Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Thromb Haemost ; 2024 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-39029905

RESUMEN

BACKGROUND: Platelets prevent extravasation of capillary fluids into the pulmonary interstitial tissue by sealing gaps in inflamed endothelium. This reduces respiratory distress associated with pneumonia. Streptococcus pneumoniae is the leading cause of severe community-acquired pneumonia. Pneumococci produce pneumolysin (PLY), which forms pores in membranes of eukaryotic cells including platelets. Additionally, pneumococci express neuraminidases, which cleave sialic acid residues from eukaryotic glycoproteins. In this study, we investigated the effect of desialylation on PLY binding and pore formation on platelets. MATERIAL AND METHODS: We incubated human platelets with purified neuraminidases and PLY, nonencapsulated D39/TIGR4 and isogenic mutants deficient for PLY and/or NanA. We assessed platelet desialylation, PLY binding and pore formation by flow cytometry. We also analyzed the inhibitory potential of therapeutic immunoglobulin G preparations. RESULTS: Pneumococci cause desialylation of platelet glycoproteins by neuraminidases, which is reduced by 90-100% in NanA-deficient mutants. NanC, cleaving only 2,3-linked sialic acid, induced platelet desialylation. PLY binding to platelets doubled (p=0.0166) and pore formation tripled (p=0.0373). A neuraminidase cleaving 2,3-, 2,6-, and 2,8-linked sialic acid like NanA was even more efficient. Addition of polyvalent IVIG decreased platelet desialylation induced by NanC up to 90% (p=0.263) and reduced pore formation >95% (p<0.0001) when incubated with pneumococci. Conclusion: Neuraminidases are key virulence factors of pneumococci and desialylate platelet glycoproteins, thereby unmasking PLY-binding sites. This enhances binding of PLY and pore formation showing that pneumococcal neuraminidases and PLY act in concert to kill platelets. However, human polyvalent IgG preparations are promising agents for therapeutic intervention during severe pneumococcal pneumonia.

2.
Cells ; 11(10)2022 05 13.
Artículo en Inglés | MEDLINE | ID: mdl-35626674

RESUMEN

Group B streptococci (GBS) cause a range of invasive maternal-fetal diseases during pregnancy and post-partum. However, invasive infections in non-pregnant adults are constantly increasing. These include sepsis and streptococcal toxic shock syndrome, which are often complicated by systemic coagulation and thrombocytopenia. GBS express a hyper-hemolytic ornithine rhamnolipid pigment toxin with cytolytic and coagulatory activity. Here, we investigated the effects of GBS pigment on human platelets. Infections of platelets with pigmented GBS resulted initially in platelet activation, followed by necrotic cell death. Thus, this study shows that GBS pigment kills human platelets.


Asunto(s)
Infecciones Estreptocócicas , Streptococcus agalactiae , Adulto , Femenino , Hemólisis , Humanos , Pigmentación , Activación Plaquetaria , Embarazo
3.
Cells ; 11(7)2022 03 25.
Artículo en Inglés | MEDLINE | ID: mdl-35406684

RESUMEN

Systemic infections with pathogenic or facultative pathogenic bacteria are associated with activation and aggregation of platelets leading to thrombocytopenia and activation of the clotting system. Bacterial proteins leading to platelet activation and aggregation have been identified, and while platelet receptors are recognized, induced signal transduction cascades are still often unknown. In addition to proteinaceous adhesins, pathogenic bacteria such as Staphylococcus aureus and Streptococcus pneumoniae also produce toxins such as pneumolysin and alpha-hemolysin. They bind to cellular receptors or form pores, which can result in disturbance of physiological functions of platelets. Here, we discuss the bacteria-platelet interplay in the context of adhesin-receptor interactions and platelet-activating bacterial proteins, with a main emphasis on S. aureus and S. pneumoniae. More importantly, we summarize recent findings of how S. aureus toxins and the pore-forming toxin pneumolysin of S. pneumoniae interfere with platelet function. Finally, the relevance of platelet dysfunction due to killing by toxins and potential treatment interventions protecting platelets against cell death are summarized.


Asunto(s)
Plaquetas , Streptococcus pneumoniae , Adhesinas Bacterianas/metabolismo , Proteínas Bacterianas/metabolismo , Plaquetas/metabolismo , Staphylococcus aureus , Streptococcus pneumoniae/metabolismo
4.
J Thromb Haemost ; 20(6): 1464-1475, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35303391

RESUMEN

BACKGROUND: Toxins are key virulence determinants of pathogens and can impair the function of host immune cells, including platelets. Insights into pathogen toxin interference with platelets will be pivotal to improve treatment of patients with bacterial bloodstream infections. MATERIALS AND METHODS: In this study, we deciphered the effects of Staphylococcus aureus toxins α-hemolysin, LukAB, LukDE, and LukSF on human platelets and compared the effects with the pore forming toxin pneumolysin of Streptococcus pneumoniae. Activation of platelets and loss of platelet function were investigated by flow cytometry, aggregometry, platelet viability, fluorescence microscopy, and intracellular calcium release. Thrombus formation was assessed in whole blood. RESULTS: α-hemolysin (Hla) is known to be a pore-forming toxin. Hla-induced calcium influx initially activates platelets as indicated by CD62P and αIIbß3 integrin activation, but also induces finally alterations in the phenotype of platelets. In contrast to Hla and pneumolysin, S. aureus bicomponent pore-forming leukocidins LukAB, LukED, and LukSF do not bind to platelets and had no significant effect on platelet activation and viability. The presence of small amounts of Hla (0.2 µg/ml) in whole blood abrogates thrombus formation indicating that in systemic infections with S. aureus the stability of formed thrombi is impaired. Damage of platelets by Hla was not neutralized by intravenous immune globulins. CONCLUSION: Our findings might be of clinical relevance for S. aureus induced endocarditis. Stabilizing the aortic-valve thrombi by inhibiting Hla-induced impairment of platelets might reduce the risk for septic (micro-)embolization.


Asunto(s)
Infecciones Estafilocócicas , Trombosis , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/farmacología , Calcio , Proteínas Hemolisinas/metabolismo , Proteínas Hemolisinas/farmacología , Humanos , Leucocidinas/metabolismo , Staphylococcus aureus
5.
Thromb Haemost ; 122(7): 1147-1158, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-34918314

RESUMEN

Platelets play an important role in the development and progression of respiratory distress. Functional platelets are known to seal inflammatory endothelial gaps and loss of platelet function has been shown to result in loss of integrity of pulmonary vessels. This leads to fluid accumulation in the pulmonary interstitium, eventually resulting in respiratory distress. Streptococcus pneumoniae is one of the major pathogens causing community-acquired pneumonia. Previously, we have shown that its major toxin pneumolysin forms pores in platelet membranes and renders them nonfunctional. In vitro, this process was inhibited by polyvalent intravenous immunoglobulins (IVIGs). In this study, we compared the efficacy of a standard IVIG preparation (IVIG, 98% immunoglobulin G [IgG]; Privigen, CSL Behring, United States) and an IgM/IgA-enriched immunoglobulin preparation (21% IgA, 23% IgM, 56% IgG; trimodulin, Biotest AG, Germany) to inhibit pneumolysin-induced platelet destruction. Platelet destruction and functionality were assessed by flow cytometry, intracellular calcium release, aggregometry, platelet viability, transwell, and flow chamber assays. Overall, both immunoglobulin preparations efficiently inhibited pneumolysin-induced platelet destruction. The capacity to antagonize pneumolysin mainly depended on the final IgG content. As both polyvalent immunoglobulin preparations efficiently prevent pneumolysin-induced platelet destruction and maintain platelet function in vitro, they represent promising candidates for clinical studies on supportive treatment of pneumococcal pneumonia to reduce progression of respiratory distress.


Asunto(s)
Inmunoglobulinas Intravenosas , Síndrome de Dificultad Respiratoria , Proteínas Bacterianas/farmacología , Humanos , Inmunoglobulina A , Inmunoglobulina G , Inmunoglobulina M , Inmunoglobulinas Intravenosas/farmacología , Estreptolisinas
6.
Blood Adv ; 4(24): 6315-6326, 2020 12 22.
Artículo en Inglés | MEDLINE | ID: mdl-33351126

RESUMEN

Community-acquired pneumonia by primary or superinfections with Streptococcus pneumoniae can lead to acute respiratory distress requiring mechanical ventilation. The pore-forming toxin pneumolysin alters the alveolar-capillary barrier and causes extravasation of protein-rich fluid into the interstitial pulmonary tissue, which impairs gas exchange. Platelets usually prevent endothelial leakage in inflamed pulmonary tissue by sealing inflammation-induced endothelial gaps. We not only confirm that S pneumoniae induces CD62P expression in platelets, but we also show that, in the presence of pneumolysin, CD62P expression is not associated with platelet activation. Pneumolysin induces pores in the platelet membrane, which allow anti-CD62P antibodies to stain the intracellular CD62P without platelet activation. Pneumolysin treatment also results in calcium efflux, increase in light transmission by platelet lysis (not aggregation), loss of platelet thrombus formation in the flow chamber, and loss of pore-sealing capacity of platelets in the Boyden chamber. Specific anti-pneumolysin monoclonal and polyclonal antibodies inhibit these effects of pneumolysin on platelets as do polyvalent human immunoglobulins. In a post hoc analysis of the prospective randomized phase 2 CIGMA trial, we show that administration of a polyvalent immunoglobulin preparation was associated with a nominally higher platelet count and nominally improved survival in patients with severe S pneumoniae-related community-acquired pneumonia. Although, due to the low number of patients, no definitive conclusion can be made, our findings provide a rationale for investigation of pharmacologic immunoglobulin preparations to target pneumolysin by polyvalent immunoglobulin preparations in severe community-acquired pneumococcal pneumonia, to counteract the risk of these patients becoming ventilation dependent. This trial was registered at www.clinicaltrials.gov as #NCT01420744.


Asunto(s)
Activación Plaquetaria , Estreptolisinas , Proteínas Bacterianas , Humanos , Inmunoglobulinas , Estudios Prospectivos
8.
J Virol ; 92(17)2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-29950419

RESUMEN

The flexible regulation of cellular metabolic pathways enables cellular adaptation to changes in energy demand under conditions of stress such as posed by a virus infection. To analyze such an impact on cellular metabolism, rubella virus (RV) was used in this study. RV replication under selected substrate supplementation with glucose, pyruvate, and glutamine as essential nutrients for mammalian cells revealed its requirement for glutamine. The assessment of the mitochondrial respiratory (based on the oxygen consumption rate) and glycolytic (based on the extracellular acidification rate) rate and capacity by respective stress tests through Seahorse technology enabled determination of the bioenergetic phenotype of RV-infected cells. Irrespective of the cellular metabolic background, RV infection induced a shift of the bioenergetic state of epithelial cells (Vero and A549) and human umbilical vein endothelial cells to a higher oxidative and glycolytic level. Interestingly there was a RV strain-specific, but genotype-independent demand for glutamine to induce a significant increase in metabolic activity. While glutaminolysis appeared to be rather negligible for RV replication, glutamine could serve as donor of its amide nitrogen in biosynthesis pathways for important metabolites. This study suggests that the capacity of RVs to induce metabolic alterations could evolve differently during natural infection. Thus, changes in cellular bioenergetics represent an important component of virus-host interactions and could complement our understanding of the viral preference for a distinct host cell population.IMPORTANCE RV pathologies, especially during embryonal development, could be connected with its impact on mitochondrial metabolism. With bioenergetic phenotyping we pursued a rather novel approach in virology. For the first time it was shown that a virus infection could shift the bioenergetics of its infected host cell to a higher energetic state. Notably, the capacity to induce such alterations varied among different RV isolates. Thus, our data add viral adaptation of cellular metabolic activity to its specific needs as a novel aspect to virus-host evolution. In addition, this study emphasizes the implementation of different viral strains in the study of virus-host interactions and the use of bioenergetic phenotyping of infected cells as a biomarker for virus-induced pathological alterations.


Asunto(s)
Metabolismo Energético , Glutamina/metabolismo , Glucólisis/efectos de los fármacos , Consumo de Oxígeno/fisiología , Virus de la Rubéola/metabolismo , Células A549 , Células Endoteliales/metabolismo , Células Endoteliales/virología , Glucosa/metabolismo , Glucosa/farmacología , Glutamina/farmacología , Homeostasis , Humanos , Quinurenina/metabolismo , Redes y Vías Metabólicas/efectos de los fármacos , Mitocondrias/metabolismo , Nucleótidos/biosíntesis , Oxidación-Reducción , Estrés Oxidativo , Consumo de Oxígeno/efectos de los fármacos , Fenotipo , Ácido Pirúvico/metabolismo , Ácido Pirúvico/farmacología , Replicación Viral/efectos de los fármacos
9.
ACS Infect Dis ; 3(12): 886-897, 2017 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-29043768

RESUMEN

Human induced pluripotent stem cell (iPSC) lines are a promising model for the early phase of human embryonic development. Here, their contribution to the still incompletely understood pathogenesis of congenital virus infections was evaluated. The infection of iPSC lines with miscarriage-associated coxsackievirus B3 (CVB3) and measles virus (MV) was compared to the efficient teratogen rubella virus (RV). While CVB3 and MV were found to be cytopathogenic on iPSC lines, RV replicated without impairment of iPSC colony morphology and integrity. This so far outstanding course of infection enabled maintenance of RV-infected iPSC cultures over several passages and their subsequent differentiation to ectoderm, endoderm, and mesoderm. A modification of the metabolic profile of infected iPSC lines was the only common aspect for all three viruses. This study points toward two important aspects. First, iPSC lines represent a suitable cell culture model for early embryonic virus infection. Second, metabolic activity represents an important means for evaluation of pathogen-associated alterations in iPSC lines.


Asunto(s)
Aborto Espontáneo/etiología , Desarrollo Embrionario , Enterovirus Humano B/patogenicidad , Células Madre Pluripotentes Inducidas/virología , Virus del Sarampión/patogenicidad , Virus de la Rubéola/patogenicidad , Teratogénesis , Animales , Caspasas/fisiología , Diferenciación Celular , Supervivencia Celular , Células Cultivadas , Humanos , Células Madre Pluripotentes Inducidas/fisiología , Replicación Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA