Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
PLoS Pathog ; 19(1): e1011023, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36696456

RESUMEN

Pseudomonas aeruginosa, an opportunistic Gram-negative pathogen, is a leading cause of bacteremia with a high mortality rate. We recently reported that P. aeruginosa forms a persister-like sub-population of evaders in human plasma. Here, using a gain-of-function transposon sequencing (Tn-seq) screen in plasma, we identified and validated previously unknown factors affecting bacterial persistence in plasma. Among them, we identified a small periplasmic protein, named SrgA, whose expression leads to up to a 100-fold increase in resistance to killing. Additionally, mutants in pur and bio genes displayed higher tolerance and persistence, respectively. Analysis of several steps of the complement cascade and exposure to an outer-membrane-impermeable drug, nisin, suggested that the mutants impede membrane attack complex (MAC) activity per se. Electron microscopy combined with energy-dispersive X-ray spectroscopy (EDX) revealed the formation of polyphosphate (polyP) granules upon incubation in plasma of different size in purD and wild-type strains, implying the bacterial response to a stress signal. Indeed, inactivation of ppk genes encoding polyP-generating enzymes lead to significant elimination of persisting bacteria from plasma. Through this study, we shed light on a complex P. aeruginosa response to the plasma conditions and discovered the multifactorial origin of bacterial resilience to MAC-induced killing.


Asunto(s)
Antibacterianos , Pseudomonas aeruginosa , Humanos , Antibacterianos/farmacología , Pseudomonas aeruginosa/genética , Proteínas del Sistema Complemento , Complejo de Ataque a Membrana del Sistema Complemento
2.
J Biol Chem ; 299(8): 104956, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37356719

RESUMEN

The human complement system plays a crucial role in immune defense. However, its erroneous activation contributes to many serious inflammatory diseases. Since most unwanted complement effector functions result from C5 cleavage into C5a and C5b, development of C5 inhibitors, such as clinically approved monoclonal antibody eculizumab, are of great interest. Here, we developed and characterized two anti-C5 nanobodies, UNbC5-1 and UNbC5-2. Using surface plasmon resonance, we determined a binding affinity of 119.9 pM for UNbC5-1 and 7.7 pM for UNbC5-2. Competition experiments determined that the two nanobodies recognize distinct epitopes on C5. Both nanobodies efficiently interfered with C5 cleavage in a human serum environment, as they prevented red blood cell lysis via membrane attack complexes (C5b-9) and the formation of chemoattractant C5a. The cryo-EM structure of UNbC5-1 and UNbC5-2 in complex with C5 (3.6 Å resolution) revealed that the binding interfaces of UNbC5-1 and UNbC5-2 overlap with known complement inhibitors eculizumab and RaCI3, respectively. UNbC5-1 binds to the MG7 domain of C5, facilitated by a hydrophobic core and polar interactions, and UNbC5-2 interacts with the C5d domain mostly by salt bridges and hydrogen bonds. Interestingly, UNbC5-1 potently binds and inhibits C5 R885H, a genetic variant of C5 that is not recognized by eculizumab. Altogether, we identified and characterized two different, high affinity nanobodies against human C5. Both nanobodies could serve as diagnostic and/or research tools to detect C5 or inhibit C5 cleavage. Furthermore, the residues targeted by UNbC5-1 hold important information for therapeutic inhibition of different polymorphic variants of C5.


Asunto(s)
Anticuerpos Monoclonales , Complemento C5 , Anticuerpos de Dominio Único , Humanos , Activación de Complemento , Complemento C5/antagonistas & inhibidores , Complemento C5/genética , Complejo de Ataque a Membrana del Sistema Complemento , Proteínas del Sistema Complemento/metabolismo
4.
EMBO J ; 38(4)2019 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-30643019

RESUMEN

The immune system kills bacteria by the formation of lytic membrane attack complexes (MACs), triggered when complement enzymes cleave C5. At present, it is not understood how the MAC perturbs the composite cell envelope of Gram-negative bacteria. Here, we show that the role of C5 convertase enzymes in MAC assembly extends beyond the cleavage of C5 into the MAC precursor C5b. Although purified MAC complexes generated from preassembled C5b6 perforate artificial lipid membranes and mammalian cells, these components lack bactericidal activity. In order to permeabilize both the bacterial outer and inner membrane and thus kill a bacterium, MACs need to be assembled locally by the C5 convertase enzymes. Our data indicate that C5b6 rapidly loses the capacity to form bactericidal pores; therefore, bacterial killing requires both in situ conversion of C5 and immediate insertion of C5b67 into the membrane. Using flow cytometry and atomic force microscopy, we show that local assembly of C5b6 at the bacterial surface is required for the efficient insertion of MAC pores into bacterial membranes. These studies provide basic molecular insights into MAC assembly and bacterial killing by the immune system.


Asunto(s)
Actividad Bactericida de la Sangre , Membrana Celular/metabolismo , Convertasas de Complemento C3-C5/metabolismo , Complejo de Ataque a Membrana del Sistema Complemento/metabolismo , Bacterias Gramnegativas/crecimiento & desarrollo , Hemólisis , Permeabilidad de la Membrana Celular , Activación de Complemento , Bacterias Gramnegativas/metabolismo , Humanos
5.
PLoS Pathog ; 17(1): e1009227, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33481964

RESUMEN

Infections with Gram-negative bacteria form an increasing risk for human health due to antibiotic resistance. Our immune system contains various antimicrobial proteins that can degrade the bacterial cell envelope. However, many of these proteins do not function on Gram-negative bacteria, because the impermeable outer membrane of these bacteria prevents such components from reaching their targets. Here we show that complement-dependent formation of Membrane Attack Complex (MAC) pores permeabilizes this barrier, allowing antimicrobial proteins to cross the outer membrane and exert their antimicrobial function. Specifically, we demonstrate that MAC-dependent outer membrane damage enables human lysozyme to degrade the cell wall of E. coli. Using flow cytometry and confocal microscopy, we show that the combination of MAC pores and lysozyme triggers effective E. coli cell wall degradation in human serum, thereby altering the bacterial cell morphology from rod-shaped to spherical. Completely assembled MAC pores are required to sensitize E. coli to the antimicrobial actions of lysozyme and other immune factors, such as Human Group IIA-secreted Phospholipase A2. Next to these effects in a serum environment, we observed that the MAC also sensitizes E. coli to more efficient degradation and killing inside human neutrophils. Altogether, this study serves as a proof of principle on how different players of the human immune system can work together to degrade the complex cell envelope of Gram-negative bacteria. This knowledge may facilitate the development of new antimicrobials that could stimulate or work synergistically with the immune system.


Asunto(s)
Antiinfecciosos/farmacología , Membrana Externa Bacteriana/efectos de los fármacos , Activación de Complemento , Complejo de Ataque a Membrana del Sistema Complemento/metabolismo , Bacterias Gramnegativas/efectos de los fármacos , Antibacterianos/farmacología , Pared Celular/efectos de los fármacos , Escherichia coli/efectos de los fármacos , Escherichia coli/inmunología , Citometría de Flujo , Bacterias Gramnegativas/inmunología , Fosfolipasas A2 Grupo II/metabolismo , Humanos , Microscopía Confocal , Muramidasa/metabolismo , Neutrófilos/microbiología , Fagocitos/microbiología
6.
PLoS Pathog ; 17(11): e1010051, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34752492

RESUMEN

Complement proteins can form membrane attack complex (MAC) pores that directly kill Gram-negative bacteria. MAC pores assemble by stepwise binding of C5b, C6, C7, C8 and finally C9, which can polymerize into a transmembrane ring of up to 18 C9 monomers. It is still unclear if the assembly of a polymeric-C9 ring is necessary to sufficiently damage the bacterial cell envelope to kill bacteria. In this paper, polymerization of C9 was prevented without affecting binding of C9 to C5b-8, by locking the first transmembrane helix domain of C9. Using this system, we show that polymerization of C9 strongly enhanced damage to both the bacterial outer and inner membrane, resulting in more rapid killing of several Escherichia coli and Klebsiella strains in serum. By comparing binding of wildtype and 'locked' C9 by flow cytometry, we also show that polymerization of C9 is impaired when the amount of available C9 per C5b-8 is limited. This suggests that an excess of C9 is required to efficiently form polymeric-C9. Finally, we show that polymerization of C9 was impaired on complement-resistant E. coli strains that survive killing by MAC pores. This suggests that these bacteria can specifically block polymerization of C9. All tested complement-resistant E. coli expressed LPS O-antigen (O-Ag), compared to only one out of four complement-sensitive E. coli. By restoring O-Ag expression in an O-Ag negative strain, we show that the O-Ag impairs polymerization of C9 and results in complement-resistance. Altogether, these insights are important to understand how MAC pores kill bacteria and how bacterial pathogens can resist MAC-dependent killing.


Asunto(s)
Actividad Bactericida de la Sangre , Pared Celular/patología , Complemento C9/química , Complejo de Ataque a Membrana del Sistema Complemento/farmacología , Escherichia coli/crecimiento & desarrollo , Klebsiella/crecimiento & desarrollo , Polimerizacion , Pared Celular/efectos de los fármacos , Escherichia coli/efectos de los fármacos , Infecciones por Escherichia coli/tratamiento farmacológico , Infecciones por Escherichia coli/microbiología , Humanos , Klebsiella/efectos de los fármacos , Infecciones por Klebsiella/tratamiento farmacológico , Infecciones por Klebsiella/microbiología
7.
Chembiochem ; 23(19): e202200340, 2022 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-35877976

RESUMEN

The interactions between bacteria and their host often rely on recognition processes that involve host or bacterial glycans. Glycoengineering techniques make it possible to modify and study the glycans on the host's eukaryotic cells, but only a few are available for the study of bacterial glycans. Here, we have adapted selective exoenzymatic labeling (SEEL), a chemical reporter strategy, to label the lipooligosaccharides of the bacterial pathogen Neisseria gonorrhoeae, using the recombinant glycosyltransferase ST6Gal1, and three synthetic CMP-sialic acid derivatives. We show that SEEL treatment does not affect cell viability and can introduce an α2,6-linked sialic acid with a reporter group on the lipooligosaccharides by Western blot, flow cytometry and fluorescent microscopy. This new bacterial glycoengineering technique allows for the precise modification, here with α2,6-sialoside derivatives, and direct detection of specific surface glycans on live bacteria, which will aid in further unravelling the precise biological functions of bacterial glycans.


Asunto(s)
Ácido N-Acetilneuramínico Citidina Monofosfato , Neisseria gonorrhoeae , Ácido N-Acetilneuramínico Citidina Monofosfato/metabolismo , Glicosiltransferasas/metabolismo , Lipopolisacáridos , Ácido N-Acetilneuramínico , Polisacáridos Bacterianos/metabolismo , Ácidos Siálicos/metabolismo
8.
PLoS Pathog ; 16(6): e1008606, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32569291

RESUMEN

An important effector function of the human complement system is to directly kill Gram-negative bacteria via Membrane Attack Complex (MAC) pores. MAC pores are assembled when surface-bound convertase enzymes convert C5 into C5b, which together with C6, C7, C8 and multiple copies of C9 forms a transmembrane pore that damages the bacterial cell envelope. Recently, we found that bacterial killing by MAC pores requires local conversion of C5 by surface-bound convertases. In this study we aimed to understand why local assembly of MAC pores is essential for bacterial killing. Here, we show that rapid interaction of C7 with C5b6 is required to form bactericidal MAC pores on Escherichia coli. Binding experiments with fluorescently labelled C6 show that C7 prevents release of C5b6 from the bacterial surface. Moreover, trypsin shaving experiments and atomic force microscopy revealed that this rapid interaction between C7 and C5b6 is crucial to efficiently anchor C5b-7 to the bacterial cell envelope and form complete MAC pores. Using complement-resistant clinical E. coli strains, we show that bacterial pathogens can prevent complement-dependent killing by interfering with the anchoring of C5b-7. While C5 convertase assembly was unaffected, these resistant strains blocked efficient anchoring of C5b-7 and thus prevented stable insertion of MAC pores into the bacterial cell envelope. Altogether, these findings provide basic molecular insights into how bactericidal MAC pores are assembled and how bacteria evade MAC-dependent killing.


Asunto(s)
Actividad Bactericida de la Sangre , Membrana Celular/metabolismo , Pared Celular/metabolismo , Complemento C5/metabolismo , Complejo de Ataque a Membrana del Sistema Complemento/metabolismo , Escherichia coli/metabolismo , Proteínas del Sistema Complemento/metabolismo , Células HEK293 , Humanos
9.
Mol Plant Microbe Interact ; 27(7): 603-10, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24654978

RESUMEN

Bacterial flagellin molecules are strong inducers of innate immune responses in both mammals and plants. The opportunistic pathogen Pseudomonas aeruginosa secretes an alkaline protease called AprA that degrades flagellin monomers. Here, we show that AprA is widespread among a wide variety of bacterial species. In addition, we investigated the role of AprA in virulence of the bacterial plant pathogen P. syringae pv. tomato DC3000. The AprA-deficient DC3000 ΔaprA knockout mutant was significantly less virulent on both tomato and Arabidopsis thaliana. Moreover, infiltration of A. thaliana Col-0 leaves with DC3000 ΔaprA evoked a significantly higher level of expression of the defense-related genes FRK1 and PR-1 than did wild-type DC3000. In the flagellin receptor mutant fls2, pathogen virulence and defense-related gene activation did not differ between DC3000 and DC3000 ΔaprA. Together, these results suggest that AprA of DC3000 is important for evasion of recognition by the FLS2 receptor, allowing wild-type DC3000 to be more virulent on its host plant than AprA-deficient DC3000 ΔaprA. To provide further evidence for the role of DC3000 AprA in host immune evasion, we overexpressed the AprA inhibitory peptide AprI of DC3000 in A. thaliana to counteract the immune evasive capacity of DC3000 AprA. Ectopic expression of aprI in A. thaliana resulted in an enhanced level of resistance against wild-type DC3000, while the already elevated level of resistance against DC3000 ΔaprA remained unchanged. Together, these results indicate that evasion of host immunity by the alkaline protease AprA is important for full virulence of strain DC3000 and likely acts by preventing flagellin monomers from being recognized by its cognate immune receptor.


Asunto(s)
Arabidopsis/microbiología , Flagelina/metabolismo , Regulación Bacteriana de la Expresión Génica/fisiología , Pseudomonas syringae/fisiología , Serina Endopeptidasas/metabolismo , Solanum lycopersicum/microbiología , Regulación Enzimológica de la Expresión Génica , Enfermedades de las Plantas/inmunología , Enfermedades de las Plantas/microbiología , Pseudomonas syringae/inmunología , Serina Endopeptidasas/genética , Factores de Virulencia/genética , Factores de Virulencia/metabolismo
10.
J Immunol ; 188(1): 386-93, 2012 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-22131330

RESUMEN

The complement system rapidly detects and kills Gram-negative bacteria and supports bacterial killing by phagocytes. However, bacterial pathogens exploit several strategies to evade detection by the complement system. The alkaline protease (AprA) of Pseudomonas aeruginosa has been associated with bacterial virulence and is known to interfere with complement-mediated lysis of erythrocytes, but its exact role in bacterial complement escape is unknown. In this study, we analyzed how AprA interferes with complement activation and whether it could block complement-dependent neutrophil functions. We found that AprA potently blocked phagocytosis and killing of Pseudomonas by human neutrophils. Furthermore, AprA inhibited opsonization of bacteria with C3b and the formation of the chemotactic agent C5a. AprA specifically blocked C3b deposition via the classical and lectin pathways, whereas the alternative pathway was not affected. Serum degradation assays revealed that AprA degrades both human C1s and C2. However, repletion assays demonstrated that the mechanism of action for complement inhibition is cleavage of C2. In summary, we showed that P. aeruginosa AprA interferes with classical and lectin pathway-mediated complement activation via cleavage of C2.


Asunto(s)
Proteínas Bacterianas/inmunología , Exopeptidasas/inmunología , Neutrófilos/inmunología , Infecciones por Pseudomonas/inmunología , Pseudomonas aeruginosa/inmunología , Pseudomonas aeruginosa/patogenicidad , Factores de Virulencia/inmunología , Proteínas Bacterianas/metabolismo , Complemento C2/inmunología , Complemento C2/metabolismo , Complemento C3b/inmunología , Complemento C3b/metabolismo , Complemento C5a/inmunología , Complemento C5a/metabolismo , Lectina de Unión a Manosa de la Vía del Complemento , Exopeptidasas/metabolismo , Humanos , Evasión Inmune , Neutrófilos/metabolismo , Fagocitosis/inmunología , Infecciones por Pseudomonas/enzimología , Pseudomonas aeruginosa/enzimología , Factores de Virulencia/metabolismo
11.
PLoS Pathog ; 7(8): e1002206, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21901099

RESUMEN

The building blocks of bacterial flagella, flagellin monomers, are potent stimulators of host innate immune systems. Recognition of flagellin monomers occurs by flagellin-specific pattern-recognition receptors, such as Toll-like receptor 5 (TLR5) in mammals and flagellin-sensitive 2 (FLS2) in plants. Activation of these immune systems via flagellin leads eventually to elimination of the bacterium from the host. In order to prevent immune activation and thus favor survival in the host, bacteria secrete many proteins that hamper such recognition. In our search for Toll like receptor (TLR) antagonists, we screened bacterial supernatants and identified alkaline protease (AprA) of Pseudomonas aeruginosa as a TLR5 signaling inhibitor as evidenced by a marked reduction in IL-8 production and NF-κB activation. AprA effectively degrades the TLR5 ligand monomeric flagellin, while polymeric flagellin (involved in bacterial motility) and TLR5 itself resist degradation. The natural occurring alkaline protease inhibitor AprI of P. aeruginosa blocked flagellin degradation by AprA. P. aeruginosa aprA mutants induced an over 100-fold enhanced activation of TLR5 signaling, because they fail to degrade excess monomeric flagellin in their environment. Interestingly, AprA also prevents flagellin-mediated immune responses (such as growth inhibition and callose deposition) in Arabidopsis thaliana plants. This was due to decreased activation of the receptor FLS2 and clearly demonstrated by delayed stomatal closure with live bacteria in plants. Thus, by degrading the ligand for TLR5 and FLS2, P. aeruginosa escapes recognition by the innate immune systems of both mammals and plants.


Asunto(s)
Proteínas Bacterianas/metabolismo , Endopeptidasas/metabolismo , Flagelina/inmunología , Inmunidad Innata , Inmunidad de la Planta , Pseudomonas aeruginosa/inmunología , Animales , Proteínas de Arabidopsis/antagonistas & inhibidores , Proteínas de Arabidopsis/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/inmunología , Bovinos , Línea Celular , Endopeptidasas/genética , Endopeptidasas/inmunología , Flagelina/metabolismo , Humanos , Interleucina-8/metabolismo , Mutación , FN-kappa B/metabolismo , Proteínas Quinasas/metabolismo , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/metabolismo , Transducción de Señal , Receptor Toll-Like 5/antagonistas & inhibidores , Receptor Toll-Like 5/metabolismo
12.
Cell Microbiol ; 14(6): 902-13, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22309196

RESUMEN

Phagocytosis by neutrophils is the essential step in fighting Pseudomonas infections. The first step in neutrophil recruitment to the site infection is the interaction of P-selectin (on endothelial cells) with P-selectin glycoprotein ligand-1 (PSGL-1) on neutrophils. Pseudomonas aeruginosa secretes various proteases that degrade proteins that are essential for host defence, such as elastase and alkaline protease. Here we identify PA0572 of P. aeruginosa as an inhibitor of PSGL-1 and named this secreted hypothetical protease immunomodulating metalloprotease of P. aeruginosa or IMPa. Proteolytic activity was confirmed by cleavage of recombinant and cell-surface expressed PSGL-1. Functional inhibition was demonstrated by impaired PSGL-1-mediated rolling of IMPa-treated neutrophils under flow conditions. Next to PSGL-1, IMPa targets CD43 and CD44 that are also involved in leucocyte homing. These data indicate that IMPa prevents neutrophil extravasation and thereby protects P. aeruginosa from neutrophil attack.


Asunto(s)
Proteínas Bacterianas/metabolismo , Inmunomodulación , Metaloproteasas/metabolismo , Pseudomonas aeruginosa/enzimología , Secuencias de Aminoácidos , Proteínas Bacterianas/química , Proteínas Bacterianas/fisiología , Dominio Catalítico , Adhesión Celular , Células Cultivadas , Secuencia Conservada , Medios de Cultivo Condicionados/química , Interacciones Huésped-Patógeno , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Rodamiento de Leucocito , Leucosialina/metabolismo , Glicoproteínas de Membrana/antagonistas & inhibidores , Glicoproteínas de Membrana/metabolismo , Metaloproteasas/química , Metaloproteasas/fisiología , Neutrófilos/fisiología , Selectina-P/metabolismo , Unión Proteica , Pseudomonas aeruginosa/fisiología , Análisis de Secuencia de Proteína , Ácidos Siálicos/metabolismo
13.
Sci Rep ; 13(1): 856, 2023 01 16.
Artículo en Inglés | MEDLINE | ID: mdl-36646746

RESUMEN

Bacteriophages (phages) are viruses that specifically attack bacteria. Their use as therapeutics, which constitutes a promising alternative to antibiotics, heavily relies on selecting effective lytic phages against the pathogen of interest. Current selection techniques are laborious and do not allow for direct visualization of phage infection dynamics. Here, we present a method that circumvents these limitations. It can be scaled for high-throughput and permits monitoring of the phage infection in real time via a fluorescence signal readout. This is achieved through the use of a membrane-impermeant nucleic acid dye that stains the DNA of damaged or lysed bacteria and new phage progeny. We have tested the method on Pseudomonas aeruginosa and Klebsiella pneumoniae and show that an increase in fluorescence reflects phage-mediated killing. This is confirmed by other techniques including spot tests, colony plating, flow cytometry and metabolic activity measurements. Furthermore, we illustrate how our method may be used to compare the activity of different phages and to screen the susceptibility of clinical isolates to phage. Altogether, we present a fast, reliable way of selecting phages against Gram-negative bacteria, which may be valuable in optimizing the process of selecting phages for therapeutic use.


Asunto(s)
Bacteriófagos , Colorantes Fluorescentes , Bacteriófagos/genética , Bacterias , Antibacterianos , ADN
14.
Viruses ; 15(11)2023 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-38005888

RESUMEN

Therapeutic bacteriophages (phages) are primarily chosen based on their in vitro bacteriolytic activity. Although anti-phage antibodies are known to inhibit phage infection, the influence of other immune system components is less well known. An important anti-bacterial and anti-viral innate immune system that may interact with phages is the complement system, a cascade of proteases that recognizes and targets invading microorganisms. In this research, we aimed to study the effects of serum components such as complement on the infectivity of different phages targeting Pseudomonas aeruginosa. We used a fluorescence-based assay to monitor the killing of P. aeruginosa by phages of different morphotypes in the presence of human serum. Our results reveal that several myophages are inhibited by serum in a concentration-dependent way, while the activity of four podophages and one siphophage tested in this study is not affected by serum. By using specific nanobodies blocking different components of the complement cascade, we showed that activation of the classical complement pathway is a driver of phage inhibition. To determine the mechanism of inhibition, we produced bioorthogonally labeled fluorescent phages to study their binding by means of microscopy and flow cytometry. We show that phage adsorption is hampered in the presence of active complement. Our results indicate that interactions with complement may affect the in vivo activity of therapeutically administered phages. A better understanding of this phenomenon is essential to optimize the design and application of therapeutic phage cocktails.


Asunto(s)
Bacteriófagos , Infecciones por Pseudomonas , Fagos Pseudomonas , Humanos , Pseudomonas aeruginosa/fisiología , Fagos Pseudomonas/fisiología , Bacteriólisis , Infecciones por Pseudomonas/terapia , Infecciones por Pseudomonas/microbiología
15.
Sci Rep ; 13(1): 12618, 2023 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-37537263

RESUMEN

Due to multi-drug resistance, physicians increasingly use the last-resort antibiotic colistin to treat infections with the Gram-negative bacterium Klebsiella pneumoniae. Unfortunately, K. pneumoniae can also develop colistin resistance. Interestingly, colistin resistance has dual effects on bacterial clearance by the immune system. While it increases resistance to antimicrobial peptides, colistin resistance has been reported to sensitize certain bacteria for killing by human serum. Here we investigate the mechanisms underlying this increased serum sensitivity, focusing on human complement which kills Gram-negatives via membrane attack complex (MAC) pores. Using in vitro evolved colistin resistant strains and a fluorescent MAC-mediated permeabilization assay, we showed that two of the three tested colistin resistant strains, Kp209_CSTR and Kp257_CSTR, were sensitized to MAC. Transcriptomic and mechanistic analyses focusing on Kp209_CSTR revealed that a mutation in the phoQ gene locked PhoQ in an active state, making Kp209_CSTR colistin resistant and MAC sensitive. Detailed immunological assays showed that complement activation on Kp209_CSTR in human serum required specific IgM antibodies that bound Kp209_CSTR but did not recognize the wild-type strain. Together, our results show that developing colistin resistance affected recognition of Kp209_CSTR and its killing by the immune system.


Asunto(s)
Colistina , Infecciones por Klebsiella , Humanos , Colistina/farmacología , Colistina/uso terapéutico , Klebsiella pneumoniae/genética , Proteínas Bacterianas/farmacología , Farmacorresistencia Bacteriana/genética , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Mutación , Inmunoglobulina M/genética , Infecciones por Klebsiella/tratamiento farmacológico , Infecciones por Klebsiella/microbiología , Pruebas de Sensibilidad Microbiana
16.
Sci Rep ; 13(1): 18836, 2023 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-37914798

RESUMEN

Antibodies play a key role in the immune defence against Gram-negative bacteria. After binding to bacterial surface antigens, IgG and IgM can activate the complement system and trigger formation of lytic membrane attack complex (MAC) pores. Molecular studies to compare functional activity of antibodies on bacteria are hampered by the limited availability of well-defined antibodies against bacterial surface antigens. Therefore, we genetically engineered E. coli by expressing the StrepTagII antigen into outer membrane protein X (OmpX) and validated that these engineered bacteria were recognised by anti-StrepTagII antibodies. We then combined this antigen-antibody system with a purified complement assay to avoid interference of serum components and directly compare MAC-mediated bacterial killing via IgG1 and pentameric IgM. While both IgG1 and IgM could induce MAC-mediated killing, we show that IgM has an increased capacity to induce complement-mediated killing of E. coli compared to IgG1. While Fc mutations that enhance IgG clustering after target binding could not improve MAC formation, mutations that cause formation of pre-assembled IgG hexamers enhanced the complement activating capacity of IgG1. Altogether, we here present a system to study antibody-dependent complement activation on E. coli and show IgM's enhanced capacity over IgG to induce complement-mediated lysis of E. coli.


Asunto(s)
Anticuerpos Monoclonales , Escherichia coli , Escherichia coli/metabolismo , Anticuerpos Monoclonales/metabolismo , Proteínas del Sistema Complemento/metabolismo , Complejo de Ataque a Membrana del Sistema Complemento/metabolismo , Activación de Complemento , Inmunoglobulina G , Antígenos de Superficie/metabolismo , Inmunoglobulina M/metabolismo
17.
J Glob Antimicrob Resist ; 30: 406-413, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35840108

RESUMEN

OBJECTIVES: Our group recently developed a new group of antimicrobial peptides termed PepBiotics, of which peptides CR-163 and CR-172 showed optimized antibacterial activity against Pseudomonas aeruginosa and Staphylococcus aureus without inducing antimicrobial resistance. In this study, the antibacterial mechanism of action and the immunomodulatory activity of these two PepBiotics was explored. METHODS: RAW264.7 cells were used to determine the ability of PepBiotics to neutralize Lipopolysaccharide (LPS)-and Lipoteichoic acid (LTA)-induced activation of macrophages. Isothermal titration calorimetry and competition assays with dansyl-labeled polymyxin B determined binding characteristics to LPS and LTA. Combined bacterial killing with subsequent macrophage activation assays was performed to determine so-called 'silent killing'. Finally, flow cytometry of peptide-treated genetically engineered Escherichia coli expressing Green Fluorescent Protein (GFP) and mCherry in the cytoplasm and periplasm, respectively, further established the antimicrobial mechanism of PepBiotics. RESULTS: Both CR-163 and CR-172 were shown to have broad-spectrum activity against ESKAPE pathogens and E. coli using a membranolytic mechanism of action. PepBiotics could exothermically bind LPS/LTA and were able to replace polymyxin B. Finally, it was demonstrated that bacteria killed by PepBiotics were less prone to stimulate immune cells, contrary to gentamicin and heat-killed bacteria that still elicited a strong immune response. CONCLUSIONS: These studies highlight the multifunctional nature of the two peptide antibiotics as both broad-spectrum antimicrobial and immunomodulator. Their ability to kill bacteria and reduce unwanted subsequent immune activation is a major advantage and highlights their potential for future therapeutic use.


Asunto(s)
Antiinfecciosos , Lipopolisacáridos , Animales , Antibacterianos/farmacología , Escherichia coli/genética , Escherichia coli/metabolismo , Inmunidad , Ratones , Péptidos/farmacología , Polimixina B/farmacología , Células RAW 264.7
18.
Elife ; 112022 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-35947526

RESUMEN

The membrane attack complex (MAC or C5b-9) is an important effector of the immune system to kill invading microbes. MAC formation is initiated when complement enzymes on the bacterial surface convert complement component C5 into C5b. Although the MAC is a membrane-inserted complex, soluble forms of MAC (sMAC), or terminal complement complex (TCC), are often detected in sera of patients suffering from infections. Consequently, sMAC has been proposed as a biomarker, but it remains unclear when and how it is formed during infections. Here, we studied mechanisms of MAC formation on different Gram-negative and Gram-positive bacteria and found that sMAC is primarily formed in human serum by bacteria resistant to MAC-dependent killing. Surprisingly, C5 was converted into C5b more potently by MAC-resistant compared to MAC-sensitive Escherichia coli strains. In addition, we found that MAC precursors are released from the surface of MAC-resistant bacteria during MAC assembly. Although release of MAC precursors from bacteria induced lysis of bystander human erythrocytes, serum regulators vitronectin (Vn) and clusterin (Clu) can prevent this. Combining size exclusion chromatography with mass spectrometry profiling, we show that sMAC released from bacteria in serum is a heterogeneous mixture of complexes composed of C5b-8, up to three copies of C9 and multiple copies of Vn and Clu. Altogether, our data provide molecular insight into how sMAC is generated during bacterial infections. This fundamental knowledge could form the basis for exploring the use of sMAC as biomarker.


Asunto(s)
Complemento C5 , Infecciones por Escherichia coli , Activación de Complemento , Complejo de Ataque a Membrana del Sistema Complemento , Escherichia coli , Bacterias Grampositivas , Humanos , Vitronectina
19.
Science ; 376(6599): eabm6380, 2022 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-35587511

RESUMEN

The molecular basis of interindividual clinical variability upon infection with Staphylococcus aureus is unclear. We describe patients with haploinsufficiency for the linear deubiquitinase OTULIN, encoded by a gene on chromosome 5p. Patients suffer from episodes of life-threatening necrosis, typically triggered by S. aureus infection. The disorder is phenocopied in patients with the 5p- (Cri-du-Chat) chromosomal deletion syndrome. OTULIN haploinsufficiency causes an accumulation of linear ubiquitin in dermal fibroblasts, but tumor necrosis factor receptor-mediated nuclear factor κB signaling remains intact. Blood leukocyte subsets are unaffected. The OTULIN-dependent accumulation of caveolin-1 in dermal fibroblasts, but not leukocytes, facilitates the cytotoxic damage inflicted by the staphylococcal virulence factor α-toxin. Naturally elicited antibodies against α-toxin contribute to incomplete clinical penetrance. Human OTULIN haploinsufficiency underlies life-threatening staphylococcal disease by disrupting cell-intrinsic immunity to α-toxin in nonleukocytic cells.


Asunto(s)
Toxinas Bacterianas , Síndrome del Maullido del Gato , Endopeptidasas , Haploinsuficiencia , Proteínas Hemolisinas , Infecciones Estafilocócicas , Staphylococcus aureus , Toxinas Bacterianas/inmunología , Síndrome del Maullido del Gato/genética , Síndrome del Maullido del Gato/inmunología , Endopeptidasas/genética , Haploinsuficiencia/genética , Haploinsuficiencia/inmunología , Proteínas Hemolisinas/inmunología , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Humanos , Inmunidad Celular/genética , Necrosis , Infecciones Estafilocócicas/genética , Infecciones Estafilocócicas/inmunología , Infecciones Estafilocócicas/patología
20.
J Mol Recognit ; 24(6): 1077-86, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22038814

RESUMEN

During infection, our innate immune system is the first line of defense and has evolved to clear invading bacteria immediately. To do so, recognition is the key element. However, how does the innate immune system distinguish self from nonself, and how does it recognize all bacteria (estimated to be far over a million species)? The answer lies in the recognition of evolutionary conserved structures. In this review, we approach this phenomenon from the bacterial perspective. What are the evolutionary conserved structures in bacteria, and what strategies are there in the human innate immune system to sense these structures? We illustrate most examples both at the functional as well as at the molecular level. Furthermore, we highlight how pathogenic bacteria can evade this recognition to survive better in the human host which in turn can result in life-threatening diseases.


Asunto(s)
Bacterias/patogenicidad , Infecciones Bacterianas/inmunología , Interacciones Huésped-Patógeno/inmunología , Evasión Inmune/inmunología , Inmunidad Innata/inmunología , Bacterias/inmunología , Infecciones Bacterianas/genética , Carbohidratos/inmunología , Evolución Molecular , Humanos , Evasión Inmune/genética , Inmunidad Innata/genética , Lipopéptidos/inmunología , Lipopolisacáridos/inmunología , Staphylococcus aureus/genética , Staphylococcus aureus/inmunología , Receptores Toll-Like/genética , Receptores Toll-Like/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA