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

País de afiliação
Intervalo de ano de publicação
1.
Rev Argent Microbiol ; 56(1): 25-32, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-37704516

RESUMO

In Argentina, hemolytic uremic syndrome (HUS) caused by EHEC has the highest incidence in the world. EHEC infection has an endemo-epidemic behavior, causing 20-30% of acute bloody diarrhea syndrome in children under 5 years old. In the period 2016-2020, 272 new cases per year were notified to the National Health Surveillance System. Multiple factors are responsible for HUS incidence in Argentina including person-to-person transmission. In order to detect possible EHEC carriers, we carried out a preliminary study of the frequency of kindergarten teachers with anti-LPS antibodies against the most prevalent EHEC serotypes in Argentina. We analyzed 61 kindergarten teachers from 26 institutions from José C. Paz district, located in the suburban area of Buenos Aires province, Argentina. Fifty-one percent of the plasma samples had antibodies against O157, O145, O121 and O103 LPS: 6.4% of the positive samples had IgM isotype (n=2), 61.3% IgG isotype (n=19) and 32.3% IgM and IgG (n=10). Given that antibodies against LPS antigens are usually short-lived specific IgM detection may indicate a recent infection. In addition, the high percentage of positive samples may indicate a frequent exposure to EHEC strains in the cohort studied, as well as the existence of a large non-symptomatic population of adults carrying pathogenic strains that could contribute to the endemic behavior through person-to-person transmission. The improvement of continuous educational programs in kindergarten institutions could be a mandatory measure to reduce HUS cases not only in Argentina but also globally.


Assuntos
Escherichia coli Êntero-Hemorrágica , Infecções por Escherichia coli , Síndrome Hemolítico-Urêmica , Criança , Adulto , Humanos , Pré-Escolar , Lipopolissacarídeos , Infecções por Escherichia coli/epidemiologia , Diarreia/epidemiologia , Síndrome Hemolítico-Urêmica/epidemiologia , Imunoglobulina G , Imunoglobulina M
2.
Pediatr Nephrol ; 38(3): 739-748, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-35802271

RESUMO

BACKGROUND: Hemolytic uremic syndrome (HUS) is characterized by microangiopathic hemolysis, thrombocytopenia, and thrombus formation leading to tissue injury. HUS is classified according to its etiology as post-diarrheal or atypical HUS. Differential diagnosis of both entities continues to be a challenge for pediatric physicians. METHODS: The aim was to improve the rapid etiological diagnosis of post-diarrheal HUS cases based on the detection of Shiga toxin (Stx)-producing Escherichia coli (STEC) infection by screening of stx1/stx2 and rfbO157 in cultured stools by multiplex PCR, and the additional detection of anti-lipopolysaccharide (anti-LPS) O157, O145, and O121 antibodies by Glyco-iELISA test. In addition, we studied patients' relatives to detect circulating pathogenic strains that could contribute to HUS diagnosis and/or lead to the implementation of measures to prevent dissemination of familial outbreaks. This study describes the diagnosis of 31 HUS patients admitted to Hospital Municipal de Niños Prof Dr Ramón Exeni during the 2017-2020 period. RESULTS: Stool PCR confirmed the diagnosis of STEC associated with HUS in 38.7% of patients (12/31), while anti-LPS serology did in 88.9% (24/27). In those patients in which both methods were carried out (n = 27), a strong association between the results obtained was found. We found that 30.4% of HUS patients had at least one relative positive for STEC. CONCLUSIONS: We could identify 96.3% (26/27) of HUS cases as secondary to STEC infections when both methods (genotyping and serology) were used. The results demonstrated a high circulation of STEC in HUS families and the prevalence of the STEC O157 serotype (83%) in our pediatric cohort. A higher-resolution version of the Graphical abstract is available as Supplementary information.


Assuntos
Síndrome Hemolítico-Urêmica Atípica , Infecções por Escherichia coli , Escherichia coli Shiga Toxigênica , Criança , Humanos , Diarreia/diagnóstico , Diarreia/etiologia , Diarreia/epidemiologia , Sorogrupo , Lipopolissacarídeos , Anticorpos Antibacterianos , Infecções por Escherichia coli/complicações , Infecções por Escherichia coli/diagnóstico , Infecções por Escherichia coli/epidemiologia
3.
Clin Sci (Lond) ; 134(24): 3283-3301, 2020 12 23.
Artigo em Inglês | MEDLINE | ID: mdl-33346356

RESUMO

Host adaptation of pathogens may increase intra- and interspecies transmission. We showed previously that the passage of a clinically isolated enterohemorrhagic Escherichia coli (EHEC) O157 strain (125/99) through the gastrointestinal tract of mice increases its pathogenicity in the same host. In this work, we aimed to elucidate the underlying mechanism(s) involved in the patho-adaptation of the stool-recovered (125RR) strain. We assessed the global transcription profile by microarray and found almost 100 differentially expressed genes in 125RR strain compared with 125/99 strain. We detected an overexpression of Type Three Secretion System (TTSS) proteins at the mRNA and protein levels and demonstrated increased adhesion to epithelial cell lines for the 125RR strain. Additional key attributes of the 125RR strain were: increased motility on semisolid agar, which correlated with an increased fliC mRNA level; reduced Stx2 production at the mRNA and protein levels; increased survival at pH 2.5, as determined by acid resistance assays. We tested whether the overexpression of the LEE-encoded regulator (ler) in trans in the 125/99 strain could recreate the increased pathogenicity observed in the 125RR strain. As anticipated ler overexpression led to increased expression of TTSS proteins and bacterial adhesion to epithelial cells in vitro but also increased mortality and intestinal colonization in vivo. We conclude that this host-adaptation process required changes in several mechanisms that improved EHEC O157 fitness in the new host. The research highlights some of the bacterial mechanisms required for horizontal transmission of these zoonotic pathogens between their animal and human populations.


Assuntos
Adaptação Fisiológica , Microambiente Celular , Escherichia coli O157/fisiologia , Intestinos/microbiologia , Animais , Sistemas de Secreção Bacterianos/genética , Escherichia coli O157/genética , Escherichia coli O157/patogenicidade , Feminino , Regulação Bacteriana da Expressão Gênica , Masculino , Camundongos Endogâmicos C57BL , Fenótipo , Virulência
4.
Medicina (B Aires) ; 77(3): 185-190, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28643674

RESUMO

Shiga toxin (Stx)-producing Escherichia coli (STEC) infections are implicated in the development of the life-threatening hemolytic-uremic syndrome (HUS). Despite the magnitude of the social and economic problems caused by HUS, no licensed vaccine or effective therapy is currently available for human use. Prevention of STEC infections continues being the most important measure to reduce HUS incidence. This is especially true for Argentina where HUS incidence among children is extremely high and shows an endemic pattern. The aim of this work was to investigate serologically adult staff of kindergartens in Buenos Aires city and suburban areas in order to detect possible carriers, and to educate personnel about good practices to reduce HUS transmission. We also assessed the microbiological quality of water and meal samples from the same kindergartens. We tested 67 healthy adults, 13 water supplies and 6 meals belonging to 6 public kindergartens. We analysed hand swabs for isolation of STEC and serum samples for the presence of antibodies against Stx and lipopolysaccharide (LPS) of O157 serogroup. We identified 46 Stx2-positive individuals, but only 7 for O157 LPS. No presence of STEC pathogens was detected in hands of staff, water or meal samples.


Assuntos
Infecções por Escherichia coli/prevenção & controle , Escherichia coli O157/isolamento & purificação , Síndrome Hemolítico-Urêmica/microbiologia , Síndrome Hemolítico-Urêmica/prevenção & controle , Adulto , Argentina/epidemiologia , Criança , Surtos de Doenças , Eletroforese , Infecções por Escherichia coli/epidemiologia , Infecções por Escherichia coli/microbiologia , Infecções por Escherichia coli/transmissão , Síndrome Hemolítico-Urêmica/sangue , Síndrome Hemolítico-Urêmica/epidemiologia , Humanos , Fatores de Risco , Sorotipagem , População Urbana
5.
Antimicrob Agents Chemother ; 60(1): 459-70, 2016 01.
Artigo em Inglês | MEDLINE | ID: mdl-26525795

RESUMO

A subset of Gram-negative bacterial pathogens uses a type III secretion system (T3SS) to open up a conduit into eukaryotic cells in order to inject effector proteins. These modulate pathways to enhance bacterial colonization. In this study, we screened established bioactive compounds for any that could repress T3SS expression in enterohemorrhagic Escherichia coli (EHEC) O157. The ketolides telithromycin and, subsequently, solithromycin both demonstrated repressive effects on expression of the bacterial T3SS at sub-MICs, leading to significant reductions in bacterial binding and actin-rich pedestal formation on epithelial cells. Preincubation of epithelial cells with solithromycin resulted in significantly less attachment of E. coli O157. Moreover, bacteria expressing the T3SS were more susceptible to solithromycin, and there was significant preferential killing of E. coli O157 bacteria when they were added to epithelial cells that had been preexposed to the ketolide. This killing was dependent on expression of the T3SS. Taken together, this research indicates that the ketolide that has accumulated in epithelial cells may traffic back into the bacteria via the T3SS. Considering that neither ketolide induces the SOS response, nontoxic members of this class of antibiotics, such as solithromycin, should be considered for future testing and trials evaluating their use for treatment of EHEC infections. These antibiotics may also have broader significance for treating infections caused by other pathogenic bacteria, including intracellular bacteria, that express a T3SS.


Assuntos
Antibacterianos/farmacologia , Escherichia coli O157/efeitos dos fármacos , Cetolídeos/farmacologia , Macrolídeos/farmacologia , Bibliotecas de Moléculas Pequenas/farmacologia , Triazóis/farmacologia , Sistemas de Secreção Tipo III/antagonistas & inibidores , Animais , Antibacterianos/química , Aderência Bacteriana/efeitos dos fármacos , Células CACO-2 , Bovinos , Linhagem Celular , Descoberta de Drogas , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/microbiologia , Escherichia coli O157/genética , Escherichia coli O157/metabolismo , Expressão Gênica , Ensaios de Triagem em Larga Escala , Humanos , Cetolídeos/química , Macrolídeos/química , Testes de Sensibilidade Microbiana , Mucosa Respiratória/efeitos dos fármacos , Mucosa Respiratória/microbiologia , Triazóis/química , Sistemas de Secreção Tipo III/genética , Sistemas de Secreção Tipo III/metabolismo
6.
Int J Med Microbiol ; 305(8): 910-7, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26456732

RESUMO

Shiga toxin-producing Escherichia coli (STEC) is a food-borne pathogen that causes hemorrhagic colitis. Under some circumstances, Shiga toxin (Stx) produced within the intestinal tract enters the bloodstream, leading to systemic complications that may cause the potentially fatal hemolytic-uremic syndrome (HUS). Despite STEC human infection is characterized by acute inflammation of the colonic mucosa, little is known regarding the role of proinflammatory mediators like cysteine leukotrienes (cysLTs) in this pathology. Thus, the aim of this work was to analyze whether leukotriene C4 (LTC4) influences STEC pathogenesis in mice. We report that exogenous LTC4 pretreatment severely affected the outcome of STEC gastrointestinal infection. LTC4-pretreated (LTC4+) and STEC-infected (STEC+) mice showed an increased intestinal damage by histological studies, and a decreased survival compared to LTC4-non-pretreated (LTC4-) and STEC+ mice. LTC4+/STEC+ mice that died after the infection displayed neutrophilia and high urea levels, indicating that the cause of death was related to Stx2-toxicity. Despite the differences observed in the survival between LTC4+ and LTC4- mice after STEC infection, both groups showed the same survival after Stx2-intravenous inoculation. In addition, LTC4 pretreatment increased the permeability of mucosal intestinal barrier, as assessed by FITC-dextran absorption experiments. Altogether these results suggest that LTC4 detrimental effect on STEC infection is related to the increased passage of pathogenic factors to the bloodstream. Finally, we showed that STEC infection per se increases the endogenous LTC4 levels in the gut, suggesting that this inflammatory mediator plays a role in the pathogenicity of STEC infection in mice, mainly by disrupting the mucosal epithelial barrier.


Assuntos
Suscetibilidade a Doenças , Infecções por Escherichia coli/complicações , Síndrome Hemolítico-Urêmica/microbiologia , Síndrome Hemolítico-Urêmica/patologia , Leucotrieno C4/metabolismo , Escherichia coli Shiga Toxigênica/patogenicidade , Animais , Modelos Animais de Doenças , Infecções por Escherichia coli/imunologia , Infecções por Escherichia coli/microbiologia , Intestinos/patologia , Camundongos Endogâmicos BALB C , Análise de Sobrevida
7.
J Immunol ; 191(5): 2403-11, 2013 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-23918978

RESUMO

The striking feature of enterohemorrhagic Escherichia coli (EHEC) infections is the production of Shiga toxins (Stx) implicated in the development of the life-threatening hemolytic uremic syndrome. Despite the magnitude of the social impact of EHEC infections, no licensed vaccine or effective therapy is available for human use. One of the biggest challenges is to develop an effective and safe immunogen to ensure nontoxicity, as well as a strong input to the immune system to induce long-lasting, high-affinity Abs with anti-Stx-neutralizing capacity. The enzyme lumazine synthase from Brucella spp. (BLS) is a highly stable dimer of pentamers and a scaffold with enormous plasticity on which to display foreign Ags. Taking into account the advantages of BLS and the potential capacity of the B subunit of Stx2 to induce Abs that prevent Stx2 toxicity by blocking its entrance into the host cells, we engineered a new immunogen by inserting the B subunit of Stx2 at the amino termini of BLS. The resulting chimera demonstrated a strong capacity to induce a long-lasting humoral immune response in mice. The chimera induced Abs with high neutralizing capacity for Stx2 and its variants. Moreover, immunized mice were completely protected against i.v. Stx2 challenge, and weaned mice receiving an oral challenge with EHEC were completely protected by the transference of immune sera. We conclude that this novel immunogen represents a promising candidate for vaccine or Ab development with preventive or therapeutic ends, for use in hemolytic uremic syndrome-endemic areas or during future outbreaks caused by pathogenic strains of Stx-producing E. coli.


Assuntos
Síndrome Hemolítico-Urêmica/prevenção & controle , Complexos Multienzimáticos/imunologia , Toxina Shiga II/imunologia , Vacinas contra Shigella/imunologia , Animais , Anticorpos Antibacterianos/sangue , Anticorpos Neutralizantes/sangue , Brucella , Modelos Animais de Doenças , Escherichia coli Êntero-Hemorrágica , Feminino , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Complexos Multienzimáticos/química , Proteínas Recombinantes/química , Proteínas Recombinantes/imunologia , Toxina Shiga II/química
8.
Infect Immun ; 82(9): 3948-57, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25001607

RESUMO

Enterohemorrhagic Escherichia coli (EHEC) is a food-borne pathogen that produces Shiga toxin (Stx) and causes hemorrhagic colitis. Under some circumstances, Stx produced within the intestinal tract enters the bloodstream, leading to systemic complications that may cause the potentially fatal hemolytic-uremic syndrome. Although retinoids like vitamin A (VA) and retinoic acid (RA) are beneficial to gut integrity and the immune system, the effect of VA supplementation on gastrointestinal infections of different etiologies has been controversial. Thus, the aim of this work was to study the influence of different VA status on the outcome of an EHEC intestinal infection in mice. We report that VA deficiency worsened the intestinal damage during EHEC infection but simultaneously improved survival. Since death is associated mainly with Stx toxicity, Stx was intravenously inoculated to analyze whether retinoid levels affect Stx susceptibility. Interestingly, while VA-deficient (VA-D) mice were resistant to a lethal dose of Stx2, RA-supplemented mice were more susceptible to it. Given that peripheral blood polymorphonuclear cells (PMNs) are known to potentiate Stx2 toxicity, we studied the influence of retinoid levels on the absolute number and function of PMNs. We found that VA-D mice had decreased PMN numbers and a diminished capacity to produce reactive oxygen species, while RA supplementation had the opposite effect. These results are in line with the well-known function of retinoids in maintaining the homeostasis of the gut but support the idea that they have a proinflammatory effect by acting, in part, on the PMN population.


Assuntos
Escherichia coli Êntero-Hemorrágica/metabolismo , Infecções por Escherichia coli/metabolismo , Retinoides/metabolismo , Toxina Shiga II/metabolismo , Animais , Modelos Animais de Doenças , Infecções por Escherichia coli/microbiologia , Mucosa Intestinal/metabolismo , Intestinos/microbiologia , Camundongos , Camundongos Endogâmicos BALB C , Espécies Reativas de Oxigênio/metabolismo , Vitamina A/metabolismo
9.
IUBMB Life ; 65(10): 827-35, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24014500

RESUMO

Shiga toxin (Stx) producing Escherichia coli (STEC) is responsible to bloody diarrhea (hemorrhagic colitis) and the hemolytic uremic syndrome (HUS). STEC strains carry inducible lambda phages integrated into their genomes that encode Stx 1 and/or 2, with several allelic variants each one. O157:H7 is the serotype that was documented in the vast majority of HUS cases although non-O157 serotypes have been increasingly reported to account for HUS cases. However, the outbreak that occurred in central Europe during late spring of 2011 showed that the pathogen was E. coli O104:H4. More than 4,000 persons were infected mainly in Germany, and it produced more than 900 cases of HUS resulting in 54 deaths. E. coli O104:H4 is a hybrid organism that combines some of the virulence genes of STEC and enteroaggregative E. coli specially production of Stx2 and the adherence mechanisms to intestinal epithelium. The differences in the epidemiology and presentation of E. coli pathogen meant a challenge for public health and scientific research to increase the knowledge of HUS-pathophysiology and to improve available therapies to treat HUS.


Assuntos
Diarreia/genética , Síndrome Hemolítico-Urêmica/genética , Toxina Shiga II/genética , Escherichia coli Shiga Toxigênica/genética , Bacteriófago lambda/genética , Bacteriófago lambda/patogenicidade , Diarreia/epidemiologia , Diarreia/microbiologia , Diarreia/patologia , Surtos de Doenças , Alemanha/epidemiologia , Síndrome Hemolítico-Urêmica/epidemiologia , Síndrome Hemolítico-Urêmica/microbiologia , Síndrome Hemolítico-Urêmica/patologia , Humanos , Toxina Shiga II/metabolismo , Escherichia coli Shiga Toxigênica/patogenicidade , Virulência/genética
10.
J Clin Immunol ; 32(3): 622-31, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22228570

RESUMO

PURPOSE: The interaction of Shiga toxin (Stx) and/or lipopolysaccharide (LPS) with monocytes (Mo) may be central to the pathogenesis of hemolytic uremic syndrome (HUS), providing the cytokines necessary to sensitize endothelial cells to Stx action. We have previously demonstrated phenotypical alterations in Mo from HUS patients, including increased number of CD16+ Mo. Our aim was to investigate cytokine production in Mo from HUS patients. METHODS: We evaluated TNF-α and IL-10 intracellular contents and secretion in the different Mo subsets in mild (HUS 1) and moderate/severe (HUS 2 + 3) patients. As controls, we studied healthy (HC) and infected children (IC). We also studied Mo responsive capacity towards LPS, measuring the modulation of Mo surface molecules and cytokine production. RESULTS: In basal conditions, the intracellular measurement of TNF-α and IL-10 revealed that the highest number of cytokine-producing Mo was found in HUS 2 + 3 and IC, whereas LPS caused a similar increase in TNF-α and IL-10-producing Mo for all groups. However, when evaluating the release of TNF-α and IL-10, we found a diminished secretion capacity in the entire HUS group and IC compared to HC in basal and LPS conditions. Similarly, a lower Mo response to LPS in HUS 2 + 3 and IC groups was observed when surface markers were studied. CONCLUSION: These results indicate that Mo from severe cases of HUS, similar to IC but different to mild HUS cases, present functional changes in Mo subpopulations and abnormal responses to LPS.


Assuntos
Síndrome Hemolítico-Urêmica/imunologia , Interleucina-10/imunologia , Monócitos/imunologia , Fator de Necrose Tumoral alfa/imunologia , Criança , Pré-Escolar , Feminino , Humanos , Lactente , Interleucina-10/sangue , Lipopolissacarídeos/imunologia , Masculino , Fator de Necrose Tumoral alfa/sangue
11.
Infect Immun ; 79(3): 1280-8, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21173307

RESUMO

Severe sepsis is associated with early release of inflammatory mediators that contribute to the morbidity and mortality observed during the first stages of this syndrome. Although sepsis is a deadly, acute disease, high mortality rates have been observed in patients displaying evidence of sepsis-induced immune deactivation. Although the contribution of experimental models to the knowledge of pathophysiological and therapeutic aspects of human sepsis is undeniable, most of the current studies using animal models have focused on the acute, proinflammatory phase. We developed a murine model that reproduces the early acute phases but also the long-term consequences of human sepsis. We induced polymicrobial acute peritonitis (AP) by establishing a surgical connection between the cecum and the peritoneum, allowing the exit of intestinal bacteria. Using this model, we observed an acute phase with high mortality, leukopenia, increased interleukin-6 levels, bacteremia, and neutrophil activation. A peak of leukocytosis on day 9 or 10 revealed the persistence of the infection within the lung and liver, with inflammatory hepatic damage being shown by histological examination. Long-term (20 days) derangements in both innate and adaptive immune responses were found, as demonstrated by impaired systemic tumor necrosis factor alpha production in response to an inflammatory stimulus; a decreased primary humoral immune response and T cell proliferation, associated with an increased number of myeloid suppressor cells (Gr-1(+) CD11b(+)) in the spleen; and a low clearance capacity. This model provides a good approach to attempt novel therapeutic interventions directed to augmenting host immunity during late sepsis.


Assuntos
Modelos Animais de Doenças , Peritonite/complicações , Peritonite/imunologia , Sepse/etiologia , Sepse/imunologia , Animais , Ceco/microbiologia , Ceco/cirurgia , Citocinas/imunologia , Inflamação/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Peritônio/cirurgia , Peritonite/microbiologia , Sepse/microbiologia
12.
Medicina (B Aires) ; 71(4): 383-9, 2011.
Artigo em Espanhol | MEDLINE | ID: mdl-21893458

RESUMO

The typical form of hemolytic uremic syndrome (HUS) is the major complication of Shiga toxin-producing Escherichia coli (STEC) infections. HUS is a critical health problem in Argentina since it is the main cause of acute renal failure in children and the second cause of chronic renal failure, giving account for 20% of renal transplants in children and adolescents in our country. In spite of the extensive research in the field, the mainstay of treatment for patients with HUS is supportive therapy, and there are no specific therapies preventing or ameliorating the disease course. In this review, we present the current knowledge about pathogenic mechanisms and discuss traditional and innovative therapeutic approaches, with special focus in national status and contributions made by Argentinean groups.


Assuntos
Síndrome Hemolítico-Urêmica , Escherichia coli Shiga Toxigênica , Argentina/epidemiologia , Síndrome Hemolítico-Urêmica/epidemiologia , Síndrome Hemolítico-Urêmica/microbiologia , Síndrome Hemolítico-Urêmica/terapia , Humanos , Escherichia coli Shiga Toxigênica/patogenicidade
13.
Infect Immun ; 78(3): 1193-201, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20008539

RESUMO

Hemolytic-uremic syndrome (HUS) is generally caused by Shiga toxin (Stx)-producing Escherichia coli. Endothelial dysfunction mediated by Stx is a central aspect in HUS development. However, inflammatory mediators such as bacterial lipopolysaccharide (LPS) and polymorphonuclear neutrophils (PMN) contribute to HUS pathophysiology by potentiating Stx effects. Acute renal failure is the main feature of HUS, but in severe cases, patients can develop neurological complications, which are usually associated with death. Although the mechanisms of neurological damage remain uncertain, alterations of the blood-brain barrier associated with brain endothelial injury is clear. Astrocytes (ASTs) are the most abundant inflammatory cells of the brain that modulate the normal function of brain endothelium and neurons. The aim of this study was to evaluate the effects of Stx type 1 (Stx1) alone or in combination with LPS in ASTs. Although Stx1 induced a weak inflammatory response, pretreatment with LPS sensitized ASTs to Stx1-mediated effects. Moreover, LPS increased the level of expression of the Stx receptor and its internalization. An early inflammatory response, characterized by the release of tumor necrosis factor alpha (TNF-alpha) and nitric oxide and PMN-chemoattractant activity, was induced by Stx1 in LPS-sensitized ASTs, whereas activation, evidenced by higher levels of glial fibrillary acid protein and cell death, was induced later. Furthermore, increased adhesion and PMN-mediated cytotoxicity were observed after Stx1 treatment in LPS-sensitized ASTs. These effects were dependent on NF-kappaB activation or AST-derived TNF-alpha. Our results suggest that TNF-alpha is a pivotal effector molecule that amplifies Stx1 effects on LPS-sensitized ASTs, contributing to brain inflammation and leading to endothelial and neuronal injury.


Assuntos
Astrócitos/efeitos dos fármacos , Astrócitos/imunologia , Lipopolissacarídeos/imunologia , Toxina Shiga I/imunologia , Toxina Shiga I/toxicidade , Fator de Necrose Tumoral alfa/imunologia , Animais , Células Cultivadas , Escherichia coli/patogenicidade , Mediadores da Inflamação/metabolismo , Óxido Nítrico/metabolismo , Ratos
14.
Immunology ; 129(4): 600-9, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20102414

RESUMO

The membrane-anchored form of the chemokine fractalkine (CX(3)CL1) has been identified as a novel adhesion molecule that interacts with its specific receptor (CX(3)CR1) expressed in monocytes, T cells and natural killer cells to induce adhesion. In addition, CX(3)CL1 can be cleaved from the cell membrane to induce chemotaxis of CX(3)CR1-expressing leucocytes. Recently, marked variations in CX(3)CR1 monocyte expression have been observed during several pathological conditions. Regulation of CX(3)CR1 in monocytes during basal or inflammatory/anti-inflammatory conditions is poorly understood. The aim of this study was therefore to examine CX(3)CR1 expression during monocyte maturation and the effect of soluble mediators on this process. We found that basal expression of CX(3)CR1 in fresh monocytes was reduced during culture, and that lipopolysacchairde accelerated this effect. In contrast, interleukin-10 and interferon-gamma treatment abrogated CX(3)CR1 down-modulation, through a phosphatidylinositol 3 kinase-dependent pathway. Most importantly, CX(3)CR1 membrane expression correlated with monocyte CX(3)CL1-dependent function. Taken together, our data demonstrate that CX(3)CR1 expression in monocytes can be modulated, and suggest that alterations in their environment are able to influence CX(3)CL1-dependent functions, such as chemotaxis and adhesion, leading to changes in the kinetics, composition and/or functional status of the leucocyte infiltrate.


Assuntos
Interferon gama/farmacologia , Interleucina-10/farmacologia , Monócitos/efeitos dos fármacos , Monócitos/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Receptores de Quimiocinas/biossíntese , Receptor 1 de Quimiocina CX3C , Células Cultivadas , Relação Dose-Resposta a Droga , Humanos , Interferon gama/imunologia , Interleucina-10/imunologia , Monócitos/imunologia , Receptores de Quimiocinas/imunologia , Fatores de Tempo
15.
Vaccine ; 36(13): 1728-1735, 2018 03 20.
Artigo em Inglês | MEDLINE | ID: mdl-29483033

RESUMO

E. coli O157:H7 is a foodborne pathogen responsible for bloody diarrhea, hemorrhagic colitis and hemolytic uremic syndrome (HUS). The objective of the present work was to evaluate the ability of colostral IgG obtained from Stx2-immunized cows to prevent against E. coli O157:H7 infection and Stx2 cytotoxicity. Hyperimmune colostrum (HC) was obtained from cows intramuscularly immunized with inactivated Stx2 or vehicle for controls. Colostral IgG was purified by affinity chromatography. Specific IgG antibodies against Stx2 and bovine lactoferrin (bLF) levels in HC and the corresponding IgG (HC-IgG/bLF) were determined by ELISA. The protective effects of HC-IgG/bLF against Stx2 cytotoxicity and adhesion of E. coli O157:H7 and its Stx2-negative mutant were analyzed in HCT-8 cells. HC-IgG/bLF prevention against E. coli O157:H7 was studied in human colon and rat colon loops. Protection against a lethal dose of E. coli O157:H7 was evaluated in a weaned mice model. HC-IgG/bLF showed high anti-Stx2 titers and high bLF levels that were able to neutralize the cytotoxic effects of Stx2 in vitro and in vivo. Furthermore, HC-IgG/bLF avoided the inhibition of water absorption induced by E. coli O157:H7 in human colon and also the pathogenicity of E. coli O157:H7 and E. coli O157:H7Δstx2 in rat colon loops. Finally, HC-IgG/bLF prevented in a 100% the lethality caused by E. coli O157:H7 in a weaned mice model. Our study suggests that HC-IgG/bLF have protective effects against E. coli O157:H7 infection. These beneficial effects may be due to specific anti-Stx2 neutralizing antibodies in combination with high bLF levels. These results allow us to consider HC-IgG/bLF as a nutraceutical tool which could be used in combination with balanced supportive diets to prevent HUS. However further studies are required before recommendations can be made for therapeutic and clinical applications.


Assuntos
Anticorpos Antibacterianos/imunologia , Anticorpos Neutralizantes/imunologia , Doenças dos Bovinos/imunologia , Doenças dos Bovinos/microbiologia , Infecções por Escherichia coli/veterinária , Escherichia coli O157/imunologia , Lactoferrina/biossíntese , Toxina Shiga II/imunologia , Animais , Anticorpos Antibacterianos/biossíntese , Anticorpos Neutralizantes/biossíntese , Especificidade de Anticorpos/imunologia , Bovinos , Linhagem Celular Tumoral , Colo/imunologia , Colo/metabolismo , Colo/microbiologia , Colo/patologia , Escherichia coli O157/patogenicidade , Feminino , Síndrome Hemolítico-Urêmica/veterinária , Humanos , Imunização , Imunoglobulina G/imunologia , Masculino , Camundongos , Testes de Neutralização , Gravidez , Ratos
16.
Front Microbiol ; 9: 3104, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30619183

RESUMO

Hemolytic uremic syndrome (HUS), principally caused by shiga toxins (Stxs), is associated with Shiga toxin-producing Escherichia coli (STEC) infections. We previously reported Stx2 expression by host cells in vitro and in vivo. As the genes encoding the two Stx subunits are located in bacteriophage genomes, the aim of the current study was to evaluate the role of bacteriophage induction in HUS development in absence of an E. coli O157:H7 genomic background. Mice were inoculated with a non-pathogenic E. coli strain carrying the lysogenic bacteriophage 933W (C600Φ933W), and bacteriophage excision was induced by an antibiotic. The mice died 72 h after inoculation, having developed pathogenic damage typical of STEC infection. As well as renal and intestinal damage, markers of central nervous system (CNS) injury were observed, including aberrant immunolocalization of neuronal nuclei (NeuN) and increased expression of glial fibrillary acidic protein (GFAP). These results show that bacteriophage 933W without an E. coli O157:H7 background is capable of inducing the pathogenic damage associated with STEC infection. In addition, a novel mouse model was developed to evaluate therapeutic approaches focused on the bacteriophage as a new target.

17.
Vaccine ; 36(27): 3949-3959, 2018 06 22.
Artigo em Inglês | MEDLINE | ID: mdl-29807709

RESUMO

Escherichia coli O157:H7 is a zoonotic pathogen of global importance and the serotype of Shiga toxin-producing E.coli (STEC) most frequently associated with Hemolytic Uremic Syndrome (HUS) in humans. The main STEC reservoir is cattle. Vaccination of calves with the carboxy-terminal fraction of Intimin γ (IntC280) and EspB can reduce E.coli O157:H7 fecal shedding after experimental challenge. Shiga toxin (Stx) exerts local immunosuppressive effects in the bovine intestine and Stx2B fused to Brucella lumazine synthase (BLS-Stx2B) induces Stx2-neutralizing antibodies. To determine if an immune response against Stx could improve a vaccine's effect on fecal shedding, groups of calves were immunized with EspB + IntC280, with EspB + IntC280 + BLS-Stx2B, or kept as controls. At 24 days post vaccination calves were challenged with E.coli O157:H7. Shedding of E.coli O157:H7 was assessed in recto-anal mucosal swabs by direct plating and enrichment followed by immunomagnetic separation and multiplex PCR. Calves were euthanized 15 days after the challenge and intestinal segments were obtained to assess mucosal antibodies. Vaccination induced a significant increase of IntC280 and EspB specific antibodies in serum and intestinal mucosa in both vaccinated groups. Antibodies against Stx2B were detected in serum and intestinal mucosa of animals vaccinated with 3 antigens. Sera and intestinal homogenates were able to neutralize Stx2 verocytotoxicity compared to the control and the 2-antigens vaccinated group. Both vaccines reduced E.coli O157:H7 shedding compared to the control group. The addition of Stx2B to the vaccine formulation did not result in a superior level of protection compared to the one conferred by IntC280 and EspB alone. It remains to be determined if the inclusion of Stx2B in the vaccine alters E.coli O157:H7 shedding patterns in the long term and after recurrent low dose exposure as occurring in cattle herds.


Assuntos
Adesinas Bacterianas/imunologia , Proteínas da Membrana Bacteriana Externa/imunologia , Infecções por Escherichia coli/veterinária , Escherichia coli O157/imunologia , Proteínas de Escherichia coli/imunologia , Vacinas contra Escherichia coli/administração & dosagem , Síndrome Hemolítico-Urêmica/prevenção & controle , Toxina Shiga II/imunologia , Zoonoses/prevenção & controle , Adesinas Bacterianas/genética , Animais , Anticorpos Antibacterianos/sangue , Anticorpos Neutralizantes/sangue , Proteínas da Membrana Bacteriana Externa/genética , Derrame de Bactérias , Bovinos , Infecções por Escherichia coli/prevenção & controle , Proteínas de Escherichia coli/genética , Vacinas contra Escherichia coli/imunologia , Vacinas contra Escherichia coli/uso terapêutico , Fezes/microbiologia , Humanos , Imunidade Humoral/imunologia , Mucosa Intestinal/imunologia , Masculino , Toxina Shiga II/genética , Vacinação/veterinária , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/imunologia , Vacinas Sintéticas/uso terapêutico
18.
ScientificWorldJournal ; 7: 1155-64, 2007 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-17694250

RESUMO

Thrombotic microangiopathy and acute renal failure are cardinal features of post-diarrheal hemolytic uremic syndrome (HUS). These conditions are related to endothelial and epithelial cell damage induced by Shiga toxin (Stx), through an interaction with its globotriaosylceramide (Gb3) receptor. Although, Stx is the main pathogenic factor and necessary for HUS development, clinical and experimental evidence suggest that the inflammatory response is able to potentiate Stx toxicity. Lipopolysaccharides (LPS) and neutrophils (PMN) represent two central components of inflammation during a Gram-negative infection. In this regard, patients with high peripheral PMN counts at presentation have a poor prognosis. In the present review, we discuss the contribution of experimental models and patient's studies in an attempt to elucidate the pathogenic mechanisms of HUS.


Assuntos
Síndrome Hemolítico-Urêmica/fisiopatologia , Neutrófilos/fisiologia , Animais , Modelos Animais de Doenças , Síndrome Hemolítico-Urêmica/imunologia , Síndrome Hemolítico-Urêmica/metabolismo , Humanos , Lipopolissacarídeos/imunologia , Lipopolissacarídeos/metabolismo , Modelos Biológicos , Neutrófilos/imunologia , Neutrófilos/metabolismo
19.
PLoS One ; 12(1): e0169422, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28046078

RESUMO

Ruminants are the primary reservoir of Shiga-toxin producing Escherichia coli (STEC) O157:H7 and the main source of infection for humans. The aim of this study was to assess the immunogenic properties of a candidate vaccine consisting on the recombinant proteins of E. coli O157:H7 IntiminC280, the carboxy-terminal fraction of Intimin γ, EspB and the fusion protein between the B subunit of Stx2 and Brucella Lumazine Synthase (BLS)(BLS-Stx2B), in Holstein Fresian calves.To accomplish this goal we vaccinated calves with two doses of different vaccine formulations: 2 antigens (IntiminC280, EspB), 3 antigens (IntiminC280, EspB, BLS-Stx2B), BLS-Stx2B alone and a control non-vaccinated group. All antigens were expressed as recombinant proteins in E. coli. Specific IgG titres increased in vaccinated calves and the inclusion of BLS-Stx2B in the formulation seems to have a stimulatory effect on the humoral response to IntiminC280 and EspB after the booster. The neutralizing activity of antibodies against these two antigens was assessed in Red Blood Cell lysis assays and adherence to Hep-2 cells as a correlate of T3SS activity. Both sera from animals vaccinated with 2 or 3 antigens inhibited both virulence properties. Serological response to Stx2 was observed in animals vaccinated only with BLS-Stx2B and with 3 antigens and neutralization of Stx2 cytotoxicity was also observed in both groups. In conclusion, immunization of calves with BLS-Stx2B, IntiminC280 and EspB elicited a potent humoral response able to neutralize Shiga toxin 2 cytotoxity and the T3SS virulence properties in vitro. These results suggest that this formulation is a good candidate vaccine to reduce STEC shedding in cattle and needs to be further assessed in vivo.


Assuntos
Doenças dos Bovinos/imunologia , Bovinos/imunologia , Infecções por Escherichia coli/veterinária , Vacinas contra Escherichia coli/imunologia , Toxina Shiga II/imunologia , Sistemas de Secreção Tipo III , Adesinas Bacterianas/metabolismo , Animais , Anticorpos Antibacterianos/sangue , Proteínas da Membrana Bacteriana Externa/metabolismo , Derrame de Bactérias , Doenças dos Bovinos/microbiologia , Adesão Celular , Chlorocebus aethiops , Eritrócitos/microbiologia , Infecções por Escherichia coli/imunologia , Escherichia coli O157/patogenicidade , Proteínas de Escherichia coli/metabolismo , Imunidade Humoral , Imunoglobulina G/sangue , Masculino , Proteínas Recombinantes/imunologia , Células Vero , Virulência
20.
Toxins (Basel) ; 9(11)2017 10 25.
Artigo em Inglês | MEDLINE | ID: mdl-29068360

RESUMO

Shiga toxin (Stx), produced by Escherichia coli, is the main pathogenic factor of diarrhea-associated hemolytic uremic syndrome (HUS), which is characterized by the obstruction of renal microvasculature by platelet-fibrin thrombi. It is well known that the oxidative imbalance generated by Stx induces platelet activation, contributing to thrombus formation. Moreover, activated platelets release soluble CD40 ligand (sCD40L), which in turn contributes to oxidative imbalance, triggering the release of reactive oxidative species (ROS) on various cellular types. The aim of this work was to determine if the interaction between the oxidative response and platelet-derived sCD40L, as consequence of Stx-induced endothelium damage, participates in the pathogenic mechanism during HUS. Activated human glomerular endothelial cells (HGEC) by Stx2 induced platelets to adhere to them. Although platelet adhesion did not contribute to endothelial damage, high levels of sCD40L were released to the medium. The release of sCD40L by activated platelets was inhibited by antioxidant treatment. Furthermore, we found increased levels of sCD40L in plasma from HUS patients, which were also able to trigger the respiratory burst in monocytes in a sCD40L-dependent manner. Thus, we concluded that platelet-derived sCD40L and the oxidative response are reciprocally stimulated during Stx2-associated HUS. This process may contribute to the evolution of glomerular occlusion and the microangiopathic lesions.


Assuntos
Ligante de CD40/sangue , Células Endoteliais/efeitos dos fármacos , Síndrome Hemolítico-Urêmica/sangue , Toxina Shiga/toxicidade , Células Cultivadas , Criança , Pré-Escolar , Células Endoteliais/patologia , Feminino , Síndrome Hemolítico-Urêmica/induzido quimicamente , Humanos , Lactente , Rim/metabolismo , Rim/patologia , Masculino , Microvasos , Monócitos/metabolismo , Estresse Oxidativo , Ativação Plaquetária/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA