Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
Add more filters










Publication year range
1.
Nat Commun ; 14(1): 6693, 2023 10 23.
Article in English | MEDLINE | ID: mdl-37872209

ABSTRACT

Group A streptococcus (GAS) is a major bacterial pathogen responsible for both local and systemic infections in humans. The molecular mechanisms that contribute to disease heterogeneity remain poorly understood. Here we show that the transition from a local to a systemic GAS infection is paralleled by pathogen-driven alterations in IgG homeostasis. Using animal models and a combination of sensitive proteomics and glycoproteomics readouts, we documented the progressive accumulation of IgG cleavage products in plasma, due to extensive enzymatic degradation triggered by GAS infection in vivo. The level of IgG degradation was modulated by the route of pathogen inoculation, and mechanistically linked to the combined activities of the bacterial protease IdeS and the endoglycosidase EndoS, upregulated during infection. Importantly, we show that these virulence factors can alter the structure and function of exogenous therapeutic IgG in vivo. These results shed light on the role of bacterial virulence factors in shaping GAS pathogenesis, and potentially blunting the efficacy of antimicrobial therapies.


Subject(s)
Bacterial Proteins , Streptococcal Infections , Humans , Animals , Bacterial Proteins/metabolism , Immunoglobulin G , Streptococcal Infections/microbiology , Streptococcus pyogenes , Virulence Factors/metabolism
2.
Nat Commun ; 14(1): 5359, 2023 09 02.
Article in English | MEDLINE | ID: mdl-37660105

ABSTRACT

The incorporation of machine learning methods into proteomics workflows improves the identification of disease-relevant biomarkers and biological pathways. However, machine learning models, such as deep neural networks, typically suffer from lack of interpretability. Here, we present a deep learning approach to combine biological pathway analysis and biomarker identification to increase the interpretability of proteomics experiments. Our approach integrates a priori knowledge of the relationships between proteins and biological pathways and biological processes into sparse neural networks to create biologically informed neural networks. We employ these networks to differentiate between clinical subphenotypes of septic acute kidney injury and COVID-19, as well as acute respiratory distress syndrome of different aetiologies. To gain biological insight into the complex syndromes, we utilize feature attribution-methods to introspect the networks for the identification of proteins and pathways important for distinguishing between subtypes. The algorithms are implemented in a freely available open source Python-package ( https://github.com/InfectionMedicineProteomics/BINN ).


Subject(s)
Acute Kidney Injury , COVID-19 , Humans , Proteomics , Neural Networks, Computer , Algorithms
3.
Nat Commun ; 14(1): 3603, 2023 06 17.
Article in English | MEDLINE | ID: mdl-37330510

ABSTRACT

Sepsis is the major cause of mortality across intensive care units globally, yet details of accompanying pathological molecular events remain unclear. This knowledge gap has resulted in ineffective biomarker development and suboptimal treatment regimens to prevent and manage organ dysfunction/damage. Here, we used pharmacoproteomics to score time-dependent treatment impact in a murine Escherichia coli sepsis model after administering beta-lactam antibiotic meropenem (Mem) and/or the immunomodulatory glucocorticoid methylprednisolone (Gcc). Three distinct proteome response patterns were identified, which depended on the underlying proteotype for each organ. Gcc enhanced some positive proteome responses of Mem, including superior reduction of the inflammatory response in kidneys and partial restoration of sepsis-induced metabolic dysfunction. Mem introduced sepsis-independent perturbations in the mitochondrial proteome that Gcc counteracted. We provide a strategy for the quantitative and organotypic assessment of treatment effects of candidate therapies in relationship to dosing, timing, and potential synergistic intervention combinations during sepsis.


Subject(s)
Bacteremia , Gram-Negative Bacterial Infections , Sepsis , Mice , Animals , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Proteome , Meropenem/pharmacology , Meropenem/therapeutic use , Sepsis/drug therapy , Sepsis/complications , Gram-Negative Bacterial Infections/drug therapy , Bacteremia/drug therapy
4.
Commun Biol ; 6(1): 628, 2023 06 10.
Article in English | MEDLINE | ID: mdl-37301900

ABSTRACT

Data independent acquisition mass spectrometry (DIA-MS) has recently emerged as an important method for the identification of blood-based biomarkers. However, the large search space required to identify novel biomarkers from the plasma proteome can introduce a high rate of false positives that compromise the accuracy of false discovery rates (FDR) using existing validation methods. We developed a generalized precursor scoring (GPS) method trained on 2.75 million precursors that can confidently control FDR while increasing the number of identified proteins in DIA-MS independent of the search space. We demonstrate how GPS can generalize to new data, increase protein identification rates, and increase the overall quantitative accuracy. Finally, we apply GPS to the identification of blood-based biomarkers and identify a panel of proteins that are highly accurate in discriminating between subphenotypes of septic acute kidney injury from undepleted plasma to showcase the utility of GPS in discovery DIA-MS proteomics.


Subject(s)
Proteomics , Tandem Mass Spectrometry , Proteomics/methods , Tandem Mass Spectrometry/methods , Biomarkers , Proteome/analysis
5.
Sci Rep ; 13(1): 8801, 2023 05 31.
Article in English | MEDLINE | ID: mdl-37258541

ABSTRACT

Alveolar epithelial cells (AEC) have been implicated in pathological remodelling. We examined the capacity of AEC to produce extracellular matrix (ECM) and thereby directly contribute towards remodelling in chronic lung diseases. Cryopreserved type 2 AEC (AEC2) from healthy lungs and chronic obstructive pulmonary disease (COPD) afflicted lungs were cultured in decellularized healthy human lung slices for 13 days. Healthy-derived AEC2 were treated with transforming growth factor ß1 (TGF-ß1) to evaluate the plasticity of their ECM production. Evaluation of phenotypic markers and expression of matrisome genes and proteins were evaluated by RNA-sequencing, mass spectrometry and immunohistochemistry. The AEC2 displayed an AEC marker profile similar to freshly isolated AEC2 throughout the 13-day culture period. COPD-derived AECs proliferated as healthy AECs with few differences in gene and protein expression while retaining increased expression of disease marker HLA-A. The AEC2 expressed basement membrane components and a complex set of interstitial ECM proteins. TGF-ß1 stimuli induced a significant change in interstitial ECM production from AEC2 without loss of specific AEC marker expression. This study reveals a previously unexplored potential of AEC to directly contribute to ECM turnover by producing interstitial ECM proteins, motivating a re-evaluation of the role of AEC2 in pathological lung remodelling.


Subject(s)
Alveolar Epithelial Cells , Pulmonary Disease, Chronic Obstructive , Humans , Transforming Growth Factor beta1/metabolism , Lung/pathology , Extracellular Matrix/metabolism , Extracellular Matrix Proteins/metabolism , Pulmonary Disease, Chronic Obstructive/pathology , Epithelial Cells/metabolism
6.
mSystems ; 7(4): e0039522, 2022 08 30.
Article in English | MEDLINE | ID: mdl-35913192

ABSTRACT

Vascular dysfunction and organ failure are two distinct, albeit highly interconnected, clinical outcomes linked to morbidity and mortality in human sepsis. The mechanisms driving vascular and parenchymal damage are dynamic and display significant molecular cross talk between organs and tissues. Therefore, assessing their individual contribution to disease progression is technically challenging. Here, we hypothesize that dysregulated vascular responses predispose the organism to organ failure. To address this hypothesis, we have evaluated four major organs in a murine model of Staphylococcus aureus sepsis by combining in vivo labeling of the endothelial cell surface proteome, data-independent acquisition (DIA) mass spectrometry, and an integrative computational pipeline. The data reveal, with unprecedented depth and throughput, that a septic insult evokes organ-specific proteome responses that are highly compartmentalized, synchronously coordinated, and significantly correlated with the progression of the disease. These responses include abundant vascular shedding, dysregulation of the intrinsic pathway of coagulation, compartmentalization of the acute phase response, and abundant upregulation of glycocalyx components. Vascular cell surface proteome changes were also found to precede bacterial invasion and leukocyte infiltration into the organs, as well as to precede changes in various well-established cellular and biochemical correlates of systemic coagulopathy and tissue dysfunction. Importantly, our data suggest a potential role for the vascular proteome as a determinant of the susceptibility of the organs to undergo failure during sepsis. IMPORTANCE Sepsis is a life-threatening response to infection that results in immune dysregulation, vascular dysfunction, and organ failure. New methods are needed for the identification of diagnostic and therapeutic targets. Here, we took a systems-wide approach using data-independent acquisition (DIA) mass spectrometry to track the progression of bacterial sepsis in the vasculature leading to organ failure. Using a murine model of S. aureus sepsis, we were able to quantify thousands of proteins across the plasma and parenchymal and vascular compartments of multiple organs in a time-resolved fashion. We showcase the profound proteome remodeling triggered by sepsis over time and across these compartments. Importantly, many vascular proteome alterations precede changes in traditional correlates of organ dysfunction, opening a molecular window for the discovery of early markers of sepsis progression.


Subject(s)
Bacteremia , Sepsis , Mice , Humans , Animals , Staphylococcus aureus , Proteome , Multiple Organ Failure/metabolism , Disease Models, Animal
7.
J Proteome Res ; 21(2): 535-546, 2022 02 04.
Article in English | MEDLINE | ID: mdl-35042333

ABSTRACT

Data-independent acquisition-mass spectrometry (DIA-MS) is the method of choice for deep, consistent, and accurate single-shot profiling in bottom-up proteomics. While classic workflows for targeted quantification from DIA-MS data require auxiliary data-dependent acquisition (DDA) MS analysis of subject samples to derive prior-knowledge spectral libraries, library-free approaches based on in silico prediction promise deep DIA-MS profiling with reduced experimental effort and cost. Coverage and sensitivity in such analyses are however limited, in part, by the large library size and persistent deviations from the experimental data. We present MSLibrarian, a new workflow and tool to obtain optimized predicted spectral libraries by the integrated usage of spectrum-centric DIA data interpretation via the DIA-Umpire approach to inform and calibrate the in silico predicted library and analysis approach. Predicted-vs-observed comparisons enabled optimization of intensity prediction parameters, calibration of retention time prediction for deviating chromatographic setups, and optimization of the library scope and sample representativeness. Benchmarking via a dedicated ground-truth-embedded experiment of species-mixed proteins and quantitative ratio-validation confirmed gains of up to 13% on peptide and 8% on protein level at equivalent FDR control and validation criteria. MSLibrarian is made available as an open-source R software package, including step-by-step user instructions, at https://github.com/MarcIsak/MSLibrarian.


Subject(s)
Peptides , Proteomics , Mass Spectrometry/methods , Peptides/analysis , Proteins , Proteome/analysis , Proteomics/methods , Software
8.
mBio ; 12(5): e0118121, 2021 10 26.
Article in English | MEDLINE | ID: mdl-34544271

ABSTRACT

Hepatic failure is an important risk factor for poor outcome in septic patients. Using a chemical tagging workflow and high-resolution mass spectrometry, we demonstrate that rapid proteome remodeling of the vascular surfaces precedes hepatic damage in a murine model of Staphylococcus aureus sepsis. These early changes include vascular deposition of neutrophil-derived proteins, shedding of vascular receptors, and altered levels of heparin/heparan sulfate-binding factors. Modification of endothelial heparan sulfate, a major component of the vascular glycocalyx, diminishes neutrophil trafficking to the liver and reduces hepatic coagulopathy and organ damage during the systemic inflammatory response to infection. Modifying endothelial heparan sulfate likewise reduces neutrophil trafficking in sterile hepatic injury, reflecting a more general role of heparan sulfate contribution to the modulation of leukocyte behavior during inflammation. IMPORTANCE Vascular glycocalyx remodeling is critical to sepsis pathology, but the glycocalyx components that contribute to this process remain poorly characterized. This article shows that during Staphylococcus aureus sepsis, the liver vascular glycocalyx undergoes dramatic changes in protein composition associated with neutrophilic activity and heparin/heparan sulfate binding, all before organ damage is detectable by standard circulating liver damage markers or histology. Targeted manipulation of endothelial heparan sulfate modulates S. aureus sepsis-induced hepatotoxicity by controlling the magnitude of neutrophilic infiltration into the liver in both nonsterile and sterile injury. These data identify an important vascular glycocalyx component that impacts hepatic failure during nonsterile and sterile injury.


Subject(s)
Endothelial Cells/metabolism , Heparitin Sulfate/genetics , Heparitin Sulfate/metabolism , Neutrophil Activation , Neutrophils/pathology , Sepsis/microbiology , Staphylococcus aureus/immunology , Animals , Disease Models, Animal , Endothelial Cells/immunology , Female , Glycocalyx/metabolism , Glycocalyx/pathology , Liver/immunology , Liver/microbiology , Liver/pathology , Lung/immunology , Lung/microbiology , Lung/pathology , Male , Mice , Mice, Inbred C57BL , Staphylococcus aureus/pathogenicity
9.
Mol Cell Proteomics ; 20: 100113, 2021.
Article in English | MEDLINE | ID: mdl-34139362

ABSTRACT

Severe coronavirus disease 2019 (COVID-19) can result in pneumonia and acute respiratory failure. Accumulation of mucus in the airways is a hallmark of the disease and can result in hypoxemia. Here, we show that quantitative proteome analysis of the sputum from severe patients with COVID-19 reveal high levels of neutrophil extracellular trap (NET) components, which was confirmed by microscopy. Extracellular DNA from excessive NET formation can increase sputum viscosity and lead to acute respiratory distress syndrome. Recombinant human DNase (Pulmozyme; Roche) has been shown to be beneficial in reducing sputum viscosity and improve lung function. We treated five patients pwith COVID-19 resenting acute symptoms with clinically approved aerosolized Pulmozyme. No adverse reactions to the drug were seen, and improved oxygen saturation and recovery in all severely ill patients with COVID-19 was observed after therapy. Immunofluorescence and proteome analysis of sputum and blood plasma samples after treatment revealed a marked reduction of NETs and a set of statistically significant proteome changes that indicate reduction of hemorrhage, plasma leakage and inflammation in the airways, and reduced systemic inflammatory state in the blood plasma of patients. Taken together, the results indicate that NETs contribute to acute respiratory failure in COVID-19 and that degrading NETs may reduce dependency on external high-flow oxygen therapy in patients. Targeting NETs using recombinant human DNase may have significant therapeutic implications in COVID-19 disease and warrants further studies.


Subject(s)
COVID-19 Drug Treatment , Deoxyribonuclease I/pharmacology , Extracellular Traps/metabolism , Proteome/analysis , Aged , Blood Proteins/analysis , COVID-19/metabolism , COVID-19/therapy , Female , Fluorescent Antibody Technique , Humans , Male , Middle Aged , Recombinant Proteins/pharmacology , Severity of Illness Index , Sputum/drug effects , Sputum/metabolism , Sputum/virology , Systemic Inflammatory Response Syndrome/drug therapy , Systemic Inflammatory Response Syndrome/metabolism , Systemic Inflammatory Response Syndrome/virology
10.
iScience ; 24(4): 102339, 2021 Apr 23.
Article in English | MEDLINE | ID: mdl-33855284

ABSTRACT

Streptococcus pyogenes is a major bacterial pathogen in the human population and isolates of the clinically important M1 serotype secrete protein Streptococcal inhibitor of complement (SIC) known to interfere with human innate immunity. Here we find that SIC from M1 bacteria interacts with TLR2 and CD14 on monocytes leading to the activation of the NF-κB and p38 MAPK pathways and the release of several pro-inflammatory cytokines (e.g. TNFα and INFγ). In human plasma, SIC binds clusterin and histidine-rich glycoprotein, and whole plasma, and these two purified plasma proteins enhanced the activation of monocytes by SIC. Isolates of the M55 serotype secrete an SIC homolog, but this protein did not activate monocytes. M1 isolates are common in cases of invasive S. pyogenes infections characterized by massive inflammation, and the results of this study indicate that the pro-inflammatory property of SIC contributes to the pathology of these severe clinical conditions.

11.
J Cyst Fibros ; 20(2): 346-355, 2021 03.
Article in English | MEDLINE | ID: mdl-32727663

ABSTRACT

BACKGROUND: Cystic fibrosis (CF), involves excessive airway accumulation of neutrophils, often in parallel with severe infection caused by Pseudomonas aeruginosa. Free histones are known to possess bactericidal properties, but the degree of antibacterial activity exerted on specific lung-based pathogens is largely unknown. Neutrophils have a high content of peptidyl deiminase 4 (PADI4), which citrullinate cationic peptidyl-arginines. In histone H3.1, several positions in the NH2-terminal tail are subject to citrullination. METHODS: Full-length and segmented histone subunit H3.1 was investigated for bactericidal activity towards P. aeruginosa (strain PAO1). PADI4-induced citrullination of histone H3.1 was assessed for antibacterial activity towards P. aeruginosa. Next, the effect of neutrophil elastase (NE)-mediated proteolysis of histone H3.1 was investigated. Finally, PADI4, H3.1, and citrullinated H3.1 were examined in healthy control and CF patient lung tissues. RESULTS: Full-length histone H3.1 and sections of the histone H3.1 tail, displayed bactericidal activity towards P. aeruginosa. These antibacterial effects were reduced following citrullination by PADI4 or proteolysis by NE. Interestingly, citrullination of histone H3.1 exacerbated NE-mediated degradation. In CF lung tissue, citrullinated histone H3.1 and PADI4 immunoreactivity was abundant. Degraded histone H3.1 was detected in the sputum of CF patients but was absent in the sputum of healthy controls. CONCLUSIONS: Citrullination impairs the antibacterial activity of histone H3.1 and exacerbates its proteolytic degradation by NE. Citrullination is likely to play an important role during resolution of acute inflammation. However, in chronic inflammation akin to CF, citrullination may dampen host defense and promote pathogen survival, as exemplified by P. aeruginosa.


Subject(s)
Citrullination , Cystic Fibrosis/metabolism , Histones/metabolism , Histones/pharmacology , Protein-Arginine Deiminase Type 4/metabolism , Pseudomonas Infections/drug therapy , Case-Control Studies , Humans , Leukocyte Elastase/metabolism , Proteolysis , Pseudomonas aeruginosa/drug effects
12.
J Proteomics ; 227: 103904, 2020 09 15.
Article in English | MEDLINE | ID: mdl-32702520

ABSTRACT

Serum amyloid A (SAA) is a well-described acute phase protein induced during the acute phase response (APR) to infection. Four isoform specific genes are found in most mammals. Depending on species, SAA3 and SAA4 are generally preferentially expressed extrahepatically whereas SAA1 and SAA2 are hepatic isoforms dominating the SAA serum pool. Little is known about how specific infections affect the serum SAA isoform profile, as SAA isoform discriminating antibodies are not generally available. An antibody independent, quantitative targeted MS method (Selected Reaction Monitoring, SRM) based on available information on porcine SAA isoform genes was developed and used to profile SAA in serum samples from pigs experimentally infected with Staphylococcus aureus (Sa). While results suggest SAA2 as the main circulating porcine SAA isoform, induced around 10 times compared to non-infected controls, total SAA serum concentrations reached only around 4 µg/mL, much lower than established previously by immunoassays. This might suggest that SAA isoform variants not detected by the SRM method might be present in porcine serum. The assay allows monitoring host responses to experimental infections, infectious diseases and inflammation states in the pig at an unprecedented level of detail. It can also be used in a non-calibrated (relative quantification) format. SIGNIFICANCE: We developed an SRM MS method which for the first time allowed the specific quantification of each of the circulating porcine SAA isoforms (SAA2, SAA3, SAA4). It was found that SAA2 is the dominating circulating isoform of SAA in the pig and that, during the acute phase response to Sa infection SAA2, SAA3 and SAA4 are induced approx. 10, 15 and 2 times, respectively. Absolute levels of the isoforms as determined by SRM MS were much lower than reported previously for total SAA quantified by immunosassays, suggesting the existence of hitherto non-described SAA variants. SRM MS holds great promise for the study of the basic biology of SAA isoforms with the potential to study an even broader range of SAA variants.


Subject(s)
Serum Amyloid A Protein , Staphylococcus aureus , Acute-Phase Reaction , Animals , Mass Spectrometry , Protein Isoforms , Serum Amyloid A Protein/analysis , Swine
13.
Microb Biotechnol ; 12(5): 892-906, 2019 09.
Article in English | MEDLINE | ID: mdl-31270938

ABSTRACT

Organic pollutants (OPs) are critically toxic, bioaccumulative and globally widespread. Moreover, several OPs negatively influence aquatic wildlife. Although bacteria are major drivers of the ocean carbon cycle and the turnover of vital elements, there is limited knowledge of OP effects on heterotrophic bacterioplankton. We therefore investigated growth and gene expression responses of the Baltic Sea model bacterium Rheinheimera sp. BAL341 to environmentally relevant concentrations of distinct classes of OPs in 2-h incubation experiments. During exponential growth, exposure to a mix of polycyclic aromatic hydrocarbons, alkanes and organophosphate esters (denoted MIX) resulted in a significant decrease (between 9% and 18%) in bacterial abundance and production compared with controls. In contrast, combined exposure to perfluorooctanesulfonic acids and perfluorooctanoic acids (denoted PFAS) had no significant effect on growth. Nevertheless, MIX and PFAS exposures both induced significant shifts in gene expression profiles compared with controls in exponential growth. This involved several functional metabolism categories (e.g. stress response and fatty acids metabolism), some of which were pollutant-specific (e.g. phosphate acquisition and alkane-1 monooxygenase genes). In stationary phase, only two genes in the MIX treatment were significantly differentially expressed. The substantial direct influence of OPs on metabolism during bacterial growth suggests that widespread OPs could severely alter biogeochemical processes governed by bacterioplankton.


Subject(s)
Aquatic Organisms/drug effects , Aquatic Organisms/growth & development , Chromatiaceae/drug effects , Chromatiaceae/growth & development , Gene Expression/drug effects , Organic Chemicals/toxicity , Water Pollutants, Chemical/toxicity , Aquatic Organisms/genetics , Bacterial Load , Chromatiaceae/genetics , Gene Expression Profiling , Metabolic Networks and Pathways/genetics , Seawater/microbiology
14.
Int J Med Microbiol ; 309(7): 151325, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31257068

ABSTRACT

Aerococcus urinae is an emerging pathogen that causes urinary tract infections, bacteremia and infective endocarditis. The mechanisms through which A. urinae cause infection are largely unknown. The aims of this study were to describe the surface proteome of A. urinae and to analyse A. urinae genomes in search for genes encoding surface proteins. Two proteins, denoted Aerococcal surface protein (Asp) 1 and 2, were through the use of mass spectrometry based proteomics found to quantitatively dominate the aerococcal surface. The presence of these proteins on the surface was also shown using ELISA with serum from rabbits immunized with the recombinant Asp. These proteins had a signal sequence in the amino-terminal end and a cell wall-sorting region in the carboxy-terminal end, which contained an LPATG-motif, a hydrophobic domain and a positively charged tail. Twenty-three additional A. urinae genomes were sequenced using Illumina HiSeq technology. Six different variants of asp genes were found (denoted asp1-6). All isolates had either one or two of these asp-genes located in a conserved locus, designated Locus encoding Aerococcal Surface Proteins (LASP). The 25 genomes had in median 13 genes encoding LPXTG-proteins (range 6-24). For other Gram-positive bacteria, cell wall-anchored surface proteins with an LPXTG-motif play a key role for virulence. Thus, it will be of great interest to explore the function of the Asp proteins of A. urinae to establish a better understanding of the molecular mechanisms by which A. urinae cause disease.


Subject(s)
Aerococcus/chemistry , Bacterial Proteins/metabolism , Cell Wall/chemistry , Membrane Proteins/metabolism , Aerococcus/genetics , Aerococcus/metabolism , Aerococcus/pathogenicity , Amino Acid Motifs , Amino Acid Sequence , Animals , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Base Sequence , Cell Wall/metabolism , Enzyme-Linked Immunosorbent Assay , Genome, Bacterial/genetics , Membrane Proteins/chemistry , Membrane Proteins/genetics , Protein Sorting Signals , Proteome , Rabbits , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Virulence/genetics
15.
Nat Commun ; 10(1): 2727, 2019 06 21.
Article in English | MEDLINE | ID: mdl-31227708

ABSTRACT

A fundamental challenge in medical microbiology is to characterize the dynamic protein-protein interaction networks formed at the host-pathogen interface. Here, we generate a quantitative interaction map between the significant human pathogen, Streptococcus pyogenes, and proteins from human saliva and plasma obtained via complementary affinity-purification and bacterial-surface centered enrichment strategies and quantitative mass spectrometry. Perturbation of the network using immunoglobulin protease cleavage, mixtures of different concentrations of saliva and plasma, and different S. pyogenes serotypes and their isogenic mutants, reveals how changing microenvironments alter the interconnectivity of the interaction map. The importance of host immunoglobulins for the interaction with human complement proteins is demonstrated and potential protective epitopes of importance for phagocytosis of S. pyogenes cells are localized. The interaction map confirms several previously described protein-protein interactions; however, it also reveals a multitude of additional interactions, with possible implications for host-pathogen interactions involving other bacterial species.


Subject(s)
Antibodies, Bacterial/metabolism , Bacterial Proteins/metabolism , Host-Pathogen Interactions/immunology , Streptococcal Infections/immunology , Streptococcus pyogenes/immunology , Antibodies, Bacterial/immunology , Antigens, Bacterial/immunology , Antigens, Bacterial/metabolism , Bacterial Proteins/immunology , Chromatography, Affinity , Complement System Proteins/immunology , Complement System Proteins/metabolism , Epitope Mapping , Healthy Volunteers , Humans , Mass Spectrometry , Opsonin Proteins/immunology , Opsonin Proteins/metabolism , Protein Binding , Protein Interaction Mapping , Protein Interaction Maps/immunology , Streptococcal Infections/blood , Streptococcal Infections/microbiology , Streptococcus pyogenes/metabolism
16.
J Exp Med ; 216(7): 1615-1629, 2019 07 01.
Article in English | MEDLINE | ID: mdl-31092533

ABSTRACT

Streptococcus pyogenes (Group A streptococcus; GAS) is a human pathogen causing diseases from uncomplicated tonsillitis to life-threatening invasive infections. GAS secretes EndoS, an endoglycosidase that specifically cleaves the conserved N-glycan on IgG antibodies. In vitro, removal of this glycan impairs IgG effector functions, but its relevance to GAS infection in vivo is unclear. Using targeted mass spectrometry, we characterized the effects of EndoS on host IgG glycosylation during the course of infections in humans. Substantial IgG glycan hydrolysis occurred at the site of infection and systemically in the severe cases. We demonstrated decreased resistance to phagocytic killing of GAS lacking EndoS in vitro and decreased virulence in a mouse model of invasive infection. This is the first described example of specific bacterial IgG glycan hydrolysis during infection and thereby verifies the hypothesis that EndoS modifies antibodies in vivo. This mechanisms of immune evasion could have implications for treatment of severe GAS infections and for future efforts at vaccine development.


Subject(s)
Adaptive Immunity/immunology , Bacterial Proteins/metabolism , Glycoside Hydrolases/metabolism , Immunoglobulin G/immunology , Streptococcal Infections/immunology , Streptococcus pyogenes/immunology , Animals , Disease Models, Animal , Electrophoresis, Polyacrylamide Gel , Female , Glycosylation , Humans , Hydrolysis , Immunoglobulin G/metabolism , Immunoglobulin G/pharmacology , Limit of Detection , Mass Spectrometry , Mice , Mice, Inbred C57BL , Streptococcal Infections/microbiology , Streptococcus pyogenes/enzymology , Tonsillitis/immunology , Tonsillitis/microbiology
17.
Nat Commun ; 10(1): 1667, 2019 04 10.
Article in English | MEDLINE | ID: mdl-30971685

ABSTRACT

Neutrophils are crucial mediators of host defense that are recruited to the central nervous system (CNS) in large numbers during acute bacterial meningitis caused by Streptococcus pneumoniae. Neutrophils release neutrophil extracellular traps (NETs) during infections to trap and kill bacteria. Intact NETs are fibrous structures composed of decondensed DNA and neutrophil-derived antimicrobial proteins. Here we show NETs in the cerebrospinal fluid (CSF) of patients with pneumococcal meningitis, and their absence in other forms of meningitis with neutrophil influx into the CSF caused by viruses, Borrelia and subarachnoid hemorrhage. In a rat model of meningitis, a clinical strain of pneumococci induced NET formation in the CSF. Disrupting NETs using DNase I significantly reduces bacterial load, demonstrating that NETs contribute to pneumococcal meningitis pathogenesis in vivo. We conclude that NETs in the CNS reduce bacterial clearance and degrading NETs using DNase I may have significant therapeutic implications.


Subject(s)
Cerebrospinal Fluid/cytology , Extracellular Traps/microbiology , Immune Evasion , Meningitis, Pneumococcal/immunology , Neutrophils/immunology , Streptococcus pneumoniae/immunology , Adolescent , Adult , Aged , Animals , Borrelia burgdorferi Group/immunology , Brain/cytology , Brain/drug effects , Brain/immunology , Brain/microbiology , Cerebrospinal Fluid/immunology , Cerebrospinal Fluid/microbiology , Deoxyribonuclease I/administration & dosage , Disease Models, Animal , Extracellular Traps/drug effects , Extracellular Traps/immunology , Female , Humans , Lyme Neuroborreliosis/cerebrospinal fluid , Lyme Neuroborreliosis/immunology , Lyme Neuroborreliosis/microbiology , Male , Meningitis, Pneumococcal/cerebrospinal fluid , Meningitis, Pneumococcal/drug therapy , Meningitis, Pneumococcal/microbiology , Meningitis, Viral/cerebrospinal fluid , Meningitis, Viral/immunology , Middle Aged , Neutrophils/microbiology , Rats , Rats, Sprague-Dawley , Recombinant Proteins/administration & dosage , Spinal Puncture , Streptococcus pneumoniae/isolation & purification , Subarachnoid Hemorrhage/cerebrospinal fluid , Young Adult
18.
Microbiome ; 6(1): 173, 2018 09 28.
Article in English | MEDLINE | ID: mdl-30266101

ABSTRACT

BACKGROUND: Prokaryotes dominate the biosphere and regulate biogeochemical processes essential to all life. Yet, our knowledge about their biology is for the most part limited to the minority that has been successfully cultured. Molecular techniques now allow for obtaining genome sequences of uncultivated prokaryotic taxa, facilitating in-depth analyses that may ultimately improve our understanding of these key organisms. RESULTS: We compared results from two culture-independent strategies for recovering bacterial genomes: single-amplified genomes and metagenome-assembled genomes. Single-amplified genomes were obtained from samples collected at an offshore station in the Baltic Sea Proper and compared to previously obtained metagenome-assembled genomes from a time series at the same station. Among 16 single-amplified genomes analyzed, seven were found to match metagenome-assembled genomes, affiliated with a diverse set of taxa. Notably, genome pairs between the two approaches were nearly identical (average 99.51% sequence identity; range 98.77-99.84%) across overlapping regions (30-80% of each genome). Within matching pairs, the single-amplified genomes were consistently smaller and less complete, whereas the genetic functional profiles were maintained. For the metagenome-assembled genomes, only on average 3.6% of the bases were estimated to be missing from the genomes due to wrongly binned contigs. CONCLUSIONS: The strong agreement between the single-amplified and metagenome-assembled genomes emphasizes that both methods generate accurate genome information from uncultivated bacteria. Importantly, this implies that the research questions and the available resources are allowed to determine the selection of genomics approach for microbiome studies.


Subject(s)
Bacteria/genetics , Genome, Bacterial/genetics , Metagenome/genetics , Microbiota/genetics , Nucleic Acid Amplification Techniques , Whole Genome Sequencing/methods , Bacteria/classification , High-Throughput Nucleotide Sequencing , Oceans and Seas , RNA, Ribosomal, 16S/genetics , Sequence Alignment , Sweden
19.
J Biol Chem ; 293(35): 13578-13591, 2018 08 31.
Article in English | MEDLINE | ID: mdl-30002122

ABSTRACT

Some strains of the bacterial pathogen Streptococcus pyogenes secrete protein SIC (streptococcal inhibitor of complement), including strains of the clinically relevant M1 serotype. SIC neutralizes the effect of a number of antimicrobial proteins/peptides and interferes with the function of the host complement system. Previous studies have shown that some S. pyogenes proteins bind and modulate coagulation and fibrinolysis factors, raising the possibility that SIC also may interfere with the activity of these factors. Here we show that SIC interacts with both human thrombin and plasminogen, key components of coagulation and fibrinolysis. We found that during clot formation, SIC binds fibrin through its central region and that SIC inhibits fibrinolysis by interacting with plasminogen. Flow cytometry results indicated that SIC and plasminogen bind simultaneously to S. pyogenes bacteria, and fluorescence microscopy revealed co-localization of the two proteins at the bacterial surface. As a consequence, SIC-expressing bacteria entrapped in clots inhibit fibrinolysis, leading to delayed bacterial escape from the clots as compared with mutant bacteria lacking SIC. Moreover, within the clots SIC-expressing bacteria were protected against killing. In an animal model of subcutaneous infection, SIC-expressing bacteria exhibited a delayed systemic spread. These results demonstrate that the bacterial protein SIC interferes with coagulation and fibrinolysis and thereby enhances bacterial survival, a finding that has significant implications for S. pyogenes virulence.


Subject(s)
Bacterial Proteins/immunology , Fibrinolysis , Streptococcal Infections/immunology , Streptococcus pyogenes/immunology , Thrombosis/immunology , Animals , Complement System Proteins/immunology , Female , Fibrin/immunology , Fibrinogen/immunology , Humans , Immunity, Innate , Mice , Mice, Inbred C57BL , Streptococcal Infections/complications , Streptococcal Infections/microbiology , Thrombin/immunology , Thrombosis/complications , Thrombosis/microbiology
20.
Environ Microbiol Rep ; 10(4): 493-500, 2018 08.
Article in English | MEDLINE | ID: mdl-29733107

ABSTRACT

Disturbances are believed to be one of the main factors influencing variations in community diversity and functioning. Here we investigated if exposure to a pH press disturbance affected the composition and functional performance of a bacterial community and its resistance, recovery and resilience to a second press disturbance (salt addition). Lake bacterial assemblages were initially exposed to reduced pH in six mesocosms whereas another six mesocosms were kept as reference. Seven days after the pH disturbance, three tanks from each treatment were exposed to a salt disturbance. Both bacterial production and enzyme activity were negatively affected by the salt treatment, regardless if the communities had been subject to a previous disturbance or not. However, cell-specific enzyme activity had a higher resistance in communities pre-exposed to the pH disturbance compared to the reference treatment. In contrast, for cell-specific bacterial production resistance was not affected, but recovery was faster in the communities that had previously been exposed to the pH disturbance. Over time, bacterial community composition diverged among treatments, in response to both pH and salinity. The difference in functional recovery, resilience and resistance may depend on differences in community composition caused by the pH disturbance, niche breadth or acquired stress resistance.


Subject(s)
Bacterial Physiological Phenomena , Plankton/physiology , Stress, Physiological/physiology , Water Microbiology , Bacteria/classification , Bacteria/growth & development , Bacteria/metabolism , Carbon/metabolism , Cellulose 1,4-beta-Cellobiosidase/metabolism , Hydrogen-Ion Concentration , Lakes/microbiology , Plankton/classification , Plankton/growth & development , Plankton/metabolism , RNA, Ribosomal, 16S/genetics , Salinity , beta-Glucosidase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...