Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 110
Filtrar
1.
J Biol Chem ; 296: 100280, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33450227

RESUMEN

Protein tyrosine phosphorylation regulates the production of capsular polysaccharide, an essential virulence factor of the deadly pathogen Vibrio vulnificus. The process requires the protein tyrosine kinase Wzc and its cognate phosphatase Wzb, both of which are largely uncharacterized. Herein, we report the structures of Wzb of V. vulnificus (VvWzb) in free and ligand-bound forms. VvWzb belongs to the low-molecular-weight protein tyrosine phosphatase (LMWPTP) family. Interestingly, it contains an extra four-residue insertion in the W-loop, distinct from all known LMWPTPs. The W-loop of VvWzb protrudes from the protein body in the free structure, but undergoes significant conformational changes to fold toward the active site upon ligand binding. Deleting the four-residue insertion from the W-loop severely impaired the enzymatic activity of VvWzb, indicating its importance for optimal catalysis. However, mutating individual residues or even substituting the whole insertion with four alanine residues only modestly decreased the enzymatic activity, suggesting that the contribution of the insertion to catalysis is not determined by the sequence specificity. Furthermore, inserting the four residues into Escherichia coli Wzb at the corresponding position enhanced its activity as well, indicating that the four-residue insertion in the W-loop can act as a general activity enhancing element for other LMWPTPs. The novel W-loop type and phylogenetic analysis suggested that VvWzb and its homologs should be classified into a new group of LMWPTPs. Our study sheds new insight into the catalytic mechanism and structural diversity of the LMWPTP family and promotes the understanding of the protein tyrosine phosphorylation system in prokaryotes.


Asunto(s)
Proteínas Bacterianas/genética , Proteínas de Escherichia coli/genética , Proteínas de la Membrana/genética , Fosfoproteínas Fosfatasas/genética , Proteínas Tirosina Fosfatasas/genética , Proteínas Tirosina Quinasas/genética , Vibrio vulnificus/genética , Secuencia de Aminoácidos/genética , Proteínas Bacterianas/química , Dominio Catalítico/genética , Escherichia coli/enzimología , Escherichia coli/genética , Proteínas de Escherichia coli/química , Humanos , Ligandos , Proteínas de la Membrana/química , Modelos Moleculares , Polímeros Impresos Molecularmente/química , Fosfoproteínas Fosfatasas/química , Filogenia , Proteínas Tirosina Fosfatasas/clasificación , Proteínas Tirosina Quinasas/química , Homología de Secuencia de Aminoácido , Especificidad por Sustrato , Vibrio vulnificus/química , Vibrio vulnificus/enzimología
2.
Biochem Biophys Res Commun ; 534: 604-609, 2021 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-33213836

RESUMEN

CmoB utilizes carboxy-S-adenosyl-l-methionine (CxSAM) to carry out unusual carboxymethyl transfer to form 5-carboxymethoxyuridine (cmo5U) of several tRNA species in Gram-negative bacteria. In this report, we present three X-ray crystal structures of CmoB from Vibrio vulnificus representing different states in the course of the reaction pathway; i.e., apo-, substrate-bound, and product-bound forms. Especially, the crystal structure of apo-CmoB unveils a novel open state of the enzyme, capturing unprecedented conformational dynamics around the substrate-binding site. The apo-structure demonstrates that the open conformation favors the release of CxSAM thus representing an inactive form. Our crystal structures of CmoB complexed with CxSAM and S-adenosyl-l-homocysteine (SAH) and combined binding assay results support the proposed mechanism underlying the cofactor selectivity, where CmoB preferentially senses negative charge around amino acid residues Lys-91, Tyr-200, and Arg-315.


Asunto(s)
Proteínas Bacterianas/metabolismo , Metiltransferasas/metabolismo , ARN Bacteriano/metabolismo , ARN de Transferencia/metabolismo , Uridina/análogos & derivados , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Dominio Catalítico , Cristalografía por Rayos X , Ligandos , Metiltransferasas/química , Metiltransferasas/genética , Modelos Moleculares , Conformación Proteica , Pliegue de Proteína , ARN Bacteriano/química , ARN Bacteriano/genética , ARN de Transferencia/química , ARN de Transferencia/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , S-Adenosilmetionina/análogos & derivados , S-Adenosilmetionina/metabolismo , Uridina/química , Uridina/metabolismo , Vibrio vulnificus/enzimología , Vibrio vulnificus/genética
3.
J Biol Chem ; 294(30): 11391-11401, 2019 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-31073025

RESUMEN

Phospholipases can disrupt host membranes and are important virulence factors in many pathogens. VvPlpA is a phospholipase A2 secreted by Vibrio vulnificus and essential for virulence. Its homologs, termed thermolabile hemolysins (TLHs), are widely distributed in Vibrio bacteria, but no structural information for this virulence factor class is available. Herein, we report the crystal structure of VvPlpA to 1.4-Å resolution, revealing that VvPlpA contains an N-terminal domain of unknown function and a C-terminal phospholipase domain and that these two domains are packed closely together. The phospholipase domain adopts a typical SGNH hydrolase fold, containing the four conserved catalytic residues Ser, Gly, Asn, and His. Interestingly, the structure also disclosed that the phospholipase domain accommodates a chloride ion near the catalytic His residue. The chloride is five-coordinated in a distorted bipyramid geometry, accepting hydrogen bonds from a water molecule and the amino groups of surrounding residues. This chloride substitutes for the most common Asp/Glu residue and forms an unusual Ser-His-chloride catalytic triad in VvPlpA. The chloride may orient the catalytic His and stabilize the charge on its imidazole ring during catalysis. Indeed, VvPlpA activity depended on chloride concentration, confirming the important role of chloride in catalysis. The VvPlpA structure also revealed a large hydrophobic substrate-binding pocket that is capable of accommodating a long-chain acyl group. Our results provide the first structure of the TLH family and uncover an unusual Ser-His-chloride catalytic triad, expanding our knowledge on the biological role of chloride.


Asunto(s)
Dominio Catalítico , Cloruros/química , Histidina/química , Fosfolipasas A2/química , Serina/química , Vibrio vulnificus/enzimología , Secuencia de Aminoácidos , Conformación Proteica , Alineación de Secuencia
4.
J Biol Chem ; 293(47): 18110-18122, 2018 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-30282804

RESUMEN

Multifunctional autoprocessing repeats-in-toxin (MARTX) toxins are secreted by Gram-negative bacteria and function as primary virulence-promoting macromolecules that deliver multiple cytopathic and cytotoxic effector domains into the host cytoplasm. Among these effectors, Ras/Rap1-specific endopeptidase (RRSP) catalyzes the sequence-specific cleavage of the Switch I region of the cellular substrates Ras and Rap1 that are crucial for host innate immune defenses during infection. To dissect the molecular basis underpinning RRSP-mediated substrate inactivation, we determined the crystal structure of an RRSP from the sepsis-causing bacterial pathogen Vibrio vulnificus (VvRRSP). Structural and biochemical analyses revealed that VvRRSP is a metal-independent TIKI family endopeptidase composed of an N-terminal membrane-localization and substrate-recruitment domain (N lobe) connected via an inter-lobe linker to the C-terminal active site-coordinating core ß-sheet-containing domain (C lobe). Structure-based mutagenesis identified the 2His/2Glu catalytic residues in the core catalytic domain that are shared with other TIKI family enzymes and that are essential for Ras processing. In vitro KRas cleavage assays disclosed that deleting the N lobe in VvRRSP causes complete loss of enzymatic activity. Endogenous Ras cleavage assays combined with confocal microscopy analysis of HEK293T cells indicated that the N lobe functions both in membrane localization via the first α-helix and in substrate assimilation by altering the functional conformation of the C lobe to facilitate recruitment of cellular substrates. Collectively, these results indicate that RRSP is a critical virulence factor that robustly inactivates Ras and Rap1 and augments the pathogenicity of invading bacteria via the combined effects of its N and C lobes.


Asunto(s)
Proteínas Bacterianas/metabolismo , Endopeptidasas/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Sepsis/enzimología , Sepsis/microbiología , Vibrio vulnificus/enzimología , Proteínas de Unión al GTP rap1/metabolismo , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Toxinas Bacterianas , Endopeptidasas/química , Endopeptidasas/genética , Células HEK293 , Interacciones Huésped-Patógeno , Humanos , Dominios Proteicos , Proteínas Proto-Oncogénicas p21(ras)/química , Proteínas Proto-Oncogénicas p21(ras)/genética , Sepsis/genética , Vibrio vulnificus/química , Vibrio vulnificus/genética , Proteínas de Unión al GTP rap1/química , Proteínas de Unión al GTP rap1/genética
5.
Can J Microbiol ; 65(9): 681-690, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31075207

RESUMEN

Gastric acidity is one of the earliest host defences faced by ingested organisms, and successful pathogens need to overcome this hurdle. The objective of this study was the systematic assessment of acid-stress response of Vibrio vulnificus isolated from coastal regions of Mangaluru. Acid-shock experiments were carried out at pH 4.0 and pH 4.5, with different experimental conditions expected to produce a varied acid response. Exposure to mild acid before the acid shock was favourable to the bacteria but was dependent on cell population and pH of the media and was independent of the strains tested. Lysine-dependent acid response was demonstrated with reference to the previously identified lysine decarboxylase system. Additionally, the results showed that inoculation into oysters provided some level of protection against acid stress. Increased expression of lysine/cadaverine genes was observed upon the addition of ground oyster and was confirmed by quantitative real-time PCR. The potential role of ornithine was analyzed with regard to acid stress, but no change in the survival pattern was observed. These findings highlight the physiology of bacteria in acid stress.


Asunto(s)
Ostreidae/microbiología , Estrés Fisiológico , Vibrio vulnificus/fisiología , Ácidos , Animales , Proteínas Bacterianas/metabolismo , Carboxiliasas/metabolismo , Ambiente , Genotipo , Concentración de Iones de Hidrógeno , India , Vibrio vulnificus/enzimología , Vibrio vulnificus/genética , Vibrio vulnificus/crecimiento & desarrollo
6.
Biotechnol Lett ; 41(10): 1155-1162, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31392514

RESUMEN

OBJECTIVES: To survey a library of over-expressed nitroreductases to identify those most active with 2,4- and 2,6-dinitrotoluene substrates, as promising candidates for phytoremediation of soils and groundwater contaminated with poly-nitro toluene pollutants. RESULTS: To indirectly monitor dinitrotoluene reduction we implemented a nitroblue tetrazolium dye screen to compare relative rates of NADPH consumption for 58 nitroreductase candidates, over-expressed in a nitroreductase-deleted strain of Escherichia coli. Although the screen only provides activity data at a single substrate concentration, by altering the substrate concentration and duration of incubation we showed we could first distinguish between more-active and less-active enzymes and then discriminate between the relative rates of reduction exhibited by the most active nitroreductases in the collection. We observed that members of the NfsA and NfsB nitroreductase families were the most active with 2,4-dinitrotoluene, but that only members of the NfsB family reduced 2,6-dinitrotoluene effectively. Two NfsB family members, YfkO from Bacillus subtilis and NfsB from Vibrio vulnificus, appeared especially effective with these substrates. Purification of both enzymes as His6-tagged recombinant proteins enabled in vitro determination of Michaelis-Menten kinetic parameters with each dinitrotoluene substrate. CONCLUSIONS: Vibrio vulnificus NfsB is a particularly promising candidate for bioremediation applications, being ca. fivefold more catalytically efficient with 2,4-dinitrotoluene and over 26-fold more active with 2,6-dinitrotoluene than the benchmark E. coli nitroreductases NfsA and NfsB.


Asunto(s)
Bacillus subtilis/enzimología , Biodegradación Ambiental , Dinitrobencenos/metabolismo , Contaminantes Ambientales/metabolismo , Nitrorreductasas/análisis , Vibrio vulnificus/enzimología , Cinética , Nitrorreductasas/aislamiento & purificación , Oxidación-Reducción
7.
J Bacteriol ; 200(7)2018 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-29339417

RESUMEN

Septicemia-causing Vibrio vulnificus produces at least three exoproteases, VvpE, VvpS, and VvpM, all of which participate in interactions with human cells. Expression of VvpE and VvpS is induced in the stationary phase by multiple transcription factors, including sigma factor S, SmcR, and the cAMP-cAMP receptor protein (cAMP-CRP) complex. Distinct roles of VvpM, such as induction of apoptosis, lead us to hypothesize VvpM expression is different from that of the other exoproteases. Its transcription, which was found to be independent of sigma S, is induced at the early exponential phase and then becomes negligible upon entry into the stationary phase. SmcR and CRP were studied regarding the control of vvpM expression. Transcription of vvpM was repressed by SmcR and cAMP-CRP complex individually, which specifically bound to the regions -2 to +20 and +6 to +27, respectively, relative to the vvpM transcription initiation site. Derepression of vvpM gene expression was 10- to 40-fold greater in an smcR crp double mutant than in single-gene mutants. Therefore, these results show that the expression of V. vulnificus exoproteases is differentially regulated, and in this way, distinct proteases can engage in specific interactions with a host.IMPORTANCE An opportunistic human pathogen, Vibrio vulnificus produces multiple extracellular proteases that are involved in diverse interactions with a host. The total exoproteolytic activity is detected mainly in the supernatants of the high-cell-density cultures. However, some proteolytic activity derived from a metalloprotease, VvpM, was present in the supernatants of the low-cell-density cultures sampled at the early growth period. In this study, we present the regulatory mechanism for VvpM expression via repression by at least two transcription factors. This type of transcriptional regulation is the exact opposite of those for expression of the other V. vulnificus exoproteases. Differential regulation of each exoprotease's production then facilitates the pathogen's participation in the distinct interactions with a host.


Asunto(s)
Proteínas Bacterianas/biosíntesis , Proteínas Bacterianas/genética , Regulación Bacteriana de la Expresión Génica , Metaloendopeptidasas/biosíntesis , Metaloendopeptidasas/genética , Percepción de Quorum , Vibrio vulnificus/genética , Apoptosis , Proteína Receptora de AMP Cíclico/metabolismo , Represión Enzimática/genética , Humanos , Proteolisis , Factores de Transcripción/genética , Vibrio vulnificus/enzimología
8.
J Biol Chem ; 292(41): 17129-17143, 2017 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-28855258

RESUMEN

The marine bacterium Vibrio vulnificus causes food-borne diseases, which may lead to life-threatening septicemia in some individuals. Therefore, identifying virulence factors in V. vulnificus is of high priority. We performed a transcriptome analysis on V. vulnificus after infection of human intestinal HT29-methotrexate cells and found induction of plpA, encoding a putative phospholipase, VvPlpA. Bioinformatics, biochemical, and genetic analyses demonstrated that VvPlpA is a phospholipase A2 secreted in a type II secretion system-dependent manner. Compared with the wild type, the plpA mutant exhibited reduced mortality, systemic infection, and inflammation in mice as well as low cytotoxicity toward the human epithelial INT-407 cells. Moreover, plpA mutation attenuated the release of actin and cytosolic cyclophilin A from INT-407 cells, indicating that VvPlpA is a virulence factor essential for causing lysis and necrotic death of the epithelial cells. plpA transcription was growth phase-dependent, reaching maximum levels during the early stationary phase. Also, transcription factor HlyU and cAMP receptor protein (CRP) mediate additive activation and host-dependent induction of plpA Molecular biological analyses revealed that plpA expression is controlled via the promoter, P plpA , and that HlyU and CRP directly bind to P plpA upstream sequences. Taken together, this study demonstrated that VvPlpA is a type II secretion system-dependent secretory phospholipase A2 regulated by HlyU and CRP and is essential for the pathogenicity of V. vulnificus.


Asunto(s)
Proteínas Bacterianas/metabolismo , Fosfolipasas A2/metabolismo , Vibriosis/enzimología , Vibrio vulnificus/enzimología , Vibrio vulnificus/patogenicidad , Proteínas Bacterianas/genética , Sistemas de Secreción Bacterianos/genética , Sistemas de Secreción Bacterianos/metabolismo , Línea Celular , Humanos , Mucosa Intestinal/microbiología , Mucosa Intestinal/patología , Fosfolipasas A2/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Vibriosis/genética , Vibriosis/patología , Vibrio vulnificus/genética
9.
Infect Immun ; 86(9)2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29914931

RESUMEN

Vibrio vulnificus, an opportunistic human pathogen, produces cyclo-(l-Phe-l-Pro) (cFP), which serves as a signaling molecule controlling the ToxR-dependent expression of innate bacterial genes, and also as a virulence factor eliciting pathogenic effects on human cells by enhancing intracellular reactive oxygen species levels. We found that cFP facilitated the protection of V. vulnificus against hydrogen peroxide. At a concentration of 1 mM, cFP enhanced the level of the transcriptional regulator RpoS, which in turn induced expression of katG, encoding hydroperoxidase I, an enzyme that detoxifies H2O2 to overcome oxidative stress. We found that cFP upregulated the transcription of the histone-like proteins vHUα and vHUß through the cFP-dependent regulator LeuO. LeuO binds directly to upstream regions of vhuA and vhuB to enhance transcription. vHUα and vHUß then enhance the level of RpoS posttranscriptionally by stabilizing the mRNA. This cFP-mediated ToxR-LeuO-vHUαß-RpoS pathway also upregulates genes known to be members of the RpoS regulon, suggesting that cFP acts as a cue for the signaling pathway responsible for both the RpoS and the LeuO regulons. Taken together, this study shows that cFP plays an important role as a virulence factor, as well as a signal for the protection of the cognate pathogen.


Asunto(s)
Estrés Oxidativo , Péptidos Cíclicos/farmacología , Peroxidasas/genética , Percepción de Quorum , Transducción de Señal , Vibrio vulnificus/enzimología , Proteínas Bacterianas/genética , Dipéptidos/farmacología , Regulación Bacteriana de la Expresión Génica , Factor sigma/genética , Factores de Transcripción/genética , Vibrio vulnificus/genética , Factores de Virulencia/genética
10.
Protein Expr Purif ; 149: 13-16, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29635038

RESUMEN

Vibrio vulnificus is a human pathogen causing fatal septicemia with edematous and hemorrhagic skin damage. Among multiple virulence factors, an extracellular metalloprotease termed as V. vulnificus protease (VVP) is known to play a crucial role in eliciting the skin damage. The mature VVP (413 aa) is composed of two domains, the N-terminal core domain with proteolytic activity and the C-terminal domain mediates efficient attachment to protein substrates. However, VVP is produced as an inactive precursor (609 aa) with a signal peptide (24 aa) and propeptide (172 aa). In order to clarify the function of propeptide, a series of DNA fragments encoding the VVP precursor and its various domains were designed and the proteins were expressed in vitro by using cell-free translational system. The results indicated that the propeptide might function as an intramolecular chaperon to promote the proper folding of both N-terminal and C-terminal domains. The obtained results also suggest that the propeptide, itself was unstable and thus digested easily by the enzymes present in cell lysate used for cell-free system. Additionally, the C-terminal domain in VVP found to inhibit the folding of the N-terminal domain in absence of propeptide.


Asunto(s)
Sistema Libre de Células , Metaloproteasas/metabolismo , Vibrio vulnificus/enzimología , Expresión Génica , Metaloproteasas/genética , Péptidos/genética , Péptidos/metabolismo , Procesamiento Proteico-Postraduccional , Factores de Virulencia/metabolismo
11.
Biochemistry ; 56(21): 2747-2757, 2017 05 30.
Artículo en Inglés | MEDLINE | ID: mdl-28459538

RESUMEN

Ras/Rap1-specific endopeptidase (RRSP) is a cytotoxic effector domain of the multifunctional autoprocessing repeats-in-toxin (MARTX) toxin of highly virulent strains of Vibrio vulnificus. RRSP blocks RAS-MAPK kinase signaling by cleaving Ras and Rap1 within the switch I region between Y32 and D33. Although the RRSP processing site is highly conserved among small GTPases, only Ras and Rap1 have been identified as proteolytic substrates. Here we report that residues Y32 and D33 at the scissile bond play an important role in RRSP substrate recognition, while the nucleotide state of Ras has an only minimal effect. In addition, substrate specificity is generated by residues across the entire switch I region. Indeed, swapping the Ras switch I region into either RalA or RhoA, GTPases that are not recognized by RRSP, generated chimeras that are substrates of RRSP. However, a difference in the processing efficiency of Ras switch I in the context of Ras, RalA, or RhoA indicates that protein regions outside Ras switch I also contribute to efficient RRSP substrate recognition. Moreover, we show that synthetic peptides corresponding to the Ras and Rap1, but not RalA, switch I regions are cleaved by RRSP, demonstrating sequence-specific substrate recognition. In conclusion, this work demonstrates that the GTPase recognition of RRSP is independent of the nucleotide state and is mainly driven by the Ras and Rap1 switch I loop and also influenced by additional protein-protein interactions, increasing the substrate specificity of RRSP.


Asunto(s)
Toxinas Bacterianas/química , Toxinas Bacterianas/metabolismo , Endopeptidasas/química , Endopeptidasas/metabolismo , Vibrio vulnificus/enzimología , Proteínas de Unión al GTP rap1/metabolismo , Proteínas ras/metabolismo , Humanos , Modelos Moleculares , Especificidad por Sustrato , Proteínas de Unión al GTP rap1/química , Proteínas ras/química
12.
Protein Expr Purif ; 133: 121-131, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28302513

RESUMEN

Undecaprenyl pyrophosphate phosphatase (UppP), a cell membrane integral enzyme, catalyzes the dephosphorylation of undecaprenyl pyrophosphate to undecaprenyl phosphate, which is an essential carrier lipid in bacterial cell wall synthesis. We previously purified E. coli UppP and concluded that its catalytic site is likely located in the periplasm. To search for additional natural UppP homologs to elucidate what constitutes a common catalytic mechanism and to gain a better chance of obtaining high-resolution crystal structural information, we expressed and purified recombinant Vibrio vulnificus UppP using E. coli as a host. Mutagenesis analysis demonstrates that the proposed catalytic residues Gln-13, Glu-17, His-26 and Arg-166 are directly involved in enzyme catalysis. Additionally, mutations of most of the conserved serine and glycine residues within the proposed catalytic site (S22A, G163A and S165A) lead to complete inactivity, very low activity (<1.3% of the wild type) or no protein expression at all (G163R and G168A), whereas S23A and S167A retain enzyme activity (65% and 34%). Kinetic analysis indicates that S23A and S167A result in 1.4- and 5-fold decreases in kcat, whereas the substrate Km value exhibits only minor changes compared with wild-type UppP, implying that they are involved in enzyme catalysis. The structural modeling and molecular dynamics simulation analyses also provide a plausible structure of the catalytic core, centered on a conserved histidine (His-26) that initiates the hydrolysis of phosphate esters, rationalizing the mutagenesis data. This conclusion can be applied generally to all bacterial UppP enzymes.


Asunto(s)
Proteínas Bacterianas , Expresión Génica , Simulación de Dinámica Molecular , Pirofosfatasas , Vibrio vulnificus , Proteínas Bacterianas/biosíntesis , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Proteínas Bacterianas/aislamiento & purificación , Cristalografía por Rayos X , Dominios Proteicos , Pirofosfatasas/biosíntesis , Pirofosfatasas/química , Pirofosfatasas/genética , Pirofosfatasas/aislamiento & purificación , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , Vibrio vulnificus/enzimología , Vibrio vulnificus/genética
13.
Microbiol Immunol ; 61(1): 1-11, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28111826

RESUMEN

Vibrio vulnificus, a gram-negative halophilic estuarine bacterium, is an opportunistic human pathogen that causes rapidly progressive fatal septicemia and necrotizing wound infection. This species also causes hemorrhagic septicemia called vibriosis in cultured eels. It has been proposed that a range of virulence factors play roles in pathogenesis during human and/or eel infection. Among these factors, a metalloprotease (V. vulnificus protease [VVP]) and a cytolytic toxin (V. vulnificus hemolysin [VVH]) are of significant importance. VVP elicits the characteristic edematous and hemorrhagic skin damage, whereas VVH exhibits powerful hemolytic and cytolytic activities and contributes to bacterial invasion from the intestine to the blood stream. In addition, a few V. vulnificus strains isolated from diseased eels have recently been found to produce a serine protease designated as V. vulnificus serine protease (VvsA) instead of VVP. Similarly to VVP, VvsA may possess various toxic activities such as collagenolytic, cytotoxic and edema-forming activity. In this review, regulation of V. vulnificus VVP, VVH and VvsA is clarified in terms of expression at the mRNA and protein levels. The explanation is given on the basis of the quorum sensing system, which is dependent on bacterial cell density. In addition, the roles of environmental factors and global regulators, such as histone-like nucleoid structuring protein, cyclic adeno monophosphate receptor protein, RpoS, HlyU, Fur, ToxRS, AphB and LeuO, in this regulation are outlined. The cumulative impact of these regulatory systems on the pathogenicity of V. vulnificus is here delineated.


Asunto(s)
Proteínas Hemolisinas/biosíntesis , Metaloproteasas/biosíntesis , Vibrio vulnificus/metabolismo , Animales , Proteínas Bacterianas/biosíntesis , Proteínas Bacterianas/genética , Proteínas Hemolisinas/genética , Humanos , Metaloendopeptidasas/biosíntesis , Metaloendopeptidasas/genética , Metaloproteasas/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Vibrio vulnificus/enzimología , Vibrio vulnificus/genética , Factores de Virulencia/genética
14.
J Biol Chem ; 290(30): 18708-20, 2015 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-26041774

RESUMEN

We describe a novel insulin-degrading enzyme, SidC, that contributes to the proliferation of the human bacterial pathogen Vibrio vulnificus in a mouse model. SidC is phylogenetically distinct from other known insulin-degrading enzymes and is expressed and secreted specifically during host infection. Purified SidC causes a significant decrease in serum insulin levels and an increase in blood glucose levels in mice. A comparison of mice infected with wild type V. vulnificus or an isogenic sidC-deletion strain showed that wild type bacteria proliferated to higher levels. Additionally, hyperglycemia leads to increased proliferation of V. vulnificus in diabetic mice. Consistent with these observations, the sid operon was up-regulated in response to low glucose levels through binding of the cAMP-receptor protein (CRP) complex to a region upstream of the operon. We conclude that glucose levels are important for the survival of V. vulnificus in the host, and that this pathogen uses SidC to actively manipulate host endocrine signals, making the host environment more favorable for bacterial survival and growth.


Asunto(s)
Proliferación Celular/genética , Interacciones Huésped-Patógeno/genética , Insulisina/genética , Ratones Endogámicos NOD/genética , Vibrio vulnificus/enzimología , Animales , Glucemia/metabolismo , Diabetes Mellitus Experimental/enzimología , Diabetes Mellitus Experimental/microbiología , Diabetes Mellitus Experimental/patología , Modelos Animales de Enfermedad , Regulación Bacteriana de la Expresión Génica , Humanos , Insulina/sangre , Insulisina/química , Insulisina/aislamiento & purificación , Ratones , Ratones Endogámicos NOD/microbiología , Vibriosis/genética , Vibriosis/microbiología , Vibriosis/patología , Vibrio vulnificus/genética , Vibrio vulnificus/patogenicidad
15.
J Biol Chem ; 290(35): 21231-43, 2015 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-26160170

RESUMEN

Family 2 polysaccharide lyases (PL2s) preferentially catalyze the ß-elimination of homogalacturonan using transition metals as catalytic cofactors. PL2 is divided into two subfamilies that have been generally associated with secretion, Mg(2+) dependence, and endolysis (subfamily 1) and with intracellular localization, Mn(2+) dependence, and exolysis (subfamily 2). When present within a genome, PL2 genes are typically found as tandem copies, which suggests that they provide complementary activities at different stages along a catabolic cascade. This relationship most likely evolved by gene duplication and functional divergence (i.e. neofunctionalization). Although the molecular basis of subfamily 1 endolytic activity is understood, the adaptations within the active site of subfamily 2 enzymes that contribute to exolysis have not been determined. In order to investigate this relationship, we have conducted a comparative enzymatic analysis of enzymes dispersed within the PL2 phylogenetic tree and elucidated the structure of VvPL2 from Vibrio vulnificus YJ016, which represents a transitional member between subfamiles 1 and 2. In addition, we have used ancestral sequence reconstruction to functionally investigate the segregated evolutionary history of PL2 progenitor enzymes and illuminate the molecular evolution of exolysis. This study highlights that ancestral sequence reconstruction in combination with the comparative analysis of contemporary and resurrected enzymes holds promise for elucidating the origins and activities of other carbohydrate active enzyme families and the biological significance of cryptic metabolic pathways, such as pectinolysis within the zoonotic marine pathogen V. vulnificus.


Asunto(s)
Bacterias/enzimología , Evolución Molecular , Polisacárido Liasas/genética , Polisacárido Liasas/metabolismo , Secuencia de Aminoácidos , Bacterias/química , Bacterias/genética , Bacterias/metabolismo , Magnesio/metabolismo , Manganeso/metabolismo , Modelos Moleculares , Datos de Secuencia Molecular , Filogenia , Polisacárido Liasas/química , Conformación Proteica , Alineación de Secuencia , Vibrio vulnificus/química , Vibrio vulnificus/enzimología , Vibrio vulnificus/genética , Vibrio vulnificus/metabolismo
16.
Mol Microbiol ; 97(2): 330-46, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25869813

RESUMEN

An exoprotease of Vibrio vulnificus, VvpS, exhibits an autolytic function during the stationary phase. To understand how vvpS expression is controlled, the regulators involved in vvpS transcription and their regulatory mechanisms were investigated. LeuO was isolated in a ligand-fishing experiment, and experiments using a leuO-deletion mutant revealed that LeuO represses vvpS transcription. LeuO bound the extended region including LeuO-binding site (LBS)-I and LBS-II. Further screening of additional regulators revealed that SmcR and cyclic adenosine monophosphate-receptor protein (CRP) play activating roles in vvpS transcription. SmcR and CRP bound the regions overlapping LBS-I and -II, respectively. In addition, the LeuO occupancy of LBS-I and LBS-II was competitively exchanged by SmcR and CRP, respectively. To examine the mechanism of stationary-phase induction of vvpS expression, in vivo levels of three transcription factors were monitored. Cellular level of LeuO was maximal at exponential phase, while those of SmcR and CRP were maximal at stationary phase and relatively constant after the early-exponential phase, respectively. Thus, vvpS transcription was not induced during the exponential phase by high cellular content of LeuO. When entering the stationary phase, however, LeuO content was significantly reduced and repression by LeuO was relieved through simultaneous binding of SmcR and CRP to LBS-I and -II, respectively.


Asunto(s)
Exopeptidasas/biosíntesis , Factores de Transcripción/metabolismo , Vibrio vulnificus/metabolismo , Proteínas Bacterianas/metabolismo , Inducción Enzimática , Exopeptidasas/genética , Exopeptidasas/metabolismo , Regulación Bacteriana de la Expresión Génica , Regulación Enzimológica de la Expresión Génica , Unión Proteica , Serina Proteasas/biosíntesis , Serina Proteasas/genética , Serina Proteasas/metabolismo , Vibrio vulnificus/enzimología , Vibrio vulnificus/genética , Vibrio vulnificus/crecimiento & desarrollo
17.
Mol Microbiol ; 96(3): 497-512, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25626927

RESUMEN

Vibrio vulnificus contains two coproporphyrinogen III oxidases (CPOs): O2-dependent HemF and O2-independent HemN. The growth of the hemF mutant HF1 was similar to wild-type cells at pH 7.5 under 2% O2 conditions where HemN was active and had a half-life of 64 min. However, HF1 did not grow when the medium pH decreased to pH 5.0, where oxidative stress affects endogenous S-adenosylmethionine (SAM) levels. The growth of HF1 was restored not only by elevating the expression of MnSOD but also through the exogenous addition of SAM. For HF1 to grow under these SAM-limiting conditions, a mutation arose in hemN, encoding HemNY74F . Refolding of the denatured enzymes in vitro revealed that the apparent binding affinity of HemNY74F for the cofactor SAM1, which coordinates the 4Fe-4S cluster, was approximately sixfold higher than that of HemN. The Km of HemNY74F for the co-substrate SAM2, which provides radicals for CPO reactions, was threefold lower than that of HemN. Thus, affinities for both SAM1 and SAM2 were higher with the Y74F mutation. Taken together, when SAM is limiting, HemN is apparently nonfunctional, and heme synthesis is continued by HemF.


Asunto(s)
Coenzimas/metabolismo , Coproporfirinógeno Oxidasa/metabolismo , Hemo/biosíntesis , S-Adenosilmetionina/metabolismo , Vibrio vulnificus/enzimología , Vibrio vulnificus/metabolismo , Coproporfirinógeno Oxidasa/genética , Medios de Cultivo/química , Eliminación de Gen , Concentración de Iones de Hidrógeno , Cinética , Mutación Missense , Oxígeno/metabolismo , Unión Proteica , Supresión Genética , Vibrio vulnificus/crecimiento & desarrollo
18.
Mol Microbiol ; 96(2): 293-305, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25598011

RESUMEN

The bacterial phosphoenolpyruvate:sugar phosphotransferase system (PTS) consists of two general energy-coupling proteins [enzyme I and histidine phosphocarrier protein (HPr)] and several sugar-specific enzyme IIs. Although, in addition to the phosphorylation-coupled transport of sugars, various regulatory roles of PTS components have been identified in Escherichia coli, much less is known about the PTS in the opportunistic human pathogen Vibrio vulnificus. In this study, we have identified pyruvate kinase A (PykA) as a binding partner of HPr in V. vulnificus. The interaction between HPr and PykA was strictly dependent on the presence of inorganic phosphate, and only dephosphorylated HPr interacted with PykA. Experiments involving domain swapping between the PykAs of V. vulnificus and E. coli revealed the requirement for the C-terminal domain of V. vulnificus PykA for a specific interaction with V. vulnificus HPr. Dephosphorylated HPr decreased the Km of PykA for phosphoenolpyruvate by approximately fourfold without affecting Vmax . Taken together, these findings indicate that the V. vulnificus PTS catalyzing the first step of glycolysis stimulates the final step of glycolysis in the presence of glucose through the direct interaction of dephospho-HPr with the C-terminal domain of PykA.


Asunto(s)
Proteínas Bacterianas/metabolismo , Glucosa/metabolismo , Sistema de Fosfotransferasa de Azúcar del Fosfoenolpiruvato/metabolismo , Piruvato Quinasa/metabolismo , Vibriosis/microbiología , Vibrio vulnificus/enzimología , Proteínas Bacterianas/genética , Regulación Bacteriana de la Expresión Génica , Histidina/metabolismo , Humanos , Sistema de Fosfotransferasa de Azúcar del Fosfoenolpiruvato/genética , Fosforilación , Piruvato Quinasa/genética , Vibrio vulnificus/genética , Vibrio vulnificus/metabolismo
19.
Curr Microbiol ; 72(6): 716-22, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26888524

RESUMEN

RNase E plays an important role in the degradation and processing of RNA in Escherichia coli. The enzymatic activity of RNase E is controlled by the protein inhibitors RraA and RraB. The marine pathogenic bacterium Vibrio vulnificus also contains homologs of RNase E and RraA, designated as RNase EV, RraAV1, and RraAV2. Here, we report that RraAV1 actively inhibits the enzymatic activity of RNase EV in vivo and in vitro by interacting with the C-terminal domain of RNase EV. Coexpression of RraAV1 reduced ribonucleolytic activity in the cells overproducing RNase EV and consequently restored normal growth of these cells. An in vitro cleavage assay further demonstrated that RraAV1 efficiently inhibits the ribonucleolytic activity of RNase EV on BR10 + hpT, a synthetic oligonucleotide containing the RNase E cleavage site of RNA I. Our findings suggest that RraAV1 plays an active role in RNase EV-mediated RNA cleavage in V. vulnificus.


Asunto(s)
Proteínas Bacterianas/metabolismo , Endorribonucleasas/metabolismo , Escherichia coli/genética , Vibrio vulnificus/enzimología , Proteínas Bacterianas/genética , Endorribonucleasas/genética , Escherichia coli/enzimología , Escherichia coli/metabolismo , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Regulación Bacteriana de la Expresión Génica , ARN Bacteriano/genética , ARN Bacteriano/metabolismo , Vibrio vulnificus/genética
20.
J Biol Chem ; 289(52): 36263-74, 2014 Dec 26.
Artículo en Inglés | MEDLINE | ID: mdl-25398878

RESUMEN

Peroxiredoxins (Prxs) are ubiquitous antioxidant enzymes that reduce toxic peroxides. A new Vibrio vulnificus Prx, named Prx3, was identified and characterized in this study. Biochemical and mutational analyses revealed that Prx3 reduces H2O2, utilizing glutaredoxin 3 (Grx3) and glutathione (GSH) as reductants, and requires only N-terminal peroxidatic cysteine for its catalysis. These results, combined with the monomeric size of Prx3 observed under non-reducing conditions, suggested that Prx3 is a Grx3/GSH-dependent 1-Cys Prx and oxidized without forming intermolecular disulfide bonds. The prx3 mutation impaired growth in the medium containing peroxides and reduced virulence in mice, indicating that Prx3 is essential for survival under oxidative stress and pathogenesis of V. vulnificus. The Fe-S cluster regulator IscR activates prx3 by direct binding to a specific binding sequence centered at -44 from the transcription start site. The binding sequence was homologous to the Type 2 IscR-binding sequence, most likely recognized by the Fe-S clusterless apo-IscR in Escherichia coli. The iscR3CA mutant, chromosomally encoding the apo-locked IscR, exhibited 3-fold higher levels of activation of prx3 than the wild type and accumulated more IscR3CA protein in cells. The IscR-dependent activation of prx3 by aerobic growth and iron starvation was also associated with the increase in cellular levels of IscR protein. Taken together, the results suggested that IscR senses iron starvation as well as reactive oxygen species and shifts to the apo-form, which leads to the increase of cellular IscR and in turn prx3 expression, contributing to the survival and virulence of V. vulnificus during pathogenesis.


Asunto(s)
Proteínas Bacterianas/fisiología , Peroxiredoxina III/fisiología , Vibrio vulnificus/enzimología , Secuencia de Aminoácidos , Animales , Proteínas Bacterianas/química , Secuencia de Bases , Sitios de Unión , Femenino , Regulación Bacteriana de la Expresión Génica , Hierro/metabolismo , Ratones Endogámicos ICR , Viabilidad Microbiana , Datos de Secuencia Molecular , Estrés Oxidativo , Peroxiredoxina III/química , Regiones Promotoras Genéticas , Transcripción Genética , Vibriosis/microbiología , Vibrio vulnificus/patogenicidad , Virulencia
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda