Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mol Microbiol ; 121(5): 865-881, 2024 05.
Artigo em Inglês | MEDLINE | ID: mdl-38366323

RESUMO

In the human pathogen Staphylococcus aureus, branched-chain fatty acids (BCFAs) are the most abundant fatty acids in membrane phospholipids. Strains deficient for BCFAs synthesis experience auxotrophy in laboratory culture and attenuated virulence during infection. Furthermore, the membrane of S. aureus is among the main targets for antibiotic therapy. Therefore, determining the mechanisms involved in BCFAs synthesis is critical to manage S. aureus infections. Here, we report that the overexpression of SAUSA300_2542 (annotated to encode an acyl-CoA synthetase) restores BCFAs synthesis in strains lacking the canonical biosynthetic pathway catalyzed by the branched-chain α-keto acid dehydrogenase (BKDH) complex. We demonstrate that the acyl-CoA synthetase activity of MbcS activates branched-chain carboxylic acids (BCCAs), and is required by S. aureus to utilize the isoleucine derivative 2-methylbutyraldehyde to restore BCFAs synthesis in S. aureus. Based on the ability of some staphylococci to convert branched-chain aldehydes into their respective BCCAs and our findings demonstrating that branched-chain aldehydes are in fact BCFAs precursors, we propose that MbcS promotes the scavenging of exogenous BCCAs and mediates BCFA synthesis via a de novo alternative pathway.


Assuntos
Aldeídos , Ácidos Carboxílicos , Coenzima A Ligases , Ácidos Graxos , Staphylococcus aureus , Staphylococcus aureus/metabolismo , Staphylococcus aureus/genética , Staphylococcus aureus/enzimologia , Coenzima A Ligases/metabolismo , Coenzima A Ligases/genética , Aldeídos/metabolismo , Ácidos Graxos/metabolismo , Ácidos Graxos/biossíntese , Ácidos Carboxílicos/metabolismo , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/genética , Vias Biossintéticas , Infecções Estafilocócicas/microbiologia , Humanos
2.
Infect Immun ; 91(4): e0004623, 2023 04 18.
Artigo em Inglês | MEDLINE | ID: mdl-36975788

RESUMO

The regulation of membrane protein activity for cellular functions is critically dependent on the composition of phospholipid membranes. Cardiolipin, a unique phospholipid found in bacterial membranes and mitochondrial membranes of eukaryotes, plays a crucial role in stabilizing membrane proteins and maintaining their function. In the human pathogen Staphylococcus aureus, the SaeRS two-component system (TCS) controls the expression of key virulence factors essential for the bacterium's virulence. The SaeS sensor kinase activates the SaeR response regulator via phosphoryl transfer to bind its gene target promoters. In this study, we report that cardiolipin is critical for sustaining the full activity of SaeRS and other TCSs in S. aureus. The sensor kinase protein SaeS binds directly to cardiolipin and phosphatidylglycerol, enabling SaeS activity. Elimination of cardiolipin from the membrane reduces SaeS kinase activity, indicating that bacterial cardiolipin is necessary for modulating the kinase activities of SaeS and other sensor kinases during infection. Moreover, the deletion of cardiolipin synthase genes cls1 and cls2 leads to reduced cytotoxicity to human neutrophils and lower virulence in a mouse model of infection. These findings suggest a model where cardiolipin modulates the kinase activity of SaeS and other sensor kinases after infection to adapt to the hostile environment of the host and expand our knowledge of how phospholipids contribute to membrane protein function.


Assuntos
Cardiolipinas , Fatores de Transcrição , Animais , Camundongos , Humanos , Cardiolipinas/metabolismo , Fatores de Transcrição/genética , Staphylococcus aureus , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Proteínas Quinases/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Regulação Bacteriana da Expressão Gênica
3.
bioRxiv ; 2023 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-36778227

RESUMO

The composition of phospholipid membranes is critical to regulating the activity of membrane proteins for cellular functions. Cardiolipin is a unique phospholipid present within the bacterial membrane and mitochondria of eukaryotes and plays a role in maintaining the function and stabilization of membrane proteins. Here, we report that, in the human pathogen Staphylococcus aureus, cardiolipin is required for full activity of the SaeRS two-component system (TCS). Deletion of the cardiolipin synthase genes, cls1 , and cls2 , reduces the basal activity of SaeRS and other TCSs. Cardiolipin is an indispensable requisite for Sae activation mediated by human neutrophil peptides (HNPs) in the stationary growth phase but not mandatory for Sae induction in the exponential growth phase. Ectopic expression with cls2 , but not with cls1 , in the cls1 cls2 double mutant fully restores Sae activity. Elimination of cardiolipin from the membranes results in decreased kinase activity of the sensor protein SaeS. Purified SaeS protein directly binds to cardiolipin as well as phosphatidylglycerol. A strain lacking cls2 or cls1cls2 renders S. aureus less cytotoxic to human neutrophils and less virulent in a mouse model of infection. Our findings suggest that cardiolipin enables a pathogen to confer virulence by modulating the kinase activity of SaeS and other sensor kinases upon infection.

4.
mBio ; 13(5): e0147222, 2022 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-36135382

RESUMO

Staphylococcus aureus is a ubiquitous Gram-positive bacterium and an opportunistic human pathogen. S. aureus pathogenesis relies on a complex network of regulatory factors that adjust gene expression. Two important factors in this network are CodY, a repressor protein responsive to nutrient availability, and the SaeRS two-component system (TCS), which responds to neutrophil-produced factors. Our previous work revealed that CodY regulates the secretion of many toxins indirectly via Sae through an unknown mechanism. We report that disruption of codY results in increased levels of phosphorylated SaeR (SaeR~P) and that codY mutant cell membranes contain a higher percentage of branched-chain fatty acids (BCFAs) than do wild-type membranes, prompting us to hypothesize that changes to membrane composition modulate the activity of the SaeS sensor kinase. Disrupting the lpdA gene encoding dihydrolipoyl dehydrogenase, which is critical for BCFA synthesis, significantly reduced the abundance of SaeR, phosphorylated SaeR, and BCFAs in the membrane, resulting in reduced toxin production and attenuated virulence. Lower SaeR levels could be explained in part by reduced stability. Sae activity in the lpdA mutant could be complemented genetically and chemically with exogenous short- or full-length BCFAs. Intriguingly, lack of lpdA also alters the activity of other TCSs, suggesting a specific BCFA requirement managing the basal activity of multiple TCSs. These results reveal a novel method of posttranscriptional virulence regulation via BCFA synthesis, potentially linking CodY activity to multiple virulence regulators in S. aureus. IMPORTANCE Two-component systems (TCSs) are an essential way that bacteria sense and respond to their environment. These systems are usually composed of a membrane-bound histidine kinase that phosphorylates a cytoplasmic response regulator. Because most of the histidine kinases are embedded in the membrane, lipids can allosterically regulate the activity of these sensors. In this study, we reveal that branched-chain fatty acids (BCFAs) are required for the activation of multiple TCSs in Staphylococcus aureus. Using both genetic and biochemical data, we show that the activity of the virulence activator SaeS and the phosphorylation of its response regulator SaeR are reduced in a branched-chain keto-acid dehydrogenase complex mutant and that defects in BCFA synthesis have far-reaching consequences for exotoxin secretion and virulence. Finally, we show that mutation of the global nutritional regulator CodY alters BCFA content in the membrane, revealing a potential mechanism of posttranscriptional regulation of the Sae system by CodY.


Assuntos
Infecções Estafilocócicas , Staphylococcus aureus , Humanos , Staphylococcus aureus/metabolismo , Regulação Bacteriana da Expressão Gênica , Histidina Quinase/metabolismo , Di-Hidrolipoamida Desidrogenase/genética , Di-Hidrolipoamida Desidrogenase/metabolismo , Histidina/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Infecções Estafilocócicas/microbiologia , Ácidos Graxos/metabolismo , Exotoxinas/metabolismo
5.
Antibiotics (Basel) ; 10(10)2021 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-34680778

RESUMO

In the Gram-positive pathogen Staphylococcus aureus, FtsH, a membrane-bound metalloprotease, plays a critical role in bacterial virulence and stress resistance. This protease is also known to sensitize methicillin-resistant Staphylococcus aureus (MRSA) to ß-lactam antibiotics; however, the molecular mechanism is not known. Here, by the analysis of FtsH substrate mutants, we found that FtsH sensitizes MRSA specifically to ß-lactams by degrading YpfP, the enzyme synthesizing the anchor molecule for lipoteichoic acid (LTA). Both the overexpression of FtsH and the disruption of ypfP-sensitized MRSA to ß-lactams were observed. The knockout mutation in ftsH and ypfP increased the thickness of the cell wall. The ß-lactam sensitization coincided with the production of aberrantly large LTA molecules. The combination of three mutations in the rpoC, vraB, and SAUSA300_2133 genes blocked the ß-lactam-sensitizing effect of FtsH. Murine infection with the ypfP mutant could be treated by oxacillin, a ß-lactam antibiotic ineffective against MRSA; however, the effective concentration of oxacillin differed depending on the S. aureus strain. Our study demonstrated that the ß-lactam sensitizing effect of FtsH is due to its digestion of YpfP. It also suggests that the larger LTA molecules are responsible for the ß-lactam sensitization phenotype, and YpfP is a viable target for developing novel anti-MRSA drugs.

6.
Sci Rep ; 10(1): 17555, 2020 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-33046771

RESUMO

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

7.
J Bacteriol ; 202(9)2020 04 09.
Artigo em Inglês | MEDLINE | ID: mdl-32094161

RESUMO

In bacteria, chromosomal DNA resides in the cytoplasm, and most transcription factors are also found in the cytoplasm. However, some transcription factors, called membrane-bound transcription factors (MTFs), reside in the cytoplasmic membrane. Here, we report the identification of a new MTF in the Gram-positive pathogen Staphylococcus aureus and its regulation by the protease FtsH. The MTF, named MbtS (membrane-bound transcription factor of Staphylococcus aureus), is encoded by SAUSA300_2640 and predicted to have an N-terminal DNA binding domain and three transmembrane helices. The MbtS protein was degraded by membrane vesicles containing FtsH or by the purified FtsH. MbtS bound to an inverted repeat sequence in its promoter region, and the DNA binding was essential for its transcription. Transcriptional comparison between the ftsH deletion mutant and the ftsH mbtS double mutant showed that MbtS could alter the transcription of over 200 genes. Although the MbtS protein was not detected in wild-type (WT) cells grown in a liquid medium, the protein was detected in some isolated colonies on an agar plate. In a murine model of a skin infection, the disruption of mbtS increased the lesion size. Based on these results, we concluded that MbtS is a new S. aureus MTF whose activity is proteolytically regulated by FtsH.IMPORTANCEStaphylococcus aureus is an important pathogenic bacterium causing various diseases in humans. In the bacterium, transcription is typically regulated by the transcription factors located in the cytoplasm. In this study, we report an atypical transcription factor identified in S. aureus Unlike most other transcription factors, the newly identified transcription factor is located in the cytoplasmic membrane, and its activity is proteolytically controlled by the membrane-bound AAA+ protease FtsH. The newly identified MTF, named MbtS, has the potential to regulate the transcription of over 200 genes. This study provides a molecular mechanism by which a protease affects bacterial transcription and illustrates the diversity of the bacterial transcriptional regulation.


Assuntos
ATPases Associadas a Diversas Atividades Celulares/metabolismo , Proteínas de Bactérias/metabolismo , Membrana Celular/enzimologia , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/enzimologia , Fatores de Transcrição/metabolismo , ATPases Associadas a Diversas Atividades Celulares/genética , Animais , Proteínas de Bactérias/genética , Membrana Celular/genética , Membrana Celular/metabolismo , Feminino , Regulação Bacteriana da Expressão Gênica , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteólise , Staphylococcus aureus/genética , Fatores de Transcrição/genética
8.
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
9.
J Med Chem ; 61(23): 10473-10487, 2018 12 13.
Artigo em Inglês | MEDLINE | ID: mdl-30388007

RESUMO

As an alternative strategy to fight antibiotic resistance, two-component systems (TCSs) have emerged as novel targets. Among TCSs, master virulence regulators that control the expression of multiple virulence factors are considered as excellent antivirulence targets. In Staphylococcus aureus, virulence factor expression is tightly regulated by a few master regulators, including the SaeRS TCS. In this study, we used a SaeRS GFP-reporter system to screen natural compound inhibitors of SaeRS, and identified xanthoangelol B 1, a prenylated chalcone from Angelica keiskei as a hit. We have synthesized 1 and its derivative PM-56 and shown that 1 and PM-56 both had excellent inhibitory potency against the SaeRS TCS, as demonstrated by various in vitro and in vivo experiments. As a mode of action, 1 and PM-56 were shown to bind directly to SaeS and inhibit its histidine kinase activity, which suggests a possibility of a broad spectrum inhibitor of histidine kinases.


Assuntos
Chalcona/análogos & derivados , Desenho de Fármacos , Staphylococcus aureus/efeitos dos fármacos , Staphylococcus aureus/metabolismo , Fatores de Virulência/biossíntese , Antibacterianos/síntese química , Antibacterianos/química , Antibacterianos/farmacologia , Chalcona/síntese química , Chalcona/química , Chalcona/farmacologia , Técnicas de Química Sintética
10.
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
11.
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
12.
Artigo em Inglês | MEDLINE | ID: mdl-28555174

RESUMO

Biofilm causes hospital-associated infections on indwelling medical devices. In Staphylococcus aureus, Biofilm formation is controlled by intricately coordinated network of regulating systems, of which the ATP-dependent protease ClpP shows an inhibitory effect. Here, we demonstrate that the inhibitory effect of ClpP on biofilm formation is through Agr and the cell wall hydrolase Sle1. Biofilm formed by clpP mutant consists of proteins and extracellular DNA (eDNA). The increase of the protein was, at least in part, due to the reduced protease activity of the mutant, which was caused by the decreased activity of agr. On the other hand, the increase of eDNA was due to increased cell lysis caused by the higher level of Sle1. Indeed, as compared with wild type, the clpP mutant excreted an increased level of eDNA, and showed higher sensitivity to Triton-induced autolysis. The deletion of sle1 in the clpP mutant decreased the biofilm formation, the level of eDNA, and the Triton-induced autolysis to wild-type levels. Despite the increased biofilm formation capability, however, the clpP mutant showed significantly reduced virulence in a murine model of subcutaneous foreign body infection, indicating that the increased biofilm formation capability cannot compensate for the intrinsic functions of ClpP during infection.


Assuntos
Proteínas de Bactérias/efeitos dos fármacos , Biofilmes/efeitos dos fármacos , Parede Celular/efeitos dos fármacos , Endopeptidase Clp/antagonistas & inibidores , Regulação Bacteriana da Expressão Gênica/efeitos dos fármacos , Hidrolases/efeitos dos fármacos , Staphylococcus aureus/efeitos dos fármacos , Transativadores/efeitos dos fármacos , Animais , Autólise , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Parede Celular/metabolismo , Modelos Animais de Doenças , Endopeptidase Clp/genética , Endopeptidase Clp/fisiologia , Genes Bacterianos/genética , Hidrolases/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Mutação , Staphylococcus aureus/patogenicidade , Staphylococcus aureus/fisiologia , Transativadores/metabolismo , Virulência/genética
13.
J Am Chem Soc ; 139(10): 3790-3795, 2017 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-28218837

RESUMO

Staphylococcus aureus, a major human pathogen, has been the cause of serious infectious diseases with a high mortality rate. Although genetics is a key means to study S. aureus physiology, such as drug resistance and pathogenesis, genetic manipulation in S. aureus is always time-consuming and labor-intensive. Here we report a CRISPR/Cas9 system (pCasSA) for rapid and efficient genome editing, including gene deletion, insertion, and single-base substitution mutation in S. aureus. The designed pCasSA system is amenable to the assembly of spacers and repair arms by Golden Gate assembly and Gibson assembly, respectively, enabling rapid construction of the plasmids for editing. We further engineered the pCasSA system to be an efficient transcription inhibition system for gene knockdown and possible genome-wide screening. The development of the CRISPR/Cas9-mediated genome editing and transcription inhibition tools will dramatically accelerate drug-target exploration and drug development.


Assuntos
Sistemas CRISPR-Cas/genética , Staphylococcus aureus/genética , Edição de Genes , Engenharia Genética , Mutação , Staphylococcus aureus/efeitos dos fármacos , Transcrição Gênica/efeitos dos fármacos , Transcrição Gênica/genética
15.
Genes (Basel) ; 7(10)2016 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-27706107

RESUMO

In the Gram-positive pathogenic bacterium Staphylococcus aureus, the SaeRS twocomponent system (TCS) plays a major role in controlling the production of over 20 virulence factors including hemolysins, leukocidins, superantigens, surface proteins, and proteases. The SaeRS TCS is composed of the sensor histidine kinase SaeS, response regulator SaeR, and two auxiliary proteins SaeP and SaeQ. Since its discovery in 1994, the sae locus has been studied extensively, and its contributions to staphylococcal virulence and pathogenesis have been well documented and understood; however, the molecular mechanism by which the SaeRS TCS receives and processes cognate signals is not. In this article, therefore, we review the literature focusing on the signaling mechanism and its interaction with other global regulators.

16.
Nat Commun ; 7: 11000, 2016 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-26987594

RESUMO

Most low GC Gram-positive bacteria possess an essential walKR two-component system (TCS) for signal transduction involved in regulating cell wall homoeostasis. Despite the well-established intracellular regulatory mechanism, the role of this TCS in extracellular signal recognition and factors that modulate the activity of this TCS remain largely unknown. Here we identify the extracellular receptor of the kinase 'WalK' (erWalK) as a key hub for bridging extracellular signal input and intracellular kinase activity modulation in Staphylococcus aureus. Characterization of the crystal structure of erWalK revealed a canonical Per-Arnt-Sim (PAS) domain for signal sensing. Single amino-acid mutation of potential signal-transduction residues resulted in severely impaired function of WalKR. A small molecule derived from structure-based virtual screening against erWalK is capable of selectively activating the walKR TCS. The molecular level characterization of erWalK will not only facilitate exploration of natural signal(s) but also provide a template for rational design of erWalK inhibitors.


Assuntos
Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Transdução de Sinais , Staphylococcus aureus/metabolismo , Benzofenonas/farmacologia , Cristalografia por Raios X , Regulação Bacteriana da Expressão Gênica/efeitos dos fármacos , Proteínas Mutantes/metabolismo , Mutação/genética , Estrutura Terciária de Proteína , Transdução de Sinais/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas/farmacologia , Staphylococcus aureus/genética , Staphylococcus aureus/ultraestrutura , Transcrição Gênica/efeitos dos fármacos
17.
Antimicrob Agents Chemother ; 60(4): 2222-31, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26810659

RESUMO

Cationic antimicrobial peptides (CAMPs) are an essential part of the innate immune system. Some Gram-negative enteric pathogens, such asSalmonella enterica, show intrinsic resistance to CAMPs. However, the molecular basis of intrinsic resistance is poorly understood, largely due to a lack of information about the genes involved. In this study, using a microarray-based genomic technique, we screened the Keio collection of 3,985Escherichia colimutants for altered susceptibility to human neutrophil peptide 1 (HNP-1) and identifiedenvCandzapBas novel genetic determinants of intrinsic CAMP resistance. In CAMP killing assays, anE. coliΔenvCEcor ΔzapBEcmutant displayed a distinct profile of increased susceptibility to both LL-37 and HNP-1. Both mutants, however, displayed wild-type resistance to polymyxin B and human ß-defensin 3 (HBD3), suggesting that the intrinsic resistance mediated by EnvC or ZapB is specific to certain CAMPs. A correspondingSalmonellaΔenvCSemutant showed similarly increased CAMP susceptibility. TheenvCmutants of bothE. coliandS. entericadisplayed increased surface negativity and hydrophobicity, which partly explained the increased CAMP susceptibility. However, the ΔenvCEcmutant, but not the ΔenvCSemutant, was defective in outer membrane permeability, excluding this defect as a common factor contributing to the increased CAMP susceptibility. Animal experiments showed that theSalmonellaΔenvCSemutant had attenuated virulence. Taken together, our results indicate that the role ofenvCin intrinsic CAMP resistance is likely conserved among Gram-negative enteric bacteria, demonstrate the importance of intrinsic CAMP resistance for full virulence ofS. enterica, and provide insight into distinct mechanisms of action of CAMPs.


Assuntos
Antibacterianos/farmacologia , Proteínas de Bactérias/biossíntese , Farmacorresistência Bacteriana/genética , Endopeptidases/biossíntese , Salmonella enterica/efeitos dos fármacos , alfa-Defensinas/farmacologia , Peptídeos Catiônicos Antimicrobianos/farmacologia , Proteínas de Bactérias/genética , Proteínas de Bactérias/isolamento & purificação , Proteínas de Ciclo Celular , Permeabilidade da Membrana Celular , Endopeptidases/genética , Endopeptidases/isolamento & purificação , Escherichia coli/efeitos dos fármacos , Escherichia coli/genética , Escherichia coli/metabolismo , Escherichia coli/patogenicidade , Expressão Gênica , Interações Hidrofóbicas e Hidrofílicas , Testes de Sensibilidade Microbiana , Mutação , Polimixina B/farmacologia , Salmonella enterica/genética , Salmonella enterica/metabolismo , Salmonella enterica/patogenicidade , Eletricidade Estática , Virulência , beta-Defensinas/farmacologia , Catelicidinas
18.
PLoS Pathog ; 11(7): e1005026, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26147796

RESUMO

Calprotectin, the most abundant cytoplasmic protein in neutrophils, suppresses the growth of Staphylococcus aureus by sequestering the nutrient metal ions Zn and Mn. Here we show that calprotectin can also enhance the activity of the SaeRS two component system (TCS), a signaling system essential for production of over 20 virulence factors in S. aureus. The activity of the SaeRS TCS is repressed by certain divalent ions found in blood or neutrophil granules; however, the Zn bound-form of calprotectin relieves this repression. During staphylococcal encounter with murine neutrophils or staphylococcal infection of the murine peritoneal cavity, calprotectin increases the activity of the SaeRS TCS as well as the production of proinflammatory cytokines such as IL-1ß and TNF-α, resulting in higher murine mortality. These results suggest that, under certain conditions, calprotectin can be exploited by S. aureus to increase bacterial virulence and host mortality.


Assuntos
Interações Hospedeiro-Parasita/fisiologia , Complexo Antígeno L1 Leucocitário/imunologia , Proteínas Quinases/imunologia , Infecções Estafilocócicas/imunologia , Animais , Proteínas de Bactérias , Western Blotting , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Complexo Antígeno L1 Leucocitário/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Microscopia de Fluorescência , Reação em Cadeia da Polimerase , Proteínas Quinases/metabolismo , Infecções Estafilocócicas/metabolismo , Staphylococcus aureus/imunologia , Staphylococcus aureus/metabolismo , Staphylococcus aureus/patogenicidade , Virulência/fisiologia
19.
PLoS Pathog ; 11(4): e1004799, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25849574

RESUMO

Bacterial pathogens often employ two-component systems (TCSs), typically consisting of a sensor kinase and a response regulator, to control expression of a set of virulence genes in response to changing host environments. In Staphylococcus aureus, the SaeRS TCS is essential for in vivo survival of the bacterium. The intramembrane-sensing histidine kinase SaeS contains, along with a C-terminal kinase domain, a simple N-terminal domain composed of two transmembrane helices and a nine amino acid-long extracytoplasmic linker peptide. As a molecular switch, SaeS maintains low but significant basal kinase activity and increases its kinase activity in response to inducing signals such as human neutrophil peptide 1 (HNP1). Here we show that the linker peptide of SaeS controls SaeS's basal kinase activity and that the amino acid sequence of the linker peptide is highly optimized for its function. Without the linker peptide, SaeS displays aberrantly elevated kinase activity even in the absence of the inducing signal, and does not respond to HNP1. Moreover, SaeS variants with alanine substitution of the linker peptide amino acids exhibit altered basal kinase activity and/or irresponsiveness to HNP1. Biochemical assays reveal that those SaeS variants have altered autokinase and phosphotransferase activities. Finally, animal experiments demonstrate that the linker peptide-mediated fine tuning of SaeS kinase activity is critical for survival of the pathogen. Our results indicate that the function of the linker peptide in SaeS is a highly evolved feature with very optimized amino acid sequences, and we propose that, in other SaeS-like intramembrane sensing histidine kinases, the extracytoplasmic linker peptides actively fine-control their kinases.


Assuntos
Proteínas Quinases/metabolismo , Infecções Estafilocócicas/metabolismo , Staphylococcus aureus/patogenicidade , Sequência de Aminoácidos , Animais , Proteínas de Bactérias , Western Blotting , Citometria de Fluxo , Regulação Bacteriana da Expressão Gênica/fisiologia , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Neutrófilos/microbiologia , Reação em Cadeia da Polimerase , Proteínas Quinases/genética , Infecções Estafilocócicas/genética , Staphylococcus aureus/genética , Virulência/fisiologia
20.
mBio ; 5(5): e01485-14, 2014 Oct 07.
Artigo em Inglês | MEDLINE | ID: mdl-25293763

RESUMO

The histone-like nucleoid-structuring (H-NS) protein binds to horizontally acquired genes in the bacterium Salmonella enterica serovar Typhimurium, silencing their expression. We now report that overcoming the silencing effects of H-NS imposes a delay in the expression of genes activated by the transcriptional regulator PhoP. We determine that PhoP-activated genes ancestral to Salmonella are expressed before those acquired horizontally. This expression timing reflects the in vivo occupancy of the corresponding promoters by the PhoP protein. These results are surprising because some of these horizontally acquired genes reached higher mRNA levels than ancestral genes expressed earlier and were transcribed from promoters harboring PhoP-binding sites with higher in vitro affinity for the PhoP protein. Our findings challenge the often-made assumption that for genes coregulated by a given transcription factor, early genes are transcribed to higher mRNA levels than those transcribed at later times. Moreover, they provide a singular example of how gene ancestry can impact expression timing. Importance: We report that gene ancestry dictates the expression behavior of genes under the direct control of the Salmonella transcriptional regulator PhoP. That is, ancestral genes are transcribed before horizontally acquired genes. This reflects both the need to overcome silencing by the H-NS protein of the latter genes and the architecture of the corresponding promoters. Unexpectedly, transcription levels do not reflect transcription timing. Our results illustrate how a bacterium can exhibit an elaborate temporal expression behavior among genes coregulated by a transcription factor even though the products encoded by the target genes do not participate in a morphological or developmental pathway.


Assuntos
Proteínas de Bactérias/metabolismo , Proteínas de Ligação a DNA/metabolismo , Regulação Bacteriana da Expressão Gênica , Salmonella typhimurium/enzimologia , Salmonella typhimurium/genética , Transcrição Gênica , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...