Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Cell Host Microbe ; 32(3): 411-424.e10, 2024 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-38307020

RESUMO

Intracellular Salmonella experiencing oxidative stress downregulates aerobic respiration. To maintain cellular energetics during periods of oxidative stress, intracellular Salmonella must utilize terminal electron acceptors of lower energetic value than molecular oxygen. We show here that intracellular Salmonella undergoes anaerobic respiration during adaptation to the respiratory burst of the phagocyte NADPH oxidase in macrophages and in mice. Reactive oxygen species generated by phagocytes oxidize methionine, generating methionine sulfoxide. Anaerobic Salmonella uses the molybdenum cofactor-containing DmsABC enzymatic complex to reduce methionine sulfoxide. The enzymatic activity of the methionine sulfoxide reductase DmsABC helps Salmonella maintain an alkaline cytoplasm that supports the synthesis of the antioxidant hydrogen sulfide via cysteine desulfuration while providing a source of methionine and fostering redox balancing by associated dehydrogenases. Our investigations demonstrate that nontyphoidal Salmonella responding to oxidative stress exploits the anaerobic metabolism associated with dmsABC gene products, a pathway that has accrued inactivating mutations in human-adapted typhoidal serovars.


Assuntos
Metionina/análogos & derivados , NADPH Oxidases , Fagócitos , Animais , Camundongos , Humanos , Anaerobiose , Fagócitos/metabolismo , Metionina/metabolismo , Salmonella typhimurium/metabolismo , Respiração
2.
Sci Adv ; 6(9): eaaz0260, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-32133408

RESUMO

Cytostasis is the most salient manifestation of the potent antimicrobial activity of nitric oxide (NO), yet the mechanism by which NO disrupts bacterial cell division is unknown. Here, we show that in respiring Escherichia coli, Salmonella, and Bacillus subtilis, NO arrests the first step in division, namely, the GTP-dependent assembly of the bacterial tubulin homolog FtsZ into a cytokinetic ring. FtsZ assembly fails in respiring cells because NO inactivates inosine 5'-monophosphate dehydrogenase in de novo purine nucleotide biosynthesis and quinol oxidases in the electron transport chain, leading to drastic depletion of nucleoside triphosphates, including the GTP needed for the polymerization of FtsZ. Despite inhibiting respiration and dissipating proton motive force, NO does not destroy Z ring formation and only modestly decreases nucleoside triphosphates in glycolytic cells, which obtain much of their ATP by substrate-level phosphorylation and overexpress inosine 5'-monophosphate dehydrogenase. Purine metabolism dictates the susceptibility of early morphogenic steps in cytokinesis to NO toxicity.


Assuntos
Bacillus subtilis/metabolismo , Citocinese/efeitos dos fármacos , Escherichia coli/metabolismo , Óxido Nítrico/farmacologia , Salmonella/metabolismo , Bacillus subtilis/genética , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Citocinese/genética , Proteínas do Citoesqueleto/genética , Proteínas do Citoesqueleto/metabolismo , Escherichia coli/genética , Consumo de Oxigênio/efeitos dos fármacos , Consumo de Oxigênio/genética , Força Próton-Motriz/efeitos dos fármacos , Força Próton-Motriz/genética , Salmonella/genética
3.
mBio ; 11(1)2020 02 25.
Artigo em Inglês | MEDLINE | ID: mdl-32098823

RESUMO

Guanosine tetraphosphate (ppGpp) and guanosine pentaphosphate (pppGpp), together named (p)ppGpp, regulate diverse aspects of Salmonella pathogenesis, including synthesis of nutrients, resistance to inflammatory mediators, and expression of secretion systems. In Salmonella, these nucleotide alarmones are generated by the synthetase activities of RelA and SpoT proteins. In addition, the (p)ppGpp hydrolase activity of the bifunctional SpoT protein is essential to preserve cell viability. The contribution of SpoT to physiology and pathogenesis has proven elusive in organisms such as Salmonella, because the hydrolytic activity of this RelA and SpoT homologue (RSH) is vital to prevent inhibitory effects of (p)ppGpp produced by a functional RelA. Here, we describe the biochemical and functional characterization of a spoT-Δctd mutant Salmonella strain encoding a SpoT protein that lacks the C-terminal regulatory elements collectively referred to as "ctd." Salmonella expressing the spoT-Δctd variant hydrolyzes (p)ppGpp with similar kinetics to those of wild-type bacteria, but it is defective at synthesizing (p)ppGpp in response to acidic pH. Salmonella spoT-Δctd mutants have virtually normal adaptations to nutritional, nitrosative, and oxidative stresses, but poorly induce metal cation uptake systems and Salmonella pathogenicity island 2 (SPI-2) genes in response to the acidic pH of the phagosome. Importantly, spoT-Δctd mutant Salmonella replicates poorly intracellularly and is attenuated in a murine model of acute salmonellosis. Collectively, these investigations indicate that (p)ppGpp synthesized by SpoT serves a unique function in the adaptation of Salmonella to the intracellular environment of host phagocytes that cannot be compensated by the presence of a functional RelA.IMPORTANCE Pathogenic bacteria experience nutritional challenges during colonization and infection of mammalian hosts. Binding of the alarmone nucleotide guanosine tetraphosphate (ppGpp) to RNA polymerase coordinates metabolic adaptations and virulence gene transcription, increasing the fitness of diverse Gram-positive and Gram-negative bacteria as well as that of actinomycetes. Gammaproteobacteria such as Salmonella synthesize ppGpp by the combined activities of the closely related RelA and SpoT synthetases. Due to its profound inhibitory effects on growth, ppGpp must be removed; in Salmonella, this process is catalyzed by the vital hydrolytic activity of the bifunctional SpoT protein. Because SpoT hydrolase activity is essential in cells expressing a functional RelA, we have a very limited understanding of unique roles these two synthetases may assume during interactions of bacterial pathogens with their hosts. We describe here a SpoT truncation mutant that lacks ppGpp synthetase activity and all C-terminal regulatory domains but retains excellent hydrolase activity. Our studies of this mutant reveal that SpoT uniquely senses the acidification of phagosomes, inducing virulence programs that increase Salmonella fitness in an acute model of infection. Our investigations indicate that the coexistence of RelA/SpoT homologues in a bacterial cell is driven by the need to mount a stringent response to a myriad of physiological and host-specific signatures.


Assuntos
Proteínas de Bactérias/metabolismo , Ligases/metabolismo , Fagossomos/metabolismo , Pirofosfatases/metabolismo , Salmonella/metabolismo , Animais , Sobrevivência Celular , Modelos Animais de Doenças , Bactérias Gram-Negativas/metabolismo , Bactérias Gram-Positivas/metabolismo , Guanosina Pentafosfato/genética , Guanosina Pentafosfato/metabolismo , Guanosina Tetrafosfato/genética , Guanosina Tetrafosfato/metabolismo , Imunidade Inata , Ligases/genética , Camundongos , Pirofosfatases/genética , Salmonella/genética , Fator de Transcrição RelA/metabolismo , Virulência/genética
4.
Sci Rep ; 8(1): 9465, 2018 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-29930310

RESUMO

The repressive activity of ancestral histone-like proteins helps integrate transcription of foreign genes with discrepant AT content into existing regulatory networks. Our investigations indicate that the AT-rich discriminator region located between the -10 promoter element and the transcription start site of the regulatory gene ssrA plays a distinct role in the balanced expression of the Salmonella pathogenicity island-2 (SPI2) type III secretion system. The RNA polymerase-binding protein DksA activates the ssrAB regulon post-transcriptionally, whereas the alarmone guanosine tetraphosphate (ppGpp) relieves the negative regulation imposed by the AT-rich ssrA discriminator region. An increase in the GC-content of the ssrA discriminator region enhances ssrAB transcription and SsrB translation, thus activating the expression of downstream SPI2 genes. A Salmonella strain expressing a GC-rich ssrA discriminator region is attenuated in mice and grows poorly intracellularly. The combined actions of ppGpp and DksA on SPI2 expression enable Salmonella to grow intracellularly, and cause disease in a murine model of infection. Collectively, these findings indicate that (p)ppGpp relieves the negative regulation associated with the AT-rich discriminator region in the promoter of the horizontally-acquired ssrA gene, whereas DksA activates ssrB gene expression post-transcriptionally. The combined effects of (p)ppGpp and DksA on the ssrAB locus facilitate a balanced SPI2 virulence gene transcription that is essential for Salmonella pathogenesis.


Assuntos
Regulação Bacteriana da Expressão Gênica , Ilhas Genômicas , Guanosina Tetrafosfato/metabolismo , Salmonella/genética , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Regiões Promotoras Genéticas , Regulon , Salmonella/metabolismo , Salmonella/patogenicidade , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Sistemas de Secreção Tipo III/genética , Sistemas de Secreção Tipo III/metabolismo , Fatores de Virulência/genética , Fatores de Virulência/metabolismo
5.
mBio ; 9(1)2018 02 27.
Artigo em Inglês | MEDLINE | ID: mdl-29487237

RESUMO

The adaptations that protect pathogenic microorganisms against the cytotoxicity of nitric oxide (NO) engendered in the immune response are incompletely understood. We show here that salmonellae experiencing nitrosative stress suffer dramatic losses of the nucleoside triphosphates ATP, GTP, CTP, and UTP while simultaneously generating a massive burst of the alarmone nucleotide guanosine tetraphosphate. RelA proteins associated with ribosomes overwhelmingly synthesize guanosine tetraphosphate in response to NO as a feedback mechanism to transient branched-chain amino acid auxotrophies. Guanosine tetraphosphate activates the transcription of valine biosynthetic genes, thereby reestablishing branched-chain amino acid biosynthesis that enables the translation of the NO-consuming flavohemoglobin Hmp. Guanosine tetraphosphate synthesized by RelA protects salmonellae from the metabolic stress inflicted by reactive nitrogen species generated in the mammalian host response. This research illustrates the importance of nucleotide metabolism in the adaptation of salmonellae to the nutritional stress imposed by NO released in the innate host response.IMPORTANCE Nitric oxide triggers dramatic drops in nucleoside triphosphates, the building blocks that power DNA replication; RNA transcription; translation; cell division; and the biosynthesis of fatty acids, lipopolysaccharide, and peptidoglycan. Concomitantly, this diatomic gas stimulates a burst of guanosine tetraphosphate. Global changes in nucleotide metabolism may contribute to the potent bacteriostatic activity of nitric oxide. In addition to inhibiting numerous growth-dependent processes, guanosine tetraphosphate positively regulates the transcription of branched-chain amino acid biosynthesis genes, thereby facilitating the translation of antinitrosative defenses that mediate recovery from nitrosative stress.


Assuntos
Óxido Nítrico/toxicidade , Estresse Nitrosativo , Nucleotídeos/metabolismo , Salmonella typhimurium/efeitos dos fármacos , Salmonella typhimurium/fisiologia , Viabilidade Microbiana/efeitos dos fármacos , Estresse Fisiológico
6.
mBio ; 7(6)2016 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-27999164

RESUMO

In the course of an infection, Salmonella enterica occupies diverse anatomical sites with various concentrations of oxygen (O2) and nitric oxide (NO). These diatomic gases compete for binding to catalytic metal groups of quinol oxidases. Enterobacteriaceae express two evolutionarily distinct classes of quinol oxidases that differ in affinity for O2 and NO as well as stoichiometry of H+ translocated across the cytoplasmic membrane. The investigations presented here show that the dual function of bacterial cytochrome bd in bioenergetics and antinitrosative defense enhances Salmonella virulence. The high affinity of cytochrome bd for O2 optimizes respiratory rates in hypoxic cultures, and thus, this quinol oxidase maximizes bacterial growth under O2-limiting conditions. Our investigations also indicate that cytochrome bd, rather than cytochrome bo, is an intrinsic component of the adaptive antinitrosative toolbox of Salmonella Accordingly, induction of cytochrome bd helps Salmonella grow and respire in the presence of inhibitory NO. The combined antinitrosative defenses of cytochrome bd and the flavohemoglobin Hmp account for a great part of the adaptations that help Salmonella recover from the antimicrobial activity of NO. Moreover, the antinitrosative defenses of cytochrome bd and flavohemoglobin Hmp synergize to promote Salmonella growth in systemic tissues. Collectively, our investigations indicate that cytochrome bd is a critical means by which Salmonella resists the nitrosative stress that is engendered in the innate response of mammalian hosts while it concomitantly allows for proper O2 utilization in tissue hypoxia. IMPORTANCE: It is becoming quite apparent that metabolism is critically important to the virulence potential of pathogenic microorganisms. Bacterial cells use a variety of terminal electron acceptors to power electron transport chains and metabolic processes. Of all the electron acceptors available to bacteria, utilization of O2 yields the most energy while diversifying the type of substrates that a pathogen can use. Recent investigations have demonstrated important roles for bd-type quinol oxidases with high affinity for O2 in bacterial pathogenesis. The investigations presented here have revealed that cytochrome bd potentiates virulence of a clinically relevant bacterial pathogen by fueling bioenergetics of prokaryotic cells while protecting the respiratory chain against NO toxicity. The adaptive antinitrosative defenses afforded by cytochrome bd synergize with other NO-detoxifying systems to preserve cellular bioenergetics, thereby promoting bacterial virulence in tissue hypoxia.


Assuntos
Grupo dos Citocromos b/metabolismo , Grupo dos Citocromos d/metabolismo , Metabolismo Energético , Óxido Nítrico/metabolismo , Oxigênio/metabolismo , Salmonella enterica/metabolismo , Salmonella enterica/patogenicidade , Animais , Complexo de Proteínas da Cadeia de Transporte de Elétrons , Humanos , Hipóxia , Imunidade Inata , Oxirredução , Oxirredutases/metabolismo , Consumo de Oxigênio , Salmonella enterica/crescimento & desenvolvimento , Estresse Fisiológico
7.
PLoS Negl Trop Dis ; 8(8): e3079, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25121731

RESUMO

Our investigations show that nonlethal concentrations of nitric oxide (NO) abrogate the antibiotic activity of ß-lactam antibiotics against Burkholderia pseudomallei, Escherichia coli and nontyphoidal Salmonella enterica serovar Typhimurium. NO protects B. pseudomallei already exposed to ß-lactams, suggesting that this diatomic radical tolerizes bacteria against the antimicrobial activity of this important class of antibiotics. The concentrations of NO that elicit antibiotic tolerance repress consumption of oxygen (O2), while stimulating hydrogen peroxide (H2O2) synthesis. Transposon insertions in genes encoding cytochrome c oxidase-related functions and molybdenum assimilation confer B. pseudomallei a selective advantage against the antimicrobial activity of the ß-lactam antibiotic imipenem. Cumulatively, these data support a model by which NO induces antibiotic tolerance through the inhibition of the electron transport chain, rather than by potentiating antioxidant defenses as previously proposed. Accordingly, pharmacological inhibition of terminal oxidases and nitrate reductases tolerizes aerobic and anaerobic bacteria to ß-lactams. The degree of NO-induced ß-lactam antibiotic tolerance seems to be inversely proportional to the proton motive force (PMF), and thus the dissipation of ΔH+ and ΔΨ electrochemical gradients of the PMF prevents ß-lactam-mediated killing. According to this model, NO generated by IFNγ-primed macrophages protects intracellular Salmonella against imipenem. On the other hand, sublethal concentrations of imipenem potentiate the killing of B. pseudomallei by NO generated enzymatically from IFNγ-primed macrophages. Our investigations indicate that NO modulates the antimicrobial activity of ß-lactam antibiotics.


Assuntos
Antibacterianos/farmacologia , Burkholderia pseudomallei/efeitos dos fármacos , Imipenem/farmacologia , Interferon gama/farmacologia , Macrófagos/imunologia , Óxido Nítrico/fisiologia , Salmonella typhimurium/efeitos dos fármacos , Animais , Células Cultivadas , Camundongos , Estresse Oxidativo , Força Próton-Motriz
8.
Infect Immun ; 82(1): 333-40, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24166960

RESUMO

Herein we report an important role for the ferric uptake regulator (Fur) in the resistance of Salmonella enterica serovar Typhimurium to the reactive nitrogen species produced by inducible nitric oxide (NO) synthase in an NRAMP1(r) murine model of acute systemic infection. The expression of fur protected Salmonella grown under normoxic and hypoxic conditions against the bacteriostatic activity of NO. The hypersusceptibility of fur-deficient Salmonella to the cytotoxic actions of NO coincides with a marked repression of respiratory activity and the reduced ability of the bacteria to detoxify NO. A fur mutant Salmonella strain contained reduced levels of the terminal quinol oxidases of the electron transport chain. Addition of the heme precursor δ-aminolevulinic acid restored the cytochrome content, respiratory activity, NO consumption, and wild-type growth in bacteria undergoing nitrosative stress. The innate antinitrosative defenses regulated by Fur added to the adaptive response associated with the NO-detoxifying activity of the flavohemoprotein Hmp. Our investigations indicate that, in addition to playing a critical role in iron homeostasis, Fur is an important antinitrosative determinant of Salmonella pathogenesis.


Assuntos
Proteínas de Bactérias/fisiologia , Proteínas Repressoras/fisiologia , Infecções por Salmonella/microbiologia , Salmonella typhimurium/patogenicidade , Análise de Variância , Animais , Proteínas de Bactérias/imunologia , Modelos Animais de Doenças , Camundongos , Óxido Nítrico Sintase Tipo II , Estresse Oxidativo/fisiologia , Proteínas Repressoras/deficiência , Proteínas Repressoras/imunologia , Infecções por Salmonella/fisiopatologia , Salmonella typhimurium/imunologia , Estresse Fisiológico/fisiologia
9.
Mol Microbiol ; 87(3): 609-22, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23217033

RESUMO

We found herein that the intracytoplasmic pool of the low-molecular-weight (LMW) thiol glutathione (GSH) is readily oxidized in Salmonella exposed to nitric oxide (NO). The hypersusceptibility of gshA and gshB mutants lacking γ-glutamylcysteine and glutathione synthetases to NO and S-nitrosoglutathione indicates that GSH antagonizes the bacteriostatic activity of reactive nitrogen species. Metabolites of the GSH biosynthetic pathway do not affect the enzymatic activity of classical NO targets such as quinol oxidases. In contrast, LMW thiols diminish the nitrosative stress experienced by enzymes, such as glutamine oxoglutarate amidotransferase, that contain redox active cysteines. LMW thiols also preserve the transcription of Salmonella pathogenicity island 2 gene targets from the inhibitory activity of nitrogen oxides. These findings are consistent with the idea that GSH scavenges reactive nitrogen species (RNS) other than NO. Compared with the adaptive response afforded by inducible systems such as the hmp-encoded flavohaemoprotein, gshA, encoding the first step of GSH biosynthesis, is constitutively expressed in Salmonella. An acute model of salmonellosis has revealed that the antioxidant and antinitrosative properties associated with the GSH biosynthetic pathway represent a first line of Salmonella resistance against reactive oxygen and nitrogen species engendered in the context of a functional NRAMP1(R) divalent metal transporter.


Assuntos
Glutationa/metabolismo , Óxido Nítrico/metabolismo , Oxidantes/metabolismo , Salmonella/fisiologia , Compostos de Sulfidrila/metabolismo , Peso Molecular , Óxido Nítrico/antagonistas & inibidores , Nitrosação , Oxidantes/antagonistas & inibidores , Oxirredução , Espécies Reativas de Nitrogênio/antagonistas & inibidores , Espécies Reativas de Oxigênio/antagonistas & inibidores , Salmonella/efeitos dos fármacos , Salmonella/metabolismo
10.
Nitric Oxide ; 27(1): 25-31, 2012 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-22521523

RESUMO

Burkholderia pseudomallei infections are fastidious to treat with conventional antibiotic therapy, often involving a combination of drugs and long-term regimes. Bacterial genetic determinants contribute to the resistance of B. pseudomallei to many classes of antibiotics. In addition, anaerobiosis and hypoxia in abscesses typical of melioidosis select for persistent populations of B. pseudomallei refractory to a broad spectrum of antibacterials. We tested the susceptibility of B. pseudomallei to the drugs hydroxyurea, spermine NONOate and DETA NONOate that release nitric oxide (NO). Our investigations indicate that B. pseudomallei are killed by NO in a concentration and time-dependent fashion. The cytoxicity of this diatomic radical against B. pseudomallei depends on both the culture medium and growth phase of the bacteria. Rapidly growing, but not stationary phase, B. pseudomallei are readily killed upon exposure to the NO donor spermine NONOate. NO also has excellent antimicrobial activity against anaerobic B. pseudomallei. In addition, persistent bacteria highly resistant to most conventional antibiotics are remarkably susceptible to NO. Sublethal concentrations of NO inhibited the enzymatic activity of [4Fe-4S]-cofactored aconitase of aerobic and anaerobic B. pseudomallei. The strong anti-B. pseudomallei activity of NO described herein merits further studies on the application of NO-based antibiotics for the treatment of melioidosis.


Assuntos
Antibacterianos/farmacologia , Burkholderia pseudomallei/efeitos dos fármacos , Óxido Nítrico/farmacologia , Aconitato Hidratase/efeitos dos fármacos , Aconitato Hidratase/metabolismo , Anaerobiose , Antibacterianos/metabolismo , Burkholderia pseudomallei/fisiologia , Meios de Cultura , Humanos , Melioidose/microbiologia , Testes de Sensibilidade Microbiana , Óxido Nítrico/metabolismo , Doadores de Óxido Nítrico/metabolismo , Doadores de Óxido Nítrico/farmacologia , Compostos Nitrosos/metabolismo , Compostos Nitrosos/farmacologia , Espermina/análogos & derivados , Espermina/metabolismo , Espermina/farmacologia
11.
PLoS One ; 6(6): e21523, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21720554

RESUMO

Burkholderia pseudomallei and Burkholderia mallei are category B select agents and must be studied under BSL3 containment in the United States. They are typically resistant to multiple antibiotics, and the antibiotics used to treat B. pseudomallei or B. mallei infections may not be used as selective agents with the corresponding Burkholderia species. Here, we investigated alanine racemase deficient mutants of B. pseudomallei and B. mallei for development of non-antibiotic-based genetic selection methods and for attenuation of virulence. The genome of B. pseudomallei K96243 has two annotated alanine racemase genes (bpsl2179 and bpss0711), and B. mallei ATCC 23344 has one (bma1575). Each of these genes encodes a functional enzyme that can complement the alanine racemase deficiency of Escherichia coli strain ALA1. Herein, we show that B. pseudomallei with in-frame deletions in both bpsl2179 and bpss0711, or B. mallei with an in-frame deletion in bma1575, requires exogenous D-alanine for growth. Introduction of bpsl2179 on a multicopy plasmid into alanine racemase deficient variants of either Burkholderia species eliminated the requirement for D-alanine. During log phase growth without D-alanine, the viable counts of alanine racemase deficient mutants of B. pseudomallei and B. mallei decreased within 2 hours by about 1000-fold and 10-fold, respectively, and no viable bacteria were present at 24 hours. We constructed several genetic tools with bpsl2179 as a selectable genetic marker, and we used them without any antibiotic selection to construct an in-frame ΔflgK mutant in the alanine racemase deficient variant of B. pseudomallei K96243. In murine peritoneal macrophages, wild type B. mallei ATCC 23344 was killed much more rapidly than wild type B. pseudomallei K96243. In addition, the alanine racemase deficient mutant of B. pseudomallei K96243 exhibited attenuation versus its isogenic parental strain with respect to growth and survival in murine peritoneal macrophages.


Assuntos
Alanina Racemase/genética , Antibacterianos/farmacologia , Burkholderia mallei/enzimologia , Burkholderia pseudomallei/enzimologia , Mutação/genética , Alanina/farmacologia , Alanina Racemase/química , Sequência de Aminoácidos , Animais , Burkholderia mallei/efeitos dos fármacos , Burkholderia mallei/genética , Burkholderia mallei/ultraestrutura , Burkholderia pseudomallei/efeitos dos fármacos , Burkholderia pseudomallei/genética , Burkholderia pseudomallei/ultraestrutura , Deleção de Genes , Genes Bacterianos/genética , Loci Gênicos/genética , Marcadores Genéticos , Macrófagos Peritoneais/efeitos dos fármacos , Macrófagos Peritoneais/microbiologia , Macrófagos Peritoneais/ultraestrutura , Camundongos , Viabilidade Microbiana/efeitos dos fármacos , Dados de Sequência Molecular , Ácido Periódico/farmacologia , Plasmídeos/genética , Reação em Cadeia da Polimerase , Reprodutibilidade dos Testes , Alinhamento de Sequência
12.
BMC Microbiol ; 11: 58, 2011 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-21418628

RESUMO

BACKGROUND: Salmonella enterica serovar Typhimurium (S. Typhimurium) is a Gram-negative pathogen that must successfully adapt to the broad fluctuations in the concentration of dissolved dioxygen encountered in the host. In Escherichia coli, ArcA (Aerobic Respiratory Control) helps the cells to sense and respond to the presence of dioxygen. The global role of ArcA in E. coli is well characterized; however, little is known about its role in anaerobically grown S. Typhimurium. RESULTS: We compared the transcriptional profiles of the virulent wild-type (WT) strain (ATCC 14028s) and its isogenic arcA mutant grown under anaerobic conditions. We found that ArcA directly or indirectly regulates 392 genes (8.5% of the genome); of these, 138 genes are poorly characterized. Regulation by ArcA in S. Typhimurium is similar, but distinct from that in E. coli. Thus, genes/operons involved in core metabolic pathways (e.g., succinyl-CoA, fatty acid degradation, cytochrome oxidase complexes, flagellar biosynthesis, motility, and chemotaxis) were regulated similarly in the two organisms. However, genes/operons present in both organisms, but regulated differently by ArcA in S. Typhimurium included those coding for ethanolamine utilization, lactate transport and metabolism, and succinate dehydrogenases. Salmonella-specific genes/operons regulated by ArcA included those required for propanediol utilization, flagellar genes (mcpAC, cheV), Gifsy-1 prophage genes, and three SPI-3 genes (mgtBC, slsA, STM3784). In agreement with our microarray data, the arcA mutant was non-motile, lacked flagella, and was as virulent in mice as the WT. Additionally, we identified a set of 120 genes whose regulation was shared with the anaerobic redox regulator, Fnr. CONCLUSION(S): We have identified the ArcA regulon in anaerobically grown S. Typhimurium. Our results demonstrated that in S. Typhimurium, ArcA serves as a transcriptional regulator coordinating cellular metabolism, flagella biosynthesis, and motility. Furthermore, ArcA and Fnr share in the regulation of 120 S. Typhimurium genes.


Assuntos
Regulação Bacteriana da Expressão Gênica , Regulon , Proteínas Repressoras/metabolismo , Salmonella typhimurium/crescimento & desenvolvimento , Salmonella typhimurium/genética , Anaerobiose , Animais , Feminino , Deleção de Genes , Perfilação da Expressão Gênica , Redes e Vias Metabólicas/genética , Camundongos , Camundongos Endogâmicos C57BL , Análise em Microsséries , Proteínas Repressoras/genética , Salmonella typhimurium/metabolismo
13.
J Bacteriol ; 193(2): 497-505, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21075923

RESUMO

Iron is an essential element for the survival of living cells. However, excess iron is toxic, and its uptake is exquisitely regulated by the ferric uptake regulator, Fur. In Salmonella, the Salmonella pathogenicity island 1 (SPI-1) encodes a type three secretion system, which is required for invasion of host epithelial cells in the small intestine. A major activator of SPI-1 is HilA, which is encoded within SPI-1. One known regulator of hilA is Fur. The mechanism of hilA regulation by Fur is unknown. We report here that Fur is required for virulence in Salmonella enterica serovar Typhimurium and that Fur is required for the activation of hilA, as well as of other HilA-dependent genes, invF and sipC. The Fur-dependent regulation of hilA was independent of PhoP, a known repressor of hilA. Instead, the expression of the gene coding for the histone-like protein, hns, was significantly derepressed in the fur mutant. Indeed, the activation of hilA by Fur was dependent on 28 nucleotides located upstream of hns. Moreover, we used chromatin immunoprecipitation to show that Fur bound, in vivo, to the upstream region of hns in a metal-dependent fashion. Finally, deletion of fur in an hns mutant resulted in Fur-independent activation of hilA. In conclusion, Fur activates hilA by repressing the expression of hns.


Assuntos
Proteínas de Bactérias/biossíntese , Proteínas de Bactérias/metabolismo , Proteínas de Ligação a DNA/metabolismo , Regulação Bacteriana da Expressão Gênica , Proteínas Repressoras/metabolismo , Salmonella typhimurium/fisiologia , Transativadores/biossíntese , Fatores de Virulência/biossíntese , Animais , Proteínas de Bactérias/genética , Imunoprecipitação da Cromatina , DNA Bacteriano/metabolismo , Proteínas de Ligação a DNA/biossíntese , Modelos Animais de Doenças , Técnicas de Inativação de Genes , Genes Bacterianos , Ilhas Genômicas , Camundongos , Camundongos Endogâmicos C3H , Óperon , Ligação Proteica , Proteínas Repressoras/genética , Salmonelose Animal/microbiologia , Salmonella typhimurium/patogenicidade , Análise de Sobrevida , Fatores de Transcrição/biossíntese , Virulência
14.
Proc Natl Acad Sci U S A ; 107(32): 14396-401, 2010 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-20660761

RESUMO

We show herein that the Salmonella pathogenicity island 2 (SPI2) response regulator SsrB undergoes S-nitrosylation upon exposure of Salmonella to acidified nitrite, a signal encountered by this enteropathogen in phagosomes of macrophages. Mutational analysis has identified Cys(203) in the C-terminal dimerization domain of SsrB as the redox-active residue responding to nitric oxide (NO) congeners generated in the acidification of nitrite. Peroxynitrite and products of the autooxidation of NO in the presence of oxygen, but not hydrogen peroxide, inhibit the DNA-binding capacity of SsrB, demonstrating the selectivity of the reaction of Cys(203) with reactive nitrogen species (RNS). These findings identify the two-component response regulator SsrB Cys(203) as a thiol-based redox sensor. A C203S substitution protects SsrB against the attack of RNS while preserving its DNA-binding capacity. When exposed to SPI2-inducing conditions, Salmonella expressing the wild-type ssrB allele or the ssrB C203S variant sustain transcription of the sifA, sspH2, and srfJ effector genes. Nonetheless, compared with the strain expressing a redox-resistant SsrB C203S variant, wild-type Salmonella bearing the NO-responsive allele exhibit increased fitness when exposed to RNS in an NRAMP(R), C3H/HeN murine model of acute oral infection. Given the widespread occurrence of the wild-type allele in Salmonella enterica, these findings indicate that SsrB Cys(203) increases Salmonella virulence by serving as a redox sensor of NO resulting from the host immune response to oral infection.


Assuntos
Proteínas de Bactérias/fisiologia , Óxido Nítrico/metabolismo , Salmonella typhimurium/patogenicidade , Fatores de Transcrição/fisiologia , Interações Hospedeiro-Patógeno , Imunidade , Oxirredução , Infecções por Salmonella
15.
PLoS One ; 3(4): e1976, 2008 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-18398486

RESUMO

BACKGROUND: Much remains to be known about the mechanisms by which O(2)-dependent host defenses mediate broad antimicrobial activity. METHODOLOGY/PRINCIPAL FINDINGS: We show herein that reactive nitrogen species (RNS) generated by inducible nitric oxide (NO) synthase (iNOS) account for the anti-Burkholderia mallei activity of IFNgamma-primed macrophages. Inducible NOS-mediated intracellular killing may represent direct bactericidal activity, because B. mallei showed an exquisite sensitivity to NO generated chemically. Exposure of B. mallei to sublethal concentrations of NO upregulated transcription of [Fe-S] cluster repair genes, while damaging the enzymatic activity of the [Fe-S] protein aconitase. To test whether [Fe-S] clusters are critical targets for RNS-dependent killing of B. mallei, a mutation was constructed in the NO-induced, [Fe-S] cluster repair regulator iscR. Not only was the iscR mutant hypersusceptible to iNOS-mediated killing, but its aconitase pool was readily oxidized by NO donors as compared to wild-type controls. Although killed by authentic H(2)O(2), which also oxidizes [Fe-S] clusters, B. mallei appear to be resilient to NADPH oxidase-mediated cytotoxicity. The poor respiratory burst elicited by this bacterium likely explains why the NADPH oxidase is nonessential to the killing of B. mallei while it is still confined within phagosomes. CONCLUSIONS/SIGNIFICANCE: Collectively, these findings have revealed a disparate role for NADPH oxidase and iNOS in the innate macrophage response against the strict aerobe B. mallei. To the best of our knowledge, this is the first instance in which disruption of [Fe-S] clusters is demonstrated as cause of the bactericidal activity of NO congeners.


Assuntos
Burkholderia mallei/metabolismo , Proteínas Ferro-Enxofre/química , Óxido Nítrico/metabolismo , Aconitato Hidratase/metabolismo , Animais , Humanos , Peróxido de Hidrogênio/farmacologia , Macrófagos/metabolismo , Macrófagos/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Modelos Biológicos , Modelos Genéticos , NADPH Oxidases/metabolismo , Oxigênio/metabolismo , Fagossomos/metabolismo , Superóxidos/metabolismo
16.
Methods Enzymol ; 437: 521-38, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18433645

RESUMO

Nitric oxide (NO*) is a critical component of mammalian host defense that is produced in macrophages and other cells comprising the innate immune system. Isolated mammalian macrophages have been utilized to measure the kinetics of NO production and to demonstrate NO-related antimicrobial actions. Some microorganisms possess enzymes to detoxify nitrogen oxides, and mutant strains lacking these enzymes can be used to demonstrate the importance of these mechanisms for intracellular bacterial survival. This chapter describes techniques with which to analyze the antimicrobial actions of nitric oxide in murine and human macrophages and in laboratory mice.


Assuntos
Anti-Infecciosos/farmacologia , Macrófagos/efeitos dos fármacos , Óxido Nítrico/farmacologia , Animais , Humanos , Ativação de Macrófagos/efeitos dos fármacos , Camundongos , Camundongos Knockout , Testes de Sensibilidade Microbiana/métodos , Viabilidade Microbiana/efeitos dos fármacos , Óxido Nítrico Sintase Tipo II/genética , Salmonelose Animal/metabolismo , Salmonella enterica
17.
J Biol Chem ; 283(12): 7682-9, 2008 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-18198179

RESUMO

Aerobic metabolism generates biologically challenging reactive oxygen species (ROS) by the endogenous autooxidation of components of the electron transport chain (ETC). Basal levels of oxidative stress can dramatically rise upon activation of the NADPH oxidase-dependent respiratory burst. To minimize ROS toxicity, prokaryotic and eukaryotic organisms express a battery of low-molecular-weight thiol scavengers, a legion of detoxifying catalases, peroxidases, and superoxide dismutases, as well as a variety of repair systems. We present herein blockage of bacterial respiration as a novel strategy that helps the intracellular pathogen Salmonella survive extreme oxidative stress conditions. A Salmonella strain bearing mutations in complex I NADH dehydrogenases is refractory to the early NADPH oxidase-dependent antimicrobial activity of IFNgamma-activated macrophages. The ability of NADH-rich, complex I-deficient Salmonella to survive oxidative stress is associated with resistance to peroxynitrite (ONOO(-)) and hydrogen peroxide (H(2)O(2)). Inhibition of respiration with nitric oxide (NO) also triggered a protective adaptive response against oxidative stress. Expression of the NDH-II dehydrogenase decreases NADH levels, thereby abrogating resistance of NO-adapted Salmonella to H(2)O(2). NADH antagonizes the hydroxyl radical (OH(.)) generated in classical Fenton chemistry or spontaneous decomposition of peroxynitrous acid (ONOOH), while fueling AhpCF alkylhydroperoxidase. Together, these findings identify the accumulation of NADH following the NO-mediated inhibition of Salmonella's ETC as a novel antioxidant strategy. NO-dependent respiratory arrest may help mitochondria and a plethora of organisms cope with oxidative stress engendered in situations as diverse as aerobic respiration, ischemia reperfusion, and inflammation.


Assuntos
Proteínas de Bactérias/metabolismo , Macrófagos Peritoneais/enzimologia , NADPH Desidrogenase/metabolismo , Óxido Nítrico/metabolismo , Estresse Oxidativo , Peroxirredoxinas/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Explosão Respiratória , Salmonelose Animal/enzimologia , Salmonella typhimurium/enzimologia , Animais , Proteínas de Bactérias/genética , Resistência Microbiana a Medicamentos/genética , Complexo de Proteínas da Cadeia de Transporte de Elétrons/genética , Complexo de Proteínas da Cadeia de Transporte de Elétrons/metabolismo , Interferon gama/farmacologia , Macrófagos Peritoneais/microbiologia , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , NADPH Desidrogenase/genética , NADPH Oxidase 2 , NADPH Oxidases/genética , NADPH Oxidases/metabolismo , Óxido Nítrico Sintase Tipo II/genética , Óxido Nítrico Sintase Tipo II/metabolismo , Estresse Oxidativo/genética , Peroxirredoxinas/genética , Ácido Peroxinitroso/farmacologia , Explosão Respiratória/genética , Salmonelose Animal/genética , Salmonella typhimurium/genética
18.
Immunobiology ; 212(9-10): 759-69, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-18086377

RESUMO

We show here that the nitric oxide (NO)-detoxifying Hmp flavohemoprotein increases by 3-fold the transcription of the Salmonella pathogenicity island 2 (SPI2) in macrophages expressing a functional inducible NO synthase (iNOS). However, Hmp does not prevent NO-related repression of SPI2 transcription in IFNgamma-primed phagocytes, despite preserving intracellular transcription of sdhA sdhB subunits of Salmonella succinate dehydrogenase within both control and IFNgamma-primed phagocytes. To shed light into the seemingly paradoxical role that Hmp plays in protecting intracellular SPI2 expression in various populations of macrophages, N(2)O(3) was quantified as an indicator of the nitrosative potential of Salmonella-infected phagocytes in different states of activation. Hmp was found to prevent the formation of 300nM N(2)O(3)/h/bacteria in IFNgamma-primed macrophages, accounting for about a 60% reduction of the nitrosative power of activated phagocytes. Utilization of the vacuolar ATPase inhibitor bafilomycin indicates that a fourth of the approximately 200nM N(2)O(3)/h sustained by IFNgamma-primed macrophages is generated in endosomal compartments via condensation of HNO(2). In sharp contrast, control macrophages infected with wild-type Salmonella produce as little N(2)O(3) as iNOS-deficient controls. Collectively, these findings indicate that the NO-metabolizing activity of Salmonella Hmp is functional in both control and IFNgamma-primed macrophages. Nonetheless, a substantial amount of the NO generated by IFNgamma-primed macrophages gives rise to N(2)O(3), a species that not only enhances the nitrosative potential of activated phagocytes but also avoids detoxification by Salmonella Hmp.


Assuntos
Interferon gama/imunologia , Ativação de Macrófagos , Macrófagos/imunologia , Macrófagos/metabolismo , Óxidos de Nitrogênio/metabolismo , Salmonella typhimurium/imunologia , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Flavoproteínas/genética , Flavoproteínas/metabolismo , Hemeproteínas/metabolismo , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , Fagócitos/imunologia , Fagócitos/metabolismo , Succinato Desidrogenase/genética , Succinato Desidrogenase/metabolismo
19.
Infect Immun ; 75(11): 5346-52, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17698574

RESUMO

We have identified acid sphingomyelinase (ASM) as an important player in the early and late anti-Salmonella activity of macrophages. A functional ASM participated in the killing activity of macrophages against wild-type Salmonella enterica serovar Typhimurium. The role of ASM in early macrophage killing of Salmonella appears to be linked to an active NADPH phagocyte oxidase enzymatic complex, since the flavoprotein inhibitor diphenyleneiodonium not only blocked a productive respiratory burst but also abrogated the survival advantage of Salmonella in macrophages lacking ASM. Lack of ASM activity also increased the intracellular survival of an isogenic DeltaspiC::FRT Salmonella strain deficient in a translocator and effector of the Salmonella pathogenicity island 2 (SPI2) type III secretion system, suggesting that the antimicrobial activity associated with ASM is manifested regardless of the SPI2 status of the bacteria. Constitutively expressed ASM is responsible for the role that this lipid-metabolizing hydrolase plays in the innate host defense of macrophages against Salmonella. Accordingly, the ASM activity and intracellular concentration and composition of ceramide, gangliosides, and neutral sphingolipids did not increase upon Salmonella infection. Salmonella triggered, nonetheless, a significant increase in the secreted fraction of ASM. Collectively, these findings have elucidated a novel role for constitutive ASM in the anti-Salmonella activity of murine macrophages.


Assuntos
Macrófagos/imunologia , Viabilidade Microbiana/imunologia , Salmonella typhimurium/imunologia , Esfingomielina Fosfodiesterase/imunologia , Animais , Proteínas de Bactérias/genética , Ceramidas/análise , Citosol/química , Gangliosídeos/análise , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , NADPH Oxidases/antagonistas & inibidores , NADPH Oxidases/imunologia , Espécies Reativas de Oxigênio/metabolismo , Esfingolipídeos/análise , Esfingomielina Fosfodiesterase/deficiência , Esfingomielina Fosfodiesterase/genética , Esfingomielina Fosfodiesterase/metabolismo
20.
Infect Immun ; 75(6): 2708-16, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17403871

RESUMO

To better understand the evolution of a systemic memory response to a mucosal pathogen, we monitored antigen-specific OT1 CD8 T-cell responses to a fusion of the SspH2 protein and the peptide SIINFEKL stably expressed from the chromosome of Salmonella enterica and loaded into the class I pathway of antigen presentation of professional phagocytes through the Salmonella pathogenicity island 2 type III secretion system (TTSS). This strategy has revealed that effector memory CD8 T cells with low levels of CD62L expression (CD62L(low)) are maintained in systemic sites months after vaccination in response to low-grade infections with Salmonella. However, the CD8 T-cell pool eventually declines. Low numbers of central memory cells surviving after prolonged resting from an antigen encounter can nevertheless reconstitute the systemic effector memory pool in a route-specific recall response to cognate antigens encountered in the gut. Accordingly, populations of CD62L(high) interleukin-7 receptor-positive progenitor central memory cells grafted into naïve mice expand in response to orally administered Salmonella expressing the chromosomal translational fusion of sspH2 and the sequence encoding the SIINFEKL peptide but fail to proliferate following systemic stimulation. Moreover, populations of systemic memory CD8 T cells restricted to Salmonella in oral vaccines selectively expand in response to cognate antigens presented by cells isolated from mesenteric lymph nodes (MLN). Together, these findings have revealed the imprinting of systemic CD8 central memory T-cell recall responses against enteropathogens by MLN. MLN restriction represents a novel mechanism by which systemic CD8 T-cell immunity is confined to periods of high risk for extraintestinal dissemination.


Assuntos
Proteínas de Bactérias/metabolismo , Linfócitos T CD8-Positivos/imunologia , Mucosa Gástrica/imunologia , Memória Imunológica , Proteínas de Membrana/metabolismo , Infecções por Salmonella/imunologia , Salmonella enterica/imunologia , Animais , Mucosa Gástrica/microbiologia , Camundongos , Especificidade de Órgãos , Infecções por Salmonella/microbiologia , Infecções por Salmonella/patologia , Salmonella enterica/classificação , Salmonella enterica/genética , Salmonella enterica/patogenicidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA