Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Int J Med Microbiol ; 308(7): 940-946, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29983334

RESUMEN

The life-threatening sequela of hemorrhagic colitis induced by Shiga toxins (Stx)-producing Escherichia coli (STEC) infections in humans is hemolytic uremic syndrome (HUS), the main cause of acute renal failure in early childhood. The key step in the pathogenesis of HUS is the appearance of Stx in the blood of infected patients because these powerful virulence factors are capable of inducing severe microangiopathic lesions in the kidney. During precocious toxemia, which occurs in patients before the onset of HUS during the intestinal phase, Stx bind to several different circulating cells. An early response of these cells might include the release of proinflammatory mediators associated with the development of HUS. Here, we show that primary human monocytes stimulated with Shiga toxin 1a (Stx1a) through the glycolipid receptor globotriaosylceramide released larger amounts of proinflammatory molecules (IL-1ß, TNFα, IL-6, G-CSF, CXCL8, CCL2, CCL4) than Stx1a-treated neutrophils. The mediators (except IL-1ß) are among the top six proinflammatory mediators found in the sera from patients with HUS in different studies. The molecules appear to be involved in different pathogenetic steps of HUS, i.e. sensitization of renal endothelial cells to the toxin actions (IL-1ß, TNFα), activation of circulating monocytes and neutrophils (CXCL8, CCL2, CCL4) and increase in neutrophil counts in patients with poor prognosis (G-CSF). Hence, a role of circulating monocytes in the very early phases of the pathogenetic process culminating with HUS can be envisaged. Impairment of the events of precocious toxemia would prevent or reduce the risk of HUS in STEC-infected children.


Asunto(s)
Citocinas/sangre , Síndrome Hemolítico-Urémico/patología , Monocitos/metabolismo , Toxina Shiga I/metabolismo , Escherichia coli Shiga-Toxigénica/patogenicidad , Trihexosilceramidas/metabolismo , Células Cultivadas , Citocinas/metabolismo , Síndrome Hemolítico-Urémico/microbiología , Humanos , Interleucina-8/sangre , Neutrófilos/metabolismo
2.
J Immunol ; 196(3): 1177-85, 2016 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-26695372

RESUMEN

Hemolytic uremic syndrome (HUS) is the life-threatenig sequela of intestinal infections by Shiga toxin (Stx)-producing Escherichia coli (STEC) in children. Human neutrophils specifically bind Stx through TLR4, the receptor of LPS. The binding could be considered protective (Stx sequestration) or harmful (toxin delivery to target organs). The amount of Stx on neutrophils is in equilibrium with the amount of Stx present in the gut, and it is also related to renal and neurologic symptoms. The TLR4-mediated interaction of LPS with innate immune cells is hampered by the well-known antibiotic polymyxin B. In this study, we show that the same antibiotic impairs the binding of Stx to neutrophils, also blocking their functional effects (release of CXCL8, formation of neutrophil/platelet aggregates) involved in HUS pathogenesis. Controls for contaminating LPS in Stx-induced neutrophil responses inhibited by polymyxin B were performed. Stx interact with human neutrophils through their A chain, since these leukocytes do not express globotriaosylceramide, the specific receptor for Stx B chains. Consistently, polymyxin B blocked the enzymatic activity of Stx1, Stx2, Stx1 A chain, and the analogous plant protein gelonin, whereas the antibiotic did not show any protective effect on Stx-induced cytotoxicity in globotriaosylceramide-expressing Raji cells. Antibiotic administration is not recommended in human STEC infections during the prodromal intestinal phase, and the toxicity of polymyxin B could further discourage its therapeutic use. However, nontoxic, nonbactericidal polymyxin derivatives have been developed and might be used in animal models of STEC infection to study their efficacy in preventing the onset of HUS during the systemic blood phase of Stx.


Asunto(s)
Antibacterianos/farmacología , Síndrome Hemolítico-Urémico/inmunología , Neutrófilos/efectos de los fármacos , Polimixina B/farmacología , Toxina Shiga/toxicidad , Animales , Citometría de Flujo , Síndrome Hemolítico-Urémico/tratamiento farmacológico , Humanos , Ratones , Neutrófilos/inmunología
3.
Biochem Biophys Res Commun ; 483(3): 936-940, 2017 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-28082201

RESUMEN

Protein synthesis is one of the main cellular functions inhibited during hypertonic challenge. The subsequent accumulation of the compatible osmolyte betaine during the later adaptive response allows not only recovery of translation but also its stimulation. In this paper, we show that betaine modulates translation by enhancing the formation of cap-independent 48 S pre-initiation complexes, leaving cap-dependent 48 S pre-initiation complexes basically unchanged. In the presence of betaine, CrPV IRES- and sodium-dependent neutral amino acid transporter-2 (SNAT2) 5'-UTR-driven translation is 2- and 1.5-fold stimulated in MCF7 cells, respectively. Thus, betaine could provide an advantage in translation of messengers coding for proteins implicated in the response of cells to different stressors, which are often recognized by ribosomal 40 S subunit through simplified cap-independent mechanisms.


Asunto(s)
Betaína/metabolismo , Betaína/farmacología , Biosíntesis de Proteínas/efectos de los fármacos , Caperuzas de ARN/metabolismo , Regiones no Traducidas 5' , Sistema de Transporte de Aminoácidos A/metabolismo , Animales , Sistema Libre de Células , Humanos , Soluciones Hipertónicas , Luciferasas/genética , Luciferasas/metabolismo , Células MCF-7 , Presión Osmótica , Polirribosomas/metabolismo , Biosíntesis de Proteínas/genética , Conejos , Reticulocitos/efectos de los fármacos , Reticulocitos/metabolismo
4.
J Immunol ; 191(9): 4748-58, 2013 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-24068665

RESUMEN

Hemolytic uremic syndrome (HUS) caused by intestinal Shiga toxin-producing Escherichia coli infections is a worldwide health problem, as dramatically exemplified by the German outbreak occurred in summer 2011 and by a constant burden of cases in children. Shiga toxins (Stx) play a pivotal role in HUS by triggering endothelial damage in kidney and brain through globotriaosylceramide (Gb3Cer) receptor targeting. Moreover, Stx interact with human neutrophils, as experimentally demonstrated in vitro and as observed in patients with HUS. A neutrophil-protective role on endothelial damage (sequestration of circulating toxins) and a causative role in toxin delivery from the gut to the kidney (piggyback transport) have been suggested in different studies. However, the receptor that recognizes Stx in human neutrophils, which do not express Gb3Cer, has not been identified. In this study, by competition and functional experiments with appropriate agonists and antagonists (LPS, anti-TLR4 Abs, respectively), we have identified TLR4 as the receptor that specifically recognizes Stx1 and Stx2 in human neutrophils. Accordingly, these treatments displaced both toxin variants from neutrophils and, upon challenge with Stx1 or Stx2, neutrophils displayed the same pattern of cytokine expression as in response to LPS (assessed by quantitative RT-PCR, ELISA, or multiplexed Luminex-based immunoassays). Moreover, data were supported by adequate controls excluding any potential interference of contaminating LPS in Stx-binding and activation of neutrophils. The identification of the Stx-receptor on neutrophils provides additional elements to foster the understanding of the pathophysiology of HUS and could have an important effect on the development of therapeutic strategies.


Asunto(s)
Neutrófilos/metabolismo , Toxina Shiga I/inmunología , Toxina Shiga II/inmunología , Receptor Toll-Like 4/inmunología , Anticuerpos Monoclonales , Citocinas/metabolismo , Escherichia coli/inmunología , Escherichia coli/metabolismo , Infecciones por Escherichia coli/inmunología , Síndrome Hemolítico-Urémico/inmunología , Síndrome Hemolítico-Urémico/microbiología , Humanos , Lipopolisacáridos , Neutrófilos/inmunología , Trihexosilceramidas/metabolismo
5.
Microorganisms ; 12(1)2024 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-38257980

RESUMEN

Candida spp. are an important opportunistic pathogen that can represent a possible cause of severe infections, especially in immunocompromised individuals. The clinical impact of Candida spp. depends, in part, on the ability to form biofilms, communities of nestled cells into the extracellular matrix. In this study, we compared the biofilm formation ability of 83 strains of Candida spp. isolated from blood cultures and other materials, such as respiratory samples, urine, and exudate, and their sensitivity to fluconazole (FLZ). Strains were divided into tertiles to establish cut-offs to classify isolates as low, moderate, or high biofilm producers (<0.26, 0.266-0.839, >0.839) and biofilms with low, moderate, or high metabolic activity (<0.053, 0.053-0.183, >0.183). A non-linear relationship between biofilm production and metabolic activity was found in C. glabrata and C. tropicalis. In addition, the increase in minimum biofilm eradication concentrations (MBEC50) compared to the Minor Inhibitory Concentration (PMIC) of the planktonic form in Candida spp. confirms the role of biofilm in the induction of resistance to FLZ.

6.
Biochem Biophys Res Commun ; 430(2): 466-9, 2013 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-23237800

RESUMEN

Lactate dehydrogenase A (LDH-A) binds single stranded DNA (ssDNA) and stimulates cell transcription. Binding is prevented by NADH, suggesting that the coenzyme site is involved in the interaction LDH-A/ssDNA. We recently identified an inhibitor of LDH-A enzymatic activity (Galloflavin, GF) which occupies the NADH site. In the experiments reported here we studied whether GF can also hinder the binding of LDH-A to ssDNA and investigated its effects on RNA synthesis in cultured cells. Using a filter binding assay we observed that 4 µM GF inhibited the binding of human LDH-A to a single stranded [(3)H]DNA sample by 50%. After only 0.5-1h, 50-100 µM GF inhibited RNA synthesis in SW620 cells maintained in a medium in which galactose substituted glucose. In these culture conditions, SW620 cells did not produce lactic acid and effects caused by the inhibition of the enzymatic activity of LDH-A could be excluded. Novel LDH-A inhibitors which hinder aerobic glycolysis of cancer cells are at present actively searched. Our results suggest that: (i) inhibitors which bind the NADH site can exert their antiproliferative activity not only by blocking aerobic glycolysis but also by causing an inhibition of RNA synthesis independent from the effect on glycolysis; (ii) GF can be a useful tool to study the biological role of LDH-A binding to ssDNA.


Asunto(s)
ADN de Cadena Simple/metabolismo , Isocumarinas/farmacología , L-Lactato Deshidrogenasa/antagonistas & inhibidores , ARN/antagonistas & inhibidores , Línea Celular Tumoral , Humanos , Isoenzimas/antagonistas & inhibidores , Isoenzimas/metabolismo , L-Lactato Deshidrogenasa/metabolismo , Lactato Deshidrogenasa 5 , Unión Proteica/efectos de los fármacos , ARN/biosíntesis
7.
Viruses ; 15(2)2023 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-36851504

RESUMEN

Literature offers plenty of cases of immunocompromised patients, who develop chronic and severe SARS-CoV-2 infections. The aim of this study is to provide further insight into SARS-CoV-2 evolutionary dynamic taking into exam a subject suffering from follicular lymphoma, who developed a persistent infection for over 7 months. Eight nasopharyngeal swabs were obtained, and were analyses by qRT-PCR for diagnostic purposes. All of them were considered eligible (Ct < 30) for NGS sequencing. Sequence analysis showed that all sequences matched the B.1.617.2 AY.122 lineage, but they differed by few mutations identifying three genetically similar subpopulations, which evolved during the course of infection, demonstrating that prolonged replication is paralleled with intra-host virus evolution. These evidences support the hypothesis that SARS-CoV-2 adaptive capacities are able to shape a heterogeneous viral population in the context of immunocompromised patients. Spill-over of viral variants with enhanced transmissibility or immune escape capacities from these subjects is plausible.


Asunto(s)
COVID-19 , Humanos , SARS-CoV-2/genética , Huésped Inmunocomprometido , Mutación
8.
Microorganisms ; 11(1)2023 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-36677483

RESUMEN

The recent emergence of a number of new SARS-CoV-2 variants resulting from recombination between two distinct parental lineages or sub-lineages within the same lineage has sparked the debate regarding potential enhanced viral infectivity and immune escape. Among these, XBB, recombinant of BA.2.10 and BA.2.75, has caused major concern in some countries due to its rapid increase in prevalence. In this study, we tested XBB escape capacity from mRNA-vaccine-induced (BNT162b2) neutralising antibodies compared to B.1 ancestral lineage and another co-circulating variant (B.1.1.529 BA.5) by analysing sera collected 30 days after the second dose in 92 healthcare workers. Our data highlighted an enhanced and statistically significant immune escape ability of the XBB recombinant. Although these are preliminary results, this study highlights the importance of immune escape monitoring of new and forthcoming variants and of the reformulation of existing vaccines.

9.
Int J Infect Dis ; 131: 65-70, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-36924839

RESUMEN

OBJECTIVES: Recombination related to coinfection is a huge driving force in determining the virus genetic variability, particularly in conditions of partial immune control, leading to prolonged infection. Here, we characterized a distinctive mutational pattern, highly suggestive of Delta-Omicron double infection, in a lymphoma patient. METHODS: The specimen was characterized through a combined approach, analyzing the results of deep sequencing in primary sample, viral culture, and plaque assay. RESULTS: Bioinformatic analysis on the sequences deriving from the primary sample supports the hypothesis of a double viral population within the host. Plaque assay on viral culture led to the isolation of a recombinant strain deriving from Delta and Omicron lineages, named XS, which virtually replaced its parent lineages within a single viral propagation. CONCLUSION: It is impossible to establish whether the recombination event happened within the host or in vitro; however, it is important to monitor co-infections, especially in the exceptional intrahost environment of patients who are immunocompromised, as strong driving forces of viral evolution.


Asunto(s)
COVID-19 , Coinfección , Humanos , SARS-CoV-2/genética , Huésped Inmunocomprometido , Biología Computacional
10.
Viruses ; 15(8)2023 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-37631974

RESUMEN

Since the first SARS-CoV-2 outbreak, mutations such as single nucleotide polymorphisms (SNPs) and insertion/deletions (INDELs) have changed and characterized the viral genome sequence, structure and protein folding leading to the onset of new variants. The presence of those alterations challenges not only the clinical field but also the diagnostic demand due to failures in gene detection or incompleteness of polymerase chain reaction (PCR) results. In particular, the analysis of understudied genes such as N and the investigation through whole-genome next generation sequencing (WG-NGS) of regions more prone to mutate can help in the identification of new or reacquired mutations, with the aim of designing robust and long-lasting primers. In 48 samples of SARS-CoV-2 (including Alpha, Delta and Omicron variants), a lack of N gene amplification was observed in the genomes analyzed through WG-NGS. Three gene regions were detected hosting the highest number of SNPs and INDELs. In several cases, the latter can interfere deeply with both the sensitivity of diagnostic methodologies and the final protein folding. The monitoring over time of the viral evolution and the reacquisition among different variants of the same mutations or different alterations within the same genomic positions can be relevant to avoid unnecessary consumption of resources.


Asunto(s)
SARS-CoV-2 , Humanos , COVID-19/diagnóstico , COVID-19/epidemiología , Prueba de COVID-19 , Genómica , Reacción en Cadena de la Polimerasa , SARS-CoV-2/genética
11.
J Biol Chem ; 286(40): 34514-21, 2011 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-21832076

RESUMEN

Shiga toxins (Stx) play an important role in the pathogenesis of hemolytic uremic syndrome, a life-threatening renal sequela of human intestinal infection caused by specific Escherichia coli strains. Stx target a restricted subset of human endothelial cells that possess the globotriaosylceramide receptor, like that in renal glomeruli. The toxins, composed of five B chains and a single enzymatic A chain, by removing adenines from ribosomes and DNA, trigger apoptosis and the production of pro-inflammatory cytokines in target cells. Because bacteria are confined to the gut, the toxins move to the kidney through the circulation. Polymorphonuclear leukocytes (PMN) have been indicated as the carriers that "piggyback" shuttle toxins to the kidney. However, there is no consensus on this topic, because not all laboratories have been able to reproduce the Stx/PMN interaction. Here, we demonstrate that conformational changes of Shiga toxin 1, with reduction of α-helix content and exposition to solvent of hydrophobic tryptophan residues, cause a loss of PMN binding activity. The partially unfolded toxin was found to express both enzymatic and globotriaosylceramide binding activities being fully active in intoxicating human endothelial cells; this suggests the presence of a distinct PMN-binding domain. By reviewing functional and structural data, we suggest that A chain moieties close to Trp-203 are recognized by PMN. Our findings could help explain the conflicting results regarding Stx/PMN interactions, especially as the groups reporting positive results obtained Stx by single-step affinity chromatography, which could have preserved the correct folding of Stx with respect to more complicated multi-step purification methods.


Asunto(s)
Neutrófilos/citología , Toxina Shiga I/metabolismo , Toxinas Shiga/metabolismo , Adenina/química , Toxinas Bacterianas/metabolismo , Dicroismo Circular , Células Endoteliales/citología , Escherichia coli/genética , Colorantes Fluorescentes/farmacología , Síndrome Hemolítico-Urémico/metabolismo , Humanos , Cinética , Neutrófilos/metabolismo , Conformación Proteica , Estructura Secundaria de Proteína , Ricina/química , Toxina Shiga , Venas Umbilicales/citología
12.
Diagn Microbiol Infect Dis ; 102(1): 115540, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34649189

RESUMEN

Five SARS-CoV-2-positive samples showed N-gene drop-out with a RT-PCR multiplex test. WGS found all samples to harbor a deletion in the same region of the N gene, which is likely to impair the efficiency of amplification. This highlights the need for a continued surveillance of viral evolution and diagnostic test performance.


Asunto(s)
Prueba de COVID-19 , COVID-19/virología , SARS-CoV-2/genética , COVID-19/diagnóstico , Pruebas Diagnósticas de Rutina , Genoma Viral , Humanos , Reacción en Cadena de la Polimerasa Multiplex , Mutación Puntual , Secuenciación Completa del Genoma
13.
Sci Rep ; 12(1): 12479, 2022 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-35864211

RESUMEN

The ongoing evolution of SARS-CoV-2 and the emergence of new viral variants bearing specific escape mutations responsible for immune evasion from antibody neutralisation has required a more accurate characterisation of the immune response as one of the evolutive forces behind viral adaptation to a largely immunised human population. In this work, culturing in the presence of neutralising sera vigorously promoted mutagenesis leading to the acquisition of known escape mutations on the spike as well as new presumptive escape mutations on structural proteins whose role as target of the neutralizing antibody response might have been thus far widely neglected. From this perspective, this study, in addition to tracing the past evolution of the species back to interactions with neutralising antibody immune response, also offers a glimpse into future evolutive scenarios.


Asunto(s)
COVID-19 , SARS-CoV-2 , Anticuerpos Neutralizantes , Anticuerpos Antivirales , COVID-19/genética , Humanos , Mutación , Pruebas de Neutralización , SARS-CoV-2/genética , Glicoproteína de la Espiga del Coronavirus/genética
14.
Microorganisms ; 10(8)2022 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-36013991

RESUMEN

The incidence of total joint arthroplasty is increasing over time since the last decade and expected to be more than 4 million by 2030. As a consequence, the detection of infections associated with surgical interventions is increasing and prosthetic joint infections are representing both a clinically and economically challenging problem. Many pathogens, from bacteria to fungi, elicit the immune system response and produce a polymeric matrix, the biofilm, that serves as their protection. In the last years, the implementation of diagnostic methodologies reduced the error rate and the turn-around time: polymerase chain reaction, targeted or broad-spectrum, and next-generation sequencing have been introduced and they represent a robust approach nowadays that frees laboratories from the unique approach based on culture-based techniques.

15.
Biochem J ; 432(1): 173-80, 2010 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-20809900

RESUMEN

The main cause of acute renal failure in children is HUS (haemolytic uraemic syndrome), a consequence of intestinal infections with Escherichia coli strains producing Stx (Shiga toxins). Stx released in the gut by the non-invasive bacteria reach the bloodstream and are targeted to cerebral and renal endothelium triggering HUS. PMN (polymorphonuclear leucocytes) seem to be involved in Stx delivery through an unidentified membrane receptor (Kd=10⁻8 M; 2×105 binding sites) which does not allow internalization. Some experts in the field have defined the Stx-PMN interaction as non-specific and of little biological significance. In the present study, we show that the A chain of ricin, the well-known plant RIP (ribosome-inactivating protein), interacts with PMN (Kd=10⁻9 M; 2×105 binding sites) competing for the same receptor that recognizes Stx, whereas diphtheria toxin and several agonists of TLRs (Toll-like receptors) or the mannose receptor were ineffective. No toxic effects of ricin A chain on PMN were observed, as assessed by measuring protein synthesis and the rate of spontaneous apoptosis of leucocytes. Moreover, two single-chain RIPs (gelonin and saporin S6) had the same competing effect. Thus RIPs and Stx1 share structural similarities, the same enzymatic activity and a common receptor on PMN. These observations reveal that the Stx-PMN interaction is specific, confirming that PMN recognize molecular patterns common to different foreign molecules.


Asunto(s)
Neutrófilos/metabolismo , Receptores de Superficie Celular/metabolismo , Ricina/metabolismo , Toxina Shiga I/metabolismo , Apoptosis/efectos de los fármacos , Unión Competitiva/efectos de los fármacos , Toxina Diftérica/metabolismo , Toxina Diftérica/farmacología , Citometría de Flujo , Humanos , Radioisótopos de Yodo , Lectinas Tipo C/agonistas , Lectinas Tipo C/metabolismo , Receptor de Manosa , Lectinas de Unión a Manosa/agonistas , Lectinas de Unión a Manosa/metabolismo , Neutrófilos/efectos de los fármacos , Unión Proteica/efectos de los fármacos , Biosíntesis de Proteínas/efectos de los fármacos , Ensayo de Unión Radioligante , Receptores de Superficie Celular/agonistas , Ricina/farmacología , Toxina Shiga I/farmacología , Receptores Toll-Like/agonistas , Receptores Toll-Like/metabolismo
16.
Thromb Haemost ; 120(1): 107-120, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31858520

RESUMEN

Hemolytic uremic syndrome (HUS), the leading cause of acute renal failure in children (< 3 years), is mainly related to Shiga toxins (Stx)-producing Escherichia coli (STEC) infections. STEC are confined to the gut resulting in hemorrhagic colitis, whereas Stx are delivered in blood to target kidney and brain, with unclear mechanisms, triggering HUS in 5 to 15% of infected children. Stx were found on circulating cells, free in sera (soluble Stx) or in blood cell-derived microvesicles (particulate Stx), whereby the relationship between these forms of circulating toxins is unclear. Here, we have examined 2,846 children with bloody diarrhea and found evidence of STEC infection in 5%. Twenty patients were enrolled to study the natural course of STEC infections before the onset of HUS. In patients, Stx were found to be associated to circulating cells and/or free and functionally active in sera. In most children, Stx were bound to neutrophils when high amounts of toxins were found in feces. Time-course analysis showed that Stx increased transiently in patients' sera while the decrease of toxin amount on leukocytes was observed. Notably, patients who recovered (85%) displayed different settings than those who developed HUS (15%). The distinctive feature of the latter group was the presence in blood of particulate Stx2 (Stx2 sedimented at g-forces corresponding to 1 µm microvesicles) the day before diagnosis of HUS, during the release phase of toxins from circulating cells. This observation strongly suggests the involvement of blood cell-derived particulate Stx2 in the transition from hemorrhagic colitis to HUS.


Asunto(s)
Infecciones por Escherichia coli/metabolismo , Síndrome Hemolítico-Urémico/metabolismo , Riñón/metabolismo , Neutrófilos/metabolismo , Material Particulado/sangre , Toxina Shiga II/sangre , Escherichia coli Shiga-Toxigénica/fisiología , Adolescente , Línea Celular , Niño , Preescolar , ADN Bacteriano/genética , Heces/microbiología , Femenino , Humanos , Lactante , Recién Nacido , Riñón/patología , Masculino , Toxina Shiga II/genética
17.
Oxid Med Cell Longev ; 2019: 6528106, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31396304

RESUMEN

In the cold environments of the interstellar medium, a variety of molecules in which a hydrogen (H) atom has been replaced by its heavier isotope deuterium (D) can be found. From its emergence, life had to counteract the toxic action of many agents, which posed a constant threat to its development and propagation. Oxygen-reactive species are archaic toxicants that lead to protein damage and genomic instability. Most of the oxidative lesions involve cleavage of C-H bonds and H abstraction. According to free radical chemistry principles, the substitution of D for H in oxidation-sensitive positions of cellular components should confer protection against the oxidative attack without compromising the chemical identity of the compounds. Here, we show that deuterated nucleosides and proteins protect from oxidative damage. Our data suggest a new, subtle but likely role of D in terrestrial life's evolution in that its inclusion in critical biomolecules might have facilitated their resistance during the infinite generations of life entities, cells, and organisms.


Asunto(s)
Deuterio/química , Estrés Oxidativo , Supervivencia Celular/efectos de los fármacos , Sistema Libre de Células , Daño del ADN/efectos de los fármacos , Radicales Libres/química , Productos Finales de Glicación Avanzada/análisis , Humanos , Células Jurkat , Nucleósidos/química , Nucleósidos/metabolismo , Nucleósidos/farmacología , Estrés Oxidativo/efectos de los fármacos , Carbonilación Proteica , Proteínas/química , Proteínas/metabolismo
18.
Toxins (Basel) ; 10(9)2018 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-30231570

RESUMEN

Shiga toxin 2a (Stx2a) is the main virulence factor produced by pathogenic Escherichia coli strains (Stx-producing E. coli, STEC) responsible for hemorrhagic colitis and the life-threatening sequela hemolytic uremic syndrome in children. The toxin released in the intestine by STEC targets the globotriaosylceramide receptor (Gb3Cer) present on the endothelial cells of the brain and the kidney after a transient blood phase during which Stx2a interacts with blood components, such as neutrophils, which, conversely, recognize Stx through Toll-like receptor 4 (TLR4). Among non-cellular blood constituents, human amyloid P component (HuSAP) is considered a negative modulating factor that specifically binds Stx2a and impairs its toxic action. Here, we show that the soluble extracellular domain of TLR4 inhibits the binding of Stx2a to neutrophils, assessed by indirect flow cytometric analysis. Moreover, by using human sensitive Gb3Cer-expressing cells (Raji cells) we found that the complex Stx2a/soluble TLR4 escaped from capture by HuSAP allowing the toxin to target and damage human cells, as assayed by measuring translation inhibition, the typical Stx-induced functional impairment. Thus, soluble TLR4 stood out as a positive modulating factor for Stx2a. In the paper, these findings have been discussed in the context of the pathogenesis of hemolytic uremic syndrome.


Asunto(s)
Componente Amiloide P Sérico/metabolismo , Toxina Shiga II/toxicidad , Receptor Toll-Like 4/metabolismo , Línea Celular Tumoral , Humanos , Neutrófilos/metabolismo , Dominios Proteicos
19.
Toxins (Basel) ; 7(11): 4564-76, 2015 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-26556372

RESUMEN

Shiga toxins (Stx) have a definite role in the development of hemolytic uremic syndrome in children with hemorrhagic colitis caused by pathogenic Stx-producing Escherichia coli (STEC) strains. The dramatic effects of these toxins on the microvasculature of different organs, particularly of the kidney, are well known, whereas there is no consensus on the mechanism by which Stx reach the endothelia of target organs and/or indirectly injure these body sites. We hereby describe a quick (4 h), radioactive, Raji cell-based method designed for the detection of Stx in human sera. The assay monitors the translation impairment induced by these powerful inhibitors of protein synthesis, which are identified properly by neutralizing their activity with specific monoclonal antibodies. By this method, we detected for the first time the functional activity of Stx in sera of STEC-infected patients during hemorrhagic colitis. Recent research has pointed to a dynamic process of Stx-induced renal intoxication in which concurrent and interactive steps are involved. Our rapid and specific method could be useful for studying the kinetics of Stx during the natural course of STEC infection and the interplay between Stx activity in serum and Stx presence in different blood fractions (neutrophils, monocytes, platelets, leukocyte-platelet aggregates, microvesicles, lipoproteins).


Asunto(s)
Síndrome Hemolítico-Urémico/tratamiento farmacológico , Toxinas Shiga/sangre , Toxinas Shiga/toxicidad , Anticuerpos Monoclonales/análisis , Anticuerpos Monoclonales/farmacología , Línea Celular , Niño , Infecciones por Escherichia coli/sangre , Infecciones por Escherichia coli/tratamiento farmacológico , Síndrome Hemolítico-Urémico/microbiología , Septicemia Hemorrágica/sangre , Humanos , Inhibidores de la Síntesis de la Proteína/sangre , Inhibidores de la Síntesis de la Proteína/farmacología , Inhibidores de la Síntesis de la Proteína/toxicidad , Toxinas Shiga/antagonistas & inhibidores , Escherichia coli Shiga-Toxigénica/patogenicidad
20.
Toxins (Basel) ; 5(2): 431-44, 2013 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-23430607

RESUMEN

Shiga toxin 1 (Stx1), produced by pathogenic Escherichia coli, targets a restricted subset of human cells, which possess the receptor globotriaosylceramide (Gb3Cer/CD77), causing hemolytic uremic syndrome. In spite of the high toxicity, Stx1 has been proposed in the treatment of Gb3Cer/CD77-expressing lymphoma. Here, we demonstrate in a Burkitt lymphoma cell model expressing this receptor, namely Raji cells, that Stx1, at quasi-non-toxic concentrations (0.05-0.1 pM), inhibits the repair of mafosfamide-induced DNA alkylating lesions, synergistically potentiating the cytotoxic activity of the anticancer drug. Conversely, human promyelocytic leukemia cells HL-60, which do not express Gb3Cer/CD77, were spared by the toxin as previously demonstrated for CD34+ human progenitor cells, and hence, in this cancer model, no additive nor synergistic effects were observed with the combined Stx1/mafosfamide treatment. Our findings suggest that Stx1 could be used to improve the mafosfamide-mediated purging of Gb3Cer/CD77+ tumor cells before autologous bone marrow transplantation.


Asunto(s)
Antineoplásicos/administración & dosificación , Ciclofosfamida/análogos & derivados , Inhibidores de la Síntesis de la Proteína/administración & dosificación , Toxina Shiga I/administración & dosificación , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Ciclofosfamida/administración & dosificación , Reparación del ADN/efectos de los fármacos , Sinergismo Farmacológico , Células HL-60 , Células Endoteliales de la Vena Umbilical Humana , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA