Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros










Intervalo de año de publicación
1.
PLoS Pathog ; 19(12): e1011877, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38127952

RESUMEN

Shiga-toxin producing Escherichia coli (STEC) infections can cause from bloody diarrhea to Hemolytic Uremic Syndrome. The STEC intestinal infection triggers an inflammatory response that can facilitate the development of a systemic disease. We report here that neutrophils might contribute to this inflammatory response by secreting Interleukin 1 beta (IL-1ß). STEC stimulated neutrophils to release elevated levels of IL-1ß through a mechanism that involved the activation of caspase-1 driven by the NLRP3-inflammasome and neutrophil serine proteases (NSPs). Noteworthy, IL-1ß secretion was higher at lower multiplicities of infection. This secretory profile modulated by the bacteria:neutrophil ratio, was the consequence of a regulatory mechanism that reduced IL-1ß secretion the higher were the levels of activation of both caspase-1 and NSPs, and the production of NADPH oxidase-dependent reactive oxygen species. Finally, we also found that inhibition of NSPs significantly reduced STEC-triggered IL-1ß secretion without modulating the ability of neutrophils to kill the bacteria, suggesting NSPs might represent pharmacological targets to be evaluated to limit the STEC-induced intestinal inflammation.


Asunto(s)
Infecciones por Escherichia coli , Escherichia coli O157 , Síndrome Hemolítico-Urémico , Interleucina-1beta , Escherichia coli Shiga-Toxigénica , Humanos , Caspasas , Infecciones por Escherichia coli/metabolismo , Infecciones por Escherichia coli/microbiología , Síndrome Hemolítico-Urémico/metabolismo , Síndrome Hemolítico-Urémico/microbiología , Neutrófilos , Interleucina-1beta/metabolismo
2.
Front Pediatr ; 11: 1210158, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37425258

RESUMEN

Introduction: Hemolytic uremic syndrome (HUS) is a condition that results in acute kidney failure mainly in children, which is caused by Shiga toxin-producing Escherichia coli and inflammatory response. Although anti-inflammatory mechanisms are triggered, studies on the implication in HUS are scarce. Interleukin-10 (IL-10) regulates inflammation in vivo, and the interindividual differences in its expression are related to genetic variants. Notably, the single nucleotide polymorphism (SNP) rs1800896 -1082 (A/G), located in the IL-10 promoter, regulates cytokine expression. Methods: Plasma and peripheral blood mononuclear cells (PBMC) were collected from healthy children and HUS patients exhibiting hemolytic anemia, thrombocytopenia, and kidney damage. Monocytes identified as CD14+ cells were analyzed within PBMC by flow cytometry. IL-10 levels were quantified by ELISA, and SNP -1082 (A/G) was analyzed by allele-specific PCR. Results: Circulating IL-10 levels were increased in HUS patients, but PBMC from these patients exhibited a lower capacity to secrete this cytokine compared with those from healthy children. Interestingly, there was a negative association between the circulating levels of IL-10 and inflammatory cytokine IL-8. We observed that circulating IL-10 levels were threefold higher in HUS patients with -1082G allele in comparison to AA genotype. Moreover, there was relative enrichment of GG/AG genotypes in HUS patients with severe kidney failure. Discussion: Our results suggest a possible contribution of SNP -1082 (A/G) to the severity of kidney failure in HUS patients that should be further evaluated in a larger cohort.

3.
Front Cell Infect Microbiol ; 13: 1143918, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37260706

RESUMEN

Introduction: Shiga-toxin (Stx) producing Escherichia coli (STEC) O157:H7 is the most frequent serotype associated with hemolytic uremic syndrome (HUS) after gastrointestinal infections. Protection against HUS secondary to STEC infections has been experimentally assayed through the generation of different vaccine formulations. With focus on patients, the strategies have been mainly oriented to inhibit production of Stx or its neutralization. However, few approaches have been intended to block gastrointestinal phase of this disease, which is considered the first step in the pathogenic cascade of HUS. The aim of this work was to assay H7 flagellin as a mucosal vaccine candidate to prevent the systemic complications secondary to E. coli O157:H7 infections. Materials and methods: The cellular and humoral immune response after H7 nasal immunization in mice were studied by the analysis of systemic and intestinal specific antibody production, as well as cytokine production and lymphocyte proliferation against H7 flagellin ex vivo. Results: Immunized mice developed a strong and specific anti-H7 IgG and IgA response, at systemic and mucosal level, as well as a cellular Th1/Th2/Th17 response. H7 induced activation of bone marrow derived dendritic cells in vitro and a significant delayed-type hypersensitivity (DTH) response in immunized mice. Most relevant, immunized mice were completely protected against the challenge with an E. coli O157:H7 virulent strain in vivo, and surviving mice presented high titres of anti-H7 and Stx antibodies. Discussion: These results suggest that immunization avoids HUS outcome and allows to elicit a specific immune response against other virulence factors.


Asunto(s)
Enfermedades Transmisibles , Infecciones por Escherichia coli , Escherichia coli O157 , Enfermedades Gastrointestinales , Síndrome Hemolítico-Urémico , Escherichia coli Shiga-Toxigénica , Animales , Ratones , Flagelina , Infecciones por Escherichia coli/prevención & control , Inmunización , Síndrome Hemolítico-Urémico/prevención & control
4.
Infect Immun ; 89(5)2021 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-33619029

RESUMEN

Enterohemorrhagic Escherichia coli (EHEC) infections can result in a wide range of clinical presentations despite that EHEC strains belong to the O157:H7 serotype, one of the most pathogenic forms. Although pathogen virulence influences disease outcome, we emphasize the concept of host-pathogen interactions, which involve resistance or tolerance mechanisms in the host that determine total host fitness and bacterial virulence. Taking advantage of the genetic differences between mouse strains, we analyzed the clinical progression in C57BL/6 and BALB/c weaned mice infected with an E. coli O157:H7 strain. We carefully analyzed colonization with several bacterial doses, clinical parameters, intestinal histology, and the integrity of the intestinal barrier, as well as local and systemic levels of antibodies to pathogenic factors. We demonstrated that although both strains had comparable susceptibility to Shiga toxin (Stx) and the intestinal bacterial burden was similar, C57BL/6 showed increased intestinal damage, alteration of the integrity of the intestinal barrier, and impaired renal function that resulted in increased mortality. The increased survival rate in the BALB/c strain was associated with an early specific antibody response as part of a tolerance mechanism.


Asunto(s)
Infecciones por Escherichia coli/inmunología , Infecciones por Escherichia coli/microbiología , Escherichia coli O157/inmunología , Interacciones Huésped-Patógeno , Tolerancia Inmunológica , Animales , Susceptibilidad a Enfermedades , Escherichia coli O157/patogenicidad , Interacciones Huésped-Patógeno/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Toxina Shiga , Especificidad de la Especie , Virulencia
5.
Clin Sci (Lond) ; 135(3): 575-588, 2021 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-33496327

RESUMEN

Hemolytic Uremic Syndrome (HUS), a disease triggered by Shiga toxin (Stx), is characterized by hemolytic anemia, thrombocytopenia and renal failure. The inflammatory response mediated by polymorphonuclear neutrophils (PMNs) and monocytes is essential to HUS onset. Still, the role of anti-inflammatory cytokines is less clear. The deficiency of IL-10, an anti-inflammatory cytokine, leads to severe pathology in bacterial infections but also to beneficial effects in models of sterile injury. The aim of this work was to analyze the role of IL-10 during HUS. Control and IL-10 lacking mice (IL-10-/-) were intravenously injected with Stx type 2 (Stx2) and survival rate was evaluated. PMN and circulating and renal pro- and anti-inflammatory factors were analyzed by FACS and enzyme-linked immunosorbent assay (ELISA) respectively. IL-10-/- mice showed a higher survival associated with lower renal damage reflected by reduced plasma urea and creatinine levels than control mice. Circulating PMN increased at 72 h in both mouse strains accompanied by an up-regulation of CD11b in control mice. In parallel, renal PMN were significantly increased only in control mice after toxin. Plasma TNF-α, IL-6 and corticosterone levels were higher increased in IL-10-/- than control mice. Simultaneously renal TNF-α raised constantly but was accompanied by increased TGF-ß levels in IL-10-/- mice. These results demonstrate that the profile of circulating and renal cytokines after Stx2 differed between strains suggesting that balance of these factors could participate in renal protection. We conclude that IL-10 absence has a protective role in an experimental model of HUS by reducing PMN recruitment into kidney and renal damage, and increasing mice survival.


Asunto(s)
Síndrome Hemolítico-Urémico/inducido químicamente , Interleucina-10/metabolismo , Toxina Shiga II/toxicidad , Animales , Corticosterona/sangre , Síndrome Hemolítico-Urémico/patología , Interleucina-10/genética , Interleucina-6/sangre , Riñón/química , Riñón/patología , Ratones Endogámicos BALB C , Ratones Noqueados , Neutrófilos , Tasa de Supervivencia , Factor de Crecimiento Transformador beta , Factor de Necrosis Tumoral alfa/sangre
6.
Toxins (Basel) ; 12(1)2020 01 14.
Artículo en Inglés | MEDLINE | ID: mdl-31947665

RESUMEN

Enterohemorrhagic Escherichia coli (EHEC) strains are food-borne pathogens that can cause different clinical conditions. Shiga toxin 2a and/or 2c (Stx2)-producing E. coli O157:H7 is the serotype most frequently associated with severe human disease. In this work we analyzed the hypothesis that host cells participate in Stx2 production, cell damage, and inflammation during EHEC infection. With this aim, macrophage-differentiated THP-1 cells and the intestinal epithelial cell line HCT-8 were incubated with E. coli O157:H7. A time course analysis of cellular and bacterial survival, Stx2 production, stx2 transcription, and cytokine secretion were analyzed in both human cell lines. We demonstrated that macrophages are able to internalize and kill EHEC. Simultaneously, Stx2 produced by internalized bacteria played a major role in macrophage death. In contrast, HCT-8 cells were completely resistant to EHEC infection. Besides, macrophages and HCT-8 infected cells produce IL-1ß and IL-8 inflammatory cytokines, respectively. At the same time, bacterial stx2-specific transcripts were detected only in macrophages after EHEC infection. The interplay between bacteria and host cells led to Stx production, triggering of inflammatory response and cell damage, all of which could contribute to a severe outcome after EHEC infections.


Asunto(s)
Escherichia coli O157 , Interacciones Microbiota-Huesped , Inmunomodulación/fisiología , Toxinas Shiga/toxicidad , Línea Celular , Citocinas , Escherichia coli Enterohemorrágica , Infecciones por Escherichia coli , Humanos , Inflamación , Macrófagos
7.
Mol Immunol ; 108: 23-33, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30776726

RESUMEN

Membrane expression of fractalkine (CX3CL1)-receptor (CX3CR1) is relevant in monocytes (Mo) because CX3CR1-CX3CL1 interactions might participate on both, homeostatic and pathologic conditions. We have previously demonstrated that CX3CR1 levels are decreased during culture and when Mo are differentiated into dendritic cells, but enhanced when differentiated into macrophages. Regarding soluble factors, lipopolysaccharide (LPS) accelerated the loss of CX3CR1, while interleukin (IL)-10 and Interferon-gamma (IFN-γ) prevented it. However, the comprehensive knowledge about the intracellular pathways that underlay the level of CX3CR1 expression in Mo is still incomplete. In the current work, we studied the effect of anti-inflammatory cytokines (IL-4, IL-13, IL-10), alone or together with IFN- γ on CX3CR1 expression. We found that only IL-10 and IFN-γ separately were able to prevent CX3CR1 down-modulation during culture of human Mo. Besides, Mo incubated with IL-10 plus IFN-γ showed the highest CX3CR1 expression by cell, suggesting cooperation between two different mechanism used by both cytokines. By studying intracellular mechanisms triggered by IL-10 and IFN-γ, we demonstrated that they specifically induced PI3K-dependent serine-phosphorylation of signal transducer and activator of transcription (STAT)3 or STAT1, respectively. Moreover, chemical inhibitors of STAT1 or STAT3 abrogated IFN-γ or IL-10 effects on CX3CR1 expression. Strikingly, only IL-10 increased CX3CR1 mRNA level, as consequence of augmenting mRNA stability. CX3CR1 mRNA increase was PI3K-dependent, supporting the causal link between the action of IL-10 at the CX3CR1 transcript and CX3CR1 protein level on Mo. Thus, both cytokines up-regulate CX3CR1 expression on human Mo by different intracellular mechanisms.


Asunto(s)
Receptor 1 de Quimiocinas CX3C/metabolismo , Interferón gamma/metabolismo , Interleucina-10/metabolismo , Monocitos/metabolismo , Regulación hacia Arriba , Receptor 1 de Quimiocinas CX3C/genética , Humanos , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factores de Transcripción STAT/metabolismo , Serina/metabolismo
8.
Toxins (Basel) ; 10(12)2018 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-30513821

RESUMEN

Shiga toxin (Stx) producing Escherichia coli (STEC) is responsible for causing hemolytic uremic syndrome (HUS), a life-threatening thrombotic microangiopathy characterized by thrombocytopenia, hemolytic anemia, and acute renal failure after bacterially induced hemorrhagic diarrhea. Until now, there has been neither an effective treatment nor method of prevention for the deleterious effects caused by Stx intoxication. Antibodies are well recognized as affinity components of therapeutic drugs; thus, a previously obtained recombinant human FabC11:Stx2 fragment was used to neutralize Stx2 in vitro in a Vero cell viability assay. Herein, we demonstrated that this fragment neutralized, in a dose-dependent manner, the cytotoxic effects of Stx2 on human glomerular endothelial cells, on human proximal tubular epithelial cells, and prevented the morphological alterations induced by Stx2. FabC11:Stx2 protected mice from a lethal dose of Stx2 by toxin-antibody pre-incubation. Altogether, our results show the ability of a new encouraging molecule to prevent Stx-intoxication symptoms during STEC infection.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Anticuerpos Neutralizantes/farmacología , Fragmentos Fab de Inmunoglobulinas/farmacología , Toxina Shiga II/toxicidad , Animales , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Células Endoteliales/efectos de los fármacos , Humanos , Glomérulos Renales/citología , Ratones Endogámicos BALB C , Proteínas Recombinantes/farmacología
9.
Pediatr Nephrol ; 33(11): 2057-2071, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-29372302

RESUMEN

Hemolytic uremic syndrome (HUS) is defined as a triad of noninmune microangiopathic hemolytic anemia, thrombocytopenia, and acute kidney injury. The most frequent presentation is secondary to Shiga toxin (Stx)-producing Escherichia coli (STEC) infections, which is termed postdiarrheal, epidemiologic or Stx-HUS, considering that Stx is the necessary etiological factor. After ingestion, STEC colonize the intestine and produce Stx, which translocates across the intestinal epithelium. Once Stx enters the bloodstream, it interacts with renal endothelial and epithelial cells, and leukocytes. This review summarizes the current evidence about the involvement of inflammatory components as central pathogenic factors that could determine outcome of STEC infections. Intestinal inflammation may favor epithelial leakage and subsequent passage of Stx to the systemic circulation. Vascular damage triggered by Stx promotes not only release of thrombin and increased fibrin concentration but also production of cytokines and chemokines by endothelial cells. Recent evidence from animal models and patients strongly indicate that several immune cells types may participate in HUS physiopathology: neutrophils, through release of proteases and reactive oxygen species (ROS); monocytes/macrophages through secretion of cytokines and chemokines. In addition, high levels of Bb factor and soluble C5b-9 (sC5b-9) in plasma as well as complement factors adhered to platelet-leukocyte complexes, microparticles and microvesicles, suggest activation of the alternative pathway of complement. Thus, acute immune response secondary to STEC infection, the Stx stimulatory effect on different immune cells, and inflammatory stimulus secondary to endothelial damage all together converge to define a strong inflammatory status that worsens Stx toxicity and disease.


Asunto(s)
Infecciones por Escherichia coli/inmunología , Síndrome Hemolítico-Urémico/inmunología , Microvasos/patología , Escherichia coli Shiga-Toxigénica/inmunología , Animales , Vía Alternativa del Complemento/inmunología , Citocinas/inmunología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Células Endoteliales/inmunología , Células Endoteliales/patología , Endotelio Vascular/citología , Endotelio Vascular/inmunología , Endotelio Vascular/patología , Células Epiteliales/inmunología , Células Epiteliales/patología , Infecciones por Escherichia coli/sangre , Infecciones por Escherichia coli/microbiología , Infecciones por Escherichia coli/patología , Síndrome Hemolítico-Urémico/sangre , Síndrome Hemolítico-Urémico/microbiología , Síndrome Hemolítico-Urémico/patología , Humanos , Mucosa Intestinal/microbiología , Riñón/irrigación sanguínea , Riñón/inmunología , Riñón/patología , Microvasos/citología , Microvasos/inmunología , Escherichia coli Shiga-Toxigénica/aislamiento & purificación
10.
Toxins, v. 10, n. 12, 508, dez. 2018
Artículo en Inglés | Sec. Est. Saúde SP, SESSP-IBPROD, Sec. Est. Saúde SP | ID: bud-2640

RESUMEN

Shiga toxin (Stx) producing Escherichia coli (STEC) is responsible for causing hemolytic uremic syndrome (HUS), a life-threatening thrombotic microangiopathy characterized by thrombocytopenia, hemolytic anemia, and acute renal failure after bacterially induced hemorrhagic diarrhea. Until now, there has been neither an effective treatment nor method of prevention for the deleterious effects caused by Stx intoxication. Antibodies are well recognized as affinity components of therapeutic drugs; thus, a previously obtained recombinant human FabC11:Stx2 fragment was used to neutralize Stx2 in vitro in a Vero cell viability assay. Herein, we demonstrated that this fragment neutralized, in a dose-dependent manner, the cytotoxic effects of Stx2 on human glomerular endothelial cells, on human proximal tubular epithelial cells, and prevented the morphological alterations induced by Stx2. FabC11:Stx2 protected mice from a lethal dose of Stx2 by toxin-antibody pre-incubation. Altogether, our results show the ability of a new encouraging molecule to prevent Stx-intoxication symptoms during STEC infection.

11.
Toxins ; 10(12): 508, 2018.
Artículo en Inglés | Sec. Est. Saúde SP, SESSP-IBPROD, Sec. Est. Saúde SP | ID: but-ib15765

RESUMEN

Shiga toxin (Stx) producing Escherichia coli (STEC) is responsible for causing hemolytic uremic syndrome (HUS), a life-threatening thrombotic microangiopathy characterized by thrombocytopenia, hemolytic anemia, and acute renal failure after bacterially induced hemorrhagic diarrhea. Until now, there has been neither an effective treatment nor method of prevention for the deleterious effects caused by Stx intoxication. Antibodies are well recognized as affinity components of therapeutic drugs; thus, a previously obtained recombinant human FabC11:Stx2 fragment was used to neutralize Stx2 in vitro in a Vero cell viability assay. Herein, we demonstrated that this fragment neutralized, in a dose-dependent manner, the cytotoxic effects of Stx2 on human glomerular endothelial cells, on human proximal tubular epithelial cells, and prevented the morphological alterations induced by Stx2. FabC11:Stx2 protected mice from a lethal dose of Stx2 by toxin-antibody pre-incubation. Altogether, our results show the ability of a new encouraging molecule to prevent Stx-intoxication symptoms during STEC infection.

12.
Curr Pharm Des ; 22(34): 5294-5299, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27356777

RESUMEN

BACKGROUND: Hemolytic Uremic Syndrome (HUS) caused by infections with Shiga toxin (Stx)-producing E. coli is a life-threatening complication characterized by acute renal failure, thrombocytopenia and hemolytic anemia. Stx is the main pathogenic factor. Therefore, the mouse model by intravenous administration of a single lethal dose of Stx is often used to explore its pathogenic mechanisms. OBJECTIVE: The aim of this work was to develop an alternative mouse model of Stx type 2 (Stx2) intoxication to evaluate new therapeutic strategies. METHODS AND RESULTS: One lethal dose of Stx2 was divided in four daily doses. We observed a dose-dependent toxicity characterized by neutrophilia, leukocytopenia and renal damage. Most importantly, we demonstrated that the polyclonal anti-Stx2 serum was able to protect mice from fatal evolution even when administered together the third dose of Stx2. CONCLUSION: This model would provide an advantage for evaluation of therapeutic strategies. Furthermore, the results presented herein suggest that appropriate treatment with anti-Stx2 agents following the appearance of initial clinical signs may block the ongoing outcome or may alleviate disease in patients who have just been diagnosed with HUS. However, the delay in the onset of therapy would be unsafe.


Asunto(s)
Modelos Animales de Enfermedad , Síndrome Hemolítico-Urémico/inducido químicamente , Síndrome Hemolítico-Urémico/tratamiento farmacológico , Toxina Shiga II/toxicidad , Animales , Relación Dosis-Respuesta a Droga , Síndrome Hemolítico-Urémico/patología , Inyecciones Intravenosas , Ratones , Ratones Endogámicos BALB C , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/toxicidad , Toxina Shiga II/administración & dosificación , Toxina Shiga II/inmunología
13.
J Innate Immun ; 8(4): 400-11, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27230920

RESUMEN

Hemolytic uremic syndrome (HUS), a vascular disease characterized by hemolytic anemia, thrombocytopenia, and acute renal failure, is caused by enterohemorrhagic Shiga toxin (Stx)-producing bacteria, which mainly affect children. Besides Stx, the inflammatory response mediated by neutrophils (PMN) is essential to HUS evolution. PMN can release neutrophil extracellular traps (NET) composed of DNA, histones, and other proteins. Since NET are involved in infectious and inflammatory diseases, the aim of this work was to investigate the contribution of NET to HUS. Plasma from HUS patients contained increased levels of circulating free-DNA and nucleosomes in comparison to plasma from healthy children. Neutrophils from HUS patients exhibited a greater capacity to undergo spontaneous NETosis. NET activated human glomerular endothelial cells, stimulating secretion of the proinflammatory cytokines IL-6 and IL-8. Stx induced PMN activation as judged by its ability to trigger reactive oxygen species production, increase CD11b and CD66b expression, and induce NETosis in PMN from healthy donors. During HUS, NET can contribute to the inflammatory response and thrombosis in the microvasculature and thus to renal failure. Intervention strategies to inhibit inflammatory mechanisms mediated by PMN, such as NETosis, could have a potential therapeutic impact towards amelioration of the severity of HUS.


Asunto(s)
Infecciones Bacterianas/inmunología , Células Endoteliales/inmunología , Trampas Extracelulares/inmunología , Síndrome Hemolítico-Urémico/inmunología , Riñón/patología , Neutrófilos/inmunología , Toxina Shiga/inmunología , Lesión Renal Aguda , Anemia Hemolítica , Apoptosis , Células Cultivadas , Niño , Humanos , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Activación Neutrófila , Neutrófilos/microbiología , Especies Reactivas de Oxígeno/metabolismo , Trombocitopenia
14.
Clin Sci (Lond) ; 129(3): 235-44, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25748554

RESUMEN

Haemolytic uraemic syndrome (HUS) is the major complication of Escherichia coli gastrointestinal infections that are Shiga toxin (Stx) producing. Monocytes contribute to HUS evolution by producing cytokines that sensitize endothelial cells to Stx action and migration to the injured kidney. As CC chemokine receptors (CCRs) are involved in monocyte recruitment to injured tissue, we analysed the contribution of these receptors to the pathogenesis of HUS. We analysed CCR1, CCR2 and CCR5 expression in peripheral monocytes from HUS patients during the acute period, with healthy children as controls. We observed an increased expression of CCRs per cell in monocytes from HUS patients, accompanied by an increase in the absolute number of monocytes CCR1+, CCR2+ and CCR5+. It is interesting that prospective analysis confirmed that CCR1 expression positively correlated with HUS severity. The evaluation of chemokine levels in plasma showed that regulated on activation of normal T-cell-expressed and -secreted (RANTES) protein was reduced in plasma from patients with severe HUS, and this decrease correlated with thrombocytopenia. Finally, the expression of the higher CCRs was accompanied by a loss of functionality which could be due to a mechanism for desensitization to compensate for altered receptor expression. The increase in CCR expression correlates with HUS severity, suggesting that the dysregulation of these receptors might contribute to an increased risk of renal damage. Activated monocytes could be recruited by chemokines and then receptors could be dysregulated. The dysregulation of CCRs and their ligands observed during the acute period suggests that a chemokine pathway would participate in HUS development.


Asunto(s)
Quimiocinas/inmunología , Síndrome Hemolítico-Urémico/metabolismo , Monocitos/metabolismo , Receptores de Quimiocina/metabolismo , Movimiento Celular , Niño , Preescolar , Femenino , Expresión Génica/fisiología , Síndrome Hemolítico-Urémico/inmunología , Humanos , Riñón/metabolismo , Masculino , Monocitos/citología , Estudios Prospectivos
15.
Cell Mol Immunol ; 12(6): 669-80, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25502213

RESUMEN

Circulating monocytes (Mos) may continuously repopulate macrophage (MAC) or dendritic cell (DC) populations to maintain homeostasis. MACs and DCs are specialized cells that play different and complementary immunological functions. Accordingly, they present distinct migratory properties. Specifically, whereas MACs largely remain in tissues, DCs are capable of migrating from peripheral tissues to lymphoid organs. The aim of this work was to analyze the expression of the fractalkine receptor (CX3CR1) during the monocytic differentiation process. Freshly isolated Mos express high levels of both CX3CR1 mRNA and protein. During the Mo differentiation process, CX3CR1 is downregulated in both DCs and MACs. However, MACs showed significantly higher CX3CR1 expression levels than did DC. We also observed an antagonistic CX3CR1 regulation by interferon (IFN)-γ and interleukin (IL)-4 during MAC activation through the classical and alternative MAC pathways, respectively. IFN-γ inhibited the loss of CX3CR1, but IL-4 induced it. Additionally, we demonstrated an association between CX3CR1 expression and apoptosis prevention by soluble fractalkine (sCX3CL1) in Mos, DCs and MACs. This is the first report demonstrating sequential and differential CX3CR1 modulation during Mo differentiation. Most importantly, we demonstrated a functional link between CX3CR1 expression and cell survival in the presence of sCX3CL1.


Asunto(s)
Células Dendríticas/citología , Macrófagos/citología , Monocitos/citología , Receptores de Quimiocina/genética , Apoptosis/efectos de los fármacos , Receptor 1 de Quimiocinas CX3C , Diferenciación Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Regulación de la Expresión Génica , Humanos , Interferón gamma/genética , Interferón gamma/inmunología , Interferón gamma/farmacología , Interleucina-4/genética , Interleucina-4/inmunología , Interleucina-4/farmacología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Monocitos/efectos de los fármacos , Monocitos/inmunología , Especificidad de Órganos , Cultivo Primario de Células , Receptores de Quimiocina/inmunología , Transducción de Señal
16.
Med Microbiol Immunol ; 203(3): 145-54, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24399245

RESUMEN

Hemolytic uremic syndrome (HUS) is the major complication of gastrointestinal infections with enterohemorrhagic Escherichia coli (EHEC) and is mediated by the production of Shiga toxins (Stx). Although it has been previously reported that not only HUS patients but healthy children have anti-Stx antibodies, very little is known about how these infections impact on mucosal immune system to generate a specific immune response. This work aimed to evaluate the immune responses elicited after a single oral dose of EHEC in a mouse model of HUS at weaning. We found sequential activation of T and B lymphocytes together with an increased percentage of IgA-bearing B cells in Peyer's patches and mesenteric lymph nodes. We also found fecal anti-EHEC IgA and serum anti-Stx2 IgG in EHEC-inoculated mice. Besides, these mice were partially protected against an intravenous challenge with Stx2. These data demonstrate that one episode of EHEC infection is enough to induce activation in the gut-associated lymphoid tissue, especially the B cell compartment, and lead to the production of specific IgA in mucosal tissue and the generation of systemic protection against Stx2 in a percentage of intragastrically inoculated mice. These data also support the epidemiologic observation that a second episode of HUS is very rare.


Asunto(s)
Sangre/inmunología , Vacunas contra Escherichia coli/administración & dosificación , Vacunas contra Escherichia coli/inmunología , Síndrome Hemolítico-Urémico/prevención & control , Mucosa Intestinal/inmunología , Escherichia coli Shiga-Toxigénica/inmunología , Administración Oral , Animales , Anticuerpos Antibacterianos/análisis , Anticuerpos Antibacterianos/sangre , Linfocitos B/inmunología , Modelos Animales de Enfermedad , Heces/química , Femenino , Inmunoglobulina A/análisis , Inmunoglobulina G/sangre , Masculino , Ratones Endogámicos BALB C , Ganglios Linfáticos Agregados/citología , Ganglios Linfáticos Agregados/inmunología , Suero/química , Linfocitos T/inmunología
17.
Infect Immun ; 82(4): 1491-9, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24421050

RESUMEN

Hemolytic-uremic syndrome (HUS) is defined as the triad of anemia, thrombocytopenia, and acute kidney injury. Enterohemorrhagic Shiga toxin (Stx)-producing Escherichia coli (EHEC), which causes a prodromal hemorrhagic enteritis, remains the most common etiology of the typical or epidemic form of HUS. Because no licensed vaccine or effective therapy is presently available for human use, we recently developed a novel immunogen based on the B subunit of Shiga toxin 2 (Stx2B) and the enzyme lumazine synthase from Brucella spp. (BLS) (BLS-Stx2B). The aim of this study was to analyze maternal immunization with BLS-Stx2B as a possible approach for transferring anti-Stx2 protection to the offspring. BALB/c female mice were immunized with BLS-Stx2B before mating. Both dams and pups presented comparable titers of anti-Stx2B antibodies in sera and fecal extracts. Moreover, pups were totally protected against a lethal dose of systemic Stx2 injection up to 2 to 3 months postpartum. In addition, pups were resistant to an oral challenge with an Stx2-producing EHEC strain at weaning and did not develop any symptomatology associated with Stx2 toxicity. Fostering experiments demonstrated that anti-Stx2B neutralizing IgG antibodies were transmitted through breast-feeding. Pups that survived the EHEC infection due to maternally transferred immunity prolonged an active and specific immune response that protected them against a subsequent challenge with intravenous Stx2. Our study shows that maternal immunization with BLS-Stx2B was very effective at promoting the transfer of specific antibodies, and suggests that preexposure of adult females to this immunogen could protect their offspring during the early phase of life.


Asunto(s)
Infecciones por Escherichia coli/inmunología , Síndrome Hemolítico-Urémico/prevención & control , Inmunidad Materno-Adquirida/inmunología , Inmunización/métodos , Toxina Shiga II/inmunología , Vacunas contra la Shigella/inmunología , Animales , Anticuerpos Antibacterianos/análisis , Brucella/inmunología , Modelos Animales de Enfermedad , Escherichia coli Enterohemorrágica , Femenino , Síndrome Hemolítico-Urémico/microbiología , Ratones , Ratones Endogámicos BALB C , Complejos Multienzimáticos/inmunología , Proteínas Recombinantes de Fusión/inmunología
18.
Pediatr Nephrol ; 21(8): 1082-92, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16703367

RESUMEN

Uninephrectomy (Unx) is followed by the compensatory renal growth (CRG) of the remaining kidney. Previous evidence has shown that during CRG, renal tissue is resistant to a variety of pathologies. We tested the hypothesis that the functional changes that take place during CRG could attenuate Shiga toxin (Stx) toxicity in a mouse model of Stx2-induced hemolytic uremic syndrome (HUS). The participation of nitric oxide (NO) was analyzed. After CRG induction with Unx, mice were exposed to a lethal dose of Stx2, and the degree of renal damage and mortality was measured. Stx2 effects on the growth, renal blood flow (RBF) and NO synthase (NOS) intrarenal expression in the remaining kidney were then studied. The induction of CRG strongly prevented Stx2-mediated mortality and renal damage. Administration of the NOS inhibitor NG-nitro-L-arginine methyl ester (L-NAME) during CRG partially impaired the protection. Both Stx2 and L-NAME interfered with the hypertrophic and hyperplastic responses to Unx, as well as with the increase in RBF. In intact mice, Stx2 decreased renal perfusion, inhibited endothelial NOS basal expression and enhanced inducible NOS expression; all of these effects were attenuated by prior Unx. It is concluded that during CRG mice are highly protected against Stx2 toxicity and lethality. The protective capacity of CRG could be related to the enhancement of renal perfusion and preservation of eNOS renal expression, counterbalancing two major pathogenic mechanisms of Stx2.


Asunto(s)
Síndrome Hemolítico-Urémico/inducido químicamente , Síndrome Hemolítico-Urémico/prevención & control , Riñón/crecimiento & desarrollo , Toxina Shiga II/toxicidad , Animales , Síndrome Hemolítico-Urémico/enzimología , Masculino , Ratones , Óxido Nítrico/fisiología
19.
Buenos Aires; Ministerio de Salud de la Nación; 2004. (120386).
Monografía en Español | ARGMSAL | ID: biblio-993395

Asunto(s)
Becas
20.
Buenos Aires; Ministerio de Salud de la Nación; 2003. (120385).
Monografía en Español | ARGMSAL | ID: biblio-993396

RESUMEN

El objetivo del trabajo consiste en estudiar los mecanismos anti-inflamatorios endógenos en la modulación de la respuesta inflamatoria durante el SUH. Dentro de este contexto el objetivo particular es analizar los mecanismos a través de los cuales los glucocorticoides (GC) disminuyen la toxicidad inducida por la toxina Shiga tipo 2 (Stx2).


Asunto(s)
Síndrome Hemolítico-Urémico , Becas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...