Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
2.
Proc Natl Acad Sci U S A ; 112(46): E6369-78, 2015 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-26578797

RESUMEN

The outcome of an infection depends on host recognition of the pathogen, hence leading to the activation of signaling pathways controlling defense responses. A long-held belief is that the modification of the lipid A moiety of the lipopolysaccharide could help Gram-negative pathogens to evade innate immunity. However, direct evidence that this happens in vivo is lacking. Here we report the lipid A expressed in the tissues of infected mice by the human pathogen Klebsiella pneumoniae. Our findings demonstrate that Klebsiella remodels its lipid A in a tissue-dependent manner. Lipid A species found in the lungs are consistent with a 2-hydroxyacyl-modified lipid A dependent on the PhoPQ-regulated oxygenase LpxO. The in vivo lipid A pattern is lost in minimally passaged bacteria isolated from the tissues. LpxO-dependent modification reduces the activation of inflammatory responses and mediates resistance to antimicrobial peptides. An lpxO mutant is attenuated in vivo thereby highlighting the importance of this lipid A modification in Klebsiella infection biology. Colistin, one of the last options to treat multidrug-resistant Klebsiella infections, triggers the in vivo lipid A pattern. Moreover, colistin-resistant isolates already express the in vivo lipid A pattern. In these isolates, LpxO-dependent lipid A modification mediates resistance to colistin. Deciphering the lipid A expressed in vivo opens the possibility of designing novel therapeutics targeting the enzymes responsible for the in vivo lipid A pattern.


Asunto(s)
Infecciones por Klebsiella/metabolismo , Klebsiella pneumoniae/metabolismo , Lípido A/biosíntesis , Lípido A/química , Animales , Humanos , Infecciones por Klebsiella/genética , Infecciones por Klebsiella/patología , Klebsiella pneumoniae/genética , Lípido A/genética , Pulmón/microbiología , Ratones , Estructura Molecular , Especificidad de Órganos
3.
PLoS Pathog ; 11(1): e1004627, 2015 01.
Artículo en Inglés | MEDLINE | ID: mdl-25633080

RESUMEN

Klebsiella pneumoniae is a significant human pathogen, in part due to high rates of multidrug resistance. RamA is an intrinsic regulator in K. pneumoniae established to be important for the bacterial response to antimicrobial challenge; however, little is known about its possible wider regulatory role in this organism during infection. In this work, we demonstrate that RamA is a global transcriptional regulator that significantly perturbs the transcriptional landscape of K. pneumoniae, resulting in altered microbe-drug or microbe-host response. This is largely due to the direct regulation of 68 genes associated with a myriad of cellular functions. Importantly, RamA directly binds and activates the lpxC, lpxL-2 and lpxO genes associated with lipid A biosynthesis, thus resulting in modifications within the lipid A moiety of the lipopolysaccharide. RamA-mediated alterations decrease susceptibility to colistin E, polymyxin B and human cationic antimicrobial peptide LL-37. Increased RamA levels reduce K. pneumoniae adhesion and uptake into macrophages, which is supported by in vivo infection studies, that demonstrate increased systemic dissemination of ramA overexpressing K. pneumoniae. These data establish that RamA-mediated regulation directly perturbs microbial surface properties, including lipid A biosynthesis, which facilitate evasion from the innate host response. This highlights RamA as a global regulator that confers pathoadaptive phenotypes with implications for our understanding of the pathogenesis of Enterobacter, Salmonella and Citrobacter spp. that express orthologous RamA proteins.


Asunto(s)
Proteínas Bacterianas/genética , Farmacorresistencia Bacteriana Múltiple/genética , Interacciones Huésped-Patógeno/genética , Klebsiella pneumoniae/genética , Lipopolisacáridos/metabolismo , Animales , Antibacterianos/farmacología , Péptidos Catiónicos Antimicrobianos/farmacología , Secuencia de Bases , Células Cultivadas , Regulación Bacteriana de la Expresión Génica , Genes Bacterianos , Infecciones por Klebsiella/genética , Proteínas de Transporte de Membrana/genética , Proteínas de Transporte de Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Polimixinas/farmacología , Regulón
4.
J Biol Chem ; 290(27): 16678-97, 2015 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-25971969

RESUMEN

Klebsiella pneumoniae is an etiologic agent of community-acquired and nosocomial pneumonia. It has been shown that K. pneumoniae infections are characterized by reduced early inflammatory response. Recently our group has shown that K. pneumoniae dampens the activation of inflammatory responses by antagonizing the activation of the NF-κB canonical pathway. Our results revealed that K. pneumoniae capsule polysaccharide (CPS) was necessary but not sufficient to attenuate inflammation. To identify additional Klebsiella factors required to dampen inflammation, we standardized and applied a high-throughput gain-of-function screen to examine a Klebsiella transposon mutant library. We identified 114 mutants that triggered the activation of NF-κB. Two gene ontology categories accounted for half of the loci identified in the screening: metabolism and transport genes (32% of the mutants) and envelope-related genes (17%). Characterization of the mutants revealed that the lack of the enterobactin siderophore was linked to a reduced CPS expression, which in turn underlined the NF-κB activation induced by the mutant. The lipopolysaccharide (LPS) O-polysaccharide and the pullulanase (PulA) type 2 secretion system (T2SS) are required for full effectiveness of the immune evasion. Importantly, these factors do not play a redundant role. The fact that LPS O-polysaccharide and T2SS mutant-induced responses were dependent on TLR2-TLR4-MyD88 activation suggested that LPS O-polysaccharide and PulA perturbed Toll-like receptor (TLR)-dependent recognition of K. pneumoniae. Finally, we demonstrate that LPS O-polysaccharide and pulA mutants are attenuated in the pneumonia mouse model. We propose that LPS O-polysaccharide and PulA T2SS could be new targets for the design of new antimicrobials. Increasing TLR-governed defense responses might provide also selective alternatives for the management of K. pneumoniae pneumonia.


