Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Elife ; 102021 01 06.
Article in English | MEDLINE | ID: mdl-33404502

ABSTRACT

The hydrothermal vent tubeworm Riftia pachyptila hosts a single 16S rRNA phylotype of intracellular sulfur-oxidizing symbionts, which vary considerably in cell morphology and exhibit a remarkable degree of physiological diversity and redundancy, even in the same host. To elucidate whether multiple metabolic routes are employed in the same cells or rather in distinct symbiont subpopulations, we enriched symbionts according to cell size by density gradient centrifugation. Metaproteomic analysis, microscopy, and flow cytometry strongly suggest that Riftia symbiont cells of different sizes represent metabolically dissimilar stages of a physiological differentiation process: While small symbionts actively divide and may establish cellular symbiont-host interaction, large symbionts apparently do not divide, but still replicate DNA, leading to DNA endoreduplication. Moreover, in large symbionts, carbon fixation and biomass production seem to be metabolic priorities. We propose that this division of labor between smaller and larger symbionts benefits the productivity of the symbiosis as a whole.


Subject(s)
Bacterial Physiological Phenomena , Polychaeta/microbiology , Symbiosis , Animals , Bacteria/isolation & purification , Hydrothermal Vents/microbiology
2.
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
3.
PLoS One ; 15(5): e0233854, 2020.
Article in English | MEDLINE | ID: mdl-32470006

ABSTRACT

Alpha-toxin (Hla) is a major virulence factor of Staphylococcus aureus (S. aureus) and plays an important role in S. aureus-induced pneumonia. It binds as a monomer to the cell surface of eukaryotic host cells and forms heptameric transmembrane pores. Sensitivities toward the toxin of various types of potential host cells have been shown to vary substantially, and the reasons for these differences are unclear. We used three human model airway epithelial cell lines (16HBE14o-, S9, A549) to correlate cell sensitivity (measured as rate of paracellular gap formation in the cell layers) with Hla monomer binding, presence of the potential Hla receptors ADAM10 or α5ß1 integrin, presence of the toxin-stabilizing factor caveolin-1 as well as plasma membrane lipid composition (phosphatidylserine/choline, sphingomyelin). The abundance of ADAM10 correlated best with gap formation or cell sensitivities, respectively, when the three cell types were compared. Caveolin-1 or α5ß1 integrin did not correlate with toxin sensitivity. The relative abundance of sphingomyelin in plasma membranes may also be used as a proxi for cellular sensitivity against alpha-toxin as sphingomyelin abundances correlated well with the intensities of alpha-toxin mediated gap formation in the cell layers.


Subject(s)
Bacterial Toxins/metabolism , Bacterial Toxins/toxicity , Cell Membrane/metabolism , Epithelial Cells/metabolism , Hemolysin Proteins/metabolism , Hemolysin Proteins/toxicity , Host-Pathogen Interactions , Respiratory System/pathology , A549 Cells , Caveolin 1/metabolism , Cell Membrane/drug effects , Cell Size , Epithelial Cells/drug effects , Host-Pathogen Interactions/drug effects , Humans , Models, Biological , Phospholipids/metabolism , Protein Binding , Receptors, Cell Surface/metabolism
4.
Environ Microbiol ; 22(6): 2312-2328, 2020 06.
Article in English | MEDLINE | ID: mdl-32249531

ABSTRACT

Upon competence-inducing nutrient-limited conditions, only part of the Bacillus subtilis population becomes competent. Here, we separated the two subpopulations by fluorescence-assisted cell sorting (FACS). Using RNA-seq, we confirmed the previously described ComK regulon. We also found for the first time significantly downregulated genes in the competent subpopulation. The downregulated genes are not under direct control by ComK but have higher levels of corresponding antisense RNAs in the competent subpopulation. During competence, cell division and replication are halted. By investigating the proteome during competence, we found higher levels of the regulators of cell division, MinD and Noc. The exonucleases SbcC and SbcD were also primarily regulated at the post-transcriptional level. In the competent subpopulation, yhfW was newly identified as being highly upregulated. Its absence reduces the expression of comG, and has a modest, but statistically significant effect on the expression of comK. Although expression of yhfW is higher in the competent subpopulation, no ComK-binding site is present in its promoter region. Mutants of yhfW have a small but significant defect in transformation. Metabolomic analyses revealed significant reductions in tricarboxylic acid (TCA) cycle metabolites and several amino acids in a ΔyhfW mutant. RNA-seq analysis of ΔyhfW revealed higher expression of the NAD synthesis genes nadA, nadB and nadC.


Subject(s)
Bacillus subtilis/genetics , Bacterial Proteins/genetics , RNA, Untranslated , Bacillus subtilis/metabolism , Down-Regulation , Regulon , Up-Regulation
5.
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
6.
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
7.
Int J Med Microbiol ; 308(6): 664-674, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29941384

ABSTRACT

Staphylococcus aureus, an opportunistic pathogen is able to invade into and persist inside non-professional phagocytic cells. To do so, this bacterium possesses a wide range of secreted virulence factors which enable attachment to the host as well as intracellular survival. Hence, a monitoring of virulence factors specifically produced upon internalization might reveal targets for prevention or therapy of S. aureus infections. However, previous proteome approaches enriching S. aureus from lysed host cells after infection did not cover secreted virulence factors. Therefore, we used density gradient centrifugation and mass spectrometry to identify S. aureus HG001 proteins which were secreted into compartments of infected human bronchial epithelial S9 cells. Because shotgun mass spectrometry revealed only few bacterial proteins amongst 1905 host proteins, we used highly sensitive and selective single reaction monitoring mass spectrometry as an alternative approach and quantified 37 bacterial proteins within the S. aureus containing host cell compartment 2.5 h and 6.5 h post infection. Among them were secreted bacterial virulence factors like lipases, pore forming toxins, and secreted adhesins which are usually hard to detect from infected sample material by proteomics approaches due to their low abundance. S. aureus adapted its proteome to improve its response to oxidative and cell wall stress occurring inside the host, but also, increased the amounts of some adhesins and pore-forming toxins, required for attachment and host cell lysis.


Subject(s)
Bacterial Proteins/analysis , Epithelial Cells/microbiology , Host-Pathogen Interactions , Staphylococcus aureus/chemistry , Biological Transport , Bronchi/cytology , Bronchi/microbiology , Cell Line , Cells, Cultured , Centrifugation, Density Gradient , Humans , Mass Spectrometry , Proteome/analysis , Proteomics , Virulence Factors/analysis
8.
Mol Cell Biol ; 38(5)2018 03 01.
Article in English | MEDLINE | ID: mdl-29203643

ABSTRACT

The BCL11B gene encodes a Krüppel-like, sequence-specific zinc finger (ZF) transcription factor that acts as either a repressor or an activator, depending on its posttranslational modifications. The importance of BCL11B in numerous biological processes in multiple organs has been well established in mouse knockout models. The phenotype of the first de novo monoallelic germ line missense mutation in the BCL11B gene (encoding N441K) strongly implies that the mutant protein acts in a dominant-negative manner by neutralizing the unaffected protein through the formation of a nonfunctional dimer. Using a Förster resonance energy transfer-assisted fluorescence-activated cell sorting (FACS-FRET) assay and affinity purification followed by mass spectrometry (AP-MS), we show that the N-terminal CCHC zinc finger motif is necessary and sufficient for the formation of the BCL11B dimer. Mutation of the CCHC ZF in BCL11B abolishes its transcription-regulatory activity. In addition, unlike wild-type BCL11B, this mutant is incapable of inducing cell cycle arrest and protecting against DNA damage-driven apoptosis. Our results confirm the BCL11B dimerization hypothesis and prove its importance for BCL11B function. By mapping the relevant regions to the CCHC domain, we describe a previously unidentified mechanism of transcription factor homodimerization.


Subject(s)
Repressor Proteins/genetics , Repressor Proteins/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Amino Acid Sequence , Cell Culture Techniques , DNA-Binding Proteins/metabolism , Dimerization , Fluorescence Resonance Energy Transfer/methods , Germ-Line Mutation , Humans , Mass Spectrometry/methods , Mutation, Missense , Protein Domains , Transcription Factors/genetics , Transcription Factors/metabolism , Zinc Fingers
9.
Sci Rep ; 7(1): 9718, 2017 09 08.
Article in English | MEDLINE | ID: mdl-28887440

ABSTRACT

Data-independent acquisition mass spectrometry promises higher performance in terms of quantification and reproducibility compared to data-dependent acquisition mass spectrometry methods. To enable high-accuracy quantification of Staphylococcus aureus proteins, we have developed a global ion library for data-independent acquisition approaches employing high-resolution time of flight or Orbitrap instruments for this human pathogen. We applied this ion library resource to investigate the time-resolved adaptation of S. aureus to the intracellular niche in human bronchial epithelial cells and in a murine pneumonia model. In epithelial cells, abundance changes for more than 400 S. aureus proteins were quantified, revealing, e.g., the precise temporal regulation of the SigB-dependent stress response and differential regulation of translation, fermentation, and amino acid biosynthesis. Using an in vivo murine pneumonia model, our data-independent acquisition quantification analysis revealed for the first time the in vivo proteome adaptation of S. aureus. From approximately 2.15 × 105 S. aureus cells, 578 proteins were identified. Increased abundance of proteins required for oxidative stress response, amino acid biosynthesis, and fermentation together with decreased abundance of ribosomal proteins and nucleotide reductase NrdEF was observed in post-infection samples compared to the pre-infection state.


Subject(s)
Bacterial Proteins/metabolism , Host-Pathogen Interactions , Proteome , Proteomics , Staphylococcal Infections/metabolism , Staphylococcal Infections/microbiology , Staphylococcus aureus/metabolism , Animals , Computational Biology/methods , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Humans , Ions/metabolism , Mice , Peptides , Proteomics/methods , Respiratory Mucosa/metabolism , Respiratory Mucosa/microbiology
10.
Data Brief ; 7: 1031-1037, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27761493

ABSTRACT

To simultaneously obtain proteome data of host and pathogen from an internalization experiment, human alveolar epithelial A549 cells were infected with Staphylococcus aureus HG001 which carried a plasmid (pMV158GFP) encoding a continuously expressed green fluorescent protein (GFP). Samples were taken hourly between 1.5 h and 6.5 h post infection. By fluorescence activated cell sorting GFP-expressing bacteria could be enriched from host cell debris, but also infected host cells could be separated from those which did not carry bacteria after contact (exposed). Additionally, proteome data of A549 cells which were not exposed to S. aureus but underwent the same sample processing steps are provided as a control. Time-resolved changes in bacterial protein abundance were quantified in a label-free approach. Proteome adaptations of host cells were monitored by comparative analysis to a stable isotope labeled cell culture (SILAC) standard. Proteins were extracted from the cells, digested proteolytically, measured by nanoLC-MS/MS, and subsequently identified by database search and then quantified. The data presented here are related to a previously published research article describing the interplay of S. aureus HG001 and human epithelial cells (Surmann et al., 2015 [1]). They have been deposited to the ProteomeXchange platform with the identifiers PRIDE: http://www.ebi.ac.uk/pride/archive/projects/PXD002384 for the S. aureus HG001 proteome dataset and PRIDE: http://www.ebi.ac.uk/pride/archive/projects/PXD002388 for the A549 proteome dataset.

11.
Cytometry A ; 89(10): 932-940, 2016 10.
Article in English | MEDLINE | ID: mdl-27643682

ABSTRACT

Staphylococcus aureus is a Gram-positive opportunistic pathogen that is able to cause a broad range of infectious diseases in humans. Furthermore, S. aureus is able to survive inside nonprofessional phagocytic host cell which serve as a niche for the pathogen to hide from the immune system and antibiotics therapies. Modern OMICs technologies provide valuable tools to investigate host-pathogen interactions upon internalization. However, these experiments are often hampered by limited capabilities to retrieve bacteria from such an experimental setting. Thus, the aim of this study was to develop a labeling strategy allowing fast detection and quantitation of S. aureus in cell lysates or infected cell lines by flow cytometry for subsequent proteome analyses. Therefore, S. aureus cells were labeled with the DNA stain SYTO® 9, or Vancomycin BODIPY® FL (VMB), a glycopeptide antibiotic binding to most Gram-positive bacteria which was conjugated to a fluorescent dye. Staining of S. aureus HG001 with SYTO 9 allowed counting of bacteria from pure cultures but not in cell lysates from infection experiments. In contrast, with VMB it was feasible to stain bacteria from pure cultures as well as from samples of infection experiments. VMB can also be applied for histocytochemistry analysis of formaldehyde fixed cell layers grown on coverslips. Proteome analyses of S. aureus labeled with VMB revealed that the labeling procedure provoked only minor changes on proteome level and allowed cell sorting and analysis of S. aureus from infection settings with sensitivity similar to continuous gfp expression. Furthermore, VMB labeling allowed precise counting of internalized bacteria and can be employed for downstream analyses, e.g., proteomics, of strains not easily amendable to genetic manipulation such as clinical isolates. © 2016 International Society for Advancement of Cytometry.


Subject(s)
Fluorescent Dyes/metabolism , Host-Pathogen Interactions/physiology , Proteome/metabolism , Staining and Labeling/methods , Staphylococcal Infections/microbiology , Staphylococcus aureus/metabolism , Adult , Aged, 80 and over , Bacterial Proteins/metabolism , Cell Line , DNA, Bacterial/genetics , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Female , Flow Cytometry/methods , Humans , Male , Proteomics/methods , Staphylococcal Infections/metabolism
12.
J Proteomics ; 128: 203-17, 2015 Oct 14.
Article in English | MEDLINE | ID: mdl-26244908

ABSTRACT

Infectious diseases caused by pathogens such as Staphylococcus aureus are still a major threat for human health. Proteome analyses allow detailed monitoring of the molecular interplay between pathogen and host upon internalization. However, the investigation of the responses of both partners is complicated by the large excess of host cell proteins compared to bacterial proteins as well as by the fact that only a fraction of host cells are infected. In the present study we infected human alveolar epithelial A549 cells with S. aureus HG001 pMV158GFP and separated intact bacteria from host cell debris or infected from non-infected A549 cells by cell sorting to enable detailed proteome analysis. During the first 6.5h in the intracellular milieu S. aureus displayed reduced growth rate, induction of the stringent response, adaptation to microaerobic conditions as well as cell wall stress. Interestingly, both truly infected host cells and those not infected but exposed to secreted S. aureus proteins and host cell factors showed differences in the proteome pattern compared to A549 cells which had never been in contact with S. aureus. However, adaptation reactions were more pronounced in infected compared to non-infected A549 bystander cells.


Subject(s)
Epithelial Cells/metabolism , Epithelial Cells/microbiology , Pneumonia, Staphylococcal/metabolism , Proteome/metabolism , Respiratory Mucosa/metabolism , Staphylococcus aureus/metabolism , Cell Line , Cytokines/metabolism , Host-Pathogen Interactions , Humans , Pneumonia, Staphylococcal/microbiology , Respiratory Mucosa/microbiology
13.
Front Microbiol ; 5: 392, 2014.
Article in English | MEDLINE | ID: mdl-25136337

ABSTRACT

Staphylococcus aureus is a human pathogen that can cause a wide range of diseases. Although formerly regarded as extracellular pathogen, it has been shown that S. aureus can also be internalized by host cells and persist within these cells. In the present study, we comparatively analyzed survival and physiological adaptation of S. aureus HG001 after internalization by two human lung epithelial cell lines (S9 and A549), and human embryonic kidney cells (HEK 293). Combining enrichment of bacteria from host-pathogen assays by cell sorting and quantitation of the pathogen's proteome by mass spectrometry we characterized S. aureus adaptation during the initial phase between 2.5 h and 6.5 h post-infection. Starting with about 2 × 10(6) bacteria, roughly 1450 S. aureus proteins, including virulence factors and metabolic enzymes were identified by spectral comparison and classical database searches. Most of the bacterial adaptation reactions, such as decreased levels of ribosomal proteins and metabolic enzymes or increased amounts of proteins involved in arginine and lysine biosynthesis, enzymes coding for terminal oxidases and stress responsive proteins or activation of the sigma factor SigB were observed after internalization into any of the three cell lines studied. However, differences were noted in central carbon metabolism including regulation of fermentation and threonine degradation. Since these differences coincided with different intracellular growth behavior, complementary profiling of the metabolome of the different non-infected host cell types was performed. This revealed similar levels of intracellular glucose but host cell specific differences in the amounts of amino acids such as glycine, threonine or glutamate. With this comparative study we provide an impression of the common and specific features of the adaptation of S. aureus HG001 to specific host cell environments as a starting point for follow-up studies with different strain isolates and regulatory mutants.

14.
Int J Med Microbiol ; 304(2): 177-87, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24480029

ABSTRACT

Staphylococcus aureus is a versatile pathogen that can be a commensal but also cause a wide range of different infections. This broad disease spectrum is a reflection of the complex regulation of a large collection of virulence factors that together with metabolic fitness allow adaptation to different niches. The alternative sigma factor SigB is one of the global regulators mediating this adaptation. However, even if SigB contributes to expression of many virulence factors its importance for successful infection greatly varies with the strain and the infection setting analyzed. We have recently established a proteomics workflow that combines high efficiency cell sorting with sensitive mass spectrometry and allows monitoring of global proteome adaptations with roughly one million bacterial cells. Thus, we can now approach the adaptation of pathogens to the intracellular milieu. In the current study this proteomics workflow was used in conjunction with qRT-PCR and confocal fluorescence microscopy to comparatively analyze the adaptation of the S. aureus wild type strain HG001 and its isogenic sigB mutant to the intracellular milieu of human S9 bronchial epithelial cells. The study revealed fast and transient activation of SigB following internalization by human host cells and the requirement of SigB for intracellular growth. Loss of SigB triggered proteome changes reflecting the different residual growth rates of wild type and sigB mutant, respectively, the resistance to methicillin, adaptation to oxidative stress and protein quality control mechanisms.


Subject(s)
Bacterial Proteins/biosynthesis , Endocytosis , Epithelial Cells/microbiology , Host-Pathogen Interactions , Proteome/analysis , Sigma Factor/biosynthesis , Staphylococcus aureus/physiology , Adaptation, Physiological , Bacterial Proteins/genetics , Cell Line , Gene Deletion , Gene Expression Profiling , Humans , Microscopy, Confocal , Real-Time Polymerase Chain Reaction , Sigma Factor/genetics
15.
Cytometry A ; 85(2): 140-50, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24347542

ABSTRACT

Throughout the world, infections caused by bacteria such as Staphylococcus aureus are a major cause of morbidity and mortality. In order to gain some understanding of the complicated physiological link between host and pathogen, modern techniques such as confocal microscopy and sophisticated OMICs technologies are suitable. However, labeling of pathogens such as S. aureus with green fluorescent protein, for example, or the generation of a reliable antibody, which are prerequisites for the application of reproducible isolation techniques, does not always succeed. Here, we present a universal approach for monitoring pathogen traffic after internalization into host cells by fluorescence microscopy and for isolation of bacteria from host-pathogen interaction assays using gold or ferric oxide-core, poly(vinyl alcohol) coated, and fluorescence-labeled nanoparticles (NP). The incubation of S. aureus HG001 with those NP had only minor effects on the bacterial growth in vitro. Quantitative proteome analysis after 24 h of NP incubation revealed that presence of NP provoked only marginal changes in the proteome pattern. The method presented enabled us to investigate the behavior of S. aureus HG001 during infection of S9 human epithelial cells by means of fluorescence microscopy and proteomics using magnetic separation or cell sorting.


Subject(s)
Ferric Compounds/chemistry , Gold/chemistry , Host-Pathogen Interactions , Metal Nanoparticles/chemistry , Staining and Labeling/methods , Staphylococcus aureus/growth & development , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cell Line , Epithelial Cells/cytology , Epithelial Cells/microbiology , Flow Cytometry , Fluorescent Dyes/chemistry , Gene Expression , Humans , Magnets , Microscopy, Fluorescence , Polyvinyl Alcohol/chemistry , Proteome/genetics , Proteome/metabolism , Staphylococcus aureus/ultrastructure
16.
PLoS One ; 8(5): e65109, 2013.
Article in English | MEDLINE | ID: mdl-23724126

ABSTRACT

The role of regulatory T cells (Tregs) in bacterial sepsis remains controversial because antibody-mediated depletion experiments gave conflicting results. We employed DEREG mice (DEpletion of REGulatory T cells) and a caecal ligation and puncture model to elucidate the role of CD4(+)Foxp3(+) Tregs in sepsis. In DEREG mice natural Tregs can be visualized easily and selectively depleted by diphtheria toxin because the animals express the diphtheria toxin receptor and enhanced green fluorescent protein as a fusion protein under the control of the foxp3 locus. We confirmed rapid Treg-activation and an increased ratio of Tregs to Teffs in sepsis. Nevertheless, 24 h after sepsis induction, Treg-depleted and control mice showed equally strong inflammation, immune cell immigration into the peritoneum and bacterial dissemination. During the first 36 h of disease survival was not influenced by Treg-depletion. Later, however, only Treg-competent animals recovered from the insult. We conclude that the suppressive capacity of Tregs is not sufficient to control overwhelming inflammation and early mortality, but is a prerequisite for the recovery from severe sepsis.


Subject(s)
Forkhead Transcription Factors/metabolism , Sepsis/immunology , T-Lymphocytes, Regulatory/immunology , Adoptive Transfer , Animals , Cecum/pathology , Cell Movement , Cytokines/blood , Ligation , Lymphocyte Activation/immunology , Lymphocyte Depletion , Mice , Mice, Inbred C57BL , Peritoneum/pathology , Punctures , Sepsis/blood , Sepsis/pathology , Survival Analysis
17.
Methods ; 61(3): 244-50, 2013 Jun 15.
Article in English | MEDLINE | ID: mdl-23643866

ABSTRACT

The development of a mass spectrometric workflow for the sensitive identification and quantitation of the kinetics of changes in metaproteomes, or in particular bacterial pathogens after internalization by host cells, is described. This procedure employs three essential stages: (i) SILAC pulse-chase labeling and infection assay; (ii) isolation of bacteria by GFP-assisted cell sorting; (iii) mass spectrometry-based proteome analysis. This approach displays greater sensitivity than techniques relying on conventional cell sorting and protein separation, due to an efficient combination of a filtration-based purification and an on-membrane digestion. We exemplary describe the use of the workflow for the identification and quantitation of the proteome of 106 cells of Staphylococcus aureus after internalization by S9 human bronchial epithelial cells. With minor modifications, the workflow described can be applied for the characterization of other host-pathogen pairs, permitting identification and quantitation of hundreds of bacterial proteins over a time range of several hours post infection.


Subject(s)
Bacterial Proteins/isolation & purification , Bronchi/microbiology , Epithelial Cells/microbiology , Peptides/isolation & purification , Proteomics/methods , Staphylococcus aureus/chemistry , Adaptation, Physiological , Arginine/chemistry , Arginine/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Bronchi/chemistry , Bronchi/cytology , Carbon Isotopes , Cell Line , Epithelial Cells/chemistry , Epithelial Cells/cytology , Host-Pathogen Interactions , Humans , Isotope Labeling , Lysine/chemistry , Lysine/metabolism , Mass Spectrometry , Peptides/chemistry , Staphylococcus aureus/growth & development , Staphylococcus aureus/metabolism , Time Factors
18.
Cell Microbiol ; 15(7): 1253-65, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23347173

ABSTRACT

Soluble virulence-associated factors of Staphylococcus aureus like haemolysin A (Hla) induce secretion of chemo/cytokines from airway epithelial cells. To elucidate the potential roles of specific signalling pathways in this response, we treated 16HBE14o-, S9 or A549 cells with recombinant Hla (rHla). In a dose-dependent manner, rHla induced secretion of IL-8 in all three cell types, but IL-6 release only in 16HBE14o- and S9 cells. rHla-mediated secretion of IL-8 and IL-6 was suppressed by pre-incubation of cells with inhibitors of Erk type or p38 MAP kinases, indicating that activation of these signalling pathways is essential for IL-8 release in all three cell types and for IL-6 release in 16HBE14o- and S9 cells. The rHla-mediated phosphorylation and activation of p38 MAP kinase seem to depend on elevations in [Ca(2+)]i, an early response in rHla-treated cells. Inhibitors of calmodulin or calcium/calmodulin-dependent kinase II attenuated rHla-mediated release of IL-8 in 16HBE14o- and A549 cells and of IL-6 in 16HBE14o- cells. This indicates that rHla may mediate simultaneous activation of calmodulin-dependent processes as additional prerequisites for chemo/cytokine secretion.However, the inhibitors of calmodulin-dependent signalling did not affect rHla-induced p38 MAP kinase phosphorylation, indicating that this pathway works in parallel with p38 MAP kinase.


Subject(s)
Bacterial Toxins/immunology , Epithelial Cells/immunology , Extracellular Signal-Regulated MAP Kinases/metabolism , Hemolysin Proteins/immunology , Host-Pathogen Interactions , Interleukin-6/metabolism , Interleukin-8/metabolism , MAP Kinase Signaling System , Cell Line , Epithelial Cells/microbiology , Humans , Staphylococcus aureus/immunology
19.
J Biomed Mater Res B Appl Biomater ; 100(1): 41-50, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22114061

ABSTRACT

Magnesium-based bioabsorbable cardiovascular stents have been developed to overcome limitations of permanent metallic stents, such as late stent thrombosis. During stent degradation, endothelial and smooth muscle cells will be exposed to locally high magnesium concentrations with yet unknown physiological consequences. Here, we investigated the effects of elevated magnesium concentrations on human coronary artery endothelial and smooth muscle cell (HCAEC, HCASMC) growth and gene expression. In the course of 24 h after incubation with magnesium chloride solutions (1 or 10 mM) intracellular magnesium level in HCASMC raised from 0.55 ± 0.25 mM (1 mM) to 1.38 ± 0.95 mM (10 mM), while no increase was detected in HCAEC. Accordingly, a DNA microarray-based study identified 69 magnesium regulated transcripts in HCAEC, but 2172 magnesium regulated transcripts in HCASMC. Notably, a significant regulation of various growth factors and extracellular matrix components was observed. In contrast, viability and proliferation of HCAEC were increased at concentrations of up to 25 mM magnesium chloride, while in HCASMC viability and proliferation appeared to be unaffected. Taken together, our data indicate that magnesium halts smooth muscle cell proliferation and stimulates endothelial cell proliferation, which might translate into a beneficial effect in the setting of stent associated vascular injury.


Subject(s)
Absorbable Implants , Cell Proliferation , Coronary Vessels/metabolism , Endothelial Cells/metabolism , Magnesium , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Stents , Cell Survival , Cells, Cultured , Coronary Vessels/cytology , Coronary Vessels/injuries , Endothelial Cells/cytology , Gene Expression Regulation , Humans , Materials Testing/methods , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/cytology
20.
PLoS One ; 5(9)2010 Sep 02.
Article in English | MEDLINE | ID: mdl-20824091

ABSTRACT

BACKGROUND: The expression of BCL11B was reported in T-cells, neurons and keratinocytes. Aberrations of BCL11B locus leading to abnormal gene transcription were identified in human hematological disorders and corresponding animal models. Recently, the elevated levels of Bcl11b protein have been described in a subset of squameous cell carcinoma cases. Despite the rapidly accumulating knowledge concerning Bcl11b biology, the contribution of this protein to normal or transformed cell homeostasis remains open. METHODOLOGY/PRINCIPAL FINDINGS: Here, by employing an overexpression strategy we revealed formerly unidentified features of Bcl11b. Two different T-cell lines were forced to express BCL11B at levels similar to those observed in primary T-cell leukemias. This resulted in markedly increased resistance to radiomimetic drugs while no influence on death-receptor apoptotic pathway was observed. Apoptosis resistance triggered by BCL11B overexpression was accompanied by a cell cycle delay caused by accumulation of cells at G1. This cell cycle restriction was associated with upregulation of CDKN1C (p57) and CDKN2C (p18) cyclin dependent kinase inhibitors. Moreover, p27 and p130 proteins accumulated and the SKP2 gene encoding a protein of the ubiquitin-binding complex responsible for their degradation was repressed. Furthermore, the expression of the MYCN oncogene was silenced which resulted in significant depletion of the protein in cells expressing high BCL11B levels. Both cell cycle restriction and resistance to DNA-damage-induced apoptosis coincided and required the histone deacetylase binding N-terminal domain of Bcl11b. The sensitivity to genotoxic stress could be restored by the histone deacetylase inhibitor trichostatine A. CONCLUSIONS: The data presented here suggest a potential role of BCL11B in tumor survival and encourage developing Bcl11b-inhibitory approaches as a potential tool to specifically target chemoresistant tumor cells.


Subject(s)
Drug Resistance, Neoplasm , G1 Phase , Gene Expression , Leukemia, T-Cell/genetics , Leukemia, T-Cell/physiopathology , Repressor Proteins/genetics , Tumor Suppressor Proteins/genetics , Apoptosis , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , DNA Damage , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Humans , Leukemia, T-Cell/drug therapy , Leukemia, T-Cell/metabolism , Protein Binding , Protein Structure, Tertiary , Repressor Proteins/chemistry , Repressor Proteins/metabolism , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...