Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 190
Filtrar
1.
PLoS Pathog ; 20(6): e1011883, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38838057

RESUMO

ATP-binding cassette (ABC) transport systems are crucial for bacteria to ensure sufficient uptake of nutrients that are not produced de novo or improve the energy balance. The cell surface of the pathobiont Streptococcus pneumoniae (pneumococcus) is decorated with a substantial array of ABC transporters, critically influencing nasopharyngeal colonization and invasive infections. Given the auxotrophic nature of pneumococci for certain amino acids, the Ami ABC transporter system, orchestrating oligopeptide uptake, becomes indispensable in host compartments lacking amino acids. The system comprises five exposed Oligopeptide Binding Proteins (OBPs) and four proteins building the ABC transporter channel. Here, we present a structural analysis of all the OBPs in this system. Multiple crystallographic structures, capturing both open and closed conformations along with complexes involving chemically synthesized peptides, have been solved at high resolution providing insights into the molecular basis of their diverse peptide specificities. Mass spectrometry analysis of oligopeptides demonstrates the unexpected remarkable promiscuity of some of these proteins when expressed in Escherichia coli, displaying affinity for a wide range of peptides. Finally, a model is proposed for the complete Ami transport system in complex with its various OBPs. We further disclosed, through in silico modelling, some essential structural changes facilitating oligopeptide transport into the cellular cytoplasm. Thus, the structural analysis of the Ami system provides valuable insights into the mechanism and specificity of oligopeptide binding by the different OBPs, shedding light on the intricacies of the uptake mechanism and the in vivo implications for this human pathogen.


Assuntos
Transportadores de Cassetes de Ligação de ATP , Proteínas de Bactérias , Oligopeptídeos , Streptococcus pneumoniae , Streptococcus pneumoniae/metabolismo , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Oligopeptídeos/metabolismo , Transportadores de Cassetes de Ligação de ATP/metabolismo , Transportadores de Cassetes de Ligação de ATP/química , Cristalografia por Raios X , Modelos Moleculares , Lipoproteínas
2.
PLoS Biol ; 21(1): e3001990, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36716340

RESUMO

Competence development in the human pathogen Streptococcus pneumoniae controls several features such as genetic transformation, biofilm formation, and virulence. Competent bacteria produce so-called "fratricins" such as CbpD that kill noncompetent siblings by cleaving peptidoglycan (PGN). CbpD is a choline-binding protein (CBP) that binds to phosphorylcholine residues found on wall and lipoteichoic acids (WTA and LTA) that together with PGN are major constituents of the pneumococcal cell wall. Competent pneumococci are protected against fratricide by producing the immunity protein ComM. How competence and fratricide contribute to virulence is unknown. Here, using a genome-wide CRISPRi-seq screen, we show that genes involved in teichoic acid (TA) biosynthesis are essential during competence. We demonstrate that LytR is the major enzyme mediating the final step in WTA formation, and that, together with ComM, is essential for immunity against CbpD. Importantly, we show that key virulence factors PspA and PspC become more surface-exposed at midcell during competence, in a CbpD-dependent manner. Together, our work supports a model in which activation of competence is crucial for host adherence by increased surface exposure of its various CBPs.


Assuntos
Streptococcus pneumoniae , Fatores de Virulência , Humanos , Streptococcus pneumoniae/genética , Fatores de Virulência/genética , Fatores de Virulência/metabolismo , N-Acetil-Muramil-L-Alanina Amidase/química , N-Acetil-Muramil-L-Alanina Amidase/genética , N-Acetil-Muramil-L-Alanina Amidase/metabolismo , Colina/metabolismo , Parede Celular/metabolismo , Proteínas de Bactérias/metabolismo
3.
J Infect Dis ; 2024 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-38679601

RESUMO

Streptococcus pneumoniae is a leading cause of morbidity and mortality in children and older adults. Yet knowledge on the development of pneumococcal protein-specific antibody responses throughout life is limited. To investigate this, we measured serum IgG levels to 55 pneumococcal proteins in 11-month old infants (n=73), 24-month old children (n=101), parents (n=99), adults without children <6 years of age (n= 99) and older adults aged >60 years (n=100). Our findings revealed low IgG levels in infancy, with distinct development patterns peaking in adults. A decrease in levels was observed for 27 antigens towards older age. Adults and older adults had increased IgG levels during pneumococcal carriage and at increased exposure risk to S. pneumoniae. Carriage was a stronger predictor than exposure or age for antibody responses. These findings highlight the dynamic nature of naturally acquired humoral immunity to pneumococcal proteins throughout life, offering insights for age-targeted interventions.

4.
Infect Immun ; 91(9): e0015423, 2023 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-37551971

RESUMO

Streptococcus pneumoniae is a Gram-positive opportunistic pathogen that can colonize the upper respiratory tract. It is a leading cause of a wide range of infectious diseases, including community-acquired pneumonia and meningitis. Pneumococcal infections cause 1-2 million deaths per year, most of which occur in developing countries. Here, we focused on three choline-binding proteins (CBPs), i.e., PspC, PspA, and LytA. These pneumococcal proteins have different surface-exposed regions but share related choline-binding anchors. These surface-exposed pneumococcal proteins are in direct contact with host cells and have diverse functions. We explored the role of the three CBPs on adhesion and pathogenicity in a human host by performing relevant imaging and functional analyses, such as electron microscopy, confocal laser scanning microscopy, and functional quantitative assays, targeting biofilm formation and the hemolytic capacity of S. pneumoniae. In vitro biofilm formation assays and electron microscopy experiments were used to examine the ability of knockout mutant strains lacking the lytA, pspC, or pspA genes to adhere to surfaces. We found that LytA plays an important role in robust synthesis of the biofilm matrix. PspA and PspC appeared crucial for the hemolytic effects of S. pneumoniae on human red blood cells. Furthermore, all knockout mutants caused less damage to endothelial cells than wild-type bacteria, highlighting the significance of each CPB for the overall pathogenicity of S. pneumoniae. Hence, in addition to their structural function within the cell wall of S. pneumoniae, each of these three surface-exposed CBPs controls or mediates multiple steps during bacterial pathogenesis.


Assuntos
Infecções Pneumocócicas , Streptococcus pneumoniae , Humanos , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Células Endoteliais/metabolismo , Colina/metabolismo , Proteínas de Bactérias/metabolismo , Infecções Pneumocócicas/microbiologia , Proteínas de Membrana/metabolismo , Eritrócitos
5.
J Neuroinflammation ; 20(1): 267, 2023 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-37978545

RESUMO

BACKGROUND: Brain pericytes participate in the regulation of cerebral blood flow and the maintenance of blood-brain barrier integrity. Because of their perivascular localization, their receptor repertoire, and their potential ability to respond to inflammatory and infectious stimuli by producing various cytokines and chemokines, these cells are also thought to play an active role in the immune response to brain infections. This assumption is mainly supported by in vitro studies, investigations in in vivo disease models are largely missing. Here, we analysed the role of brain pericytes in pneumococcal meningitis, in vitro and in vivo in two animal models of pneumococcal meningitis. METHODS: Primary murine and human pericytes were stimulated with increasing concentrations of different serotypes of Streptococcus pneumoniae in the presence or absence of Toll-like receptor inhibitors and their cell viability and cytokine production were monitored. To gain insight into the role of pericytes in brain infection in vivo, we performed studies in a zebrafish embryo model of pneumococcal meningitis in which pericytes were pharmacologically depleted. Furthermore, we analyzed the impact of genetically induced pericyte ablation on disease progression, intracranial complications, and brain inflammation in an adult mouse model of this disease. RESULTS: Both murine and human pericytes reacted to pneumococcal exposure with the release of selected cytokines. This cytokine release is pneumolysin-dependent, TLR-dependent in murine (but not human) pericytes and can be significantly increased by macrophage-derived IL-1b. Pharmacological depletion of pericytes in zebrafish embryos resulted in increased cerebral edema and mortality due to pneumococcal meningitis. Correspondingly, in an adult mouse meningitis model, a more pronounced blood-brain barrier disruption and leukocyte infiltration, resulting in an unfavorable disease course, was observed following genetic pericyte ablation. The degree of leukocyte infiltration positively correlated with an upregulation of chemokine expression in the brains of pericyte-depleted mice. CONCLUSIONS: Our findings show that pericytes play a protective role in pneumococcal meningitis by impeding leukocyte migration and preventing blood-brain barrier breaching. Thus, preserving the integrity of the pericyte population has the potential as a new therapeutic strategy in pneumococcal meningitis.


Assuntos
Meningite Pneumocócica , Humanos , Animais , Camundongos , Barreira Hematoencefálica/metabolismo , Peixe-Zebra/metabolismo , Pericitos/metabolismo , Streptococcus pneumoniae , Citocinas/metabolismo , Quimiocinas/metabolismo , Leucócitos/metabolismo
6.
Metabolomics ; 18(6): 39, 2022 06 10.
Artigo em Inglês | MEDLINE | ID: mdl-35687250

RESUMO

INTRODUCTION: Respiratory tract infections are a worldwide health problem for humans and animals. Different cell types produce lipid mediators in response to infections, which consist of eicosanoids like hydroxyeicosatetraenoic acids (HETEs) or oxylipins like hydroxydocosahexaenoic acids (HDHAs). Both substance classes possess immunomodulatory functions. However, little is known about their role in respiratory infections. OBJECTIVES: Here, we aimed to analyze the lipid mediator imprint of different organs of C57BL/6J mice after intranasal mono-infections with Streptococcus pneumoniae (pneumococcus), Staphylococcus aureus or Influenza A virus (IAV) as wells as pneumococcal-IAV co-infection. METHODS: C57BL/6J mice were infected with different pathogens and lungs, spleen, and plasma were collected. Lipid mediators were analyzed using HPLC-MS/MS. In addition, spatial-distribution of sphingosine 1-phosphate (S1P) and ceramide 1-phosphates (C1P) in tissue samples was examined using MALDI-MS-Imaging. The presence of bacterial pathogens in the lung was confirmed via immunofluorescence staining. RESULTS: We found IAV specific changes for different HDHAs and HETEs in mouse lungs as well as enhanced levels of 20-HETE in severe S. aureus infection. Moreover, MALDI-MS-Imaging analysis showed an accumulation of C1P and a decrease of S1P during co-infection in lung and spleen. Long chain C1P was enriched in the red and not in the white pulp of the spleen. CONCLUSIONS: Lipid mediator analysis showed that host synthesis of bioactive lipids is in part specific for a certain pathogen, in particular for IAV infection. Furthermore, MS-Imaging displayed great potential to study infections and revealed changes of S1P and C1P in lungs and spleen of co-infected animals, which was not described before.


Assuntos
Coinfecção , Vírus da Influenza A , Infecções Respiratórias , Animais , Metabolômica , Camundongos , Camundongos Endogâmicos C57BL , Staphylococcus aureus , Streptococcus pneumoniae , Espectrometria de Massas em Tandem
7.
PLoS Pathog ; 15(7): e1007987, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31356624

RESUMO

Streptococcus pneumoniae (pneumococci) is a leading cause of severe bacterial meningitis in many countries worldwide. To characterize the repertoire of fitness and virulence factors predominantly expressed during meningitis we performed niche-specific analysis of the in vivo proteome in a mouse meningitis model, in which bacteria are directly inoculated into the cerebrospinal fluid (CSF) cisterna magna. We generated a comprehensive mass spectrometry (MS) spectra library enabling bacterial proteome analysis even in the presence of eukaryotic proteins. We recovered 200,000 pneumococci from CSF obtained from meningitis mice and by MS we identified 685 pneumococci proteins in samples from in vitro filter controls and 249 in CSF isolates. Strikingly, the regulatory two-component system ComDE and substrate-binding protein AliB of the oligopeptide transporter system were exclusively detected in pneumococci recovered from the CSF. In the mouse meningitis model, AliB-, ComDE-, or AliB-ComDE-deficiency resulted in attenuated meningeal inflammation and disease severity when compared to wild-type pneumococci indicating the crucial role of ComDE and AliB in pneumococcal meningitis. In conclusion, we show here mechanisms of pneumococcal adaptation to a defined host compartment by a proteome-based approach. Further, this study provides the basis of a promising strategy for the identification of protein antigens critical for invasive disease caused by pneumococci and other meningeal pathogens.


Assuntos
Proteínas de Bactérias/fisiologia , Proteínas de Transporte/fisiologia , Lipoproteínas/fisiologia , Meningite Pneumocócica/microbiologia , Streptococcus pneumoniae/fisiologia , Streptococcus pneumoniae/patogenicidade , Fatores de Virulência/fisiologia , Animais , Proteínas de Bactérias/genética , Proteínas de Transporte/genética , Genes Bacterianos , Interações entre Hospedeiro e Microrganismos/fisiologia , Humanos , Lipoproteínas/deficiência , Lipoproteínas/genética , Masculino , Meningite Pneumocócica/líquido cefalorraquidiano , Camundongos , Camundongos Endogâmicos C57BL , Mutação , Proteômica , Regulon , Streptococcus pneumoniae/genética , Virulência/genética , Virulência/fisiologia , Fatores de Virulência/genética
8.
Blood ; 133(9): 978-989, 2019 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-30573633

RESUMO

Heparin-induced thrombocytopenia (HIT) is caused by platelet-activating anti-platelet factor 4 (PF4)/heparin antibodies. Platelet activation assays that use "washed" platelets are more sensitive for detecting HIT antibodies than platelet-rich plasma (PRP)-based assays. Moreover, heparin-exposed patients vary considerably with respect to the risk of PF4/heparin immunization and, among antibody-positive patients, the risk of subsequent "breakthrough" of clinical HIT with manifestation of thrombocytopenia. We used washed platelets and PRP, standard laboratory HIT tests, and physicochemical methods to identify a plasma factor interfering with PF4/heparin complexes and anti-PF4/heparin antibody-platelet interaction, thus explaining differences in functional assays. To investigate a modulating risk for PF4/heparin immunization and breakthrough of HIT, we also tested 89 plasmas from 2 serosurveillance trials. Fibronectin levels were measured in 4 patient groups exhibiting different degrees of heparin-dependent immunization and expression of HIT. The heat-labile plasma protein, fibronectin, inhibited PF4 binding to platelets in a dose-dependent fashion, particularly in washed (vs PRP) systems. Fibronectin also inhibited PF4/heparin binding to platelets, anti-PF4/heparin antibody binding to PF4/heparin complexes, and anti-PF4/heparin antibody-induced platelet activation as a result of PF4/heparin complex disruption. In addition, plasma fibronectin levels increased progressively among the following 4 patient groups: enzyme-linked immunosorbent assay (ELISA)+/serotonin-release assay (SRA)+/HIT+ < ELISA+/SRA+/HIT- ∼ ELISA+/SRA-/HIT- < ELISA-/SRA-/HIT-. Altogether, these findings suggest that fibronectin interferes with PF4/heparin complex formation and anti-PF4/heparin antibody-induced platelet activation. Reduced fibronectin levels in washed platelet assays help to explain the greater sensitivity of washed platelet (vs PRP) assays for HIT. More importantly, lower plasma fibronectin levels could represent a risk factor for PF4/heparin immunization and clinical breakthrough of HIT.


Assuntos
Anticorpos Monoclonais/imunologia , Plaquetas/imunologia , Fibronectinas/imunologia , Heparina/imunologia , Fator Plaquetário 4/imunologia , Trombocitopenia/patologia , Anticorpos Monoclonais/metabolismo , Anticoagulantes , Plaquetas/metabolismo , Estudos de Casos e Controles , Fibronectinas/metabolismo , Heparina/efeitos adversos , Humanos , Ativação Plaquetária , Fator Plaquetário 4/metabolismo , Prognóstico , Trombocitopenia/induzido quimicamente , Trombocitopenia/imunologia
9.
J Infect Dis ; 222(10): 1702-1712, 2020 10 13.
Artigo em Inglês | MEDLINE | ID: mdl-32445565

RESUMO

BACKGROUND: In tissue infections, adenosine triphosphate (ATP) is released into extracellular space and contributes to purinergic chemotaxis. Neutrophils are important players in bacterial clearance and are recruited to the site of tissue infections. Pneumococcal infections can lead to uncontrolled hyperinflammation of the tissue along with substantial tissue damage through excessive neutrophil activation and uncontrolled granule release. We aimed to investigate the role of ATP in neutrophil response to pneumococcal infections. METHODS: Primary human neutrophils were exposed to the pneumococcal strain TIGR4 and its pneumolysin-deficient mutant or directly to different concentrations of recombinant pneumolysin. Neutrophil activation was assessed by measurement of secreted azurophilic granule protein resistin and profiling of the secretome, using mass spectrometry. RESULTS: Pneumococci are potent inducers of neutrophil degranulation. Pneumolysin was identified as a major trigger of neutrophil activation. This process is partially lysis independent and inhibited by ATP. Pneumolysin and ATP interact with each other in the extracellular space leading to reduced neutrophil activation. Proteome analyses of the neutrophil secretome confirmed that ATP inhibits pneumolysin-dependent neutrophil activation. CONCLUSIONS: Our findings suggest that despite its cytolytic activity, pneumolysin serves as a potent neutrophil activating factor. Extracellular ATP mitigates pneumolysin-induced neutrophil activation.


Assuntos
Trifosfato de Adenosina/metabolismo , Ativação de Neutrófilo/efeitos dos fármacos , Infecções Pneumocócicas/metabolismo , Estreptolisinas/efeitos adversos , Proteínas de Bactérias/efeitos adversos , Morte Celular , Humanos , Neutrófilos/metabolismo , Neutrófilos/microbiologia , Streptococcus pneumoniae
10.
J Proteome Res ; 19(4): 1435-1446, 2020 04 03.
Artigo em Inglês | MEDLINE | ID: mdl-32154730

RESUMO

To understand bacterial reactions to environmental stress or infection-related processes, it is necessary to identify the involved proteins. In mass spectrometry-based proteomics, the method of choice for spectra-to-peptide-match is database search, but in recent times, spectral libraries have come into focus. Here, we built a mass spectral library from Streptococcus pneumoniae D39, reflecting 76% of the theoretical proteome of the organism. Besides the proteins themselves, posttranslational protein modifications especially reveal central hubs of regulation in bacterial pathogenesis. Here, for example, phosphorylation events are involved in the signal transduction and regulation of virulence. Although there have been major advances in phosphoproteomics, identification of this modification is still challenging. To enhance the number of phosphorylated peptides, which can be reproducibly detected, a comprehensive mass spectral library of the S. pneumoniae D39 phosphoproteome has been compiled in addition to the comprehensive total proteome mass spectral library. The phosphopeptide library was manually validated, and the data quality was additionally proven by analyses of synthetic phosphorylated peptides. In total, 128 phosphorylated proteins were revealed, of which many are involved in glycolysis, purine metabolism, protein biosynthesis, and virulence. The publicly available, thoroughly validated spectral libraries are an excellent resource to improve and speed up future investigations on the proteome and phosphoproteome of pneumococci.


Assuntos
Fosfoproteínas , Streptococcus pneumoniae , Espectrometria de Massas , Fosforilação , Proteoma , Proteômica
11.
J Biol Chem ; 294(34): 12795-12806, 2019 08 23.
Artigo em Inglês | MEDLINE | ID: mdl-31289122

RESUMO

Bacterial pneumonia is a global health challenge that causes up to 2 million deaths each year. Purinergic signaling plays a pivotal role in healthy alveolar epithelium. Here, we used fluorophore-based analysis and live-cell calcium imaging to address the question of whether the bacterial pathogen Streptococcus pneumoniae directly interferes with purinergic signaling in alveolar epithelial cells. Disturbed purinergic signaling might result in pathophysiologic changes like edema formation and atelectasis, which are commonly seen in bacterial pneumonia. Purine receptors are mainly activated by ATP, mediating a cytosolic calcium response. We found that this purinergic receptor P2Y2-mediated response is suppressed in the presence of S. pneumoniae in A549 and isolated primary alveolar cells in a temperature-dependent manner. Downstream inositol 3-phosphate (IP3) signaling appeared to be unaffected, as calcium signaling via protease-activated receptor 2 remained unaltered. S. pneumoniae-induced suppression of the P2Y2-mediated calcium response depended on the P2Y2 phosphorylation sites Ser-243, Thr-344, and Ser-356, which are involved in receptor desensitization and internalization. Spinning-disk live-cell imaging revealed that S. pneumoniae induces P2Y2 translocation into the cytosol. In conclusion, our results show that S. pneumoniae directly inhibits purinergic signaling by inducing P2Y2 phosphorylation and internalization, resulting in the suppression of the calcium response of alveolar epithelial cells to ATP, thereby affecting cellular integrity and function.


Assuntos
Células Epiteliais Alveolares/metabolismo , Receptores Purinérgicos P2Y2/metabolismo , Streptococcus pneumoniae/metabolismo , Células A549 , Trifosfato de Adenosina/metabolismo , Animais , Sinalização do Cálcio , Células Cultivadas , Humanos , Masculino , Ratos , Ratos Sprague-Dawley , Receptores Purinérgicos P2Y2/genética
12.
Acta Neuropathol ; 140(2): 183-208, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32529267

RESUMO

Bacterial meningitis is a deadly disease most commonly caused by Streptococcus pneumoniae, leading to severe neurological sequelae including cerebral edema, seizures, stroke, and mortality when untreated. Meningitis is initiated by the transfer of S. pneumoniae from blood to the brain across the blood-cerebrospinal fluid barrier or the blood-brain barrier (BBB). The underlying mechanisms are still poorly understood. Current treatment strategies include adjuvant dexamethasone for inflammation and cerebral edema, followed by antibiotics. The success of dexamethasone is however inconclusive, necessitating new therapies for controlling edema, the primary reason for neurological complications. Since we have previously shown a general activation of hypoxia inducible factor (HIF-1α) in bacterial infections, we hypothesized that HIF-1α, via induction of vascular endothelial growth factor (VEGF) is involved in transmigration of pathogens across the BBB. In human, murine meningitis brain samples, HIF-1α activation was observed by immunohistochemistry. S. pneumoniae infection in brain endothelial cells (EC) resulted in in vitro upregulation of HIF-1α/VEGF (Western blotting/qRT-PCR) associated with increased paracellular permeability (fluorometry, impedance measurements). This was supported by bacterial localization at cell-cell junctions in vitro and in vivo in brain ECs from mouse and humans (confocal, super-resolution, electron microscopy, live-cell imaging). Hematogenously infected mice showed increased permeability, S. pneumoniae deposition in the brain, along with upregulation of genes in the HIF-1α/VEGF pathway (RNA sequencing of brain microvessels). Inhibition of HIF-1α with echinomycin, siRNA in bEnd5 cells or using primary brain ECs from HIF-1α knock-out mice revealed reduced endothelial permeability and transmigration of S. pneumoniae. Therapeutic rescue using the HIF-1α inhibitor echinomycin resulted in increased survival and improvement of BBB function in S. pneumoniae-infected mice. We thus demonstrate paracellular migration of bacteria across BBB and a critical role for HIF-1α/VEGF therein and hence propose targeting this pathway to prevent BBB dysfunction and ensuing brain damage in infections.


Assuntos
Barreira Hematoencefálica , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Meningite Pneumocócica , Streptococcus pneumoniae , Migração Transendotelial e Transepitelial/fisiologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Barreira Hematoencefálica/metabolismo , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Fator A de Crescimento do Endotélio Vascular/metabolismo
13.
Immunity ; 34(2): 258-68, 2011 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-21349432

RESUMO

Activated mast cells trigger edema in allergic and inflammatory disease. We report a paracrine mechanism by which mast cell-released heparin increases vascular permeability in vivo. Heparin activated the protease factor XII, which initiates bradykinin formation in plasma. Targeting factor XII or kinin B2 receptors abolished heparin-triggered leukocyte-endothelium adhesion and interfered with a mast cell-driven drop in blood pressure in rodents. Intravital laser scanning microscopy and tracer measurements showed heparin-driven fluid extravasation in mouse skin microvessels. Ablation of factor XII or kinin B2 receptors abolished heparin-induced skin edema and protected mice from allergen-activated mast cell-driven leakage. In contrast, heparin and activated mast cells induced excessive edema in mice deficient in the major inhibitor of factor XII, C1 esterase inhibitor. Allergen exposure triggered edema attacks in hereditary angioedema patients, lacking C1 esterase inhibitor. The data indicate that heparin-initiated bradykinin formation plays a fundamental role in mast cell-mediated diseases.


Assuntos
Bradicinina/biossíntese , Síndrome de Vazamento Capilar/fisiopatologia , Permeabilidade Capilar/fisiologia , Heparina/fisiologia , Mastócitos/metabolismo , Anafilaxia Cutânea Passiva/fisiologia , Animais , Bradicinina/genética , Síndrome de Vazamento Capilar/etiologia , Adesão Celular , Proteína Inibidora do Complemento C1/fisiologia , Edema/etiologia , Edema/fisiopatologia , Células Endoteliais/patologia , Ativação Enzimática , Fator XII/fisiologia , Heparina/metabolismo , Hipotensão/etiologia , Hipotensão/fisiopatologia , Imunoglobulina E/imunologia , Sistema Calicreína-Cinina/fisiologia , Leucócitos/fisiologia , Masculino , Camundongos , Comunicação Parácrina/fisiologia , Plasma , Ratos , Transdução de Sinais/fisiologia , Pele/irrigação sanguínea
14.
Eur J Clin Microbiol Infect Dis ; 39(10): 1865-1878, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32409955

RESUMO

The aim of our study was to investigate phenotypic and genotypic features of streptococci misidentified (misID) as Streptococcus pneumoniae, obtained over 20 years from hospital patients in Poland. Sixty-three isolates demonstrating microbiological features typical for pneumococci (optochin susceptibility and/or bile solubility) were investigated by phenotypic tests, lytA and 16S rRNA gene polymorphism and whole-genome sequencing (WGS). All isolates had a 6-bp deletion in the lytA 3' terminus, characteristic for Mitis streptococc and all but two isolates lacked the pneumococcal signature cytosine at nucleotide position 203 in the 16S rRNA genes. The counterparts of psaA and ply were present in 100% and 81.0% of isolates, respectively; the spn9802 and spn9828 loci were characteristic for 49.2% and 38.1% of isolates, respectively. Phylogenetic trees and networks, based on the multilocus sequence analysis (MLSA) scheme, ribosomal multilocus sequence typing (rMLST) scheme and core-genome analysis, clearly separated investigated isolates from S. pneumoniae and demonstrated the polyclonal character of misID streptococci, associated with the Streptococcus pseudopneumoniae and Streptococcus mitis groups. While the S. pseudopneumoniae clade was relatively well defined in all three analyses, only the core-genome analysis revealed the presence of another cluster comprising a fraction of misID streptococci and a strain proposed elsewhere as a representative of a novel species in the Mitis group. Our findings point to complex phylogenetic and taxonomic relationships among S. mitis-like bacteria and support the notion that this group may in fact consist of several distinct species.


Assuntos
Infecções Estreptocócicas/epidemiologia , Streptococcus mitis/isolamento & purificação , Streptococcus/isolamento & purificação , Técnicas de Tipagem Bacteriana , Erros de Diagnóstico , Feminino , Humanos , Masculino , Filogenia , Polônia/epidemiologia , RNA Ribossômico 16S , Infecções Estreptocócicas/microbiologia , Streptococcus/genética , Streptococcus mitis/genética , Streptococcus pneumoniae/genética , Streptococcus pneumoniae/isolamento & purificação
15.
J Biol Chem ; 293(27): 10620-10629, 2018 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-29764936

RESUMO

The bacterial lung pathogen Streptococcus pneumoniae has a unique nutritional requirement for exogenous choline and attaches phosphorylcholine (P-Cho) residues to the GalpNAc moieties of its teichoic acids (TAs) in its cell wall. Two phosphorylcholine transferases, LicD1 and LicD2, mediate the attachment of P-Cho to the O-6 positions of the two GalpNAc residues present in each repeating unit of pneumococcal TAs (pnTAs), of which only LicD1 has been determined to be essential. At the molecular level, the specificity of the P-Cho attachment to pnTAs by LicD1 and LicD2 remains still elusive. Here, using detailed structural analyses of pnTAs from a LicD2-deficient strain, we confirmed the specificity in the attachment of P-Cho residues to pnTA. LicD1 solely transfers P-Cho to α-d-GalpNAc moieties, whereas LicD2 attaches P-Cho to ß-d-GalpNAc. Further, we investigated the role of the pneumococcal phosphorylcholine esterase (Pce) in the modification of the P-Cho substitution pattern of pnTAs. To clarify the specificity of Pce-mediated P-Cho hydrolysis, we evaluated different concentrations and pH conditions for the treatment of pneumococcal lipoteichoic acid with purified Pce. We show that Pce can hydrolyze both P-Cho residues of the terminal repeat of the pnTA chain and almost all P-Cho residues bound to ß-d-GalpNAc in vitro However, hydrolysis in vivo was restricted to the terminal repeat. In summary, our findings indicate that LicD1 and LicD2 specifically transfer P-Cho to α-d-GalpNAc and ß-d-GalpNAc moieties, respectively, and that Pce removes distinct P-Cho substituents from pnTAs.


Assuntos
Proteínas de Bactérias/metabolismo , Lipopolissacarídeos/química , Fosforilcolina/química , Receptores de Superfície Celular/metabolismo , Streptococcus pneumoniae/enzimologia , Ácidos Teicoicos/química , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Lipopolissacarídeos/metabolismo , Mutação , Fosforilcolina/metabolismo , Receptores de Superfície Celular/química , Receptores de Superfície Celular/genética , Ácidos Teicoicos/metabolismo
16.
Infect Immun ; 87(1)2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30323030

RESUMO

Complement is a critical component of antimicrobial immunity. Various complement regulatory proteins prevent host cells from being attacked. Many pathogens have acquired the ability to sequester complement regulators from host plasma to evade complement attack. We describe here how Streptococcus pneumoniae adopts a strategy to prevent the formation of the C3 convertase C4bC2a by the rapid conversion of surface bound C4b and iC4b into C4dg, which remains bound to the bacterial surface but no longer forms a convertase complex. Noncapsular virulence factors on the pneumococcus are thought to facilitate this process by sequestering C4b-binding protein (C4BP) from host plasma. When S. pneumoniae D39 was opsonized with human serum, the larger C4 activation products C4b and iC4b were undetectable, but the bacteria were liberally decorated with C4dg and C4BP. With targeted deletions of either PspA or PspC, C4BP deposition was markedly reduced, and there was a corresponding reduction in C4dg and an increase in the deposition of C4b and iC4b. The effect was greatest when PspA and PspC were both knocked out. Infection experiments in mice indicated that the deletion of PspA and/or PspC resulted in the loss of bacterial pathogenicity. Recombinant PspA and PspC both bound serum C4BP, and both led to increased C4b and reduced C4dg deposition on S. pneumoniae D39. We conclude that PspA and PspC help the pneumococcus to evade complement attack by binding C4BP and so inactivating C4b.


Assuntos
Proteínas de Bactérias/metabolismo , Proteína de Ligação ao Complemento C4b/metabolismo , Complemento C4b/antagonistas & inibidores , Evasão da Resposta Imune , Streptococcus pneumoniae/imunologia , Animais , Modelos Animais de Doenças , Humanos , Camundongos , Infecções Pneumocócicas/microbiologia , Ligação Proteica , Streptococcus pneumoniae/patogenicidade
17.
J Infect Dis ; 217(3): 358-370, 2018 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-28968817

RESUMO

Pneumococcal hemolytic uremic syndrome (HUS) in children is caused by infections with Streptococcus pneumoniae. Because endothelial cell damage is a hallmark of HUS, we studied how HUS-inducing pneumococci derived from infant HUS patients during the acute phase disrupt the endothelial layer. HUS pneumococci efficiently bound human plasminogen. These clinical isolates of HUS pneumococci efficiently bound human plasminogen via the bacterial surface proteins Tuf and PspC. When activated to plasmin at the bacterial surface, the active protease degraded fibrinogen and cleaved C3b. Here, we show that PspC is a pneumococcal plasminogen receptor and that plasmin generated on the surface of HUS pneumococci damages endothelial cells, causing endothelial retraction and exposure of the underlying matrix. Thus, HUS pneumococci damage endothelial cells in the blood vessels and disturb local complement homeostasis. Thereby, HUS pneumococci promote a thrombogenic state that drives HUS pathology.


Assuntos
Aderência Bacteriana , Proteínas de Bactérias/metabolismo , Células Endoteliais/patologia , Fibrinolisina/metabolismo , Síndrome Hemolítico-Urêmica/microbiologia , Plasminogênio/metabolismo , Streptococcus pneumoniae/fisiologia , Pré-Escolar , Feminino , Humanos , Infecções Pneumocócicas/microbiologia , Ligação Proteica , Streptococcus pneumoniae/isolamento & purificação
18.
J Biol Chem ; 292(14): 5770-5783, 2017 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-28209711

RESUMO

Streptococcus pneumoniae serotype 3 strains emerge frequently within clinical isolates of invasive diseases. Bacterial invasion into deeper tissues is associated with colonization and immune evasion mechanisms. Thus, pneumococci express a versatile repertoire of surface proteins sequestering and interacting specifically with components of the human extracellular matrix and serum. Hic, a PspC-like pneumococcal surface protein, possesses vitronectin and factor H binding activity. Here, we show that heterologously expressed Hic domains interact, similar to the classical PspC molecule, with human matricellular thrombospondin-1 (hTSP-1). Binding studies with isolated human thrombospondin-1 and various Hic domains suggest that the interaction between hTSP-1 and Hic differs from binding to vitronectin and factor H. Binding of Hic to hTSP-1 is inhibited by heparin and chondroitin sulfate A, indicating binding to the N-terminal globular domain or type I repeats of hTSP-1. Competitive inhibition experiments with other pneumococcal hTSP-1 adhesins demonstrated that PspC and PspC-like Hic recognize similar domains, whereas PavB and Hic can bind simultaneously to hTSP-1. In conclusion, Hic binds specifically hTSP-1; however, truncation in the N-terminal part of Hic decreases the binding activity, suggesting that the full length of the α-helical regions of Hic is required for an optimal interaction.


Assuntos
Adesinas Bacterianas/metabolismo , Proteínas de Bactérias/metabolismo , Plaquetas/metabolismo , Proteínas de Transporte/metabolismo , Streptococcus pneumoniae/metabolismo , Trombospondina 1/metabolismo , Humanos , Ligação Proteica
19.
BMC Genomics ; 19(1): 10, 2018 01 03.
Artigo em Inglês | MEDLINE | ID: mdl-29298677

RESUMO

BACKGROUND: In recent years, the idea of a highly immunogenic protein-based vaccine to combat Streptococcus pneumoniae and its severe invasive infectious diseases has gained considerable interest. However, the target proteins to be included in a vaccine formulation have to accomplish several genetic and immunological characteristics, (such as conservation, distribution, immunogenicity and protective effect), in order to ensure its suitability and effectiveness. This study aimed to get comprehensive insights into the genomic organization, population distribution and genetic conservation of all pneumococcal surface-exposed proteins, genetic regulators and other virulence factors, whose important function and role in pathogenesis has been demonstrated or hypothesized. RESULTS: After retrieving the complete set of DNA and protein sequences reported in the databases GenBank, KEGG, VFDB, P2CS and Uniprot for pneumococcal strains whose genomes have been fully sequenced and annotated, a comprehensive bioinformatic analysis and systematic comparison has been performed for each virulence factor, stand-alone regulator and two-component regulatory system (TCS) encoded in the pan-genome of S. pneumoniae. A total of 25 S. pneumoniae strains, representing different pneumococcal phylogenetic lineages and serotypes, were considered. A set of 92 different genes and proteins were identified, classified and studied to construct a pan-genomic variability map (variome) for S. pneumoniae. Both, pneumococcal virulence factors and regulatory genes, were well-distributed in the pneumococcal genome and exhibited a conserved feature of genome organization, where replication and transcription are co-oriented. The analysis of the population distribution for each gene and protein showed that 49 of them are part of the core genome in pneumococci, while 43 belong to the accessory-genome. Estimating the genetic variability revealed that pneumolysin, enolase and Usp45 (SP_2216 in S. p. TIGR4) are the pneumococcal virulence factors with the highest conservation, while TCS08, TCS05, and TCS02 represent the most conserved pneumococcal genetic regulators. CONCLUSIONS: The results identified well-distributed and highly conserved pneumococcal virulence factors as well as regulators, representing promising candidates for a new generation of serotype-independent protein-based vaccine(s) to combat pneumococcal infections.


Assuntos
Variação Genética , Streptococcus pneumoniae/genética , Fatores de Virulência/genética , Proteínas de Bactérias/genética , Mapeamento Cromossômico , Genes Bacterianos , Genes Reguladores , Genoma Bacteriano , Filogenia , Streptococcus pneumoniae/classificação , Streptococcus pneumoniae/patogenicidade
20.
Mol Microbiol ; 105(6): 839-859, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28657670

RESUMO

Colonization of mucosal respiratory surfaces is a prerequisite for the human pathobiont Streptococcus pneumoniae (the pneumococcus) to cause severe invasive infections. The arsenal of pneumococcal adhesins interacts with a multitude of extracellular matrix proteins. A paradigm for pneumococci is their interaction with the adhesive glycoprotein fibronectin, which facilitates bacterial adherence to host cells. Here, we deciphered the molecular interaction between fibronectin and pneumococcal fibronectin-binding proteins (FnBPs) PavA and PavB respectively. We show in adherence and binding studies that the pneumococcal interaction with fibronectin is a non-human specific trait. PavA and PavB target at least 13 out of 15 type III fibronectin domains as demonstrated in ligand overlay assays, surface plasmon resonance studies and SPOT peptide arrays. Strikingly, both pneumococcal FnBPs recognize similar peptides in targeted type III repeats. Structural comparisons revealed that the targeted type III repeat epitopes cluster on the inner strands of both ß-sheets forming the fibronectin domains. Importantly, synthetic peptides of FnIII1 , FnIII5 or FnIII15 bind directly to FnBPs PavA and PavB respectively. In conclusion, our study suggests a common pattern of molecular interactions between pneumococcal FnBPs and fibronectin. The specific epitopes recognized in this study can potentially be tested as antimicrobial targets in further scientific endeavours.


Assuntos
Proteínas de Bactérias/metabolismo , Domínio de Fibronectina Tipo III/fisiologia , Fibronectinas/metabolismo , Adesinas Bacterianas/metabolismo , Aderência Bacteriana/genética , Aderência Bacteriana/fisiologia , Proteínas de Bactérias/genética , Proteínas de Transporte/metabolismo , Domínio de Fibronectina Tipo III/genética , Interações Hospedeiro-Patógeno , Humanos , Ligação Proteica/genética , Ligação Proteica/fisiologia , Domínios Proteicos , Streptococcus pneumoniae/metabolismo , Fatores de Virulência/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA