Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 179
Filtrar
Mais filtros

Intervalo de ano de publicação
1.
Microb Pathog ; 192: 106683, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38735447

RESUMO

Bacteria possess the ability to develop diverse and ingenious strategies to outwit the host immune system, and proteases are one of the many weapons employed by bacteria. This study sought to identify S. agalactiae additional serine protease and determine its role in virulence. The S. agalactiae THN0901 genome features one S8 family serine peptidase B (SfpB), acting as a secreted and externally exposed entity. A S8 family serine peptidase mutant strain (ΔsfpB) and complement strain (CΔsfpB) were generated through homologous recombination. Compared to the wild-type strain THN0901, the absorption of EtBr dyes was significantly reduced (P < 0.01) in ΔsfpB, implying an altered cell membrane permeability. In addition, the ΔsfpB strain had a significantly lower survival rate in macrophages (P < 0.01) and a 61.85 % lower adhesion ability to the EPC cells (P < 0.01) compared to THN0901. In the in vivo colonization experiment using tilapia as a model, 210 fish were selected and injected with different bacterial strains at a concentration of 3 × 106 CFU/tail. At 6, 12, 24, 48, 72 and 96 h post-injection, three fish were randomly selected from each group and their brain, liver, spleen, and kidney tissues were isolated. Subsequently, it was demonstrated that the ΔsfpB strain exhibited a markedly diminished capacity for colonization in tilapia. Additionally, the cumulative mortality of ΔsfpB in fish after intraperitoneal injection was reduced by 19.92-23.85 %. In conclusion, the findings in this study have demonstrated that the SfpB plays a significant role in S. agalactiae cell membrane stability and immune evasion. The immune evasion is fundamental for the development and transmission of invasive diseases, the serine protease SfpB may be a promising candidate for the development of antimicrobial agents to reduce the transmission of S. agalactiae.


Assuntos
Membrana Celular , Doenças dos Peixes , Evasão da Resposta Imune , Infecções Estreptocócicas , Streptococcus agalactiae , Streptococcus agalactiae/genética , Streptococcus agalactiae/patogenicidade , Streptococcus agalactiae/enzimologia , Streptococcus agalactiae/imunologia , Animais , Virulência , Infecções Estreptocócicas/microbiologia , Infecções Estreptocócicas/imunologia , Membrana Celular/metabolismo , Doenças dos Peixes/microbiologia , Doenças dos Peixes/imunologia , Aderência Bacteriana , Macrófagos/microbiologia , Macrófagos/imunologia , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Serina Proteases/genética , Serina Proteases/metabolismo , Fatores de Virulência/genética , Fatores de Virulência/metabolismo , Camundongos
2.
Glycobiology ; 31(11): 1435-1443, 2021 12 18.
Artigo em Inglês | MEDLINE | ID: mdl-34280262

RESUMO

Glycosaminoglycans (GAGs), such as hyaluronan (HA) and heparan sulfate (HS), are a large group of polysaccharides found in the extracellular matrix and on the cell surface. The turnover of these molecules is controlled by de novo synthesis and catabolism through specific endoglycosidases, which are the keys to our understanding of the homeostasis of GAGs and could hold opportunities for therapeutic intervention. Herein, we describe assays for endoglycosidases using nonreducing end fluorophore-labeled GAGs, in which GAGs were labeled via incorporation of GlcNAz by specific synthases and cycloaddition of alkyne fluorophores and then digested with corresponding endoglycosidases. Assays of various HA-specific hyaluronidases (HYALs), including PH-20 or SPAM1, and HS-specific heparanase (HPSE) are presented. We demonstrated the distinctive pH profiles, substrate specificities and specific activities of these enzymes and provided evidence that both HYAL3 and HYAL4 are authentic hyaluronidases. In addition, while all HYALs are active on high-molecular-weight HA, they are active on low-molecular-weight HA. Subsequently, we defined a new way of measuring the activities of HYALs. Our results indicate that the activities of HYALs must be under strict pH regulation. Our quantitative methods of measuring the activity GAG endoglycosidases could bring the opportunity of designing novel therapeutics by targeting these important enzymes.


Assuntos
Glucuronidase/metabolismo , Proteoglicanas de Heparan Sulfato/metabolismo , Ácido Hialurônico/metabolismo , Hialuronoglucosaminidase/metabolismo , Imagem Óptica , Eletroforese em Gel de Poliacrilamida , Concentração de Íons de Hidrogênio , Pasteurella multocida/enzimologia , Proteínas Recombinantes/metabolismo , Streptococcus agalactiae/enzimologia , Especificidade por Substrato
3.
J Antimicrob Chemother ; 74(4): 931-934, 2019 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-30649407

RESUMO

OBJECTIVES: We have previously identified group B Streptococcus (GBS) clinical isolates with reduced penicillin susceptibility (PRGBS) that were non-susceptible to cefotaxime; however, the rates of cefotaxime and ceftriaxone non-susceptibility among PRGBS isolates have never been reported. Therefore, we first determined the MICs of 22 antibacterial drugs/compounds for 74 PRGBS isolates and then determined the rates of cefotaxime and ceftriaxone non-susceptibility among these isolates. METHODS: We used 74 clinical PRGBS isolates, previously collected in Japan and confirmed to harbour relevant amino acid substitutions in PBP2X. We also used 80 penicillin-susceptible GBS (PSGBS) clinical isolates as controls. The MICs of 22 antibacterial drugs/compounds for all 154 GBS isolates were determined via microdilution and/or agar dilution methods, as recommended by the CLSI. RESULTS: The rates of non-susceptibility/resistance to ampicillin, cefotaxime, ceftriaxone and levofloxacin for the 80 PSGBS isolates were 0%, 0%, 0% and 30%, respectively, but were 15% (P = 0.0003), 28% (P < 0.0001), 36% (P < 0.0001) and 93% (P < 0.0001) for the 74 PRGBS isolates, respectively. No PRGBS isolates were identified to be non-susceptible to meropenem, doripenem, vancomycin, quinupristin/dalfopristin, daptomycin or linezolid. CONCLUSIONS: We found that cefotaxime- and ceftriaxone-non-susceptible PRGBS isolates occur at relatively high rates in Japan. Importantly, this finding suggests that the range of drugs likely to be effective in treating PRGBS infections may be limited compared with those available for PSGBS infections; therefore, clinicians should exercise care when considering drug choice and efficacy for PRGBS infections.


Assuntos
Antibacterianos/farmacologia , Cefotaxima/farmacologia , Ceftriaxona/farmacologia , Penicilinas/farmacologia , Streptococcus agalactiae/efeitos dos fármacos , Resistência beta-Lactâmica , Humanos , Japão/epidemiologia , Testes de Sensibilidade Microbiana , Mutação de Sentido Incorreto , Proteínas de Ligação às Penicilinas/genética , Prevalência , Infecções Estreptocócicas/epidemiologia , Infecções Estreptocócicas/microbiologia , Streptococcus agalactiae/enzimologia , Streptococcus agalactiae/genética , Streptococcus agalactiae/isolamento & purificação
4.
Microb Pathog ; 127: 359-367, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-30553015

RESUMO

GAPDH being a key enzyme in the glycolytic pathway is one of the surface adhesins of many Gram-positive bacteria including Streptococcus agalactiae. This anchorless adhesin is known to bind to host plasminogen (PLG) and fibrinogen (Fg), which enhances the virulence and modulates the host immune system. The crystal structure of the recombinant GAPDH from S. agalactiae (SagGAPDH) was determined at 2.6 Šresolution by molecular replacement. The structure was found to be highly conserved with a typical NAD binding domain and a catalytic domain. In this paper, using biolayer interferometry studies, we report that the multifunctional SagGAPDH enzyme binds to a variety of host molecules such as PLG, Fg, laminin, transferrin and mucin with a KD value of 4.4 × 10-7 M, 9.8 × 10-7 M, 1 × 10-5 M, 9.7 × 10-12 M and 1.4 × 10-7 M respectively. The ligand affinity blots reveal that SagGAPDH binds specifically to α and ß subunits of Fg and the competitive binding ELISA assay reveals that the Fg and PLG binding sites on GAPDH does not overlap each other. The PLG binding motif of GAPDH varies with organisms, however positively charged residues in the hydrophobic surroundings is essential for PLG binding. The lysine analogue competitive binding assay and lysine succinylation experiments deciphered the role of SagGAPDH lysines in PLG binding. On structural comparison with S. pneumoniae GAPDH, K171 of SagGAPDH is being predicted to be involved in PLG binding. Further SagGAPDH exhibited enzymatic activity in the presence of Fg, PLG and transferrin. This suggests that these host molecules does not mask the active site and bind at some other region of GAPDH.


Assuntos
Adesinas Bacterianas/química , Adesinas Bacterianas/metabolismo , Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Proteínas da Matriz Extracelular/metabolismo , Gliceraldeído-3-Fosfato Desidrogenase (Fosforiladora)/química , Gliceraldeído-3-Fosfato Desidrogenase (Fosforiladora)/metabolismo , Sítios de Ligação , Cristalografia por Raios X , Cinética , Modelos Moleculares , Ligação Proteica , Conformação Proteica , Domínios Proteicos , Streptococcus agalactiae/enzimologia
5.
Biochemistry ; 57(4): 461-468, 2018 01 30.
Artigo em Inglês | MEDLINE | ID: mdl-29320164

RESUMO

Streptide is a ribosomally synthesized and post-translationally modified peptide with a unique cyclization motif consisting of an intramolecular lysine-tryptophan cross-link. Three radical S-adenosylmethionine enzymes, StrB, AgaB, and SuiB from different species of Streptococcus, have been shown to install this modification onto their respective precursor peptides in a leader-dependent fashion. Herein, we conduct detailed investigations to differentiate among several plausible mechanistic proposals, specifically addressing radical versus electrophilic addition to the indole during cross-link formation, the role of substrate side chains in binding in the enzyme active site, and the identity of the catalytic base in the reaction cycle. Our results are consistent with a radical electrophilic aromatic substitution mechanism for the key carbon-carbon bond-forming step. They also elaborate on other mechanistic features that underpin this unique and synthetically challenging post-translational modification.


Assuntos
Proteínas de Bactérias/metabolismo , Lisina/metabolismo , Streptococcus agalactiae/enzimologia , Streptococcus suis/enzimologia , Triptofano/metabolismo , Sequência de Aminoácidos , Proteínas de Bactérias/química , Catálise , Domínio Catalítico , Modelos Químicos , Modelos Moleculares , Conformação Proteica , Precursores de Proteínas/metabolismo
6.
Infect Immun ; 86(3)2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29229728

RESUMO

Clustered regularly interspaced palindromic repeats (CRISPR) and their associated cas genes have been demonstrated to regulate self-genes and virulence in many pathogens. In this study, we found that inactivation of cas9 caused reduced adhesion and intracellular survival of the piscine Streptococcus agalactiae strain GD201008-001 and significantly decreased the virulence of this strain in zebrafish and mice. Further investigation indicated that the regR transcriptional regulator was upregulated in the Δcas9 mutant. As regR mediates the repression of hyaluronidase, a critical factor involved in opening the blood-brain barrier (BBB) in mice, cas9-mediated repression of regR transcription is important for S. agalactiae to open the BBB and thereby cause meningitis in animals. This study expands our understanding of endogenous gene regulation mediated by CRISPR-Cas systems in bacteria.


Assuntos
Proteínas de Bactérias/metabolismo , Endonucleases/metabolismo , Infecções Estreptocócicas/microbiologia , Streptococcus agalactiae/enzimologia , Streptococcus agalactiae/patogenicidade , Fatores de Transcrição/metabolismo , Transcrição Gênica , Animais , Proteínas de Bactérias/genética , Barreira Hematoencefálica/microbiologia , Sistemas CRISPR-Cas , Endonucleases/genética , Feminino , Regulação Bacteriana da Expressão Gênica , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Streptococcus agalactiae/genética , Fatores de Transcrição/genética , Virulência , Peixe-Zebra
7.
Pak J Pharm Sci ; 31(4(Supplementary)): 1597-1602, 2018 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-30058554

RESUMO

Streptokinase (SK) is a fibrinolytic protein used for the treatment of cardiovascular disorders. In the present study, enhanced production of SK was achieved by determining the optimum fermentation conditions for the maximum growth of Streptococcus agalactiae EBL-31 using response surface methodology (RSM). Four process variables (pH, temperature, incubation time and inoculum size) with five levels were evaluated in 30 experimental runs. Central composite rotatable design (CCRD) was employed to predict the effect of independent variables on SK activity. The statistical evaluation by ANOVA showed that the model was fit as the effect of single factors, quadratic effects and most of the interactions among variables. The value ofR2 (0.9988) indicated the satisfactory interaction between the experimental and predicted responses. Furthermore, the model F value (902.67) and coefficient of variation (1.92) clearly showed that the model is significant (p =>0.0001). The functional activity of SK was determined by spectrophotometric analysis and maximum SK production was obtained at pH-7.0, temperature- 37.5oC, an incubation time of 36 hours and 2.5 mL inoculum size. Hence it was concluded that the optimization of culture conditions through RSM increases the production of SK by 2.01-fold. Production of SK by fermentation is an economical choice to be used for the treatment of cardiovascular diseases.


Assuntos
Química Farmacêutica/métodos , Fermentação/fisiologia , Streptococcus agalactiae/enzimologia , Estreptoquinase/biossíntese , Humanos , Streptococcus agalactiae/genética , Estreptoquinase/genética
8.
J Biol Chem ; 290(10): 6281-92, 2015 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-25605731

RESUMO

Glycosaminoglycans in mammalian extracellular matrices are degraded to their constituents, unsaturated uronic (glucuronic/iduronic) acids and amino sugars, through successive reactions of bacterial polysaccharide lyase and unsaturated glucuronyl hydrolase. Genes coding for glycosaminoglycan-acting lyase, unsaturated glucuronyl hydrolase, and the phosphotransferase system are assembled into a cluster in the genome of pathogenic bacteria, such as streptococci and clostridia. Here, we studied the streptococcal metabolic pathway of unsaturated uronic acids and the structure/function relationship of its relevant isomerase and dehydrogenase. Two proteins (gbs1892 and gbs1891) of Streptococcus agalactiae strain NEM316 were overexpressed in Escherichia coli, purified, and characterized. 4-Deoxy-l-threo-5-hexosulose-uronate (Dhu) nonenzymatically generated from unsaturated uronic acids was converted to 2-keto-3-deoxy-d-gluconate via 3-deoxy-d-glycero-2,5-hexodiulosonate through successive reactions of gbs1892 isomerase (DhuI) and gbs1891 NADH-dependent reductase/dehydrogenase (DhuD). DhuI and DhuD enzymatically corresponded to 4-deoxy-l-threo-5-hexosulose-uronate ketol-isomerase (KduI) and 2-keto-3-deoxy-d-gluconate dehydrogenase (KduD), respectively, involved in pectin metabolism, although no or low sequence identity was observed between DhuI and KduI or between DhuD and KduD, respectively. Genes for DhuI and DhuD were found to be included in the streptococcal genetic cluster, whereas KduI and KduD are encoded in clostridia. Tertiary and quaternary structures of DhuI and DhuD were determined by x-ray crystallography. Distinct from KduI ß-barrels, DhuI adopts an α/ß/α-barrel structure as a basic scaffold similar to that of ribose 5-phosphate isomerase. The structure of DhuD is unable to accommodate the substrate/cofactor, suggesting that conformational changes are essential to trigger enzyme catalysis. This is the first report on the bacterial metabolism of glycosaminoglycan-derived unsaturated uronic acids by isomerase and dehydrogenase.


Assuntos
Glicosaminoglicanos/química , Isomerases/química , Oxirredutases/química , Infecções Estreptocócicas/enzimologia , Streptococcus agalactiae/enzimologia , Cristalografia por Raios X , Matriz Extracelular/química , Matriz Extracelular/metabolismo , Glucuronatos/química , Glucuronatos/metabolismo , Glicosaminoglicanos/metabolismo , Ácido Idurônico/química , Ácido Idurônico/metabolismo , Isomerases/metabolismo , Oxirredutases/metabolismo , Infecções Estreptocócicas/patologia , Streptococcus agalactiae/química , Streptococcus agalactiae/patogenicidade , Especificidade por Substrato , Ácidos Urônicos/química , Ácidos Urônicos/metabolismo
9.
FASEB J ; 29(11): 4629-40, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26202865

RESUMO

Group B Streptococcus (GBS) expresses 3 structurally distinct pilus types (1, 2a, and 2b) identified as important virulence factors and vaccine targets. These pili are heterotrimeric polymers, covalently assembled on the cell wall by sortase (Srt) enzymes. We investigated the pilus-2b biogenesis mechanism by using a multidisciplinary approach integrating genetic, biochemical, and structural studies to dissect the role of the 2 pilus-2b-associated Srts. We show that only 1 sortase (SrtC1-2b) is responsible for pilus protein polymerization, whereas the second one (Srt2-2b) does not act as a pilin polymerase, but similarly to the housekeeping class A Srt (SrtA), it is involved in cell-wall pilus anchoring by targeting the minor ancillary subunit. Based on its function and sequence features, Srt2-2b does not belong to class C Srts (SrtCs), nor is it a canonical member of any other known family of Srts. We also report the crystal structure of SrtC1-2b at 1.9 Å resolution. The overall fold resembles the typical structure of SrtCs except for the N-terminal lid region that appears in an open conformation displaced from the active site. Our findings reveal that GBS pilus type 2b biogenesis differs significantly from the current model of pilus assembly in gram-positive pathogens.


Assuntos
Proteínas de Bactérias/química , Cisteína Endopeptidases/química , Fímbrias Bacterianas/enzimologia , Streptococcus agalactiae/enzimologia , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Cristalografia por Raios X , Cisteína Endopeptidases/genética , Cisteína Endopeptidases/metabolismo , Fímbrias Bacterianas/genética , Estrutura Terciária de Proteína , Streptococcus agalactiae/genética
10.
J Bacteriol ; 197(3): 510-7, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25404702

RESUMO

Serine-rich repeat glycoproteins (SRRPs) are highly conserved in streptococci and staphylococci. Glycosylation of SRRPs is important for bacterial adhesion and pathogenesis. Streptococcus agalactiae is the leading cause of bacterial sepsis and meningitis among newborns. Srr2, an SRRP from S. agalactiae strain COH1, has been implicated in bacterial virulence. Four genes (gtfA, gtfB, gtfC, and gtfD) located downstream of srr2 share significant homology with genes involved in glycosylation of other SRRPs. We have shown previously that gtfA and gtfB encode two glycosyltransferases, GtfA and GtfB, that catalyze the transfer of GlcNAc residues to the Srr2 polypeptide. However, the function of other glycosyltransferases in glycosylation of Srr2 is unknown. In this study, we determined that GtfC catalyzed the direct transfer of glucosyl residues to Srr2-GlcNAc. The GtfC crystal structure was solved at 2.7 Å by molecular replacement. Structural analysis revealed a loop region at the N terminus as a putative acceptor substrate binding domain. Deletion of this domain rendered GtfC unable to bind to its substrate Srr2-GlcNAc, concurrently abolished the glycosyltransferase activity of GtfC, and also altered glycosylation of Srr2. Furthermore, deletion of the corresponding regions from GtfC homologs also abolished their substrate binding and enzymatic activity, indicating that this region is functionally conserved. In summary, we have determined that GtfC is important for the glycosylation of Srr2 and identified a conserved loop region that is crucial for acceptor substrate binding from GtfC homologs in streptococci. These findings shed new mechanistic insight into this family of glycosyltransferases.


Assuntos
Proteínas de Bactérias/metabolismo , Glucosiltransferases/metabolismo , Streptococcus agalactiae/enzimologia , Streptococcus agalactiae/metabolismo , Sítios de Ligação , Cristalografia por Raios X , Análise Mutacional de DNA , Glucosiltransferases/química , Glucosiltransferases/genética , Ligação Proteica , Conformação Proteica , Deleção de Sequência , Streptococcus agalactiae/genética
11.
J Biol Chem ; 289(9): 5479-89, 2014 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-24429288

RESUMO

Streptococcus agalactiae (Group B Streptococcus) is a commensal of the human intestine and vagina of adult women but is the leading cause of invasive infection in neonates. This Gram-positive bacterium displays a set of virulence-associated surface proteins involved in the interaction with the host, such as adhesion to host cells, invasion of tissues, or subversion of the immune system. In this study, we characterized a cell wall-localized protein as an ecto-5'-nucleoside diphosphate phosphohydrolase (NudP) involved in the degradation of extracellular nucleotides which are central mediators of the immune response. Biochemical characterization of recombinant NudP revealed a Mn(2+)-dependent ecto-5'-nucleotidase activity on ribo- and deoxyribonucleoside 5'-mono- and 5'-diphosphates with a substrate specificity different from that of known orthologous enzymes. Deletion of the gene coding the housekeeping enzyme sortase A led to the release of NudP into the culture supernatant, confirming that this enzyme is anchored to the cell wall by its non-canonical LPXTN motif. The NudP ecto-5'-nucleotidase activity is reminiscent of the reactions performed by the mammalian ectonucleotidases CD39 and CD73 involved in regulating the extracellular level of ATP and adenosine. We further demonstrated that the absence of NudP activity decreases bacterial survival in mouse blood, a process dependent on extracellular adenosine. In vivo assays in animal models of infection showed that NudP activity is critical for virulence. These results demonstrate that Group B Streptococcus expresses a specific ecto-5'-nucleotidase necessary for its pathogenicity and highlight the diversity of reactions performed by this enzyme family. These results suggest that bacterial pathogens have developed specialized strategies to subvert the mammalian immune response controlled by the extracellular nucleotide signaling pathways.


Assuntos
Adenosina/metabolismo , Viabilidade Microbiana , N-Glicosil Hidrolases/metabolismo , Streptococcus agalactiae/enzimologia , Adenosina/genética , Motivos de Aminoácidos , Animais , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , N-Glicosil Hidrolases/genética , N-Glicosil Hidrolases/imunologia , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/fisiologia , Streptococcus agalactiae/genética , Streptococcus agalactiae/imunologia
12.
J Intern Med ; 278(3): 264-76, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25683265

RESUMO

BACKGROUND: Type 1 narcolepsy is a neurological disorder characterized by excessive daytime sleepiness and cataplexy associated with the HLA allele DQB1*06:02. Genetic predisposition along with external triggering factors may drive autoimmune responses, ultimately leading to the selective loss of hypocretin-positive neurons. OBJECTIVE: The aim of this study was to investigate potential aetiological factors in Swedish cases of postvaccination (Pandemrix) narcolepsy defined by interferon-gamma (IFNγ) production from immune cells in response to molecularly defined targets. METHODS: Cellular reactivity defined by IFNγ production was examined in blood from 38 (HLA-DQB1*06:02(+) ) Pandemrix-vaccinated narcolepsy cases and 76 (23 HLA-DQB1*06:02(+) and 53 HLA-DQB1*06:02(-) ) control subjects, matched for age, sex and exposure, using a variety of different antigens: ß-haemolytic group A streptococcal (GAS) antigens (M5, M6 and streptodornase B), influenza (the pandemic A/H1N1/California/7/09 NYMC X-179A and A/H1N1/California/7/09 NYMC X-181 vaccine antigens, previous Flu-A and -B vaccine targets, A/H1N1/Brisbane/59/2007, A/H1N1/Solomon Islands/3/2006, A/H3N2/Uruguay/716/2007, A/H3N2/Wisconsin/67/2005, A/H5N1/Vietnam/1203/2004 and B/Malaysia/2506/2004), noninfluenza viral targets (CMVpp65, EBNA-1 and EBNA-3) and auto-antigens (hypocretin peptide, Tribbles homolog 2 peptide cocktail and extract from rat hypothalamus tissue). RESULTS: IFN-γ production was significantly increased in whole blood from narcolepsy cases in response to streptococcus serotype M6 (P = 0.0065) and streptodornase B protein (P = 0.0050). T-cell recognition of M6 and streptodornase B was confirmed at the single-cell level by intracellular cytokine (IL-2, IFNγ, tumour necrosis factor-alpha and IL-17) production after stimulation with synthetic M6 or streptodornase B peptides. Significantly, higher (P = 0.02) titres of serum antistreptolysin O were observed in narcolepsy cases, compared to vaccinated controls. CONCLUSION: ß-haemolytic GAS may be involved in triggering autoimmune responses in patients who developed narcolepsy symptoms after vaccination with Pandemrix in Sweden, characterized by a Streptococcus pyogenes M-type-specific IFN-γ cellular immune response.


Assuntos
Narcolepsia/imunologia , Streptococcus agalactiae/imunologia , Estreptodornase e Estreptoquinase/imunologia , Adolescente , Adulto , Idoso , Antiestreptolisina/sangue , Criança , Feminino , Humanos , Interferon gama/biossíntese , Interferon gama/sangue , Masculino , Pessoa de Meia-Idade , Narcolepsia/epidemiologia , Fragmentos de Peptídeos/biossíntese , Fragmentos de Peptídeos/sangue , Sorotipagem , Streptococcus agalactiae/enzimologia , Suécia/epidemiologia
13.
Nature ; 458(7234): 83-6, 2009 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-19262672

RESUMO

Antimicrobial drugs targeting the reportedly essential type II fatty acid synthesis (FASII) pathway have been recently acclaimed for their efficacy against infections caused by multiresistant Gram-positive bacteria. Our findings show that the strategy for antibiotic development based on FASII pathway targets is fundamentally flawed by the fact that exogenous fatty acids fully bypass inhibition of this pathway in both in vitro and in vivo conditions. We demonstrate that major Gram-positive pathogens-such as streptococci, pneumococci, enterococci and staphylococci-overcome drug-induced FASII pathway inhibition when supplied with exogenous fatty acids, and human serum proves to be a highly effective source of fatty acids. For opportunist pathogen Streptococcus agalactiae, growth in serum leads to an overall decrease of FASII gene expression. No antibiotic inhibitor could have a stronger effect than the inactivation of the target gene, so we challenged the role of FASII using deletion mutants. Our results unequivocally show that the FASII target enzymes are dispensable in vivo during S. agalactiae infection. The results of this study largely compromise the use of FASII-based antimicrobials for treating sepsis caused by Gram-positive pathogens.


Assuntos
Antibacterianos/farmacologia , Farmacorresistência Bacteriana , Ácidos Graxos/biossíntese , Bactérias Gram-Positivas/efeitos dos fármacos , Animais , Ácidos Graxos/análise , Ácidos Graxos/química , Ácidos Graxos/farmacologia , Regulação Bacteriana da Expressão Gênica/efeitos dos fármacos , Genes Bacterianos/genética , Bactérias Gram-Positivas/genética , Bactérias Gram-Positivas/metabolismo , Bactérias Gram-Positivas/patogenicidade , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Testes de Sensibilidade Microbiana , Ratos , Ratos Sprague-Dawley , Reprodutibilidade dos Testes , Sepse/tratamento farmacológico , Sepse/microbiologia , Soro/química , Soro/microbiologia , Infecções Estreptocócicas/tratamento farmacológico , Infecções Estreptocócicas/microbiologia , Streptococcus agalactiae/efeitos dos fármacos , Streptococcus agalactiae/enzimologia , Streptococcus agalactiae/genética , Streptococcus agalactiae/metabolismo , Especificidade por Substrato , Virulência/genética
14.
J Dairy Sci ; 98(12): 8560-4, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26409975

RESUMO

The objective of this study was to determine the lipolytic activity on milk fat of 2 bovine mastitis pathogens, that is, Staphylococcus aureus and Streptococcus agalactiae. The lipolytic activity was determined by 2 different techniques, that is, thin-layer chromatography and an extraction-titration method, in an experimental model using the most commonly occurring field strains of the 2 mastitic bacteria isolated from Swedish dairy farms. The microorganisms were inoculated into bacteria-free control milk and incubated at 37°C to reflect physiological temperatures in the mammary gland. Levels of free fatty acids (FFA) were analyzed at time of inoculation (t=0) and after 2 and 6h of incubation, showing significant increase in FFA levels. After 2h the FFA content had increased by approximately 40% in milk samples inoculated with Staph. aureus and Strep. agalactiae, and at 6h the pathogens had increased FFA levels by 47% compared with the bacteria-free control milk. Changes in lipid composition compared with the bacteria-free control were investigated at 2 and 6h of incubation. Diacylglycerols, triacylglycerols, and phospholipids increased significantly after 6h incubation with the mastitis bacteria, whereas cholesterol and sterol esters decreased. Our results suggest that during mammary infections with Staph. aureus and Strep. agalactiae, the action of lipases originating from the mastitis pathogens will contribute significantly to milk fat lipolysis and thus to raw milk deterioration.


Assuntos
Lipólise , Mastite Bovina/microbiologia , Leite/química , Staphylococcus aureus/enzimologia , Streptococcus agalactiae/enzimologia , Animais , Bovinos , Ácidos Graxos não Esterificados/metabolismo , Feminino , Lipase/metabolismo , Lipídeos/análise , Leite/microbiologia , Infecções Estafilocócicas/veterinária , Staphylococcus aureus/isolamento & purificação , Streptococcus agalactiae/isolamento & purificação , Suécia
15.
Genet Mol Res ; 14(2): 6003-17, 2015 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-26125800

RESUMO

Cytidine monophosphate (CMP) N-acetylneuraminic acid (NeuNAc) synthetase, which is encoded by the neuA gene, can catalyze the activation of sialic acid with CMP, and plays an important role in Streptococcus agalactiae infection pathogenesis. To study the structure and function of the S. agalactiae neuA gene, we isolated it from diseased tilapia, amplified it using polymerase chain reaction (PCR) with specific primers, and cloned it into a pMD19-T vector. The recombinant plasmid was confirmed by PCR and restriction enzyme digestion, and identified by sequencing. Molecular characterization analyses of the neuA nucleotide amino acid sequence were performed using bioinformatic tools and an online server. The results showed that the neuA nucleotide sequence contained a complete coding region, which comprised 1242 bp, encoding 413 amino acids (aa). The aa sequence was highly conserved and contained a Glyco_tranf_GTA_type superfamily and an SGNH_hydrolase superfamily conserved domain, which are related to sialic acid activation catalysis. The NeuA protein possessed many important sites related to post-translational modification, including 28 potential phosphorylation sites and 2 potential N-glycosylation sites, had no signal peptides or transmembrane regions, and was predicted to reside in the cytoplasm. Moreover, the protein had some B-cell epitopes, which suggests its potential in development of a vaccine against S. agalactiae infection. The codon usage frequency of neuA differed greatly in Escherichia coli and Homo sapiens genes, and neuA may be more efficiently expressed in eukaryotes (yeast). S. agalactiae neuA from tilapia maintains high structural homology and sequence identity with CMP-NeuNAc synthetases from other bacteria.


Assuntos
Acetiltransferases/genética , Sequência de Aminoácidos , Biologia Computacional , Streptococcus agalactiae/enzimologia , Animais , Clonagem Molecular , Escherichia coli/genética , Proteínas de Escherichia coli/genética , Humanos , Fases de Leitura Aberta , Homologia de Sequência de Aminoácidos , Streptococcus agalactiae/genética , Tilápia/microbiologia
16.
Acta Crystallogr D Biol Crystallogr ; 70(Pt 11): 2937-49, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25372684

RESUMO

The group B pathogen Streptococcus agalactiae commonly populates the human gut and urogenital tract, and is a major cause of infection-based mortality in neonatal infants and in elderly or immunocompromised adults. Nuclease A (GBS_NucA), a secreted DNA/RNA nuclease, serves as a virulence factor for S. agalactiae, facilitating bacterial evasion of the human innate immune response. GBS_NucA efficiently degrades the DNA matrix component of neutrophil extracellular traps (NETs), which attempt to kill and clear invading bacteria during the early stages of infection. In order to better understand the mechanisms of DNA substrate binding and catalysis of GBS_NucA, the high-resolution structure of a catalytically inactive mutant (H148G) was solved by X-ray crystallography. Several mutants on the surface of GBS_NucA which might influence DNA substrate binding and catalysis were generated and evaluated using an imidazole chemical rescue technique. While several of these mutants severely inhibited nuclease activity, two mutants (K146R and Q183A) exhibited significantly increased activity. These structural and biochemical studies have greatly increased our understanding of the mechanism of action of GBS_NucA in bacterial virulence and may serve as a foundation for the structure-based drug design of antibacterial compounds targeted to S. agalactiae.


Assuntos
Proteínas de Bactérias/química , Endonucleases/química , Infecções Estreptocócicas/microbiologia , Streptococcus agalactiae/química , Fatores de Virulência/química , Sequência de Aminoácidos , Proteínas de Bactérias/genética , Cristalografia por Raios X , Endonucleases/genética , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Mutação Puntual , Conformação Proteica , Alinhamento de Sequência , Streptococcus agalactiae/enzimologia , Streptococcus agalactiae/genética , Fatores de Virulência/genética
17.
Mol Microbiol ; 89(3): 518-31, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23772975

RESUMO

Most bacteria of the genus Streptococcus are opportunistic pathogens, and some of them produce extracellular DNases, which may be important for virulence. Genome analyses of Streptococcus agalactiae (GBS) neonate isolate NEM316 revealed the presence of seven genes putatively encoding secreted DNases, although their functions, if any, are unknown. In this study, we observed that respiration growth of GBS led to the extracellular accumulation of a putative nuclease, identified as being encoded by the gbs0661 gene. When overproduced in Lactococcus lactis, the protein was found to be a divalent cation-requiring, pH-stable and heat-stable nuclease that we named Nuclease A (NucA). Substitution of the histidine(148) by alanine reduced nuclease activity of the GBS wild-type strain, indicating that NucA is the major nuclease ex vivo. We determined that GBS is able to degrade the DNA matrix comprising the neutrophil extracellular trap (NET). The nucA(H148A) mutant was impaired for this function, implicating NucA in the virulence of GBS. In vivo infection studies confirmed that NucA is required for full infection, as the mutant strain allowed increased bacterial clearance from lung tissue and decreased mortality in infected mice. These results show that NucA is involved in NET escape and is needed for full virulence.


Assuntos
Proteínas de Bactérias/metabolismo , Desoxirribonucleases/metabolismo , Neutrófilos/imunologia , Infecções Estreptocócicas/imunologia , Streptococcus agalactiae/patogenicidade , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Proteínas de Bactérias/genética , Desoxirribonucleases/genética , Humanos , Evasão da Resposta Imune , Pulmão/microbiologia , Camundongos , Dados de Sequência Molecular , Neutrófilos/microbiologia , Infecções Estreptocócicas/microbiologia , Streptococcus agalactiae/enzimologia , Streptococcus agalactiae/genética , Receptor Toll-Like 9/imunologia , Virulência
18.
Microbiology (Reading) ; 160(Pt 7): 1298-1315, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24760965

RESUMO

We have referenced and described Streptococcus agalactiae transposable elements encoding DDE transposases. These elements belonged to nine families of insertion sequences (ISs) and to a family of conjugative transposons (TnGBSs). An overview of the physiological impact of the insertion of all these elements is provided. DDE-transposable elements affect S. agalactiae in a number of aspects of its capability to adapt to various environments and modulate the expression of several virulence genes, the scpB-lmB genomic region and the genes involved in capsule expression and haemolysin transport being the targets of several different mobile elements. The referenced mobile elements modify S. agalactiae behaviour by transferring new gene(s) to its genome, by modifying the expression of neighbouring genes at the integration site or by promoting genomic rearrangements. Transposition of some of these elements occurs in vivo, suggesting that by dynamically regulating some adaptation and/or virulence genes, they improve the ability of S. agalactiae to reach different niches within its host and ensure the 'success' of the infectious process.


Assuntos
Adaptação Fisiológica , Proteínas de Bactérias/genética , Elementos de DNA Transponíveis/genética , Genoma Bacteriano/genética , Infecções Estreptocócicas/microbiologia , Streptococcus agalactiae/fisiologia , Proteínas de Bactérias/metabolismo , Humanos , Família Multigênica , Streptococcus agalactiae/enzimologia , Streptococcus agalactiae/genética , Transposases/genética , Transposases/metabolismo , Fatores de Virulência/genética , Fatores de Virulência/metabolismo
19.
J Biol Chem ; 287(34): 29035-44, 2012 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-22761440

RESUMO

Phosphoglycerate kinase (PGK), present on the surface of group B streptococcus (GBS), has previously been demonstrated to bind the host proteins actin and plasminogen. The actin and plasminogen binding sites of GBS-PGK were identified using truncated GBS-PGK molecules, followed by peptide mapping. These experiments identified two actin and plasminogen binding sites located between amino acids 126-134 and 204-208 of the 398-amino acid-long GBS-PGK molecule. Substitution of the lysine residues within these regions with alanine resulted in significantly reduced binding to both actin and plasminogen. In addition, conversion of the glutamic acid residue at amino acid 133 to proline, the amino acid found at this position for the PGK protein of Streptococcus pneumoniae, also resulted in significantly reduced binding to actin and plasminogen. These results demonstrate that the lysine residues at amino acid positions 126, 127, 130, 204, and 208 along with the glutamic acid residue at amino acid position 133 are necessary for actin and plasminogen binding by GBS-PGK.


Assuntos
Actinas/química , Proteínas de Bactérias/química , Fosfoglicerato Quinase/química , Plasminogênio/química , Streptococcus agalactiae/enzimologia , Actinas/genética , Actinas/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Humanos , Mapeamento de Peptídeos/métodos , Fosfoglicerato Quinase/genética , Fosfoglicerato Quinase/metabolismo , Plasminogênio/genética , Plasminogênio/metabolismo , Ligação Proteica , Streptococcus agalactiae/genética , Streptococcus pneumoniae/enzimologia , Streptococcus pneumoniae/genética
20.
J Antimicrob Chemother ; 68(7): 1533-6, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23449828

RESUMO

OBJECTIVES: Group B Streptococcus (GBS; Streptococcus agalactiae) has been regarded as uniformly susceptible to penicillins. However, we recently reported the existence of GBS with reduced penicillin susceptibility (PRGBS), with amino acid substitutions in penicillin-binding protein (PBP) 2X. Although most PRGBS show high MICs of ceftizoxime (4-64 mg/L) and cefotaxime (0.12-1 mg/L), those for strain B1 are exceptionally high (ceftizoxime MIC ≥256 mg/L and cefotaxime MIC 2 mg/L). We previously found an amino acid substitution (G539S) neighbouring the conserved K540TG motif in PBP1A in addition to the PRGBS-specific amino acid substitution Q557E in PBP2X of B1. The aim of this study was to reveal the effect of the amino acid substitutions in PBP1A and PBP2X of B1 on the high cephalosporin resistance. METHODS: A ceftizoxime competition assay was performed to reveal the PBPs that are the main targets of ceftizoxime. We generated two allelic exchange mutants from ß-lactam-susceptible GBS BAA-611. BAA-611 (B1PBP2X) contained the PBP2X gene derived from B1 and BAA-611 (B1PBP2X, B1PBP1A) contained both the PBP2X and the PBP1A gene derived from B1. These allelic exchange mutants and strain B1 were subjected to susceptibility testing. RESULTS: The ceftizoxime competition assay revealed that PBP1A and PBP2X were the main targets of ceftizoxime. Although the MICs of ceftizoxime and cefotaxime for BAA-611 (B1PBP2X) were 64 and 0.5 mg/L, respectively, BAA-611 (B1PBP2X, B1PBP1A) showed high cephalosporin resistance (ceftizoxime MIC ≥256 mg/L and cefotaxime MIC 2 mg/L) comparable to B1. CONCLUSIONS: The high cephalosporin resistance of GBS was caused by amino acid substitutions in PBP1A and PBP2X.


Assuntos
Resistência às Cefalosporinas , Proteínas de Ligação às Penicilinas/genética , Proteínas de Ligação às Penicilinas/metabolismo , Peptidil Transferases/genética , Peptidil Transferases/metabolismo , Streptococcus agalactiae/efeitos dos fármacos , Streptococcus agalactiae/enzimologia , Substituição de Aminoácidos , Testes de Sensibilidade Microbiana , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Mutação de Sentido Incorreto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA