Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
IUCrJ ; 11(Pt 2): 260-274, 2024 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-38446458

RESUMO

The discovery of lytic polysaccharide monooxygenases (LPMOs), a family of copper-dependent enzymes that play a major role in polysaccharide degradation, has revealed the importance of oxidoreductases in the biological utilization of biomass. In fungi, a range of redox proteins have been implicated as working in harness with LPMOs to bring about polysaccharide oxidation. In bacteria, less is known about the interplay between redox proteins and LPMOs, or how the interaction between the two contributes to polysaccharide degradation. We therefore set out to characterize two previously unstudied proteins from the shipworm symbiont Teredinibacter turnerae that were initially identified by the presence of carbohydrate binding domains appended to uncharacterized domains with probable redox functions. Here, X-ray crystal structures of several domains from these proteins are presented together with initial efforts to characterize their functions. The analysis suggests that the target proteins are unlikely to function as LPMO electron donors, raising new questions as to the potential redox functions that these large extracellular multi-haem-containing c-type cytochromes may perform in these bacteria.


Assuntos
Gammaproteobacteria , Oxirredução , Oxigenases de Função Mista , Polissacarídeos
2.
Essays Biochem ; 67(3): 585-595, 2023 04 18.
Artigo em Inglês | MEDLINE | ID: mdl-36748351

RESUMO

The plant cell wall is rich in carbohydrates and many fungi and bacteria have evolved to take advantage of this carbon source. These carbohydrates are largely locked away in polysaccharides and so these organisms deploy a range of enzymes that can liberate individual sugars from these challenging substrates. Glycoside hydrolases (GHs) are the enzymes that are largely responsible for bringing about this sugar release; however, 12 years ago, a family of enzymes known as lytic polysaccharide monooxygenases (LPMOs) were also shown to be of key importance in this process. LPMOs are copper-dependent oxidative enzymes that can introduce chain breaks within polysaccharide chains. Initial work demonstrated that they could activate O2 to attack the substrate through a reaction that most likely required multiple electrons to be delivered to the enzyme. More recently, it has emerged that LPMO kinetics are significantly improved if H2O2 is supplied to the enzyme as a cosubstrate instead of O2. Only a single electron is required to activate an LPMO and H2O2 cosubstrate and the enzyme has been shown to catalyse multiple turnovers following the initial one-electron reduction of the copper, which is not possible if O2 is used. This has led to further studies of the roles of the electron donor in LPMO biochemistry, and this review aims to highlight recent findings in this area and consider how ongoing research could impact our understanding of the interplay between redox processes in nature.


Assuntos
Elétrons , Oxigenases de Função Mista , Oxigenases de Função Mista/química , Peróxido de Hidrogênio , Cobre , Polissacarídeos
3.
RSC Chem Biol ; 4(1): 56-64, 2023 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-36685256

RESUMO

Protein N-termini provide uniquely reactive motifs for single site protein modification. Though a number of reactions have been developed to target this site, the selectivity, generality, and stability of the conjugates formed has not been studied. We have therefore undertaken a comprehensive comparative study of the most promising methods for N-terminal protein modification, and find that there is no 'one size fits all' approach, necessitating reagent screening for a particular protein or application. Moreover, we observed limited stability in all cases, leading to a need for continued innovation and development in the bioconjugation field.

4.
Biochem J ; 478(14): 2927-2944, 2021 07 30.
Artigo em Inglês | MEDLINE | ID: mdl-34240737

RESUMO

The release of glucose from lignocellulosic waste for subsequent fermentation into biofuels holds promise for securing humankind's future energy needs. The discovery of a set of copper-dependent enzymes known as lytic polysaccharide monooxygenases (LPMOs) has galvanised new research in this area. LPMOs act by oxidatively introducing chain breaks into cellulose and other polysaccharides, boosting the ability of cellulases to act on the substrate. Although several proteins have been implicated as electron sources in fungal LPMO biochemistry, no equivalent bacterial LPMO electron donors have been previously identified, although the proteins Cbp2D and E from Cellvibrio japonicus have been implicated as potential candidates. Here we analyse a small c-type cytochrome (CjX183) present in Cellvibrio japonicus Cbp2D, and show that it can initiate bacterial CuII/I LPMO reduction and also activate LPMO-catalyzed cellulose-degradation. In the absence of cellulose, CjX183-driven reduction of the LPMO results in less H2O2 production from O2, and correspondingly less oxidative damage to the enzyme than when ascorbate is used as the reducing agent. Significantly, using CjX183 as the activator maintained similar cellulase boosting levels relative to the use of an equivalent amount of ascorbate. Our results therefore add further evidence to the impact that the choice of electron source can have on LPMO action. Furthermore, the study of Cbp2D and other similar proteins may yet reveal new insight into the redox processes governing polysaccharide degradation in bacteria.


Assuntos
Proteínas de Bactérias/metabolismo , Cellvibrio/enzimologia , Grupo dos Citocromos c/metabolismo , Oxigenases de Função Mista/metabolismo , Polissacarídeos Bacterianos/metabolismo , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Biocatálise , Celulose/metabolismo , Cellvibrio/genética , Grupo dos Citocromos c/química , Grupo dos Citocromos c/genética , Peróxido de Hidrogênio/metabolismo , Isoenzimas/química , Isoenzimas/genética , Isoenzimas/metabolismo , Oxigenases de Função Mista/química , Oxigenases de Função Mista/genética , Modelos Moleculares , Oligossacarídeos/metabolismo , Oxirredução , Oxigênio/metabolismo , Domínios Proteicos , Espectrofotometria/métodos , Especificidade por Substrato
5.
ACS Chem Biol ; 16(7): 1152-1158, 2021 07 16.
Artigo em Inglês | MEDLINE | ID: mdl-34151573

RESUMO

Antimycins are anticancer compounds produced by a hybrid nonribosomal peptide synthetase/polyketide synthase (NRPS/PKS) pathway. The biosynthesis of these compounds is well characterized, with the exception of the standalone ß-ketoreductase enzyme AntM that is proposed to catalyze the reduction of the C8 carbonyl of the antimycin scaffold. Inactivation of antM and structural characterization suggested that rather than functioning as a post-PKS tailoring enzyme, AntM acts upon the terminal biosynthetic intermediate while it is tethered to the PKS acyl carrier protein. Mutational analysis identified two amino acid residues (Tyr185 and Phe223) that are proposed to serve as checkpoints controlling substrate access to the AntM active site. Aromatic checkpoint residues are conserved in uncharacterized standalone ß-ketoreductases, indicating that they may also act concomitantly with synthesis of the scaffold. These data provide novel mechanistic insights into the functionality of standalone ß-ketoreductases and will enable their reprogramming for combinatorial biosynthesis.


Assuntos
Oxirredutases do Álcool/metabolismo , Antimicina A/análogos & derivados , Oxirredutases do Álcool/química , Oxirredutases do Álcool/genética , Sequência de Aminoácidos , Antimicina A/biossíntese , Antimicina A/metabolismo , Biocatálise , Domínio Catalítico , Biologia Computacional , Cristalografia por Raios X , Simulação de Acoplamento Molecular , Mutação , Ligação Proteica , Alinhamento de Sequência , Streptomyces/enzimologia , Especificidade por Substrato/genética
6.
Dalton Trans ; 49(11): 3413-3422, 2020 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-32125319

RESUMO

Probing the detailed interaction between lytic polysaccharide monooxygenases (LPMOs) and their polysaccharide substrates is key to revealing further insights into the mechanism of action of this class of enzymes on recalcitrant biomass. This investigation is somewhat hindered, however, by the insoluble nature of the substrates, which precludes the use of most optical spectroscopic techniques. Herein, we report a new semi-oriented EPR method which evaluates directly the binding of cellulose-active LPMOs to crystalline cellulose. We make use of the intrinsic order of cellulose fibres in Apium graveolens (celery) to orient the LPMO with respect to the magnetic field of an EPR spectrometer. The subsequent angle-dependent changes observed in the EPR spectra can then be related to the orientation of the g matrix principal directions with respect to the magnetic field of the spectrometer and, hence, to the binding of the enzyme onto the cellulose fibres. This method, which does not require specific modification of standard CW-EPR equipment, can be used as a general procedure to investigate LPMO-cellulose interactions.


Assuntos
Celulose/química , Oxigenases de Função Mista/química , Polissacarídeos/química , Apium/química , Celulose/metabolismo , Espectroscopia de Ressonância de Spin Eletrônica , Campos Magnéticos , Oxigenases de Função Mista/metabolismo , Polissacarídeos/metabolismo
7.
IUCrJ ; 6(Pt 6): 1120-1133, 2019 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-31709067

RESUMO

Indanomycin is biosynthesized by a hybrid nonribosomal peptide synthase/polyketide synthase (NRPS/PKS) followed by a number of 'tailoring' steps to form the two ring systems that are present in the mature product. It had previously been hypothesized that the indane ring of indanomycin was formed by the action of IdmH using a Diels-Alder reaction. Here, the crystal structure of a selenomethionine-labelled truncated form of IdmH (IdmH-Δ99-107) was solved using single-wavelength anomalous dispersion (SAD) phasing. This truncated variant allows consistent and easy crystallization, but importantly the structure was used as a search model in molecular replacement, allowing the full-length IdmH structure to be determined to 2.7 Šresolution. IdmH is a homodimer, with the individual protomers consisting of an α+ß barrel. Each protomer contains a deep hydrophobic pocket which is proposed to constitute the active site of the enzyme. To investigate the reaction catalysed by IdmH, 88% of the backbone NMR resonances were assigned, and using chemical shift perturbation of [15N]-labelled IdmH it was demonstrated that indanomycin binds in the active-site pocket. Finally, combined quantum mechanical/molecular mechanical (QM/MM) modelling of the IdmH reaction shows that the active site of the enzyme provides an appropriate environment to promote indane-ring formation, supporting the assignment of IdmH as the key Diels-Alderase catalysing the final step in the biosynthesis of indanomycin through a similar mechanism to other recently characterized Diels-Alderases involved in polyketide-tailoring reactions. An animated Interactive 3D Complement (I3DC) is available in Proteopedia at https://proteopedia.org/w/Journal:IUCrJ:S2052252519012399.

8.
Biotechnol Biofuels ; 12: 232, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31583018

RESUMO

BACKGROUND: The quest for novel enzymes for cellulosic biomass-degradation has recently been focussed on lytic polysaccharide monooxygenases (LPMOs/PMOs), Cu-containing proteins that catalyse the oxidative degradation of otherwise recalcitrant polysaccharides using O2 or H2O2 as a co-substrate. RESULTS: Although classical saprotrophic fungi and bacteria have been a rich source of lytic polysaccharide monooxygenases (LPMOs), we were interested to see if LPMOs from less evident bio-environments could be discovered and assessed for their cellulolytic activity in a biofuel context. In this regard, the marine shipworm Lyrodus pedicellatus represents an interesting source of new enzymes, since it must digest wood particles ingested during its natural tunnel boring behaviour and plays host to a symbiotic bacterium, Teredinibacter turnerae, the genome of which has revealed a multitude of enzymes dedicated to biomass deconstruction. Here, we show that T. turnerae encodes a cellulose-active AA10 LPMO. The 3D structure, at 1.4 Å resolution, along with its EPR spectrum is distinct from other AA10 polysaccharide monooxygenases insofar as it displays a "histidine-brace" catalytic apparatus with changes to the surrounding coordination sphere of the copper. Furthermore, TtAA10A possesses a second, surface accessible, Cu site 14 Å from the classical catalytic centre. Activity measurements show that the LPMO oxidises cellulose and thereby significantly augments the rate of degradation of cellulosic biomass by classical glycoside hydrolases. CONCLUSION: Shipworms are wood-boring marine molluscs that can live on a diet of lignocellulose. Bacterial symbionts of shipworms provide many of the enzymes needed for wood digestion. The shipworm symbiont T. turnerae produces one of the few LPMOs yet described from the marine environment, notably adding to the capability of shipworms to digest recalcitrant polysaccharides.

9.
J Biol Chem ; 294(45): 17117-17130, 2019 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-31471321

RESUMO

Lytic polysaccharide monooxygenases (LPMOs) are redox-enzymes involved in biomass degradation. All characterized LPMOs possess an active site of two highly conserved histidine residues coordinating a copper ion (the histidine brace), which are essential for LPMO activity. However, some protein sequences that belong to the AA9 LPMO family display a natural N-terminal His to Arg substitution (Arg-AA9). These are found almost entirely in the phylogenetic fungal class Agaricomycetes, associated with wood decay, but no function has been demonstrated for any Arg-AA9. Through bioinformatics, transcriptomic, and proteomic analyses we present data, which suggest that Arg-AA9 proteins could have a hitherto unidentified role in fungal degradation of lignocellulosic biomass in conjunction with other secreted fungal enzymes. We present the first structure of an Arg-AA9, LsAA9B, a naturally occurring protein from Lentinus similis The LsAA9B structure reveals gross changes in the region equivalent to the canonical LPMO copper-binding site, whereas features implicated in carbohydrate binding in AA9 LPMOs have been maintained. We obtained a structure of LsAA9B with xylotetraose bound on the surface of the protein although with a considerably different binding mode compared with other AA9 complex structures. In addition, we have found indications of protein phosphorylation near the N-terminal Arg and the carbohydrate-binding site, for which the potential function is currently unknown. Our results are strong evidence that Arg-AA9s function markedly different from canonical AA9 LPMO, but nonetheless, may play a role in fungal conversion of lignocellulosic biomass.


Assuntos
Histidina , Oxigenases de Função Mista/química , Oxigenases de Função Mista/metabolismo , Polissacarídeos/metabolismo , Motivos de Aminoácidos , Sequência de Aminoácidos , Substituição de Aminoácidos , Sítios de Ligação , Ligantes , Oxigenases de Função Mista/genética , Modelos Moleculares , Fosforilação , Filogenia
10.
J Mol Biol ; 431(5): 981-995, 2019 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-30668971

RESUMO

Dietary fiber is an important food source for members of the human gut microbiome. Members of the dominant Bacteroidetes phylum capture diverse polysaccharides via the action of multiple cell surface proteins encoded within polysaccharide utilization loci (PUL). The independent activities of PUL-encoded glycoside hydrolases (GHs) and surface glycan-binding proteins (SGBPs) for the harvest of various glycans have been studied in detail, but how these proteins work together to coordinate uptake is poorly understood. Here, we combine genetic and biochemical approaches to discern the interplay between the BoGH9 endoglucanase and the xyloglucan-binding proteins SGBP-A and SGBP-B from the Bacteroides ovatus xyloglucan utilization locus (XyGUL). The expression of BoGH9, a weakly active xyloglucanase in isolation, is required in a strain that expresses a non-binding version of SGBP-A (SGBP-A*). The crystal structure of the BoGH9 enzyme suggests the molecular basis for its robust activity on mixed-linkage ß-glucan compared to xyloglucan. However, catalytically inactive site-directed mutants of BoGH9 fail to complement the deletion of the active BoGH9 in a SGBP-A* strain. We also find that SGBP-B is needed in an SGBP-A* background to support growth on xyloglucan, but that the non-binding SGBP-B* protein acts in a dominant negative manner to inhibit growth on xyloglucan. We postulate a model whereby the SGBP-A, SGBP-B, and BoGH9 work together at the cell surface, likely within a discrete complex, and that xyloglucan binding by SGBP-B and BoGH9 may facilitate the orientation of the xyloglucan for transfer across the outer membrane.


Assuntos
Proteínas de Bactérias/metabolismo , Bacteroides/metabolismo , Microbioma Gastrointestinal/fisiologia , Trato Gastrointestinal/microbiologia , Glucanos/metabolismo , Proteínas de Membrana/metabolismo , Polissacarídeos/metabolismo , Xilanos/metabolismo , Transporte Biológico/fisiologia , Membrana Celular/metabolismo , Celulase/metabolismo , Fibras na Dieta/metabolismo , Trato Gastrointestinal/metabolismo , Glicosídeo Hidrolases/metabolismo , Humanos
11.
Methods Enzymol ; 613: 63-90, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30509474

RESUMO

Lytic polysaccharide monooxygenases (LPMOs, also known as PMOs) are a recently discovered family of enzymes that play a key role in the breakdown of polysaccharide substrates. The ability of LPMOs to introduce chain breaks, using an oxidative mechanism, has particularly attracted attention as the world seeks more cost-effective and environmentally friendly ways of producing second-generation biofuels for the future. LPMOs are copper-dependent enzymes and have an unusual active site which includes the N-terminal residue of the protein in the copper coordination sphere. This N-terminal histidine side chain is also methylated in fungal enzymes, the molecular reason for which is still a debated topic. The production of these enzymes poses several challenges if we are to understand their chemical mechanisms. Here, we describe the methods that have been used in the field to produce LPMOs and provide information on the workflows that we use for our electron paramagnetic resonance (EPR) spectroscopy experiments. EPR has been a particularly powerful tool in the study of these enzymes and our objective with this chapter is to provide some helpful information for researchers for whom this technique might be daunting or theoretically difficult to access.


Assuntos
Oxigenases de Função Mista/metabolismo , Proteínas Recombinantes/metabolismo , Espectroscopia de Ressonância de Spin Eletrônica , Escherichia coli/genética , Escherichia coli/metabolismo , Oxigenases de Função Mista/genética , Proteínas Recombinantes/genética , Especificidade por Substrato
12.
Acta Crystallogr D Struct Biol ; 74(Pt 10): 946-955, 2018 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-30289404

RESUMO

The biological conversion of lignocellulosic matter into high-value chemicals or biofuels is of increasing industrial importance as the sector slowly transitions away from nonrenewable sources. Many industrial processes involve the use of cellulolytic enzyme cocktails - a selection of glycoside hydrolases and, increasingly, polysaccharide oxygenases - to break down recalcitrant plant polysaccharides. ORFs from the genome of Teredinibacter turnerae, a symbiont hosted within the gills of marine shipworms, were identified in order to search for enzymes with desirable traits. Here, a putative T. turnerae glycoside hydrolase from family 8, hereafter referred to as TtGH8, is analysed. The enzyme is shown to be active against ß-1,4-xylan and mixed-linkage (ß-1,3,ß-1,4) marine xylan. Kinetic parameters, obtained using high-performance anion-exchange chromatography with pulsed amperometric detection and 3,5-dinitrosalicyclic acid reducing-sugar assays, show that TtGH8 catalyses the hydrolysis of ß-1,4-xylohexaose with a kcat/Km of 7.5 × 107 M-1 min-1 but displays maximal activity against mixed-linkage polymeric xylans, hinting at a primary role in the degradation of marine polysaccharides. The three-dimensional structure of TtGH8 was solved in uncomplexed and xylobiose-, xylotriose- and xylohexaose-bound forms at approximately 1.5 Šresolution; the latter was consistent with the greater kcat/Km for hexasaccharide substrates. A 2,5B boat conformation observed in the -1 position of bound xylotriose is consistent with the proposed conformational itinerary for this class of enzyme. This work shows TtGH8 to be effective at the degradation of xylan-based substrates, notably marine xylan, further exemplifying the potential of T. turnerae for effective and diverse biomass degradation.


Assuntos
Endo-1,4-beta-Xilanases/química , Gammaproteobacteria/enzimologia , Bacilos Gram-Negativos Anaeróbios Facultativos/enzimologia , Proteínas de Bactérias/química , Biomassa , Glicosídeo Hidrolases/química , Cinética , Células Vegetais/metabolismo , Plantas/química , Plantas/metabolismo , Polissacarídeos/metabolismo , Conformação Proteica , Xilanos/metabolismo
13.
Nat Commun ; 9(1): 756, 2018 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-29472725

RESUMO

Thermobia domestica belongs to an ancient group of insects and has a remarkable ability to digest crystalline cellulose without microbial assistance. By investigating the digestive proteome of Thermobia, we have identified over 20 members of an uncharacterized family of lytic polysaccharide monooxygenases (LPMOs). We show that this LPMO family spans across several clades of the Tree of Life, is of ancient origin, and was recruited by early arthropods with possible roles in remodeling endogenous chitin scaffolds during development and metamorphosis. Based on our in-depth characterization of Thermobia's LPMOs, we propose that diversification of these enzymes toward cellulose digestion might have endowed ancestral insects with an effective biochemical apparatus for biomass degradation, allowing the early colonization of land during the Paleozoic Era. The vital role of LPMOs in modern agricultural pests and disease vectors offers new opportunities to help tackle global challenges in food security and the control of infectious diseases.


Assuntos
Artrópodes/enzimologia , Proteínas de Insetos/metabolismo , Oxigenases de Função Mista/metabolismo , Polissacarídeos/metabolismo , Animais , Artrópodes/genética , Artrópodes/crescimento & desenvolvimento , Biodegradação Ambiental , Biomassa , Celulose/metabolismo , Quitina/metabolismo , Evolução Molecular , Genes de Insetos , Proteínas de Insetos/química , Proteínas de Insetos/genética , Insetos/enzimologia , Insetos/genética , Insetos/crescimento & desenvolvimento , Oxigenases de Função Mista/química , Oxigenases de Função Mista/genética , Modelos Moleculares , Filogenia , Proteômica
15.
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
16.
Nat Chem Biol ; 13(6): 610-612, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28346405

RESUMO

O-GlcNAc hydrolase (OGA) removes O-linked N-acetylglucosamine (O-GlcNAc) from a myriad of nucleocytoplasmic proteins. Through co-expression and assembly of OGA fragments, we determined the three-dimensional structure of human OGA, revealing an unusual helix-exchanged dimer that lays a structural foundation for an improved understanding of substrate recognition and regulation of OGA. Structures of OGA in complex with a series of inhibitors define a precise blueprint for the design of inhibitors that have clinical value.


Assuntos
Modelos Moleculares , beta-N-Acetil-Hexosaminidases/química , Acetilglucosamina/metabolismo , Sítios de Ligação , Ativação Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Células HEK293 , Humanos , Ligantes , Ligação Proteica , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Estrutura Terciária de Proteína , beta-N-Acetil-Hexosaminidases/genética , beta-N-Acetil-Hexosaminidases/metabolismo
17.
Dalton Trans ; 45(42): 16904-16912, 2016 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-27722375

RESUMO

The enzymatic deconstruction of recalcitrant polysaccharide biomass is central to the conversion of these substrates for societal benefit, such as in biofuels. Traditional models for enzyme-catalysed polysaccharide degradation involved the synergistic action of endo-, exo- and processive glycoside hydrolases working in concert to hydrolyse the substrate. More recently this model has been succeeded by one featuring a newly discovered class of mononuclear copper enzymes: lytic polysaccharide monooxygenases (LPMOs; classified as Auxiliary Activity (AA) enzymes in the CAZy classification). In 2013, the structure of an LPMO from Bacillus amyloliquefaciens, BaAA10, was solved with the Cu centre photoreduced to Cu(i) in the X-ray beam. Here we present the catalytic activity of BaAA10. We show that it is a chitin-active LPMO, active on both α and ß chitin, with the Cu(ii) binding with low nM KD, and the substrate greatly increasing the thermal stability of the enzyme. A spiral data collection strategy has been used to facilitate access to the previously unobservable Cu(ii) state of the active centre, revealing a coordination geometry around the copper which is distorted from axial symmetry, consistent with the previous findings from EPR spectroscopy.


Assuntos
Bacillus amyloliquefaciens/metabolismo , Proteínas de Bactérias/metabolismo , Quitina/metabolismo , Oxigenases de Função Mista/metabolismo , Bacillus amyloliquefaciens/química , Proteínas de Bactérias/química , Domínio Catalítico , Cobre/química , Cobre/metabolismo , Cristalografia por Raios X , Estabilidade Enzimática , Oxigenases de Função Mista/química , Modelos Moleculares , Polissacarídeos/metabolismo , Conformação Proteica , Especificidade por Substrato
18.
Open Biol ; 6(7)2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27466444

RESUMO

The human gastrointestinal tract harbours myriad bacterial species, collectively termed the microbiota, that strongly influence human health. Symbiotic members of our microbiota play a pivotal role in the digestion of complex carbohydrates that are otherwise recalcitrant to assimilation. Indeed, the intrinsic human polysaccharide-degrading enzyme repertoire is limited to various starch-based substrates; more complex polysaccharides demand microbial degradation. Select Bacteroidetes are responsible for the degradation of the ubiquitous vegetable xyloglucans (XyGs), through the concerted action of cohorts of enzymes and glycan-binding proteins encoded by specific xyloglucan utilization loci (XyGULs). Extending recent (meta)genomic, transcriptomic and biochemical analyses, significant questions remain regarding the structural biology of the molecular machinery required for XyG saccharification. Here, we reveal the three-dimensional structures of an α-xylosidase, a ß-glucosidase, and two α-l-arabinofuranosidases from the Bacteroides ovatus XyGUL. Aided by bespoke ligand synthesis, our analyses highlight key adaptations in these enzymes that confer individual specificity for xyloglucan side chains and dictate concerted, stepwise disassembly of xyloglucan oligosaccharides. In harness with our recent structural characterization of the vanguard endo-xyloglucanse and cell-surface glycan-binding proteins, the present analysis provides a near-complete structural view of xyloglucan recognition and catalysis by XyGUL proteins.


Assuntos
Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Bacteroides/enzimologia , Glucanos/metabolismo , Xilanos/metabolismo , Arabinose/análogos & derivados , Arabinose/química , Bacteroides/química , Cristalografia por Raios X , Trato Gastrointestinal/microbiologia , Humanos , Modelos Moleculares , Conformação Proteica , Especificidade por Substrato , Xilosidases/química , beta-Glucosidase/química
19.
J Biol Chem ; 291(24): 12838-12850, 2016 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-27129229

RESUMO

Copper-dependent lytic polysaccharide monooxygenases (LPMOs) are enzymes that oxidatively deconstruct polysaccharides. The active site copper in LPMOs is coordinated by a histidine-brace. This utilizes the amino group and side chain of the N-terminal His residue with the side chain of a second His residue to create a T-shaped arrangement of nitrogen ligands. We report a structural, kinetic, and thermodynamic appraisal of copper binding to the histidine-brace in an auxiliary activity family 10 (AA10) LPMO from Streptomyces lividans (SliLPMO10E). Unexpectedly, we discovered the existence of two apo-SliLPMO10E species in solution that can each bind copper at a single site with distinct kinetic and thermodynamic (exothermic and endothermic) properties. The experimental EPR spectrum of copper-bound SliLPMO10E requires the simulation of two different line shapes, implying two different copper-bound species, indicative of three and two nitrogen ligands coordinating the copper. Amino group coordination was probed through the creation of an N-terminal extension variant (SliLPMO10E-Ext). The kinetics and thermodynamics of copper binding to SliLPMO10E-Ext are in accord with copper binding to one of the apo-forms in the wild-type protein, suggesting that amino group coordination is absent in the two-nitrogen coordinate form of SliLPMO10E. Copper binding to SliLPMO10B was also investigated, and again it revealed the presence of two apo-forms with kinetics and stoichiometry of copper binding identical to that of SliLPMO10E. Our findings highlight that heterogeneity exists in the active site copper coordination sphere of LPMOs that may have implications for the mechanism of loading copper in the cell.


Assuntos
Proteínas de Bactérias/química , Cobre/química , Histidina/química , Oxigenases de Função Mista/química , Compostos Organometálicos/química , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Domínio Catalítico , Cobre/metabolismo , Cristalografia por Raios X , Espectroscopia de Ressonância de Spin Eletrônica , Regulação Bacteriana da Expressão Gênica , Regulação Enzimológica da Expressão Gênica , Histidina/metabolismo , Cinética , Espectrometria de Massas , Oxigenases de Função Mista/genética , Oxigenases de Função Mista/metabolismo , Modelos Moleculares , Mutação , Compostos Organometálicos/metabolismo , Polissacarídeos/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína , Espectrometria de Fluorescência , Streptomyces lividans/enzimologia , Streptomyces lividans/genética , Especificidade por Substrato , Termodinâmica
20.
Nat Chem Biol ; 12(4): 298-303, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26928935

RESUMO

Lytic polysaccharide monooxygenases (LPMOs) are copper-containing enzymes that oxidatively break down recalcitrant polysaccharides such as cellulose and chitin. Since their discovery, LPMOs have become integral factors in the industrial utilization of biomass, especially in the sustainable generation of cellulosic bioethanol. We report here a structural determination of an LPMO-oligosaccharide complex, yielding detailed insights into the mechanism of action of these enzymes. Using a combination of structure and electron paramagnetic resonance spectroscopy, we reveal the means by which LPMOs interact with saccharide substrates. We further uncover electronic and structural features of the enzyme active site, showing how LPMOs orchestrate the reaction of oxygen with polysaccharide chains.


Assuntos
Celulose/metabolismo , Quitina/metabolismo , Oxigenases de Função Mista/metabolismo , Sequência de Aminoácidos , Aspergillus oryzae/enzimologia , Aspergillus oryzae/genética , Sítios de Ligação , Domínio Catalítico , Cobre/metabolismo , Cristalografia por Raios X , Transferência Ressonante de Energia de Fluorescência , Lentinula/enzimologia , Lentinula/genética , Oxigenases de Função Mista/química , Oxigenases de Função Mista/genética , Modelos Moleculares , Dados de Sequência Molecular , Oligossacarídeos/química , Oxirredução , Especificidade por Substrato
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...