Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Infect Immun ; 91(4): e0004623, 2023 04 18.
Artículo en Inglés | MEDLINE | ID: mdl-36975788

RESUMEN

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.


Asunto(s)
Cardiolipinas , Factores de Transcripción , Animales , Ratones , Humanos , Cardiolipinas/metabolismo , Factores de Transcripción/genética , Staphylococcus aureus , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Proteínas Quinasas/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Regulación Bacteriana de la Expresión Génica
2.
Mol Microbiol ; 117(5): 1196-1212, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35366366

RESUMEN

Staphylococcus aureus is a Gram-positive commensal that can also cause a variety of infections in humans. S. aureus virulence factor gene expression is under tight control by a complex regulatory network, which includes, sigma factors, sRNAs, and two-component systems (TCS). Previous work in our laboratory demonstrated that overexpression of the sRNA tsr37 leads to an increase in bacterial aggregation. Here, we demonstrate that the clumping phenotype is dependent on a previously unannotated 88 amino acid protein encoded within the tsr37 sRNA transcript (which we named ScrA for S. aureus clumping regulator A). To investigate the mechanism of action of ScrA we performed proteomics and transcriptomics in a ScrA overexpressing strain and show that a number of surface adhesins are upregulated, while secreted proteases are downregulated. Results also showed upregulation of the SaeRS TCS, suggesting that ScrA is influencing SaeRS activity. Overexpression of ScrA in a saeR mutant abrogates the clumping phenotype confirming that ScrA functions via the Sae system. Finally, we identified the ArlRS TCS as a positive regulator of scrA expression. Collectively, our results show that ScrA is an activator of the SaeRS system and suggests that ScrA may act as an intermediary between the ArlRS and SaeRS systems.


Asunto(s)
ARN Pequeño no Traducido , Infecciones Estafilocócicas , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Expresión Génica , Regulación Bacteriana de la Expresión Génica/genética , Humanos , Proteínas Quinasas/metabolismo , ARN Pequeño no Traducido/metabolismo , Staphylococcus aureus/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Virulencia/genética
3.
Appl Environ Microbiol ; 89(11): e0097823, 2023 11 29.
Artículo en Inglés | MEDLINE | ID: mdl-37889047

RESUMEN

IMPORTANCE: Staphylococcus aureus is a formidable pathogen responsible for a wide range of infections, and the emergence of antibiotic-resistant strains has posed significant challenges in treating these infections. In this study, we have established a novel dual reporter system capable of concurrently monitoring the activities of two critical virulence regulators in S. aureus. By incorporating both reporters into a single screening platform, we provide a time- and cost-efficient approach for assessing the activity of compounds against two distinct targets in a single screening round. This innovative dual reporter system presents a promising strategy for the identification of molecules capable of modulating virulence gene expression in S. aureus, potentially expediting the development of antivirulence therapies.


Asunto(s)
Infecciones Estafilocócicas , Staphylococcus aureus , Humanos , Staphylococcus aureus/metabolismo , Factores de Virulencia/genética , Proteínas Bacterianas/metabolismo , Virulencia , Colorantes/metabolismo , Regulación Bacteriana de la Expresión Génica
4.
Mol Microbiol ; 116(5): 1378-1391, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34626146

RESUMEN

In Staphylococcus aureus, the two-component system SaeRS is responsible for regulating various virulence factors essential for the success of this pathogen. SaeRS can be stimulated by neutrophil-derived products but has also recently been shown to be inactivated by the presence of free fatty acids. A mechanism for how fatty acids negatively impacts SaeRS has not been described. We found that unsaturated fatty acids, as well as fatty acids not commonly found in Staphylococcal membranes, prevent the activation of SaeRS at a lower concentration than their saturated counterparts. These fatty acids can negatively impact SaeRS without altering the respiratory capacity of the bacterium. To uncover a potential mechanism for how fatty acids impact SaeRS function/activity, we utilized a naturally occurring point mutation found in S. aureus as well as chimeric SaeS proteins. Using these tools, we identified that the native transmembrane domains of SaeS dictate the transcriptional response to fatty acids in S. aureus. Our data support a model where free fatty acids alter the activity of the two-component system SaeRS directly through the sensor kinase SaeS and is dependent on the transmembrane domains of the protein.


Asunto(s)
Proteínas Bacterianas/efectos de los fármacos , Proteínas Bacterianas/metabolismo , Ácidos Grasos/farmacología , Proteínas Quinasas/efectos de los fármacos , Proteínas Quinasas/metabolismo , Staphylococcus aureus/metabolismo , Factores de Transcripción/efectos de los fármacos , Factores de Transcripción/metabolismo , Regulación Bacteriana de la Expresión Génica , Humanos , Respiración , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/enzimología , Staphylococcus aureus/patogenicidad , Virulencia
5.
Protein Expr Purif ; 177: 105765, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32987120

RESUMEN

The SaeRS two-component system in Staphylococcus aureus controls the expression of a series of virulence factors, such as hemolysins, proteases, and coagulase. The response regulator, SaeR, belongs to the OmpR family with an N-terminal regulatory domain and a C-terminal DNA binding domain. To improve the production and stability of the recombinant protein SaeR, l-arginine (L-Arg) was added to the purification buffers. L-Arg enhanced the solubility and stability of the recombinant protein SaeR. The thermal denaturation temperature of SaeR in 10 mM L-Arg buffer was significantly increased compared to the buffer without L-Arg. Microscale Thermophoresis (MST) analysis results showed that the SaeR protein could bind to the P1 promoter under both phosphorylated and non-phosphorylated status in buffer containing 10 mM L-Arg. These results illustrate an effective method to purify SaeR and other proteins.


Asunto(s)
Arginina/química , Proteínas Bacterianas/genética , ADN Bacteriano/genética , Regulación Bacteriana de la Expresión Génica , Proteínas Quinasas/genética , Staphylococcus aureus/genética , Factores de Transcripción/genética , Proteínas Bacterianas/metabolismo , Clonación Molecular , ADN Bacteriano/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Expresión Génica , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Fosforilación , Regiones Promotoras Genéticas , Unión Proteica , Desnaturalización Proteica , Dominios Proteicos , Proteínas Quinasas/metabolismo , Estabilidad Proteica , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Solubilidad , Staphylococcus aureus/metabolismo , Factores de Transcripción/metabolismo
6.
J Bacteriol ; 199(5)2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-28031278

RESUMEN

In previous studies, we identified the fatty acid kinase virulence factor regulator B (VfrB) as a potent regulator of α-hemolysin and other virulence factors in Staphylococcus aureus In this study, we demonstrated that VfrB is a positive activator of the SaeRS two-component regulatory system. Analysis of vfrB, saeR, and saeS mutant strains revealed that VfrB functions in the same pathway as SaeRS. At the transcriptional level, the promoter activities of SaeRS class I (coa) and class II (hla) target genes were downregulated during the exponential growth phase in the vfrB mutant, compared to the wild-type strain. In addition, saePQRS expression was decreased in the vfrB mutant strain, demonstrating a need for this protein in the autoregulation of SaeRS. The requirement for VfrB-mediated activation was circumvented when SaeS was constitutively active due to an SaeS (L18P) substitution. Furthermore, activation of SaeS via human neutrophil peptide 1 (HNP-1) overcame the dependence on VfrB for transcription from class I Sae promoters. Consistent with the role of VfrB in fatty acid metabolism, hla expression was decreased in the vfrB mutant with the addition of exogenous myristic acid. Lastly, we determined that aspartic acid residues D38 and D40, which are predicted to be key to VfrB enzymatic activity, were required for VfrB-mediated α-hemolysin production. Collectively, this study implicates VfrB as a novel accessory protein needed for the activation of SaeRS in S. aureusIMPORTANCE The SaeRS two-component system is a key regulator of virulence determinant production in Staphylococcus aureus Although the regulon of this two-component system is well characterized, the activation mechanisms, including the specific signaling molecules, remain elusive. Elucidating the complex regulatory circuit of SaeRS regulation is important for understanding how the system contributes to disease causation by this pathogen. To this end, we have identified the fatty acid kinase VfrB as a positive regulatory modulator of SaeRS-mediated transcription of virulence factors in S. aureus In addition to describing a new regulatory aspect of SaeRS, this study establishes a link between fatty acid kinase activity and virulence factor regulation.


Asunto(s)
Proteínas Bacterianas/metabolismo , Regulación Bacteriana de la Expresión Génica/fisiología , Proteínas Quinasas/metabolismo , Staphylococcus aureus/metabolismo , Secuencia de Aminoácidos , Proteínas Bacterianas/genética , Secuencia Conservada , Proteínas Quinasas/genética , Staphylococcus aureus/genética , Factores de Transcripción
7.
Int J Med Microbiol ; 306(6): 367-80, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27265234

RESUMEN

ST30 (CC30)-SCCmec IV (USA1100) is one of the most common community-acquired methicillin-resistant Staphylococcus aureus (CA-MRSA) lineages. ST30 isolates typically carry lukSF-PV genes encoding the Panton-Valentine leukocidin (PVL) and are responsible for outbreaks of invasive infections worldwide. In this study, twenty CC30 isolates were analyzed. All were very susceptible to non-ß-lactam antimicrobials, 18/20 harbored the lukSF-PV genes, only 1/20 exhibited agr-rnaIII dysfunction, and the majority was not able to form biofilm on inert surfaces. Analysis of lukSF-PV temporal regulation revealed that opposite to other CA-MRSA isolates, these genes were more highly expressed in early log phase than in stationary phase. This inverted lukSF-PV temporal expression was associated with a similar pattern of saeRS expression in the ST30 isolates, namely high level expression in log phase and reduced expression in stationary phase. Reduced saeRS expression in stationary phase was associated with low expression levels of the sae regulators, agr and agr-upregulator sarA, which activate the stationary phase sae-P1 promoter and overexpression of agr-RNAIII restored the levels of saeR and lukSF-PV trancripts in stationary phase. Altered SaeRS activity in the ST30 isolates was attributed to amino acid substitutions (N227S, E268K and S351T) in the HTPase_c domain of SaeS (termed SaeS(SKT)). Complementation of a USA300 saeS mutant with the saeS(SKT) and saeS alleles under the direction of the log phase sae-P3 promoter revealed that saeR and lukSF-PV transcription levels were more significantly activated by saeS(SKT) than saeS. In summary our data identify a unique saeS allele (saeS(SKT)) which appears to override cell-density dependent SaeR and PVL expression in ST30 CA-MRSA isolates. Further studies to determine the contribution of saeS(SKT) allele to the pathogenesis of infections caused by ST30 isolates are merited.


Asunto(s)
Proteínas Bacterianas/metabolismo , Toxinas Bacterianas/metabolismo , Exotoxinas/metabolismo , Regulación Bacteriana de la Expresión Génica , Leucocidinas/metabolismo , Staphylococcus aureus Resistente a Meticilina/genética , Proteínas Quinasas/metabolismo , Alelos , Proteínas Bacterianas/genética , Toxinas Bacterianas/genética , Recuento de Células , Exotoxinas/genética , Perfilación de la Expresión Génica , Humanos , Leucocidinas/genética , Staphylococcus aureus Resistente a Meticilina/crecimiento & desarrollo , Proteínas Quinasas/genética , Factores de Transcripción
8.
J Basic Microbiol ; 56(4): 358-68, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26898187

RESUMEN

Staphylococcus epidermidis, which is a causative pathogen of nosocomial infection, expresses its virulent traits such as biofilm and autolysis regulated by two-component signal transduction system SaeRS. In this study, the S. epidermidis SaeRS was identified to negatively regulate the expression of genes involved in competence (comF, murF), cytolysis (lrgA), and autolysis (lytS) by DNA microarray or real-time RT-PCR analysis. In addition, saeRS mutant showed increased competence and higher susceptibility to antibiotics such as penicillin and oxacillin than the wild-type strain. The study will be helpful for understanding the characterization of the SaeRS in S. epidermidis.


Asunto(s)
Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Penicilina G/farmacología , Staphylococcus epidermidis/efectos de los fármacos , Staphylococcus epidermidis/metabolismo , Antibacterianos/farmacología , Proteínas Bacterianas/efectos de los fármacos , Biopelículas/efectos de los fármacos , ADN Bacteriano/genética , Regulación Bacteriana de la Expresión Génica , Pruebas de Sensibilidad Microbiana , Oxacilina/farmacología , Transducción de Señal , Staphylococcus epidermidis/genética
9.
J Infect Dis ; 212(6): 934-8, 2015 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-25722299

RESUMEN

While Staphylococcus aureus accelerates human neutrophil cell death, the underlying host- and pathogen-derived mechanisms remain incompletely defined. Previous studies demonstrated that the S. aureus SaeR/S sensory system is essential for pathogen survival following neutrophil phagocytosis. Herein, we demonstrate that the SaeR/S system promoted accelerated cell death, suppressed phosphorylation of nuclear factor-κB, and reduced interleukin-8 (IL-8) production in human neutrophils. Treatment of neutrophils with recombinant IL-8 significantly reduced bacterial burden and apoptosis. Our findings demonstrate a mechanism by which S. aureus suppresses the early neutrophil-derived IL-8 response to disrupt cell fate and promote disease.


Asunto(s)
Muerte Celular/fisiología , Interleucina-8/metabolismo , Neutrófilos/fisiología , Staphylococcus aureus/fisiología , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Células Cultivadas , Regulación de la Expresión Génica/fisiología , Humanos , Interleucina-8/genética , FN-kappa B/metabolismo , Proteínas Quinasas/genética , Proteínas Quinasas/metabolismo , Factores de Transcripción
10.
Virulence ; 15(1): 2411540, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-39359063

RESUMEN

The ability to form robust biofilms and secrete a diverse array of virulence factors are key pathogenic determinants of Staphylococcus aureus, causing a wide range of infectious diseases. Here, we characterized cwrA as a VraR-regulated gene encoding a cell wall inhibition-responsive protein (CwrA) using electrophoretic mobility shift assays. We constructed cwrA deletion mutants in the genetic background of methicillin-resistant S. aureus (MRSA) and methicillin-sensitive S. aureus (MSSA) strains. Phenotypic analyses indicated that deletion of cwrA led to impaired biofilm formation, which was correlated with polysaccharide intercellular adhesin (PIA). Besides, the results of real-time quantitative PCR (RT-qPCR) and ß-galactosidase activity assay revealed that CwrA promoted biofilm formation by influence the ica operon activity in S. aureus. Furthermore, cwrA deletion mutants released less extracellular DNA (eDNA) in the biofilm because of their reduced autolytic activity compared to the wild-type (WT) strains. We also found that cwrA deletion mutant more virulence than the parental strain because of its enhanced hemolytic activity. Mechanistically, this phenotypic alteration is related to activation of the SaeRS two-component system, which positively regulates the transcriptional levels of genes encoding membrane-damaging toxins. Overall, our results suggest that CwrA plays an important role in modulating biofilm formation and hemolytic activity in S. aureus.


Asunto(s)
Proteínas Bacterianas , Biopelículas , Pared Celular , Regulación Bacteriana de la Expresión Génica , Infecciones Estafilocócicas , Staphylococcus aureus , Factores de Virulencia , Biopelículas/crecimiento & desarrollo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Staphylococcus aureus/patogenicidad , Staphylococcus aureus/genética , Virulencia , Pared Celular/metabolismo , Factores de Virulencia/genética , Factores de Virulencia/metabolismo , Infecciones Estafilocócicas/microbiología , Animales , Ratones , Staphylococcus aureus Resistente a Meticilina/genética , Staphylococcus aureus Resistente a Meticilina/patogenicidad , Operón , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Polisacáridos Bacterianos/metabolismo , Polisacáridos Bacterianos/genética , Eliminación de Gen , Femenino , Proteínas Quinasas
11.
Emerg Microbes Infect ; 12(2): 2276335, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37882148

RESUMEN

Diabetic foot infections are a common complication of diabetes. Staphylococcus aureus is frequently isolated from diabetic foot infections and commonly colonizes human nares. According to the study, the nasal microbiome analysis revealed that diabetic patients had a significantly altered nasal microbial composition and diversity. Typically, the fasting blood glucose (FBG) level had an impact on the abundance and sequence type (ST) of S. aureus in diabetic patients. We observed that highly virulent S. aureus ST7 strains were more frequently colonized in diabetic patients, especially those with poorly controlled FBG, while ST59 was dominant in healthy individuals. S. aureus ST7 strains were more resistant to human antimicrobial peptides and formed stronger biofilms than ST59 strains. Critically, S. aureus ST7 strains displayed higher virulence compared to ST59 strains in vivo. The dominance of S. aureus ST7 strains in hyperglycemic environment is due to the higher activity of the SaeRS two-component system (TCS). S. aureus ST7 strains outcompeted ST59 both in vitro, and in nasal colonization model in diabetic mice, which was abolished by the deletion of the SaeRS TCS. Our data indicated that highly virulent S. aureus strains preferentially colonize diabetic patients with poorly controlled FBG through SaeRS TCS. Detection of S. aureus colonization and elimination of colonizing S. aureus are critical in the care of diabetic patients with high FBG.


Asunto(s)
Diabetes Mellitus Experimental , Pie Diabético , Staphylococcus aureus Resistente a Meticilina , Infecciones Estafilocócicas , Humanos , Animales , Ratones , Staphylococcus aureus , Diabetes Mellitus Experimental/complicaciones , Nariz , Cavidad Nasal
12.
Front Microbiol ; 14: 1121621, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37138609

RESUMEN

There are a variety of regulatory systems in bacteria, among which the two-component system (TCS) can sense external environmental changes and make a series of physiological and biochemical reactions, which is crucial for the life activities of bacteria. As a member of TCS, SaeRS is considered to be an important virulence factor in Staphylococcus aureus, but its function in tilapia (Oreochromis niloticus)-derived Streptococcus agalactiae remains unknown. To explore the role of SaeRS in regulating virulence in the two-component system (TCS) of S. agalactiae from tilapia, ΔSaeRS mutant strain and CΔSaeRS complementary strain were constructed by homologous recombination. The results showed that the abilities of growth and biofilm formation of ΔSaeRS strain were significantly decreased when cultured in a brain heart infusion (BHI) medium (P < 0.01). Also, the survival rate of the ΔSaeRS strain in blood was decreased when compared with the wild strain S. agalactiae THN0901. Under the higher infection dose, the accumulative mortality of tilapia caused by the ΔSaeRS strain was significantly decreased (23.3%), of which THN0901 and CΔSaeRS strains were 73.3%. The results of competition experiments in tilapia showed that the invasion and colonization abilities of the ΔSaeRS strain were also dramatically lower than those of the wild strain (P < 0.01). Compared with the THN0901, the mRNA expression levels of virulence factors (fbsB, sip, cylE, bca, etc.) in the ΔSaeRS strain were significantly down-regulated (P < 0.01). SaeRS is one of the virulence factors of S. agalactiae. It plays a role in promoting host colonization and achieving immune evasion during the infection of tilapia, which provides a basis for exploring the pathogenic mechanism of S. agalactiae infected with tilapia.

13.
Microbiol Spectr ; 11(3): e0525522, 2023 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-37154710

RESUMEN

Staphylococcus aureus is a Gram-positive commensal and opportunistic pathogen able to cause diseases ranging from mild skin infections to life-threatening endocarditis and toxic shock syndrome. The ability to cause such an array of diseases is due to the complex S. aureus regulatory network controlling an assortment of virulence factors, including adhesins, hemolysins, proteases, and lipases. This regulatory network is controlled by both protein and RNA elements. We previously identified a novel regulatory protein called ScrA, which, when overexpressed, leads to the increased activity and expression of the SaeRS regulon. In this study, we further explore the role of ScrA and examine the consequences to the bacterial cell of scrA gene disruption. These results demonstrate that scrA is required for several virulence-related processes, and in many cases, the phenotypes of the scrA mutant are inverse to those observed in cells overexpressing ScrA. Interestingly, while the majority of ScrA-mediated phenotypes appear to rely on the SaeRS system, our results also indicate that ScrA may also act independently of SaeRS when regulating hemolytic activity. Finally, using a murine model of infection, we demonstrate that scrA is required for virulence, potentially in an organ-specific manner. IMPORTANCE Staphylococcus aureus is the cause of several potentially life-threatening infections. An assortment of toxins and virulence factors allows such a wide range of infections. However, an assortment of toxins or virulence factors requires complex regulation to control expression under all of the different conditions encountered by the bacterium. Understanding the intricate web of regulatory systems allows the development of novel approaches to combat S. aureus infections. Here, we have shown that the small protein ScrA, which was previously identified by our laboratory, influences several virulence-related functions through the SaeRS global regulatory system. These findings add ScrA to the growing list of virulence regulators in S. aureus.


Asunto(s)
Infecciones Estafilocócicas , Staphylococcus aureus , Animales , Ratones , Virulencia , Staphylococcus aureus/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Factores de Transcripción/metabolismo , Factores de Virulencia/metabolismo , Infecciones Estafilocócicas/microbiología
14.
Emerg Microbes Infect ; 11(1): 1402-1415, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-35508433

RESUMEN

Staphylococcus aureus is an important human commensal bacteria colonizing the human body, especially the nasal cavity. The nasal carriage can be a source of S. aureus bacteremia. However, the bacterial factors contributing to nasal colonization are not completely understood. By analysing S. aureus strains from the nasal cavity of the children, young adults, and seniors, we found that the low activity of the SaeRS two-component system (TCS) is an important determinant for S. aureus to colonize in seniors. The senior group isolates of S. aureus showed a rather distinct sequence type composition as compared with other age group isolates. The senior group isolates showed not only a lower gene carriage of enterotoxins a, c, and q but also lower hemolytic activity against human red blood cells. Of regulators affecting hemolysin production (i.e. agr, saeRS, rot, rsp, and sarS), only the SaeRS TCS showed an age-dependent decrease of activity. The decreased virulence and better colonization ability of the senior group isolates of S. aureus were confirmed in the mouse model. The senior group isolates showed the lowest survival and the best adhesion and colonizing ability. Also, the senior nasal secretions supported S. aureus survival better than the child and young adult nasal secretions. These results indicated that the senior nasal cavity favours colonization of S. aureus with higher adhesion and lower virulence, to which the reduced SaeRS TCS activity contributes. Taken together, our results illustrate an example of bacterial adaptation to the changing host environment.


Asunto(s)
Infecciones Estafilocócicas , Staphylococcus aureus , Animales , Humanos , Ratones , Cavidad Nasal , Nariz , Infecciones Estafilocócicas/microbiología , Virulencia , Factores de Virulencia/genética
15.
Infect Drug Resist ; 15: 2853-2864, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35677526

RESUMEN

Introduction: Staphylococcus aureus is an opportunistic pathogen that can cause life-threatening bloodstream infections such as sepsis and endocarditis. In recent years, the emergence and increase of methicillin-resistant and multidrug-resistant S. aureus has posed a great challenge to the antibiotic treatment of infectious diseases. Anti-virulence strategies targeting virulence factors are an effective new therapy for the treatment of S. aureus infections. Results: In this study, we constructed a NWMN2330 deletion mutant (Newman-ΔNWMN2330) and a complement (Newman-ΔNWMN2330-C) of S. aureus Newman to study the role of NWMN2330 in the virulence of S. aureus. Through transcriptome sequencing, it was found that the expression of 224 genes in Newman-ΔNWMN2330 was significantly different (>2-fold) compared with S. aureus Newman, and these differentially expressed genes were related to multiple functions of S. aureus. And we found that NWMN2330 could positively regulate the expression of S. aureus hla gene. Therefore, the deletion mutant Newman-ΔNWMN2330 exhibited lower hemolytic activity and lower α-toxin production than Newman. Newman-ΔNWMN2330 also exhibited lower lethality and pathogenicity in worm survival experiments and nude mouse skin abscess model. RT-qPCR results showed that compared with the wild-type strain, the expression of saeRS and hla in Newman-ΔNWMN2330 strain was significantly reduced at the mRNA level, which preliminarily indicated that NWMN2330 promoted the expression of hla by up-regulating saeRS. Discussion: In general, our results indicated that NWMN2330 may be associated with the virulence of Staphylococcus aureus by increasing the expression of hla and saeRS.

16.
Cell Rep ; 41(9): 111721, 2022 11 29.
Artículo en Inglés | MEDLINE | ID: mdl-36450248

RESUMEN

Influenza infection is substantially worsened by the onset of secondary pneumonia caused by bacteria, such as methicillin-resistant Staphylococcus aureus (MRSA). The bidirectional interaction between the influenza-injured lung microenvironment and MRSA is poorly understood. By conditioning MRSA ex vivo in bronchoalveolar lavage fluid collected from mice at various time points of influenza infection, we found that the influenza-injured lung microenvironment dynamically induces MRSA to increase cytotoxin expression while decreasing metabolic pathways. LukAB, a SaeRS two-component system-dependent cytotoxin, is particularly important to the severity of post-influenza MRSA pneumonia. LukAB's activity is likely shaped by the post-influenza lung microenvironment, as LukAB binds to (and is activated by) heparan sulfate (HS) oligosaccharide sequences shed from the epithelial glycocalyx after influenza. Our findings indicate that post-influenza MRSA pneumonia is shaped by bidirectional host-pathogen interactions: host injury triggers changes in bacterial expression of toxins, the activity of which may be shaped by host-derived HS fragments.


Asunto(s)
Coinfección , Gripe Humana , Staphylococcus aureus Resistente a Meticilina , Neumonía Bacteriana , Animales , Ratones , Humanos , Gripe Humana/complicaciones , Virulencia , Neumonía Bacteriana/complicaciones , Citotoxinas , Heparitina Sulfato , Pulmón
17.
mBio ; 13(5): e0147222, 2022 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-36135382

RESUMEN

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.


Asunto(s)
Infecciones Estafilocócicas , Staphylococcus aureus , Humanos , Staphylococcus aureus/metabolismo , Regulación Bacteriana de la Expresión Génica , Histidina Quinasa/metabolismo , Dihidrolipoamida Deshidrogenasa/genética , Dihidrolipoamida Deshidrogenasa/metabolismo , Histidina/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Infecciones Estafilocócicas/microbiología , Ácidos Grasos/metabolismo , Exotoxinas/metabolismo
18.
Pathogens ; 10(11)2021 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-34832552

RESUMEN

Coagulase is a critical factor for distinguishing Staphylococcus aureus and coagulase-negative Staphylococcus. Our previous studies demonstrated that the null mutation of coagulase (coa) or its direct regulator, SaeRS, significantly enhanced the ability of S. aureus (CA-MRSA 923) to survive in human blood in vitro. This led us to further investigate the role of coagulase and its direct regulator, SaeRS, in the pathogenicity of CA-MRSA 923 in bacteremia during infection. In this study, we found that the null mutation of coa significantly decreased the mortality of CA-MRSA 923; moreover, the single null mutation of saeRS and the double deletion of coa/saeRS abolished the virulence of CA-MRSA 923. Moreover, the mice infected with either the saeRS knockout or the coa/saeRS double knockout mutant exhibited fewer histological lesions and less neutrophils infiltration in the infected kidneys compared to those infected with the coa knockout mutant or their parental control. Furthermore, we examined the impact of coa and saeRS on bacterial survival in vitro. The null mutation of coa had no impact on bacterial survival in mice blood, whereas the deletion mutation of saeRS or coa/saeRS significantly enhanced bacterial survival in mice blood. These data indicate that SaeRS plays a key role in the lethality of CA-MRSA 923 bacteremia, and that coagulase is one of the important virulence factors that is regulated by SaeRS and contributes to the pathogenicity of CA-MRSA 923.

19.
Cell Host Microbe ; 29(6): 930-940.e4, 2021 06 09.
Artículo en Inglés | MEDLINE | ID: mdl-33852876

RESUMEN

Staphylococcus aureus commonly infects the skin, but the host-pathogen interactions controlling bacterial growth remain unclear. S. aureus virulence is regulated by the Agr quorum-sensing system that controls factors including phenol-soluble modulins (PSMs), a group of cytotoxic peptides. We found a differential requirement for Agr and PSMα for pathogen growth in the skin. In neutrophil-deficient mice, S. aureus growth on the epidermis was unaffected, but the pathogen penetrated the dermis through mechanisms that require PSMα. In the dermis, pathogen expansion required Agr in wild-type mice, but not in neutrophil-deficient mice. Agr limited oxidative and non-oxidative killing in neutrophils by inhibiting pathogen late endosome localization and promoting phagosome escape. Unlike Agr, the SaeR/S virulence program was dispensable for growth in the epidermis and promoted dermal pathogen expansion independently of neutrophils. Thus, S. aureus growth and invasion are differentially regulated with Agr limiting intracellular killing within neutrophils to promote pathogen expansion in the dermis and subcutaneous tissue.


Asunto(s)
Proteínas Bacterianas/metabolismo , Neutrófilos/fisiología , Piel/microbiología , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/fisiología , Staphylococcus aureus/patogenicidad , Transactivadores/metabolismo , Virulencia , Animales , Toxinas Bacterianas/metabolismo , Interacciones Huésped-Patógeno , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación , Proteínas Quinasas/metabolismo , Percepción de Quorum , Factores de Transcripción/metabolismo
20.
Front Microbiol ; 11: 25, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32117092

RESUMEN

Staphylococcus aureus is an important pathogen in hospital and community infections. Fusidic acid is particularly effective in treating skin and wound infections caused by staphylococci. The purpose of our study was to clarify the effect of fusidic acid on the biofilm formation and α-toxin expression of S. aureus at subinhibitory concentrations [1/64, 1/32, and 1/16 × minimum inhibitory concentration (MIC)]. A total of 504 genes greater than a twofold or less than twofold change in expression of S. aureus effected by subinhibitory concentrations of fusidic acid were found, including 232 up-regulated genes and 272 down-regulated genes, which were determined by transcriptome sequencing. Our results showed subinhibitory concentrations of fusidic acid significantly inhibited the expression of hla, spa, icaA, and cidA at the mRNA level in clinical S. aureus strains tested. And subinhibitory concentrations of fusidic acid can significantly reduce the hemolysis activity and α-toxin production of S. aureus. In addition, the subinhibitory concentrations of fusidic acid significantly inhibited biofilm formation, autolysis, cell aggregation, and polysaccharide intercellular adhesin (PIA) production of S. aureus. Moreover, fusidic acid effectively reduces the damage of mouse skin lesion area. Furthermore, fusidic acid reduced the expression of the two-component regulatory system saeRS and staphylococcal accessory gene regulator (sarA). In conclusion, our results suggested that the subinhibitory concentrations of fusidic acid may reduce the virulence of S. aureus by down-regulating sarA and saeRS to reduce biofilm formation and α-toxin expression, which will provide a theoretical basis for the clinical treatment of S. aureus infection. This is the first report that fusidic acid has an inhibitory effect on the virulence of S. aureus, and this broadens the clinical application of fusidic acid.

SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda