Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 138
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Nature ; 621(7980): 840-848, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37674084

RESUMO

In both cancer and infections, diseased cells are presented to human Vγ9Vδ2 T cells through an 'inside out' signalling process whereby structurally diverse phosphoantigen (pAg) molecules are sensed by the intracellular domain of butyrophilin BTN3A11-4. Here we show how-in both humans and alpaca-multiple pAgs function as 'molecular glues' to promote heteromeric association between the intracellular domains of BTN3A1 and the structurally similar butyrophilin BTN2A1. X-ray crystallography studies visualized that engagement of BTN3A1 with pAgs forms a composite interface for direct binding to BTN2A1, with various pAg molecules each positioned at the centre of the interface and gluing the butyrophilins with distinct affinities. Our structural insights guided mutagenesis experiments that led to disruption of the intracellular BTN3A1-BTN2A1 association, abolishing pAg-mediated Vγ9Vδ2 T cell activation. Analyses using structure-based molecular-dynamics simulations, 19F-NMR investigations, chimeric receptor engineering and direct measurement of intercellular binding force revealed how pAg-mediated BTN2A1 association drives BTN3A1 intracellular fluctuations outwards in a thermodynamically favourable manner, thereby enabling BTN3A1 to push off from the BTN2A1 ectodomain to initiate T cell receptor-mediated γδ T cell activation. Practically, we harnessed the molecular-glue model for immunotherapeutics design, demonstrating chemical principles for developing both small-molecule activators and inhibitors of human γδ T cell function.


Assuntos
Butirofilinas , Ativação Linfocitária , Fosfoproteínas , Receptores de Antígenos de Linfócitos T gama-delta , Linfócitos T , Animais , Humanos , Antígenos CD/imunologia , Antígenos CD/metabolismo , Butirofilinas/imunologia , Butirofilinas/metabolismo , Camelídeos Americanos/imunologia , Simulação de Dinâmica Molecular , Fosfoproteínas/imunologia , Fosfoproteínas/metabolismo , Receptores de Antígenos de Linfócitos T gama-delta/imunologia , Receptores de Antígenos de Linfócitos T gama-delta/metabolismo , Linfócitos T/citologia , Linfócitos T/imunologia , Linfócitos T/metabolismo , Cristalografia por Raios X , Ressonância Magnética Nuclear Biomolecular , Termodinâmica
2.
Immunity ; 50(4): 1043-1053.e5, 2019 04 16.
Artigo em Inglês | MEDLINE | ID: mdl-30902636

RESUMO

Human Vγ9Vδ2 T cells respond to microbial infections and malignancy by sensing diphosphate-containing metabolites called phosphoantigens, which bind to the intracellular domain of butyrophilin 3A1, triggering extracellular interactions with the Vγ9Vδ2 T cell receptor (TCR). Here, we examined the molecular basis of this "inside-out" triggering mechanism. Crystal structures of intracellular butyrophilin 3A proteins alone or in complex with the potent microbial phosphoantigen HMBPP or a synthetic analog revealed key features of phosphoantigens and butyrophilins required for γδ T cell activation. Analyses with chemical probes and molecular dynamic simulations demonstrated that dimerized intracellular proteins cooperate in sensing HMBPP to enhance the efficiency of γδ T cell activation. HMBPP binding to butyrophilin doubled the binding force between a γδ T cell and a target cell during "outside" signaling, as measured by single-cell force microscopy. Our findings provide insight into the "inside-out" triggering of Vγ9Vδ2 T cell activation by phosphoantigen-bound butyrophilin, facilitating immunotherapeutic drug design.


Assuntos
Antígenos CD/química , Butirofilinas/química , Ativação Linfocitária , Organofosfatos/metabolismo , Subpopulações de Linfócitos T/imunologia , Antígenos CD/metabolismo , Sítios de Ligação , Butirofilinas/metabolismo , Cristalografia por Raios X , Dimerização , Desenho de Fármacos , Humanos , Ligação de Hidrogênio , Imunoterapia , Modelos Moleculares , Simulação de Dinâmica Molecular , Mutagênese Sítio-Dirigida , Conformação Proteica , Domínios Proteicos , Isoformas de Proteínas/química , Processamento de Proteína Pós-Traducional , Receptores de Antígenos de Linfócitos T gama-delta , Análise de Célula Única , Relação Estrutura-Atividade , Subpopulações de Linfócitos T/metabolismo
3.
Appl Environ Microbiol ; 88(9): e0249721, 2022 05 10.
Artigo em Inglês | MEDLINE | ID: mdl-35435717

RESUMO

Nonheme iron- and α-ketoglutarate (αKG)-dependent halogenases (NHFeHals), which catalyze the regio- and stereoselective halogenation of the unactivated C(sp3)-H bonds, exhibit tremendous potential in the challenging asymmetric halogenation. AdeV from Actinomadura sp. ATCC 39365 is the first identified carrier protein-free NHFeHal that catalyzes the chlorination of nucleotide 2'-deoxyadenosine-5'-monophosphate (2'-dAMP) to afford 2'-chloro-2'-deoxyadenosine-5'-monophosphate. Here, we determined the complex crystal structures of AdeV/FeII/Cl and AdeV/FeII/Cl/αKG at resolutions of 1.76 and 1.74 Å, respectively. AdeV possesses a typical ß-sandwich topology with H194, H252, αKG, chloride, and one water molecule coordinating FeII in the active site. Molecular docking, mutagenesis, and biochemical analyses reveal that the hydrophobic interactions and hydrogen bond network between the substrate-binding pocket and the adenine, deoxyribose, and phosphate moieties of 2'-dAMP are essential for substrate recognition. Residues H111, R177, and H192 might play important roles in the second-sphere interactions that control reaction partitioning. This study provides valuable insights into the catalytic selectivity of AdeV and will facilitate the rational engineering of AdeV and other NHFeHals for synthesis of halogenated nucleotides. IMPORTANCE Halogenated nucleotides are a group of important antibiotics and are clinically used as antiviral and anticancer drugs. AdeV is the first carrier protein-independent nonheme iron- and α-ketoglutarate (αKG)-dependent halogenase (NHFeHal) that can selectively halogenate nucleotides and exhibits restricted substrate specificity toward several 2'-dAMP analogues. Here, we determined the complex crystal structures of AdeV/FeII/Cl and AdeV/FeII/Cl/αKG. Molecular docking, mutagenesis, and biochemical analyses provide important insights into the catalytic selectivity of AdeV. This study will facilitate the rational engineering of AdeV and other carrier protein-independent NHFeHals for synthesis of halogenated nucleotides.


Assuntos
Halogenação , Ácidos Cetoglutáricos , Proteínas de Transporte , Compostos Ferrosos , Halogênios , Ferro/química , Simulação de Acoplamento Molecular , Nucleotídeos
4.
Biochem Biophys Res Commun ; 579: 54-61, 2021 11 19.
Artigo em Inglês | MEDLINE | ID: mdl-34587555

RESUMO

1,2-ß-Mannobiose phosphorylases (1,2-ß-MBPs) from glycoside hydrolase 130 (GH130) family are important bio-catalysts in glycochemistry applications owing to their ability in synthesizing oligomannans. Here, we report the crystal structure of a thermostable 1,2-ß-MBP from Thermoanaerobacter sp. X-514 termed Teth514_1789 to reveal the molecular basis of its higher thermostability and mechanism of action. We also solved the enzyme complexes of mannose, mannose-1-phosphate (M1P) and 1,4-ß-mannobiose to manifest the enzyme-substrate interaction networks of three main subsites. Notably, a Zn ion that should be derived from crystallization buffer was found in the active site and coordinates the phosphate moiety of M1P. Nonetheless, this Zn-coordination should reflect an inhibitory status as supplementing Zn severely impairs the enzyme activity. These results indicate that the effects of metal ions should be taken into consideration when applying Teth514_1789 and other related enzymes. Based on the structure, a reliable model of Teth514_1788 that shares 61.7% sequence identity to Teth514_1789 but displays a different substrate preference was built. Analyzing the structural features of these two closely related enzymes, we hypothesized that the length of a loop fragment that covers the entrance of the catalytic center might regulate the substrate selectivity. In conclusion, these information provide in-depth understanding of GH130 1,2-ß-MBPs and should serve as an important guidance for enzyme engineering for further applications.


Assuntos
Thermoanaerobacter/enzimologia , beta-Manosidase/química , Sítios de Ligação , Catálise , Domínio Catalítico , Glicosídeo Hidrolases/química , Íons , Ligantes , Mananas/química , Manose/química , Manosefosfatos/química , Fosforilases/química , Plasmídeos/metabolismo , Conformação Proteica , Reprodutibilidade dos Testes , Eletricidade Estática , Temperatura , Zinco/química
5.
Biochem Biophys Res Commun ; 534: 73-78, 2021 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-33310191

RESUMO

Glycosylation catalyzed by uridine diphosphate-dependent glycosyltransferases (UGT) contributes to the chemical and functional diversity of a number of natural products. Bacillus subtilis Bs-YjiC is a robust and versatile UGT that holds potentials in the biosynthesis of unnatural bioactive ginsenosides. To understand the molecular mechanism underlying the substrate promiscuity of Bs-YjiC, we solved crystal structures of Bs-YjiC and its binary complex with uridine diphosphate (UDP) at resolution of 2.18 Å and 2.44 Å, respectively. Bs-YjiC adopts the classical GT-B fold containing the N-terminal and C-terminal domains that accommodate the sugar acceptor and UDP-glucose, respectively. Molecular docking indicates that the spacious sugar-acceptor binding pocket of Bs-YjiC might be responsible for its broad substrate spectrum and unique glycosylation patterns toward protopanaxadiol-(PPD) and PPD-type ginsenosides. Our study reveals the structural basis for the aglycone promiscuity of Bs-YjiC and will facilitate the protein engineering of Bs-YjiC to synthesize novel bioactive glycosylated compounds.


Assuntos
Bacillus subtilis/enzimologia , Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Glicosiltransferases/química , Glicosiltransferases/metabolismo , Proteínas de Bactérias/genética , Sítios de Ligação , Cristalografia por Raios X , Ginsenosídeos/química , Ginsenosídeos/metabolismo , Glicosilação , Glicosiltransferases/genética , Modelos Moleculares , Simulação de Acoplamento Molecular , Domínios Proteicos , Sapogeninas/metabolismo , Especificidade por Substrato , Difosfato de Uridina/química , Difosfato de Uridina/metabolismo , Uridina Difosfato Glucose/metabolismo
6.
Chembiochem ; 22(8): 1317-1328, 2021 04 16.
Artigo em Inglês | MEDLINE | ID: mdl-33232569

RESUMO

Cytochrome P450s are heme-thiolate enzymes that participate in carbon source assimilation, natural compound biosynthesis and xenobiotic metabolism in all kingdoms of life. P450s can catalyze various reactions by using a wide range of organic compounds, thus exhibiting great potential in biotechnological applications. The catalytic reactions of P450s are driven by electron equivalents that are sourced from pyridine nucleotides and delivered by cognate or matching redox partners (RPs). The electron transfer (ET) route from RPs to P450s involves one or more redox center-containing domains. As the rate of ET is one of the main determinants of P450 efficacy, an in-depth understanding of the P450 ET pathway should increase our knowledge of these important enzymes and benefit their further applications. Here, the various P450 RP systems along with current understanding of their ET routes will be reviewed. Notably, state-of-the-art structural studies of the two main types of self-sufficient P450 will also be summarized.


Assuntos
Sistema Enzimático do Citocromo P-450/metabolismo , Biocatálise , Sistema Enzimático do Citocromo P-450/química , Transporte de Elétrons , Humanos , Modelos Moleculares
7.
Biochem Biophys Res Commun ; 521(1): 31-36, 2020 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-31653344

RESUMO

The epimerase MoeE5 from Streptomyces viridosporus converts UDP-glucuronic acid (UDP-GlcA) to UDP-galacturonic acid (UDP-GalA) to provide the first sugar in synthesizing moenomycin, a potent inhibitor against bacterial peptidoglycan glycosyltransferases. The enzyme belongs to the UDP-hexose 4-epimerase family, and uses NAD+ as its cofactor. Here we present the complex crystal structures of MoeE5/NAD+/UDP-GlcA and MoeE5/NAD+/UDP-glucose, determined at 1.48 Šand 1.66 Šresolution. The cofactor NAD+ is bound to the N-terminal Rossmann-fold domain and the substrate is bound to the smaller C-terminal domain. In both crystals the C4 atom of the sugar moiety of the substrate is in close proximity to the C4 atom of the nicotinamide of NAD+, and the O4 atom of the sugar is also hydrogen bonded to the side chain of Tyr154, suggesting a productive binding mode. As the first complex structure of this protein family with a bound UDP-GlcA in the active site, it shows an extensive hydrogen-bond network between the enzyme and the substrate. We further built a model with the product UDP-GalA, and found that the unique Arg192 of MoeE5 might play an important role in the catalytic pathway. Consequently, MoeE5 is likely a specific epimerase for UDP-GlcA to UDP-GalA conversion, rather than a promiscuous enzyme as some other family members.


Assuntos
Antibacterianos/biossíntese , Oligossacarídeos/biossíntese , UDPglucose 4-Epimerase/metabolismo , Antibacterianos/química , Cristalografia por Raios X , Modelos Moleculares , Oligossacarídeos/química , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Streptomyces/enzimologia , Especificidade por Substrato , UDPglucose 4-Epimerase/química , UDPglucose 4-Epimerase/genética
8.
Biochem Biophys Res Commun ; 529(2): 156-161, 2020 08 20.
Artigo em Inglês | MEDLINE | ID: mdl-32703404

RESUMO

Thebaine synthase 2 (THS2) that can transform (7S)-salutaridinol 7-O-acetate to thebaine catalyzes the final step of thebaine biosynthesis in Papaver somniferum. Here, the crystal structures of THS2 and its complex with thebaine are reported, revealing the interaction network in the substrate-binding pocket. Subsequent docking and QM/MM studies was performed to further explore the catalytic mechanism of THS2. Our results suggest that T105 may abstract the proton of C4-OH from the substrate under the assistance of H89. The resulting C4-O- phenolate anion then attacks the nearby C5, and triggers intramolecular SN2' syn displacement with the elimination of O-acetyl group. Moreover, the latter SN2' reaction is the rate-determining step of the whole enzymatic reaction with an overall energy barrier of 18.8 kcal/mol. These findings are of pivotal importance to understand the mechanism of action of thebaine biosynthesis, and would guide enzyme engineering to enhance the production of opiate alkaloids via metabolic engineering.


Assuntos
Ligases/metabolismo , Papaver/enzimologia , Proteínas de Plantas/metabolismo , Tebaína/metabolismo , Cristalografia por Raios X , Ligases/química , Modelos Moleculares , Papaver/química , Papaver/metabolismo , Proteínas de Plantas/química , Conformação Proteica , Teoria Quântica
9.
Biochem Biophys Res Commun ; 532(1): 108-113, 2020 10 29.
Artigo em Inglês | MEDLINE | ID: mdl-32828542

RESUMO

African Swine Fever Virus (ASFV) is an enveloped double-stranded DNA icosahedral virus that causes the devastating hemorrhagic fever of pigs. ASFV infections severely impact swine production and cause an enormous economic loss, but no effective vaccine and therapeutic regimen is available. pA151R is a non-structural protein of ASFV, which is expressed at both early and late stages of viral infection. Significantly, pA151R may play a key role in ASFV replication and virus assembly as suppressing pA151R expression can reduce virus replication. However, little is known about the functional and structural mechanisms of pA151R because it shares a very low sequence identity to known structures. It was proposed that pA151R might participate in the redox pathway owing to the presence of a thioredoxin active site feature, the WCTKC motif. In this study, we determined the crystal structure of pA151R. Based on the crystal structure, we found that pA151R comprises of a central five-stranded ß-sheet packing against two helices on one side and an incompact C-terminal region containing the WCTKC motif on the other side. Notably, two cysteines in the WCTKC motif, an additional cysteine C116 from the ß7-ß8 loop together with ND1 of H109 coordinate a Zn2+ ion to form a Zn-binding motif. These findings suggest that the structure of pA151R is significantly different from that of typical thioredoxins. Our structure should provide molecular insights into the understanding of functional and structural mechanisms of pA151R from ASFV and shall benefit the development of prophylactic and therapeutic anti-ASFV agents.


Assuntos
Vírus da Febre Suína Africana/química , Proteínas não Estruturais Virais/química , Febre Suína Africana/virologia , Vírus da Febre Suína Africana/genética , Vírus da Febre Suína Africana/fisiologia , Animais , Sítios de Ligação/genética , Cristalografia por Raios X , Genes Virais , Modelos Moleculares , Conformação Proteica , Multimerização Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Eletricidade Estática , Homologia Estrutural de Proteína , Sus scrofa , Suínos , Tiorredoxinas/química , Tiorredoxinas/genética , Tiorredoxinas/fisiologia , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/fisiologia
10.
Biochem Biophys Res Commun ; 527(3): 799-804, 2020 06 30.
Artigo em Inglês | MEDLINE | ID: mdl-32423809

RESUMO

When administrated orally, the vasodilating drug diltiazem can be metabolized into diacetyl diltiazem in the presence of Bacteroides thetaiotaomicron, a human gut microbe. The removal of acetyl group from the parent drug is carried out by the GDSL/SGNH-family hydrolase BT4096. Here the crystal structure of the enzyme was solved by mercury soaking and single-wavelength anomalous diffraction. The protein folds into two parts. The N-terminal part comprises the catalytic domain which is similar to other GDSL/SGNH hydrolases. The flanking C-terminal part is made up of a ß-barrel subdomain and an α-helical subdomain. Structural comparison shows that the catalytic domain is most akin to acetyl-xylooligosaccharide esterase and allows a plausible binding mode of diltiazem to be proposed. The ß-barrel subdomain is similar in topology to the immunoglobulin-like domains, including some carbohydrate-binding modules, of various bacterial glycoside hydrolases. Consequently, BT4096 might originally function as an oligosaccharide deacetylase with additional subdomains that could enhance substrate binding, and it acts on diltiazem just by accident.


Assuntos
Proteínas de Bactérias/metabolismo , Bacteroides thetaiotaomicron/enzimologia , Diltiazem/metabolismo , Microbioma Gastrointestinal , Hidrolases/metabolismo , Vasodilatadores/metabolismo , Acetilação , Proteínas de Bactérias/química , Bacteroides thetaiotaomicron/química , Bacteroides thetaiotaomicron/metabolismo , Domínio Catalítico , Humanos , Hidrolases/química , Modelos Moleculares , Conformação Proteica , Especificidade por Substrato
11.
Chembiochem ; 21(20): 2950-2956, 2020 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-32421914

RESUMO

A 2,3-dihydroxybenzoic acid decarboxylase from Fusarium oxysporum (2,3-DHBD_Fo) has a relatively high catalytic efficiency for the decarboxylation of 2,3-dihydroxybenzoic acid (DHBA) and carboxylation of catechol, thus it has a different substrate spectrum from other benzoic acid decarboxylases. We have determined the structures of 2,3-DHBD_Fo in its apo form and complexes with catechol or 2,5-dihydroxybenzoic acid at 1.55, 1.97, and 2.45 Šresolution, respectively. The crystal structures of 2,3-DHBD_Fo show that the enzyme exists as a homotetramer, and each active center has a Zn2+ ion coordinated by E8, H167, D291 and three water molecules. This is different from 2,6-DHBD from Rhizobium sporomusa, in which the Zn2+ ion is also coordinated with H10. Surprisingly, mutation of A10 of 2,3-DHBD_Fo to His resulted in almost complete loss of the enzyme activity. Enzyme-substrate docking and site-directed mutation studies indicate that residue R233Δ interacts with the 3-hydroxy group of 2,3-DHBA, and plays an important role in substrate recognition for this enzyme, thus revealing the molecular basis 2,3-dihydroxybenzoic acid decarboxylase.


Assuntos
Carboxiliases/química , Fusarium/enzimologia , Carboxiliases/genética , Carboxiliases/metabolismo , Cristalografia por Raios X , Simulação de Acoplamento Molecular , Conformação Proteica , Especificidade por Substrato
12.
Biochemistry ; 58(38): 3943-3954, 2019 09 24.
Artigo em Inglês | MEDLINE | ID: mdl-31436959

RESUMO

Members of an important group of industrial enzymes, Rhizopus lipases, exhibit valuable hydrolytic features that underlie their biological functions. Particularly important is their N-terminal polypeptide segment (NTPS), which is required for secretion and proper folding but is removed in the process of enzyme maturation. A second common feature of this class of lipases is the α-helical "lid", which regulates the accessibility of the substrate to the enzyme active site. Some Rhizopus lipases also exhibit "interfacial activation" by micelle and/or aggregate surfaces. While it has long been recognized that the NTPS is critical for function, its dynamic features have frustrated efforts to characterize its structure by X-ray crystallography. Here, we combine nuclear magnetic resonance spectroscopy and X-ray crystallography to determine the structure and dynamics of Rhizopus chinensis lipase (RCL) with its 27-residue NTPS prosequence (r27RCL). Both r27RCL and the truncated mature form of RCL (mRCL) exhibit biphasic interfacial activation kinetics with p-nitrophenyl butyrate (pNPB). r27RCL exhibits a substrate binding affinity significantly lower than that of mRCL due to stabilization of the closed lid conformation by the NTPS. In contrast to previous predictions, the NTPS does not enhance lipase activity by increasing surface hydrophobicity but rather inhibits activity by forming conserved interactions with both the closed lid and the core protein structure. Single-site mutations and kinetic studies were used to confirm that the NTPS serves as internal competitive inhibitor and to develop a model of the associated process of interfacial activation. These structure-function studies provide the basis for engineering RCL lipases with enhanced catalytic activities.


Assuntos
Proteínas Fúngicas/química , Microbiologia Industrial , Lipase/química , Peptídeos/química , Rhizopus/enzimologia , Sequência de Aminoácidos , Cristalografia por Raios X , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Hidrólise , Cinética , Lipase/genética , Lipase/metabolismo , Ressonância Magnética Nuclear Biomolecular , Peptídeos/genética , Peptídeos/metabolismo , Relação Estrutura-Atividade , Especificidade por Substrato
13.
Biochem Biophys Res Commun ; 511(4): 800-805, 2019 04 16.
Artigo em Inglês | MEDLINE | ID: mdl-30837154

RESUMO

The antibiotic moenomycin A is a phosphoglycerate derivative with a C25-moenocinyl chain and a branched oligosaccharide. Formation of the C25-chain is catalyzed by the enzyme MoeN5 with geranyl pyrophosphate (GPP) and the sugar-linked 2-Z,E-farnesyl-3-phosphoglycerate (FPG) as its substrates. Previous complex crystal structures with GPP and long-chain alkyl glycosides suggested that GPP binds to the S1 site in a similar way as in most other α-helical prenyltransferases (PTs), and FPG is likely to assume a bent conformation in the S2 site. However, two FPG derivatives synthesized in the current study were found in the S1 site rather than S2 in their complex crystal structures with MoeN5. Apparently S1 is the preferred site for prenyl-containing ligand, and S2 binding may proceed only after S1 is occupied. Thus, like most trans-type PTs, MoeN5 may employ a sequential ionization-condensation-elimination mechanism that involves a carbocation intermediate.


Assuntos
Proteínas de Bactérias/metabolismo , Dimetilaliltranstransferase/metabolismo , Streptomyces/metabolismo , 2,3-Difosfoglicerato/química , 2,3-Difosfoglicerato/metabolismo , Sequência de Aminoácidos , Proteínas de Bactérias/química , Bambermicinas/metabolismo , Cristalografia por Raios X , Dimetilaliltranstransferase/química , Simulação de Acoplamento Molecular , Conformação Proteica , Alinhamento de Sequência , Streptomyces/química , Especificidade por Substrato
14.
Biochem Biophys Res Commun ; 515(4): 621-626, 2019 08 06.
Artigo em Inglês | MEDLINE | ID: mdl-31178134

RESUMO

The polyprenoid glycan carriers are produced by cis-prenyltransferases (cis-PTs), which function as heterodimers in metazoa and fungi or homodimers in bacteria, but both are found in plants, protista and archaea. Heterodimeric cis-PTs comprise catalytic and non-catalytic subunits while homodimeric enzymes contain two catalytic subunits. The non-catalytic subunits of cis-PT shows low sequence similarity to known cis-PTs and their structure information is of great interests. Here we report the crystal structure of Nus1, the non-catalytic subunit of cis-PT from Saccharomyces cerevisiae. We also investigate the heterodimer formation and active site conformation by constructing a homology model of Nus1 and its catalytic subunit. Nus1 does not contain an active site, but its C-terminus may participate in catalysis by interacting with the substrates bound to the catalytic subunit. These results provide important basis for further investigation of heterodimeric cis-PTs.


Assuntos
Alquil e Aril Transferases/química , Dimetilaliltranstransferase/química , Proteínas de Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/enzimologia , Catálise , Domínio Catalítico , Ligação Proteica , Multimerização Proteica
15.
Biochem Biophys Res Commun ; 512(3): 517-523, 2019 05 07.
Artigo em Inglês | MEDLINE | ID: mdl-30904161

RESUMO

Invasive aspergillosis (IA) is a life-threatening disease impacting immunocompromised individuals. Standard treatments of IA, including polyenes and azoles, suffer from high toxicity and emerging resistance, leading to the need to develop new antifungal agents with novel mechanisms of action. Ergosterol biosynthesis is a classic target for antifungals, and squalene synthase (SQS) catalyzes the first committed step in ergosterol biosynthesis in Aspergillus spp. making SQS of interest in the context of antifungal development. Here, we cloned, expressed, purified and characterized SQS from the pathogen Aspergillus flavus (AfSQS), confirming that it produced squalene. To identify potential leads targeting AfSQS, we tested known squalene synthase inhibitors, zaragozic acid and the phosphonosulfonate BPH-652, finding that they were potent inhibitors. We then screened a library of 744 compounds from the National Cancer Institute (NCI) Diversity Set V for inhibition activity. 20 hits were identified and IC50 values were determined using dose-response curves. 14 compounds that interfered with the assay were excluded and the remaining 6 compounds were analyzed for drug-likeness, resulting in one compound, celastrol, which had an AfSQS IC50 value of 830 nM. Enzyme inhibition kinetics revealed that celastrol binds to AfSQS in a noncompetitive manner, but did not bind covalently. Since celastrol is also known to inhibit growth of the highly virulent Aspergillus fumigatus by inhibiting flavin-dependent monooxygenase siderophore A (SidA, under iron starvation conditions), it may be a promising multi-target lead for antifungal development.


Assuntos
Antifúngicos/farmacologia , Aspergillus flavus/enzimologia , Inibidores Enzimáticos/farmacologia , Farnesil-Difosfato Farnesiltransferase/antagonistas & inibidores , Farnesil-Difosfato Farnesiltransferase/metabolismo , Aspergilose/tratamento farmacológico , Aspergilose/microbiologia , Aspergillus flavus/genética , Aspergillus flavus/metabolismo , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Clonagem Molecular , Farnesil-Difosfato Farnesiltransferase/genética , Humanos , Modelos Moleculares , Terapia de Alvo Molecular , Triterpenos Pentacíclicos , Ácidos Tricarboxílicos/farmacologia , Triterpenos/farmacologia
16.
Biochem Biophys Res Commun ; 516(3): 801-805, 2019 08 27.
Artigo em Inglês | MEDLINE | ID: mdl-31256936

RESUMO

Hydroalkoxylation is a useful and efficient reaction which generates C-O bond and produces cyclic ethers, the common structural elements of natural products. The dedicative enzyme which can catalyze enantioselective hydroalkoxylation named PhnH was recently identified in the herqueinone biosynthetic gene from Penicillium herquei. It catalyzes addition of a phenol to the terminal olefin on substrate to produce a dihydrobenzofuran. Here, the crystal structure of PhnH is reported and the putative substrate-binding pocket is illustrated. Through docking experiment, possible substrate-binding poses are displayed and the catalytic mechanism is therefore proposed. Our findings form the basis for further studies of enantioselective hydroalkoxylation enzymes.


Assuntos
Proteínas Fúngicas/química , Penicillium/enzimologia , Fenalenos/síntese química , Álcoois/química , Benzofuranos/química , Sítios de Ligação , Catálise , Clonagem Molecular , Cristalografia por Raios X , Escherichia coli/genética , Escherichia coli/metabolismo , Éteres Cíclicos/síntese química , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Expressão Gênica , Vetores Genéticos/química , Vetores Genéticos/metabolismo , Simulação de Acoplamento Molecular , Penicillium/química , Fenalenos/metabolismo , Fenóis/química , Ligação Proteica , Conformação Proteica em alfa-Hélice , Conformação Proteica em Folha beta , Domínios e Motivos de Interação entre Proteínas , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Estereoisomerismo , Especificidade por Substrato
17.
Org Biomol Chem ; 17(8): 2070-2076, 2019 02 20.
Artigo em Inglês | MEDLINE | ID: mdl-30628619

RESUMO

LepI is a novel multifunctional enzyme that catalyzes stereoselective dehydration, Diels-Alder reaction, and retro-Claisen rearrangement. Here we report the crystal structure of LepI in complex with its co-factor S-adenosyl methionine (SAM). LepI forms a tetramer via the N-terminal helical domain and binds to a SAM molecule in the C-terminal catalytic domain. The binding modes of various LepI substrates are investigated by docking simulations, which suggest that the substrates are bound via both hydrophobic and hydrophilic forces, as well as cation-π interactions with the positively charged SAM. The reaction starts with a dehydration step in which H133 possibly deprotonates the pyridone hydroxyl group of the substrate, while D296 might protonate an alkyl-chain hydroxyl group. Subsequent pericyclization may be facilitated by the correct fold of the substrate's alkyl chain and a thermodynamic driving force towards σ-bonds at the expense of π-bonds. These results provide structural insights into LepI catalysis and are important in understanding the mechanism of enzymatic pericyclization.


Assuntos
Aspergillus nidulans/enzimologia , Benzopiranos/metabolismo , Proteínas Fúngicas/metabolismo , Piridonas/metabolismo , S-Adenosilmetionina/metabolismo , Sequência de Aminoácidos , Aspergillus nidulans/química , Aspergillus nidulans/metabolismo , Vias Biossintéticas , Domínio Catalítico , Cristalografia por Raios X , Reação de Cicloadição , Proteínas Fúngicas/química , Simulação de Acoplamento Molecular , Conformação Proteica , Multimerização Proteica , Estereoisomerismo
18.
Biochem Biophys Res Commun ; 495(2): 1782-1788, 2018 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-29229390

RESUMO

FamD1 is a novel CloQ/NphB-family indole prenyltransferase which involves in hapalindole-type alkaloid biosynthesis. Here the native FamD1 structure and three protein-ligand complexes are analyzed to investigate the molecular basis of substrate binding and catalysis. FamD1 adopts a typical ABBA architecture of aromatic prenyltransferase, in which the substrate-binding chamber is found in the central ß-barrel. The indole-containing acceptor substrate is bound adjacent to the prenyl donor. Based on the complex structures, a catalytic mechanism of FamD1 is proposed. Functional implications on the sister enzyme FamD2 are also discussed.


Assuntos
Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Dimetilaliltranstransferase/química , Dimetilaliltranstransferase/metabolismo , Alcaloides Indólicos/metabolismo , Proteínas de Bactérias/genética , Domínio Catalítico , Cristalografia por Raios X , Cianobactérias/enzimologia , Cianobactérias/genética , Dimetilaliltranstransferase/genética , Alcaloides Indólicos/química , Modelos Moleculares , Conformação Proteica , Dobramento de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
19.
Angew Chem Int Ed Engl ; 57(46): 15060-15064, 2018 11 12.
Artigo em Inglês | MEDLINE | ID: mdl-30222239

RESUMO

Found recently in stignomatales, the Stig cyclases catalyze the Cope rearrangement and intramolecular cyclization to produce complex indole alkaloids. Five crystal structures were solved of subfamily 1 and 2 Stig cyclases, which adopt a ß-sandwich fold like the non-catalytic carbohydrate-binding motif. Several complex structures were also determined of indole-based compounds, which are bound to the hydrophobic terminal cavity, where a conserved Asp residue makes an H-bond to the indole N and triggers the acid-catalyzed Cope rearrangement. Through analyzing the enzyme-ligand interactions and mutagenesis experiments, several aromatic residues were found important in catalysis. Apart from a common substrate binding mode and catalytic mechanism, potential subfamily variations that may attribute to the different product specificity are implicated. These results shall expand our scope of enzymology, in particular for further investigation of the biosynthetic Cope rearrangement.


Assuntos
Proteínas de Bactérias/química , Cianobactérias/enzimologia , Proteínas de Bactérias/metabolismo , Sítios de Ligação , Cristalografia por Raios X , Cianobactérias/química , Cianobactérias/metabolismo , Ciclização , Alcaloides Indólicos/química , Alcaloides Indólicos/metabolismo , Modelos Moleculares , Conformação Proteica , Especificidade por Substrato
20.
Angew Chem Int Ed Engl ; 57(3): 683-687, 2018 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-29215779

RESUMO

We report the first X-ray crystallographic structure of the "head-to-middle" prenyltransferase, isosesquilavandulyl diphosphate synthase, involved in biosynthesis of the merochlorin class of antibiotics. The protein adopts the ζ or cis-prenyl transferase fold but remarkably, unlike tuberculosinol adenosine synthase and other cis-prenyl transferases (e.g. cis-farnesyl, decaprenyl, undecaprenyl diphosphate synthases), the large, hydrophobic side chain does not occupy a central hydrophobic tunnel. Instead, it occupies a surface pocket oriented at 90° to the hydrophobic tunnel. Product chain-length control is achieved by squeezing out the ligand from the conventional allylic S1 binding site, with proton abstraction being achieved using a diphosphate-Asn-Ser relay. The structures revise and unify our thinking as to the mechanism of action of many other prenyl transferases and may also be of use in engineering new merochlorin-class antibiotics.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA