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










Publication year range
1.
N Biotechnol ; 64: 17-26, 2021 Sep 25.
Article in English | MEDLINE | ID: mdl-33992842

ABSTRACT

Notch signalling is a well-established oncogenic pathway, and its ligand Delta-like 1 (DLL1) is overexpressed in estrogen receptor-positive (ER+) breast cancers and associated with poor patient prognosis. Hence, DLL1 has become an interesting therapeutic target for breast cancer. Here, the development of specific functional blocking anti-DLL1 antibodies with potential activity against ER+ breast cancer cells is reported. Human DLL1 proteins, containing the essential regions for binding to the Notch receptor and Notch signalling activation, were produced and used to select specific scFv antibody fragments by phage display. Fifteen unique scFvs were identified and reformatted into full IgGs. Characterization of these antibodies by ELISA, surface plasmon resonance and flow cytometry enabled selection of three specific anti-DLL1 IgGs, sharing identical VH regions, with nM affinities. Cellular assays on ER+ breast cancer MCF-7 cells showed that one of the IgGs (IgG-69) was able to partially impair DLL1-mediated activation of the Notch pathway, as determined by Notch reporter and RT-qPCR assays, and to attenuate cell growth. Treatment of MCF-7 cells with IgG-69 reduced mammosphere formation, suggesting that it decreases the breast cancer stem cell subpopulation. These results support the use of this strategy to develop and identify potential anti-DLL1 antibodies candidates against breast cancer.


Subject(s)
Breast Neoplasms , Calcium-Binding Proteins/immunology , Cell Surface Display Techniques , Immunoglobulin G/biosynthesis , Membrane Proteins/immunology , Female , Humans , Ligands , MCF-7 Cells
2.
Biotechnol Bioeng ; 118(6): 2202-2219, 2021 06.
Article in English | MEDLINE | ID: mdl-33624859

ABSTRACT

Serological assays are valuable tools to study SARS-CoV-2 spread and, importantly, to identify individuals that were already infected and would be potentially immune to a virus reinfection. SARS-CoV-2 Spike protein and its receptor binding domain (RBD) are the antigens with higher potential to develop SARS-CoV-2 serological assays. Moreover, structural studies of these antigens are key to understand the molecular basis for Spike interaction with angiotensin converting enzyme 2 receptor, hopefully enabling the development of COVID-19 therapeutics. Thus, it is urgent that significant amounts of this protein became available at the highest quality. In this study, we produced Spike and RBD in two human derived cell hosts: HEK293-E6 and Expi293F™. We evaluated the impact of different and scalable bioprocessing approaches on Spike and RBD production yields and, more importantly, on these antigens' quality attributes. Using negative and positive sera collected from human donors, we show an excellent performance of the produced antigens, assessed in serologic enzyme-linked immunosorbent assay (ELISA) tests, as denoted by the high specificity and sensitivity of the test. We show robust Spike productions with final yields of approx. 2 mg/L of culture that were maintained independently of the production scale or cell culture strategy. To the best of our knowledge, the final yield of 90 mg/L of culture obtained for RBD production, was the highest reported to date. An in-depth characterization of SARS-CoV-2 Spike and RBD proteins was performed, namely the antigen's oligomeric state, glycosylation profiles, and thermal stability during storage. The correlation of these quality attributes with ELISA performance show equivalent reactivity to SARS-CoV-2 positive serum, for all Spike and RBD produced, and for all storage conditions tested. Overall, we provide straightforward protocols to produce high-quality SARS-CoV-2 Spike and RBD antigens, that can be easily adapted to both academic and industrial settings; and integrate, for the first time, studies on the impact of bioprocess with an in-depth characterization of these proteins, correlating antigen's glycosylation and biophysical attributes to performance of COVID-19 serologic tests.


Subject(s)
Antigens, Viral/biosynthesis , Glycosylation , Spike Glycoprotein, Coronavirus/biosynthesis , Cold Temperature , Enzyme-Linked Immunosorbent Assay/standards , Freezing , HEK293 Cells , Humans , Protein Conformation , Protein Stability , Recombinant Proteins/biosynthesis , Recombinant Proteins/standards , SARS-CoV-2 , Serologic Tests/standards , Spike Glycoprotein, Coronavirus/standards
3.
Virulence ; 9(1): 312-317, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29020514

ABSTRACT

RICs are a family of bacterial proteins involved in the repair of iron centers containing proteins damaged by the antimicrobial reactive species liberated by the innate immune system of infected hosts. Staphylococcus aureus is a human pathogen with increasing antibiotic resistance that also contains a RIC-like protein. In this work, we show that the survival of S. aureus within macrophages decreases upon inactivation of ric, and that the viability was restored to levels similar to the wild-type strain by reintroduction of ric via in trans complementation. Importantly, in macrophages that do not produce reactive oxygen species, the lower survival of the ric mutant was no longer observed. In lung epithelial cells, the intracellular viability of the S. aureus ric mutant was also shown to be lower than that of the wild-type. The wax moth larvae Galleria mellonella infected with S. aureus ric mutant presented an approximately 2.5-times higher survival when compared to the wild-type strain. Moreover, significantly lower bacterial loads were determined in the larvae hemolymph infected with strains not expressing ric, and complementation assays confirmed that this behavior was related to RIC. Furthermore, expression of the S. aureus ric gene within the larvae increased along the course of infection with a ~20-fold increase after 8 h of infection. Altogether, the data show that RIC is important for the virulence of S. aureus.


Subject(s)
Bacterial Proteins/metabolism , Staphylococcal Infections/pathology , Staphylococcus aureus/pathogenicity , Virulence Factors/metabolism , Animals , Bacterial Load , Bacterial Proteins/genetics , Cells, Cultured , Disease Models, Animal , Epithelial Cells/immunology , Epithelial Cells/microbiology , Gene Deletion , Genetic Complementation Test , Hemolymph/microbiology , Humans , Lepidoptera , Macrophages/immunology , Macrophages/microbiology , Microbial Viability , Staphylococcal Infections/microbiology , Staphylococcus aureus/genetics , Survival Analysis , Virulence , Virulence Factors/genetics
4.
Protist ; 167(3): 222-33, 2016 06.
Article in English | MEDLINE | ID: mdl-27124376

ABSTRACT

Trichomonas vaginalis, the causative parasite of one of the most prevalent sexually transmitted diseases is, so far, the only protozoan encoding two putative Repair of Iron Centres (RIC) proteins. Homologs of these proteins have been shown to protect bacteria from the chemical stress imposed by mammalian immunity. In this work, the biochemical and functional characterisation of the T. vaginalis RICs revealed that the two proteins have different properties. Expression of ric1 is induced by nitrosative stress but not by hydrogen peroxide, while ric2 transcription remained unaltered under similar conditions. T. vaginalis RIC1 contains a di-iron centre, but RIC2 apparently does not. Only RIC1 resembles bacterial RICs on spectroscopic profiling and repairing ability of oxidatively-damaged iron-sulfur clusters. Unexpectedly, RIC2 was found to bind DNA plasmid and T. vaginalis genomic DNA, a function proposed to be related with its leucine zipper domain. The two proteins also differ in their cellular localization: RIC1 is expressed in the cytoplasm only, and RIC2 occurs both in the nucleus and cytoplasm. Therefore, we concluded that the two RIC paralogs have different roles in T. vaginalis, with RIC2 showing an unprecedented DNA binding ability when compared with all other until now studied RICs.


Subject(s)
Protozoan Proteins/metabolism , Trichomonas vaginalis/metabolism , Amino Acid Motifs , Amino Acid Sequence , Cell Nucleus/chemistry , Cytoplasm/chemistry , DNA/metabolism , Gene Expression Profiling , Iron/metabolism , Microscopy, Fluorescence , Protein Binding , Protozoan Proteins/genetics , Sequence Alignment , Spectrum Analysis , Transcription, Genetic
5.
Dalton Trans ; 45(4): 1455-66, 2016 Jan 28.
Article in English | MEDLINE | ID: mdl-26673556

ABSTRACT

Transition metal carbonyl complexes used as CO-releasing molecules (CORMs) for biological and therapeutic applications may exhibit interesting antimicrobial activity. However, understanding the chemical traits and mechanisms of action that rule this activity is required to establish a rationale for the development of CORMs into useful antibiotics. In this work the bactericidal activity, the toxicity to eukaryotic cells, and the ability of CORMs to deliver CO to bacterial and eukaryotic cells were analysed for a set of seven CORMs that differ in the transition metal, ancillary ligands and the CO release profile. Most of these CORMs exhibited bactericidal properties that decrease in the following order: CORM-2 > CORM-3 > ALF062 > ALF850 > ALF186 > ALF153 > [Fe(SBPy3)(CO)](BF4)2. A similar yet not entirely coincident decreasing order was found for their induction of intracellular reactive oxygen species (ROS) in E. coli. In contrast, studies in model animal cells showed that for any given CORM, the level of intracellular ROS generated was negligible when compared with that measured inside bacteria. Importantly, these CORMs were in general not toxic to eukaryotic cells, namely murine macrophages, kidney LLC-PK1 epithelial cells, and liver cell line HepG2. CORM-2 and CORM-3 delivered CO to the intracellular space of both E. coli and the two types of tested eukaryotic cells, yet toxicity was only elicited in the case of E. coli. CO delivered by ALF186 into the intercellular space did not enter E. coli cells and the compound was not toxic to either bacteria or to eukaryotic cells. The Fe(ii) carbonyl complex [Fe(SBPy3)(CO)](2+) had the reverse, undesirable toxicity profile, being unexpectedly toxic to eukaryotic cells and non-toxic to E. coli. ALF153, the most stable complex in the whole set, was essentially devoid of toxicity or ROS induction ability in all cells. These results suggest that CORMs have a relevant therapeutic potential as antimicrobial drugs since (i) they can show opposite toxicity profiles towards bacteria and eukaryotic cells; (ii) their activity can be modulated through manipulation of the ancillary ligands, as shown with the three {Ru(CO)3}(2+) and two zerovalent Mo based CORMs; and (iii) their toxicity to eukaryotic cells can be made acceptably low. With this new approach, this work contributes to the understanding of the roots of the bactericidal action of CORMs and helps in establishing strategies for their development into a new class of antibiotics.


Subject(s)
Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Carbon Monoxide/chemistry , Cells/drug effects , Escherichia coli/drug effects , Organometallic Compounds/adverse effects , Organometallic Compounds/pharmacology , Animals , Anti-Bacterial Agents/adverse effects , Anti-Bacterial Agents/chemical synthesis , Cell Survival/drug effects , Cells/cytology , Dose-Response Relationship, Drug , Epithelial Cells/drug effects , Escherichia coli/cytology , Escherichia coli/growth & development , Hep G2 Cells , Humans , Macrophages/drug effects , Mice , Microbial Sensitivity Tests , Molecular Conformation , Organometallic Compounds/chemical synthesis , Organometallic Compounds/chemistry , Structure-Activity Relationship
6.
FEBS Lett ; 589(4): 426-31, 2015 Feb 13.
Article in English | MEDLINE | ID: mdl-25583388

ABSTRACT

Repair of Iron Centres (RICs) are a widely-spread family of diiron proteins involved in the protection of iron-sulphur-containing enzymes from nitrosative and oxidative stress. Here, homology-based modelling was used to predict putative ligands of the RIC diiron centre in E. coli. Site-directed mutagenesis studies showed that several conserved residues modulate the spectroscopic properties of the diiron centre, and mutations in H129, E133 and E208 abrogated RIC ability to protect aconitase. Taken together, these data led to a structural model of a diiron centre inserted in a four-helix bundle fold and coordinated by H84, H129, H160, H204, E133 and E208. Moreover, two µ-carboxylate bridges involving E133 and E208 were found to be required for assembly of a stable diiron centre.


Subject(s)
Escherichia coli Proteins/chemistry , Escherichia coli , Iron/chemistry , Amino Acid Sequence , Conserved Sequence , Coordination Complexes , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Structural Homology, Protein
7.
Curr Microbiol ; 69(2): 164-8, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24682218

ABSTRACT

In Staphylococcus aureus, the intracellular siderophore staphyloferrin B, which has been shown to chelate iron-bound to serum transferrin, is transported into cells by the SirABC system. In this work, we have analysed the role of the Sir transporter under stress conditions that resemble those imposed by the mammalian innate immune system. We show that exposure of S. aureus to oxidative and nitrosative stress generated by hydrogen peroxide and S-nitrosoglutathione, respectively, induced the expression of the sirA gene. The disruption of the sir operon led to a strain with lower viability and decreased resistance to oxidative stress. S. aureus sir null mutant was also analysed during infection of murine macrophages and shown to contribute to S. aureus survival inside macrophages. Altogether, our results indicate that the Sir transport system confers protection against reactive oxygen species, therefore, contributing to the virulence of S. aureus.


Subject(s)
Drug Tolerance , Membrane Transport Proteins/metabolism , Oxidants/toxicity , Oxidative Stress , Siderophores/metabolism , Staphylococcus aureus/physiology , Animals , Cell Line , Gene Expression Profiling , Gene Knockout Techniques , Macrophages/immunology , Macrophages/microbiology , Membrane Transport Proteins/genetics , Mice , Microbial Viability/drug effects , Nitroso Compounds/toxicity , Staphylococcus aureus/genetics
8.
PLoS One ; 9(4): e95222, 2014.
Article in English | MEDLINE | ID: mdl-24740378

ABSTRACT

Escherichia coli RIC (Repair of Iron Centers) is a diiron protein previously reported to be involved in the repair of iron-sulfur proteins damaged by oxidative or nitrosative stresses, and proposed to act as an iron donor. This possible role of RIC was now examined specifically by evaluating its ability to donate iron ions to apo-iron-sulfur proteins, determining the iron binding constants and assessing the lability of its iron ions. We show, by UV-visible, EPR and resonance Raman spectroscopies that RIC may participate in the synthesis of an iron-sulfur cluster in the apo-forms of the spinach ferredoxin and IscU when in the presence of the sulfide donating system IscS and L-cysteine. Iron binding assays allowed determining the as-isolated and fully reduced RIC dissociation constants for the ferric and ferrous iron of 10-27 M and 10-13 M, respectively. Mössbauer studies revealed that the RIC iron ions are labile, namely when the center is in the mixed-valence redox form as compared with the (µ-oxo) diferric one. Altogether, these results suggest that RIC is capable of delivering iron for the formation of iron-sulfur clusters.


Subject(s)
Escherichia coli Proteins/metabolism , Escherichia coli/metabolism , Gene Expression Regulation, Bacterial , Iron-Sulfur Proteins/metabolism , Iron/metabolism , Sulfur/metabolism , Carbon-Sulfur Lyases/genetics , Carbon-Sulfur Lyases/metabolism , Cysteine/metabolism , Escherichia coli/genetics , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Ferredoxins/genetics , Ferredoxins/metabolism , Iron-Sulfur Proteins/chemistry , Iron-Sulfur Proteins/genetics , Kinetics , Oxidation-Reduction , Plant Proteins/genetics , Plant Proteins/metabolism , Protein Binding , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Spinacia oleracea/chemistry
9.
J Bacteriol ; 195(11): 2684-90, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23564166

ABSTRACT

Desulfovibrio species are Gram-negative anaerobic sulfate-reducing bacteria that colonize the human gut. Recently, Desulfovibrio spp. have been implicated in gastrointestinal diseases and shown to stimulate the epithelial immune response, leading to increased production of inflammatory cytokines by macrophages. Activated macrophages are key cells of the immune system that impose nitrosative stress during phagocytosis. Hence, we have analyzed the in vitro and in vivo responses of Desulfovibrio vulgaris Hildenborough to nitric oxide (NO) and the role of the hybrid cluster proteins (HCP1 and HCP2) and rubredoxin oxygen oxidoreductases (ROO1 and ROO2) in NO protection. Among the four genes, hcp2 was the gene most highly induced by NO, and the hcp2 transposon mutant exhibited the lowest viability under conditions of NO stress. Studies in murine macrophages revealed that D. vulgaris survives incubation with these phagocytes and triggers NO production at levels similar to those stimulated by the cytokine gamma interferon (IFN-γ). Furthermore, D. vulgaris hcp and roo mutants exhibited reduced viability when incubated with macrophages, revealing that these gene products contribute to the survival of D. vulgaris during macrophage infection.


Subject(s)
Bacterial Proteins/metabolism , Desulfovibrio vulgaris/physiology , Desulfovibrionaceae Infections/microbiology , Iron-Sulfur Proteins/metabolism , Macrophages/microbiology , NADH, NADPH Oxidoreductases/genetics , Nitric Oxide/metabolism , Animals , Bacterial Proteins/genetics , Cell Line , Desulfovibrio vulgaris/drug effects , Desulfovibrio vulgaris/genetics , Desulfovibrio vulgaris/growth & development , Desulfovibrionaceae Infections/immunology , Gene Expression Regulation, Bacterial , Humans , Iron-Sulfur Proteins/genetics , Macrophages/immunology , Macrophages/metabolism , Mice , Microbial Sensitivity Tests , Microbial Viability , Mutagenesis, Insertional , NADH, NADPH Oxidoreductases/metabolism , Nitric Oxide/pharmacology , Nitrites/analysis , Nitrites/metabolism , Oxidative Stress , Phenotype , Stress, Physiological
10.
Appl Microbiol Biotechnol ; 97(6): 2563-73, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23389340

ABSTRACT

Staphylococcus aureus is a pathogen responsible for severe community- and nosocomially acquired infections. To fight pathogen intrusion, the innate immune system uses a plethora of weapons, with the generation of oxidative and nitrosative stresses among the most efficient. In this work, the S. aureus genome-wide transcriptional responses to oxidative stress generated by hydrogen peroxide, to nitrosative stress imposed by S-nitrosoglutathione (GSNO), and to the combination of the two were investigated using microarray analysis. The results showed that these stresses have a significant impact on the transcriptome of S. aureus. Hydrogen peroxide modified mainly the mRNA abundance of genes involved in oxidative detoxification and DNA metabolism, which together represent 14 % of the total number of upregulated genes. GSNO caused significant alteration of the expression of gene products with regulatory function. However, the simultaneous addition of GSNO and hydrogen peroxide was found to cause the more significant transcriptomic alteration, affecting ∼10 % of the total transcriptome. In particular, exposure of S. aureus to GSNO plus hydrogen peroxide modified the transcription of genes associated with cell envelope and iron metabolism, including induction of ftnA and dps genes that encode iron-storage and oxidative-protecting proteins. Further studies revealed that when exposed to combined GSNO-hydrogen peroxide stresses, S. aureus has decreased viability, which is enhanced in the presence of iron, and low siderophore activity. Altogether, this study revealed, for the first time, how the combined oxidative and nitrosative stresses inflicted during phagocytosis interfere at the transcriptional level with the S. aureus cellular metabolism.


Subject(s)
Hydrogen Peroxide/toxicity , S-Nitrosoglutathione/toxicity , Staphylococcus aureus/drug effects , Transcriptome , Metabolic Networks and Pathways/genetics , Microarray Analysis , Microbial Viability/drug effects , Nitrosation , Oxidative Stress , Staphylococcus aureus/genetics , Staphylococcus aureus/physiology
11.
PLoS One ; 8(12): e83157, 2013.
Article in English | MEDLINE | ID: mdl-24386154

ABSTRACT

Helicobacter pylori is a pathogen that establishes long life infections responsible for chronic gastric ulcer diseases and a proved risk factor for gastric carcinoma. The therapeutic properties of carbon-monoxide releasing molecules (CORMs) led us to investigate their effect on H. pylori. We show that H. pylori 26695 is susceptible to two widely used CORMs, namely CORM-2 and CORM-3. Also, several H. pylori clinical isolates were killed by CORM-2, including those resistant to metronidazole. Moreover, sub-lethal doses of CORM-2 combined with metronidazole, amoxicillin and clarithromycin was found to potentiate the effect of the antibiotics. We further demonstrate that the mechanisms underpinning the antimicrobial effect of CORMs involve the inhibition of H. pylori respiration and urease activity. In vivo studies done in key cells of the innate immune system, such as macrophages, showed that CORM-2, either alone or when combined with metronidazole, strongly reduces the ability of H. pylori to infect animal cells. Hence, CORMs have the potential to kill antibiotic resistant strains of H. pylori.


Subject(s)
Anti-Bacterial Agents/pharmacology , Carbon Monoxide/pharmacology , Helicobacter pylori/drug effects , Drug Resistance, Bacterial , Drug Therapy, Combination , Helicobacter pylori/metabolism , Metronidazole/pharmacology , Microbial Sensitivity Tests , Oxygen Consumption , Urease/metabolism
12.
FEMS Microbiol Lett ; 336(1): 1-10, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22774863

ABSTRACT

Carbon monoxide-releasing molecules (CO-RMs) are, in general, transition metal carbonyl complexes that liberate controlled amounts of CO. In animal models, CO-RMs have been shown to reduce myocardial ischaemia, inflammation and vascular dysfunction, and to provide a protective effect in organ transplantation. Moreover, CO-RMs are bactericides that kill both Gram-positive and Gram-negative bacteria such as Staphylococcus aureus and Pseudomonas aeruginosa. Herein are reviewed the microbial genetic and biochemical responses associated with CO-RM-mediated cell death. Particular emphasis is given to the data revealing that CO-RMs induce the generation of reactive oxygen species (ROS), which contribute to the antibacterial activity of these compounds.


Subject(s)
Anti-Bacterial Agents/chemistry , Bacteria/metabolism , Carbon Monoxide/pharmacology , Reactive Oxygen Species/metabolism , Bacteria/drug effects , Gene Expression Regulation, Bacterial/drug effects , Oxidative Stress/drug effects
13.
Anaerobe ; 18(4): 454-8, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22706208

ABSTRACT

Intracellular free iron, is under aerobic conditions and via the Fenton reaction a catalyst for the formation of harmful reactive oxygen species. In this article, we analyzed the relation between intracellular iron storage and oxidative stress response in the sulfate reducing bacterium Desulfovibrio vulgaris Hildenborough, an anaerobe that is often found in oxygenated niches. To this end, we investigated the role of the iron storage protein bacterioferritin using transcriptomic and physiological approaches. We observed that transcription of bacterioferritin is strongly induced upon exposure of cells to an oxygenated atmosphere. When grown in the presence of high concentrations of oxygen the D. vulgaris bacterioferritin mutant exhibited, in comparison with the wild type strain, lower viability and a higher content of intracellular reactive oxygen species. Furthermore, the bacterioferritin gene is under the control of the oxidative stress response regulator D. vulgaris PerR. Altogether the data revealed a previously unrecognized ability for the iron storage bacterioferritin to contribute to the oxygen tolerance exhibited by D. vulgaris.


Subject(s)
Bacterial Proteins/metabolism , Cytochrome b Group/metabolism , Desulfovibrio vulgaris/metabolism , Ferritins/metabolism , Oxygen/metabolism , Adaptation, Physiological , Bacterial Proteins/genetics , Cytochrome b Group/genetics , Desulfovibrio vulgaris/genetics , Ferritins/genetics , Gene Expression Regulation, Bacterial , Genes, Bacterial , Microbial Viability , Oxidative Stress , Reactive Oxygen Species/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Time Factors , Transcription, Genetic
14.
J Biol Chem ; 286(30): 26708-17, 2011 Jul 29.
Article in English | MEDLINE | ID: mdl-21646348

ABSTRACT

CO-releasing molecules (CO-RMs) were previously shown by us to be more potent bactericides than CO gas. This suggests a mechanism of action for CO-RM, which either potentiates the activity of CO or uses another CO-RM-specific effect. We have also reported that CORM-2 induces the expression of genes related to oxidative stress. In the present study we intend to establish whether the generation of reactive oxygen species by CO-RMs may indeed result in the inhibition of bacterial cellular function. We now report that two CO-RMs (CORM-2 and ALF062) stimulate the production of ROS in Escherichia coli, an effect that is abolished by addition of antioxidants. Furthermore, deletion of genes encoding E. coli systems involved in reactive oxygen species scavenging, namely catalases and superoxide dismutases, potentiates the lethality of CORM-2 due to an increase of intracellular ROS content. CORM-2 also induces the expression of the E. coli DNA repair/SOS system recA, and its inactivation enhances toxicity of CORM-2. Moreover, fluorescence microscopy images reveal that CORM-2 causes DNA lesions to bacterial cells. We also demonstrate that cells treated with CORM-2 contain higher levels of free iron arising from destruction of iron-sulfur proteins. Importantly, we show that CO-RMs generate hydroxyl radicals in a cell-free solution, a process that is abolished by scavenging CO. Altogether, we provide a novel insight into the molecular basis of CO-RMs action by showing that their bactericidal properties are linked to cell damage inflicted by the oxidative stress that they are able to generate.


Subject(s)
Anti-Bacterial Agents/pharmacokinetics , Carbon Monoxide/metabolism , Escherichia coli K12/metabolism , Organometallic Compounds/pharmacology , Reactive Oxygen Species/metabolism , Catalase/genetics , Catalase/metabolism , Escherichia coli K12/genetics , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Oxidative Stress/drug effects , SOS Response, Genetics/drug effects , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism
15.
J Bacteriol ; 192(6): 1527-33, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20097858

ABSTRACT

In this work, we report that flavohemoglobin contributes to the azole susceptibility of Staphylococcus aureus. We first observed that deletion of the flavohemoglobin gene leads to an increase in the viability of imidazole-treated S. aureus cells and that reversion to the wild-type phenotype occurs upon expression of flavohemoglobin from a multicopy plasmid. Further spectroscopic analyses showed that miconazole, the most efficient azole antibiotic against S. aureus, ligates to heme of both oxidized and reduced flavohemoglobin. The binding of miconazole to oxidized flavohemoglobin, with an association constant of 1.7 x 10(6) M(-1), typical of a tight, specific binding equilibrium, results in augmentation of the superoxide production by the enzyme. These results are corroborated by in vivo studies showing that imidazole-treated S. aureus cells expressing flavohemoglobin contain a larger amount of reactive oxygen species. Moreover, it was observed that the survival of miconazole-treated S. aureus internalized by murine macrophages is higher for cells lacking flavohemoglobin. Altogether, the present data revealed that in S. aureus, flavohemoglobin enhances the antimicrobial activity of imidazoles via an increase of intracellular oxidative stress.


Subject(s)
Anti-Bacterial Agents/pharmacology , Azoles/pharmacology , Bacterial Proteins/metabolism , Hemeproteins/metabolism , Oxidative Stress/drug effects , Staphylococcus aureus/drug effects , Staphylococcus aureus/metabolism , Animals , Macrophages/microbiology , Mice , Reactive Oxygen Species
16.
J Bacteriol ; 191(10): 3403-6, 2009 May.
Article in English | MEDLINE | ID: mdl-19286809

ABSTRACT

In this report we show that inactivation of the putative nitroreductase SA0UHSC_00833 (ntrA) increases the sensitivity of Staphylococcus aureus to S-nitrosoglutathione (GSNO) and augments its resistance to nitrofurans. S. aureus NtrA is a bifunctional enzyme that exhibits nitroreductase and GSNO reductase activity. A phylogenetic analysis suggests that NtrA is a member of a novel family of nitroreductases that seems to play a dual role in vivo, promoting nitrofuran activation and protecting the cell against transnitrosylation.


Subject(s)
Aldehyde Oxidoreductases/metabolism , Nitroreductases/metabolism , Staphylococcus aureus/enzymology , Aldehyde Oxidoreductases/classification , Aldehyde Oxidoreductases/genetics , Mutation , Nitrofurans/pharmacology , Nitroreductases/classification , Nitroreductases/genetics , Oligonucleotide Array Sequence Analysis , Phylogeny , Staphylococcus aureus/drug effects , Staphylococcus aureus/genetics
17.
Microbiology (Reading) ; 155(Pt 3): 813-824, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19246752

ABSTRACT

We recently reported that carbon monoxide (CO) has bactericidal activity. To understand its mode of action we analysed the gene expression changes occurring when Escherichia coli, grown aerobically and anaerobically, is treated with the CO-releasing molecule CORM-2 (tricarbonyldichlororuthenium(II) dimer). Microarray analysis shows that the E. coli CORM-2 response is multifaceted, with a high number of differentially regulated genes spread through several functional categories, namely genes involved in inorganic ion transport and metabolism, regulators, and genes implicated in post-translational modification, such as chaperones. CORM-2 has a higher impact in E. coli cells grown anaerobically, as judged by the repression of genes belonging to eight functional classes which are not seen in the response of aerobically CORM-2-treated cells. The biological relevance of the variations caused by CORM-2 was substantiated by studying the CORM-2 sensitivity of selected E. coli mutants. The results show that the deletion of redox-sensing regulators SoxS and OxyR increased the sensitivity to CORM-2 and suggest that while SoxS plays an important role in protection against CORM-2 under both growth conditions, OxyR seems to participate only in the aerobic CORM-2 response. Under anaerobic conditions, we found that the heat-shock proteins IbpA and IbpB contribute to CORM-2 defence since the deletion of these genes increases the sensitivity of the strain. The induction of several met genes and the hypersensitivity to CORM-2 of the DeltametR, DeltametI and DeltametN mutant strains suggest that CO has effects on the methionine metabolism of E. coli. CORM-2 also affects the transcription of several E. coli biofilm-related genes and increases biofilm formation in E. coli. In particular, the absence of tqsA or bhsA increases the resistance of E. coli to CORM-2, and deletion of tsqA leads to a strain that has lost its capacity to form biofilm upon treatment with CORM-2. In spite of the relatively stable nature of the CO molecule, our results show that CO is able to trigger a significant alteration in the transcriptome of E. coli which necessarily has effects in several key metabolic pathways.


Subject(s)
Carbon Monoxide/metabolism , Escherichia coli/genetics , Gene Expression Profiling , Organometallic Compounds/pharmacology , Biofilms , Escherichia coli/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Gene Expression Regulation, Bacterial , Genes, Bacterial , Genes, Regulator , Genetic Complementation Test , Methionine/metabolism , Microbial Viability , Mutation , Oligonucleotide Array Sequence Analysis , Phenotype , RNA, Bacterial/genetics
18.
Methods Enzymol ; 436: 203-16, 2008.
Article in English | MEDLINE | ID: mdl-18237634

ABSTRACT

Biotically, bacteria encounter nitrogen-reactive species in environments where denitrification occurs or when nitric oxide (NO) is generated by the mammal NO synthase, particularly during the infectious processes. In bacteria, flavohemoglobins have been shown to be one of the major systems responsible for the scavenging of these chemical species, either in aerobic or in anaerobic environments. Staphylococcus aureus, a pathogenic bacterium with high impact on human heath, also contains a gene encoding a homologue of a flavohemoprotein. The study of the recombinant protein and of the knockout mutant strain allowed us to conclude that, in spite of sharing similar physicochemical properties with the other known flavohemoglobins, the S. aureus flavohemoglobin requires microaerobic conditions to protect this bacterium from the nocive effects of the nitrosative stress.


Subject(s)
Bacterial Proteins/chemistry , Hemeproteins/chemistry , Staphylococcus aureus/chemistry , Amino Acid Sequence , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Base Sequence , Chemical Phenomena , Chemistry, Physical , DNA, Bacterial/genetics , Gene Deletion , Gene Expression , Genes, Bacterial , Genetic Complementation Test , Hemeproteins/genetics , Hemeproteins/metabolism , Molecular Sequence Data , Nitric Oxide/metabolism , Nitric Oxide/pharmacology , Nitrosation , Oxidation-Reduction , RNA, Bacterial/genetics , RNA, Bacterial/isolation & purification , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , S-Nitrosoglutathione/pharmacology , Sequence Homology, Amino Acid , Staphylococcus aureus/drug effects , Staphylococcus aureus/genetics , Staphylococcus aureus/metabolism
19.
Antimicrob Agents Chemother ; 51(12): 4303-7, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17923486

ABSTRACT

Carbon monoxide (CO) is endogenously produced in the human body, mainly from the oxidation of heme catalyzed by heme oxygenase (HO) enzymes. The induction of HO and the consequent increase in CO production play important physiological roles in vasorelaxation and neurotransmission and in the immune system. The exogenous administration of CO gas and CO-releasing molecules (CO-RMs) has been shown to induce vascular effects and to alleviate hypoxia-reoxygenation injury of mammalian cells. In particular, due to its anti-inflammatory, antiapoptotic, and antiproliferative properties, CO inhibits ischemic-reperfusion injury and provides potent cytoprotective effects during organ and cell transplantation. In spite of these findings regarding the physiology and biology of mammals, nothing is known about the action of CO on bacteria. In the present work, we examined the effect of CO on bacterial cell proliferation. Cell growth experiments showed that CO caused the rapid death of the two pathogenic bacteria tested, Escherichia coli and Staphylococcus aureus, particularly when delivered through organometallic CO-RMs. Of importance is the observation that the effectiveness of the CO-RMs was greater in near-anaerobic environments, as many pathogens are anaerobic organisms and pathogen colonization occurs in environments with low oxygen concentrations. Our results constitute the first evidence that CO can be utilized as an antimicrobial agent. We anticipate our results to be the starting point for the development of novel types of therapeutic drugs designed to combat antibiotic-resistant pathogens, which are widespread and presently a major public health concern.


Subject(s)
Carbon Monoxide/chemistry , Carbon Monoxide/pharmacology , Organometallic Compounds/chemistry , Organometallic Compounds/pharmacology , Aerobiosis , Anaerobiosis , Anti-Infective Agents/chemistry , Anti-Infective Agents/pharmacology , Escherichia coli/drug effects , Microbial Sensitivity Tests , Microbial Viability/drug effects , Molecular Structure , Staphylococcus aureus/drug effects
20.
FEBS Lett ; 580(7): 1817-21, 2006 Mar 20.
Article in English | MEDLINE | ID: mdl-16516202

ABSTRACT

Flavohemoglobins and flavodiiron proteins are two families of enzymes involved in nitrosative detoxification. However, the physiological oxygen-related conditions under which they work and their relative role are still a matter of debate. To address this question we analyzed the function of the putative flavohemoprotein of Staphylococcus aureus, an organism that lacks a flavodiiron-like gene. In this report we show that the recombinant protein contains all features typical of canonical flavohemoglobins and that the transcription of flavohemoglobin gene was upregulated by nitrosative stress in an oxygen-dependent manner. However, and in contrast to other bacterial flavohemoglobins, the S. aureus protein has no apparent role in aerobic nitrosative protection, being only beneficial when cells of S. aureus are submitted to nitrosative stress in a microaerophilic environment. The in vivo data corroborates the proposal that Hmp acts physiologically as a denitrosylase.


Subject(s)
Aerobiosis , Bacterial Proteins/physiology , Hemeproteins/physiology , Nitrogen Oxides/pharmacology , Staphylococcus aureus/metabolism , Oxygen/pharmacology , Recombinant Proteins , Transcription, Genetic , Up-Regulation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...