J Biol Chem ; 296: 100203, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33334876

RESUMO

The exopolysaccharide poly-ß-(1→6)-N-acetylglucosamine (PNAG) is a major structural determinant of bacterial biofilms responsible for persistent and nosocomial infections. The enzymatic dispersal of biofilms by PNAG-hydrolyzing glycosidase enzymes, such as Dispersin B (DspB), is a possible approach to treat biofilm-dependent bacterial infections. The cationic charge resulting from partial de-N-acetylation of native PNAG is critical for PNAG-dependent biofilm formation. We recently demonstrated that DspB has increased catalytic activity on de-N-acetylated PNAG oligosaccharides, but the molecular basis for this increased activity is not known. Here, we analyze the role of anionic amino acids surrounding the catalytic pocket of DspB in PNAG substrate recognition and hydrolysis using a combination of site-directed mutagenesis, activity measurements using synthetic PNAG oligosaccharide analogs, and in vitro biofilm dispersal assays. The results of these studies support a model in which bound PNAG is weakly associated with a shallow anionic groove on the DspB protein surface with recognition driven by interactions with the -1 GlcNAc residue in the catalytic pocket. An increased rate of hydrolysis for cationic PNAG was driven, in part, by interaction with D147 on the anionic surface. Moreover, we identified that a DspB mutant with improved hydrolysis of fully acetylated PNAG oligosaccharides correlates with improved in vitro dispersal of PNAG-dependent Staphylococcus epidermidis biofilms. These results provide insight into the mechanism of substrate recognition by DspB and suggest a method to improve DspB biofilm dispersal activity by mutation of the amino acids within the anionic binding surface.


Assuntos
Aggregatibacter actinomycetemcomitans/metabolismo , Aminoácidos/metabolismo , Proteínas de Bactérias/metabolismo , Glicosídeo Hidrolases/metabolismo , beta-Glucanas/metabolismo , Biofilmes , Hidrólise , Modelos Moleculares
4.
J Biol Chem ; 295(13): 4316-4326, 2020 03 27.
Artigo em Inglês | MEDLINE | ID: mdl-31871050

RESUMO

Recent work exploring protein sequence space has revealed a new glycoside hydrolase (GH) family (GH164) of putative mannosidases. GH164 genes are present in several commensal bacteria, implicating these genes in the degradation of dietary glycans. However, little is known about the structure, mechanism of action, and substrate specificity of these enzymes. Herein we report the biochemical characterization and crystal structures of the founding member of this family (Bs164) from the human gut symbiont Bacteroides salyersiae. Previous reports of this enzyme indicated that it has α-mannosidase activity, however, we conclusively show that it cleaves only ß-mannose linkages. Using NMR spectroscopy, detailed enzyme kinetics of WT and mutant Bs164, and multiangle light scattering we found that it is a trimeric retaining ß-mannosidase, that is susceptible to several known mannosidase inhibitors. X-ray crystallography revealed the structure of Bs164, the first known structure of a GH164, at 1.91 Å resolution. Bs164 is composed of three domains: a (ß/α)8 barrel, a trimerization domain, and a ß-sandwich domain, representing a previously unobserved structural-fold for ß-mannosidases. Structures of Bs164 at 1.80-2.55 Å resolution in complex with the inhibitors noeuromycin, mannoimidazole, or 2,4-dinitrophenol 2-deoxy-2-fluoro-mannoside reveal the residues essential for specificity and catalysis including the catalytic nucleophile (Glu-297) and acid/base residue (Glu-160). These findings further our knowledge of the mechanisms commensal microbes use for nutrient acquisition.


Assuntos
Glicosídeo Hidrolases/genética , Conformação Proteica , Relação Estrutura-Atividade , beta-Manosidase/genética , Sequência de Aminoácidos/genética , Bacteroides/enzimologia , Bacteroides/ultraestrutura , Catálise , Domínio Catalítico/genética , Cristalografia por Raios X , Glicosídeo Hidrolases/química , Glicosídeo Hidrolases/ultraestrutura , Humanos , Cinética , Modelos Moleculares , Especificidade por Substrato , beta-Manosidase/química , beta-Manosidase/ultraestrutura
5.
J Biol Chem ; 295(31): 10638-10652, 2020 07 31.
Artigo em Inglês | MEDLINE | ID: mdl-32518157

RESUMO

Infections by many bacterial pathogens rely on their ability to degrade host glycans by producing glycoside hydrolases (GHs). Here, we discovered a conserved multifunctional GH, SsGalNagA, containing a unique combination of two family 32 carbohydrate-binding modules (CBM), a GH16 domain and a GH20 domain, in the zoonotic pathogen Streptococcus suis 05ZYH33. Enzymatic assays revealed that the SsCBM-GH16 domain displays endo-(ß1,4)-galactosidase activity specifically toward the host-derived αGal epitope Gal(α1,3)Gal(ß1,4)Glc(NAc)-R, whereas the SsGH20 domain has a wide spectrum of exo-ß-N-acetylhexosaminidase activities, including exo-(ß1,3)-N-acetylglucosaminidase activity, and employs this activity to act in tandem with SsCBM-GH16 on the αGal-epitope glycan. Further, we found that the CBM32 domain adjacent to the SsGH16 domain is indispensable for SsGH16 catalytic activity. Surface plasmon resonance experiments uncovered that both CBM32 domains specifically bind to αGal-epitope glycan, and together they had a KD of 3.5 mm toward a pentasaccharide αGal-epitope glycan. Cell-binding and αGal epitope removal assays revealed that SsGalNagA efficiently binds to both swine erythrocytes and tracheal epithelial cells and removes the αGal epitope from these cells, suggesting that SsGalNagA functions in nutrient acquisition or alters host signaling in S. suis Both binding and removal activities were blocked by an αGal-epitope glycan. SsGalNagA is the first enzyme reported to sequentially act on a glycan containing the αGal epitope. These findings shed detailed light on the evolution of GHs and an important host-pathogen interaction.


Assuntos
Proteínas de Bactérias/química , Epitopos/química , Glicosídeo Hidrolases/química , Polissacarídeos Bacterianos/química , Streptococcus suis/química , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Dissacarídeos/química , Dissacarídeos/genética , Dissacarídeos/metabolismo , Epitopos/genética , Epitopos/metabolismo , Glicosídeo Hidrolases/genética , Glicosídeo Hidrolases/metabolismo , Camundongos , Polissacarídeos Bacterianos/genética , Polissacarídeos Bacterianos/metabolismo , Domínios Proteicos , Coelhos , Streptococcus suis/genética , Streptococcus suis/metabolismo , Suínos
6.
J Biol Chem ; 294(23): 9172-9185, 2019 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-31010825

RESUMO

The 6-deoxy sugar l-rhamnose (l-Rha) is found widely in plant and microbial polysaccharides and natural products. The importance of this and related compounds in host-pathogen interactions often means that l-Rha plays an essential role in many organisms. l-Rha is most commonly biosynthesized as the activated sugar nucleotide uridine 5'-diphospho-ß-l-rhamnose (UDP-ß-l-Rha) or thymidine 5'-diphospho-ß-l-rhamnose (TDP-ß-l-Rha). Enzymes involved in the biosynthesis of these sugar nucleotides have been studied in some detail in bacteria and plants, but the activated form of l-Rha and the corresponding biosynthetic enzymes have yet to be explored in algae. Here, using sugar-nucleotide profiling in two representative algae, Euglena gracilis and the toxin-producing microalga Prymnesium parvum, we show that levels of UDP- and TDP-activated l-Rha differ significantly between these two algal species. Using bioinformatics and biochemical methods, we identified and characterized a fusion of the RmlC and RmlD proteins, two bacteria-like enzymes involved in TDP-ß-l-Rha biosynthesis, from P. parvum Using this new sequence and also others, we explored l-Rha biosynthesis among algae, finding that although most algae contain sequences orthologous to plant-like l-Rha biosynthesis machineries, instances of the RmlC-RmlD fusion protein identified here exist across the Haptophyta and Gymnodiniaceae families of microalgae. On the basis of these findings, we propose potential routes for the evolution of nucleoside diphosphate ß-l-Rha (NDP-ß-l-Rha) pathways among algae.


Assuntos
Proteínas de Algas/metabolismo , Carboidratos Epimerases/metabolismo , Haptófitas/metabolismo , Ramnose/biossíntese , Proteínas de Algas/genética , Carboidratos Epimerases/classificação , Carboidratos Epimerases/genética , Filogenia , Plastídeos/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Ramnose/química , Simbiose
7.
J Biol Chem ; 294(52): 19978-19987, 2019 12 27.
Artigo em Inglês | MEDLINE | ID: mdl-31740581

RESUMO

Glucuronoyl esterases (GEs) catalyze the cleavage of ester linkages between lignin and glucuronic acid moieties on glucuronoxylan in plant biomass. As such, GEs represent promising biochemical tools in industrial processing of these recalcitrant resources. However, details on how GEs interact and catalyze degradation of their natural substrates are sparse, calling for thorough enzyme structure-function studies. Presented here is a structural and mechanistic investigation of the bacterial GE OtCE15A. GEs belong to the carbohydrate esterase family 15 (CE15), which is in turn part of the larger α/ß-hydrolase superfamily. GEs contain a Ser-His-Asp/Glu catalytic triad, but the location of the catalytic acid in GEs has been shown to be variable, and OtCE15A possesses two putative catalytic acidic residues in the active site. Through site-directed mutagenesis, we demonstrate that these residues are functionally redundant, possibly indicating the evolutionary route toward new functionalities within the family. Structures determined with glucuronate, in both native and covalently bound intermediate states, and galacturonate provide insights into the catalytic mechanism of CE15. A structure of OtCE15A with the glucuronoxylooligosaccharide 23-(4-O-methyl-α-d-glucuronyl)-xylotriose (commonly referred to as XUX) shows that the enzyme can indeed interact with polysaccharides from the plant cell wall, and an additional structure with the disaccharide xylobiose revealed a surface binding site that could possibly indicate a recognition mechanism for long glucuronoxylan chains. Collectively, the results indicate that OtCE15A, and likely most of the CE15 family, can utilize esters of glucuronoxylooligosaccharides and support the proposal that these enzymes work on lignin-carbohydrate complexes in plant biomass.


Assuntos
Proteínas de Bactérias/metabolismo , Esterases/metabolismo , Lignina/metabolismo , Verrucomicrobia/enzimologia , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Sítios de Ligação , Domínio Catalítico , Dissacarídeos/química , Dissacarídeos/metabolismo , Esterases/química , Esterases/genética , Ácido Glucurônico/química , Ácido Glucurônico/metabolismo , Cinética , Lignina/química , Simulação de Dinâmica Molecular , Mutagênese Sítio-Dirigida , Oligossacarídeos/química , Oligossacarídeos/metabolismo , Especificidade por Substrato
8.
J Biol Chem ; 294(37): 13833-13849, 2019 09 13.
Artigo em Inglês | MEDLINE | ID: mdl-31416836

RESUMO

Aspergillus fumigatus is an opportunistic fungal pathogen that causes both chronic and acute invasive infections. Galactosaminogalactan (GAG) is an integral component of the A. fumigatus biofilm matrix and a key virulence factor. GAG is a heterogeneous linear α-1,4-linked exopolysaccharide of galactose and GalNAc that is partially deacetylated after secretion. A cluster of five co-expressed genes has been linked to GAG biosynthesis and modification. One gene in this cluster, ega3, is annotated as encoding a putative α-1,4-galactosaminidase belonging to glycoside hydrolase family 114 (GH114). Herein, we show that recombinant Ega3 is an active glycoside hydrolase that disrupts GAG-dependent A. fumigatus and Pel polysaccharide-dependent Pseudomonas aeruginosa biofilms at nanomolar concentrations. Using MS and functional assays, we demonstrate that Ega3 is an endo-acting α-1,4-galactosaminidase whose activity depends on the conserved acidic residues, Asp-189 and Glu-247. X-ray crystallographic structural analysis of the apo Ega3 and an Ega3-galactosamine complex, at 1.76 and 2.09 Å resolutions, revealed a modified (ß/α)8-fold with a deep electronegative cleft, which upon ligand binding is capped to form a tunnel. Our structural analysis coupled with in silico docking studies also uncovered the molecular determinants for galactosamine specificity and substrate binding at the -2 to +1 binding subsites. The findings in this study increase the structural and mechanistic understanding of the GH114 family, which has >600 members encoded by plant and opportunistic human pathogens, as well as in industrially used bacteria and fungi.


Assuntos
Aspergillus fumigatus/metabolismo , Glicosídeo Hidrolases/genética , Hexosaminidases/metabolismo , Aspergillus fumigatus/genética , Aspergillus fumigatus/ultraestrutura , Biofilmes/efeitos dos fármacos , Cristalografia por Raios X/métodos , Proteínas Fúngicas/genética , Fungos/metabolismo , Glicosídeo Hidrolases/metabolismo , Hexosaminidases/farmacologia , Hexosaminidases/ultraestrutura , Polissacarídeos/metabolismo , Virulência
9.
Appl Environ Microbiol ; 86(23)2020 11 10.
Artigo em Inglês | MEDLINE | ID: mdl-32917756

RESUMO

Laminarin is an abundant algal polysaccharide that serves as carbon storage and fuel to meet the nutrition demands of heterotrophic microbes. Laminarin depolymerization catalyzed by microbial extracellular enzymes initiates remineralization, a key process in ocean biogeochemical cycles. Here, we described a glycoside hydrolase 16 (GH16) family laminarinase from a marine alga-associated Flavobacterium at the biochemical and structural levels. We found that the endolytic enzyme cleaved laminarin with a preference for ß-1,3-glycoside linkages and showed transglycosylation activity across a broad range of acceptors. We also solved and compared high-resolution crystal structures of laminarinase in the apo form and in complex with ß-1,3-tetrasaccharides, revealing an expanded catalytic cleft formed following substrate binding. Moreover, structure and mutagenesis studies identified multiple specific contacts between the enzyme and glucosyl residues essential for the substrate specificity for ß-1,3-glucan. These results provide novel insights into the structural requirements for substrate binding and catalysis of GH16 family laminarinase, enriching our understanding of bacterial utilization of algal laminarin.IMPORTANCE Heterotrophic bacterial communities are key players in marine biogeochemical cycling due to their ability to remineralize organic carbon. Processing of complex organic matter requires heterotrophic bacteria to produce extracellular enzymes with precise specificity to depolymerize substrates to sizes sufficiently small for uptake. Thus, extracellular enzymatic hydrolysis initiates microbe-driven heterotrophic carbon cycling. In this study, based on biochemical and structural analyses, we revealed the depolymerization mechanism of ß-1,3-glucan, a carbon reserve in algae, by laminarinase from an alga-associated marine Flavobacterium The findings provide new insights into the substrate recognition and catalysis of bacterial laminarinase and promote a better understanding of how extracellular enzymes are involved in organic matter cycling.


Assuntos
Proteínas de Bactérias/metabolismo , Celulases/metabolismo , Flavobacteriaceae/enzimologia , Proteínas de Bactérias/química , Celulases/química , Conformação Proteica , Especificidade por Substrato
10.
J Biol Chem ; 293(42): 16277-16290, 2018 10 19.
Artigo em Inglês | MEDLINE | ID: mdl-30171074

RESUMO

Sialic acids are a family of more than 50 structurally distinct acidic sugars on the surface of all vertebrate cells where they terminate glycan chains and are exposed to many interactions with the surrounding environment. In particular, sialic acids play important roles in cell-cell and host-pathogen interactions. The sialic acids or related nonulosonic acids have been observed in Deuterostome lineages, Eubacteria, and Archaea but are notably absent from plants. However, the structurally related C8 acidic sugar 3-deoxy-d-manno-2-octulosonic acid (Kdo) is present in Gram-negative bacteria and plants as a component of bacterial lipopolysaccharide and pectic rhamnogalacturonan II in the plant cell wall. Until recently, sialic acids were not thought to occur in algae, but as in plants, Kdo has been observed in algae. Here, we report the de novo biosynthesis of the deaminated sialic acid, 3-deoxy-d-glycero-d-galacto-2-nonulosonic acid (Kdn), in the toxin-producing microalga Prymnesium parvum Using biochemical methods, we show that this alga contains CMP-Kdn and identified and recombinantly expressed the P. parvum genes encoding Kdn-9-P synthetase and CMP-Kdn synthetase enzymes that convert mannose-6-P to CMP-Kdn. Bioinformatics analysis revealed sequences related to those of the two P. parvum enzymes, suggesting that sialic acid biosynthesis is likely more widespread among microalgae than previously thought and that this acidic sugar may play a role in host-pathogen interactions involving microalgae. Our findings provide evidence that P. parvum has the biosynthetic machinery for de novo production of the deaminated sialic acid Kdn and that sialic acid biosynthesis may be common among microalgae.


Assuntos
Haptófitas/metabolismo , Microalgas/metabolismo , Ácido N-Acetilneuramínico/biossíntese , Vias Biossintéticas , Monofosfato de Citidina/análogos & derivados , Monofosfato de Citidina/biossíntese , Ácidos Neuramínicos
11.
J Biol Chem ; 293(11): 4026-4036, 2018 03 16.
Artigo em Inglês | MEDLINE | ID: mdl-29382716

RESUMO

Ulvan is a major cell wall component of green algae of the genus Ulva, and some marine bacteria encode enzymes that can degrade this polysaccharide. The first ulvan-degrading lyases have been recently characterized, and several putative ulvan lyases have been recombinantly expressed, confirmed as ulvan lyases, and partially characterized. Two families of ulvan-degrading lyases, PL24 and PL25, have recently been established. The PL24 lyase LOR_107 from the bacterial Alteromonadales sp. strain LOR degrades ulvan endolytically, cleaving the bond at the C4 of a glucuronic acid. However, the mechanism and LOR_107 structural features involved are unknown. We present here the crystal structure of LOR_107, representing the first PL24 family structure. We found that LOR_107 adopts a seven-bladed ß-propeller fold with a deep canyon on one side of the protein. Comparative sequence analysis revealed a cluster of conserved residues within this canyon, and site-directed mutagenesis disclosed several residues essential for catalysis. We also found that LOR_107 uses the His/Tyr catalytic mechanism, common to several PL families. We captured a tetrasaccharide substrate in the structures of two inactive mutants, which indicated a two-step binding event, with the first substrate interaction near the top of the canyon coordinated by Arg320, followed by sliding of the substrate into the canyon toward the active-site residues. Surprisingly, the LOR_107 structure was very similar to that of the PL25 family PLSV_3936, despite only ∼14% sequence identity between the two enzymes. On the basis of our structural and mutational analyses, we propose a catalytic mechanism for LOR_107 that differs from the typical His/Tyr mechanism.


Assuntos
Alteromonadaceae/enzimologia , Mutação , Polissacarídeo-Liases/química , Polissacarídeo-Liases/metabolismo , Polissacarídeos/metabolismo , Catálise , Domínio Catalítico , Cristalografia por Raios X , Modelos Moleculares , Mutagênese Sítio-Dirigida , Polissacarídeo-Liases/genética , Conformação Proteica , Relação Estrutura-Atividade , Especificidade por Substrato
12.
J Biol Chem ; 293(29): 11564-11573, 2018 07 20.
Artigo em Inglês | MEDLINE | ID: mdl-29875159

RESUMO

Ulvan is a complex sulfated polysaccharide present in the cell wall of green algae of the genus Ulva (Chlorophyta). The first ulvan-degrading polysaccharide lyases were identified several years ago, and more were discovered through genome sequencing of marine bacteria. Ulvan lyases are now grouped in three polysaccharide lyase (PL) families in the CAZy database, PL24, PL25, and PL28. The recently determined structures of the representative lyases from families PL24 and PL25 show that they adopt a seven-bladed ß-propeller fold and utilize the His/Tyr catalytic mechanism. No structural information is yet available for PL28 ulvan lyases. NLR48 from Nonlabens ulvanivorans belongs to PL28 together with its close paralog, NLR42. Biochemical studies of NLR42 have revealed that it can cleave ulvan next to both uronic acid epimers. We report the crystal structure of ulvan lyase NLR48 at 1.9-Å resolution. It has a ß-jelly roll fold with an extended, deep, and positively charged substrate-binding cleft. Putative active-site residues were identified from the sequence conservation pattern, and their role was confirmed by site-directed mutagenesis. The structure of an inactive K162M mutant with a tetrasaccharide substrate showed the substrate occupying the "-" subsites. Comparison with lyases from other PL families with ß-jelly roll folds supported assignment of the active site and explained its ability to degrade ulvan next to either epimer of uronic acid. NLR48 contains the His/Tyr catalytic machinery with Lys162 and Tyr281 playing the catalytic base/acid roles.


Assuntos
Flavobacteriaceae/enzimologia , Polissacarídeo-Liases/metabolismo , Domínio Catalítico , Cristalografia por Raios X , Flavobacteriaceae/química , Flavobacteriaceae/metabolismo , Modelos Moleculares , Oligossacarídeos/metabolismo , Polissacarídeo-Liases/química , Conformação Proteica , Especificidade por Substrato
13.
J Biol Chem ; 293(14): 5150-5159, 2018 04 06.
Artigo em Inglês | MEDLINE | ID: mdl-29444825

RESUMO

Periplasmic solute-binding proteins in bacteria are involved in the active transport of nutrients into the cytoplasm. In marine bacteria of the genus Vibrio, a chitooligosaccharide-binding protein (CBP) is thought to be the major solute-binding protein controlling the rate of chitin uptake in these bacteria. However, the molecular mechanism of the CBP involvement in chitin metabolism has not been elucidated. Here, we report the structure and function of a recombinant chitooligosaccharide-binding protein from Vibrio harveyi, namely VhCBP, expressed in Escherichia coli Isothermal titration calorimetry revealed that VhCBP strongly binds shorter chitooligosaccharides ((GlcNAc) n , where n = 2, 3, and 4) with affinities that are considerably greater than those for glycoside hydrolase family 18 and 19 chitinases but does not bind longer ones, including insoluble chitin polysaccharides. We also found that VhCBP comprises two domains with flexible linkers and that the domain-domain interface forms the sugar-binding cleft, which is not long extended but forms a small cavity. (GlcNAc)2 bound to this cavity, apparently triggering a closed conformation of VhCBP. Trp-363 and Trp-513, which stack against the two individual GlcNAc rings, likely make a major contribution to the high affinity of VhCBP for (GlcNAc)2 The strong chitobiose binding, followed by the conformational change of VhCBP, may facilitate its interaction with an active-transport system in the inner membrane of Vibrio species.


Assuntos
Quitina/química , Vibrio/metabolismo , Sequência de Aminoácidos , Metabolismo dos Carboidratos/fisiologia , Carboidratos , Proteínas de Transporte/metabolismo , Quitina/análogos & derivados , Quitina/metabolismo , Quitinases/metabolismo , Quitosana , Cristalografia por Raios X/métodos , Modelos Moleculares , Oligossacarídeos , Periplasma/metabolismo , Relação Estrutura-Atividade
14.
J Biol Chem ; 292(32): 13271-13283, 2017 08 11.
Artigo em Inglês | MEDLINE | ID: mdl-28637865

RESUMO

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.


Assuntos
Proteínas de Bactérias/metabolismo , Bacteroides thetaiotaomicron/enzimologia , Loci Gênicos , Modelos Moleculares , Mucoproteínas/metabolismo , Polissacarídeo-Liases/metabolismo , Ramnose/metabolismo , Sequência de Aminoácidos , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Biocatálise , Domínio Catalítico , Sequência Conservada , Cristalografia por Raios X , Bases de Dados de Proteínas , Hidrólise , Isoenzimas , Cinética , Filogenia , Proteínas de Plantas/metabolismo , Polissacarídeo-Liases/química , Polissacarídeo-Liases/genética , Conformação Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Estereoisomerismo , Especificidade por Substrato , Tirosina
15.
Regul Toxicol Pharmacol ; 98: 140-150, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-30048706

RESUMO

A safety assessment was conducted for a symthetic variant Cytophaga sp. α-amylase enzyme expressed in Bacillus licheniformis and formulated into two distinct product formats: whole broth (a preparation in which the production organism is completely inactivated, but containing residual cell debris) and clarified preparation (from which the production organism is completely removed). The enzyme was improved via modern biotechnology techniques for use in the endohydrolysis of starch, glycogen, related polysaccharides and oligosaccharides. Applications range from carbohydrate processing, including the manufacture of sweeteners, fermentation to produce organic acids, amino acids and their salts, and potable or fuel alcohol, with resulting co-products (distillers' grains and corn gluten feed/meal) destined for use in animal feed. The toxicological studies summarized in this article (90-day rodent oral gavage and in vitro genotoxicity studies) noted no test article-related adverse effects and thus substantiate the safety of the α-amylase in not only the clarified form but also as a whole-broth preparation. Consistent with the decision tree analysis for enzymes produced with modern biotechnology techniques, this paper provides supporting information that this variant amylase with homology to an amylase from a potentially pathogenic organism (Cytophaga sp.) can be safely produced in an expression host that belongs to a Safe Strain Lineage, for safe use as processing aid to manufacture human and animal food.


Assuntos
Bacillus licheniformis/enzimologia , Proteínas de Bactérias/biossíntese , Cytophaga/enzimologia , alfa-Amilases/toxicidade , Administração Oral , Animais , Bacillus licheniformis/genética , Proteínas de Bactérias/genética , Cytophaga/genética , Feminino , Genes Bacterianos , Humanos , Microbiologia Industrial , Linfócitos/efeitos dos fármacos , Masculino , Testes de Mutagenicidade , Ratos , alfa-Amilases/genética
16.
J Biol Chem ; 291(3): 1289-306, 2016 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-26567911

RESUMO

Many pectin methylesterases (PMEs) are expressed in plants to modify plant cell-wall pectins for various physiological roles. These pectins are also attacked by PMEs from phytopathogens and phytophagous insects. The de-methylesterification by PMEs of the O6-methyl ester groups of the homogalacturonan component of pectin, exposing galacturonic acids, can occur processively or non-processively, respectively, describing sequential versus single de-methylesterification events occurring before enzyme-substrate dissociation. The high resolution x-ray structures of a PME from Aspergillus niger in deglycosylated and Asn-linked N-acetylglucosamine-stub forms reveal a 10⅔-turn parallel ß-helix (similar to but with less extensive loops than bacterial, plant, and insect PMEs). Capillary electrophoresis shows that this PME is non-processive, halophilic, and acidophilic. Molecular dynamics simulations and electrostatic potential calculations reveal very different behavior and properties compared with processive PMEs. Specifically, uncorrelated rotations are observed about the glycosidic bonds of a partially de-methyl-esterified decasaccharide model substrate, in sharp contrast to the correlated rotations of processive PMEs, and the substrate-binding groove is negatively not positively charged.


Assuntos
Aspergillus niger/enzimologia , Hidrolases de Éster Carboxílico/química , Proteínas Fúngicas/química , Modelos Moleculares , Sequência de Aminoácidos , Substituição de Aminoácidos , Hidrolases de Éster Carboxílico/genética , Hidrolases de Éster Carboxílico/metabolismo , Domínio Catalítico , Sequência Consenso , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Concentração de Íons de Hidrogênio , Simulação de Dinâmica Molecular , Dados de Sequência Molecular , Mutação , Concentração Osmolar , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Conformação Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Alinhamento de Sequência , Homologia Estrutural de Proteína , Especificidade por Substrato , Difração de Raios X
17.
J Biol Chem ; 291(30): 15551-63, 2016 07 22.
Artigo em Inglês | MEDLINE | ID: mdl-27231344

RESUMO

Extremophiles are expected to represent a source of enzymes having unique functional properties. The hypothetical protein NIS_0185, termed NitAly in this study, was identified as an alginate lyase-homolog protein in the genomic database of ϵ-Proteobacteria Nitratiruptor sp. SB155-2, which was isolated from deep-sea hydrothermal vents at a water depth of 1,000 m. Among the characterized alginate lyases in the polysaccharide lyase family 7 (PL-7), the amino acid sequence of NitAly showed the highest identity (39%) with that of red alga Pyropia yezoensis alginate lyase PyAly. Recombinant NitAly (rNitAly) was successfully expressed in Escherichia coli Purified rNitAly degraded alginate in an endolytic manner. Among alginate block types, polyM was preferable to polyG and polyMG as a substrate, and its end degradation products were mainly tri-, tetra-, and penta-saccharides. The optimum temperature and pH values were 70 °C and around 6, respectively. A high concentration of NaCl (0.8-1.4 m) was required for maximum activity. In addition, a 50% loss of activity was observed after incubation at 67 °C for 30 min. Heat stability was decreased in the presence of 5 mm DTT, and Cys-80 and Cys-232 were identified as the residues responsible for heat stability but not lyase activity. Introducing two cysteines into PyAly based on homology modeling using Pseudomonas aeruginosa alginate lyase PA1167 as the template enhanced its heat stability. Thus, NitAly is a functional alginate lyase, with its unique optimum conditions adapted to its environment. These insights into the heat stability of NitAly could be applied to improve that of other PL-7 alginate lyases.


Assuntos
Proteínas de Bactérias/química , Epsilonproteobacteria/enzimologia , Temperatura Alta , Fontes Hidrotermais/microbiologia , Polissacarídeo-Liases/química , Proteínas de Bactérias/genética , Estabilidade Enzimática , Epsilonproteobacteria/genética , Concentração de Íons de Hidrogênio , Oceanos e Mares , Polissacarídeo-Liases/genética , Domínios Proteicos
18.
J Biol Chem ; 291(43): 22572-22582, 2016 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-27601466

RESUMO

Ribose can be used for energy or as a component of several important biomolecules, but for it to be used in either capacity it must first be phosphorylated by ribokinase (RBSK). RBSK proteins are part of the phosphofructokinase-B (pfkB) family of carbohydrate kinases. Sequence comparisons of pfkB proteins from the model plant Arabidopsis thaliana with the human and Escherichia coli RBSK identified a single candidate RBSK, At1g17160 (AtRBSK). AtRBSK is more similar to predicted RBSKs from other plant species and known mammalian and prokaryotic RBSK than to all other PfkB proteins in Arabidopsis AtRBSK contains a predicted chloroplast transit peptide, and we confirmed plastid localization using AtRBSK fused to YFP. Structure prediction software verified that the AtRBSK sequence mapped onto a known RBSK structure. Kinetic parameters of purified recombinant AtRBSK were determined to be Kmribose = 150 µm ± 17 µm, KmATP = 45 µm ± 5.6 µm, and kcat = 2.0 s-1 Substrate inhibition was observed for AtRBSK (KiATP = 2.44 mm ± 0.36 mm), as has been demonstrated for other RBSK proteins. Ribose accumulated in Arabidopsis plants lacking AtRBSK. Such plants grew normally unless media was supplemented with ribose, which led to chlorosis and growth inhibition. Both chlorosis and ribose accumulation were abolished upon the introduction of a transgene expressing AtRBSK-MYC, demonstrating that the loss of protein is responsible for ribose hypersensitivity. Ribose accumulation in plants lacking AtRBSK was reduced in plants also deficient in the nucleoside ribohydrolase NSH1, linking AtRBSK activity to nucleoside metabolism.


Assuntos
Proteínas de Arabidopsis/metabolismo , Arabidopsis/enzimologia , Nucleosídeos/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Ribose/metabolismo , Arabidopsis/genética , Proteínas de Arabidopsis/genética , Escherichia coli/enzimologia , Escherichia coli/genética , Humanos , Nucleosídeos/genética , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Ribose/genética
19.
J Biol Chem ; 291(45): 23709-23718, 2016 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-27629413

RESUMO

By inspection of the predicted proteome of the fungus Myceliophthora thermophila C1 for vanillyl-alcohol oxidase (VAO)-type flavoprotein oxidases, a putative oligosaccharide oxidase was identified. By homologous expression and subsequent purification, the respective protein could be obtained. The protein was found to contain a bicovalently bound FAD cofactor. By screening a large number of carbohydrates, several mono- and oligosaccharides could be identified as substrates. The enzyme exhibits a strong substrate preference toward xylooligosaccharides; hence it is named xylooligosaccharide oxidase (XylO). Chemical analyses of the product formed upon oxidation of xylobiose revealed that the oxidation occurs at C1, yielding xylobionate as product. By elucidation of several XylO crystal structures (in complex with a substrate mimic, xylose, and xylobiose), the residues that tune the unique substrate specificity and regioselectivity could be identified. The discovery of this novel oligosaccharide oxidase reveals that the VAO-type flavoprotein family harbors oxidases tuned for specific oligosaccharides. The unique substrate profile of XylO hints at a role in the degradation of xylan-derived oligosaccharides by the fungus M. thermophila C1.


Assuntos
Glucuronatos/metabolismo , Oligossacarídeos/metabolismo , Oxirredutases/metabolismo , Sordariales/enzimologia , Sequência de Aminoácidos , Cristalografia por Raios X , Dissacarídeos/metabolismo , Flavina-Adenina Dinucleotídeo/metabolismo , Modelos Moleculares , Oxirredução , Oxirredutases/química , Conformação Proteica , Alinhamento de Sequência , Sordariales/química , Sordariales/metabolismo , Especificidade por Substrato , Xilanos/metabolismo
20.
J Biol Chem ; 290(44): 26638-48, 2015 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-26354439

RESUMO

The most abundant carbohydrate product of cellulosic biomass pyrolysis is the anhydrosugar levoglucosan (1,6-anhydro-ß-d-glucopyranose), which can be converted to glucose 6-phosphate by levoglucosan kinase (LGK). In addition to the canonical kinase phosphotransfer reaction, the conversion requires cleavage of the 1,6-anhydro ring to allow ATP-dependent phosphorylation of the sugar O6 atom. Using x-ray crystallography, we show that LGK binds two magnesium ions in the active site that are additionally coordinated with the nucleotide and water molecules to result in ideal octahedral coordination. To further verify the metal binding sites, we co-crystallized LGK in the presence of manganese instead of magnesium and solved the structure de novo using the anomalous signal from four manganese atoms in the dimeric structure. The first metal is required for catalysis, whereas our work suggests that the second is either required or significantly promotes the catalytic rate. Although the enzyme binds its sugar substrate in a similar orientation to the structurally related 1,6-anhydro-N-acetylmuramic acid kinase (AnmK), it forms markedly fewer bonding interactions with the substrate. In this orientation, the sugar is in an optimal position to couple phosphorylation with ring cleavage. We also observed a second alternate binding orientation for levoglucosan, and in these structures, ADP was found to bind with lower affinity. These combined observations provide an explanation for the high Km of LGK for levoglucosan. Greater knowledge of the factors that contribute to the catalytic efficiency of LGK can be used to improve applications of this enzyme for levoglucosan-derived biofuel production.


Assuntos
Celulose/química , Proteínas Fúngicas/química , Glucose-6-Fosfato/química , Glucose/análogos & derivados , Lipomyces/química , Fosfotransferases/química , Biocatálise , Biomassa , Celulose/metabolismo , Cristalografia por Raios X , Escherichia coli/genética , Escherichia coli/metabolismo , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Expressão Gênica , Glucose/química , Glucose/metabolismo , Glucose-6-Fosfato/metabolismo , Cinética , Lipomyces/enzimologia , Magnésio/química , Magnésio/metabolismo , Manganês/química , Manganês/metabolismo , Modelos Moleculares , Fosforilação , Fosfotransferases/genética , Fosfotransferases/metabolismo , Ligação Proteica , Conformação Proteica , Multimerização Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA