Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 145
Filtrar
1.
FASEB J ; 36(3): e22197, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35147989

RESUMEN

Neonatal meningitis-associated Escherichia coli (NMEC) is among the leading causes of bacterial meningitis and sepsis in newborn infants. Several virulence factors have been identified as common among NMEC, and have been shown to play an important role in the development of bacteremia and/or meningitis. However, there is significant variability in virulence factor expression between NMEC isolates, and relatively little research has been done to assess the impact of variable virulence factor expression on immune cell activation and the outcome of infection. Here, we investigated the role of NMEC strain-dependent P2X receptor (P2XR) signaling on the outcome of infection in neonatal mice. We found that alpha-hemolysin (HlyA)-expressing NMEC (HlyA+ ) induced robust P2XR-dependent macrophage cell death in vitro, while HlyA- NMEC did not. P2XR-dependent cell death was inflammasome independent, suggesting an uncoupling of P2XR and inflammasome activation in the context of NMEC infection. In vivo inhibition of P2XRs was associated with increased mortality in neonatal mice infected with HlyA+ NMEC, but had no effect on the survival of neonatal mice infected with HlyA- NMEC. Furthermore, we found that P2XR-dependent protection against HlyA+ NMEC in vivo required macrophages, but not neutrophils or NLRP3. Taken together, these data suggest that HlyA+ NMEC activates P2XRs which in turn confers macrophage-dependent protection against infection in neonates. In addition, our findings indicate that strain-dependent virulence factor expression should be taken into account when studying the immune response to NMEC.


Asunto(s)
Proteínas de Escherichia coli/toxicidad , Proteínas Hemolisinas/toxicidad , Inflamasomas/metabolismo , Meningitis por Escherichia coli/metabolismo , Sepsis Neonatal/metabolismo , Receptores Purinérgicos P2X/metabolismo , Animales , Células Cultivadas , Escherichia coli K12 , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Proteínas Hemolisinas/genética , Proteínas Hemolisinas/metabolismo , Macrófagos/metabolismo , Meningitis por Escherichia coli/microbiología , Ratones , Ratones Endogámicos C57BL , Sepsis Neonatal/microbiología , Receptores Purinérgicos P2X/genética
2.
Stem Cells ; 39(4): 482-496, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33373490

RESUMEN

Enterotoxigenic Escherichia coli causes severe infectious diarrhea with high morbidity and mortality in newborn and weanling pigs mainly through the production of heat-stable enterotoxins (STs). However, the precise regulatory mechanisms involved in ST-induced intestinal epithelium injury remain unclear. Consequently, we conducted the experiments in vivo (mice), ex vivo (mouse and porcine enteroids), and in vitro (MODE-K and IPEC-J2 cells) to explore the effect of STp (one type of STa) on the integrity of the intestinal epithelium. The results showed that acute STp exposure led to small intestinal edema, disrupted intestinal integrity, induced crypt cell expansion into spheroids, and downregulated Wnt/ß-catenin activity in the mice. Following a similar trend, the enteroid-budding efficiency and the expression of Active ß-catenin, ß-catenin, Lgr5, PCNA, and KRT20 were significantly decreased after STp treatment, as determined ex vivo. In addition, STp inhibited cell proliferation, induced cell apoptosis, destroyed cell barriers, and reduced Wnt/ß-catenin activity by downregulating its membrane receptor Frizzled7 (FZD7). In contrast, Wnt/ß-catenin reactivation protected the IPEC-J2 cells from STp-induced injury. Taking these findings together, we conclude that STp inhibits intestinal stem cell expansion to disrupt the integrity of the intestinal mucosa through the downregulation of the Wnt/ß-catenin signaling pathway.


Asunto(s)
Toxinas Bacterianas/toxicidad , Edema/genética , Enterotoxinas/toxicidad , Proteínas de Escherichia coli/toxicidad , Receptores Frizzled/genética , Mucosa Intestinal/efectos de los fármacos , Organoides/efectos de los fármacos , Células Madre/efectos de los fármacos , beta Catenina/genética , Animales , Línea Celular , Proliferación Celular/efectos de los fármacos , Edema/inducido químicamente , Edema/metabolismo , Edema/patología , Escherichia coli Enterotoxigénica/química , Escherichia coli Enterotoxigénica/patogenicidad , Receptores Frizzled/metabolismo , Regulación de la Expresión Génica , Absorción Intestinal/efectos de los fármacos , Mucosa Intestinal/citología , Mucosa Intestinal/metabolismo , Queratina-20/genética , Queratina-20/metabolismo , Ratones , Organoides/citología , Organoides/metabolismo , Antígeno Nuclear de Célula en Proliferación/genética , Antígeno Nuclear de Célula en Proliferación/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal , Esferoides Celulares/efectos de los fármacos , Esferoides Celulares/metabolismo , Esferoides Celulares/patología , Células Madre/citología , Células Madre/metabolismo , Porcinos , beta Catenina/metabolismo
3.
Proc Natl Acad Sci U S A ; 115(31): E7323-E7330, 2018 07 31.
Artículo en Inglés | MEDLINE | ID: mdl-30012608

RESUMEN

Pore-forming toxins (PFTs) form nanoscale pores across target membranes causing cell death. Cytolysin A (ClyA) from Escherichia coli is a prototypical α-helical toxin that contributes to cytolytic phenotype of several pathogenic strains. It is produced as a monomer and, upon membrane exposure, undergoes conformational changes and finally oligomerizes to form a dodecameric pore, thereby causing ion imbalance and finally cell death. However, our current understanding of this assembly process is limited to studies in detergents, which do not capture the physicochemical properties of biological membranes. Here, using single-molecule imaging and molecular dynamics simulations, we study the ClyA assembly pathway on phospholipid bilayers. We report that cholesterol stimulates pore formation, not by enhancing initial ClyA binding to the membrane but by selectively stabilizing a protomer-like conformation. This was mediated by specific interactions by cholesterol-interacting residues in the N-terminal helix. Additionally, cholesterol stabilized the oligomeric structure using bridging interactions in the protomer-protomer interfaces, thereby resulting in enhanced ClyA oligomerization. This dual stabilization of distinct intermediates by cholesterol suggests a possible molecular mechanism by which ClyA achieves selective membrane rupture of eukaryotic cell membranes. Topological similarity to eukaryotic membrane proteins suggests evolution of a bacterial α-toxin to adopt eukaryotic motifs for its activation. Broad mechanistic correspondence between pore-forming toxins hints at a wider prevalence of similar protein membrane insertion mechanisms.


Asunto(s)
Colesterol/química , Proteínas de Escherichia coli/toxicidad , Proteínas Hemolisinas/toxicidad , Membrana Celular/efectos de los fármacos , Proteínas de Escherichia coli/química , Proteínas Hemolisinas/química , Membrana Dobles de Lípidos/química , Simulación de Dinámica Molecular , Multimerización de Proteína
4.
Biochemistry ; 59(44): 4213-4224, 2020 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-33108867

RESUMEN

α-Hemolysin (HlyA) is an extracellular protein toxin secreted by uropathogenic strains of Escherichia coli that inserts into membranes of eukaryotic cells. The main goal of this work was to investigate the involvement of tryptophan (W) residues in the hemolytic activity of HlyA. We investigated the hemolytic activity of six single-point mutant proteins, in which one of the four Ws was replaced by cysteine (C) or leucine (L). We also analyzed the photoinactivation of HlyA with pterin (Ptr), an endogenous photosensitizer, as a method of unspecific oxidation of W and tyrosine (Y) residues. HlyA photoinactivation was analyzed by ultraviolet-visible spectrophotometry, hemolytic activity measurement, fluorescence spectroscopy, and electrophoretic analysis. The results indicate that Ws are important in the hemolytic process. Specifically, the chemical structure of the amino acid at position 578 is important for the acylation of HlyA at residue K563. Furthermore, the exposure of HlyA to ultraviolet radiation, with energy similar to that experienced under sun exposure, in the presence of Ptr induces the inactivation of the toxin, causing chemical changes in, at least, W and Y, the rate of damage to W residues being faster than that observed for Y residues. This work not only deepens our understanding of the structure-function relationship of the toxin but also introduces the possibility of using photoinactivation of HlyA for potential applications such as obtaining innocuous molecules for vaccine production and the elimination of the toxin from contaminated surfaces and drinking water.


Asunto(s)
Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/toxicidad , Proteínas Hemolisinas/química , Proteínas Hemolisinas/toxicidad , Luz , Triptófano , Sustitución de Aminoácidos , Proteínas de Escherichia coli/genética , Proteínas Hemolisinas/genética , Hemólisis/efectos de los fármacos , Hemólisis/efectos de la radiación
5.
Am J Physiol Gastrointest Liver Physiol ; 319(2): G121-G132, 2020 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-32567324

RESUMEN

Nongenomic glucocorticoid (GC) and serum- and glucocorticoid-inducible kinase 1 (SGK1) signaling regulate ion transport, but CFTR has not been investigated in the intestine. We examined GC, SGK1, and phosphatidylinositol 3-kinase (PI3K) kinase signaling of CFTR ion transport in native intestine and the role of GCs on mRNA, protein, surface expression, and cyclic guanosine monophosphate (cGMP)-elicited diarrhea. Rats were treated with dexamethasone (DEXA; 2 mg/kg ip) or DMSO for 1, 4, and 24 h. Cyclic adenosine monophosphate (cAMP)-activated ion transport was examined in the presence or absence of SGK1 and PI3K inhibitors. Phosphorylation of SGK1, phosphoinositide-dependent kinase 1, and Akt kinases was confirmed by immunoblots using phosphor-specific antibodies. Tissue lysates were analyzed by mass spectrometry. CFTR and SGK1 mRNA were measured by quantitative PCR. Changes in total and surface CFTR protein were determined. The role of GC in cGMP-activated CFTR ion transport was examined. GC synergistically increased CFTR ion transport by SGK1 and PI3K signaling and increased CFTR protein without altering SGK1 or CFTR mRNA. GC induced highest levels of CFTR protein at 4 h that were associated with marked increase in surface CFTR, phosphorylation of the ubiquitin ligase neural precursor cell expressed developmentally downregulated 4-like (Nedd4-2), and 14-3-3ε, supporting their roles in surface retention and stability. Coimmunoprecipitation of CFTR, Nedd4-2, and 14-3-3ε indicated that assembly of this complex is a likely effector of the SGK and Akt pathways. Mass spectrometry identified phosphorylated peptides in relevant proteins. GC-SGK1 potently regulates CFTR in the intestine and is implicated in diarrheal disease.NEW & NOTEWORTHY This is the first study to examine the mechanisms of glucocorticoid, serum- and glucocorticoid-inducible kinase 1, and nongenomic kinase signaling of CFTR in the native intestine. We identified unique and druggable intestine-specific factors of the pathway that are targets for treating stress-induced diarrhea.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Dexametasona/toxicidad , Diarrea/etiología , Dimetilsulfóxido/toxicidad , Proteínas Inmediatas-Precoces/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas 14-3-3/genética , Proteínas 14-3-3/metabolismo , Animales , Toxinas Bacterianas/toxicidad , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Diarrea/inducido químicamente , Enterotoxinas/toxicidad , Proteínas de Escherichia coli/toxicidad , Regulación de la Expresión Génica/efectos de los fármacos , Proteínas Inmediatas-Precoces/genética , Masculino , Ubiquitina-Proteína Ligasas Nedd4/genética , Ubiquitina-Proteína Ligasas Nedd4/metabolismo , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Transporte de Proteínas , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora/genética , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora/metabolismo , Ratas , Ratas Sprague-Dawley , Intercambiador 3 de Sodio-Hidrógeno/genética , Intercambiador 3 de Sodio-Hidrógeno/metabolismo
6.
Biochem Biophys Res Commun ; 525(4): 1068-1073, 2020 05 14.
Artículo en Inglés | MEDLINE | ID: mdl-32184018

RESUMEN

Subtilase cytotoxin (SubAB) is a member of bacterial AB5 toxin produced by certain enterohemorrhagic E. coli strains which cleaves host chaperone BiP in endoplasmic reticulum (ER), leading to ER stress-mediated cytotoxicity. Previous study suggested that protein disulfide isomerase (PDI), an enzyme which catalyzes the formation and breakage of disulfide bonds in proteins, regulates AB5 toxin such as cholera toxin by unfolding of A subunit, leading to its translocation into cytosol to induce disease. Although SubAB targets ER and has similar A subunit to that of other AB5 toxins, it is unclear whether PDI can modulate the SubAB function. Here we determined the role of PDI on SubAB-induced BiP cleavage, ER stress response and cytotoxicity in HeLa cells. We found that PDI knockdown significantly suppressed SubAB-induced BiP cleavage and eIF2α phosphorylation. The accumulation of SubAB in ER was perturbed upon PDI knockdown. Finally, cell viability assay showed that PDI knockdown and PDI inhibitor canceled the SubAB-induced cytotoxicity. Present results suggested that SubAB, after cellular uptake, translocates into ER and interacts with BiP that might be modulated by PDI. Identification of pivotal role of host proteins on bacterial toxin to elicit its pathogenesis is necessary basis for development of potential chemotherapy and new diagnostic strategy for control of toxin-producing bacterial infections.


Asunto(s)
Estrés del Retículo Endoplásmico/efectos de los fármacos , Proteínas de Escherichia coli/toxicidad , Proteína Disulfuro Isomerasas/metabolismo , Subtilisinas/toxicidad , Muerte Celular/genética , Supervivencia Celular/efectos de los fármacos , Chaperón BiP del Retículo Endoplásmico , Estrés del Retículo Endoplásmico/genética , Factor 2 Eucariótico de Iniciación/metabolismo , Técnicas de Silenciamiento del Gen , Células HeLa , Proteínas de Choque Térmico/metabolismo , Interacciones Microbiota-Huesped/genética , Humanos , MAP Quinasa Quinasa 4/metabolismo , Fosforilación , Proteína Disulfuro Isomerasas/antagonistas & inhibidores , Proteína Disulfuro Isomerasas/genética , ARN Interferente Pequeño
7.
Biochem J ; 476(22): 3455-3473, 2019 11 29.
Artículo en Inglés | MEDLINE | ID: mdl-31661116

RESUMEN

Alpha hemolysin (HlyA) is the major virulence factor of uropathogenic Escherichia coli (UPEC) strains. Once in circulation, a low concentration of the toxin induces an increase in intracellular calcium that activates calpains - which proteolyse cytoskeleton proteins - and also favours the exposure of phosphatidylserine (PS) in the outer leaflet of erythrocyte membranes. All these events are considered part of eryptosis, as well as the delivery of microvesicles (MVs). Within this context, we studied the delivery of MVs by erythrocytes treated with sublytic concentrations of HlyA and demonstrated that HlyA-treated erythrocytes secrete MVs of diameter ∼200 nm containing HlyA and PS by a mechanism involving an increment of intracellular calcium concentration and purinergic receptor activation. Despite the presence of toxin in their membrane, HlyA-MVs are not hemolytically active and do not induce ATP release in untreated erythrocytes, thus suggesting that the delivery of HlyA-MVs might act as a protective mechanism on the part of erythrocytes that removes the toxin from the membrane to prevent the spread of infection. Although erythrocytes have been found to eliminate denatured hemoglobin and several membrane proteins by shedding MVs, the present work has revealed for the first time that an exogenous protein, such as a toxin, is eliminated by this process. This finding sheds light on the mechanism of action of the toxin and serves to further elucidate the consequences of UPEC infection in patients exhibiting HlyA-related diseases.


Asunto(s)
Micropartículas Derivadas de Células/metabolismo , Eritrocitos/efectos de los fármacos , Infecciones por Escherichia coli/metabolismo , Infecciones por Escherichia coli/microbiología , Proteínas de Escherichia coli/toxicidad , Proteínas Hemolisinas/toxicidad , Micropartículas Derivadas de Células/efectos de los fármacos , Eritrocitos/citología , Eritrocitos/metabolismo , Escherichia coli/metabolismo , Infecciones por Escherichia coli/fisiopatología , Proteínas de Escherichia coli/metabolismo , Proteínas Hemolisinas/metabolismo , Hemólisis/efectos de los fármacos , Humanos , Fosfatidilserinas/metabolismo
8.
Protein Expr Purif ; 161: 70-77, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31054315

RESUMEN

Transcription factors play a crucial role in control of life of a bacterial cell, working as switchers to a different life style or pathogenicity. To reconstruct the network of regulatory events taking place in changing growth conditions, we need to know regulons of as many transcription factors as possible, and motifs recognized by them. Experimentally this can be attained via ChIP-seq in vivo, SELEX and DNAse I footprinting in vitro. All these approaches require large amounts of purified proteins. However, overproduction of transcription factors leading to their extensive binding to the regulatory elements on the DNA make them toxic to a bacterial cell thus significantly complicating production of a soluble protein. Here, on the example of three regulators from Escherichia coli, UxuR, ExuR, and LeuO, we show that stable production of toxic transcription factors in a soluble fraction can be significantly enhanced by holding the expression of a recombinant protein back at the early stages of bacterial growth. This can be achieved by cloning genes together with their regulatory regions containing repressor sites, with subsequent growth in a very rich media where activity of excessive regulators is not crucial, followed by induction with a very low concentration of an inducer. Schemes of further purification of these proteins were developed, and functional activity was confirmed.


Asunto(s)
Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/aislamiento & purificación , Escherichia coli/genética , Factores de Transcripción/genética , Factores de Transcripción/aislamiento & purificación , Escherichia coli/crecimiento & desarrollo , Escherichia coli/metabolismo , Proteínas de Escherichia coli/metabolismo , Proteínas de Escherichia coli/toxicidad , Regulación Bacteriana de la Expresión Génica , Operón , Factores de Transcripción/metabolismo , Factores de Transcripción/toxicidad
9.
EMBO Rep ; 18(11): 1978-1990, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28912123

RESUMEN

Most type VI secretion systems (T6SSs) described to date are protein delivery apparatuses that mediate bactericidal activities. Several T6SSs were also reported to mediate virulence activities, although only few anti-eukaryotic effectors have been described. Here, we identify three T6SSs in the marine bacterium Vibrio proteolyticus and show that T6SS1 mediates bactericidal activities under warm marine-like conditions. Using comparative proteomics, we find nine potential T6SS1 effectors, five of which belong to the polymorphic MIX-effector class. Remarkably, in addition to six predicted bactericidal effectors, the T6SS1 secretome includes three putative anti-eukaryotic effectors. One of these is a MIX-effector containing a cytotoxic necrotizing factor 1 domain. We demonstrate that T6SS1 can use this MIX-effector to target phagocytic cells, resulting in morphological changes and actin cytoskeleton rearrangements. In conclusion, the V. proteolyticus T6SS1, a system homologous to one found in pathogenic vibrios, uses a suite of polymorphic effectors that target both bacteria and eukaryotic neighbors.


Asunto(s)
Proteínas Bacterianas/genética , Toxinas Bacterianas/genética , Cromosomas Bacterianos/química , Proteínas de Escherichia coli/genética , Regulación Bacteriana de la Expresión Génica , Sistemas de Secreción Tipo VI/genética , Vibrio/genética , Citoesqueleto de Actina/efectos de los fármacos , Citoesqueleto de Actina/ultraestructura , Animales , Antibacterianos/metabolismo , Antibacterianos/toxicidad , Organismos Acuáticos , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/toxicidad , Toxinas Bacterianas/química , Toxinas Bacterianas/metabolismo , Toxinas Bacterianas/toxicidad , Mapeo Cromosómico , Técnicas de Cocultivo , Escherichia coli/efectos de los fármacos , Escherichia coli/crecimiento & desarrollo , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Proteínas de Escherichia coli/toxicidad , Ratones , Fagocitos/citología , Fagocitos/efectos de los fármacos , Dominios Proteicos , Células RAW 264.7 , Sistemas de Secreción Tipo VI/química , Sistemas de Secreción Tipo VI/metabolismo , Vibrio/metabolismo , Vibrio/patogenicidad , Virulencia
10.
Br J Nutr ; 122(2): 152-161, 2019 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-31006408

RESUMEN

The present study was carried out to evaluate the effect of dietary supplementation of Scutellaria baicalensis extracts (SBE) on intestinal health in terms of morphology, barrier integrity and immune responses in weaned piglets challenged with Escherichia coli K88. A total of seventy-two weaned piglets were assigned into two groups to receive a basal diet without including antibiotic additives or the basal diet supplemented 1000 mg SBE/kg diet for 14 d. On day 15, twelve healthy piglets from each group were selected to expose to oral administration of either 10 ml 1 × 109 colony-forming units of E. coli K88 or the vehicle control. After 48 h of E.coli K88 challenge, blood was sampled, and then all piglets were killed humanely for harvesting jejunal and ileal samples. Dietary supplementation of SBE significantly decreased diarrhoea frequency and improved feed conversion ratio (P < 0·05). SBE supplementation to E.coli K88-challenged piglets improved villous height and villous height/crypt depth (P < 0·05), recovered the protein expression of occludin and zonula occludens-2 in both the jejunum and ileum (P < 0·05), and mitigated the increases in plasma IL-1ß, TNF-α, IL-6, IgA and IgG (P < 0·05). Meanwhile, dietary SBE effectively inhibited the stimulation of NF-κB, P38 and TNF-α as well as IL-1ß in the small intestine of piglets challenged by E. coli K88 and prevented the activation of NF-κB/P38 signalling pathways (P < 0·05). Collectively, SBE supplementation can potently attenuate diarrhoea in weaning piglets and decrease inflammatory cytokine expressions through inhibiting the NF-κB and P38 signalling pathways.


Asunto(s)
Antígenos Bacterianos/toxicidad , Infecciones por Escherichia coli/veterinaria , Proteínas de Escherichia coli/toxicidad , Proteínas Fimbrias/toxicidad , Sistema de Señalización de MAP Quinasas/fisiología , FN-kappa B/fisiología , Scutellaria baicalensis/química , Enfermedades de los Porcinos/prevención & control , Animales , Diarrea/microbiología , Diarrea/prevención & control , Diarrea/veterinaria , Infecciones por Escherichia coli/inmunología , Infecciones por Escherichia coli/microbiología , Expresión Génica , Íleon/inmunología , Íleon/metabolismo , Íleon/patología , Yeyuno/inmunología , Yeyuno/metabolismo , Yeyuno/patología , Extractos Vegetales/administración & dosificación , Sus scrofa , Porcinos , Enfermedades de los Porcinos/inmunología , Enfermedades de los Porcinos/microbiología , Proteínas de Uniones Estrechas/genética , Destete
11.
Small ; 14(36): e1800890, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30091859

RESUMEN

Highly toxic protein misfolded oligomers associated with neurological disorders such as Alzheimer's and Parkinson's diseases are nowadays considered primarily responsible for promoting synaptic failure and neuronal death. Unraveling the relationship between structure and neurotoxicity of protein oligomers appears pivotal in understanding the causes of the pathological process, as well as in designing novel diagnostic and therapeutic strategies tuned toward the earliest and presymptomatic stages of the disease. Here, it is benefited from tip-enhanced Raman spectroscopy (TERS) as a surface-sensitive tool with spatial resolution on the nanoscale, to inspect the spatial organization and surface character of individual protein oligomers from two samples formed by the same polypeptide sequence and different toxicity levels. TERS provides direct assignment of specific amino acid residues that are exposed to a large extent on the surface of toxic species and buried in nontoxic oligomers. These residues, thanks to their outward disposition, might represent structural factors driving the pathogenic behavior exhibited by protein misfolded oligomers, including affecting cell membrane integrity and specific signaling pathways in neurodegenerative conditions.


Asunto(s)
Transferasas de Carboxilo y Carbamoilo/toxicidad , Proteínas de Escherichia coli/toxicidad , Nanopartículas/química , Pliegue de Proteína , Multimerización de Proteína , Espectrometría Raman/métodos , Pliegue de Proteína/efectos de los fármacos
12.
Microb Pathog ; 105: 96-99, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28212863

RESUMEN

YghJ, also known as SslE (Secreted and surface associated lipoprotein) is a cell surface associated and secreted lipoprotein harbouring M60 metalloprotease domain. Though the gene is known to be conserved among both pathogenic and commensal Escherichia coli isolates, the expression and secretion of YghJ was found to be higher among diverse E. coli pathotypes. YghJ, secreted from intestinal pathogens such as enterotoxigenic E. coli (ETEC) and enteropathogenic E. coli (EPEC) has been demonstrated to possess mucinase activity and hence facilitates colonization of these enteric pathogens to intestinal epithelial cells. Importantly, YghJ is also reported to be secreted from extraintestinal pathogenic E. coli isolates. In our previous study we have shown that YghJ, purified from a neonatal septicemic E. coli isolate could trigger induction of various proinflammatory cytokines in vitro. This led us to investigate the role of YghJ in causing in vivo tissue hemorrhage. In the present study, we validate the earlier in vitro finding and have showed that YghJ can cause extensive tissue damage in mouse ileum and is also able to induce significant fluid accumulation in a dose dependent manner in a mouse ileal loop (MIL) assay. Hence, our present study not only confirms the pathogenic potential of YghJ in sepsis pathophysiology but also indicates the enterotoxic ability of YghJ which makes it an important virulence determinant of intestinal pathogenic E. coli.


Asunto(s)
Proteínas de Escherichia coli/toxicidad , Escherichia coli/enzimología , Escherichia coli/patogenicidad , Hemorragia/inducido químicamente , Íleon/microbiología , Íleon/patología , Metaloproteasas/toxicidad , Animales , Ratones Endogámicos BALB C , Factores de Virulencia/metabolismo
13.
Nucleic Acids Res ; 43(16): 8002-12, 2015 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-26261214

RESUMEN

Bacterial type II toxin-antitoxin modules are protein-protein complexes whose functions are finely tuned by rapidly changing environmental conditions. E. coli toxin YafQ is suppressed under steady state growth conditions by virtue of its interaction with its cognate antitoxin, DinJ. During stress, DinJ is proteolytically degraded and free YafQ halts translation by degrading ribosome-bound mRNA to slow growth until the stress has passed. Although structures of the ribosome with toxins RelE and YoeB have been solved, it is unclear what residues among ribosome-dependent toxins are essential for mediating both recognition of the ribosome and the mRNA substrate given their low sequence identities. Here we show that YafQ coordinates binding to the 70S ribosome via three surface-exposed patches of basic residues that we propose directly interact with 16S rRNA. We demonstrate that YafQ residues H50, H63, D67 and H87 participate in acid-base catalysis during mRNA hydrolysis and further show that H50 and H63 functionally complement as general bases to initiate the phosphodiester cleavage reaction. Moreover YafQ residue F91 likely plays an important role in mRNA positioning. In summary, our findings demonstrate the plasticity of ribosome-dependent toxin active site residues and further our understanding of which toxin residues are important for function.


Asunto(s)
Toxinas Bacterianas/química , Proteínas de Escherichia coli/química , ARN Mensajero/metabolismo , Ribosomas/química , Secuencia de Aminoácidos , Toxinas Bacterianas/metabolismo , Toxinas Bacterianas/toxicidad , Secuencia Conservada , Proteínas de Escherichia coli/metabolismo , Proteínas de Escherichia coli/toxicidad , Hidrólisis , Unión Proteica , División del ARN , Ribosomas/efectos de los fármacos , Ribosomas/metabolismo
14.
J Biol Chem ; 290(52): 30783-96, 2015 Dec 25.
Artículo en Inglés | MEDLINE | ID: mdl-26499796

RESUMEN

LepB is a key membrane component of the cellular secretion machinery, which releases secreted proteins into the periplasm by cleaving the inner membrane-bound leader. We showed that LepB is also an essential component of the machinery hijacked by the tRNase colicin D for its import. Here we demonstrate that this non-catalytic activity of LepB is to promote the association of the central domain of colicin D with the inner membrane before the FtsH-dependent proteolytic processing and translocation of the toxic tRNase domain into the cytoplasm. The novel structural role of LepB results in a stable interaction with colicin D, with a stoichiometry of 1:1 and a nanomolar Kd determined in vitro. LepB provides a chaperone-like function for the penetration of several nuclease-type bacteriocins into target cells. The colicin-LepB interaction is shown to require only a short peptide sequence within the central domain of these bacteriocins and to involve residues present in the short C-terminal Box E of LepB. Genomic screening identified the conserved LepB binding motif in colicin-like ORFs from 13 additional bacterial species. These findings establish a new paradigm for the functional adaptability of an essential inner-membrane enzyme.


Asunto(s)
Toxinas Bacterianas/metabolismo , Bacteriocinas/metabolismo , Citoplasma/metabolismo , Desoxirribonucleasas/metabolismo , Proteínas de Escherichia coli/metabolismo , Escherichia coli/enzimología , Proteínas de la Membrana/metabolismo , Ribonucleasas/metabolismo , Serina Endopeptidasas/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Toxinas Bacterianas/química , Toxinas Bacterianas/genética , Toxinas Bacterianas/toxicidad , Bacteriocinas/genética , Transporte Biológico , Citoplasma/química , Citoplasma/genética , Desoxirribonucleasas/genética , Escherichia coli/química , Escherichia coli/genética , Escherichia coli/metabolismo , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/toxicidad , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Datos de Secuencia Molecular , Unión Proteica , Estructura Terciaria de Proteína , Ribonucleasas/química , Ribonucleasas/genética , Alineación de Secuencia , Serina Endopeptidasas/química , Serina Endopeptidasas/genética
15.
Infect Immun ; 84(11): 3114-3130, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27528275

RESUMEN

α-Hemolysin (HlyA) from Escherichia coli and leukotoxin A (LtxA) from Aggregatibacter actinomycetemcomitans are important virulence factors in ascending urinary tract infections and aggressive periodontitis, respectively. The extracellular signaling molecule ATP is released immediately after insertion of the toxins into plasma membranes and, via P2X receptors, is essential for the erythrocyte damage inflicted by these toxins. Moreover, ATP signaling is required for the ensuing recognition and phagocytosis of damaged erythrocytes by the monocytic cell line THP-1. Here, we investigate how these toxins affect THP-1 monocyte function. We demonstrate that both toxins trigger early ATP release and a following increase in the intracellular Ca2+ concentration ([Ca2+]i) in THP-1 monocytes. The HlyA- and LtxA-induced [Ca2+]i response is diminished by the P2 receptor antagonist in a pattern that fits the functional P2 receptor expression in these cells. Both toxins are capable of lysing THP-1 cells, with LtxA being more aggressive. Either desensitization or blockage of P2X1, P2X4, or P2X7 receptors markedly reduces toxin-induced cytolysis. This pattern is paralleled in freshly isolated human monocytes from healthy volunteers. Interestingly, only a minor fraction of the toxin-damaged THP-1 monocytes eventually lyse. P2X7 receptor inhibition generally prevents cell damage, except from a distinct cell shrinkage that prevails in response to the toxins. Moreover, we find that preexposure to HlyA preserves the capacity of THP-1 monocytes to phagocytose damaged erythrocytes and may induce readiness to discriminate between damaged and healthy erythrocytes. These findings suggest a new pharmacological target for protecting monocytes during exposure to pore-forming cytolysins during infection or injury.


Asunto(s)
Aggregatibacter actinomycetemcomitans/metabolismo , Toxinas Bacterianas/toxicidad , Escherichia coli/metabolismo , Proteínas Hemolisinas/toxicidad , Monocitos/efectos de los fármacos , Receptores Purinérgicos P2X/fisiología , Toxinas Bacterianas/metabolismo , Muerte Celular/efectos de los fármacos , Citoplasma/metabolismo , Citotoxinas/metabolismo , Eritrocitos/metabolismo , Proteínas de Escherichia coli/metabolismo , Proteínas de Escherichia coli/toxicidad , Exotoxinas/metabolismo , Exotoxinas/toxicidad , Proteínas Hemolisinas/metabolismo , Hemólisis/fisiología , Humanos , Monocitos/metabolismo
16.
BMC Microbiol ; 16(1): 158, 2016 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-27439312

RESUMEN

BACKGROUND: Pet is a toxin from the family of Serine Protease Autotransporters of Enterobacteriaceae which was initially identified in Enteroaggregative Escherichia coli strains. This protease exhibits enterotoxin properties, damages the cell cytoskeleton and induces intestinal epithelium alterations, which are associated with a severe inflammatory process. An in-vitro study was conducted to evaluate the effect of Pet on the migration of human peripheral blood monocytes-derived macrophages and its participation in the activation of the early inflammatory response and cytokine expression. RESULTS: In the macrophage migration activation assay, Pet produced a similar effect to that induced by opsonized zymosan (ZAS). Regarding the cytokine expression, an increase of IL-8, TNF-α (pro-inflammatory) and IL-10 (anti-inflammatory) was identified. In addition to the above results, the nuclear translocation of NF-kB pp65 was also identified. These events are probably related to the inflammatory response identified in the histological examination of intestine rat samples inoculated with Pet during a ligated loop assay. CONCLUSION: The results showed that Pet participates as an immunostimulant molecule for macrophages, which activates both their mobility and cytokine expression. These observations suggest that the toxin participates in the inflammatory process that is observed during the host infection by EAEC Pet producing.


Asunto(s)
Toxinas Bacterianas/química , Toxinas Bacterianas/toxicidad , Enterotoxinas/química , Enterotoxinas/toxicidad , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/toxicidad , Escherichia coli/enzimología , Activación de Macrófagos/efectos de los fármacos , Serina Endopeptidasas/química , Serina Endopeptidasas/toxicidad , Animales , Toxinas Bacterianas/metabolismo , Línea Celular , Quimiotaxis/efectos de los fármacos , Citocinas/biosíntesis , Citocinas/inmunología , Citoesqueleto/metabolismo , Enterotoxinas/metabolismo , Proteínas de Escherichia coli/metabolismo , Humanos , Inmunidad Innata , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/patología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/metabolismo , Fagocitosis , Ratas , Ratas Sprague-Dawley , Serina Endopeptidasas/metabolismo , Zimosan
17.
Immunology ; 145(2): 258-67, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25580516

RESUMEN

Enterohaemorrhagic Escherichia coli (EHEC) O157:H7 infection in humans can cause acute haemorrhagic colitis and severe haemolytic uraemic syndrome. The role of enterohaemolysin (Ehx) in the pathogenesis of O157:H7-mediated disease in humans remains undefined. Recent studies have revealed the importance of the inflammatory response in O157:H7 pathogenesis in humans. We previously reported that Ehx markedly induced interleukin-1ß (IL-1ß) production in human macrophages. Here, we investigated the disparity in Ehx-induced IL-1ß production between human and mouse macrophages and explored the underlying mechanism regarding the activation of NOD-like receptor family, pyrin domain containing 3 (NLRP3) inflammasomes. In contrast to the effects on human differentiated THP-1 cells and peripheral blood mononuclear cells, Ehx exerted no effect on IL-1ß production in mouse macrophages and splenocytes because of a disparity in pro-IL-1ß cleavage into mature IL-1ß upon caspase-1 activation. Additionally, Ehx significantly contributed to O157:H7-induced ATP release from THP-1 cells, which was not detected in mouse macrophages. Confocal microscopy demonstrated that Ehx was a key inducer of cathepsin B release in THP-1 cells but not in mouse IC-21 cells upon O157:H7 challenge. Inhibitor experiments indicated that O157:H7-induced IL-1ß production was largely dependent upon caspase-1 activation and partially dependent upon ATP signalling and cathepsin B release, which were both involved in NLRP3 activation. Moreover, inhibition of K(+) efflux drastically diminished O157:H7-induced IL-1ß production and cytotoxicity. The findings in this study may shed light on whether and how the Ehx contributes to the development of haemolytic uraemic syndrome in human O157:H7 infection.


Asunto(s)
Proteínas Portadoras/inmunología , Escherichia coli O157 , Proteínas de Escherichia coli/toxicidad , Proteínas Hemolisinas/toxicidad , Síndrome Hemolítico-Urémico/inmunología , Interleucina-1beta/inmunología , Macrófagos/inmunología , Animales , Caspasa 1/inmunología , Catepsina B/inmunología , Línea Celular Tumoral , Síndrome Hemolítico-Urémico/patología , Humanos , Inflamasomas/inmunología , Macrófagos/patología , Ratones , Proteína con Dominio Pirina 3 de la Familia NLR , Especificidad de la Especie
18.
Mol Microbiol ; 91(3): 596-605, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24308852

RESUMEN

A type II toxin-antitoxin system in Escherichia coli, rnlA-rnlB, functions as an anti-phage mechanism. RnlA is a toxin with an endoribonuclease activity and the cognate RnlB inhibits RnlA toxicity in E. coli cells. After bacteriophage T4 infection, RnlA is activated by the disappearance of RnlB, resulting in the rapid degradation of T4 mRNAs and consequently no T4 propagation, when T4 dmd is defective: Dmd is an antitoxin against RnlA for promoting own propagation. Previous studies suggested that the activation of RnlA after T4 infection was regulated by multiple components. Here, we provide the evidence that RNase HI is an essential factor for activation of RnlA. The dmd mutant phage could grow on ΔrnhA (encoding RNase HI) cells, in which RnlA-mediated mRNA cleavage activity was defective. RNase HI bound to RnlA in vivo and enhanced the RNA cleavage activity of RnlA in vitro. In addition, ectopic expression of RnlA in ΔrnlAB ΔrnhA cells has less effect on cell toxicity and RnlA-mediated mRNA degradation than in ΔrnlAB cells. This is the first example of a direct factor for activation of a toxin.


Asunto(s)
Proteínas de Escherichia coli/toxicidad , Escherichia coli/enzimología , Ribonucleasa H/metabolismo , Bacteriófago T4/crecimiento & desarrollo , Escherichia coli/genética , Eliminación de Gen , Ribonucleasa H/genética
19.
Infect Immun ; 82(12): 5308-16, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25287923

RESUMEN

Enterotoxigenic Escherichia coli (ETEC) is a significant cause of diarrheal disease and death, especially in children in developing countries. ETEC causes disease by colonizing the small intestine and producing heat-labile toxin (LT), heat-stable toxin (ST), or both LT and ST (LT+ST). The majority of ETEC strains produce both ST and LT. Despite the prevalence of LT+ST-producing organisms, few studies have examined the physiologic or immunologic consequences of simultaneous exposure to these two potent enterotoxins. In the current report, we demonstrate that when LT and ST are both present, they increase water movement into the intestinal lumen over and above the levels observed with either toxin alone. As expected, cultured intestinal epithelial cells increased their expression of intracellular cyclic GMP (cGMP) when treated with ST and their expression of intracellular cyclic AMP (cAMP) when treated with LT. When both toxins were present, cGMP levels but not cAMP levels were synergistically elevated compared with the levels of expression caused by the corresponding single-toxin treatment. Our data also demonstrate that the levels of inflammatory cytokines produced by intestinal epithelial cells in response to LT are significantly reduced in animals exposed to both enterotoxins. These findings suggest that there may be complex differences between the epithelial cell intoxication and, potentially, secretory outcomes induced by ETEC strains expressing LT+ST compared with strains that express LT or ST only. Our results also reveal a novel mechanism wherein ST production may reduce the hosts' ability to mount an effective innate or adaptive immune response to infecting organisms.


Asunto(s)
Toxinas Bacterianas/toxicidad , Citocinas/metabolismo , Escherichia coli Enterotoxigénica/fisiología , Enterotoxinas/toxicidad , Células Epiteliales/efectos de los fármacos , Proteínas de Escherichia coli/toxicidad , Nucleótidos Cíclicos/metabolismo , Agua/metabolismo , Animales , Línea Celular , Escherichia coli Enterotoxigénica/inmunología , Células Epiteliales/metabolismo , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C
20.
Infect Immun ; 82(7): 2802-14, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24733098

RESUMEN

Enteropathogenic Escherichia coli (EPEC) primarily infects children in developing countries and causes diarrhea that can be deadly. EPEC pathogenesis occurs through type III secretion system (T3SS)-mediated injection of effectors into intestinal epithelial cells (IECs); these effectors alter actin dynamics, modulate the immune response, and disrupt tight junction (TJ) integrity. The resulting compromised barrier function and increased gastrointestinal (GI) permeability may be responsible for the clinical symptoms of infection. Type I interferon (IFN) mediates anti-inflammatory activities and serves essential functions in intestinal immunity and homeostasis; however, its role in the immune response to enteric pathogens, such as EPEC, and its impact on IEC barrier function have not been examined. Here, we report that IFN-ß is induced following EPEC infection and regulates IEC TJ proteins to maintain barrier function. The EPEC T3SS effector NleD counteracts this protective activity by inhibiting IFN-ß induction and enhancing tumor necrosis factor alpha to promote barrier disruption. The endoribonuclease RNase L is a key mediator of IFN induction and action that promotes TJ protein expression and IEC barrier integrity. EPEC infection inhibits RNase L in a T3SS-dependent manner, providing a mechanism by which EPEC evades IFN-induced antibacterial activities. This work identifies novel roles for IFN-ß and RNase L in IEC barrier functions that are targeted by EPEC effectors to escape host defense mechanisms and promote virulence. The IFN-RNase L axis thus represents a potential therapeutic target for enteric infections and GI diseases involving compromised barrier function.


Asunto(s)
Endorribonucleasas/metabolismo , Escherichia coli Enteropatógena/fisiología , Interferón beta/metabolismo , Mucosa Intestinal/citología , Mucosa Intestinal/fisiología , Células CACO-2 , Endorribonucleasas/genética , Células Epiteliales/fisiología , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Proteínas de Escherichia coli/toxicidad , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Interferón beta/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA