Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
Add more filters










Publication year range
1.
Appl Environ Microbiol ; 90(5): e0011824, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38568076

ABSTRACT

Bacteria have two routes for the l-methionine biosynthesis. In one route called the direct sulfuration pathway, acetylated l-homoserine is directly converted into l-homocysteine. The reaction using H2S as the second substrate is catalyzed by a pyridoxal 5'-phosphate-dependent enzyme, O-acetylhomoserine sulfhydrylase (OAHS). In the present study, we determined the enzymatic functions and the structures of OAHS from Lactobacillus plantarum (LpOAHS). The LpOAHS enzyme exhibited the highest catalytic activity under the weak acidic pH condition. In addition, crystallographic analysis revealed that the enzyme takes two distinct structures, open and closed forms. In the closed form, two acidic residues are sterically clustered. The proximity may cause the electrostatic repulsion, inhibiting the formation of the closed form under the neutral to the basic pH conditions. We concluded that the pH-dependent regulation mechanism using the two acidic residues contributes to the acidophilic feature of the enzyme. IMPORTANCE: In the present study, we can elucidate the pH-dependent regulation mechanism of the acidophilic OAHS. The acidophilic feature of the enzyme is caused by the introduction of an acidic residue to the neighborhood of the key acidic residue acting as a switch for the structural interconversion. The strategy may be useful in the field of protein engineering to change the optimal pH of the enzymes. In addition, this study may be useful for the development of antibacterial drugs because the l-methionine synthesis essential for bacteria is inhibited by the OAHS inhibitors. The compounds that can inhibit the interconversion between the open and closed forms of OAHS may become antibacterial drugs.


Subject(s)
Bacterial Proteins , Lactobacillus plantarum , Lactobacillus plantarum/enzymology , Lactobacillus plantarum/genetics , Lactobacillus plantarum/metabolism , Hydrogen-Ion Concentration , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/chemistry , Carbon-Oxygen Lyases
2.
Protein Sci ; 31(6): e4338, 2022 06.
Article in English | MEDLINE | ID: mdl-35634777

ABSTRACT

DcsB, an enzyme produced from the d-cycloserine biosynthetic gene cluster, displays moderate similarity to arginase in the sequence and three-dimensional structure. Arginase is a ubiquitous enzyme hydrolyzing l-arginine to generate l-ornithine and urea, whereas DcsB hydrolyzes Nω -hydroxy-l-arginine (l-NOHA), an arginase inhibitor, to generate l-ornithine and hydroxyurea. We determined the crystal structure of DcsB associated with l-ornithine and that with the tetrahedral derivative of 2(S)-amino-6-boronohexanoic acid, whose boron atom forms a covalent bond with an oxygen atom bridging two manganese ions at the active center. The substrate-binding pocket of DcsB is narrower than that of arginase, suggesting that DcsB is unsuitable for the binding of l-NOHA in an inhibitory manner. The transition state-like structure demonstrated that Asp210 and Glu241 have a role to trap a positively charged ion near the dimanganese cluster. Kinetic analysis using the mutated DcsB showed that the enzyme employs different catalytic mechanisms under the neutral and alkaline pH conditions. Glu241 in DcsB is likely involved in the recognition of the hydroxyguanidino group of l-NOHA, whereas Asp210, in cooperation with Glu241, seems to contribute to the reactivity toward the protonated l-NOHA, which is a preferable species under the neutral pH conditions. After entering of the protonated l-NOHA to the substrate-binding pocket of DcsB, a hydronium ion may be trapped at the positive ion-binding site. Then, the ion serves as a specific acid catalyst to facilitate the collapse of the tetrahedral intermediate of l-NOHA.


Subject(s)
Arginase , Arginine , Amino Acids , Arginase/chemistry , Arginase/genetics , Arginine/metabolism , Catalysis , Kinetics , Ornithine
3.
Proteins ; 90(4): 912-918, 2022 04.
Article in English | MEDLINE | ID: mdl-34877716

ABSTRACT

The O-ureidoserine racemase (DcsC) is an enzyme found from the biosynthetic gene cluster of antitubercular agent d-cycloserine. Although DcsC is homologous to diaminopimelate epimerase (DapF) that catalyzes the interconversion between ll- and dl-diaminopimelic acid, it specifically catalyzes the interconversion between O-ureido-l-serine and its enantiomer. Here we determined the crystal structure of DcsC at a resolution of 2.12 Å, implicating that the catalytic mechanism of DcsC shares similarity with that of DapF. Comparing the structure of the active center of DcsC to that of DapF, Thr72, Thr198, and Tyr219 of DcsC are likely to be involved in the substrate specificity.


Subject(s)
Cycloserine , Racemases and Epimerases , Biosynthetic Pathways , Crystallography, X-Ray , Cycloserine/chemistry , Cycloserine/metabolism , Multigene Family , Racemases and Epimerases/genetics , Racemases and Epimerases/metabolism , Serine/metabolism
4.
J Virol ; 95(19): e0081521, 2021 09 09.
Article in English | MEDLINE | ID: mdl-34287046

ABSTRACT

Sendai virus (SeV), belonging to the Respirovirus genus of the family Paramyxoviridae, harbors an accessory protein, named C protein, which facilitates viral pathogenicity in mice. In addition, the C protein is known to stimulate the budding of virus-like particles by binding to the host ALG-2 interacting protein X (Alix), a component of the endosomal sorting complexes required for transport (ESCRT) machinery. However, small interfering RNA (siRNA)-mediated gene knockdown studies suggested that neither Alix nor C protein is related to SeV budding. In the present study, we determined the crystal structure of a complex comprising the C-terminal half of the C protein (Y3) and the Bro1 domain of Alix at a resolution of 2.2 Å to investigate the role of the complex in SeV budding. The structure revealed that a novel consensus sequence, LXXW, which is conserved among Respirovirus C proteins, is important for Alix binding. SeV possessing a mutated C protein with reduced Alix-binding affinity showed impaired virus production, which correlated with the binding affinity. Infectivity analysis showed a 160-fold reduction at 12 h postinfection compared with nonmutated virus, while C protein competes with CHMP4, one subunit of the ESCRT-III complex, for binding to Alix. All together, these results highlight the critical role of C protein in SeV budding. IMPORTANCE Human parainfluenza virus type I (hPIV1) is a respiratory pathogen affecting young children, immunocompromised patients, and the elderly, with no available vaccines or antiviral drugs. Sendai virus (SeV), a murine counterpart of hPIV1, has been studied extensively to determine the molecular and biological properties of hPIV1. These viruses possess a multifunctional accessory protein, C protein, which is essential for stimulating viral reproduction, but its role in budding remains controversial. In the present study, the crystal structure of the C-terminal half of the SeV C protein associated with the Bro1 domain of Alix, a component of cell membrane modulating machinery ESCRT, was elucidated. Based on the structure, we designed mutant C proteins with different binding affinities to Alix and showed that the interaction between C and Alix is vital for viral budding. These findings provide new insights into the development of new antiviral drugs against hPIV1.


Subject(s)
Calcium-Binding Proteins/chemistry , Calcium-Binding Proteins/metabolism , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/metabolism , Endosomal Sorting Complexes Required for Transport/chemistry , Endosomal Sorting Complexes Required for Transport/metabolism , Sendai virus/physiology , Viral Proteins/chemistry , Viral Proteins/metabolism , Virus Release , Amino Acid Sequence , Animals , Binding, Competitive , Cell Line , Crystallography, X-Ray , Humans , Interferon-alpha/genetics , Interferon-alpha/metabolism , Interferon-beta/genetics , Interferon-beta/metabolism , Models, Molecular , Protein Binding , Protein Conformation , Protein Domains , Sendai virus/chemistry , Sendai virus/genetics , Sendai virus/metabolism , Signal Transduction , Virion/physiology
5.
Int J Biol Macromol ; 183: 1861-1870, 2021 Jul 31.
Article in English | MEDLINE | ID: mdl-34089758

ABSTRACT

Tyrosinase (Ty) and catechol oxidase (CO) are members of type-3 copper enzymes. While Ty catalyzes both phenolase and catecholase reactions, CO catalyzes only the latter reaction. In the present study, Ty was found to catalyze the catecholase reaction, but hardly the phenolase reaction in the presence of the metallochaperon called "caddie protein (Cad)". The ability of the substrates to dissociate the motif shielding the active-site pocket seems to contribute critically to the substrate specificity of Ty. In addition, a mutation at the N191 residue, which forms a hydrogen bond with a water molecule near the active center, decreased the inherent ratio of phenolase versus catecholase activity. Unlike the wild-type complex, reaction intermediates were not observed when the catalytic reaction toward the Y98 residue of Cad was progressed in the crystalline state. The increased basicity of the water molecule may be necessary to inhibit the proton transfer from the conjugate acid to a hydroxide ion bridging the two copper ions. The deprotonation of the substrate hydroxyl by the bridging hydroxide seems to be significant for the efficient catalytic cycle of the phenolase reaction.


Subject(s)
Catechol Oxidase/chemistry , Catechol Oxidase/metabolism , Monophenol Monooxygenase/chemistry , Monophenol Monooxygenase/metabolism , Streptomyces/enzymology , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Binding Sites , Catalysis , Catalytic Domain , Catechol Oxidase/genetics , Crystallography, X-Ray , Hydrogen Bonding , Metallochaperones/metabolism , Models, Molecular , Monophenol Monooxygenase/genetics , Mutation , Protein Binding , Protein Conformation , Streptomyces/genetics , Substrate Specificity , Water/chemistry
6.
J Biol Chem ; 296: 100698, 2021.
Article in English | MEDLINE | ID: mdl-33895142

ABSTRACT

Homologues of the Oscillatoria agardhii agglutinin (OAA) lectins contain a sequence repeat of ∼66 amino acids, with the number of tandem repeats varying across family members. OAA homologues bind high-mannose glycans on viral surface proteins, thereby interfering with viral entry into host cells. As such, OAA homologues have potential utility as antiviral agents, but a more detailed understanding of their structure-function relationships would enable us to develop improved constructs. Here, we determined the X-ray crystal structure of free and glycan-bound forms of Pseudomonas taiwanensis lectin (PTL), an OAA-family lectin consisting of two tandem repeats. Like other OAA-family lectins, PTL exhibited a ß-barrel-like structure with two symmetrically positioned glycan-binding sites at the opposite ends of the barrel. Upon glycan binding, the conformation of PTL undergoes a more significant change than expected from previous OAA structural analysis. Moreover, the electron density of the bound glycans suggested that the binding affinities are different at the two binding sites. Next, based on analysis of these structures, we used site-specific mutagenesis to create PTL constructs expected to increase the population with a conformation suitable for glycan binding. The engineered PTLs were examined for their antiviral activity against the influenza virus. Interestingly, some exhibited stronger activity compared with that of the parent PTL. We propose that our approach is effective for the generation of potential microbicides with enhanced antiviral activity.


Subject(s)
Antiviral Agents/metabolism , Antiviral Agents/pharmacology , Lectins/metabolism , Lectins/pharmacology , Polysaccharides/metabolism , Protein Engineering , Antiviral Agents/chemistry , Crystallography, X-Ray , Lectins/chemistry , Lectins/genetics , Models, Molecular , Orthomyxoviridae/drug effects , Protein Binding , Protein Conformation, beta-Strand
7.
Sci Rep ; 10(1): 14886, 2020 09 10.
Article in English | MEDLINE | ID: mdl-32913258

ABSTRACT

The reverse transsulfuration pathway, which is composed of cystathionine ß-synthase (CBS) and cystathionine γ-lyase (CGL), plays a role to synthesize L-cysteine using L-serine and the sulfur atom in L-methionine. A plant-derived lactic acid bacterium Lactobacillus plantarum SN35N has been previously found to harbor the gene cluster encoding the CBS- and CGL-like enzymes. In addition, it has been demonstrated that the L. plantarum CBS can synthesize cystathionine from O-acetyl-L-serine and L-homocysteine. The aim of this study is to characterize the enzymatic functions of the L. plantarum CGL. We have found that the enzyme has the high γ-lyase activity toward cystathionine to generate L-cysteine, together with the ß-lyase activity toward L-cystine to generate L-cysteine persulfide. By the crystallographic analysis of the inactive CGL K194A mutant complexed with cystathionine, we have found the residues which recognize the distal amino and carboxyl groups of cystathionine or L-cystine. The PLP-bound substrates at the active site may take either the binding pose for the γ- or ß-elimination reaction, with the former being the major reaction in the case of cystathionine.


Subject(s)
Cystathionine gamma-Lyase/metabolism , Lactobacillus plantarum/enzymology , Catalysis , Crystallography, X-Ray , Cystathionine/metabolism , Cystathionine gamma-Lyase/chemistry , Homocysteine/metabolism , Serine/analogs & derivatives , Serine/metabolism , Substrate Specificity
8.
Acta Crystallogr D Struct Biol ; 76(Pt 6): 506-514, 2020 Jun 01.
Article in English | MEDLINE | ID: mdl-32496212

ABSTRACT

DcsB, one of the enzymes encoded in the D-cycloserine (D-CS) biosynthetic gene cluster, displays a high sequence homology to arginase, which contains two manganese ions in the active site. However, DcsB hydrolyzes Nω-hydroxy-L-arginine, but not L-arginine, to supply hydroxyurea for the biosynthesis of D-CS. Here, the crystal structure of DcsB was determined at a resolution of 1.5 Šusing anomalous scattering from the manganese ions. In the crystal structure, DscB generates an artificial dimer created by the open and closed forms. Gel-filtration analysis demonstrated that DcsB is a monomeric protein, unlike arginase, which forms a trimeric structure. The active center containing the binuclear manganese cluster differs between DcsB and arginase. In DcsB, one of the ligands of the MnA ion is a cysteine, while the corresponding residue in arginase is a histidine. In addition, DcsB has no counterpart to the histidine residue that acts as a general acid/base during the catalytic reaction of arginase. The present study demonstrates that DcsB has a unique active site that differs from that of arginase.


Subject(s)
Amidohydrolases/chemistry , Arginine/analogs & derivatives , Bacterial Proteins/chemistry , Cycloserine/biosynthesis , Streptomyces/enzymology , Amidohydrolases/genetics , Arginine/metabolism , Bacterial Proteins/genetics , Protein Structure, Tertiary , Streptomyces/genetics , Substrate Specificity
9.
FEBS J ; 287(13): 2763-2778, 2020 07.
Article in English | MEDLINE | ID: mdl-31793174

ABSTRACT

In the biosynthetic pathway of an antitubercular antibiotic d-cycloserine (d-CS), O-ureido-d-serine (d-OUS) is converted to d-CS. We have previously demonstrated that DcsG, classified into the ATP-grasp superfamily enzyme, catalyzes the ring formation to generate d-CS, which is accompanied by the cleavage of a bond in the urea moiety of d-OUS to remove a carbamoyl group. Although the general ATP-grasp enzymes catalyze an ATP-dependent ligation reaction between two substrates, DcsG catalyzes specifically the generation of an intramolecular covalent bond. In the present study, cyanate was found in the reaction mixture, suggesting that carbamoyl group is eliminated as an isocyanic acid during the reaction. By the crystallographic and mutational investigations of DcsG, we anticipate the residues necessary for the binding of d-OUS. An acylphosphate intermediate must be bound at the narrow pocket of DcsG in a folded conformation, inducing the bond cleavage and the new bond formation to generate cyanate and d-CS, respectively. DATABASE: Structural data are available in Protein Data Bank database under the accession number 6JIL.


Subject(s)
Adenosine Triphosphate/metabolism , Biosynthetic Pathways , Cycloserine/biosynthesis , Ligases/metabolism , Adenosine Triphosphate/chemistry , Binding Sites , Biocatalysis , Crystallography, X-Ray , Cyclization , Ligases/chemistry , Ligases/genetics , Models, Molecular , Mutation , Protein Binding , Protein Conformation
10.
Sci Rep ; 9(1): 4854, 2019 03 19.
Article in English | MEDLINE | ID: mdl-30890721

ABSTRACT

We investigated the role of the resistance-nodulation-cell division superfamily (RND) efflux system on intrinsic multidrug resistance in Serratia marcescens. We identified eight putative RND efflux system genes in the S. marcescens Db10 genome that included the previously characterized systems, sdeXY, sdeAB, and sdeCDE. Six out of the eight genes conferred multidrug resistance on KAM32, a drug hypersensitive strain of Escherichia coli. Five out of the eight genes conferred resistance to benzalkonium, suggesting the importance of RND efflux systems in biocide resistance in S. marcescens. The energy-dependent efflux activities of five of the pumps were examined using a rhodamine 6 G efflux assay. When expressed in the tolC-deficient strain of E. coli, KAM43, none of the genes conferred resistance on E. coli. When hasF, encoding the S. marcescens TolC ortholog, was expressed in KAM43, all of the genes conferred resistance on E. coli, suggesting that HasF is a major outer membrane protein that is used by all RND efflux systems in this organism. We constructed a sdeXY deletion mutant from a derivative strain of the clinically isolated multidrug-resistant S. marcescens strain and found that the sdeXY deletion mutant was sensitive to a broad spectrum of antimicrobial agents.


Subject(s)
Drug Resistance, Multiple, Bacterial/genetics , Escherichia coli/genetics , Serratia Infections/genetics , Serratia marcescens/drug effects , Anti-Bacterial Agents/pharmacology , Bacterial Outer Membrane Proteins/genetics , Benzalkonium Compounds/pharmacology , Cell Division/drug effects , Escherichia coli/drug effects , Genome, Bacterial/drug effects , Genome, Bacterial/genetics , Humans , Microbial Sensitivity Tests , Multidrug Resistance-Associated Proteins , Serratia Infections/drug therapy , Serratia Infections/microbiology , Serratia marcescens/genetics , Serratia marcescens/pathogenicity
11.
Food Chem ; 260: 166-173, 2018 Sep 15.
Article in English | MEDLINE | ID: mdl-29699658

ABSTRACT

Phenoloxidases (POs) play a crucial role in melanization of crustaceans. There are at least two types of POs characterized in crustaceans: the conventional type (POα here) that is expressed in hemocytes and POß, a secreted protein synthesized in the hepatopancreas. We investigated the source of PO activity in the hemolymph of a lobster and determined the kinetic parameters of mono- and di-PO activities. In the lobster hemolymph, POα, which formed a hexamer similar to both POß and hemocyanin, contributed to PO activity, whereas the amount of POß was low. Kinetic analyses using purified prophenoloxidase of crustaceans showed that lobster POα has a higher rate constant, while shrimp POß has higher specificity in both mono- and di-PO reactions, when tyramine and dopamine were employed as substrates. There should be at least two types of PO molecules in crustacean hemolymph, but the dominant PO molecule type varies among species.


Subject(s)
Hemolymph/enzymology , Monophenol Monooxygenase/metabolism , Palinuridae/enzymology , Animals , Catechol Oxidase , Dopamine/metabolism , Enzyme Precursors , Hemocyanins/metabolism , Hemocytes/enzymology , Kinetics , Penaeidae/enzymology , Species Specificity , Substrate Specificity , Tyramine/metabolism
12.
Appl Environ Microbiol ; 84(7)2018 04 01.
Article in English | MEDLINE | ID: mdl-29352085

ABSTRACT

We have previously shown that the lactic acid bacterium Lactobacillus brevis 174A, isolated from Citrus iyo fruit, produces a bacteriocin designated brevicin 174A, which is comprised of two antibacterial polypeptides (designated brevicins 174A-ß and 174A-γ). We have also found a gene cluster, composed of eight open reading frames (ORFs), that contains genes for the biosynthesis of brevicin 174A, self-resistance to its own bacteriocin, and two transcriptional regulatory proteins. Some lactic acid bacterial strains have a system to start the production of bacteriocin at an adequate stage of growth. Generally, the system consists of a membrane-bound histidine protein kinase (HPK) that senses a specific environmental stimulus and a corresponding response regulator (RR) that mediates the cellular response. We have previously shown that although the HPK- and RR-encoding genes are not found on the brevicin 174A biosynthetic gene cluster in the 174A strain, two putative regulatory genes, designated breD and breG, are in the gene cluster. In the present study, we demonstrate that the expression of brevicin 174A production and self-resistance is positively controlled by two transcriptional regulatory proteins, designated BreD and BreG. BreD is expressed together with BreE as the self-resistance determinant of L. brevis 174A. DNase I footprinting analysis and a promoter assay demonstrated that BreD binds to the breED promoter as a positive autoregulator. The present study also demonstrates that BreG, carrying a transmembrane domain, binds to the common promoter of breB and breC, encoding brevicins 174A-ß and 174A-γ, respectively, for positive regulation.IMPORTANCE The problem of the appearance of bacteria that are resistant to practical antibiotics and the increasing demand for safe foods have increased interest in replacing conventional antibiotics with bacteriocin produced by the lactic acid bacteria. This antibacterial substance can inhibit the growth of pathogenic bacteria without side effects on the human body. The bacteriocin that is produced by a Citrus iyo-derived Lactobacillus brevis strain inhibits the growth of pathogenic bacteria such as Listeria monocytogenes, Staphylococcus aureus, and Streptococcus mutans In general, lactic acid bacterial strains have a system to start the production of bacteriocin at an adequate stage of growth, which is called a quorum-sensing system. The system consists of a membrane-bound histidine protein kinase that senses a specific environmental stimulus and a corresponding response regulator that mediates the cellular response. The present study demonstrates that the expression of the genes encoding bacteriocin biosynthesis and the self-resistance determinant is positively controlled by two transcriptional regulatory proteins.


Subject(s)
Bacterial Proteins/genetics , Gene Expression Regulation, Bacterial , Genes, Bacterial/genetics , Levilactobacillus brevis/genetics , Bacterial Proteins/metabolism , Bacteriocins/genetics , Bacteriocins/metabolism , Levilactobacillus brevis/metabolism
13.
PLoS Biol ; 16(12): e3000077, 2018 12.
Article in English | MEDLINE | ID: mdl-30596633

ABSTRACT

Tyrosinase (EC 1.14.18.1), a copper-containing monooxygenase, catalyzes the conversion of phenol to the corresponding ortho-quinone. The Streptomyces tyrosinase is generated as a complex with a "caddie" protein that facilitates the transport of two copper ions into the active center. In our previous study, the Tyr98 residue in the caddie protein, which is accommodated in the pocket of active center of tyrosinase, has been found to be converted to a reactive quinone through the formations of the µ-η2:η2-peroxo-dicopper(II) and Cu(II)-dopasemiquinone intermediates. Until now-despite extensive studies for the tyrosinase reaction based on the crystallographic analysis, low-molecular-weight models, and computer simulations-the catalytic mechanism has been unable to be made clear at an atomic level. To make the catalytic mechanism of tyrosinase clear, in the present study, the cryo-trapped crystal structures were determined at very high resolutions (1.16-1.70 Å). The structures suggest the existence of an important step for the tyrosinase reaction that has not yet been found: that is, the hydroxylation reaction is triggered by the movement of CuA, which induces the syn-to-anti rearrangement of the copper ligands after the formation of µ-η2:η2-peroxo-dicopper(II) core. By the rearrangement, the hydroxyl group of the substrate is placed in an equatorial position, allowing the electrophilic attack to the aromatic ring by the Cu2O2 oxidant.


Subject(s)
Copper/metabolism , Monophenol Monooxygenase/physiology , Monophenol Monooxygenase/ultrastructure , Benzoquinones/metabolism , Binding Sites/physiology , Catalysis , Crystallography, X-Ray/methods , Hydroxylation , Ligands , Models, Molecular , Monophenol Monooxygenase/metabolism , Phenols/chemistry , Streptomyces/genetics , Streptomyces/metabolism , Tyrosine/metabolism
14.
J Biol Chem ; 292(48): 19752-19766, 2017 12 01.
Article in English | MEDLINE | ID: mdl-28978648

ABSTRACT

Sendai virus (SeV), which causes respiratory diseases in rodents, possesses the C protein that blocks the signal transduction of interferon (IFN), thereby escaping from host innate immunity. We previously demonstrated by using protein crystallography that two molecules of Y3 (the C-terminal half of the C protein) can bind to the homodimer of the N-terminal domain of STAT1 (STAT1ND), elucidating the mechanism of inhibition of IFN-γ signal transduction. SeV C protein also blocks the signal transduction of IFN-α/ß by inhibiting the phosphorylation of STAT1 and STAT2, although the mechanism for the inhibition is unclear. Therefore, we sought to elucidate the mechanism of inhibition of the IFN signal transduction via STAT1 and STAT2. Small angle X-ray scattering analysis indicated that STAT1ND associates with the N-terminal domain of STAT2 (STAT2ND) with the help of a Gly-rich linker. We generated a linker-less recombinant protein possessing a STAT1ND:STAT2ND heterodimeric structure via an artificial disulfide bond. Analytical size-exclusion chromatography and surface plasmon resonance revealed that one molecule of Y3 can associate with a linker-less recombinant protein. We propose that one molecule of C protein associates with the STAT1:STAT2 heterodimer, inducing a conformational change to an antiparallel form, which is easily dephosphorylated. This suggests that association of C protein with the STAT1ND:STAT2ND heterodimer is an important factor to block the IFN-α/ß signal transduction.


Subject(s)
Interferon Type I/metabolism , STAT1 Transcription Factor/metabolism , STAT2 Transcription Factor/metabolism , Sendai virus/metabolism , Signal Transduction , Viral Proteins/metabolism , Cell Line , Crystallography, X-Ray , Dimerization , Humans , Phosphorylation , Protein Conformation , STAT1 Transcription Factor/chemistry , STAT2 Transcription Factor/chemistry
15.
Biochemistry ; 56(41): 5593-5603, 2017 10 17.
Article in English | MEDLINE | ID: mdl-28902505

ABSTRACT

Tyrosinase (EC 1.14.18.1), which possesses two copper ions at the active center, catalyzes a rate-limiting reaction of melanogenesis, that is, the conversion of a phenol to the corresponding ortho-quinone. The enzyme from the genus Streptomyces is generated as a complex with a "caddie" protein that assists the transport of two copper ions into the active center. In this complex, the Tyr98 residue in the caddie protein was found to be accommodated in the pocket of the active center of tyrosinase, probably in a manner similar to that of l-tyrosine as a genuine substrate of tyrosinase. Under physiological conditions, the addition of the copper ion to the complex releases tyrosinase from the complex, in accordance with the aggregation of the caddie protein. The release of the copper-bound tyrosinase was found to be accelerated by adding reducing agents under aerobic conditions. Mass spectroscopic analysis indicated that the Tyr98 residue was converted to a reactive quinone, and resonance Raman spectroscopic analysis indicated that the conversion occurred through the formations of µ-η2:η2-peroxo-dicopper(II) and Cu(II)-semiquinone. Electron paramagnetic resonance analysis under anaerobic conditions and Fourier transform infrared spectroscopic analysis using CO as a structural probe under anaerobic conditions indicated that the copper transportation process to the active center is a reversible event in the tyrosinase/caddie complex. Aggregation of the caddie protein, which is triggered by the conversion of the Tyr98 residue to dopaquinone, may ensure the generation of fully activated tyrosinase.


Subject(s)
Bacterial Proteins/metabolism , Carrier Proteins/metabolism , Copper/metabolism , Models, Molecular , Monophenol Monooxygenase/metabolism , Streptomyces/enzymology , Amino Acid Substitution , Apoenzymes/chemistry , Apoenzymes/genetics , Apoenzymes/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Benzoquinones/chemistry , Benzoquinones/metabolism , Binding Sites , Carrier Proteins/chemistry , Carrier Proteins/genetics , Catalytic Domain , Copper/chemistry , Dihydroxyphenylalanine/analogs & derivatives , Dihydroxyphenylalanine/chemistry , Dihydroxyphenylalanine/metabolism , Enzyme Activation/drug effects , Monophenol Monooxygenase/chemistry , Monophenol Monooxygenase/genetics , Mutation , Oxidation-Reduction , Protein Aggregates/drug effects , Protein Multimerization/drug effects , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Reducing Agents/chemistry , Solubility , Tyrosine/chemistry , Tyrosine/metabolism
16.
Protein Sci ; 26(4): 763-783, 2017 04.
Article in English | MEDLINE | ID: mdl-28127810

ABSTRACT

Cystathionine ß-synthase (CBS) catalyzes the formation of l-cystathionine from l-serine and l-homocysteine. The resulting l-cystathionine is decomposed into l-cysteine, ammonia, and α-ketobutylic acid by cystathionine γ-lyase (CGL). This reverse transsulfuration pathway, which is catalyzed by both enzymes, mainly occurs in eukaryotic cells. The eukaryotic CBS and CGL have recently been recognized as major physiological enzymes for the generation of hydrogen sulfide (H2 S). In some bacteria, including the plant-derived lactic acid bacterium Lactobacillus plantarum, the CBS- and CGL-encoding genes form a cluster in their genomes. Inactivation of these enzymes has been reported to suppress H2 S production in bacteria; interestingly, it has been shown that H2 S suppression increases their susceptibility to various antibiotics. In the present study, we characterized the enzymatic properties of the L. plantarum CBS, whose amino acid sequence displays a similarity with those of O-acetyl-l-serine sulfhydrylase (OASS) that catalyzes the generation of l-cysteine from O-acetyl-l-serine (l-OAS) and H2 S. The L. plantarum CBS shows l-OAS- and l-cysteine-dependent CBS activities together with OASS activity. Especially, it catalyzes the formation of H2 S in the presence of l-cysteine and l-homocysteine, together with the formation of l-cystathionine. The high affinity toward l-cysteine as a first substrate and tendency to use l-homocysteine as a second substrate might be associated with its enzymatic ability to generate H2 S. Crystallographic and mutational analyses of CBS indicate that the Ala70 and Glu223 residues at the substrate binding pocket are important for the H2 S-generating activity.


Subject(s)
Bacterial Proteins , Cystathionine beta-Synthase , Lactobacillus plantarum , Multigene Family , Mutation, Missense , Amino Acid Substitution , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Binding Sites , Crystallography, X-Ray , Cystathionine/metabolism , Cystathionine beta-Synthase/chemistry , Cystathionine beta-Synthase/genetics , Cystathionine beta-Synthase/metabolism , Hydrogen Sulfide/chemistry , Hydrogen Sulfide/metabolism , Hydrolases/genetics , Hydrolases/metabolism , Lactobacillus plantarum/enzymology , Lactobacillus plantarum/genetics
17.
Biochem Biophys Rep ; 5: 266-271, 2016 Mar.
Article in English | MEDLINE | ID: mdl-28955833

ABSTRACT

We have previously isolated a lactic acid bacterium (LAB), Pediococcus pentosaceus LP28, from the longan fruit Euphoria longana. Since the plant-derived LAB strain produces an extracellular polysaccharide (EPS), in this study, we analyzed the chemical structure and the biosynthesizing genes for the EPS. The EPS, which was purified from the LP28 culture broth, was classified into acidic and neutral EPSs with a molecular mass of about 50 kDa and 40 kDa, respectively. The acidic EPS consisted of glucose, galactose, mannose, and N-acetylglucosamine moieties. Interestingly, since pyruvate residue was detected in the hydrolyzed acidic EPS, one of the four sugars may be modified with pyruvate. On the other hand, the neutral EPS consisted of glucose, mannose, and N-acetylglucosamine; pyruvate was scarcely detected in the polysaccharide molecule. As a first step to deduce the probiotic function of the EPS together with the biosynthesis, we determined the whole genome sequence of the LP28 strain, demonstrating that the genome is a circular DNA, which is composed of 1,774,865 bp (1683 ORFs) with a GC content of 37.1%. We also found that the LP28 strain harbors a plasmid carrying 6 ORFs composed of 5366 bp with a GC content of 36.5%. By comparing all of the genome sequences among the LP28 strain and four strains of P. pentosaceus reported previously, we found that 53 proteins in the LP28 strain display a similarity of less than 50% with those in the four P. pentosaceus strains. Significantly, 4 of the 53 proteins, which may be enzymes necessary for the EPS production on the LP28 strain, were absent in the other four P. pentosaceus strains and displayed less than 50% similarity with other LAB species. The EPS-biosynthetic gene cluster detected only in the LP28 genome consisted of 12 ORFs containing a priming enzyme, five glycosyltransferases, and a putative polysaccharide pyruvyltransferase.

18.
Biol Pharm Bull ; 38(12): 1902-9, 2015.
Article in English | MEDLINE | ID: mdl-26632181

ABSTRACT

In the present study, we isolated a lactic acid bacterium (LAB) from a citrus iyo fruit and identified it as Lactobacillus brevis. This plant-derived LAB strain, designated 174A, produces bacteriocin consisting of two polypeptides designated brevicin 174A-ß and 174A-γ. Although each polypeptide itself displays antibacterial activity, the ability is enhanced 100 fold by mixing both polypeptides at a 1 : 1 ratio. Significantly, brevicin 174A inhibits even the growth of several pathogenic bacteria that are more resistant to a lantibiotic bacteriocin, nisin A, which is commonly utilized as a preservative added to foodstuffs. Structural analysis of the 174A bacteriocin using a program that predicts secondary structure suggests that both component polypeptides have a positively charged N-terminal region, as well as two cysteine residues in both the N- and C-terminals. Judging from a mutational analysis of the antibacterial polypeptides, these unique amino acid sequences of 174A-ß might be important for the expression of the synergistic activity that occurs in the presence of the two polypeptides combined.


Subject(s)
Anti-Bacterial Agents/analysis , Bacteriocins/analysis , Citrus/microbiology , Fruit/microbiology , Levilactobacillus brevis/metabolism , Peptides/analysis , Amino Acid Sequence , Anti-Bacterial Agents/pharmacology , Bacteriocins/pharmacology , DNA Mutational Analysis , Drug Synergism , Genes, Bacterial , Lactic Acid , Levilactobacillus brevis/genetics , Molecular Sequence Data , Mutation , Peptides/pharmacology
20.
J Virol ; 89(22): 11487-99, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26339056

ABSTRACT

UNLABELLED: Sendai virus (SeV) C protein inhibits the signal transduction pathways of interferon alpha/beta (IFN-α/ß) and IFN-γ by binding to the N-terminal domain of STAT1 (STAT1ND), thereby allowing SeV to escape from host innate immunity. Here we determined the crystal structure of STAT1ND associated with the C-terminal half of the C protein (Y3 [amino acids 99 to 204]) at a resolution of 2.0 Å. This showed that two molecules of Y3 symmetrically bind to each niche created between two molecules of the STAT1ND dimer. Molecular modeling suggested that an antiparallel form of the full-length STAT1 dimer can bind only one Y3 molecule and that a parallel form can bind two Y3 molecules. Affinity analysis demonstrated anticooperative binding of two Y3 molecules with the STAT1 dimer, which is consistent with the hypothetical model that the second Y3 molecule can only target the STAT1 dimer in a parallel form. STAT1 with excess amounts of Y3 was prone to inhibit the dephosphorylation at Tyr(701) by a phosphatase. In an electrophoretic mobility shift assay, tyrosine-phosphorylated STAT1 (pY-STAT1) with Y3 associated with the γ-activated sequence, probably as high-molecular-weight complexes (HMWCs), which may account for partial inhibition of a reporter assay from IFN-γ by Y3. Our study suggests that the full-length C protein interferes with the domain arrangement of the STAT1 dimer, leading to the accumulation of pY-STAT1 and the formation of HMWCs. In addition, we discuss the mechanism by which phosphorylation of STAT2 is inhibited in the presence of the C protein after stimulation by IFN-α/ß. IMPORTANCE: Sendai virus, a paramyxovirus that causes respiratory diseases in rodents, possesses the C protein, which inhibits the signal transduction pathways of interferon alpha/beta (IFN-α/ß) and IFN-γ by binding to the transcription factor STAT1. In virus-infected cells, phosphorylation of STAT1 at the Tyr(701) residue is potently enhanced, although transcription by STAT1 is inert. Here, we determined the crystal structure of the N-terminal domain of STAT1 associated with the C-terminal half of the C protein. Molecular modeling and experiments suggested that the two C proteins bind to and stabilize the parallel form of the STAT1 dimer, which are likely to be phosphorylated at Tyr(701), further inducing high-molecular-weight complex formation and inhibition of transcription by IFN-γ. We also discuss the possible mechanism of inhibition of the IFN-α/ß pathways by the C protein. This is the first structural report of the C protein, suggesting a mechanism of evasion of the paramyxovirus from innate immunity.


Subject(s)
Interferon-alpha/antagonists & inhibitors , Interferon-beta/antagonists & inhibitors , Interferon-gamma/antagonists & inhibitors , STAT1 Transcription Factor/antagonists & inhibitors , Viral Proteins/ultrastructure , Binding Sites , Cell Line , Crystallography, X-Ray , Electrophoretic Mobility Shift Assay , HEK293 Cells , Humans , Interferon-alpha/metabolism , Interferon-beta/metabolism , Models, Molecular , Phosphorylation , Protein Binding , Protein Structure, Tertiary , STAT1 Transcription Factor/metabolism , STAT1 Transcription Factor/ultrastructure , STAT2 Transcription Factor/metabolism , Sendai virus/metabolism , Signal Transduction/physiology , Viral Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...