Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
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
2.
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
3.
Thromb Haemost ; 115(4): 752-61, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26467272

RESUMO

Anti-platelet factor 4 (PF4)/heparin antibodies are not only the cause of heparin-induced thrombocytopenia but might also play a role in the antibacterial host defence. Recently, marginal zone (MZ) B cells were identified to be crucial for anti-PF4/heparin IgG antibody production in mice. Combining human studies and a murine model of polymicrobial sepsis we further characterised the far less investigated anti-PF4/heparin IgM immune response. We detected anti-PF4/heparin IgM antibodies in the sera of paediatric patients < 6 months of age after cardiac surgery and in sera of splenectomised mice subjected to polymicrobial sepsis. In addition, PF4/heparin-specific IgM B cells were not only found in murine spleen, but also in peritoneum and bone marrow upon in vitro stimulation. Together, this indicates involvement of additional B cell populations, as MZ B cells are not fully developed in humans until the second year of life and are restricted to the spleen in mice. Moreover, PF4/heparin-specific B cells were detected in human cord blood upon in vitro stimulation and PF4-/- mice produced anti-PF4/heparin IgM antibodies after polymicrobial sepsis. In conclusion, the anti-PF4/heparin IgM response is a potential innate immune reaction driven by a B cell population distinct from MZ B cells.


Assuntos
Linfócitos B/imunologia , Coinfecção/imunologia , Imunoglobulina M/sangue , Fator Plaquetário 4/imunologia , Sepse/imunologia , Trombocitopenia/imunologia , Animais , Formação de Anticorpos , Modelos Animais de Doenças , Feminino , Humanos , Imunidade Inata , Lactente , Recém-Nascido , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator Plaquetário 4/genética , Trombocitopenia/induzido quimicamente
4.
Thromb Res ; 134(1): 174-81, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24816371

RESUMO

INTRODUCTION: The key feature of heparin-induced thrombocytopenia (HIT) is the production of antibodies (Ab) against the platelet factor 4 (PF4)/heparin complex. These Ab are directed against neoepitopes of the PF4 tetramer, which are induced by the complex formation with heparin. To study this humoral immune response in greater detail, either in a murine immunization model or in human blood samples, reliable and specific immune assays to detect specifically Ab against the PF4/heparin complexes, but not PF4 alone are required. MATERIALS AND METHODS: We established fluid-phase enzyme-immunoassays in which the soluble biotinylated antigen, PF4/heparin, is firstly captured by specific Ab, and secondly directly detected with enzyme-conjugated streptavidin. RESULTS: The use of this fluid-phase principle allowed a higher specificity than the traditional solid-phase enzyme-immunoassays in terms of Ab binding to murine PF4/heparin compared to murine PF4 alone. This fluid-phase approach applied to the detection of specific murine PF4/heparin Ab-secreting cells (ASC) identified the spleen as the main lymphatic organ that contributes to the PF4/heparin Ab response in mice. IgG ASC specific for PF4/heparin are very transiently detectable in mice, which might explain why anti-PF4/heparin IgG Ab typically disappear within 100 days in humans. Furthermore, this fluid-phase approach was successfully transferred to detect human PF4/heparin-specific Ab. CONCLUSION: The fluid-phase principle for the specific detection of anti-PF4/heparin IgG and IgM Ab enables new and improved assays for HIT research in men and mice. At least in mice PF4/heparin antibodies are produced by transient B cells.


Assuntos
Anticorpos/sangue , Células Produtoras de Anticorpos/metabolismo , Heparina/análise , Fator Plaquetário 4/análise , Trombocitopenia/induzido quimicamente , Animais , Feminino , Heparina/imunologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Fator Plaquetário 4/imunologia , Trombocitopenia/imunologia
5.
J Vis Exp ; (84): e51174, 2014 Feb 23.
Artigo em Inglês | MEDLINE | ID: mdl-24637643

RESUMO

Pneumonia is one of the major health care problems in developing and industrialized countries and is associated with considerable morbidity and mortality. Despite advances in knowledge of this illness, the availability of intensive care units (ICU), and the use of potent antimicrobial agents and effective vaccines, the mortality rates remain high(1). Streptococcus pneumoniae is the leading pathogen of community-acquired pneumonia (CAP) and one of the most common causes of bacteremia in humans. This pathogen is equipped with an armamentarium of surface-exposed adhesins and virulence factors contributing to pneumonia and invasive pneumococcal disease (IPD). The assessment of the in vivo role of bacterial fitness or virulence factors is of utmost importance to unravel S. pneumoniae pathogenicity mechanisms. Murine models of pneumonia, bacteremia, and meningitis are being used to determine the impact of pneumococcal factors at different stages of the infection. Here we describe a protocol to monitor in real-time pneumococcal dissemination in mice after intranasal or intraperitoneal infections with bioluminescent bacteria. The results show the multiplication and dissemination of pneumococci in the lower respiratory tract and blood, which can be visualized and evaluated using an imaging system and the accompanying analysis software.


Assuntos
Medições Luminescentes/métodos , Pneumonia Pneumocócica/microbiologia , Streptococcus pneumoniae/patogenicidade , Fatores de Virulência/análise , Animais , Bacteriemia/sangue , Bacteriemia/microbiologia , Modelos Animais de Doenças , Feminino , Pulmão/microbiologia , Camundongos , Pneumonia Pneumocócica/sangue , Streptococcus pneumoniae/química , Streptococcus pneumoniae/metabolismo , Fatores de Virulência/metabolismo
6.
EMBO Mol Med ; 5(12): 1852-70, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24136784

RESUMO

The respiratory pathogen Streptococcus pneumoniae has evolved efficient mechanisms to resist oxidative stress conditions and to displace other bacteria in the nasopharynx. Here we characterize at physiological, functional and structural levels two novel surface-exposed thioredoxin-family lipoproteins, Etrx1 and Etrx2. The impact of both Etrx proteins and their redox partner methionine sulfoxide reductase SpMsrAB2 on pneumococcal pathogenesis was assessed in mouse virulence studies and phagocytosis assays. The results demonstrate that loss of function of either both Etrx proteins or SpMsrAB2 dramatically attenuated pneumococcal virulence in the acute mouse pneumonia model and that Etrx proteins compensate each other. The deficiency of Etrx proteins or SpMsrAB2 further enhanced bacterial uptake by macrophages, and accelerated pneumococcal killing by H2 O2 or free methionine sulfoxides (MetSO). Moreover, the absence of both Etrx redox pathways provokes an accumulation of oxidized SpMsrAB2 in vivo. Taken together our results reveal insights into the role of two extracellular electron pathways required for reduction of SpMsrAB2 and surface-exposed MetSO. Identification of this system and its target proteins paves the way for the design of novel antimicrobials.


Assuntos
Proteínas de Bactérias/metabolismo , Streptococcus pneumoniae/metabolismo , Animais , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Domínio Catalítico , Cristalografia por Raios X , Modelos Animais de Doenças , Feminino , Peróxido de Hidrogênio/farmacologia , Macrófagos/imunologia , Macrófagos/fisiologia , Metionina/análogos & derivados , Metionina/farmacologia , Camundongos , Dados de Sequência Molecular , Estresse Oxidativo/efeitos dos fármacos , Fagocitose , Pneumonia/imunologia , Pneumonia/microbiologia , Pneumonia/mortalidade , Estrutura Terciária de Proteína , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Streptococcus pneumoniae/patogenicidade , Análise de Sobrevida , Virulência
7.
Blood ; 120(5): 930-1, 2012 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-22859710

RESUMO

Antiplatelet factor 4 (PF4) antibodies have an important role in the most frequent drug-induced immune disorder, heparin-induced thrombocytopenia (HIT). In this issue of Blood, Sachais and coworkers propose a new feature that may explain why only some anti-PF4 antibodies are pathogenic.(1) In addition to epitope specificity-determining affinity and a high titer, the ability of antibodies to promote formation of their own target antigens seems to be a key factor for pathogenicity.


Assuntos
Anticorpos/metabolismo , Afinidade de Anticorpos/fisiologia , Trombocitopenia/etiologia , Trombocitopenia/imunologia , Animais , Humanos
8.
Microbiology (Reading) ; 158(Pt 3): 771-780, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22222496

RESUMO

Lactococcus lactis is a non-pathogenic bacterium that is used in the food industry but is also used as a heterologous host to reveal protein functions of pathogenic bacteria. The adhesin PspC from Streptococcus pneumoniae is a choline-binding protein that is non-covalently anchored to the bacterial cell wall. To assess the exclusive impact of pneumococcal surface protein C (PspC) on the interplay with its host we generated recombinant L. lactis producing a nisin-inducible and covalently anchored variant of PspC on the lactococcal cell surface. A translational fusion of the 5'-end of pspC3.4 with the 3'-end of hic (pspC11.4) was designed to decorate the surface of L. lactis with a chimeric PspC. The PspC3.4 part comprises the first 281 aa residues of PspC3.4, while the Hic sequence consists of the proline-rich and sortase-anchored domain. The results demonstrated that PspC is sufficient for adhesion and subsequent invasion of host epithelial cells expressing the human polymeric Ig receptor (hpIgR). Moreover, invasion via hpIgR was even more pronounced when the chimeric PspC was produced by lactococci compared with pneumococci. This study shows also for the first time that PspC plays no significant role during phagocytosis by macrophages. In contrast, recruitment of Factor H via the PspC chimer has a dramatic effect on phagocytosis of recombinant but not wild-type lactococci, as Factor H interacts specifically with the amino-terminal part of PspC and mediates the contact with phagocytes. Furthermore, L. lactis expressing PspC increased intracellular calcium levels in pIgR-expressing epithelial cells, thus resembling the effect of pneumococci, which induced release of Ca(2+) from intracellular stores via the PspC-pIgR mechanism. In conclusion, expression of the chimeric PspC confers adhesive properties to L. lactis and indicates the potential of L. lactis as a suitable host to study the impact of individual bacterial factors on their capacity to interfere with the host and manipulate eukaryotic epithelial cells.


Assuntos
Aderência Bacteriana , Proteínas de Bactérias/metabolismo , Expressão Gênica , Lactococcus lactis/patogenicidade , Streptococcus pneumoniae/patogenicidade , Fatores de Virulência/metabolismo , Adesivos , Animais , Proteínas de Bactérias/genética , Linhagem Celular , Cães , Células Epiteliais/microbiologia , Humanos , Lactococcus lactis/genética , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Receptores de Imunoglobulina Polimérica/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Streptococcus pneumoniae/genética , Virulência , Fatores de Virulência/genética
9.
J Biol Chem ; 285(30): 23486-95, 2010 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-20504767

RESUMO

Streptococcus pneumoniae, a human pathogen, recruits complement regulator factor H to its bacterial cell surface. The bacterial PspC protein binds Factor H via short consensus repeats (SCR) 8-11 and SCR19-20. In this study, we define how bacterially bound Factor H promotes pneumococcal adherence to and uptake by epithelial cells or human polymorphonuclear leukocytes (PMNs) via a two-step process. First, pneumococcal adherence to epithelial cells was significantly reduced by heparin and dermatan sulfate. However, none of the glycosaminoglycans affected binding of Factor H to pneumococci. Adherence of pneumococci to human epithelial cells was inhibited by monoclonal antibodies recognizing SCR19-20 of Factor H suggesting that the C-terminal glycosaminoglycan-binding region of Factor H mediates the contact between pneumococci and human cells. Blocking of the integrin CR3 receptor, i.e. CD11b and CD18, of PMNs or CR3-expressing epithelial cells reduced significantly the interaction of pneumococci with both cell types. Similarly, an additional CR3 ligand, Pra1, derived from Candida albicans, blocked the interaction of pneumococci with PMNs. Strikingly, Pra1 inhibited also pneumococcal uptake by lung epithelial cells but not adherence. In addition, invasion of Factor H-coated pneumococci required the dynamics of host-cell actin microfilaments and was affected by inhibitors of protein-tyrosine kinases and phosphatidylinositol 3-kinase. In conclusion, pneumococcal entry into host cells via Factor H is based on a two-step mechanism. The first and initial contact of Factor H-coated pneumococci is mediated by glycosaminoglycans expressed on the surface of human cells, and the second step, pneumococcal uptake, is integrin-mediated and depends on host signaling molecules such as phosphatidylinositol 3-kinase.


Assuntos
Fator H do Complemento/metabolismo , Streptococcus pneumoniae/metabolismo , Actinas/metabolismo , Animais , Aderência Bacteriana , Transporte Biológico , Células CHO , Linhagem Celular , Fator H do Complemento/química , Sequência Consenso , Cricetinae , Cricetulus , Citoesqueleto/metabolismo , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Glicosaminoglicanos/metabolismo , Heparina/química , Heparina/metabolismo , Humanos , Antígeno de Macrófago 1/metabolismo , Neutrófilos/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Tirosina Quinases/metabolismo , Sequências Repetitivas de Aminoácidos , Transdução de Sinais , Solubilidade , Streptococcus pneumoniae/fisiologia
10.
Mol Microbiol ; 77(1): 22-43, 2010 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-20444103

RESUMO

The genomic analysis of Streptococcus pneumoniae strains identified the Pneumococcal adherence and virulence factor B (PavB), whose repetitive sequences, designated Streptococcal Surface REpeats (SSURE), interact with human fibronectin. Here, we showed the gene in all tested pneumococci and identified that the observed differences in the molecular mass of PavB rely on the number of repeats, ranging from five to nine SSURE. PavB interacted with fibronectin and plasminogen in a dose-dependent manner as shown by using various SSURE peptides. In addition, we identified PavB as colonization factor. Mice infected intranasally with DeltapavB pneumococci showed significantly increased survival times compared with wild-type bacteria. Importantly, the pavB-mutant showed a delay in transmigration to the lungs as observed in real-time using bioluminescent pneumococci and decreased colonization rates in a nasopharyngeal carriage model. In co-infection experiments the wild-type out-competed the pavB-mutant and infections of epithelial cells demonstrated that PavB contributes to adherence to host cell. Blocking experiments suggested a function of PavB as adhesin, which was confirmed by direct binding of SSURE peptides to host cells. Finally, PavB may represent a new vaccine candidate as SSURE peptides reacted with human sera. Taken together, PavB is a surface-exposed adhesin, which contributes to pneumococcal colonization and infections of the respiratory airways.


Assuntos
Adesinas Bacterianas/fisiologia , Aderência Bacteriana , Nasofaringe/microbiologia , Infecções Pneumocócicas/microbiologia , Sistema Respiratório/microbiologia , Streptococcus pneumoniae/patogenicidade , Fatores de Virulência/fisiologia , Adesinas Bacterianas/química , Adesinas Bacterianas/genética , Animais , Portador Sadio/microbiologia , DNA Bacteriano/química , DNA Bacteriano/genética , Células Epiteliais/microbiologia , Feminino , Fibronectinas/metabolismo , Humanos , Camundongos , Dados de Sequência Molecular , Peso Molecular , Plasminogênio/metabolismo , Pneumonia Pneumocócica/microbiologia , Ligação Proteica , Sequências Repetitivas de Aminoácidos , Análise de Sequência de DNA , Análise de Sobrevida , Fatores de Virulência/química , Fatores de Virulência/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...