Asunto(s)
Proteínas Bacterianas/genética , Infecciones por Klebsiella/inmunología , Klebsiella pneumoniae/genética , FN-kappa B/inmunología , Animales , Proteínas Bacterianas/inmunología , Femenino , Genómica , Humanos , Evasión Inmune , Infecciones por Klebsiella/genética , Infecciones por Klebsiella/microbiología , Klebsiella pneumoniae/inmunología , Lipopolisacáridos/inmunología , Ratones , FN-kappa B/antagonistas & inhibidores , FN-kappa B/genética , Transducción de Señal
5.
Cell Microbiol ; 17(11): 1537-60, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26045209

RESUMEN

Klebsiella pneumoniae is an important cause of community-acquired and nosocomial pneumonia. Evidence indicates that Klebsiella might be able to persist intracellularly within a vacuolar compartment. This study was designed to investigate the interaction between Klebsiella and macrophages. Engulfment of K. pneumoniae was dependent on host cytoskeleton, cell plasma membrane lipid rafts and the activation of phosphoinositide 3-kinase (PI3K). Microscopy studies revealed that K. pneumoniae resides within a vacuolar compartment, the Klebsiella-containing vacuole (KCV), which traffics within vacuoles associated with the endocytic pathway. In contrast to UV-killed bacteria, the majority of live bacteria did not co-localize with markers of the lysosomal compartment. Our data suggest that K. pneumoniae triggers a programmed cell death in macrophages displaying features of apoptosis. Our efforts to identify the mechanism(s) whereby K. pneumoniae prevents the fusion of the lysosomes to the KCV uncovered the central role of the PI3K-Akt-Rab14 axis to control the phagosome maturation. Our data revealed that the capsule is dispensable for Klebsiella intracellular survival if bacteria were not opsonized. Furthermore, the environment found by Klebsiella within the KCV triggered the down-regulation of the expression of cps. Altogether, this study proves evidence that K. pneumoniae survives killing by macrophages by manipulating phagosome maturation that may contribute to Klebsiella pathogenesis.


Asunto(s)
Klebsiella pneumoniae/inmunología , Klebsiella pneumoniae/fisiología , Lisosomas/metabolismo , Macrófagos/microbiología , Viabilidad Microbiana , Animales , Células Cultivadas , Interacciones Huésped-Patógeno , Humanos , Macrófagos/inmunología , Macrófagos/patología , Ratones , Vacuolas/microbiología
6.
PLoS Pathog ; 8(10): e1002978, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23133372

RESUMEN

Pathogenic bacteria may modify their surface to evade the host innate immune response. Yersinia enterocolitica modulates its lipopolysaccharide (LPS) lipid A structure, and the key regulatory signal is temperature. At 21°C, lipid A is hexa-acylated and may be modified with aminoarabinose or palmitate. At 37°C, Y. enterocolitica expresses a tetra-acylated lipid A consistent with the 3'-O-deacylation of the molecule. In this work, by combining genetic and mass spectrometric analysis, we establish that Y. enterocolitica encodes a lipid A deacylase, LpxR, responsible for the lipid A structure observed at 37°C. Western blot analyses indicate that LpxR exhibits latency at 21°C, deacylation of lipid A is not observed despite the expression of LpxR in the membrane. Aminoarabinose-modified lipid A is involved in the latency. 3-D modelling, docking and site-directed mutagenesis experiments showed that LpxR D31 reduces the active site cavity volume so that aminoarabinose containing Kdo(2)-lipid A cannot be accommodated and, therefore, not deacylated. Our data revealed that the expression of lpxR is negatively controlled by RovA and PhoPQ which are necessary for the lipid A modification with aminoarabinose. Next, we investigated the role of lipid A structural plasticity conferred by LpxR on the expression/function of Y. enterocolitica virulence factors. We present evidence that motility and invasion of eukaryotic cells were reduced in the lpxR mutant grown at 21°C. Mechanistically, our data revealed that the expressions of flhDC and rovA, regulators controlling the flagellar regulon and invasin respectively, were down-regulated in the mutant. In contrast, the levels of the virulence plasmid (pYV)-encoded virulence factors Yops and YadA were not affected in the lpxR mutant. Finally, we establish that the low inflammatory response associated to Y. enterocolitica infections is the sum of the anti-inflammatory action exerted by pYV-encoded YopP and the reduced activation of the LPS receptor by a LpxR-dependent deacylated LPS.


Asunto(s)
Hidrolasas de Éster Carboxílico/genética , Hidrolasas de Éster Carboxílico/metabolismo , Lípido A/química , Lípido A/metabolismo , Yersinia enterocolitica/metabolismo , Yersinia enterocolitica/patogenicidad , Acilación , Adhesinas Bacterianas/biosíntesis , Animales , Arabinosa/análogos & derivados , Proteínas Bacterianas/metabolismo , Regulación Bacteriana de la Expresión Génica , Células HeLa , Humanos , Lipopolisacáridos/química , Lipopolisacáridos/metabolismo , Macrófagos/metabolismo , Ratones , Mutagénesis Sitio-Dirigida , Mutación , Ácidos Palmíticos , Temperatura , Factores de Transcripción/metabolismo , Factores de Virulencia/metabolismo , Yersiniosis/genética , Yersiniosis/inmunología , Yersiniosis/microbiología , Yersinia enterocolitica/genética , Yersinia enterocolitica/inmunología
7.
PLoS Pathog ; 8(5): e1002675, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22589715

RESUMEN

Innate immunity recognizes bacterial molecules bearing pathogen-associated molecular patterns to launch inflammatory responses leading to the activation of adaptive immunity. However, the lipopolysaccharide (LPS) of the gram-negative bacterium Brucella lacks a marked pathogen-associated molecular pattern, and it has been postulated that this delays the development of immunity, creating a gap that is critical for the bacterium to reach the intracellular replicative niche. We found that a B. abortus mutant in the wadC gene displayed a disrupted LPS core while keeping both the LPS O-polysaccharide and lipid A. In mice, the wadC mutant induced proinflammatory responses and was attenuated. In addition, it was sensitive to killing by non-immune serum and bactericidal peptides and did not multiply in dendritic cells being targeted to lysosomal compartments. In contrast to wild type B. abortus, the wadC mutant induced dendritic cell maturation and secretion of pro-inflammatory cytokines. All these properties were reproduced by the wadC mutant purified LPS in a TLR4-dependent manner. Moreover, the core-mutated LPS displayed an increased binding to MD-2, the TLR4 co-receptor leading to subsequent increase in intracellular signaling. Here we show that Brucella escapes recognition in early stages of infection by expressing a shield against recognition by innate immunity in its LPS core and identify a novel virulence mechanism in intracellular pathogenic gram-negative bacteria. These results also encourage for an improvement in the generation of novel bacterial vaccines.


Asunto(s)
Brucella abortus/inmunología , Brucella abortus/patogenicidad , Evasión Inmune , Inmunidad Innata , Lipopolisacáridos/metabolismo , Animales , Sistemas de Secreción Bacterianos , Brucella abortus/genética , Brucelosis/microbiología , Brucelosis/patología , Células Dendríticas/inmunología , Células Dendríticas/microbiología , Femenino , Inflamación/inmunología , Lípido A/metabolismo , Lipopolisacáridos/inmunología , Macrófagos/inmunología , Macrófagos/microbiología , Proteínas de la Membrana/genética , Proteínas de la Membrana/inmunología , Ratones , Ratones Endogámicos BALB C
8.
Infect Immun ; 81(10): 3552-65, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23836821

RESUMEN

The implementation of infection models that approximate human disease is essential for understanding pathogenesis at the molecular level and for testing new therapies before they are entered into clinical stages. Insects are increasingly being used as surrogate hosts because they share, with mammals, essential aspects of the innate immune response to infections. We examined whether the larva of the wax moth Galleria mellonella could be used as a host model to conceptually approximate Klebsiella pneumoniae-triggered pneumonia. We report that the G. mellonella model is capable of distinguishing between pathogenic and nonpathogenic Klebsiella strains. Moreover, K. pneumoniae infection of G. mellonella models some of the known features of Klebsiella-induced pneumonia, i.e., cell death associated with bacterial replication, avoidance of phagocytosis by phagocytes, and the attenuation of host defense responses, chiefly the production of antimicrobial factors. Similar to the case for the mouse pneumonia model, activation of innate responses improved G. mellonella survival against subsequent Klebsiella challenge. Virulence factors necessary in the mouse pneumonia model were also implicated in the Galleria model. We found that mutants lacking capsule polysaccharide, lipid A decorations, or the outer membrane proteins OmpA and OmpK36 were attenuated in Galleria. All mutants activated G. mellonella defensive responses. The Galleria model also allowed us to monitor Klebsiella gene expression. The expression levels of cps and the loci implicated in lipid A remodeling peaked during the first hours postinfection, in a PhoPQ- and PmrAB-governed process. Taken together, these results support the utility of G. mellonella as a surrogate host for assessing infections with K. pneumoniae.


Asunto(s)
Klebsiella pneumoniae/patogenicidad , Mariposas Nocturnas/microbiología , Animales , Interacciones Huésped-Patógeno , Larva/microbiología
9.
J Bacteriol ; 194(12): 3173-88, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22505678

RESUMEN

Antimicrobial peptides (APs) belong to the arsenal of weapons of the innate immune system against infections. In the case of gram-negative bacteria, APs interact with the anionic lipid A moiety of the lipopolysaccharide (LPS). In yersiniae most virulence factors are temperature regulated. Studies from our laboratory demonstrated that Yersinia enterocolitica is more susceptible to polymyxin B, a model AP, when grown at 37°C than at 22°C (J. A. Bengoechea, R. Díaz, and I. Moriyón, Infect. Immun. 64:4891-4899, 1996), and here we have extended this observation to other APs, not structurally related to polymyxin B. Mechanistically, we demonstrate that the lipid A modifications with aminoarabinose and palmitate are downregulated at 37°C and that they contribute to AP resistance together with the LPS O-polysaccharide. Bacterial loads of lipid A mutants in Peyer's patches, liver, and spleen of orogastrically infected mice were lower than those of the wild-type strain at 3 and 7 days postinfection. PhoPQ and PmrAB two-component systems govern the expression of the loci required to modify lipid A with aminoarabinose and palmitate, and their expressions are also temperature regulated. Our findings support the notion that the temperature-dependent regulation of loci controlling lipid A modifications could be explained by H-NS-dependent negative regulation alleviated by RovA. In turn, our data also demonstrate that PhoPQ and PmrAB regulate positively the expression of rovA, the effect of PhoPQ being more important. However, rovA expression reached wild-type levels in the phoPQ pmrAB mutant background, hence indicating the existence of an unknown regulatory network controlling rovA expression in this background.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/farmacología , Proteínas Bacterianas/metabolismo , Proteínas de Unión al ADN/metabolismo , Farmacorresistencia Bacteriana , Regulación Bacteriana de la Expresión Génica , Factores de Transcripción/metabolismo , Yersinia enterocolitica/efectos de los fármacos , Animales , Arabinosa/análogos & derivados , Arabinosa/metabolismo , Carga Bacteriana , Modelos Animales de Enfermedad , Lípido A/metabolismo , Hígado/microbiología , Ratones , Antígenos O/metabolismo , Palmitatos/metabolismo , Ganglios Linfáticos Agregados/microbiología , Polimixina B/farmacología , Bazo/microbiología , Temperatura , Yersiniosis/inmunología , Yersiniosis/microbiología
10.
J Biol Chem ; 286(12): 9956-67, 2011 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-21278256

RESUMEN

Outer membrane protein A (OmpA) is a class of proteins highly conserved among the Enterobacteriaceae family and throughout evolution. Klebsiella pneumoniae is a capsulated gram-negative pathogen. It is an important cause of community-acquired and nosocomial pneumonia. Evidence indicates that K. pneumoniae infections are characterized by a lack of an early inflammatory response. Data from our laboratory indicate that K. pneumoniae CPS helps to suppress the host inflammatory response. However, it is unknown whether K. pneumoniae employs additional factors to modulate host inflammatory responses. Here, we report that K. pneumoniae OmpA is important for immune evasion in vitro and in vivo. Infection of A549 and normal human bronchial cells with 52OmpA2, an ompA mutant, increased the levels of IL-8. 52145-Δwca(K2)ompA, which does not express CPS and ompA, induced the highest levels of IL-8. Both mutants could be complemented. In vivo, 52OmpA2 induced higher levels of tnfα, kc, and il6 than the wild type. ompA mutants activated NF-κB, and the phosphorylation of p38, p44/42, and JNK MAPKs and IL-8 induction was via NF-κB-dependent and p38- and p44/42-dependent pathways. 52OmpA2 engaged TLR2 and -4 to activate NF-κB, whereas 52145-Δwca(K2)ompA activated not only TLR2 and TLR4 but also NOD1. Finally, we demonstrate that the ompA mutant is attenuated in the pneumonia mouse model. The results of this study indicate that K. pneumoniae OmpA contributes to attenuate airway cell responses. This may facilitate pathogen survival in the hostile environment of the lung.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/metabolismo , Infecciones por Klebsiella/metabolismo , Klebsiella pneumoniae/metabolismo , Neumonía Bacteriana/metabolismo , Mucosa Respiratoria/metabolismo , Animales , Proteínas de la Membrana Bacteriana Externa/genética , Proteínas de la Membrana Bacteriana Externa/inmunología , Femenino , Células HEK293 , Humanos , Evasión Inmune/genética , Evasión Inmune/inmunología , Inflamación/genética , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/microbiología , Interleucina-8/biosíntesis , Interleucina-8/genética , Interleucina-8/inmunología , Infecciones por Klebsiella/genética , Infecciones por Klebsiella/inmunología , Klebsiella pneumoniae/genética , Klebsiella pneumoniae/inmunología , Ratones , Quinasas de Proteína Quinasa Activadas por Mitógenos/genética , Quinasas de Proteína Quinasa Activadas por Mitógenos/inmunología , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Mutación , FN-kappa B/genética , FN-kappa B/inmunología , FN-kappa B/metabolismo , Proteína Adaptadora de Señalización NOD1/genética , Proteína Adaptadora de Señalización NOD1/inmunología , Proteína Adaptadora de Señalización NOD1/metabolismo , Neumonía Bacteriana/genética , Neumonía Bacteriana/inmunología , Neumonía Bacteriana/microbiología , Mucosa Respiratoria/inmunología , Mucosa Respiratoria/microbiología , Receptores Toll-Like/genética , Receptores Toll-Like/inmunología , Receptores Toll-Like/metabolismo
11.
Infect Immun ; 79(9): 3718-32, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21708987

RESUMEN

Antimicrobial peptides (APs) impose a threat to the survival of pathogens, and it is reasonable to postulate that bacteria have developed strategies to counteract them. Polymyxins are becoming the last resort to treat infections caused by multidrug-resistant Gram-negative bacteria and, similar to APs, they interact with the anionic lipopolysaccharide. Given that polymyxins and APs share the initial target, it is possible that bacterial defense mechanisms against polymyxins will be also effective against host APs. We sought to determine whether exposure to polymyxin will increase Klebsiella pneumoniae resistance to host APs. Indeed, exposure of K. pneumoniae to polymyxin induces cross-resistance not only to polymyxin itself but also to APs present in the airways. Polymyxin treatment upregulates the expression of the capsule polysaccharide operon and the loci required to modify the lipid A with aminoarabinose and palmitate with a concomitant increase in capsule and lipid A species containing such modifications. Moreover, these surface changes contribute to APs resistance and also to polymyxin-induced cross-resistance to APs. Bacterial loads of lipid A mutants in trachea and lungs of intranasally infected mice were lower than those of wild-type strain. PhoPQ, PmrAB, and the Rcs system govern polymyxin-induced transcriptional changes, and there is a cross talk between PhoPQ and the Rcs system. Our findings support the notion that Klebsiella activates a defense program against APs that is controlled by three signaling systems. Therapeutic strategies directed to prevent the activation of this program could be a new approach worth exploring to facilitate the clearance of the pathogen from the airways.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/farmacología , Proteínas Bacterianas/metabolismo , Infecciones por Klebsiella/microbiología , Klebsiella pneumoniae/efectos de los fármacos , Klebsiella pneumoniae/patogenicidad , Polimixinas/farmacología , Animales , Antibacterianos/farmacología , Arabinosa/análogos & derivados , Arabinosa/metabolismo , Cápsulas Bacterianas/biosíntesis , Cápsulas Bacterianas/genética , Carga Bacteriana , Farmacorresistencia Bacteriana Múltiple , Femenino , Klebsiella pneumoniae/metabolismo , Lípido A/análisis , Pulmón/microbiología , Espectrometría de Masas , Ratones , Ratones Endogámicos C57BL , Pruebas de Sensibilidad Microbiana , Ácido Palmítico/análisis , Tráquea/microbiología , Factores de Virulencia/biosíntesis
12.
Antimicrob Agents Chemother ; 55(7): 3370-9, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21576434

RESUMEN

Colistin resistance is rare in Acinetobacter baumannii, and little is known about its mechanism. We investigated the role of PmrCAB in this trait, using (i) resistant and susceptible clinical strains, (ii) laboratory-selected mutants of the type strain ATCC 19606 and of the clinical isolate ABRIM, and (iii) a susceptible/resistant pair of isogenic clinical isolates, Ab15/133 and Ab15/132, isolated from the same patient. pmrAB sequences in all the colistin-susceptible isolates were identical to reference sequences, whereas resistant clinical isolates harbored one or two amino acid replacements variously located in PmrB. Single substitutions in PmrB were also found in resistant mutants of strains ATCC 19606 and ABRIM and in the resistant clinical isolate Ab15/132. No mutations in PmrA or PmrC were found. Reverse transcriptase (RT)-PCR identified increased expression of pmrA (4- to 13-fold), pmrB (2- to 7-fold), and pmrC (1- to 3-fold) in resistant versus susceptible organisms. Matrix-assisted laser desorption ionization-time of flight (MALDI-TOF) mass spectrometry showed the addition of phosphoethanolamine to the hepta-acylated form of lipid A in the resistant variants and in strain ATCC 19606 grown under low-Mg(2+) induction conditions. pmrB gene knockout mutants of the colistin-resistant ATCC 19606 derivative showed >100-fold increased susceptibility to colistin and 5-fold decreased expression of pmrC; they also lacked the addition of phosphoethanolamine to lipid A. We conclude that the development of a moderate level of colistin resistance in A. baumannii requires distinct genetic events, including (i) at least one point mutation in pmrB, (ii) upregulation of pmrAB, and (iii) expression of pmrC, which lead to addition of phosphoethanolamine to lipid A.


Asunto(s)
Acinetobacter baumannii/efectos de los fármacos , Acinetobacter baumannii/metabolismo , Antibacterianos/farmacología , Proteínas Bacterianas/metabolismo , Colistina/farmacología , Etanolaminas/metabolismo , Lípido A/metabolismo , Acinetobacter baumannii/genética , Proteínas Bacterianas/genética , Farmacorresistencia Bacteriana/genética , Datos de Secuencia Molecular , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
13.
Biomolecules ; 11(4)2021 04 18.
Artículo en Inglés | MEDLINE | ID: mdl-33919637

RESUMEN

Galectins bind various pathogens through recognition of distinct carbohydrate structures. In this work, we examined the binding of four human galectins to the Gram-negative bacteria Klebsiella pneumoniae (Kpn) and non-typeable Haemophilus influenzae (NTHi), which display different surface glycans. In particular, Kpn cells are covered by a polysaccharide capsule and display an O-chain-containing lipopolysaccharide (LPS), whereas NTHi is not capsulated and its LPS, termed lipooligosacccharide (LOS), does not contain O-chain. Binding assays to microarray-printed bacteria revealed that galectins-3, -4, and -8, but not galectin-1, bind to Kpn and NTHi cells, and confocal microscopy attested binding to bacterial cells in suspension. The three galectins bound to array-printed Kpn LPS. Moreover, analysis of galectin binding to mutant Kpn cells evidenced that the O-chain is the docking point for galectins on wild type Kpn. Galectins-3, -4, and -8 also bound the NTHi LOS. Microarray-assisted comparison of the binding to full-length and truncated LOSs, as well as to wild type and mutant cells, supported LOS involvement in galectin binding to NTHi. However, deletion of the entire LOS oligosaccharide chain actually increased binding to NTHi cells, indicating the availability of other ligands on the bacterial surface, as similarly inferred for Kpn cells devoid of both O-chain and capsule. Altogether, the results illustrate galectins' versatility for recognizing different bacterial structures, and point out the occurrence of so far overlooked galectin ligands on bacterial surfaces.


Asunto(s)
Galectinas/metabolismo , Haemophilus influenzae/metabolismo , Klebsiella pneumoniae/metabolismo , Lipopolisacáridos/metabolismo , Sitios de Unión , Galectinas/química , Humanos , Lipopolisacáridos/química , Unión Proteica
14.
Infect Immun ; 78(6): 2768-81, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20385763

RESUMEN

Yersinia enterocolitica is an important human pathogen. Y. enterocolitica must adapt to the host environment, and temperature is an important cue regulating the expression of most Yersinia virulence factors. Here, we report that Y. enterocolitica 8081 serotype O:8 synthesized tetra-acylated lipid A at 37 degrees C but that hexa-acylated lipid A predominated at 21 degrees C. By mass spectrometry and genetic methods, we have shown that the Y. enterocolitica msbB, htrB, and lpxP homologues encode the acyltransferases responsible for the addition of C(12), C(14) and C(16:1), respectively, to lipid A. The expression levels of the acyltransferases were temperature regulated. Levels of expression of msbB and lpxP were higher at 21 degrees C than at 37 degrees C, whereas the level of expression of htrB was higher at 37 degrees C. At 21 degrees C, an lpxP mutant was the strain most susceptible to polymyxin B, whereas at 37 degrees C, an htrB mutant was the most susceptible. We present evidence that the lipid A acylation status affects the expression of Yersinia virulence factors. Thus, expression of flhDC, the flagellar master regulatory operon, was downregulated in msbB and lpxP mutants, with a concomitant decrease in motility. Expression of the phospholipase yplA was also downregulated in both mutants. inv expression was downregulated in msbB and htrB mutants, and consistent with this finding, invasion of HeLa cells was diminished. However, the expression of rovA, the positive regulator of inv, was not affected in the mutants. The levels of pYV-encoded virulence factors Yops and YadA in the acyltransferase mutants were not affected. Finally, we show that only the htrB mutant was attenuated in vivo.


Asunto(s)
Lípido A/metabolismo , Factores de Virulencia/metabolismo , Yersinia enterocolitica/patogenicidad , Acilación , Aciltransferasas/genética , Aciltransferasas/metabolismo , Animales , Antibacterianos/farmacología , Proteínas Bacterianas/metabolismo , Flagelina/biosíntesis , Eliminación de Gen , Perfilación de la Expresión Génica , Células HeLa , Humanos , Lípido A/química , Locomoción , Masculino , Espectrometría de Masas , Ratones , Ratones Endogámicos BALB C , Polimixina B/farmacología , Temperatura , Virulencia , Yersiniosis/microbiología , Yersiniosis/patología , Yersinia enterocolitica/efectos de los fármacos , Yersinia enterocolitica/efectos de la radiación
15.
Infect Immun ; 78(3): 1135-46, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20008534

RESUMEN

Human beta-defensins (hBDs) contribute to the protection of the respiratory tract against pathogens. It is reasonable to postulate that pathogens have developed countermeasures to resist them. Klebsiella pneumoniae capsule polysaccharide (CPS), but not the lipopolysaccharide O antigen, mediated resistance against hBD1 and hBD2. hBD3 was the most potent hBD against Klebsiella. We investigated the possibility that as a strategy for survival in the lung, K. pneumoniae may not activate the expression of hBDs. Infection of A549 and normal human bronchial cells with 52145-Deltawca(K2), a CPS mutant, increased the expression of hBD2 and hBD3. Neither the wild type nor the lipopolysaccharide O antigen mutant increased the expression of hBDs. In vivo, 52145-Deltawca(K2) induced higher levels of mBD4 and mBD14, possible mouse orthologues of hBD2 and hBD3, respectively, than the wild type. 52145-Deltawca(K2)-dependent upregulation of hBD2 occurred via NF-kappaB and mitogen-activated protein kinases (MAPKs) p44/42, Jun N-terminal protein kinase (JNK)-dependent pathways. The increase in hBD3 expression was dependent on the MAPK JNK. 52145-Deltawca(K2) engaged Toll-like receptors 2 and 4 (TLR2 and TLR4) to activate hBD2, whereas hBD3 expression was dependent on NOD1. K. pneumoniae induced the expression of CYLD and MKP-1, which act as negative regulators for 52145-Deltawca(K2)-induced expression of hBDs. Bacterial engagement of pattern recognition receptors induced CYLD and MKP-1, which may initiate the attenuation of proinflammatory pathways. The results of this study indicate that K. pneumoniae CPS not only protects the pathogen from the bactericidal action of defensins but also impedes their expression. These features of K. pneumoniae CPS may facilitate pathogen survival in the hostile environment of the lung.


Asunto(s)
Cápsulas Bacterianas/inmunología , Células Epiteliales/inmunología , Células Epiteliales/microbiología , Klebsiella pneumoniae/inmunología , beta-Defensinas/antagonistas & inhibidores , beta-Defensinas/biosíntesis , Animales , Cápsulas Bacterianas/genética , Línea Celular , Células Cultivadas , Modelos Animales de Enfermedad , Regulación hacia Abajo , Femenino , Eliminación de Gen , Genes Reporteros , Humanos , Infecciones por Klebsiella/inmunología , Luciferasas/biosíntesis , Luciferasas/genética , Ratones , Ratones Endogámicos C57BL
16.
Antimicrob Agents Chemother ; 54(1): 177-83, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19858254

RESUMEN

Respiratory infections caused by Klebsiella pneumoniae are characterized by high rates of mortality and morbidity. Management of these infections is often difficult, due to the high frequency of strains that are resistant to multiple antimicrobial agents. Multidrug efflux pumps play a major role as a mechanism of antimicrobial resistance in Gram-negative pathogens. In the present study, we investigated the role of the K. pneumoniae AcrRAB operon in antimicrobial resistance and virulence by using isogenic knockouts deficient in the AcrB component and the AcrR repressor, both derived from the virulent strain 52145R. We demonstrated that the AcrB knockout was more susceptible, not only to quinolones, but also to other antimicrobial agents, including beta-lactams, than the wild-type strain and the AcrR knockout. We further showed that the AcrB knockout was more susceptible to antimicrobial agents present in human bronchoalveolar lavage fluid and to human antimicrobial peptides than the wild-type strain and the AcrR knockout. Finally, the AcrB knockout exhibited a reduced capacity to cause pneumonia in a murine model, in contrast to the wild-type strain. The results of this study suggest that, in addition to contributing to the multidrug resistance phenotype, the AcrAB efflux pump may represent a novel virulence factor required for K. pneumoniae to resist innate immune defense mechanisms of the lung, thus facilitating the onset of pneumonia.


Asunto(s)
Antibacterianos/farmacología , Farmacorresistencia Bacteriana/genética , Klebsiella pneumoniae/genética , Klebsiella pneumoniae/patogenicidad , Animales , Líquido del Lavado Bronquioalveolar/microbiología , Medios de Cultivo , ADN Bacteriano/genética , Defensinas/farmacología , Genotipo , Humanos , Infecciones por Klebsiella/microbiología , Klebsiella pneumoniae/efectos de los fármacos , Lipopolisacáridos/biosíntesis , Ratones , Ratones Endogámicos ICR , Pruebas de Sensibilidad Microbiana , Fenotipo , Plásmidos/genética , Polimixina B/farmacología , Polisacáridos/biosíntesis , ARN Bacteriano/genética , Proteínas Represoras/genética
17.
Antimicrob Agents Chemother ; 53(1): 298-302, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19015361

RESUMEN

A Klebsiella pneumoniae ompA mutant was more susceptible to antimicrobial peptides (APs) than the wild type. Susceptibility did not result from surface changes other than the absence of OmpA. Our data suggest that OmpA is implicated in the activation of yet-unknown systems dedicated to ameliorating AP cytotoxicity.


Asunto(s)
Antibacterianos/farmacología , Péptidos Catiónicos Antimicrobianos/farmacología , Proteínas de la Membrana Bacteriana Externa/fisiología , Klebsiella pneumoniae/efectos de los fármacos , Klebsiella pneumoniae/genética , Proteínas de la Membrana Bacteriana Externa/genética , Resistencia a Medicamentos/genética , Electroforesis en Gel de Poliacrilamida , Mutación
18.
PLoS One ; 9(5): e97020, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24824990

RESUMEN

Nontypable Haemophilus influenzae (NTHi) has emerged as an important opportunistic pathogen causing infection in adults suffering obstructive lung diseases. Existing evidence associates chronic infection by NTHi to the progression of the chronic respiratory disease, but specific features of NTHi associated with persistence have not been comprehensively addressed. To provide clues about adaptive strategies adopted by NTHi during persistent infection, we compared sequential persistent isolates with newly acquired isolates in sputa from six patients with chronic obstructive lung disease. Pulse field gel electrophoresis (PFGE) identified three patients with consecutive persistent strains and three with new strains. Phenotypic characterisation included infection of respiratory epithelial cells, bacterial self-aggregation, biofilm formation and resistance to antimicrobial peptides (AMP). Persistent isolates differed from new strains in showing low epithelial adhesion and inability to form biofilms when grown under continuous-flow culture conditions in microfermenters. Self-aggregation clustered the strains by patient, not by persistence. Increasing resistance to AMPs was observed for each series of persistent isolates; this was not associated with lipooligosaccharide decoration with phosphorylcholine or with lipid A acylation. Variation was further analyzed for the series of three persistent isolates recovered from patient 1. These isolates displayed comparable growth rate, natural transformation frequency and murine pulmonary infection. Genome sequencing of these three isolates revealed sequential acquisition of single-nucleotide variants in the AMP permease sapC, the heme acquisition systems hgpB, hgpC, hup and hxuC, the 3-deoxy-D-manno-octulosonic acid kinase kdkA, the long-chain fatty acid transporter ompP1, and the phosphoribosylamine glycine ligase purD. Collectively, we frame a range of pathogenic traits and a repertoire of genetic variants in the context of persistent infection by NTHi.


Asunto(s)
Haemophilus influenzae/citología , Haemophilus influenzae/genética , Fenotipo , Enfermedad Pulmonar Obstructiva Crónica/microbiología , Adulto , Células Epiteliales Alveolares/microbiología , Análisis de Varianza , Animales , Péptidos Catiónicos Antimicrobianos/farmacología , Adhesión Bacteriana/fisiología , Secuencia de Bases , Biopelículas/crecimiento & desarrollo , Cartilla de ADN/genética , Electroforesis en Gel de Campo Pulsado , Genotipo , Haemophilus influenzae/efectos de los fármacos , Humanos , Ratones , Datos de Secuencia Molecular , Análisis de Secuencia de ADN
19.
PLoS One ; 8(2): e56847, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23457627

RESUMEN

Phagocytosis is a key process of the immune system. The human pathogen Klebsiella pneumoniae is a well known example of a pathogen highly resistant to phagocytosis. A wealth of evidence demonstrates that the capsule polysaccharide (CPS) plays a crucial role in resistance to phagocytosis. The amoeba Dictyostelium discoideum shares with mammalian macrophages the ability to phagocytose and kill bacteria. The fact that K. pneumoniae is ubiquitous in nature and, therefore, should avoid predation by amoebae, poses the question whether K. pneumoniae employs similar means to counteract amoebae and mammalian phagocytes. Here we developed an assay to evaluate K. pneumoniae-D. discoideum interaction. The richness of the growth medium affected the threshold at which the cps mutant was permissive for Dictyostelium and only at lower nutrient concentrations the cps mutant was susceptible to predation by amoebae. Given the critical role of bacterial surface elements on host-pathogen interactions, we explored the possible contribution of the lipopolysaccharide (LPS) and outer membrane proteins (OMPs) to combat phagoyctosis by D. discoideum. We uncover that, in addition to the CPS, the LPS O-polysaccharide and the first core sugar participate in Klebsiella resistance to predation by D. discoideum. K. pneumoniae LPS lipid A decorations are also necessary to avoid predation by amoebae although PagP-dependent palmitoylation plays a more important role than the lipid A modification with aminoarabinose. Mutants lacking OMPs OmpA or OmpK36 were also permissive for D. discoideium growth. Except the LPS O-polysaccharide mutants, all mutants were more susceptible to phagocytosis by mouse alveolar macrophages. Finally, we found a correlation between virulence, using the pneumonia mouse model, and resistance to phagocytosis. Altogether, this work reveals novel K. pneumoniae determinants involved in resistance to phagocytosis and supports the notion that Dictyostelium amoebae might be useful as host model to measure K. pneumoniae virulence and not only phagocytosis.


Asunto(s)
Interacciones Huésped-Patógeno , Klebsiella pneumoniae/fisiología , Fagocitos/microbiología , Animales , Arabinosa/análogos & derivados , Arabinosa/metabolismo , Proteínas de la Membrana Bacteriana Externa/metabolismo , Proteínas Bacterianas/genética , Secuencia de Carbohidratos , Dictyostelium/microbiología , Dictyostelium/fisiología , Femenino , Klebsiella pneumoniae/genética , Klebsiella pneumoniae/metabolismo , Klebsiella pneumoniae/patogenicidad , Lípido A/metabolismo , Macrófagos Alveolares/citología , Macrófagos Alveolares/microbiología , Ratones , Datos de Secuencia Molecular , Mutación , Antígenos O/química , Antígenos O/metabolismo , Ácido Palmítico/metabolismo , Fagocitos/citología , Fagocitosis , Porinas/genética
20.
PLoS One ; 5(4): e10033, 2010 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-20383325

RESUMEN

Infected airway epithelial cells up-regulate the expression of chemokines, chiefly IL-8, and antimicrobial molecules including beta-defensins (BD). Acinetobacter baumannii is a cause of hospital-acquired pneumonia. We examined whether A. baumannii induced the expressions of IL-8 and BD2 by airway epithelial cells and the receptors implicated in bacterial detection. A549 and human primary airway cells released IL-8 upon infection. A. baumannii-infected cells also increased the expression of BD2 which killed A. baummannii strains. IL-8 induction was via NF-kappaB and mitogen-activated kinases p38 and p44/42-dependent pathways. A. baumannii engaged Toll-like receptor (TLR) 2 and TLR4 pathways and A549 cells could use soluble CD14 as TLRs co-receptor. A. baumannii lipopolysaccharide stimulated IL-8 release by A549 cells and sCD14 facilitated the recognition of the lipopolysaccharide. Mass spectrometry analysis revealed that A. baumannii lipid A structure matches those with endotoxic potential. These results demonstrate that airway epithelial cells produce mediators important for A. baumannii clearance.


Asunto(s)
Acinetobacter baumannii/inmunología , Interacciones Huésped-Patógeno , Inflamación/etiología , Neumonía/microbiología , Transducción de Señal , Infecciones por Acinetobacter , Línea Celular , Células Epiteliales , Humanos , Mediadores de Inflamación/análisis , Interleucina-8/genética , Neumonía/inmunología , Sistema Respiratorio/citología , Regulación hacia Arriba/genética , beta-Defensinas/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA