Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
bioRxiv ; 2024 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-39026866

RESUMO

Bacillus anthracis , a Gram-positive facultative anaerobe and the causative agent of anthrax, multiplies to extraordinarily high numbers in vertebrate blood, resulting in considerable heme exposure. Heme is an essential nutrient and the preferred iron source for bacteria during vertebrate colonization, but its high redox potential makes it toxic in excess. To regulate heme homeostasis, many Gram-positive bacteria, including B. anthracis , rely on the two-component signaling system HssRS. HssRS comprises the heme sensing histidine kinase HssS, which modulates the activity of the HssR transcription factor to enable bacteria to circumvent heme toxicity. However, the regulation of the HssRS system remains unclear. Here we identify FapR, the transcriptional regulator of fatty acid biosynthesis, as a key factor in HssRS function. FapR plays an important role in maintaining membrane integrity and the localization of the histidine kinase HssS. Specifically, disruption of fapR leads to increased membrane rigidity, which hinders the penetration of HssRS inducers, resulting in the inactivation of HssRS. Furthermore, deletion of fapR affects the loading of HssS onto the cell membrane, compromising its heme sensing function and subsequently reducing endogenous heme biosynthesis. These findings shed light on the molecular mechanisms governing bacterial adaptation to heme stress and provide potential targets for antimicrobial intervention strategies. IMPORTANCE: Understanding the mechanisms by which B. anthracis regulates heme homeostasis is crucial for developing new strategies to combat anthrax, a serious disease affecting both humans and animals. This study uncovers the role of the transcriptional regulator FapR in maintaining membrane integrity and facilitating the proper function of the HssRS two-component signaling system, which is essential for managing heme toxicity in B. anthracis , as well as other Gram-positive pathogens. By elucidating the connection between FapR and HssRS, our findings provide new insights into the molecular adaptation of bacteria to heme stress and expand our knowledge of bacterial physiology and pathogenicity. More importantly, targeting the regulatory pathways involved in heme sensing and homeostasis presents a promising approach for developing novel therapeutics against anthrax and potentially other bacterial infections that rely on similar mechanisms.

2.
Nat Commun ; 13(1): 1491, 2022 03 21.
Artigo em Inglês | MEDLINE | ID: mdl-35314695

RESUMO

HitRS is a two-component system that responds to cell envelope damage in the human pathogen Bacillus anthracis. Here we identify an RNA-binding protein, KrrA, that regulates HitRS function by modulating the stability of the hitRS mRNA. In addition to hitRS, KrrA binds to over 70 RNAs and, directly or indirectly, affects the expression of over 150 genes involved in multiple processes, including genetic competence, sporulation, RNA turnover, DNA repair, transport, and cellular metabolism. KrrA does not exhibit detectable nuclease activity in vitro, and thus the mechanism by which it modulates mRNA stability remains unclear.


Assuntos
Bacillus anthracis , Bacillus anthracis/genética , Bacillus anthracis/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Parede Celular/metabolismo , Regulação Bacteriana da Expressão Gênica , Humanos , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo
3.
Infect Immun ; 90(1): e0056021, 2022 01 25.
Artigo em Inglês | MEDLINE | ID: mdl-34748369

RESUMO

Bacillus anthracis is the causative agent of anthrax. This Gram-positive bacterium poses a substantial risk to human health due to high mortality rates and the potential for malicious use as a bioterror weapon. To survive within the vertebrate host, B. anthracis relies on two-component system (TCS) signaling to sense host-induced stresses and respond to alterations in the environment through changes in target gene expression. HitRS and HssRS are cross-regulating TCSs in B. anthracis that respond to cell envelope disruptions and high heme levels, respectively. In this study, an unbiased and targeted genetic selection was designed to identify gene products that are involved in HitRS and HssRS signaling. This selection led to the identification of inactivating mutations within dnaJ and clpX that disrupt HitRS- and HssRS-dependent gene expression. DnaJ and ClpX are the substrate-binding subunits of the DnaJK protein chaperone and ClpXP protease, respectively. DnaJ regulates the levels of HitR and HitS to facilitate signal transduction, while ClpX specifically regulates HitS levels. Together, these results reveal that the protein homeostasis regulators, DnaJ and ClpX, function to maintain B. anthracis signal transduction activities through TCS regulation.


Assuntos
Antraz/microbiologia , Bacillus anthracis/fisiologia , Proteínas de Bactérias/metabolismo , Endopeptidase Clp/metabolismo , Proteínas de Choque Térmico HSP40/metabolismo , Transdução de Sinais , Fenômenos Fisiológicos Bacterianos , Proteínas de Bactérias/genética , Membrana Celular/metabolismo , Regulação Bacteriana da Expressão Gênica , Modelos Biológicos , Transporte Proteico , Seleção Genética
4.
PLoS Pathog ; 16(12): e1009148, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33362282

RESUMO

Two component systems (TCSs) are a primary mechanism of signal sensing and response in bacteria. Systematic characterization of an entire TCS could provide a mechanistic understanding of these important signal transduction systems. Here, genetic selections were employed to dissect the molecular basis of signal transduction by the HitRS system that detects cell envelope stress in the pathogen Bacillus anthracis. Numerous point mutations were isolated within HitRS, 17 of which were in a 50-residue HAMP domain. Mutational analysis revealed the importance of hydrophobic interactions within the HAMP domain and highlighted its essentiality in TCS signaling. In addition, these data defined residues critical for activities intrinsic to HitRS, uncovered specific interactions among individual domains and between the two signaling proteins, and revealed that phosphotransfer is the rate-limiting step for signal transduction. Furthermore, this study establishes the use of unbiased genetic selections to study TCS signaling and provides a comprehensive mechanistic understanding of an entire TCS.


Assuntos
Bacillus anthracis/fisiologia , Proteínas de Bactérias/fisiologia , Regulação Bacteriana da Expressão Gênica/fisiologia , Transdução de Sinais/fisiologia , Seleção Genética/fisiologia , Estresse Fisiológico/fisiologia
5.
mBio ; 11(2)2020 03 31.
Artigo em Inglês | MEDLINE | ID: mdl-32234818

RESUMO

Bacillus anthracis is a spore-forming bacterium that causes devastating infections and has been used as a bioterror agent. This pathogen can survive hostile environments through the signaling activity of two-component systems, which couple environmental sensing with transcriptional activation to initiate a coordinated response to stress. In this work, we describe the identification of a two-component system, EdsRS, which mediates the B. anthracis response to the antimicrobial compound targocil. Targocil is a cell envelope-targeting compound that is toxic to B. anthracis at high concentrations. Exposure to targocil causes damage to the cellular barrier and activates EdsRS to induce expression of a previously uncharacterized cardiolipin synthase, which we have named ClsT. Both EdsRS and ClsT are required for protection against targocil-dependent damage. Induction of clsT by EdsRS during targocil treatment results in an increase in cardiolipin levels, which protects B. anthracis from envelope damage. Together, these results reveal that a two-component system signaling response to an envelope-targeting antimicrobial induces production of a phospholipid associated with stabilization of the membrane. Cardiolipin is then used to repair envelope damage and promote B. anthracis viability.IMPORTANCE Compromising the integrity of the bacterial cell barrier is a common action of antimicrobials. Targocil is an antimicrobial that is active against the bacterial envelope. We hypothesized that Bacillus anthracis, a potential weapon of bioterror, senses and responds to targocil to alleviate targocil-dependent cell damage. Here, we show that targocil treatment increases the permeability of the cellular envelope and is particularly toxic to B. anthracis spores during outgrowth. In vegetative cells, two-component system signaling through EdsRS is activated by targocil. This results in an increase in the production of cardiolipin via a cardiolipin synthase, ClsT, which restores the loss of barrier function, thereby reducing the effectiveness of targocil. By elucidating the B. anthracis response to targocil, we have uncovered an intrinsic mechanism that this pathogen employs to resist toxicity and have revealed therapeutic targets that are important for bacterial defense against structural damage.


Assuntos
Antibacterianos/farmacologia , Bacillus anthracis/efeitos dos fármacos , Bacillus anthracis/fisiologia , Proteínas de Bactérias/metabolismo , Cardiolipinas/biossíntese , Quinazolinas/farmacologia , Triazóis/farmacologia , Proteínas de Bactérias/genética , Parede Celular/efeitos dos fármacos , Parede Celular/metabolismo , Regulação Bacteriana da Expressão Gênica/efeitos dos fármacos , Ordem dos Genes , Permeabilidade/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Esporos Bacterianos/efeitos dos fármacos , Esporos Bacterianos/crescimento & desenvolvimento , Transcrição Gênica
6.
FEMS Microbiol Lett ; 363(9)2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-27030728

RESUMO

Several Gram-positive pathogens scavenge host-derived heme to satisfy their nutritional iron requirement. However, heme is a toxic molecule capable of damaging the bacterial cell. Gram-positive pathogens within the phylum Firmicutes overcome heme toxicity by sensing heme through HssRS, a two-component system that regulates the heme detoxification transporter HrtAB. Here we show that heme sensing by HssRS and heme detoxification by HrtAB occur in the insect pathogen Bacillus thuringiensis We find that in B. thuringiensis, HssRS directly regulates an operon, hrmXY, encoding hypothetical membrane proteins that are not found in other Firmicutes with characterized HssRS and HrtAB systems. This novel HssRS-regulated operon or its orthologs BMB171_c3178 and BMB171_c3330 are required for maximal heme resistance. Furthermore, the activity of HrmXY is not dependent on expression of HrtAB. These results suggest that B. thuringiensis senses heme through HssRS and induces expression of separate membrane-localized systems capable of overcoming different aspects of heme toxicity.


Assuntos
Bacillus thuringiensis/genética , Bacillus thuringiensis/metabolismo , Regulação Bacteriana da Expressão Gênica , Heme/metabolismo , Bacillus thuringiensis/patogenicidade , Bacillus thuringiensis/fisiologia , Proteínas de Bactérias/genética , Transporte Biológico , Ferro/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Mutação , Óperon , Regiões Promotoras Genéticas
7.
PLoS Pathog ; 10(3): e1004044, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24675902

RESUMO

Two-component signaling systems (TCSs) are one of the mechanisms that bacteria employ to sense and adapt to changes in the environment. A prototypical TCS functions as a phosphorelay from a membrane-bound sensor histidine kinase (HK) to a cytoplasmic response regulator (RR) that controls target gene expression. Despite significant homology in the signaling domains of HKs and RRs, TCSs are thought to typically function as linear systems with little to no cross-talk between non-cognate HK-RR pairs. Here we have identified several cell envelope acting compounds that stimulate a previously uncharacterized Bacillus anthracis TCS. Furthermore, this TCS cross-signals with the heme sensing TCS HssRS; therefore, we have named it HssRS interfacing TCS (HitRS). HssRS reciprocates cross-talk to HitRS, suggesting a link between heme toxicity and cell envelope stress. The signaling between HssRS and HitRS occurs in the parental B. anthracis strain; therefore, we classify HssRS-HitRS interactions as cross-regulation. Cross-talk between HssRS and HitRS occurs at both HK-RR and post-RR signaling junctions. Finally, HitRS also regulates a previously unstudied ABC transporter implicating this transporter in the response to cell envelope stress. This chemical biology approach to probing TCS signaling provides a new model for understanding how bacterial signaling networks are integrated to enable adaptation to complex environments such as those encountered during colonization of the vertebrate host.


Assuntos
Bacillus anthracis/fisiologia , Proteínas de Bactérias/metabolismo , Regulação Bacteriana da Expressão Gênica/fisiologia , Heme/metabolismo , Transdução de Sinais/fisiologia , Parede Celular/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Estresse Fisiológico
8.
J Bacteriol ; 196(7): 1335-42, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24443529

RESUMO

The reactive nature of heme enables its use as an enzymatic cofactor while rendering excess heme toxic. The importance of heme detoxification machinery is highlighted by the presence of various types of these homeostatic systems in Gram-positive and Gram-negative microorganisms. A number of pathogens possess orthologs of the HssRS/HrtAB heme detoxification system, underscoring a potential role this system plays in the survival of bacteria in heme-rich environments such as the vertebrate host. In this work, we sought to determine the role of this system in protection against metalloporphyrin heme analogues identified by previous studies as antimicrobial agents. Our findings demonstrate that only toxic metalloporphyrins maximally activate expression of the Staphylococcus aureus heme detoxification system, suggesting that the sensing mechanism of HssRS might require a component of the associated toxicity rather than or in addition to the metalloporphyrin itself. We further establish that only a subset of toxic metalloporphyrins elicit the oxidative damage previously shown to be a significant component of heme toxicity whereas all toxic noniron metalloporphyrins inhibit bacterial respiration. Finally, we demonstrate that, despite the fact that toxic metalloporphyrin treatment induces expression of S. aureus heme detoxification machinery, the HrtAB heme export pump is unable to detoxify most of these molecules. The ineffectiveness of HrtAB against toxic heme analogues provides an explanation for their increased antimicrobial activity relative to heme. Additionally, these studies define the specificity of HssRS/HrtAB, which may provide future insight into the biochemical mechanisms of these systems.


Assuntos
Proteínas de Bactérias/metabolismo , Regulação Bacteriana da Expressão Gênica , Heme/metabolismo , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/metabolismo , Proteínas de Bactérias/genética , Transporte Biológico , Heme/análogos & derivados , Heme/toxicidade , Humanos , Infecções Estafilocócicas/metabolismo , Staphylococcus aureus/genética
9.
Proc Natl Acad Sci U S A ; 110(20): 8206-11, 2013 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-23630262

RESUMO

Staphylococcus aureus is a significant infectious threat to global public health. Acquisition or synthesis of heme is required for S. aureus to capture energy through respiration, but an excess of this critical cofactor is toxic to bacteria. S. aureus employs the heme sensor system (HssRS) to overcome heme toxicity; however, the mechanism of heme sensing is not defined. Here, we describe the identification of a small molecule activator of HssRS that induces endogenous heme biosynthesis by perturbing central metabolism. This molecule is toxic to fermenting S. aureus, including clinically relevant small colony variants. The utility of targeting fermenting bacteria is exemplified by the fact that this compound prevents the emergence of antibiotic resistance, enhances phagocyte killing, and reduces S. aureus pathogenesis. Not only is this small molecule a powerful tool for studying bacterial heme biosynthesis and central metabolism; it also establishes targeting of fermentation as a viable antibacterial strategy.


Assuntos
Fermentação , Regulação Bacteriana da Expressão Gênica , Heme/biossíntese , Naftóis/farmacologia , Pirazóis/farmacologia , Staphylococcus aureus/metabolismo , Animais , Antibacterianos/farmacologia , Proteínas de Bactérias/metabolismo , Cromatografia Líquida de Alta Pressão , Técnicas de Química Combinatória , Desenho de Fármacos , Glicólise , Heme Oxigenase (Desciclizante)/metabolismo , Concentração Inibidora 50 , Leucócitos/citologia , Espectrometria de Massas , Camundongos , Microscopia Eletrônica de Varredura , Fagócitos/metabolismo , Infecções Estafilocócicas/metabolismo , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/efeitos dos fármacos
10.
Mol Microbiol ; 86(6): 1376-92, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23043465

RESUMO

Staphylococcus aureus is a pathogen that infects multiple anatomical sites leading to a diverse array of diseases. Although vertebrates can restrict the growth of invading pathogens by sequestering iron within haem, S. aureus surmounts this challenge by employing high-affinity haem uptake systems. However, the presence of excess haem is highly toxic, necessitating tight regulation of haem levels. To overcome haem stress, S. aureus expresses the detoxification system HrtAB. In this work, a transposon screen was performed in the background of a haem-susceptible, HrtAB-deficient S. aureus strain to identify the substrate transported by this putative pump and the source of haem toxicity. While a recent report indicates that HrtAB exports haem itself, the haem-resistant mutants uncovered by the transposon selection enabled us to elucidate the cellular factors contributing to haem toxicity. All mutants identified in this screen inactivated the menaquinone (MK) biosynthesis pathway. Deletion of the final steps of this pathway revealed that quinone molecules localizing to the cell membrane potentiate haem-associated superoxide production and subsequent oxidative damage. These data suggest a model in which membrane-associated haem and quinone molecules form a redox cycle that continuously generates semiquinones and reduced haem, both of which react with atmospheric oxygen to produce superoxide.


Assuntos
Heme/toxicidade , Staphylococcus aureus/efeitos dos fármacos , Staphylococcus aureus/metabolismo , Vitamina K 2/metabolismo , Adenosina Trifosfatases/deficiência , Vias Biossintéticas/genética , Elementos de DNA Transponíveis , Deleção de Genes , Mutagênese Insercional , Estresse Oxidativo , Staphylococcus aureus/genética , Staphylococcus aureus/crescimento & desenvolvimento , Superóxidos/metabolismo
11.
J Bacteriol ; 193(15): 3871-8, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21622734

RESUMO

The bacterial pathogen Chromobacterium violaceum uses a LuxIR-type quorum-sensing system to detect and respond to changes in cell population density. CviI synthesizes the autoinducer C(10)-homoserine lactone (C(10)-HSL), and CviR is a cytoplasmic DNA binding transcription factor that activates gene expression following binding to C(10)-HSL. A number of behaviors are controlled by quorum sensing in C. violaceum. However, few genes have been shown to be directly controlled by CviR, in part because the DNA motif bound by CviR is not well characterized. Here, we define the DNA sequence required for promoter recognition by CviR. Using in vivo data generated from a library of point mutations in a CviR-regulated promoter, we find that CviR binds to a palindrome with the ideal sequence CTGNCCNNNNGGNCAG. We constructed a position weight matrix using these in vivo data and scanned the C. violaceum genome to predict CviR binding sites. We measured direct activation of the identified promoters by CviR and found that CviR controls the expression of the promoter for a chitinase, a type VI secretion-related gene, a transcriptional regulator gene, a guanine deaminase gene, and cviI. Indeed, regulation of cviI expression by CviR generates a canonical quorum-sensing positive-feedback loop.


Assuntos
Proteínas de Bactérias/metabolismo , Chromobacterium/fisiologia , DNA Bacteriano/metabolismo , Regulação Bacteriana da Expressão Gênica , Percepção de Quorum , Proteínas Repressoras/metabolismo , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Sequência de Bases , Sítios de Ligação , Chromobacterium/genética , DNA Bacteriano/genética , Dados de Sequência Molecular , Regiões Promotoras Genéticas , Ligação Proteica , Proteínas Repressoras/química , Proteínas Repressoras/genética
12.
Mol Cell ; 42(2): 199-209, 2011 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-21504831

RESUMO

Quorum-sensing bacteria communicate via small molecules called autoinducers to coordinate collective behaviors. Because quorum sensing controls virulence factor expression in many clinically relevant pathogens, membrane-permeable quorum sensing antagonists that prevent population-wide expression of virulence genes offer a potential route to novel antibacterial therapeutics. Here, we report a strategy for inhibiting quorum-sensing receptors of the widespread LuxR family. Structure-function studies with natural and synthetic ligands demonstrate that the dimeric LuxR-type transcription factor CviR from Chromobacterium violaceum is potently antagonized by molecules that bind in place of the native acylated homoserine lactone autoinducer, provided that they stabilize a closed conformation. In such conformations, each of the two DNA-binding domains interacts with the ligand-binding domain of the opposing monomer. Consequently, the DNA-binding helices are held apart by ∼60 Å, twice the ∼30 Å separation required for operator binding. This approach may represent a general strategy for the inhibition of multidomain proteins.


Assuntos
Antibacterianos/farmacologia , Biofilmes/efeitos dos fármacos , Chromobacterium/efeitos dos fármacos , Lactonas/farmacologia , Percepção de Quorum/efeitos dos fármacos , Proteínas Repressoras/antagonistas & inibidores , Transativadores/antagonistas & inibidores , 4-Butirolactona/análogos & derivados , 4-Butirolactona/metabolismo , Antibacterianos/química , Sítios de Ligação , Chromobacterium/genética , Chromobacterium/crescimento & desenvolvimento , Chromobacterium/metabolismo , Chromobacterium/patogenicidade , Cristalografia por Raios X , DNA/metabolismo , Relação Dose-Resposta a Droga , Lactonas/química , Lactonas/metabolismo , Ligantes , Modelos Moleculares , Estrutura Molecular , Mutação , Conformação Proteica , Proteínas Repressoras/química , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Relação Estrutura-Atividade , Transativadores/química , Transativadores/genética , Transativadores/metabolismo , Virulência
13.
Biomed Pharmacother ; 64(10): 672-80, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20970301

RESUMO

Human immunodeficiency virus type 1 (HIV-1) subtype C, which is most predominant in sub-Saharan Africa as well as in Asia and India, is the most prevalent subtype worldwide. A large number of transcription factor families have been shown to be involved in regulating HIV-1 gene expression in T lymphocytes and cells of the monocyte-macrophage lineage. Among these, proteins of the CCAAT/enhancer binding protein (C/EBP) family are of particular importance in regulating HIV-1 gene expression within cells of the monocytic lineage during the course of hematologic development and cellular activation. Few studies have examined the role of C/EBPs in long terminal repeat (LTR)-directed viral gene expression of HIV-1 subtypes other than subtype B. Within subtype B viruses, two functional C/EBP sites located upstream of the TATA box are required for efficient viral replication in cells of the monocyte-macrophage lineage. We report the identification of three putative subtype C C/EBP sites, upstream site 1 and 2 (C-US1 and C-US2) and downstream site 1 (C-DS1). C-US1 and C-DS1 were shown to form specific DNA-protein complexes with members of the C/EBP family (C/EBPα, ß, and δ). Functionally, within the U-937 monocytic cell line, subtype B and C LTRs were shown to be equally responsive to C/EBPß-2, although the basal activity of subtype C LTRs appeared to be higher. Furthermore, the synergistic interaction between C/EBPß-2 and Tat with the subtype C LTR was also observed in U-937 cells as previously demonstrated with the subtype B LTR.


Assuntos
Proteínas Estimuladoras de Ligação a CCAAT/genética , Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Repetição Terminal Longa de HIV , HIV-1/genética , HIV-1/metabolismo , Sequência de Bases , Sítios de Ligação , Linhagem Celular , Regulação Viral da Expressão Gênica , Humanos , Dados de Sequência Molecular , Células Precursoras de Monócitos e Macrófagos/fisiologia , Células Precursoras de Monócitos e Macrófagos/virologia , Ligação Proteica , Alinhamento de Sequência , Análise de Sequência de DNA , Linfócitos T/fisiologia , Linfócitos T/virologia , TATA Box/genética , Transcrição Gênica , Ativação Transcricional , Células U937 , Replicação Viral , Produtos do Gene tat do Vírus da Imunodeficiência Humana/genética
14.
PLoS Pathog ; 6(3): e1000802, 2010 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-20300601

RESUMO

The Staphylococcus aureus HrtAB system is a hemin-regulated ABC transporter composed of an ATPase (HrtA) and a permease (HrtB) that protect S. aureus against hemin toxicity. S. aureus strains lacking hrtA exhibit liver-specific hyper-virulence and upon hemin exposure over-express and secrete immunomodulatory factors that interfere with neutrophil recruitment to the site of infection. It has been proposed that heme accumulation in strains lacking hrtAB is the signal which triggers S. aureus to elaborate this anti-neutrophil response. However, we report here that S. aureus strains expressing catalytically inactive HrtA do not elaborate the same secreted protein profile. This result indicates that the physical absence of HrtA is responsible for the increased expression of immunomodulatory factors, whereas deficiencies in the ATPase activity of HrtA do not contribute to this process. Furthermore, HrtB expression in strains lacking hrtA decreases membrane integrity consistent with dysregulated permease function. Based on these findings, we propose a model whereby hemin-mediated over-expression of HrtB in the absence of HrtA damages the staphylococcal membrane through pore formation. In turn, S. aureus senses this membrane damage, triggering the increased expression of immunomodulatory factors. In support of this model, wildtype S. aureus treated with anti-staphylococcal channel-forming peptides produce a secreted protein profile that mimics the effect of treating DeltahrtA with hemin. These results suggest that S. aureus senses membrane damage and elaborates a gene expression program that protects the organism from the innate immune response of the host.


Assuntos
Transportadores de Cassetes de Ligação de ATP/metabolismo , Adenosina Trifosfatases/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Infecções Cutâneas Estafilocócicas/imunologia , Infecções Cutâneas Estafilocócicas/microbiologia , Staphylococcus aureus/genética , Staphylococcus aureus/imunologia , Transportadores de Cassetes de Ligação de ATP/genética , Adenosina Trifosfatases/genética , Animais , Antibacterianos/farmacologia , Movimento Celular/efeitos dos fármacos , Movimento Celular/imunologia , Exotoxinas/genética , Exotoxinas/metabolismo , Feminino , Regulação Bacteriana da Expressão Gênica , Gramicidina/farmacologia , Hemina/metabolismo , Hemina/farmacologia , Fatores Imunológicos/genética , Fatores Imunológicos/metabolismo , Proteínas de Membrana Transportadoras/genética , Camundongos , Camundongos Endogâmicos BALB C , Neutrófilos/imunologia , Neutrófilos/microbiologia , Infecções Cutâneas Estafilocócicas/tratamento farmacológico , Staphylococcus aureus/patogenicidade , Transcrição Gênica/fisiologia , Regulação para Cima/imunologia , Virulência
15.
Infect Immun ; 78(4): 1618-28, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20100857

RESUMO

The tremendous success of Staphylococcus aureus as a pathogen is due to the controlled expression of a diverse array of virulence factors. The effects of host environments on the expression of virulence factors and the mechanisms by which S. aureus adapts to colonize distinct host tissues are largely unknown. Vertebrates have evolved to sequester nutrient iron from invading bacteria, and iron availability is a signal that alerts pathogenic microorganisms when they enter the hostile host environment. Consistent with this, we report here that S. aureus senses alterations in the iron status via the ferric uptake regulator (Fur) and alters the abundance of a large number of virulence factors. These Fur-mediated changes protect S. aureus against killing by neutrophils, and Fur is required for full staphylococcal virulence in a murine model of infection. A potential mechanistic explanation for the impact of Fur on virulence is provided by the observation that Fur coordinates the reciprocal expression of cytolysins and a subset of immunomodulatory proteins. More specifically, S. aureus lacking fur exhibits decreased expression of immunomodulatory proteins and increased expression of cytolysins. These findings reveal that Fur is involved in initiating a regulatory program that organizes the expression of virulence factors during the pathogenesis of S. aureus pneumonia.


Assuntos
Proteínas de Bactérias/biossíntese , Regulação Bacteriana da Expressão Gênica , Pneumonia Estafilocócica/microbiologia , Proteínas Repressoras/fisiologia , Staphylococcus aureus/patogenicidade , Fatores de Virulência/biossíntese , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/fisiologia , Cromatografia Líquida , Modelos Animais de Doenças , Eletroforese em Gel Bidimensional , Feminino , Perfilação da Expressão Gênica , Técnicas de Inativação de Genes , Espectrometria de Massas , Camundongos , Camundongos Endogâmicos C57BL , Proteoma/análise , Proteínas Repressoras/genética , Staphylococcus aureus/fisiologia
16.
Contrib Microbiol ; 16: 120-135, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19494582

RESUMO

The important human pathogen Staphylococcus aureus is able to satisfy its nutrient iron requirement by acquiring heme from host hemoglobin in the context of infection. However, heme acquisition exposes S. aureus to heme toxicity. In order to detect the presence of toxic levels of exogenous heme, S. aureus is able to sense heme through the heme sensing system (HssRS) two-component system. Upon sensing heme, HssRS directly regulates the expression of the heme-regulated ABC transporter HrtAB, which alleviates heme toxicity. Importantly, the inability to sense or respond to heme alters the virulence of S. aureus, highlighting the importance of heme sensing and detoxification to staphylococcal pathogenesis. Furthermore, potential orthologues of the Hss and Hrt systems are found in many species of Gram-positive bacteria, a possible indication that heme stress is a challenge faced by bacteria whose habitats include host tissues rich in heme.


Assuntos
Heme/metabolismo , Percepção de Quorum/fisiologia , Transdução de Sinais/fisiologia , Staphylococcus aureus/fisiologia , Adaptação Fisiológica , Adenosina Trifosfatases/fisiologia , Evolução Biológica , Heme/toxicidade , Regiões Promotoras Genéticas , Staphylococcus aureus/efeitos dos fármacos , Staphylococcus aureus/patogenicidade
17.
Mol Microbiol ; 72(3): 763-78, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19400785

RESUMO

Bacillus anthracis proliferates to high levels within vertebrate tissues during the pathogenesis of anthrax. This growth is facilitated by the acquisition of nutrient iron from host haem. However, haem acquisition can lead to the accumulation of toxic amounts of haem within B. anthracis. Here, we show that B. anthracis resists haem toxicity by sensing haem through the HssRS two-component system, which regulates expression of the haem-detoxifying transporter HrtAB. In addition, we demonstrate that B. anthracis exhibits elevated HssRS function compared with its evolutionary relative Staphylococcus aureus. Elevated haem sensing is likely required by B. anthracis due to the significant haem sensitivity exhibited by members of the genus Bacilli. We also demonstrate that B. anthracis depends on conserved residues within the previously uncharacterized sensing domain of the histidine kinase HssS for HssS function. Finally, we show that the haem- and HssRS-regulated hrtAB promoter is activated in a murine model of anthrax. These results demonstrate the evolutionary conservation of haem sensing among multiple Gram-positive bacteria and begin to provide a mechanistic explanation for the haem resistance of B. anthracis. Further, these data suggest that haem stress is experienced by bacterial pathogens during infection.


Assuntos
Bacillus anthracis/enzimologia , Proteínas de Bactérias/metabolismo , Hemina/metabolismo , Proteínas Quinases/metabolismo , Serina Endopeptidases/metabolismo , Sequência de Aminoácidos , Animais , Bacillus anthracis/genética , Proteínas de Bactérias/genética , Regulação Bacteriana da Expressão Gênica , Teste de Complementação Genética , Histidina Quinase , Camundongos , Camundongos Endogâmicos A , Dados de Sequência Molecular , Mutagênese Insercional , Fosforilação , Regiões Promotoras Genéticas , Proteínas Quinases/genética , Serina Endopeptidases/genética , Transdução de Sinais , Staphylococcus aureus/enzimologia , Staphylococcus aureus/genética
18.
J Bacteriol ; 190(10): 3588-96, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18326576

RESUMO

During systemic infection, Staphylococcus aureus acquires nutrient iron from heme, the cofactor of vertebrate myoglobin and hemoglobin. Upon exposure to heme, S. aureus up-regulates the expression of the heme-regulated transporter, HrtAB. Strains lacking hrtAB exhibit increased sensitivity to heme toxicity, and upon heme exposure they elaborate a secreted protein response that interferes with the recruitment of neutrophils to the site of infection. Taken together, these results have led to the suggestion that hrtAB encodes an efflux system responsible for relieving the toxic effects of accumulated heme. Here we extend these observations by demonstrating that HrtA is the ATPase component of the HrtAB transport system. We show that HrtA is an Mn(2+)/Mg(2+)-dependent ATPase that functions at an optimal pH of 7.5 and exhibits in vitro temperature dependence uncommon to ABC transporter ATPases. Furthermore, we identify conserved residues within HrtA that are required for in vitro ATPase activity and are essential for the functionality of HrtA in vivo. Finally, we show that heme induces an alteration in the gene expression pattern of S. aureus Delta hrtA, implying the presence of a novel transcriptional regulatory mechanism responsible for the previously described immunomodulatory characteristics of hrtA mutants exposed to heme.


Assuntos
Adenosina Trifosfatases/genética , Adenosina Trifosfatases/metabolismo , Heme/toxicidade , Staphylococcus aureus/patogenicidade , Transportadores de Cassetes de Ligação de ATP/genética , Transportadores de Cassetes de Ligação de ATP/fisiologia , Transporte Biológico/genética , Transporte Biológico/fisiologia , Regulação Bacteriana da Expressão Gênica , Genes Bacterianos/genética , Heme/fisiologia , Staphylococcus aureus/efeitos dos fármacos , Staphylococcus aureus/crescimento & desenvolvimento , Transcrição Gênica/fisiologia
19.
Cell Host Microbe ; 1(2): 109-19, 2007 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-18005689

RESUMO

Staphylococcus aureus, a bacterium responsible for tremendous morbidity and mortality, exists as a harmless commensal in approximately 25% of humans. Identifying the molecular machinery activated upon infection is central to understanding staphylococcal pathogenesis. We describe the heme sensor system (HssRS) that responds to heme exposure and activates expression of the heme-regulated transporter (HrtAB). Inactivation of the Hss or Hrt systems leads to increased virulence in a vertebrate infection model, a phenotype that is associated with an inhibited innate immune response. We suggest that the coordinated activity of Hss and Hrt allows S. aureus to sense internal host tissues, resulting in tempered virulence to avoid excessive host tissue damage. Further, genomic analyses have identified orthologous Hss and Hrt systems in Bacillus anthracis, Listeria monocytogenes, and Enterococcus faecalis, suggesting a conserved regulatory system by which Gram-positive pathogens sense heme as a molecular marker of internal host tissue and modulate virulence.


Assuntos
Meio Ambiente , Heme/fisiologia , Heme/toxicidade , Staphylococcus aureus/patogenicidade , Aclimatação , Ferro/farmacologia , Staphylococcus aureus/efeitos dos fármacos , Staphylococcus aureus/crescimento & desenvolvimento , Virulência
20.
Proc Natl Acad Sci U S A ; 104(41): 16293-8, 2007 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-17911250

RESUMO

Helicobacter pylori VacA, a pore-forming toxin secreted by an autotransporter pathway, causes multiple alterations in human cells, contributes to the pathogenesis of peptic ulcer disease and gastric cancer, and is a candidate antigen for inclusion in an H. pylori vaccine. Here, we present a 2.4-A crystal structure of the VacA p55 domain, which has an important role in mediating VacA binding to host cells. The structure is predominantly a right-handed parallel beta-helix, a feature that is characteristic of autotransporter passenger domains but unique among known bacterial protein toxins. Notable features of VacA p55 include disruptions in beta-sheet contacts that result in five beta-helix subdomains and a C-terminal domain that contains a disulfide bond. Analysis of VacA protein sequences from unrelated H. pylori strains, including m1 and m2 forms of VacA, allows us to identify structural features of the VacA surface that may be important for interactions with host receptors. Docking of the p55 structure into a 19-A cryo-EM map of a VacA dodecamer allows us to propose a model for how VacA monomers assemble into oligomeric structures capable of membrane channel formation.


Assuntos
Proteínas de Bactérias/química , Helicobacter pylori/química , Proteínas de Bactérias/genética , Cristalografia por Raios X , Genes Bacterianos , Variação Genética , Helicobacter pylori/genética , Helicobacter pylori/patogenicidade , Humanos , Modelos Moleculares , Conformação Proteica , Estrutura Quaternária de Proteína , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...