Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 73
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Infect Immun ; 87(4)2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30670557

RESUMO

Shiga toxin (Stx)-producing Escherichia coli (STEC) causes foodborne outbreaks of bloody diarrhea. There are two major types of immunologically distinct Stxs: Stx1a and Stx2a. Stx1a is more cytotoxic to Vero cells than Stx2a, but Stx2a has a lower 50% lethal dose (LD50) in mice. Epidemiological data suggest that infections by STEC strains that produce only Stx2a progress more often to a life-threatening sequela of infection called hemolytic-uremic syndrome (HUS) than isolates that make Stx1a only or produce both Stx1a and Stx2a. In this study, we found that an E. coli O26:H11 strain that produces both Stx1a and Stx2a was virulent in streptomycin- and ciprofloxacin-treated mice and that mice were protected by administration of an anti-Stx2 antibody. However, we discovered that in the absence of ciprofloxacin, neutralization of Stx1a enhanced the virulence of the strain, a result that corroborated our previous finding that Stx1a reduces the toxicity of Stx2a by the oral route. We further found that intraperitoneal administration of the purified Stx1a B subunit delayed the mean time to death of mice intoxicated with Stx2a and reduced the cytotoxic effect of Stx2a on Vero cells. Taken together, our data suggest that Stx1a reduces both the pathogenicity of Stx2 in vivo and cytotoxicity in vitro.


Assuntos
Infecções por Escherichia coli/microbiologia , Toxina Shiga I/toxicidade , Toxina Shiga II/toxicidade , Escherichia coli Shiga Toxigênica/metabolismo , Animais , Chlorocebus aethiops , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Toxina Shiga I/metabolismo , Toxina Shiga II/metabolismo , Escherichia coli Shiga Toxigênica/genética , Escherichia coli Shiga Toxigênica/patogenicidade , Células Vero , Virulência
2.
Infect Immun ; 86(6)2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29610258

RESUMO

Bacillus cereus G9241 caused a life-threatening anthrax-like lung infection in a previously healthy human. This strain harbors two large virulence plasmids, pBCXO1 and pBC210, that are absent from typical B. cereus isolates. The pBCXO1 plasmid is nearly identical to pXO1 from Bacillus anthracis and carries genes (pagA1, lef, and cya) for anthrax toxin components (protective antigen [called PA1 in G9241], lethal factor [LF], and edema factor [EF], respectively). The plasmid also has an intact hyaluronic acid capsule locus. The pBC210 plasmid has a tetrasaccharide capsule locus, a gene for a PA1 homolog called PA2 (pagA2), and a gene (cer) for Certhrax, an ADP-ribosyltransferase toxin that inactivates vinculin. LF, EF, and Certhrax require PA for entry into cells. In this study, we asked what role PA1, PA2, LF, and Certhrax play in the pathogenicity of G9241. To answer this, we generated isogenic deletion mutations in the targeted toxin gene components and then assessed the strains for virulence in highly G9241-susceptible (A/J) and moderately G9241-sensitive (C57BL/6) mice. We found that full virulence of G9241 required PA1 and LF, while PA2 contributed minimally to pathogenesis of G9241 but could not functionally replace PA1 as a toxin-binding subunit in vivo Surprisingly, we discovered that Certhrax attenuated the virulence of G9241; i.e., a Δcer Δlef mutant strain was more virulent than a Δlef mutant strain following subcutaneous inoculation of A/J mice. Moreover, the enzymatic activity of Certhrax contributed to this phenotype. We concluded that Certhrax acts as an antivirulence factor in the anthrax-like organism B. cereus G9241.


Assuntos
ADP Ribose Transferases/metabolismo , Bacillus cereus/metabolismo , Proteínas de Bactérias/metabolismo , Toxinas Bacterianas/metabolismo , Infecções por Bactérias Gram-Positivas/microbiologia , Animais , Anticorpos Antibacterianos , Bacillus cereus/patogenicidade , Toxinas Bacterianas/genética , Escherichia coli , Regulação Bacteriana da Expressão Gênica , Camundongos , Mutação , Plasmídeos/genética , Proteínas Recombinantes , Virulência
3.
Mol Microbiol ; 102(4): 545-561, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27490458

RESUMO

AtxA is a critical transcriptional regulator of plasmid-encoded virulence genes in Bacillus anthracis. Bacillus cereus G9241, which caused an anthrax-like infection, has two virulence plasmids, pBCXO1 and pBC210, that each harbor toxin genes and a capsule locus. G9241 also produces two orthologs of AtxA: AtxA1, encoded on pBCXO1, and AtxA2, encoded on pBC210. The amino acid sequence of AtxA1 is identical to that of AtxA from B. anthracis, while the sequences of AtxA1 and AtxA2 are 79% identical and 91% similar to one another. We found by qRT-PCR that AtxA1 and AtxA2 function as positive regulators of toxin (AtxA1) and capsule operon (both) transcription in G9241 and that a ΔatxA1 mutant produced lower levels of the anthrax toxins and no hyaluronic acid capsule. Deletion of atxA1 or atxA2 decreased the virulence of spores administered intranasally or subcutaneously to C57BL/6 mice but not to A/J mice, and deletion of both genes rendered spores avirulent in A/J mice. In addition, unlike AtxA1, AtxA2 did not form stable homomultimers in vitro, although AtxA1 and AtxA2 formed heterodimers. Our data show that AtxA1 is the primary regulator of G9241 virulence factor expression and that AtxA1 and AtxA2 are both required for full virulence.


Assuntos
Bacillus cereus/patogenicidade , Proteínas de Bactérias/metabolismo , Transativadores/metabolismo , Virulência/genética , Sequência de Aminoácidos , Animais , Antígenos de Bactérias/genética , Antígenos de Bactérias/metabolismo , Bacillus anthracis/genética , Bacillus anthracis/metabolismo , Bacillus cereus/genética , Bacillus cereus/metabolismo , Cápsulas Bacterianas/metabolismo , Proteínas de Bactérias/genética , Toxinas Bacterianas/genética , Toxinas Bacterianas/metabolismo , Feminino , Camundongos , Camundongos Endogâmicos A , Camundongos Endogâmicos C57BL , Óperon/genética , Plasmídeos/genética , Transativadores/genética , Fatores de Virulência/metabolismo
4.
J Infect Dis ; 213(8): 1271-9, 2016 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-26743841

RESUMO

BACKGROUND: Shiga toxin (Stx) is the primary virulence factor of Stx-producing Escherichia coli (STEC). STEC can produce Stx1a and/or Stx2a, which are antigenically distinct. However, Stx2a-producing STEC are associated with more severe disease than strains producing both Stx1a and Stx2a. METHODS AND RESULTS: To address the hypothesis that the reason for the association of Stx2a with more severe disease is because Stx2a crosses the intestinal barrier with greater efficiency that Stx1a, we covalently labeled Stx1a and Stx2a with Alexa Fluor 750 and determined the ex vivo fluorescent intensity of murine systemic organs after oral intoxication. Surprisingly, both Stxs exhibited similar dissemination patterns and accumulated in the kidneys. We next cointoxicated mice to determine whether Stx1a could impede Stx2a. Cointoxication resulted in increased survival and an extended mean time to death, compared with intoxication with Stx2a only. The survival benefit was dose dependent, with the greatest effect observed when 5 times more Stx1a than Stx2a was delivered, and was amplified when Stx1a was delivered 3 hours prior to Stx2a. Cointoxication with an Stx1a active site toxoid also reduced Stx2a toxicity. CONCLUSIONS: These studies suggest that Stx1a reduces Stx2a-mediated toxicity, a finding that may explain why STEC that produce only Stx2a are associated with more severe disease than strains producing Stx1a and Stx2a.


Assuntos
Toxina Shiga I/farmacocinética , Toxina Shiga I/toxicidade , Toxina Shiga II/antagonistas & inibidores , Toxina Shiga II/toxicidade , Injúria Renal Aguda/induzido quimicamente , Injúria Renal Aguda/metabolismo , Administração Oral , Animais , Feminino , Rim/efeitos dos fármacos , Rim/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Toxina Shiga I/administração & dosagem , Toxina Shiga II/administração & dosagem , Escherichia coli Shiga Toxigênica , Análise de Sobrevida
5.
Proc Natl Acad Sci U S A ; 110(23): E2126-33, 2013 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-23690602

RESUMO

The likelihood that a single individual infected with the Shiga toxin (Stx)-producing, food-borne pathogen Escherichia coli O157:H7 will develop a life-threatening sequela called the hemolytic uremic syndrome is unpredictable. We reasoned that conditions that enhance Stx binding and uptake within the gut after E. coli O157:H7 infection should result in greater disease severity. Because the receptor for Stx, globotriaosylceramide, is up-regulated in the presence of butyrate in vitro, we asked whether a high fiber diet (HFD) that reportedly enhances butyrate production by normal gut flora can influence the outcome of an E. coli O157 infection in mice. To address that question, groups of BALB/c mice were fed high (10%) or low (2%) fiber diets and infected with E. coli O157:H7 strain 86-24 (Stx2+). Mice fed an HFD exhibited a 10- to 100-fold increase in colonization, lost 15% more body weight, exhibited signs of morbidity, and had 25% greater mortality relative to the low fiber diet (LFD)-fed group. Additionally, sections of intestinal tissue from HFD-fed mice bound more Stx1 and expressed more globotriaosylceramide than did such sections from LFD-fed mice. Furthermore, the gut microbiota of HFD-fed mice compared with LFD-fed mice contained reduced levels of native Escherichia species, organisms that might protect the gut from colonization by incoming E. coli O157:H7. Taken together, these results suggest that susceptibility to infection and subsequent disease after ingestion of E. coli O157:H7 may depend, at least in part, on individual diet and/or the capacity of the commensal flora to produce butyrate.


Assuntos
Butiratos/metabolismo , Fibras na Dieta/farmacologia , Suscetibilidade a Doenças/microbiologia , Infecções por Escherichia coli/prevenção & controle , Escherichia coli O157/fisiologia , Trato Gastrointestinal/microbiologia , Análise de Variância , Animais , Linhagem Celular , Primers do DNA/genética , Escherichia coli O157/metabolismo , Citometria de Fluxo , Imunofluorescência , Cromatografia Gasosa-Espectrometria de Massas , Humanos , Imuno-Histoquímica , Camundongos , Toxina Shiga/metabolismo , Especificidade da Espécie
6.
Infect Immun ; 83(4): 1661-73, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25667267

RESUMO

Uropathogenic Escherichia coli (UPEC) is the leading cause of cystitis. Cytotoxic necrotizing factor 1 (CNF1) and hemolysin (Hly) are toxins made by approximately 50% of UPEC isolates. CNF1 and Hly contribute to the robust inflammatory response in the bladders of mice challenged with UPEC strain CP9. We hypothesized that antibodies against CNF1 and/or Hly would reduce cystitis caused by CP9. To test this theory, we immunized female C3H/HeOuJ mice subcutaneously with a genetically derived Hly toxoid or genetically derived CNF1 toxoid plus sublethal doses of CNF1. We collected serum and observed increasing titers of specific and neutralizing antibodies against Hly or CNF1 over time. We challenged the mice intraurethrally with CP9 and euthanized them 24 h later. We observed 10-fold lower bacterial titers in the urine of Hly-immunized mice than in that of sham-immunized mice but no difference in kidney bacterial titers. Immunized mice also exhibited significantly less cystitis than sham-immunized mice. In CNF1-vaccinated mice, we detected neither a difference in urine or kidney bacterial titers nor a reduction in the severity of cystitis versus that of sham-immunized mice. We then passively administered an anti-CNF1 monoclonal antibody intraperitoneally to female C3H/HeOuJ mice prior to intraurethral challenge with CP9. Upon challenge, we noted no difference in colonization of the urine or kidney; however, cystitis was reduced significantly in mice treated with the anti-CNF1 antibody versus that in the bladders of mice given an isotype control antibody. Taken together, our data demonstrate that antibodies against CNF1 or Hly reduce the bladder pathology caused by UPEC.


Assuntos
Toxinas Bacterianas/imunologia , Cistite/microbiologia , Infecções por Escherichia coli/imunologia , Proteínas de Escherichia coli/imunologia , Proteínas Hemolisinas/imunologia , Soros Imunes/farmacologia , Animais , Anticorpos Monoclonais/farmacologia , Cistite/imunologia , Modelos Animais de Doenças , Infecções por Escherichia coli/microbiologia , Feminino , Soros Imunes/imunologia , Imunização Passiva , Camundongos , Camundongos Endogâmicos C3H , Bexiga Urinária/microbiologia , Bexiga Urinária/patologia , Infecções Urinárias/imunologia , Infecções Urinárias/microbiologia , Urina/microbiologia , Escherichia coli Uropatogênica/imunologia , Vacinação
7.
BMC Genomics ; 16: 947, 2015 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-26573818

RESUMO

BACKGROUND: Shiga toxin (Stx)-producing E. coli (STEC) are responsible for foodborne outbreaks that can result in severe human disease. During an outbreak, differential disease outcomes are observed after infection with the same STEC strain. One question of particular interest is why some infected people resolve infection after hemorrhagic colitis whereas others progress to the hemolytic uremic syndrome (HUS). Host age and infection dose have been implicated; however, these parameters do not appear to fully account for all of the observed variation in disease severity. Therefore, we hypothesized that additional host genetic factors may play a role in progression to HUS. METHODS AND RESULTS: To mimic the genetic diversity in the human response to infection by STEC, we measured the capacity of an O157:H7 outbreak isolate to colonize mouse strains from the advanced recombinant inbred (ARI) BXD panel. We first infected the BXD parental strains C57BL/6 J (B6) and DBA/2 J (D2) with either 86-24 (Stx2a+) or TUV86-2, an Stx2a-negative isogenic mutant. Colonization levels were determined in an intact commensal flora (ICF) infection model. We found a significant difference in colonization levels between the parental B6 and D2 strains after infection with TUV86-2 but not with 86-24. This observation suggested that a host factor that may be masked by Stx2a affects O157:H7 colonization in some genetic backgrounds. We then determined the TUV86-2 colonization levels of 24 BXD strains in the ICF model. We identified several quantitative trait loci (QTL) associated with variation in colonization by correlation analyses. We found a highly significant QTL on proximal chromosome 9 (12.5-26.7 Mb) that strongly predicts variation in colonization levels and accounts for 15-20 % of variance. Linkage, polymorphism and co-citation analyses of the mapped region revealed 36 candidate genes within the QTL, and we identified five genes that are most likely responsible for the differential colonization. CONCLUSIONS: The identification of the QTL on chromosome 9 supports our hypothesis that individual genetic makeup affects the level of colonization after infection with STEC O157:H7.


Assuntos
Mapeamento Cromossômico , DNA Recombinante/genética , Escherichia coli O157/fisiologia , Interações Hospedeiro-Patógeno , Locos de Características Quantitativas/genética , Animais , Escherichia coli O157/metabolismo , Feminino , Perfilação da Expressão Gênica , Ontologia Genética , Redes Reguladoras de Genes , Ligação Genética , Variação Genética , Síndrome Hemolítico-Urêmica/genética , Síndrome Hemolítico-Urêmica/microbiologia , Humanos , Camundongos , Camundongos Endogâmicos DBA , Toxina Shiga/metabolismo , Especificidade da Espécie , Fatores de Tempo
8.
J Infect Dis ; 210(12): 1909-19, 2014 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-25038258

RESUMO

BACKGROUND: A Shiga toxin type 2a (Stx2a)-producing enteroaggregative Escherichia coli (EAEC) strain of serotype O104:H4 caused a large outbreak in 2011 in northern Europe. Pathogenic mechanisms for this strain are unclear. We hypothesized that EAEC genes encoded on the pAA virulence plasmid promoted the translocation of Stx2a across the intestinal mucosa. METHODS: We investigated the potential contribution of pAA by using mutants of Stx-EAEC strain C227-11, either cured of the pAA plasmid or deleted for individual known pAA-encoded virulence genes (ie, aggR, aggA, and sepA). The resulting mutants were tested for their ability to induce interleukin 8 (IL-8) secretion and translocation of Stx2a across a polarized colonic epithelial (T84 cell) monolayer. RESULTS: We found that deletion of aggR or aggA significantly reduced bacterial adherence and (independently) translocation of Stx2a across the T84-cell monolayer. Moreover, deletion of aggR, aggA, sepA, or the Stx2a-encoding phage from C227-11 resulted in reduced secretion of IL-8 from the infected monolayer. CONCLUSIONS: Our data suggest that the AggR-regulated aggregative adherence fimbriae I enhance inflammation and enable the outbreak strain to both adhere to epithelial cells and translocate Stx2a across the intestinal epithelium.


Assuntos
Células Epiteliais/metabolismo , Escherichia coli/metabolismo , Transporte Proteico , Toxina Shiga II/metabolismo , Aderência Bacteriana , Linhagem Celular , Escherichia coli/classificação , Escherichia coli/genética , Escherichia coli/isolamento & purificação , Infecções por Escherichia coli/microbiologia , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Deleção de Genes , Genótipo , Alemanha , Humanos , Interleucina-8/metabolismo , Plasmídeos , Sorogrupo , Transativadores/genética , Transativadores/metabolismo , Fatores de Virulência/genética , Fatores de Virulência/metabolismo
9.
Biochemistry ; 52(13): 2309-18, 2013 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-22934824

RESUMO

Bacillus cereus G9241 was isolated from a welder suffering from an anthrax-like inhalation illness. B. cereus G9241 encodes two megaplasmids, pBCXO1 and pBC210, which are analogous to the toxin- and capsule-encoding virulence plasmids of Bacillus anthracis. Protein modeling predicted that the pBC210 LF homologue contained an ADP-ribosyltransferase (ADPr) domain. This putative bacterial ADP-ribosyltransferase domain was denoted CerADPr. Iterative modeling showed that CerADPr possessed several conserved ADP-ribosyltransferase features, including an α-3 helix, an ADP-ribosyltransferase turn-turn loop, and a "Gln-XXX-Glu" motif. CerADPr ADP-ribosylated an ~120 kDa protein in HeLa cell lysates and intact cells. EGFP-CerADPr rounded HeLa cells, elicited cytoskeletal changes, and yielded a cytotoxic phenotype, indicating that CerADPr disrupts cytoskeletal signaling. CerADPr(E431D) did not possess ADP-ribosyltransferase or NAD glycohydrolase activities and did not elicit a phenotype in HeLa cells, implicating Glu431 as a catalytic residue. These experiments identify CerADPr as a cytotoxic ADP-ribosyltransferase that disrupts the host cytoskeleton.


Assuntos
ADP Ribose Transferases/metabolismo , Bacillus cereus/enzimologia , Bacillus cereus/fisiologia , Citoesqueleto/microbiologia , Interações Hospedeiro-Patógeno , ADP Ribose Transferases/química , Sequência de Aminoácidos , Antraz/microbiologia , Bacillus cereus/isolamento & purificação , Domínio Catalítico , Células HeLa , Humanos , Dados de Sequência Molecular , Estrutura Terciária de Proteína
10.
Infect Immun ; 81(1): 99-109, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23090961

RESUMO

Cytotoxic necrotizing factor 1 (CNF1) and hemolysin (HlyA1) are toxins produced by uropathogenic Escherichia coli (UPEC). We previously showed that these toxins contribute to the inflammation and tissue damage seen in a mouse model of ascending urinary tract infection. CNF1 constitutively activates small Rho GTPases by deamidation of a conserved glutamine residue, and HlyA1 forms pores in eukaryotic cell membranes. In this study, we used cDNA microarrays of bladder tissue isolated from mice infected intraurethrally with wild-type CP9, CP9cnf1, or CP9ΔhlyA to further evaluate the role that each toxin plays in the host response to UPEC. Regardless of the strain used, we found that UPEC itself elicited a significant change in host gene expression 24 h after inoculation. The largest numbers of upregulated genes were in the cytokine and chemokine signaling and Toll-like receptor signaling pathways. CNF1 exerted a strong positive influence on expression of genes involved in innate immunity and signal transduction and a negative impact on metabolism- and transport-associated genes. HlyA1 evoked an increase in expression of genes that encode innate immunity factors and a decrease in expression of genes involved in cytoskeletal and metabolic processes. Multiplex cytokine and myeloperoxidase assays corroborated our finding that a strong proinflammatory response was elicited by all strains tested. Bladders challenged intraurethrally with purified CNF1 displayed pathology similar to but significantly less intense than the pathology that we observed in CP9-challenged mice. Our data demonstrate substantial roles for CNF1 and HlyA1 in initiation of a strong proinflammatory response to UPEC in the bladder.


Assuntos
Toxinas Bacterianas/metabolismo , Proteínas de Escherichia coli/metabolismo , Proteínas Hemolisinas/metabolismo , Bexiga Urinária/metabolismo , Escherichia coli Uropatogênica/metabolismo , Animais , Toxinas Bacterianas/imunologia , Quimiocinas/imunologia , Quimiocinas/metabolismo , Edema/genética , Edema/imunologia , Edema/metabolismo , Edema/microbiologia , Infecções por Escherichia coli/genética , Infecções por Escherichia coli/imunologia , Infecções por Escherichia coli/metabolismo , Infecções por Escherichia coli/microbiologia , Proteínas de Escherichia coli/imunologia , Feminino , Expressão Gênica/imunologia , Proteínas Hemolisinas/imunologia , Imunidade Inata/genética , Imunidade Inata/imunologia , Inflamação/genética , Inflamação/imunologia , Inflamação/metabolismo , Inflamação/microbiologia , Camundongos , Camundongos Endogâmicos C3H , Neutrófilos/imunologia , Neutrófilos/metabolismo , Neutrófilos/microbiologia , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Receptores Toll-Like/imunologia , Receptores Toll-Like/metabolismo , Regulação para Cima/imunologia , Bexiga Urinária/imunologia , Bexiga Urinária/microbiologia , Escherichia coli Uropatogênica/imunologia
11.
Infect Immun ; 81(5): 1562-74, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23439303

RESUMO

In May 2011, a large food-borne outbreak was traced to an unusual O104:H4 enteroaggregative Escherichia coli (EAEC) strain that produced Shiga toxin (Stx) type 2 (Stx2). We developed a mouse model to study the pathogenesis and treatment for this strain and examined the virulence of the isolate for Dutch belted rabbits. O104:H4 strain C227-11 was gavaged into C57BL/6 mice at 10(9) to 10(11) CFU/animal. The infected animals were then given water with ampicillin (Amp; 5 g/liter) ad libitum. The C227-11-infected, Amp-treated C57BL/6 mice exhibited both morbidity and mortality. Kidneys from mice infected with C227-11 showed acute tubular necrosis, a finding seen in mice infected with typical Stx-producing E. coli. We provided anti-Stx2 antibody after infection and found that all of the antibody-treated mice gained more weight than untreated mice and, in another study, that all of the antibody-treated animals lived, whereas 3/8 phosphate-buffered saline-treated mice died. We further compared the pathogenesis of C227-11 with that of an Stx-negative (Stx(-)) O104:H4 isolate, C734-09, and an Stx2(-) phage-cured derivative of C227-11. Whereas C227-11-infected animals lost weight or gained less weight over the course of infection and died, mice infected with either of the Stx(-) isolates did not lose weight and only one mouse died. When the Stx-positive (Stx(+)) and Stx2(-) O104:H4 strains were compared in rabbits, greater morbidity and mortality were observed in rabbits infected with the Stx2(+) isolates than the Stx2(-) isolates. In conclusion, we describe two animal models for EAEC pathogenesis, and these studies show that Stx2 is responsible for most of the virulence observed in C227-11-infected mice and rabbits.


Assuntos
Surtos de Doenças , Infecções por Escherichia coli/epidemiologia , Toxina Shiga II , Escherichia coli Shiga Toxigênica/patogenicidade , Ampicilina/uso terapêutico , Análise de Variância , Animais , Antibacterianos/uso terapêutico , Modelos Animais de Doenças , Infecções por Escherichia coli/tratamento farmacológico , Infecções por Escherichia coli/microbiologia , Feminino , Alemanha/epidemiologia , Camundongos , Camundongos Endogâmicos C57BL , Coelhos , Toxina Shiga II/metabolismo , Escherichia coli Shiga Toxigênica/metabolismo , Virulência
12.
J Clin Microbiol ; 50(9): 2951-63, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22760050

RESUMO

When Shiga toxin-producing Escherichia coli (STEC) strains emerged as agents of human disease, two types of toxin were identified: Shiga toxin type 1 (Stx1) (almost identical to Shiga toxin produced by Shigella dysenteriae type 1) and the immunologically distinct type 2 (Stx2). Subsequently, numerous STEC strains have been characterized that express toxins with variations in amino acid sequence, some of which confer unique biological properties. These variants were grouped within the Stx1 or Stx2 type and often assigned names to indicate that they were not identical in sequence or phenotype to the main Stx1 or Stx2 type. A lack of specificity or consistency in toxin nomenclature has led to much confusion in the characterization of STEC strains. Because serious outcomes of infection have been attributed to certain Stx subtypes and less so with others, we sought to better define the toxin subtypes within the main Stx1 and Stx2 types. We compared the levels of relatedness of 285 valid sequence variants of Stx1 and Stx2 and identified common sequences characteristic of each of three Stx/Stx1 and seven Stx2 subtypes. A novel, simple PCR subtyping method was developed, independently tested on a battery of 48 prototypic STEC strains, and improved at six clinical and research centers to test the reproducibility, sensitivity, and specificity of the PCR. Using a consistent schema for nomenclature of the Stx toxins and stx genes by phylogenetic sequence-based relatedness of the holotoxin proteins, we developed a typing approach that should obviate the need to bioassay each newly described toxin and that predicts important biological characteristics.


Assuntos
Reação em Cadeia da Polimerase/métodos , Toxinas Shiga/classificação , Toxinas Shiga/genética , Escherichia coli Shiga Toxigênica/genética , Terminologia como Assunto , Genótipo , Humanos , Reprodutibilidade dos Testes , Sensibilidade e Especificidade
13.
Infect Immun ; 79(8): 3012-9, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21576337

RESUMO

Bacillus cereus G9241 was isolated from a welder with a pulmonary anthrax-like illness. The organism contains two megaplasmids, pBCXO1 and pBC218. These plasmids are analogous to the Bacillus anthracis Ames plasmids pXO1 and pXO2 that encode anthrax toxins and capsule, respectively. Here we evaluated the virulence of B. cereus G9241 as well as the contributions of pBCXO1 and pBC218 to virulence. B. cereus G9241 was avirulent in New Zealand rabbits after subcutaneous inoculation and attenuated 100-fold compared to the published 50% lethal dose (LD(50)) values for B. anthracis Ames after aerosol inoculation. A/J and C57BL/6J mice were comparably susceptible to B. cereus G9241 by both subcutaneous and intranasal routes of infection. However, the LD(50)s for B. cereus G9241 in both mouse strains were markedly higher than those reported for B. anthracis Ames and more like those of the toxigenic but nonencapsulated B. anthracis Sterne. Furthermore, B. cereus G9241 spores could germinate and disseminate after intranasal inoculation into A/J mice, as indicated by the presence of vegetative cells in the spleen and blood of animals 48 h after infection. Lastly, B. cereus G9241 derivatives cured of one or both megaplasmids were highly attenuated in A/J mice. We conclude that the presence of the toxin- and capsule-encoding plasmids pBCXO1 and pBC218 in B. cereus G9241 alone is insufficient to render the strain as virulent as B. anthracis Ames. However, like B. anthracis, full virulence of B. cereus G9241 for mice requires the presence of both plasmids.


Assuntos
Antraz/patologia , Antígenos de Bactérias/biossíntese , Bacillus anthracis/metabolismo , Bacillus anthracis/patogenicidade , Bacillus cereus/metabolismo , Bacillus cereus/patogenicidade , Cápsulas Bacterianas/biossíntese , Toxinas Bacterianas/biossíntese , Administração por Inalação , Aerossóis/administração & dosagem , Animais , Antraz/microbiologia , Antígenos de Bactérias/genética , Bacillus anthracis/genética , Bacillus cereus/genética , Cápsulas Bacterianas/genética , Toxinas Bacterianas/genética , Modelos Animais de Doenças , Feminino , Dose Letal Mediana , Camundongos , Camundongos Endogâmicos A , Camundongos Endogâmicos C57BL , Plasmídeos/análise , Coelhos , Doenças dos Roedores/microbiologia , Doenças dos Roedores/patologia , Virulência , Fatores de Virulência/biossíntese , Fatores de Virulência/genética
14.
J Biomed Biotechnol ; 2011: 258185, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21274267

RESUMO

Escherichia coli O157:H7 has been responsible for multiple food- and waterborne outbreaks of diarrhea and/or hemorrhagic colitis (HC) worldwide. More importantly, a portion of E. coli O157:H7-infected individuals, particularly young children, develop a life-threatening sequela of infection called hemolytic uremic syndrome (HUS). Shiga toxin (Stx), a potent cytotoxin, is the major virulence factor linked to the presentation of both HC and HUS. Currently, treatment of E. coli O157:H7 and other Stx-producing E. coli (STEC) infections is limited to supportive care. To facilitate development of therapeutic strategies and vaccines for humans against these agents, animal models that mimic one or more aspect of STEC infection and disease are needed. In this paper, we focus on the characteristics of various mouse models that have been developed and that can be used to monitor STEC colonization, disease, pathology, or combinations of these features as well as the impact of Stx alone.


Assuntos
Modelos Animais de Doenças , Infecções por Escherichia coli/microbiologia , Infecções por Escherichia coli/patologia , Escherichia coli O157 , Toxinas Shiga , Animais , Humanos , Camundongos
15.
Infect Immun ; 78(11): 4488-99, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20732996

RESUMO

Escherichia coli O157:H7 and other Shiga toxin (Stx)-producing E. coli (STEC) bacteria are not enteroinvasive but can cause hemorrhagic colitis. In some STEC-infected individuals, a life-threatening sequela of infection called the hemolytic uremic syndrome may develop that can lead to kidney failure. This syndrome is linked to the production of Stx by the infecting organism. For Stx to reach the kidney, the toxin must first penetrate the colonic epithelial barrier. However, the Stx receptor, globotriaosylceramide (Gb3), has been thought to be absent from human intestinal epithelial cells. Thus, the mechanisms by which the toxin associates with and traverses through the intestine en route to the kidneys have been puzzling aspects of STEC pathogenesis. In this study, we initially determined that both types of Stx made by STEC, Stx1 and Stx2, do in fact bind to colonic epithelia in fresh tissue sections and to a colonic epithelial cell line (HCT-8). We also discovered that globotetraosylceramide (Gb4), a lower-affinity toxin receptor derived from Gb3, is readily detectable on the surfaces of human colonic tissue sections and HCT-8 cells. Furthermore, we found that Gb3 is present on a fraction of HCT-8 cells, where it presumably functions to bind and internalize Stx1 and Stx2. In addition, we established by quantitative real-time PCR (qRT-PCR) that both fresh colonic epithelial sections and HCT-8 cells express Gb3 synthase mRNA. Taken together, our data suggest that Gb3 may be present in small quantities in human colonic epithelia, where it may compete for Stx binding with the more abundantly expressed glycosphingolipid Gb4.


Assuntos
Colo , Galactosiltransferases/metabolismo , Globosídeos/metabolismo , Toxina Shiga I/metabolismo , Toxina Shiga II/metabolismo , Escherichia coli Shiga Toxigênica/patogenicidade , Linhagem Celular , Células Cultivadas , Colo/citologia , Colo/metabolismo , Células Epiteliais/metabolismo , Escherichia coli , Infecções por Escherichia coli , Galactosiltransferases/genética , Humanos , Técnicas de Cultura de Órgãos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
16.
Microb Pathog ; 48(3-4): 131-42, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20096770

RESUMO

Escherichia coli O157:H7 is a food-borne pathogen that can cause hemorrhagic colitis and, occasionally, hemolytic uremic syndrome, a sequela of infection that can result in renal failure and death. Here we sought to model the pathogenesis of orally-administered E. coli O157:H7 in BALB/c mice with an intact intestinal flora. First, we defined the optimal dose that permitted sustained fecal shedding of E. coli O157:H7 over 7 days ( approximately 10(9) colony forming units). Next, we monitored the load of E. coli O157:H7 in intestinal sections over time and observed that the cecum was consistently the tissue with the highest E. coli O157:H7 recovery. We then followed the expression of two key E. coli O157:H7 virulence factors, the adhesin intimin and Shiga toxin type 2, and detected both proteins early in infection when bacterial burdens were highest. Additionally, we noted that during infection, animals lost weight and approximately 30% died. Moribund animals also exhibited elevated levels of blood urea nitrogen, and, on necropsy, showed evidence of renal tubular damage. We conclude that conventional mice inoculated orally with high doses of E. coli O157:H7 can be used to model both intestinal colonization and subsequent development of certain extraintestinal manifestations of E. coli O157:H7 disease.


Assuntos
Modelos Animais de Doenças , Infecções por Escherichia coli/microbiologia , Infecções por Escherichia coli/patologia , Adesinas Bacterianas/biossíntese , Animais , Peso Corporal , Proteínas de Escherichia coli/biossíntese , Fezes/microbiologia , Feminino , Trato Gastrointestinal/microbiologia , Perfilação da Expressão Gênica , Humanos , Túbulos Renais/patologia , Camundongos , Camundongos Endogâmicos BALB C , Toxina Shiga II/biossíntese , Análise de Sobrevida , Ureia/sangue , Fatores de Virulência/biossíntese
17.
Artigo em Inglês | MEDLINE | ID: mdl-32175286

RESUMO

In this study we compared nine Shiga toxin (Stx)-producing Escherichia coli O157:H7 patient isolates for Stx levels, stx-phage insertion site(s), and pathogenicity in a streptomycin (Str)-treated mouse model. The strains encoded stx2a, stx1a and stx2a, or stx2a and stx2c. All of the strains elaborated 105-106 cytotoxic doses 50% (CD50) into the supernatant after growth in vitro as measured on Vero cells, and showed variable levels of increased toxin production after growth with sub-inhibitory levels of ciprofloxacin (Cip). The stx2a+stx2c+ isolates were 90-100% lethal for Str-treated BALB/c mice, though one isolate, JH2013, had a delayed time-to-death. The stx2a+ isolate was avirulent. Both an stx2a and a recA deletion mutant of one of the stx2a+stx2c+ strains, JH2010, exhibited at least a three-log decrease in cytotoxicity in vitro and both were avirulent in the mice. Stool from Str-treated mice infected with the highly virulent isolates were 10- to 100-fold more cytotoxic than feces from mice infected with the clinical isolate, JH2012, that made only Stx2a. Taken together these findings demonstrate that the stx2a-phage from JH2010 induces to higher levels in vivo than does the phage from JH2012. The stx1a+stx2a+ clinical isolates were avirulent and neutralization of Stx1 in stool from mice infected with those strains indicated that the toxin produced in vivo was primarily Stx1a. Treatment of mice infected with Stx1a+Stx2a+ isolates with Cip resulted in an increase in Stx2a production in vivo and lethality in the mice. Our data suggest that high levels of Stx2a in stool are predictive of virulence in mice.


Assuntos
Infecções por Escherichia coli , Escherichia coli O157 , Animais , Chlorocebus aethiops , Escherichia coli O157/genética , Fezes , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Toxina Shiga II/genética , Células Vero , Virulência
18.
Infect Immun ; 77(1): 170-9, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18955470

RESUMO

Cytotoxic necrotizing factor type 1 (CNF1) and CNF2 are toxins of pathogenic Escherichia coli that share 85% identity over 1,014 amino acids. Although both of these toxins modify GTPases, CNF1 is a more potent inducer of multinucleation in HEp-2 cells, binds more efficiently to HEp-2 cells, and, despite the conservation of amino acids (C866 and H881) required for enzymatic activity of the toxins, deamidates RhoA and Cdc42 better than CNF2. Here we exploited the differences between CNF1 and CNF2 to define the epitope on CNF1 to which the CNF1-specific neutralizing monoclonal antibody (MAb) (MAb NG8) binds and to determine the mechanism by which MAb NG8 neutralizes CNF1 activity on HEp-2 cells. For these purposes, we generated a panel of 21 site-directed mutants in which amino acids in CNF1 were exchanged for the amino acids in CNF2 between amino acids 546 and 869 and vice versa. This region of CNF1 not only is recognized by MAb NG8 but also is involved in binding of this toxin to HEp-2 cells. All the mutants retained the capacity to induce multinucleation of HEp-2 cells. However, the CNF1 double mutant with D591E and F593L mutations (CNF1(D591E F593L)) and the CNF1(H661Q) single mutant displayed drastically reduced reactivity with MAb NG8. A reverse chimeric triple mutant, CNF1(E591D L593F Q661H), imparted MAb NG8 reactivity to CNF2. MAb NG8 neutralized CNF2(E591D L593F Q661H) activity in a dose-dependent manner and reduced the binding of this chimeric toxin to HEp-2 cells. Taken together, these results pinpoint three amino acids in CNF1 that are key amino acids for recognition by neutralizing MAb NG8 and further help define a region in CNF1 that is critical for full toxin binding to HEp-2 cells.


Assuntos
Anticorpos Antibacterianos/imunologia , Anticorpos Monoclonais/imunologia , Toxinas Bacterianas/antagonistas & inibidores , Toxinas Bacterianas/imunologia , Proteínas de Escherichia coli/antagonistas & inibidores , Proteínas de Escherichia coli/imunologia , Substituição de Aminoácidos , Anticorpos Antibacterianos/metabolismo , Anticorpos Monoclonais/metabolismo , Toxinas Bacterianas/genética , Toxinas Bacterianas/metabolismo , Linhagem Celular , Mapeamento de Epitopos , Escherichia coli/imunologia , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Hepatócitos/efeitos dos fármacos , Humanos , Mutagênese Sítio-Dirigida , Ligação Proteica
19.
Infect Immun ; 77(1): 274-85, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18955476

RESUMO

The Bacillus anthracis genome encodes four superoxide dismutases (SODs), enzymes capable of detoxifying oxygen radicals. That two of these SODs, SOD15 and SODA1, are present in the outermost layers of the B. anthracis spore is indicated by previous proteomic analyses of the exosporium. Given the requirement that spores must survive interactions with reactive oxygen species generated by cells such as macrophages during infection, we hypothesized that SOD15 and SODA1 protect the spore from oxidative stress and contribute to the pathogenicity of B. anthracis. To test these theories, we constructed a double-knockout (Delta sod15 Delta sodA1) mutant of B. anthracis Sterne strain 34F2 and assessed its lethality in an A/J mouse intranasal infection model. The 50% lethal dose of the Delta sod15 Delta sodA1 strain was similar to that of the wild type (34F2), but surprisingly, measurable whole-spore SOD activity was greater than that in 34F2. A quadruple-knockout strain (Delta sod15 Delta sodA1 Delta sodC Delta sodA2) was then generated, and as anticipated, spore-associated SOD activity was diminished. Moreover, the quadruple-knockout strain, compared to the wild type, was attenuated more than 40-fold upon intranasal challenge of mice. Spore resistance to exogenously generated oxidative stress and to macrophage-mediated killing correlated with virulence in A/J mice. Allelic exchange that restored sod15 and sodA1 to their wild-type state restored wild-type characteristics. We conclude that SOD molecules within the spore afford B. anthracis protection against oxidative stress and enhance the pathogenicity of B. anthracis in the lung. We also surmise that the presence of four SOD alleles within the genome provides functional redundancy for this key enzyme.


Assuntos
Bacillus anthracis/enzimologia , Bacillus anthracis/patogenicidade , Proteínas de Bactérias/metabolismo , Estresse Oxidativo , Superóxido Dismutase/metabolismo , Fatores de Virulência/metabolismo , Animais , Antraz/microbiologia , Bacillus anthracis/efeitos dos fármacos , Bacillus anthracis/genética , Proteínas de Bactérias/genética , Linhagem Celular , Deleção de Genes , Dose Letal Mediana , Macrófagos/microbiologia , Camundongos , Camundongos Endogâmicos A , Oxidantes/toxicidade , Recombinação Genética , Superóxido Dismutase/genética , Análise de Sobrevida , Virulência , Fatores de Virulência/genética
20.
Infect Immun ; 77(7): 2730-40, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19433543

RESUMO

Monoclonal antibody (MAb) 11E10 recognizes the Shiga toxin type 2 (Stx2) A(1) subunit. The binding of 11E10 to Stx2 neutralizes both the cytotoxic and lethal activities of Stx2, but the MAb does not bind to or neutralize Stx1 despite the 61% identity and 75% similarity in the amino acids of the A(1) fragments. In this study, we sought to identify the segment or segments on Stx2 that constitute the 11E10 epitope and to determine how recognition of that region by 11E10 leads to inactivation of the toxin. Toward those objectives, we generated a set of chimeric Stx1/Stx2 molecules and then evaluated the capacity of 11E10 to recognize those hybrid toxins by Western blot analyses and to neutralize them in Vero cell cytotoxicity assays. We also compared the amino acid sequences and crystal structures of Stx1 and Stx2 for stretches of dissimilarity that might predict a binding epitope on Stx2 for 11E10. Through these assessments, we concluded that the 11E10 epitope is comprised of three noncontiguous regions surrounding the Stx2 active site. To determine how 11E10 neutralizes Stx2, we examined the capacity of 11E10/Stx2 complexes to target ribosomes. We found that the binding of 11E10 to Stx2 prevented the toxin from inhibiting protein synthesis in an in vitro assay but also altered the overall cellular distribution of Stx2 in Vero cells. We propose that the binding of MAb 11E10 to Stx2 neutralizes the effects of the toxin by preventing the toxin from reaching and/or inactivating the ribosomes.


Assuntos
Anticorpos Antibacterianos/imunologia , Anticorpos Monoclonais/imunologia , Mapeamento de Epitopos , Toxina Shiga II/antagonistas & inibidores , Toxina Shiga II/imunologia , Sequência de Aminoácidos , Animais , Chlorocebus aethiops , Masculino , Modelos Moleculares , Testes de Neutralização , Ligação Proteica , Estrutura Terciária de Proteína , Toxina Shiga II/metabolismo , Células Vero
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA