Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
Add more filters










Publication year range
1.
J Proteome Res ; 23(5): 1666-1678, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38644792

ABSTRACT

Bordetella pertussis persists inside host cells, and virulence factors are crucial for intracellular adaptation. The regulation of B. pertussis virulence factor transcription primarily occurs through the modulation of the two-component system (TCS) known as BvgAS. However, additional regulatory systems have emerged as potential contributors to virulence regulation. Here, we investigate the impact of BP1092, a putative TCS histidine kinase that shows increased levels after bacterial internalization by macrophages, on B. pertussis proteome adaptation under nonmodulating (Bvg+) and modulating (Bvg-) conditions. Using mass spectrometry, we compare B. pertussis wild-type (wt), a BP1092-deficient mutant (ΔBP1092), and a ΔBP1092 trans-complemented strain under both conditions. We find an altered abundance of 10 proteins, including five virulence factors. Specifically, under nonmodulating conditions, the mutant strain showed decreased levels of FhaB, FhaS, and Cya compared to the wt. Conversely, under modulating conditions, the mutant strain exhibited reduced levels of BvgA and BvgS compared to those of the wt. Functional assays further revealed that the deletion of BP1092 gene impaired B. pertussis ability to survive within human macrophage THP-1 cells. Taken together, our findings allow us to propose BP1092 as a novel player involved in the intricate regulation of B. pertussis virulence factors and thus in adaptation to the intracellular environment. The data have been deposited to the ProteomeXchange Consortium via the PRIDE partner repository with the data set identifier PXD041940.


Subject(s)
Bacterial Proteins , Bordetella pertussis , Histidine Kinase , Bordetella pertussis/pathogenicity , Bordetella pertussis/genetics , Histidine Kinase/metabolism , Histidine Kinase/genetics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Virulence/genetics , Gene Expression Regulation, Bacterial , Macrophages/microbiology , Humans , Proteome , Virulence Factors, Bordetella/genetics , Virulence Factors, Bordetella/metabolism , Virulence Factors/genetics , Virulence Factors/metabolism , Microbial Viability
2.
Nat Commun ; 14(1): 5818, 2023 10 02.
Article in English | MEDLINE | ID: mdl-37783679

ABSTRACT

Lower respiratory tract infections caused by Streptococcus pneumoniae (Spn) are a leading cause of death globally. Here we investigate the bronchial epithelial cellular response to Spn infection on a transcriptomic, proteomic and metabolic level. We found the NAD+ salvage pathway to be dysregulated upon infection in a cell line model, primary human lung tissue and in vivo in rodents, leading to a reduced production of NAD+. Knockdown of NAD+ salvage enzymes (NAMPT, NMNAT1) increased bacterial replication. NAD+ treatment of Spn inhibited its growth while growth of other respiratory pathogens improved. Boosting NAD+ production increased NAD+ levels in immortalized and primary cells and decreased bacterial replication upon infection. NAD+ treatment of Spn dysregulated the bacterial metabolism and reduced intrabacterial ATP. Enhancing the bacterial ATP metabolism abolished the antibacterial effect of NAD+. Thus, we identified the NAD+ salvage pathway as an antibacterial pathway in Spn infections, predicting an antibacterial mechanism of NAD+.


Subject(s)
Bacterial Infections , Nicotinamide-Nucleotide Adenylyltransferase , Respiratory Tract Infections , Humans , NAD/metabolism , Proteomics , Cytokines/metabolism , Cell Line , Adenosine Triphosphate , Nicotinamide-Nucleotide Adenylyltransferase/metabolism
3.
Proteomics ; : e2300294, 2023 Sep 29.
Article in English | MEDLINE | ID: mdl-37772677

ABSTRACT

In proteomics, fast, efficient, and highly reproducible sample preparation is of utmost importance, particularly in view of fast scanning mass spectrometers enabling analyses of large sample series. To address this need, we have developed the web application MassSpecPreppy that operates on the open science OT-2 liquid handling robot from Opentrons. This platform can prepare up to 96 samples at once, performing tasks like BCA protein concentration determination, sample digestion with normalization, reduction/alkylation and peptide elution into vials or loading specified peptide amounts onto Evotips in an automated and flexible manner. The performance of the developed workflows using MassSpecPreppy was compared with standard manual sample preparation workflows. The BCA assay experiments revealed an average recovery of 101.3% (SD: ± 7.82%) for the MassSpecPreppy workflow, while the manual workflow had a recovery of 96.3% (SD: ± 9.73%). The species mix used in the evaluation experiments showed that 94.5% of protein groups for OT-2 digestion and 95% for manual digestion passed the significance thresholds with comparable peptide level coefficient of variations. These results demonstrate that MassSpecPreppy is a versatile and scalable platform for automated sample preparation, producing injection-ready samples for proteomics research.

4.
Virulence ; 13(1): 2042-2058, 2022 12.
Article in English | MEDLINE | ID: mdl-36411449

ABSTRACT

Legionella pneumophila (L.p.) is a bacterial pathogen which is a common causative agent of pneumonia. In humans, it infects alveolar macrophages and transfers hundreds of virulence factors that interfere with cellular signalling pathways and the transcriptomic landscape to sustain its own replication. By this interaction, it has acquired eukaryote-like protein motifs by gene transfer events that partake in the pathogenicity of Legionella. In a computational screening approach for eukaryotic motifs in the transcriptome of Legionella, we identified the L.p. strain Corby protein ABQ55614 as putative histone-deacetylase and named it "suppressing modifier of histones 1" (Smh1). During infection, Smh1 is translocated from the Legionella vacuole into the host cytosol. When expressed in human macrophage THP-1 cells, Smh1 was localized predominantly in the nucleus, leading to broad histone H3 and H4 deacetylation, blunted expression of a large number of genes (e.g. IL-1ß and IL-8), and fostered intracellular bacterial replication. L.p. with a Smh1 knockdown grew normally in media but showed a slight growth defect inside the host cell. Furthermore, Smh1 showed a very potent histone deacetylation activity in vitro, e.g. at H3K14, that could be inhibited by targeted mutation of the putative catalytic center inferred by analogy with eukaryotic HDAC8, and with the deacetylase inhibitor trichostatin A. In summary, Smh1 displays functional homology with class I/II type HDACs. We identified Smh1 as a new Legionella virulence factor with a eukaryote-like histone-deacetylase activity that moderates host gene expression and might pave the way for further histone modifications.IMPORTANCELegionella pneumophila (L.p.) is a prominent bacterial pathogen, which is a common causative agent of pneumonia. In order to survive inside the host cell, the human macrophage, it profoundly interacts with host cell processes to advance its own replication. In this study, we identify a bacterial factor, Smh1, with yet unknown function as a host histone deacetylase. The activity of this factor in the host cell leads to attenuated gene expression and increased intracellular bacterial replication.


Subject(s)
Eukaryota , Legionella pneumophila , Humans , Histones/genetics , Legionella pneumophila/genetics , Eukaryotic Cells , Research , Virulence Factors/genetics , Histone Deacetylases , Repressor Proteins
5.
Pathog Dis ; 80(1)2022 08 24.
Article in English | MEDLINE | ID: mdl-35927587

ABSTRACT

Gram-negative pathogenic bacteria constitutively shed outer membrane vesicles (OMVs) which play a significant role in the host-pathogen interaction, eventually determining the outcome of the infection. We previously found that Bordetella pertussis, the etiological agent of whooping cough, survives the innate interaction with human macrophages remaining alive inside these immune cells. Adenylate cyclase (CyaA), one of the main toxins of this pathogen, was found involved in the modulation of the macrophage defense response, eventually promoting bacterial survival within the cells. We here investigated whether B. pertussis OMVs, loaded with most of the bacterial toxins and CyaA among them, modulate the macrophage response to the bacterial infection. We observed that the pre-incubation of macrophages with OMVs led to a decreased macrophage defense response to the encounter with the bacteria, in a CyaA dependent way. Our results suggest that CyaA delivered by B. pertussis OMVs dampens macrophages protective function by decreasing phagocytosis and the bactericidal capability of these host cells. By increasing the chances of bacterial survival to the innate encounter with the macrophages, B. pertussis OMVs might play a relevant role in the course of infection, promoting bacterial persistence within the host and eventually, shaping the whole infection process.


Subject(s)
Bordetella pertussis , Whooping Cough , Adenylate Cyclase Toxin , Humans , Macrophages , Virulence Factors
6.
mSphere ; 7(4): e0023522, 2022 08 31.
Article in English | MEDLINE | ID: mdl-35862816

ABSTRACT

Signaling of two-component systems by phosphoryl transfer requires interaction of the sensor kinase with the response regulator. Interaction of the C4-dicarboxylate-responsive and membrane-integral sensor kinase DcuS with the response regulator DcuR was studied. In vitro, the cytoplasmic part of DcuS (PASC-Kin) was employed. Stable complexes were formed, when either DcuS or DcuR were phosphorylated (Kd 22 ± 11 and 28 ± 7 nM, respectively). The unphosphorylated proteins produced a more labile complex (Kd 1380 ± 395 nM). Bacterial two-hybrid studies confirm interaction of DcuR with DcuS (and PASC-Kin) in vivo. The absolute contents of DcuR (197-979 pmol mg-1 protein) in the bacteria exceeded those of DcuS by more than 1 order of magnitude. According to the Kd values, DcuS exists in complex, with phosphorylated but also unphosphorylated DcuR. In live cell imaging, the predominantly freely diffusing DcuR becomes markedly less mobile after phosphorylation and activation of DcuS by fumarate. Portions of the low mobility fraction accumulated at the cell poles, the preferred location of DcuS, and other portions within the cell, representing phosphorylated DcuR bound to promoters. In the model, acitvation of DcuS increases the affinity toward DcuR, leading to DcuS-P × DcuR formation and phosphorylation of DcuR. The complex is stable enough for phosphate-transfer, but labile enough to allow exchange between DcuR from the cytosol and DcuR-P of the complex. Released DcuR-P diffuses to target promoters and binds. Uncomplexed DcuR-P in the cytosol binds to nonactivated DcuS and becomes dephosphorylated. The lower affinity between DcuR and DcuS avoids blocking of DcuS and allows rapid exchange of DcuR. IMPORTANCE Complex formation of membrane-bound sensor kinases with the response regulators represents an inherent step of signaling from the membrane to the promoters on the DNA. In the C4-dicarboxylate-sensing DcuS-DcuR two-component system, complex formation is strengthened by activation (phosphorylation) in vitro and in vivo, with trapping of the response regulator DcuR at the membrane. Single-molecule tracking of DcuR in the bacterial cell demonstrates two populations of DcuR with decreased mobility in the bacteria after activation: one at the membrane, but a second in the cytosol, likely representing DNA-bound DcuR. The data suggest a model with binding of DcuR to DcuS-P for phosphorylation, and of DcuR-P to DcuS for dephosphorylation, allowing rapid adaptation of the DcuR phosphorylation state. DcuR-P is released and transferred to DNA by 3D diffusion.


Subject(s)
DNA-Binding Proteins , Escherichia coli Proteins , Protein Kinases , Transcription Factors , DNA, Bacterial , DNA-Binding Proteins/genetics , Dicarboxylic Acid Transporters/genetics , Dicarboxylic Acid Transporters/metabolism , Escherichia coli/genetics , Escherichia coli Proteins/genetics , Fumarates/metabolism , Gene Expression Regulation, Bacterial , Promoter Regions, Genetic , Protein Kinases/metabolism , Transcription Factors/genetics
7.
J Bacteriol ; 204(1): e0018421, 2022 01 18.
Article in English | MEDLINE | ID: mdl-34633872

ABSTRACT

Staphylococcus aureus is an opportunistic pathogen that can cause life-threatening infections, particularly in immunocompromised individuals. The high-level virulence of S. aureus largely relies on its diverse and variable collection of virulence factors and immune evasion proteins, including the six serine protease-like proteins SplA to SplF. Spl proteins are expressed by most clinical isolates of S. aureus, but little is known about the molecular mechanisms by which these proteins modify the host's immune response for the benefit of the bacteria. Here, we identify SplB as a protease that inactivates central human complement proteins, i.e., C3, C4, and the activation fragments C3b and C4b, by preferentially cleaving their α-chains. SplB maintained its proteolytic activity in human serum, degrading C3 and C4. SplB further cleaved the components of the terminal complement pathway, C5, C6, C7, C8, and C9. In contrast, the important soluble human complement regulators factor H and C4b-binding protein (C4BP), as well as C1q, were left intact. Thereby, SplB reduced C3b-mediated opsonophagocytosis by human neutrophils as well as C5b-9 deposition on the bacterial surface. In conclusion, we identified the first physiological substrates of the S. aureus extracellular protease SplB. This enzyme inhibits all three complement pathways and blocks opsonophagocytosis. Thus, SplB can be considered a novel staphylococcal complement evasion protein. IMPORTANCE The success of bacterial pathogens in immunocompetent humans depends on the control and inactivation of host immunity. S. aureus, like many other pathogens, efficiently blocks host complement attack early in infection. Aiming to understand the role of the S. aureus-encoded orphan proteases of the Spl operon, we asked whether these proteins play a role in immune escape. We found that SplB inhibits all three complement activation pathways as well as the lytic terminal complement pathway. This blocks the opsonophagocytosis of the bacteria by neutrophils. We also clarified the molecular mechanisms: SplB cleaves the human complement proteins C3, C4, C5, C6, C7, C8, and C9 as well as factor B but not the complement inhibitors factor H and C4BP. Thus, we identify the first physiological substrates of the extracellular protease SplB of S. aureus and characterize SplB as a novel staphylococcal complement evasion protein.


Subject(s)
Bacterial Proteins/metabolism , Complement System Proteins/metabolism , Opsonization/physiology , Peptide Hydrolases/metabolism , Staphylococcus aureus/enzymology , Bacterial Proteins/genetics , Gene Expression Regulation, Bacterial , Gene Expression Regulation, Enzymologic , Humans , Peptide Hydrolases/genetics , Staphylococcus aureus/metabolism
8.
Int J Mol Sci ; 22(17)2021 Aug 26.
Article in English | MEDLINE | ID: mdl-34502122

ABSTRACT

Extracellular vesicles (EVs) are reminiscent of their cell of origin and thus represent a valuable source of biomarkers. However, for EVs to be used as biomarkers in clinical practice, simple, comparable, and reproducible analytical methods must be applied. Although progress is being made in EV separation methods for human biofluids, the implementation of EV assays for clinical diagnosis and common guidelines are still lacking. We conducted a comprehensive analysis of established EV separation techniques from human serum and plasma, including ultracentrifugation and size exclusion chromatography (SEC), followed by concentration using (a) ultracentrifugation, (b) ultrafiltration, or (c) precipitation, and immunoaffinity isolation. We analyzed the size, number, protein, and miRNA content of the obtained EVs and assessed the functional delivery of EV cargo. Our results demonstrate that all methods led to an adequate yield of small EVs. While no significant difference in miRNA content was observed for the different separation methods, ultracentrifugation was best for subsequent flow cytometry analysis. Immunoaffinity isolation is not suitable for subsequent protein analyses. SEC + ultracentrifugation showed the best functional delivery of EV cargo. In summary, combining SEC with ultracentrifugation gives the highest yield of pure and functional EVs and allows reliable analysis of both protein and miRNA contents. We propose this combination as the preferred EV isolation method for biomarker studies from human serum or plasma.


Subject(s)
Cell Fractionation , Chemical Fractionation , Extracellular Vesicles/metabolism , Biological Transport , Biomarkers , Cell Fractionation/methods , Chemical Fractionation/methods , Extracellular Vesicles/ultrastructure , Flow Cytometry , Humans , Liquid Biopsy/methods , Proteins/metabolism
9.
Curr Opin Infect Dis ; 34(3): 197-205, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33899754

ABSTRACT

PURPOSE OF REVIEW: An initial intracellular phase of usually extracellular bacterial pathogens displays an important strategy to hide from the host's immune system and antibiotics therapy. It helps the bacteria, including bacterial pathogens of airway diseases, to persist and eventually switch to a typical extracellular infection. Several infectious diseases of the lung are life-threatening and their control is impeded by intracellular persistence of pathogens. Thus, molecular adaptations of the pathogens to this niche but also the host's response and potential targets to interfere are of relevance. Here we discuss examples of historically considered extracellular pathogens of the respiratory airway where the intracellular survival and proliferation is well documented, including infections by Staphylococcus aureus, Bordetella pertussis, Haemophilus influenzae, Pseudomonas aeruginosa, and others. RECENT FINDINGS: Current studies focus on bacterial factors contributing to adhesion, iron acquisition, and intracellular survival as well as ways to target them for combatting the bacterial infections. SUMMARY: The investigation of common and specific mechanisms of pathogenesis and persistence of these bacteria in the host may contribute to future investigations and identifications of relevant factors and/or bacterial mechanisms to be blocked to treat or improve prevention strategies.


Subject(s)
Bacteria/metabolism , Bacterial Infections/microbiology , Respiratory Tract Infections/microbiology , Host-Pathogen Interactions , Humans , Iron/metabolism
10.
Toxins (Basel) ; 12(11)2020 11 18.
Article in English | MEDLINE | ID: mdl-33218049

ABSTRACT

Pneumonia is an acute pulmonary infection associated with high mortality and an immense financial burden on healthcare systems. Staphylococcus aureus is an opportunistic pathogen capable of inducing S. aureus pneumonia (SAP), with some lineages also showing multidrug resistance. Given the high level of antibiotic resistance, much research has been focused on targeting S. aureus virulence factors, including toxins and biofilm-associated proteins, in an attempt to develop effective SAP therapeutics. Despite several promising leads, many hurdles still remain for S. aureus vaccine research. Here, we review the state-of-the-art SAP therapeutics, highlight their pitfalls, and discuss alternative approaches of potential significance and future perspectives.


Subject(s)
Pneumonia, Staphylococcal/therapy , Staphylococcus aureus , Virulence Factors , Animals , Bacterial Vaccines/therapeutic use , Biofilms , Genomics , Humans , Metabolomics , Pneumonia, Staphylococcal/genetics , Pneumonia, Staphylococcal/metabolism , Staphylococcus aureus/pathogenicity , Staphylococcus aureus/physiology
11.
Virulence ; 11(1): 1310-1328, 2020 12.
Article in English | MEDLINE | ID: mdl-33017224

ABSTRACT

Nasopharyngeal colonization by Streptococcus pneumoniae is a prerequisite for pneumococcal transmission and disease. Current vaccines protect only against disease and colonization caused by a limited number of serotypes, consequently allowing serotype replacement and transmission. Therefore, the development of a broadly protective vaccine against colonization, transmission and disease is desired but requires a better understanding of pneumococcal adaptation to its natural niche. Hence, we measured the levels of free and protein-bound transition metals in human nasal fluid, to determine the effect of metal concentrations on the growth and proteome of S. pneumoniae. Pneumococci cultured in medium containing metal levels comparable to nasal fluid showed a highly distinct proteomic profile compared to standard culture conditions, including the increased abundance of nine conserved, putative surface-exposed proteins. AliA, an oligopeptide binding protein, was identified as the strongest protective antigen, demonstrated by the significantly reduced bacterial load in a murine colonization and a lethal mouse pneumonia model, highlighting its potential as vaccine antigen.


Subject(s)
Antigens, Bacterial/isolation & purification , Membrane Proteins/isolation & purification , Metals/pharmacology , Pneumococcal Vaccines/immunology , Streptococcus pneumoniae/drug effects , Adult , Animals , Antibodies, Bacterial/blood , Antibodies, Bacterial/immunology , Antigens, Bacterial/immunology , Culture Media/chemistry , Disease Models, Animal , Female , Humans , Male , Membrane Proteins/immunology , Metals/analysis , Mice , Mice, Inbred C57BL , Middle Aged , Nasal Lavage Fluid/chemistry , Nasopharynx/microbiology , Pneumococcal Infections/immunology , Pneumococcal Infections/prevention & control , Pneumococcal Vaccines/administration & dosage , Young Adult
12.
ACS Infect Dis ; 6(8): 2279-2290, 2020 08 14.
Article in English | MEDLINE | ID: mdl-32579327

ABSTRACT

The primary barrier that protects our lungs against infection by pathogens is a tightly sealed layer of epithelial cells. When the integrity of this barrier is disrupted as a consequence of chronic pulmonary diseases or viral insults, bacterial pathogens will gain access to underlying tissues. A major pathogen that can take advantage of such conditions is Staphylococcus aureus, thereby causing severe pneumonia. In this study, we investigated how S. aureus responds to different conditions of the human epithelium, especially nonpolarization and fibrogenesis during regeneration using an in vitro infection model. The infective process was monitored by quantification of the epithelial cell and bacterial populations, fluorescence microscopy, and mass spectrometry. The results uncover differences in bacterial internalization and population dynamics that correlate with the outcome of infection. Protein profiling reveals that, irrespective of the polarization state of the epithelial cells, the invading bacteria mount similar responses to adapt to the intracellular milieu. Remarkably, a bacterial adaptation that was associated with the regeneration state of the epithelial cells concerned the early upregulation of proteins controlled by the redox-responsive regulator Rex when bacteria were confronted with a polarized cell layer. This is indicative of the modulation of the bacterial cytoplasmic redox state to maintain homeostasis early during infection even before internalization. Our present observations provide a deeper insight into how S. aureus can take advantage of a breached epithelial barrier and show that infected epithelial cells have limited ability to respond adequately to staphylococcal insults.


Subject(s)
Staphylococcal Infections , Staphylococcus aureus , Epithelial Cells , Epithelium , Humans , Regeneration
13.
mBio ; 11(2)2020 03 24.
Article in English | MEDLINE | ID: mdl-32209695

ABSTRACT

Legionella pneumophila is an important cause of pneumonia. It invades alveolar macrophages and manipulates the immune response by interfering with signaling pathways and gene transcription to support its own replication. MicroRNAs (miRNAs) are critical posttranscriptional regulators of gene expression and are involved in defense against bacterial infections. Several pathogens have been shown to exploit the host miRNA machinery to their advantage. We therefore hypothesize that macrophage miRNAs exert positive or negative control over Legionella intracellular replication. We found significant regulation of 85 miRNAs in human macrophages upon L. pneumophila infection. Chromatin immunoprecipitation and sequencing revealed concordant changes of histone acetylation at the putative promoters. Interestingly, a trio of miRNAs (miR-125b, miR-221, and miR-579) was found to significantly affect intracellular L. pneumophila replication in a cooperative manner. Using proteome-analysis, we pinpointed this effect to a concerted downregulation of galectin-8 (LGALS8), DExD/H-box helicase 58 (DDX58), tumor protein P53 (TP53), and then MX dynamin-like GTPase 1 (MX1) by the three miRNAs. In summary, our results demonstrate a new miRNA-controlled immune network restricting Legionella replication in human macrophages.IMPORTANCE Cases of Legionella pneumophila pneumonia occur worldwide, with potentially fatal outcome. When causing human disease, Legionella injects a plethora of virulence factors to reprogram macrophages to circumvent immune defense and create a replication niche. By analyzing Legionella-induced changes in miRNA expression and genomewide chromatin modifications in primary human macrophages, we identified a cell-autonomous immune network restricting Legionella growth. This network comprises three miRNAs governing expression of the cytosolic RNA receptor DDX58/RIG-I, the tumor suppressor TP53, the antibacterial effector LGALS8, and MX1, which has been described as an antiviral factor. Our findings for the first time link TP53, LGALS8, DDX58, and MX1 in one miRNA-regulated network and integrate them into a functional node in the defense against L. pneumophila.


Subject(s)
Galectins/genetics , Host-Pathogen Interactions , Legionella pneumophila/physiology , Macrophages/microbiology , MicroRNAs/genetics , Myxovirus Resistance Proteins/genetics , Galectins/metabolism , Gene Expression Regulation/immunology , Humans , Legionnaires' Disease/microbiology , Macrophages/immunology , MicroRNAs/immunology , Myxovirus Resistance Proteins/metabolism , Proteome , Signal Transduction , THP-1 Cells , Virulence Factors
14.
J Proteomics ; 211: 103559, 2020 01 16.
Article in English | MEDLINE | ID: mdl-31669358

ABSTRACT

B. pertussis is the etiological agent of whooping cough, a highly contagious respiratory disease which remains uncontrolled worldwide. Understanding how this pathogen responds to the environmental changes and adapts to different niches found inside the host might contribute to gain insight into bacterial pathogenesis. Comparative analyses of previous transcriptomic and proteomic data suggested that post-transcriptional regulatory mechanisms modulate B. pertussis virulence in response to iron availability. Iron scarcity represents one of the major stresses faced by bacterial pathogens inside the host. In this study, we used gel-free nanoLC-MS/MS-based proteomics to investigate whether Hfq, a highly conserved post-transcriptional regulatory protein, is involved in B. pertussis adaptation to low iron environment. To this end, we compared the protein profiles of wild type B. pertussis and its isogenic hfq deletion mutant strain under iron-replete and iron-depleted conditions. Almost of 33% of the proteins identified under iron starvation was found to be Hfq-dependent. Among them, proteins involved in oxidative stress tolerance and virulence factors that play a key role in the early steps of host colonization and bacterial persistence inside the host cells. Altogether these results suggest that Hfq shapes the infective phenotype of B. pertussis. SIGNIFICANCE: In the last years, it became evident that post-transcriptional regulation of gene expression in ba cteria plays a central role in host-pathogen interactions. Hfq is a bacterial protein that regulates gene expression at post-transcriptional level found pivotal in the establishment of successful infections. In this study, we investigated the role of Hfq in Bordetella pertussis response to iron starvation, one of the main stresses imposed by the host. The data demonstrate that Hfq regulates the abundance of a significant number of B. pertussis proteins in response to iron starvation. Among them, virulence factors and proteins involved in oxidative stress tolerance, key players in host colonization and intracellular bacterial survival. Altogether, our results suggest a relevant role of Hfq in B. pertussis adaptation to the different niches found inside the host eventually granting bacterial pathogenesis.


Subject(s)
Bordetella pertussis , Proteomics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bordetella pertussis/metabolism , Gene Expression Regulation, Bacterial , Tandem Mass Spectrometry , Virulence , Virulence Factors
15.
J Proteomics ; 212: 103583, 2020 02 10.
Article in English | MEDLINE | ID: mdl-31734389

ABSTRACT

In the absence of sugars, C4-dicarboxylates (C4DC) like fumarate represent important substrates for growth of Escherichia coli. Aerobically, C4DCs are oxidized to CO2 whereas anaerobically, C4DCs are used for fumarate respiration. In order to determine the impact of fumarate under aerobic and anaerobic conditions, proteomes of E. coli W3110 grown aerobically or anaerobically with fumarate and/or the non-C4DC substrate glycerol were comparatively profiled by nanoLC-MS/MS. Membrane enrichment allowed sensitive detection of membrane proteins. A total of 1657 proteins of which 646 and 374 were assigned to the cytosol or membrane, respectively, were covered. Presence of fumarate triggered changes (≥ 2fold) to the levels of 211 and 76 proteins under aerobic and anaerobic growth, respectively. The fumarate induced changes included proteins encoded by genes regulated by the C4DC two-component system DcuS-DcuR (DctA, DcuB, FumB, FrdABC proteins) catalyzing uptake and initial catabolic steps. Many of the proteins displaying altered levels are not part of the DcuS-DcuR regulon, including proteins of citric acid cycle and associated pathways (aerobic), proteins involved in motility and chemotaxis (anaerobic), and oxidative stress. Their genes are mostly preceded by cAMP receptor protein (CRP) sites, some by DcuR-like sites. Testing of selected genes confirmed regulation by CRP and DcuS-DcuR. SIGNIFICANCE: Global protein profiling of the soluble and the membrane fraction provides a comprehensive view on the protein pattern of E. coli grown aerobically and anaerobically with or without fumarate. The results disclose during aerobic growth besides the known impact of the C4-dicarboxylates (C4DC) on carbon utilization and citric acid cycle major adaptations in amino acid metabolism. In contrast, protein alterations in the presence of fumarate under anaerobic conditions point to enhanced motility and chemotaxis. Only proteins (transporters, initial metabolic steps) feeding external C4DCs to the central pathways were regulated by the C4DC two-component system DcuS-DcuR, whereas other protein levels were controlled in an indirect manner by CRP triggered catabolite control and other mechanisms. Consequently, metabolic and transcriptional regulation by C4DCs is apparently effected by a network of the DcuS-DcuR system with important contribution by catabolite control.


Subject(s)
Escherichia coli Proteins/metabolism , Escherichia coli/growth & development , Fumarates/pharmacology , Gene Expression Regulation, Bacterial/drug effects , Proteomics/methods , Aerobiosis , Anaerobiosis , DNA-Binding Proteins/metabolism , Dicarboxylic Acids/metabolism , Dicarboxylic Acids/pharmacology , Escherichia coli/drug effects , Escherichia coli/metabolism , Fumarates/metabolism , Protein Kinases/metabolism , Tandem Mass Spectrometry/methods , Transcription Factors/metabolism
16.
Proteomics ; 19(23): e1900192, 2019 12.
Article in English | MEDLINE | ID: mdl-31532911

ABSTRACT

Proteome analyses are often hampered by the low amount of available starting material like a low bacterial cell number obtained from in vivo settings. Here, the single pot solid-phase enhanced sample preparation (SP3) protocol is adapted and combined with effective cell disruption using detergents for the proteome analysis of bacteria available in limited numbers only. Using this optimized protocol, identification of peptides and proteins for different Gram-positive and Gram-negative species can be dramatically increased and, reliable quantification can also be ensured. This adapted method is compared to already established strain-specific sample processing protocols for Staphylococcus aureus, Streptococcus suis, and Legionella pneumophila. The highest species-specific increase in identifications is observed using the adapted method with L. pneumophila samples by increasing protein and peptide identifications up to 300% and 620%, respectively. This increase is accompanied by an improvement in reproducibility of protein quantification and data completeness between replicates. Thus, this protocol is of interest for performing comprehensive proteomics analyses of low bacterial cell numbers from different settings ranging from infection assays to environmental samples.


Subject(s)
Bacteria/metabolism , Proteome/analysis , Proteomics/methods , Bacterial Proteins/metabolism , Legionella pneumophila/metabolism , Staphylococcus aureus/metabolism , Streptococcus suis/metabolism
17.
J Proteomics ; 206: 103448, 2019 08 30.
Article in English | MEDLINE | ID: mdl-31325608

ABSTRACT

Bordetella parapertussis is one of the pathogens that cause whooping cough. Even though its incidence has been rising in the last decades, this species remained poorly investigated. This study reports the first extensive proteome analysis of this bacterium. In an attempt to gain some insight into the infective phenotype, we evaluated the response of B. parapertussis to iron starvation, a critical stress the bacteria face during infection. Among other relevant findings, we observed that the adaptation to this condition involves significant changes in the abundance of two important virulence factors of this pathogen, namely, adenylate cyclase and the O-antigen. We further used the proteomic data to search for B. parapertussis proteins that are absent or classified as pseudogenes in the genome of Bordetella pertussis to unravel differences between both whooping cough causative agents. Among them, we identified proteins involved in stress resistance and virulence determinants that might help to explain the differences in the pathogenesis of these species and the lack of cross-protection of current acellular vaccines. Altogether, these results contribute to a better understanding of B. parapertussis biology and pathogenesis. SIGNIFICANCE: Whooping cough is a reemerging disease caused by both Bordetella pertussis and Bordetella parapertussis. Current vaccines fail to induce protection against B parapertussis and the incidence of this species has been rising over the years. The proteomic analysis of this study provided relevant insights into potential virulence determinants of this poorly-studied pathogen. It further identified proteins produced by B. parapertussis not present in B. pertussis, which might help to explain both the differences on their respective infectious process and the current vaccine failure. Altogether, the results of this study contribute to the better understanding of B. parapertussis pathogenesis and the eventual design of improved preventive strategies against whooping cough.


Subject(s)
Bordetella parapertussis/metabolism , Bordetella pertussis/metabolism , Iron Deficiencies , Proteomics/methods , Virulence Factors/metabolism , Bacterial Proteins/analysis , Bacterial Proteins/drug effects , Bacterial Proteins/metabolism , Bordetella parapertussis/drug effects , Bordetella parapertussis/pathogenicity , Bordetella pertussis/pathogenicity , Cells, Cultured , Culture Media/chemistry , Culture Media/pharmacology , Humans , Iron/metabolism , Iron/pharmacology , Phenotype , Proteome/analysis , Proteome/metabolism , Virulence/drug effects
18.
Mol Cell Proteomics ; 18(5): 892-908, 2019 05.
Article in English | MEDLINE | ID: mdl-30808728

ABSTRACT

Staphylococcus aureus is infamous for causing recurrent infections of the human respiratory tract. This is a consequence of its ability to adapt to different niches, including the intracellular milieu of lung epithelial cells. To understand the dynamic interplay between epithelial cells and the intracellular pathogen, we dissected their interactions over 4 days by mass spectrometry. Additionally, we investigated the dynamics of infection through live cell imaging, immunofluorescence and electron microscopy. The results highlight a major role of often overlooked temporal changes in the bacterial and host metabolism, triggered by fierce competition over limited resources. Remarkably, replicating bacteria reside predominantly within membrane-enclosed compartments and induce apoptosis of the host within ∼24 h post infection. Surviving infected host cells carry a subpopulation of non-replicating bacteria in the cytoplasm that persists. Altogether, we conclude that, besides the production of virulence factors by bacteria, it is the way in which intracellular resources are used, and how host and intracellular bacteria subsequently adapt to each other that determines the ultimate outcome of the infectious process.


Subject(s)
Bronchi/pathology , Endocytosis , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Staphylococcal Infections/pathology , Staphylococcus aureus/metabolism , Apoptosis , Bacterial Proteins/metabolism , Cell Line , Cytosol/metabolism , Epithelial Cells/ultrastructure , Host-Pathogen Interactions , Humans , Proteome/metabolism , Staphylococcus aureus/ultrastructure
19.
Methods Mol Biol ; 1841: 207-228, 2018.
Article in English | MEDLINE | ID: mdl-30259489

ABSTRACT

Proteome profiling of bacteria internalized by host cells is still a challenging task, due to low amounts of bacterial proteins in host-pathogen settings and the high amounts of contaminating host proteins. Here, we describe a workflow for the enrichment of intracellular bacteria by fluorescence activated cell sorting which in combination with highly sensitive LC-MS/MS allows monitoring of about 1200 proteins from 2 to 4 × 106 internalized bacterial cells as starting material.


Subject(s)
Adaptation, Biological , Bacterial Proteins , Host-Pathogen Interactions , Proteome , Proteomics , Bacterial Proteins/metabolism , Cell Line , Chromatography, Liquid , Humans , Mass Spectrometry , Microscopy, Fluorescence , Peptides , Proteomics/methods , Workflow
20.
PLoS One ; 13(8): e0203204, 2018.
Article in English | MEDLINE | ID: mdl-30161230

ABSTRACT

Bordetella pertussis, the causative agent of whooping cough, has the capability to survive inside the host cells. This process requires efficient adaptation of the pathogen to the intracellular environment and the associated stress. Among the proteins produced by the intracellular B. pertussis we identified a protein (BP0414) that shares homology with MgtC, a protein which was previously shown to be involved in the intracellular survival of other pathogens. To explore if BP0414 plays a role in B. pertussis intracellular survival a mutant strain defective in the production of this protein was constructed. Using standard in vitro growth conditions we found that BP0414 is required for B. pertussis growth under low magnesium availability or low pH, two environmental conditions that this pathogen might face within the host cell. Intracellular survival studies showed that MgtC is indeed involved in B. pertussis viability inside the macrophages. The use of bafilomycin A1, which inhibits phagosome acidification, abolished the survival defect of the mgtC deficient mutant strain suggesting that in intracellular B. pertussis the role of MgtC protein is mainly related to the bacterial adaptation to the acidic conditions found inside the of phagosomes. Overall, this work provides an insight into the importance of MgtC in B. pertussis pathogenesis and its contribution to bacterial survival within immune cells.


Subject(s)
Bacterial Proteins/metabolism , Bordetella pertussis/metabolism , Bacterial Proteins/genetics , Bordetella pertussis/drug effects , Bordetella pertussis/genetics , Bordetella pertussis/growth & development , Cations, Divalent/metabolism , Enzyme Inhibitors/pharmacology , Escherichia coli , Humans , Hydrogen-Ion Concentration , Macrolides/pharmacology , Macrophages/drug effects , Macrophages/microbiology , Macrophages/pathology , Magnesium/metabolism , Mutation , Sequence Homology, Amino Acid , THP-1 Cells
SELECTION OF CITATIONS
SEARCH DETAIL
...