Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Mol Cell Proteomics ; 18(5): 892-908, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30808728

RESUMEN

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.


Asunto(s)
Bronquios/patología , Endocitosis , Células Epiteliales/metabolismo , Células Epiteliales/microbiología , Infecciones Estafilocócicas/patología , Staphylococcus aureus/metabolismo , Apoptosis , Proteínas Bacterianas/metabolismo , Línea Celular , Citosol/metabolismo , Células Epiteliales/ultraestructura , Interacciones Huésped-Patógeno , Humanos , Proteoma/metabolismo , Staphylococcus aureus/ultraestructura
2.
PLoS Pathog ; 11(4): e1004870, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25923704

RESUMEN

Staphylococcus aureus is a major human pathogen that causes a range of infections from acute invasive to chronic and difficult-to-treat. Infection strategies associated with persisting S. aureus infections are bacterial host cell invasion and the bacterial ability to dynamically change phenotypes from the aggressive wild-type to small colony variants (SCVs), which are adapted for intracellular long-term persistence. The underlying mechanisms of the bacterial switching and adaptation mechanisms appear to be very dynamic, but are largely unknown. Here, we analyzed the role and the crosstalk of the global S. aureus regulators agr, sarA and SigB by generating single, double and triple mutants, and testing them with proteome analysis and in different in vitro and in vivo infection models. We were able to demonstrate that SigB is the crucial factor for adaptation in chronic infections. During acute infection, the bacteria require the simultaneous action of the agr and sarA loci to defend against invading immune cells by causing inflammation and cytotoxicity and to escape from phagosomes in their host cells that enable them to settle an infection at high bacterial density. To persist intracellularly the bacteria subsequently need to silence agr and sarA. Indeed agr and sarA deletion mutants expressed a much lower number of virulence factors and could persist at high numbers intracellularly. SigB plays a crucial function to promote bacterial intracellular persistence. In fact, ΔsigB-mutants did not generate SCVs and were completely cleared by the host cells within a few days. In this study we identified SigB as an essential factor that enables the bacteria to switch from the highly aggressive phenotype that settles an acute infection to a silent SCV-phenotype that allows for long-term intracellular persistence. Consequently, the SigB-operon represents a possible target to develop preventive and therapeutic strategies against chronic and therapy-refractory infections.


Asunto(s)
Proteínas Bacterianas/metabolismo , Endotelio Vascular/microbiología , Interacciones Huésped-Patógeno , Neutrófilos/microbiología , Osteoblastos/microbiología , Factor sigma/metabolismo , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/fisiología , Adaptación Fisiológica , Animales , Proteínas Bacterianas/genética , Línea Celular , Células Cultivadas , Endotelio Vascular/citología , Endotelio Vascular/inmunología , Endotelio Vascular/patología , Eliminación de Gen , Células Endoteliales de la Vena Umbilical Humana/citología , Células Endoteliales de la Vena Umbilical Humana/inmunología , Células Endoteliales de la Vena Umbilical Humana/microbiología , Células Endoteliales de la Vena Umbilical Humana/patología , Humanos , Ratones , Ratones Endogámicos BALB C , Mutación , Neutrófilos/citología , Neutrófilos/inmunología , Neutrófilos/patología , Osteoblastos/citología , Osteoblastos/inmunología , Osteoblastos/patología , Proteómica , Factor sigma/genética , Infecciones Estafilocócicas/inmunología , Infecciones Estafilocócicas/metabolismo , Infecciones Estafilocócicas/patología , Staphylococcus aureus/inmunología , Staphylococcus aureus/metabolismo , Factores de Tiempo , Transactivadores/genética , Transactivadores/metabolismo
3.
Mol Microbiol ; 93(6): 1259-68, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25074408

RESUMEN

With about 25 000 molecules per cell, Asp23 is one of the most abundant proteins in Staphylococcus aureus. Asp23 has been characterized as a protein that, following an alkaline shock, accumulates in the soluble protein fraction. Transcription of the asp23 gene is exclusively regulated by the alternative sigma factor σ(B) , which controls the response of the bacterium to environmental stress. Sequence analysis identified Asp23 as a member of the widely distributed Pfam DUF322 family, precluding functional predictions based on its sequence. Using fluorescence microscopy we found that Asp23 colocalized with the cell membrane of Staphylococcus aureus. Since Asp23 has no recognizable transmembrane spanning domains, we initiated a search for proteins that link Asp23 to the cell membrane. We identified SAOUHSC_02443 as the Asp23 membrane anchor and have renamed it AmaP (Asp23 membrane anchoring protein). Deletion of the asp23 gene led to an upregulation of the cell wall stress response. In summary, we have identified Asp23 as a membrane-associated protein and we suggest a function for Asp23 in cell envelope homoeostasis.


Asunto(s)
Proteínas Bacterianas/metabolismo , Pared Celular/genética , Staphylococcus aureus/metabolismo , Secuencia de Aminoácidos , Proteínas Bacterianas/genética , Membrana Celular/metabolismo , Secuencia Conservada , Regulación Bacteriana de la Expresión Génica , Datos de Secuencia Molecular , Staphylococcus aureus/citología , Staphylococcus aureus/genética
4.
Int J Med Microbiol ; 304(2): 177-87, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24480029

RESUMEN

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.


Asunto(s)
Proteínas Bacterianas/biosíntesis , Endocitosis , Células Epiteliales/microbiología , Interacciones Huésped-Patógeno , Proteoma/análisis , Factor sigma/biosíntesis , Staphylococcus aureus/fisiología , Adaptación Fisiológica , Proteínas Bacterianas/genética , Línea Celular , Eliminación de Gen , Perfilación de la Expresión Génica , Humanos , Microscopía Confocal , Reacción en Cadena en Tiempo Real de la Polimerasa , Factor sigma/genética
5.
Methods ; 61(3): 244-50, 2013 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-23643866

RESUMEN

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.


Asunto(s)
Proteínas Bacterianas/aislamiento & purificación , Bronquios/microbiología , Células Epiteliales/microbiología , Péptidos/aislamiento & purificación , Proteómica/métodos , Staphylococcus aureus/química , Adaptación Fisiológica , Arginina/química , Arginina/metabolismo , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Bronquios/química , Bronquios/citología , Isótopos de Carbono , Línea Celular , Células Epiteliales/química , Células Epiteliales/citología , Interacciones Huésped-Patógeno , Humanos , Marcaje Isotópico , Lisina/química , Lisina/metabolismo , Espectrometría de Masas , Péptidos/química , Staphylococcus aureus/crecimiento & desarrollo , Staphylococcus aureus/metabolismo , Factores de Tiempo
6.
Mol Microbiol ; 77(4): 958-71, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20572937

RESUMEN

In most organisms, dedicated multiprotein complexes, called exosome or RNA degradosome, carry out RNA degradation and processing. In addition to varying exoribonucleases or endoribonucleases, most of these complexes contain a RNA helicase. In the Gram-positive bacterium Bacillus subtilis, a RNA degradosome has recently been described; however, no RNA helicase was identified. In this work, we tested the interaction of the four DEAD box RNA helicases encoded in the B. subtilis genome with the RNA degradosome components. One of these helicases, CshA, is able to interact with several of the degradosome proteins, i.e. RNase Y, the polynucleotide phosphorylase, and the glycolytic enzymes enolase and phosphofructokinase. The determination of in vivo protein-protein interactions revealed that CshA is indeed present in a complex with polynucleotide phosphorylase. CshA is composed of two RecA-like domains that are found in all DEAD box RNA helicases and a C-terminal domain that is present in some members of this protein family. An analysis of the contribution of the individual domains of CshA revealed that the C-terminal domain is crucial both for dimerization of CshA and for all interactions with components of the RNA degradosome, including RNase Y. A transfer of this domain to CshB allowed the resulting chimeric protein to interact with RNase Y suggesting that this domain confers interaction specificity. As a degradosome component, CshA is present in the cell in similar amounts under all conditions. Taken together, our results suggest that CshA is the functional equivalent of the RhlB helicase of the Escherichia coli RNA degradosome.


Asunto(s)
Bacillus subtilis/enzimología , Endorribonucleasas/metabolismo , Complejos Multienzimáticos/metabolismo , Polirribonucleótido Nucleotidiltransferasa/metabolismo , ARN Helicasas/metabolismo , Bacillus subtilis/genética , Endorribonucleasas/genética , Complejos Multienzimáticos/genética , Polirribonucleótido Nucleotidiltransferasa/genética , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Mapeo de Interacción de Proteínas , Multimerización de Proteína , ARN Helicasas/genética
7.
Methods Mol Biol ; 1841: 207-228, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30259489

RESUMEN

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.


Asunto(s)
Adaptación Biológica , Proteínas Bacterianas , Interacciones Huésped-Patógeno , Proteoma , Proteómica , Proteínas Bacterianas/metabolismo , Línea Celular , Cromatografía Liquida , Humanos , Espectrometría de Masas , Microscopía Fluorescente , Péptidos , Proteómica/métodos , Flujo de Trabajo
8.
Sci Rep ; 7(1): 9718, 2017 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-28887440

RESUMEN

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.


Asunto(s)
Proteínas Bacterianas/metabolismo , Interacciones Huésped-Patógeno , Proteoma , Proteómica , Infecciones Estafilocócicas/metabolismo , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/metabolismo , Animales , Biología Computacional/métodos , Células Epiteliales/metabolismo , Células Epiteliales/microbiología , Humanos , Iones/metabolismo , Ratones , Péptidos , Proteómica/métodos , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/microbiología
9.
Data Brief ; 7: 1031-1037, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27761493

RESUMEN

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.

10.
J Proteomics ; 128: 203-17, 2015 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-26244908

RESUMEN

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.


Asunto(s)
Células Epiteliales/metabolismo , Células Epiteliales/microbiología , Neumonía Estafilocócica/metabolismo , Proteoma/metabolismo , Mucosa Respiratoria/metabolismo , Staphylococcus aureus/metabolismo , Línea Celular , Citocinas/metabolismo , Interacciones Huésped-Patógeno , Humanos , Neumonía Estafilocócica/microbiología , Mucosa Respiratoria/microbiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA