Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Bacteriol ; 202(15)2020 07 09.
Artigo em Inglês | MEDLINE | ID: mdl-32457050

RESUMO

In Enterococcus faecalis, the site 2 protease Eep generates sex pheromones, including cAM373. Intriguingly, in Staphylococcus aureus, a peptide similar to cAM373, named cAM373_SA, is produced from the camS gene. Here, we report that the staphylococcal Eep homolog is not only responsible for the production of cAM373_SA but also critical for staphylococcal virulence. As with other Eep proteins, the staphylococcal Eep protein has four transmembrane (TM) domains, with the predicted zinc metalloprotease active site (HEXXH) in the first TM domain. eep deletion reduced the cAM373_SA activity in the culture supernatant to the level of the camS deletion mutant. It also markedly decreased the cAM373 peptide peak in a high-performance liquid chromatography (HPLC) analysis. Proteomics analysis showed that Eep affects the production and/or the release of diverse proteins, including the signal peptidase subunit SpsB and the surface proteins SpA, SasG, and FnbA. eep deletion decreased the adherence of S. aureus to host epithelial cells; however, the adherence of the eep mutant was increased by overexpression of the surface proteins SpA, SasG, and FnbA. eep deletion reduced staphylococcal resistance to killing by human neutrophils as well as survival in a murine model of blood infection. The overexpression of the surface protein SpA in the eep mutant increased bacterial survival in the liver. Our study illustrates that in S. aureus, Eep not only generates cAM373_SA but also contributes to the survival of the bacterial pathogen in the host.IMPORTANCE The emergence of multidrug-resistant Staphylococcus aureus makes the treatment of staphylococcal infections much more difficult. S. aureus can acquire a drug resistance gene from other bacteria, such as Enterococcus faecalis Intriguingly, S. aureus produces a sex pheromone for the E. faecalis plasmid pAM373, raising the possibility that S. aureus actively promotes plasmid conjugation from E. faecalis In this study, we found that the staphylococcal Eep protein is responsible for sex pheromone processing and contributes to the survival of the bacteria in the host. These results will enhance future research on the drug resistance acquisition of S. aureus and can lead to the development of novel antivirulence drugs.


Assuntos
Proteínas de Bactérias/metabolismo , Peptídeo Hidrolases/metabolismo , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/enzimologia , Animais , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Peptídeo Hidrolases/química , Peptídeo Hidrolases/genética , Peptídeos/genética , Peptídeos/metabolismo , Domínios Proteicos , Staphylococcus aureus/genética , Staphylococcus aureus/patogenicidade , Virulência
2.
Infect Immun ; 87(12)2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31548327

RESUMO

Upon microbial infection, host immune cells recognize bacterial cell envelope components through cognate receptors. Although bacterial cell envelope components function as innate immune molecules, the role of the physical state of the bacterial cell envelope (i.e., particulate versus soluble) in host immune activation has not been clearly defined. Here, using two different forms of the staphylococcal cell envelope of Staphylococcus aureus RN4220 and USA300 LAC strains, we provide biochemical and immunological evidence that the particulate state is required for the effective activation of host innate immune responses. In a murine model of peritoneal infection, the particulate form of the staphylococcal cell envelope (PCE) induced the production of chemokine (C-X-C motif) ligand 1 (CXCL1) and CC chemokine ligand 2 (CCL2), the chemotactic cytokines for neutrophils and monocytes, respectively, resulting in a strong influx of the phagocytes into the peritoneal cavity. In contrast, compared with PCE, the soluble form of cell envelope (SCE), which was derived from PCE by treatment with cell wall-hydrolyzing enzymes, showed minimal activity. PCE also induced the secretion of calprotectin (myeloid-related protein 8/14 [MRP8/14] complex), a phagocyte-derived antimicrobial protein, into the peritoneal cavity at a much higher level than did SCE. The injected PCE particles were phagocytosed by the infiltrated neutrophils and monocytes and then delivered to mediastinal draining lymph nodes. More importantly, intraperitoneally (i.p.) injected PCE efficiently protected mice from S. aureus infection, which was abolished by the depletion of either monocytes/macrophages or neutrophils. This study demonstrated that the physical state of bacterial cells is a critical factor for efficient host immune activation and the protection of hosts from staphylococcal infections.


Assuntos
Parede Celular/imunologia , Monócitos/imunologia , Neutrófilos/imunologia , Infecções Estafilocócicas/imunologia , Staphylococcus aureus/imunologia , Animais , Quimiocina CCL2/metabolismo , Quimiocina CXCL1/metabolismo , Feminino , Imunidade Inata/imunologia , Complexo Antígeno L1 Leucocitário/metabolismo , Macrófagos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fagocitose/imunologia , Infecções Estafilocócicas/microbiologia
3.
mBio ; 10(4)2019 07 09.
Artigo em Inglês | MEDLINE | ID: mdl-31289190

RESUMO

Mannitol-1-phosphate dehydrogenase (M1PDH) is a key enzyme in Staphylococcus aureus mannitol metabolism, but its roles in pathophysiological settings have not been established. We performed comprehensive structure-function analysis of M1PDH from S. aureus USA300, a strain of community-associated methicillin-resistant S. aureus, to evaluate its roles in cell viability and virulence under pathophysiological conditions. On the basis of our results, we propose M1PDH as a potential antibacterial target. In vitro cell viability assessment of ΔmtlD knockout and complemented strains confirmed that M1PDH is essential to endure pH, high-salt, and oxidative stress and thus that M1PDH is required for preventing osmotic burst by regulating pressure potential imposed by mannitol. The mouse infection model also verified that M1PDH is essential for bacterial survival during infection. To further support the use of M1PDH as an antibacterial target, we identified dihydrocelastrol (DHCL) as a competitive inhibitor of S. aureus M1PDH (SaM1PDH) and confirmed that DHCL effectively reduces bacterial cell viability during host infection. To explain physiological functions of SaM1PDH at the atomic level, the crystal structure of SaM1PDH was determined at 1.7-Å resolution. Structure-based mutation analyses and DHCL molecular docking to the SaM1PDH active site followed by functional assay identified key residues in the active site and provided the action mechanism of DHCL. Collectively, we propose SaM1PDH as a target for antibiotic development based on its physiological roles with the goals of expanding the repertory of antibiotic targets to fight antimicrobial resistance and providing essential knowledge for developing potent inhibitors of SaM1PDH based on structure-function studies.IMPORTANCE Due to the shortage of effective antibiotics against drug-resistant Staphylococcus aureus, new targets are urgently required to develop next-generation antibiotics. We investigated mannitol-1-phosphate dehydrogenase of S. aureus USA300 (SaM1PDH), a key enzyme regulating intracellular mannitol levels, and explored the possibility of using SaM1PDH as a target for developing antibiotic. Since mannitol is necessary for maintaining the cellular redox and osmotic potential, the homeostatic imbalance caused by treatment with a SaM1PDH inhibitor or knockout of the gene encoding SaM1PDH results in bacterial cell death through oxidative and/or mannitol-dependent cytolysis. We elucidated the molecular mechanism of SaM1PDH and the structural basis of substrate and inhibitor recognition by enzymatic and structural analyses of SaM1PDH. Our results strongly support the concept that targeting of SaM1PDH represents an alternative strategy for developing a new class of antibiotics that cause bacterial cell death not by blocking key cellular machinery but by inducing cytolysis and reducing stress tolerance through inhibition of the mannitol pathway.


Assuntos
Antibacterianos/farmacologia , Manitol/metabolismo , Staphylococcus aureus Resistente à Meticilina/efeitos dos fármacos , Desidrogenase do Álcool de Açúcar/química , Desidrogenase do Álcool de Açúcar/metabolismo , Animais , Feminino , Macrófagos/microbiologia , Masculino , Staphylococcus aureus Resistente à Meticilina/enzimologia , Staphylococcus aureus Resistente à Meticilina/genética , Camundongos , Camundongos Endogâmicos C57BL , Simulação de Acoplamento Molecular , Mutação , Células RAW 264.7 , Infecções Estafilocócicas/microbiologia , Desidrogenase do Álcool de Açúcar/genética , Virulência
4.
Sci Rep ; 8(1): 2521, 2018 02 06.
Artigo em Inglês | MEDLINE | ID: mdl-29410445

RESUMO

In Staphylococcus aureus, an important Gram-positive human pathogen, the SaeRS two-component system is essential for the virulence and a good target for the development of anti-virulence drugs. In this study, we screened 12,200 small molecules for Sae inhibitors and identified two anti-cancer drugs, streptozotocin (STZ) and floxuridine (FU), as lead candidates for anti-virulence drug development against staphylococcal infections. As compared with STZ, FU was more efficient in repressing Sae-regulated promoters and protecting human neutrophils from S. aureus-mediated killing. FU inhibited S. aureus growth effectively whereas STZ did not. Intriguingly, RNA-seq analysis suggests that both compounds inhibit other virulence-regulatory systems such as Agr, ArlRS, and SarA more efficiently than they inhibit the Sae system. Both compounds induced prophages from S. aureus, indicating that they cause DNA damages. Surprisingly, a single administration of the drugs was sufficient to protect mice from staphylococcal intraperitoneal infection. Both compounds showed in vivo efficacy in a murine model of blood infection too. Finally, at the experimental dosage, neither compound showed any noticeable side effects on blood glucose level or blood cell counts. Based on these results, we concluded that STZ and FU are promising candidates for anti-virulence drug development against S. aureus infection.


Assuntos
Antibacterianos/farmacologia , Antineoplásicos/farmacologia , Proteínas de Bactérias/antagonistas & inibidores , Floxuridina/farmacologia , Infecções Estafilocócicas/tratamento farmacológico , Staphylococcus aureus/efeitos dos fármacos , Estreptozocina/farmacologia , Fatores de Transcrição/antagonistas & inibidores , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Contagem de Células Sanguíneas , Glicemia/efeitos dos fármacos , DNA Bacteriano/efeitos dos fármacos , Doxorrubicina/farmacologia , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neutrófilos/citologia , Regiões Promotoras Genéticas , Proteínas Quinases/genética , Proteínas Quinases/metabolismo , Bibliotecas de Moléculas Pequenas/farmacologia , Infecções Estafilocócicas/sangue , Staphylococcus aureus/patogenicidade , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Virulência/efeitos dos fármacos , Fatores de Virulência/genética , Fatores de Virulência/metabolismo
5.
Sci Rep ; 7(1): 8456, 2017 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-28814746

RESUMO

In the Gram-positive pathogen Staphylococcus aureus, the membrane-bound ATP-dependent metalloprotease FtsH plays a critical role in resistance to various stressors. However, the molecular mechanism of the FtsH functions is not known. Here, we identified core FtsH target proteins in S. aureus. In the strains Newman and USA300, the abundance of 33 proteins were altered in both strains, of which 11 were identified as core FtsH substrate protein candidates. In the strain Newman and some other S. aureus strains, the sensor histidine kinase SaeS has an L18P (T53C in saeS) substitution, which transformed the protein into an FtsH substrate. Due to the increase of SaeS L18P in the ftsH mutant, Eap, a sae-regulon protein, was also increased in abundance, causing the Newman-specific cell-aggregation phenotype. Regardless of the strain background, however, the ftsH mutants showed lower virulence and survival in a murine infection model. Our study illustrates the elasticity of the bacterial regulatory network, which can be rewired by a single substitution mutation.


Assuntos
Proteases Dependentes de ATP/genética , Proteínas de Bactérias/genética , Redes Reguladoras de Genes , Histidina Quinase/genética , Mutação Puntual , Staphylococcus aureus/genética , Proteases Dependentes de ATP/metabolismo , Animais , Proteínas de Bactérias/metabolismo , Feminino , Histidina Quinase/metabolismo , Camundongos Endogâmicos BALB C , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/enzimologia , Staphylococcus aureus/patogenicidade , Especificidade por Substrato , Virulência/genética
6.
Proc Natl Acad Sci U S A ; 109(23): 9095-100, 2012 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-22586129

RESUMO

Oxidation sensing and quorum sensing significantly affect bacterial physiology and host-pathogen interactions. However, little attention has been paid to the cross-talk between these two seemingly orthogonal signaling pathways. Here we show that the quorum-sensing agr system has a built-in oxidation-sensing mechanism through an intramolecular disulfide switch possessed by the DNA-binding domain of the response regulator AgrA. Biochemical and mass spectrometric analysis revealed that oxidation induces the intracellular disulfide bond formation between Cys-199 and Cys-228, thus leading to dissociation of AgrA from DNA. Molecular dynamics (MD) simulations suggest that the disulfide bond formation generates a steric clash responsible for the abolished DNA binding of the oxidized AgrA. Mutagenesis studies further established that Cys-199 is crucial for oxidation sensing. The oxidation-sensing role of Cys-199 is further supported by the observation that the mutant Staphylococcus aureus strain expressing AgrAC199S is more susceptible to H(2)O(2) owing to repression of the antioxidant bsaA gene under oxidative stress. Together, our results show that oxidation sensing is a component of the quorum-sensing agr signaling system, which serves as an intrinsic checkpoint to ameliorate the oxidation burden caused by intense metabolic activity and potential host immune response.


Assuntos
Proteínas de Bactérias/metabolismo , Dissulfetos/metabolismo , Regulação Bacteriana da Expressão Gênica/fisiologia , Interações Hospedeiro-Patógeno/fisiologia , Modelos Moleculares , Percepção de Quorum/fisiologia , Staphylococcus aureus/metabolismo , Transativadores/metabolismo , Sequência de Aminoácidos , Cisteína/metabolismo , Regulação Bacteriana da Expressão Gênica/genética , Espectrometria de Massas , Simulação de Dinâmica Molecular , Dados de Sequência Molecular , Mutagênese , Oxirredução , Staphylococcus aureus/fisiologia
7.
J Biol Chem ; 287(25): 21102-9, 2012 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-22553203

RESUMO

As a human pathogen, Staphylococcus aureus must cope with oxidative stress generated by the human immune system. Here, we report that CymR utilizes its sole Cys-25 to sense oxidative stress. Oxidation followed by thiolation of this cysteine residue leads to dissociation of CymR from its cognate promoter DNA. In contrast, the DNA binding of the CymRC25S mutant was insensitive to oxidation and thiolation, suggesting that CymR senses oxidative stress through oxidation of its sole cysteine to form a mixed disulfide with low molecular weight thiols. The determined crystal structures of the reduced and oxidized forms of CymR revealed that Cys-25 is oxidized to Cys-25-SOH in the presence of H(2)O(2). Deletion of cymR reduced the resistance of S. aureus to oxidative stresses, and the resistance was restored by expressing a C25S mutant copy of cymR. In a C25S substitution mutant, the expression of two genes, tcyP and mccB, was constitutively repressed and did not respond to hydrogen peroxide stress, whereas the expression of the genes were highly induced under oxidative stress in a wild-type strain, indicating the critical role of Cys-25 in redox signaling in vivo. Thus, CymR is another master regulator that senses oxidative stress and connects stress responses to virulence regulation in S. aureus.


Assuntos
Proteínas de Bactérias/metabolismo , Proteínas de Ligação a DNA/metabolismo , Estresse Oxidativo/fisiologia , Staphylococcus aureus/metabolismo , Staphylococcus aureus/patogenicidade , Substituição de Aminoácidos , Proteínas de Bactérias/genética , Proteínas de Bactérias/imunologia , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/imunologia , Humanos , Mutação de Sentido Incorreto , Oxirredução , Staphylococcus aureus/genética , Staphylococcus aureus/imunologia , Compostos de Sulfidrila/imunologia , Compostos de Sulfidrila/metabolismo
8.
J Am Chem Soc ; 134(1): 305-14, 2012 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-22122613

RESUMO

Oxygen sensing and redox signaling significantly affect bacterial physiology and host-pathogen interaction. Here we show that a Staphylococcus aureus two-component system, AirSR (anaerobic iron-sulfur cluster-containing redox sensor regulator, formerly YhcSR), responds to oxidation signals (O(2), H(2)O(2), NO, etc) by using a redox-active [2Fe-2S] cluster in the sensor kinase AirS. Mutagenesis studies demonstrate that the [2Fe-2S] cluster is essential for the kinase activity of AirS. We have also discovered that a homologue of IscS (SA1450) in S. aureus is active as a cysteine desulfurase, which enables the in vitro reconstitution of the [2Fe-2S] cluster in AirS. Phosphorylation assays show that the oxidized AirS with a [2Fe-2S](2+) cluster is the fully active form of the kinase but not the apo-AirS nor the reduced AirS possessing a [2Fe-2S](+) cluster. Overoxidation by prolonged exposure to O(2) or contact with H(2)O(2) or NO led to inactivation of AirS. Transcriptome analysis revealed that mutation of airR impacts the expression of ~355 genes under anaerobic conditions. Moreover, the mutant strain displayed increased resistance toward H(2)O(2), vancomycin, norfloxacin, and ciprofloxacin under anaerobic conditions. Together, our results show that S. aureus AirSR is a redox-dependent global regulatory system that plays important roles in gene regulation using a redox active Fe-S cluster under O(2)-limited conditions.


Assuntos
Proteínas de Bactérias/metabolismo , Proteínas Ferro-Enxofre/metabolismo , Oxigênio/metabolismo , Staphylococcus aureus/metabolismo , Aerobiose , Anaerobiose , Farmacorresistência Bacteriana , Meio Ambiente , Peróxido de Hidrogênio/metabolismo , Cinética , Óxido Nítrico/metabolismo , Oxirredução , Análise Espectral , Staphylococcus aureus/efeitos dos fármacos , Staphylococcus aureus/enzimologia
9.
EMBO J ; 27(20): 2656-68, 2008 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-18800056

RESUMO

Surface proteins of Gram-positive bacteria are covalently linked to the cell wall envelope by a mechanism requiring an N-terminal signal peptide and a C-terminal LPXTG motif sorting signal. We show here that surface proteins of Staphylococcus aureus arrive at two distinct destinations in the bacterial envelope, either distributed as a ring surrounding each cell or as discrete assembly sites. Proteins with ring-like distribution (clumping factor A (ClfA), Spa, fibronectin-binding protein B (FnbpB), serine-aspartate repeat protein C (SdrC) and SdrD) harbour signal peptides with a YSIRK/GS motif, whereas proteins directed to discrete assembly sites (S. aureus surface protein A (SasA), SasD, SasF and SasK) do not. Reciprocal exchange of signal peptides between surface proteins with (ClfA) or without the YSIRK/GS motif (SasF) directed recombinant products to the alternate destination, whereas mutations that altered only the YSIRK sequence had no effect. Our observations suggest that S. aureus distinguishes between signal peptides to address proteins to either the cell pole (signal peptides without YSIRK/GS) or the cross wall, the peptidoglycan layer that forms during cell division to separate new daughter cells (signal peptides with YISRK/GS motif).


Assuntos
Sinais Direcionadores de Proteínas/genética , Staphylococcus aureus/metabolismo , Motivos de Aminoácidos , Sequência de Aminoácidos , Coagulase/química , Proteínas de Membrana/metabolismo , Microscopia Confocal , Modelos Biológicos , Dados de Sequência Molecular , Peptídeos/química , Estrutura Terciária de Proteína , Proteínas Recombinantes/química , Homologia de Sequência de Aminoácidos , Propriedades de Superfície , Tripsina/química
10.
RNA ; 14(1): 98-106, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18025251

RESUMO

A2503 in 23S rRNA of the gram-negative bacterium Escherichia coli is located in a functionally important region of the ribosome, at the entrance to the nascent peptide exit tunnel. In E. coli, and likely in other species, this adenosine residue is post-transcriptionally modified to m2A. The enzyme responsible for this modification was previously unknown. We identified E. coli protein YfgB, which belongs to the radical SAM enzyme superfamily, as the methyltransferase that modifies A2503 of 23S rRNA to m2A. Inactivation of the yfgB gene in E. coli led to the loss of modification at nucleotide A2503 of 23S rRNA as revealed by primer extension analysis and thin layer chromatography. The A2503 modification was restored when YfgB protein was expressed in the yfgB knockout strain. A similar protein was shown to catalyze post-transcriptional modification of A2503 in 23S rRNA in gram-positive Staphylococcus aureus. The yfgB knockout strain loses in competition with wild type in a co-growth experiment, indicating functional importance of A2503 modification. The location of A2503 in the exit tunnel suggests its possible involvement in interaction with the nascent peptide and raises the possibility that its post-transcriptional modification may influence such an interaction.


Assuntos
Adenosina/metabolismo , Metilases de Modificação do DNA/metabolismo , Proteínas de Escherichia coli/metabolismo , RNA Bacteriano/metabolismo , RNA Ribossômico 23S/metabolismo , Sequência de Aminoácidos , Sequência de Bases , Metilases de Modificação do DNA/química , Primers do DNA , Escherichia coli/enzimologia , Escherichia coli/genética , Proteínas de Escherichia coli/química , Dados de Sequência Molecular , RNA Bacteriano/química , RNA Ribossômico 23S/química , Homologia de Sequência do Ácido Nucleico , Staphylococcus aureus/genética
11.
Nat Chem Biol ; 2(11): 591-5, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16980961

RESUMO

Staphylococcus aureus is a human pathogen responsible for most wound and hospital-acquired infections. The protein MgrA is both an important virulence determinant during infection and a regulator of antibiotic resistance in S. aureus. The crystal structure of the MgrA homodimer, solved at 2.86 A, indicates the presence of a unique cysteine residue located at the interface of the protein dimer. We discovered that this cysteine residue can be oxidized by various reactive oxygen species, such as hydrogen peroxide and organic hydroperoxide. Cysteine oxidation leads to dissociation of MgrA from DNA and initiation of signaling pathways that turn on antibiotic resistance in S. aureus. The oxidation-sensing mechanism is typically used by bacteria to counter challenges of reactive oxygen and nitrogen species. Our study reveals that in S. aureus, MgrA adopts a similar mechanism but uses it to globally regulate different defensive pathways.


Assuntos
Proteínas de Bactérias/metabolismo , Proteínas Repressoras/metabolismo , Staphylococcus aureus/química , Staphylococcus aureus/metabolismo , Proteínas de Bactérias/química , Cisteína/química , Cisteína/efeitos dos fármacos , Cisteína/metabolismo , Peróxido de Hidrogênio/química , Peróxido de Hidrogênio/farmacologia , Oxirredução , Conformação Proteica , Estrutura Terciária de Proteína , Proteínas Repressoras/química
12.
Mol Microbiol ; 54(2): 520-32, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15469521

RESUMO

The pCF10 plasmid in Enterococcus faecalis transfers from donor cells to recipients upon induction via peptide pheromone. Two plasmid-encoded negative regulators produced from the same transcript, PrgX protein and Qa RNA, repress conjugation genes in uninduced donor cells. PrgX positively autoregulates production of both itself and mature Qa RNA, and is believed to repress the prgQ promoter in a pheromone-sensitive fashion. Previous analysis of PrgX was complicated because mutations in prgX affecting regulation of conjugation also disrupted PrgX autoregulation, suggesting the two functions might be inseparable. In this study, we isolated 14 single amino acid substitutions in PrgX that reduced or eliminated repression of prgQ, without affecting autoregulation or DNA binding. PrgX was shown to bind to its cognate pheromone, cCF10, and most of the mutations lowered the affinity of PrgX for cCF10. Dimerization was affected by five of the mutations and the data indicate that it is required, but insufficient for pheromone induction. We propose a new model for the mechanism used by PrgX for regulation of the prgQ promoter, PrgX autoregulation, and Qa RNA processing.


Assuntos
Proteínas de Bactérias/metabolismo , Conjugação Genética , Enterococcus faecalis/metabolismo , Regulação Bacteriana da Expressão Gênica , Oligopeptídeos/metabolismo , Feromônios/metabolismo , Plasmídeos/metabolismo , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Dimerização , Enterococcus faecalis/genética , Genes Reporter , Oligopeptídeos/genética , Peptídeos/metabolismo , Fenótipo , Feromônios/genética , Plasmídeos/genética , Regiões Promotoras Genéticas , Ligação Proteica , Processamento Pós-Transcricional do RNA , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo
13.
Science ; 305(5690): 1626-8, 2004 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-15361626

RESUMO

Although bacteria use different iron compounds in vitGro, the possibility that microbes distinguish between these iron sources during infection has hitherto not been examined. We applied stable isotope labeling to detect source-specific iron by mass spectrometry and show that Staphylococcus aureus preferentially imports heme iron over transferrin iron. By combining this approach with computational genome analysis, we identified hts (heme transport system), a gene cluster that promotes preferred heme iron import by S. aureus. Heme iron scavenging by means of hts is required for staphylococcal pathogenesis in animal hosts, indicating that heme iron is the preferred iron source during the initiation of infection.


Assuntos
Transportadores de Cassetes de Ligação de ATP/metabolismo , Heme/metabolismo , Ferro/metabolismo , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/metabolismo , Transportadores de Cassetes de Ligação de ATP/genética , Abscesso/microbiologia , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Transporte Biológico , Caenorhabditis elegans/microbiologia , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Membrana Celular/metabolismo , Biologia Computacional , Citoplasma/metabolismo , Genes Bacterianos , Genoma Bacteriano , Hemina/metabolismo , Humanos , Isótopos de Ferro , Rim/microbiologia , Fígado/microbiologia , Espectrometria de Massas , Camundongos , Camundongos Endogâmicos BALB C , Mutagênese Insercional , Óperon , Staphylococcus aureus/genética , Staphylococcus aureus/crescimento & desenvolvimento , Staphylococcus aureus/patogenicidade , Transferrina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA