Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 50
Filter
Add more filters










Publication year range
1.
Cell Host Microbe ; 32(3): 411-424.e10, 2024 Mar 13.
Article in English | MEDLINE | ID: mdl-38307020

ABSTRACT

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.


Subject(s)
Methionine/analogs & derivatives , NADPH Oxidases , Phagocytes , Animals , Mice , Humans , Anaerobiosis , Phagocytes/metabolism , Methionine/metabolism , Salmonella typhimurium/metabolism , Respiration
2.
Cardiovasc Res ; 119(12): 2157-2164, 2023 10 16.
Article in English | MEDLINE | ID: mdl-37517059

ABSTRACT

Electronic cigarette use has grown exponentially in recent years, and while their popularity has increased, the long-term effects on the heart are yet to be fully studied and understood. Originally designed as devices to assist with those trying to quit traditional combustible cigarette use, their popularity has attracted use by teens and adolescents who traditionally have not smoked combustible cigarettes. Acute effects on the heart have been shown to be similar to traditional combustible cigarettes, including increased heart rate and blood pressure. The main components of electronic cigarettes that contribute to these arrhythmic effects are found in the e-liquid that is aerosolized and inhaled, comprised of nicotine, flavourings, and a combination of vegetable glycerin (VG) and propylene glycol (PG). Nicotine can potentially induce both ventricular and atrial arrhythmogenesis, with both the atrial and ventricular effects resulting from the interactions of nicotine and the catecholamines they release via potassium channels. Atrial arrhythmogenesis, more specifically atrial fibrillation, can also occur due to structural alterations, which happens because of nicotine downregulating microRNAs 133 and 590, both post-transcriptional growth factor repressors. Liquid flavourings and the combination of PG and VG can possibly lead to arrhythmic events by exposing users to acrolein, an aldehyde that stimulates TRPA1 that in turn causes a change towards sympathetic activation and autonomic imbalance. The design of these electronic delivery devices is constantly changing; therefore, it has proven extremely difficult to study the long-term effects on the heart caused by electronic cigarettes but will be important to understand given their rising popularity. The arrhythmic effects of electronic cigarettes appear similar to traditional cigarettes as well; however, a comprehensive review has not been compiled and is the focus of this article.


Subject(s)
Atrial Fibrillation , Electronic Nicotine Delivery Systems , Adolescent , Humans , Nicotine/adverse effects , Propylene Glycol , Glycerol
3.
Viruses ; 15(1)2022 12 30.
Article in English | MEDLINE | ID: mdl-36680147

ABSTRACT

HIV/AIDS mortality has been decreasing over the last decade. While promising, this decrease correlated directly with increased use of antiretroviral drugs. As a natural consequence of its high mutation rate, treatments provide selection pressure that promotes the natural selection of escape mutants. Individuals may acquire drug-naive strains, or those that have already mutated due to treatment. Even within a host, mutation affects HIV tropism, where initial infection begins with R5-tropic virus, but the clinical transition to AIDS correlates with mutations that lead to an X4-tropic switch. Furthermore, the high mutation rate of HIV has spelled failure for all attempts at an effective vaccine. Pre-exposure drugs are currently the most effective drug-based preventatives, but their effectiveness is also threatened by viral mutation. From attachment and entry to assembly and release, the steps in the replication cycle are also discussed to describe the drug mechanisms and mutations that arise due to those drugs. Revealing the patterns of HIV-1 mutations, their effects, and the coordinated attempt to understand and control them will lead to effective use of current preventative measures and treatment options, as well as the development of new ones.


Subject(s)
HIV Infections , HIV Seropositivity , HIV , Humans , HIV Infections/drug therapy , Mutation , Viral Tropism/genetics , Virus Replication , HIV/drug effects , HIV/genetics , Anti-HIV Agents/therapeutic use
4.
Sci Adv ; 6(9): eaaz0260, 2020 02.
Article in English | MEDLINE | ID: mdl-32133408

ABSTRACT

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.


Subject(s)
Bacillus subtilis/metabolism , Cytokinesis/drug effects , Escherichia coli/metabolism , Nitric Oxide/pharmacology , Salmonella/metabolism , Bacillus subtilis/genetics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cytokinesis/genetics , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Escherichia coli/genetics , Oxygen Consumption/drug effects , Oxygen Consumption/genetics , Proton-Motive Force/drug effects , Proton-Motive Force/genetics , Salmonella/genetics
5.
mBio ; 11(1)2020 02 25.
Article in English | MEDLINE | ID: mdl-32098823

ABSTRACT

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.


Subject(s)
Bacterial Proteins/metabolism , Ligases/metabolism , Phagosomes/metabolism , Pyrophosphatases/metabolism , Salmonella/metabolism , Animals , Cell Survival , Disease Models, Animal , Gram-Negative Bacteria/metabolism , Gram-Positive Bacteria/metabolism , Guanosine Pentaphosphate/genetics , Guanosine Pentaphosphate/metabolism , Guanosine Tetraphosphate/genetics , Guanosine Tetraphosphate/metabolism , Immunity, Innate , Ligases/genetics , Mice , Pyrophosphatases/genetics , Salmonella/genetics , Transcription Factor RelA/metabolism , Virulence/genetics
6.
Sci Rep ; 8(1): 9465, 2018 06 21.
Article in English | MEDLINE | ID: mdl-29930310

ABSTRACT

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.


Subject(s)
Gene Expression Regulation, Bacterial , Genomic Islands , Guanosine Tetraphosphate/metabolism , Salmonella/genetics , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Mice , Mice, Inbred C57BL , Promoter Regions, Genetic , Regulon , Salmonella/metabolism , Salmonella/pathogenicity , Transcription Factors/genetics , Transcription Factors/metabolism , Type III Secretion Systems/genetics , Type III Secretion Systems/metabolism , Virulence Factors/genetics , Virulence Factors/metabolism
7.
mBio ; 9(1)2018 02 27.
Article in English | MEDLINE | ID: mdl-29487237

ABSTRACT

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.


Subject(s)
Nitric Oxide/toxicity , Nitrosative Stress , Nucleotides/metabolism , Salmonella typhimurium/drug effects , Salmonella typhimurium/physiology , Microbial Viability/drug effects , Stress, Physiological
8.
mBio ; 7(6)2016 12 20.
Article in English | MEDLINE | ID: mdl-27999164

ABSTRACT

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.


Subject(s)
Cytochrome b Group/metabolism , Cytochrome d Group/metabolism , Energy Metabolism , Nitric Oxide/metabolism , Oxygen/metabolism , Salmonella enterica/metabolism , Salmonella enterica/pathogenicity , Animals , Electron Transport Chain Complex Proteins , Humans , Hypoxia , Immunity, Innate , Oxidation-Reduction , Oxidoreductases/metabolism , Oxygen Consumption , Salmonella enterica/growth & development , Stress, Physiological
9.
Nanomedicine ; 11(8): 1961-70, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26238076

ABSTRACT

This study represents a novel approach for intraoperative ovarian cancer treatment based on the combinatorial effect of a targeted photodynamic therapy (PDT) associated with suppression of the DJ-1 protein, one of the key players in the ROS defense of cancer cells. To assess the potential of the developed therapy, dendrimer-based nanoplatforms for cancer-targeted delivery of near-infrared photosensitizer, phthalocyanine, and DJ-1 siRNA have been constructed. In vitro studies revealed that therapeutic efficacy of the combinatorial approach was enhanced when compared to PDT alone and this enhancement was more pronounced in ovarian carcinoma cells, which are characterized by higher basal levels of DJ-1 protein. Moreover, the ovarian cancer tumors exposed to a single dose of combinatorial therapy were completely eradicated from the mice and the treated animals showed no evidence of cancer recurrence. Thus, the developed therapeutic approach can be potentially employed intraoperatively to eradicate unresactable cancer cells. FROM THE CLINICAL EDITOR: The complete clearance of microscopic residual tumor cells during excision surgery is important to improve survival of the patient. In this interesting paper, the authors developed a novel approach using targeted photodynamic therapy (PDT), combining a photosensitizer, phthalocyanine, and DJ-1 siRNA for the treatment of ovarian cancer. The data showed that this approach increased cancer cell killing and may pave way for future clinical studies.


Subject(s)
Indoles/therapeutic use , Intracellular Signaling Peptides and Proteins/genetics , Oncogene Proteins/genetics , Ovarian Neoplasms/therapy , Photosensitizing Agents/therapeutic use , RNA, Small Interfering/therapeutic use , Animals , Cell Line, Tumor , Dendrimers/chemistry , Dendrimers/metabolism , Drug Carriers/chemistry , Drug Carriers/metabolism , Drug Delivery Systems , Female , Humans , Indoles/administration & dosage , Isoindoles , Mice , Mice, Nude , Nanostructures/chemistry , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Ovary/metabolism , Ovary/pathology , Photochemotherapy , Photosensitizing Agents/administration & dosage , Protein Deglycase DJ-1 , RNA, Small Interfering/administration & dosage , RNAi Therapeutics , Reactive Oxygen Species/metabolism
10.
PLoS Negl Trop Dis ; 8(8): e3079, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25121731

ABSTRACT

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.


Subject(s)
Anti-Bacterial Agents/pharmacology , Burkholderia pseudomallei/drug effects , Imipenem/pharmacology , Interferon-gamma/pharmacology , Macrophages/immunology , Nitric Oxide/physiology , Salmonella typhimurium/drug effects , Animals , Cells, Cultured , Mice , Oxidative Stress , Proton-Motive Force
11.
Article in English | MEDLINE | ID: mdl-25152099

ABSTRACT

BACKGROUND: Community-based participatory research (CBPR) holds tremendous promise for addressing public health disparities. As such, there is a need for academic institutions to build lasting partnerships with community organizations. Herein we have described the process of establishing a relationship between a research university and a Black church in rural North Carolina. We then discuss Harvest of Hope, the church-based pilot garden project that emerged from that partnership. METHODS: The partnership began with a third-party effort to connect research universities with Black churches to address health disparities. Building this academic-community partnership included collaborating to determine research questions and programming priorities. Other aspects of the partnership included applying for funding together and building consensus on study budget and aims. The academic partners were responsible for administrative details and the community partners led programming and were largely responsible for participant recruitment. RESULTS: The community and academic partners collaborated to design and implement Harvest of Hope, a church-based pilot garden project involving 44 youth and adults. Community and academic partners shared responsibility for study design, recruitment, programming, and reporting of results. The successful operation of the Harvest of Hope project gave rise to a larger National Institutes of Health (NIH)-funded study, Faith, Farming and the Future (F3) involving 4 churches and 60 youth. Both projects were CBPR efforts to improve healthy food access and reducing chronic disease. This partnership continues to expand as we develop additional CBPR projects targeting physical activity, healthy eating, and environmental justice, among others. Benefits of the partnership include increased community ownership and cultural appropriateness of interventions. Challenges include managing expectations of diverse parties and adequate communication. Lessons learned and strategies for building and maintaining similar partnerships are discussed. CONCLUSIONS: The benefits of community-based research for addressing health disparities are many, and there are lessons to be learned that can strengthen community-academic partnerships.


Subject(s)
Black or African American , Community-Institutional Relations , Health Promotion/organization & administration , Health Status Disparities , Rural Population , Adolescent , Adult , Community Participation/methods , Community-Based Participatory Research , Cooperative Behavior , Diet , Exercise , Humans , North Carolina , Religion , Universities/organization & administration
12.
Infect Immun ; 82(1): 333-40, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24166960

ABSTRACT

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.


Subject(s)
Bacterial Proteins/physiology , Repressor Proteins/physiology , Salmonella Infections/microbiology , Salmonella typhimurium/pathogenicity , Analysis of Variance , Animals , Bacterial Proteins/immunology , Disease Models, Animal , Mice , Nitric Oxide Synthase Type II , Oxidative Stress/physiology , Repressor Proteins/deficiency , Repressor Proteins/immunology , Salmonella Infections/physiopathology , Salmonella typhimurium/immunology , Stress, Physiological/physiology
13.
Mol Microbiol ; 87(3): 609-22, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23217033

ABSTRACT

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.


Subject(s)
Glutathione/metabolism , Nitric Oxide/metabolism , Oxidants/metabolism , Salmonella/physiology , Sulfhydryl Compounds/metabolism , Molecular Weight , Nitric Oxide/antagonists & inhibitors , Nitrosation , Oxidants/antagonists & inhibitors , Oxidation-Reduction , Reactive Nitrogen Species/antagonists & inhibitors , Reactive Oxygen Species/antagonists & inhibitors , Salmonella/drug effects , Salmonella/metabolism
14.
Nitric Oxide ; 27(1): 25-31, 2012 Jun 30.
Article in English | MEDLINE | ID: mdl-22521523

ABSTRACT

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.


Subject(s)
Anti-Bacterial Agents/pharmacology , Burkholderia pseudomallei/drug effects , Nitric Oxide/pharmacology , Aconitate Hydratase/drug effects , Aconitate Hydratase/metabolism , Anaerobiosis , Anti-Bacterial Agents/metabolism , Burkholderia pseudomallei/physiology , Culture Media , Humans , Melioidosis/microbiology , Microbial Sensitivity Tests , Nitric Oxide/metabolism , Nitric Oxide Donors/metabolism , Nitric Oxide Donors/pharmacology , Nitroso Compounds/metabolism , Nitroso Compounds/pharmacology , Spermine/analogs & derivatives , Spermine/metabolism , Spermine/pharmacology
15.
PLoS One ; 6(6): e21523, 2011.
Article in English | MEDLINE | ID: mdl-21720554

ABSTRACT

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.


Subject(s)
Alanine Racemase/genetics , Anti-Bacterial Agents/pharmacology , Burkholderia mallei/enzymology , Burkholderia pseudomallei/enzymology , Mutation/genetics , Alanine/pharmacology , Alanine Racemase/chemistry , Amino Acid Sequence , Animals , Burkholderia mallei/drug effects , Burkholderia mallei/genetics , Burkholderia mallei/ultrastructure , Burkholderia pseudomallei/drug effects , Burkholderia pseudomallei/genetics , Burkholderia pseudomallei/ultrastructure , Gene Deletion , Genes, Bacterial/genetics , Genetic Loci/genetics , Genetic Markers , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/microbiology , Macrophages, Peritoneal/ultrastructure , Mice , Microbial Viability/drug effects , Molecular Sequence Data , Periodic Acid/pharmacology , Plasmids/genetics , Polymerase Chain Reaction , Reproducibility of Results , Sequence Alignment
16.
BMC Microbiol ; 11: 58, 2011 Mar 21.
Article in English | MEDLINE | ID: mdl-21418628

ABSTRACT

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.


Subject(s)
Gene Expression Regulation, Bacterial , Regulon , Repressor Proteins/metabolism , Salmonella typhimurium/growth & development , Salmonella typhimurium/genetics , Anaerobiosis , Animals , Female , Gene Deletion , Gene Expression Profiling , Metabolic Networks and Pathways/genetics , Mice , Mice, Inbred C57BL , Microarray Analysis , Repressor Proteins/genetics , Salmonella typhimurium/metabolism
17.
J Bacteriol ; 193(2): 497-505, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21075923

ABSTRACT

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.


Subject(s)
Bacterial Proteins/biosynthesis , Bacterial Proteins/metabolism , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Bacterial , Repressor Proteins/metabolism , Salmonella typhimurium/physiology , Trans-Activators/biosynthesis , Virulence Factors/biosynthesis , Animals , Bacterial Proteins/genetics , Chromatin Immunoprecipitation , DNA, Bacterial/metabolism , DNA-Binding Proteins/biosynthesis , Disease Models, Animal , Gene Knockout Techniques , Genes, Bacterial , Genomic Islands , Mice , Mice, Inbred C3H , Operon , Protein Binding , Repressor Proteins/genetics , Salmonella Infections, Animal/microbiology , Salmonella typhimurium/pathogenicity , Survival Analysis , Transcription Factors/biosynthesis , Virulence
18.
Proc Natl Acad Sci U S A ; 107(32): 14396-401, 2010 Aug 10.
Article in English | MEDLINE | ID: mdl-20660761

ABSTRACT

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.


Subject(s)
Bacterial Proteins/physiology , Nitric Oxide/metabolism , Salmonella typhimurium/pathogenicity , Transcription Factors/physiology , Host-Pathogen Interactions , Immunity , Oxidation-Reduction , Salmonella Infections
19.
PLoS One ; 3(4): e1976, 2008 Apr 09.
Article in English | MEDLINE | ID: mdl-18398486

ABSTRACT

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.


Subject(s)
Burkholderia mallei/metabolism , Iron-Sulfur Proteins/chemistry , Nitric Oxide/metabolism , Aconitate Hydratase/metabolism , Animals , Humans , Hydrogen Peroxide/pharmacology , Macrophages/metabolism , Macrophages/microbiology , Mice , Mice, Inbred C57BL , Models, Biological , Models, Genetic , NADPH Oxidases/metabolism , Oxygen/metabolism , Phagosomes/metabolism , Superoxides/metabolism
20.
Methods Enzymol ; 437: 521-38, 2008.
Article in English | MEDLINE | ID: mdl-18433645

ABSTRACT

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.


Subject(s)
Anti-Infective Agents/pharmacology , Macrophages/drug effects , Nitric Oxide/pharmacology , Animals , Humans , Macrophage Activation/drug effects , Mice , Mice, Knockout , Microbial Sensitivity Tests/methods , Microbial Viability/drug effects , Nitric Oxide Synthase Type II/genetics , Salmonella Infections, Animal/metabolism , Salmonella enterica
SELECTION OF CITATIONS
SEARCH DETAIL
...