Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
BMC Infect Dis ; 20(1): 908, 2020 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-33256619

RESUMO

BACKGROUND: Diarrhoea, affecting children in developing countries, is mainly caused by diarrheagenic Escherichia coli (DEC). This study principally aimed to determine the prevalence of DEC pathotypes and Extended-spectrum ß-lactamase (ESBL) genes isolated from children under 5 years old with diarrhea. METHODS: A total of 320 diarrhoea stool samples were investigated. E. coli isolates were investigated for genes specific for enterotoxigenic E. coli (ETEC), enteropathogenic E. coli (EPEC), enteroaggregative E. coli (EAEC), enteroinvasive E. coli (EIEC) and enterohemorrhagic E. coli (EHEC) using polymerase chain reaction (PCR). Furthermore, antimicrobial susceptibility testing, detection of antibiotic resistance-genes and phylogenetic typing were performed. RESULTS: Over all, DEC were isolated from 66/320 (20.6%) of the children with diarrhoea. EAEC was the predominant (47%), followed by typical EPEC (28.8%) and atypical EPEC (16.6%). Co-infection by EPEC and EAEC was detected in (7.6%) of isolates. However, ETEC, EIEC and EHEC were not detected. Phylogroup A (47%) and B2 (43.9%) were the predominant types. Multidrug-resistance (MDR) was found in 55% of DEC isolates. Extended-spectrum ß-lactamase (ESBL) genes were detected in 24 isolates (24 blaTEM and 15 blaCTX-M-15). Only one isolate harbored AmpC ß-lactamase gene (DHA gene). CONCLUSION: The study concluded that, EAEC and EPEC are important causative agents of diarrhoea in children under 5 years. MDR among DEC has the potential to be a big concern.


Assuntos
Infecções Comunitárias Adquiridas/diagnóstico , Diarreia/diagnóstico , Farmacorresistência Bacteriana Múltipla/genética , Escherichia coli Enteropatogênica/genética , Infecções por Escherichia coli/diagnóstico , Escherichia coli/genética , Filogenia , Antibacterianos/uso terapêutico , Proteínas de Bactérias/genética , Pré-Escolar , Estudos de Coortes , Coinfecção/diagnóstico , Coinfecção/microbiologia , Infecções Comunitárias Adquiridas/tratamento farmacológico , Infecções Comunitárias Adquiridas/epidemiologia , Infecções Comunitárias Adquiridas/microbiologia , Diarreia/tratamento farmacológico , Diarreia/epidemiologia , Diarreia/microbiologia , Egito/epidemiologia , Escherichia coli Enteropatogênica/enzimologia , Escherichia coli Enteropatogênica/isolamento & purificação , Escherichia coli/enzimologia , Escherichia coli/isolamento & purificação , Infecções por Escherichia coli/tratamento farmacológico , Infecções por Escherichia coli/epidemiologia , Infecções por Escherichia coli/microbiologia , Fezes/microbiologia , Feminino , Humanos , Lactente , Masculino , Testes de Sensibilidade Microbiana , Reação em Cadeia da Polimerase , Prevalência , beta-Lactamases/genética
2.
Int J Med Microbiol ; 309(3-4): 159-168, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30940425

RESUMO

Bacterial secreted proteases are the key factors that increase the virulence potential of different pathogens. Extraintestinal pathogenic E. coli (ExPEC) is a distinct pathotype that has unique ability to infect various body sites apart from the gastrointestinal tract causing several life-threatening diseases both in human and animals. Thus, understanding of ExPEC pathogenesis is crucial in effective management of disease caused by these pathogens. It is known that ExPEC possesses a broad spectrum of virulence factors including the secreted proteases which elude the host defence system. Recent studies have shown that high prevalence as well as the action of the secreted proteases influence the pathogenesis of ExPEC. However, literature on the secreted proteases present in ExPEC and their role in promoting virulence of ExPEC is rather limited. This review describes the distribution, characterization and the role of serine and metalloproteases secreted by diverse pathotypes of ExPEC, highlighting the significance of secreted proteases of ExPEC in pathogenesis.


Assuntos
Proteínas de Escherichia coli/metabolismo , Escherichia coli Extraintestinal Patogênica/enzimologia , Escherichia coli Extraintestinal Patogênica/patogenicidade , Peptídeo Hidrolases/metabolismo , Fatores de Virulência/metabolismo , Animais , Escherichia coli Enteropatogênica/enzimologia , Escherichia coli Enteropatogênica/patogenicidade , Infecções por Escherichia coli/metabolismo , Infecções por Escherichia coli/microbiologia , Humanos , Metaloproteases/metabolismo , Peptídeo Hidrolases/química , Peptídeo Hidrolases/classificação , Serina Proteases/metabolismo , Fatores de Virulência/classificação
3.
J Biol Chem ; 292(42): 17337-17350, 2017 10 20.
Artigo em Inglês | MEDLINE | ID: mdl-28860194

RESUMO

The inhibition of host innate immunity pathways is essential for the persistence of attaching and effacing pathogens such as enteropathogenic Escherichia coli (EPEC) and Citrobacter rodentium during mammalian infections. To subvert these pathways and suppress the antimicrobial response, attaching and effacing pathogens use type III secretion systems to introduce effectors targeting key signaling pathways in host cells. One such effector is the arginine glycosyltransferase NleB1 (NleBCR in C. rodentium) that modifies conserved arginine residues in death domain-containing host proteins with N-acetylglucosamine (GlcNAc), thereby blocking extrinsic apoptosis signaling. Ectopically expressed NleB1 modifies the host proteins Fas-associated via death domain (FADD), TNFRSF1A-associated via death domain (TRADD), and receptor-interacting serine/threonine protein kinase 1 (RIPK1). However, the full repertoire of arginine GlcNAcylation induced by pathogen-delivered NleB1 is unknown. Using an affinity proteomic approach for measuring arginine-GlcNAcylated glycopeptides, we assessed the global profile of arginine GlcNAcylation during ectopic expression of NleB1, EPEC infection in vitro, or C. rodentium infection in vivo NleB overexpression resulted in arginine GlcNAcylation of multiple host proteins. However, NleB delivery during EPEC and C. rodentium infection caused rapid and preferential modification of Arg117 in FADD. This FADD modification was extremely stable and insensitive to physiological temperatures, glycosidases, or host cell degradation. Despite its stability and effect on the inhibition of apoptosis, arginine GlcNAcylation did not elicit any proteomic changes, even in response to prolonged NleB1 expression. We conclude that, at normal levels of expression during bacterial infection, NleB1/NleBCR antagonizes death receptor-induced apoptosis of infected cells by modifying FADD in an irreversible manner.


Assuntos
Apoptose , Citrobacter rodentium/enzimologia , Escherichia coli Enteropatogênica/enzimologia , Infecções por Escherichia coli/metabolismo , Proteínas de Escherichia coli/metabolismo , Proteína de Domínio de Morte Associada a Fas/metabolismo , Glicosiltransferases/metabolismo , Processamento de Proteína Pós-Traducional , Fatores de Virulência/metabolismo , Citrobacter rodentium/patogenicidade , Escherichia coli Enteropatogênica/patogenicidade , Infecções por Escherichia coli/genética , Infecções por Escherichia coli/patologia , Proteínas de Escherichia coli/genética , Proteína de Domínio de Morte Associada a Fas/genética , Glicosiltransferases/genética , Células HeLa , Humanos , Estabilidade Proteica , Proteína Serina-Treonina Quinases de Interação com Receptores/genética , Proteína Serina-Treonina Quinases de Interação com Receptores/metabolismo , Proteína de Domínio de Morte Associada a Receptor de TNF/genética , Proteína de Domínio de Morte Associada a Receptor de TNF/metabolismo , Fatores de Virulência/genética
4.
J Biol Chem ; 292(27): 11423-11430, 2017 07 07.
Artigo em Inglês | MEDLINE | ID: mdl-28522607

RESUMO

Many Gram-negative bacterial pathogens use a syringe-like apparatus called a type III secretion system to inject virulence factors into host cells. Some of these effectors are enzymes that modify host proteins to subvert their normal functions. NleB is a glycosyltransferase that modifies host proteins with N-acetyl-d-glucosamine to inhibit antibacterial and inflammatory host responses. NleB is conserved among the attaching/effacing pathogens enterohemorrhagic Escherichia coli (EHEC), enteropathogenic E. coli (EPEC), and Citrobacter rodentium Moreover, Salmonella enterica strains encode up to three NleB orthologs named SseK1, SseK2, and SseK3. However, there are conflicting reports regarding the activities and host protein targets among the NleB/SseK orthologs. Therefore, here we performed in vitro glycosylation assays and cell culture experiments to compare the activities and substrate specificities of these effectors. SseK1, SseK3, EHEC NleB1, EPEC NleB1, and Crodentium NleB blocked TNF-mediated NF-κB pathway activation, whereas SseK2 and NleB2 did not. C. rodentium NleB, EHEC NleB1, and SseK1 glycosylated host GAPDH. C. rodentium NleB, EHEC NleB1, EPEC NleB1, and SseK2 glycosylated the FADD (Fas-associated death domain protein). SseK3 and NleB2 were not active against either substrate. We also found that EHEC NleB1 glycosylated two GAPDH arginine residues, Arg197 and Arg200, and that these two residues were essential for GAPDH-mediated activation of TNF receptor-associated factor 2 ubiquitination. These results provide evidence that members of this highly conserved family of bacterial virulence effectors target different host protein substrates and exhibit distinct cellular modes of action to suppress host responses.


Assuntos
Proteínas de Bactérias/metabolismo , Citrobacter rodentium/enzimologia , Escherichia coli Êntero-Hemorrágica/enzimologia , Escherichia coli Enteropatogênica/enzimologia , Proteínas de Escherichia coli/metabolismo , Salmonella enterica/enzimologia , Fatores de Virulência/metabolismo , Animais , Proteínas de Bactérias/genética , Citrobacter rodentium/genética , Citrobacter rodentium/patogenicidade , Escherichia coli Êntero-Hemorrágica/genética , Escherichia coli Êntero-Hemorrágica/patogenicidade , Escherichia coli Enteropatogênica/genética , Escherichia coli Enteropatogênica/patogenicidade , Proteínas de Escherichia coli/genética , Proteína de Domínio de Morte Associada a Fas/genética , Proteína de Domínio de Morte Associada a Fas/metabolismo , Gliceraldeído-3-Fosfato Desidrogenases/genética , Gliceraldeído-3-Fosfato Desidrogenases/metabolismo , Glicosilação , Camundongos , Células RAW 264.7 , Receptores Tipo II do Fator de Necrose Tumoral/genética , Receptores Tipo II do Fator de Necrose Tumoral/metabolismo , Salmonella enterica/genética , Salmonella enterica/patogenicidade , Fator 2 Associado a Receptor de TNF/genética , Fator 2 Associado a Receptor de TNF/metabolismo , Ubiquitinação , Fatores de Virulência/genética
5.
Nat Microbiol ; 2: 16258, 2017 01 13.
Artigo em Inglês | MEDLINE | ID: mdl-28085133

RESUMO

Cell death signalling pathways contribute to tissue homeostasis and provide innate protection from infection. Adaptor proteins such as receptor-interacting serine/threonine-protein kinase 1 (RIPK1), receptor-interacting serine/threonine-protein kinase 3 (RIPK3), TIR-domain-containing adapter-inducing interferon-ß (TRIF) and Z-DNA-binding protein 1 (ZBP1)/DNA-dependent activator of IFN-regulatory factors (DAI) that contain receptor-interacting protein (RIP) homotypic interaction motifs (RHIM) play a key role in cell death and inflammatory signalling1-3. RHIM-dependent interactions help drive a caspase-independent form of cell death termed necroptosis4,5. Here, we report that the bacterial pathogen enteropathogenic Escherichia coli (EPEC) uses the type III secretion system (T3SS) effector EspL to degrade the RHIM-containing proteins RIPK1, RIPK3, TRIF and ZBP1/DAI during infection. This requires a previously unrecognized tripartite cysteine protease motif in EspL (Cys47, His131, Asp153) that cleaves within the RHIM of these proteins. Bacterial infection and/or ectopic expression of EspL leads to rapid inactivation of RIPK1, RIPK3, TRIF and ZBP1/DAI and inhibition of tumour necrosis factor (TNF), lipopolysaccharide or polyinosinic:polycytidylic acid (poly(I:C))-induced necroptosis and inflammatory signalling. Furthermore, EPEC infection inhibits TNF-induced phosphorylation and plasma membrane localization of mixed lineage kinase domain-like pseudokinase (MLKL). In vivo, EspL cysteine protease activity contributes to persistent colonization of mice by the EPEC-like mouse pathogen Citrobacter rodentium. The activity of EspL defines a family of T3SS cysteine protease effectors found in a range of bacteria and reveals a mechanism by which gastrointestinal pathogens directly target RHIM-dependent inflammatory and necroptotic signalling pathways.


Assuntos
Apoptose , Proteínas de Escherichia coli/metabolismo , Inflamação , Necrose , Proteína Serina-Treonina Quinases de Interação com Receptores/metabolismo , Animais , Morte Celular , Citrobacter rodentium/patogenicidade , Cisteína Proteases/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Escherichia coli Enteropatogênica/enzimologia , Escherichia coli Enteropatogênica/metabolismo , Proteínas de Escherichia coli/genética , Células HEK293 , Humanos , Lipopolissacarídeos/farmacologia , Camundongos , Fosforilação , Proteína Serina-Treonina Quinases de Interação com Receptores/genética , Transdução de Sinais/efeitos dos fármacos , Fator de Necrose Tumoral alfa/metabolismo , Sistemas de Secreção Tipo III
6.
Acta Biochim Pol ; 64(1): 49-63, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-27864920

RESUMO

Enteropathogenic E. coli (EPEC) is a human pathogen using type III secretion system for delivery of proteins directly into the human host. The system contains a single ATPase, EscN, which is essential for uncoupling of proteins from their complexes with chaperones before the delivery. The structure of EscN ATPase (PDB code: 2obm) was used to screen computationally for small molecule inhibitors blocking its active site. Two lead candidates were examined but only one, Compound 54, was selected for further optimization. After extended QSAR optimization, two derivatives were found to be competitive inhibitors of EscN capable of blocking ATPase activity with a Ki below 50 µM. One candidate, WEN05-03, with a Ki=16±2 µM, was also minimally toxic to mammalian cells as determined by other assays. In the cell infection model of HeLa cells with EPEC, Compound WEN05-03 completely blocked actin cluster formation at 100 µM concentration, when analyzed by confocal microscopy. The second best inhibitor of EscN ATPase activity was WEN04-34 with a Ki=46±2 µM. However, the compound was highly toxic to the BALB/3T3 cell line. In summary, the work identifies a compound blocking bacterial ATPase in its active site without causing cellular toxicity to the host cells. It is the first report showing feasibility of using bacterial virulence system ATPase as a target for safe, non-toxic compounds and offering a proof-of-concept for non-antibiotic alternatives.


Assuntos
Adenosina Trifosfatases/antagonistas & inibidores , Antibacterianos/química , Escherichia coli Enteropatogênica/enzimologia , Proteínas de Escherichia coli/antagonistas & inibidores , Sistemas de Secreção Tipo III/química , Animais , Antibacterianos/farmacologia , Células 3T3 BALB , Domínio Catalítico , Desenho de Fármacos , Inibidores Enzimáticos/farmacologia , Células HeLa , Humanos , Camundongos , Relação Quantitativa Estrutura-Atividade
7.
Cell Commun Signal ; 14(1): 30, 2016 12 08.
Artigo em Inglês | MEDLINE | ID: mdl-27931258

RESUMO

BACKGROUND: The serine proteases HtrA/DegP secreted by the human gastrointestinal pathogens Helicobacter pylori (H. pylori) and Campylobacter jejuni (C. jejuni) cleave the mammalian cell adhesion protein E-cadherin to open intercellular adhesions. A wide range of bacteria also expresses the HtrA/DegP homologs DegQ and/or DegS, which significantly differ in structure and function. METHODS: E-cadherin shedding was investigated in infection experiments with the Gram-negative pathogens H. pylori, enteropathogenic Escherichia coli (EPEC), Salmonella enterica subsp. Enterica (S. Typhimurium), Yersinia enterocolitica (Y. enterocolitica), and Proteus mirabilis (P. mirabilis), which express different combinations of HtrAs. Annotated wild-type htrA/degP, degQ and degS genes were cloned and proteolytically inactive mutants were generated by a serine-to-alanine exchange in the active center. All HtrA variants were overexpressed and purified to compare their proteolytic activities in casein zymography and in vitro E-cadherin cleavage experiments. RESULTS: Infection of epithelial cells resulted in a strong E-cadherin ectodomain shedding as reflected by the loss of full length E-cadherin in whole cell lysates and formation of the soluble 90 kDa extracellular domain of E-cadherin (NTF) in the supernatants of infected cells. Importantly, comparing the caseinolytic and E-cadherin cleavage activities of HtrA/DegP, DegQ and DegS proteins revealed that DegP and DegQ homologs from H. pylori, S. Typhimurium, Y. enterocolitica, EPEC and P. mirabilis, but not activated DegS, cleaved E-cadherin as a substrate in vitro. CONCLUSIONS: These data indicate that E-cadherin cleavage is confined to HtrA/DegP and DegQ proteins representing an important prevalent step in bacterial pathogenesis.


Assuntos
Caderinas/metabolismo , Proteínas de Escherichia coli/metabolismo , Bactérias Gram-Negativas/enzimologia , Bactérias Gram-Negativas/fisiologia , Infecções por Bactérias Gram-Negativas/metabolismo , Proteínas de Choque Térmico/metabolismo , Proteínas Periplásmicas/metabolismo , Serina Endopeptidases/metabolismo , Sequência de Aminoácidos , Animais , Linhagem Celular , Escherichia coli Enteropatogênica/enzimologia , Escherichia coli Enteropatogênica/fisiologia , Proteínas de Escherichia coli/química , Bactérias Gram-Negativas/química , Infecções por Bactérias Gram-Negativas/patologia , Proteínas de Choque Térmico/química , Humanos , Proteínas Periplásmicas/química , Proteólise , Alinhamento de Sequência , Serina Endopeptidases/química
8.
Gut Microbes ; 7(2): 115-25, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26963626

RESUMO

Autotransporter proteins (AT) are associated with bacterial virulence attributes. Originally identified in enteroaggregative Escherichia coli (EAEC), Shigella flexneri 2a and uropathogenic E. coli, the serine protease Pic is one of these AT. We have previously detected one atypical enteropathogenic E. coli strain (BA589) carrying the pic gene. In the present study, we characterized the biological activities of Pic produced by BA589 both in vitro and in vivo. Contrarily to other Pic-producers bacteria, pic in BA589 is located on a high molecular weight plasmid. PicBA589 was able to agglutinate rabbit erythrocytes, cleave mucin and degrade complement system molecules. BA589 was able to colonize mice intestines, and an intense mucus production was observed. The BA589Δpic mutant lost the capacity to colonize as well as the above-mentioned in vitro activities. Thus, Pic represents an additional virulence factor in aEPEC strain BA589, associated with adherence, colonization and evasion from the innate immune system.


Assuntos
Escherichia coli Enteropatogênica/enzimologia , Infecções por Escherichia coli/microbiologia , Proteínas de Escherichia coli/metabolismo , Serina Endopeptidases/metabolismo , Fatores de Virulência/metabolismo , Animais , Aderência Bacteriana , Escherichia coli Enteropatogênica/genética , Escherichia coli Enteropatogênica/fisiologia , Infecções por Escherichia coli/metabolismo , Proteínas de Escherichia coli/genética , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Mucinas/metabolismo , Coelhos , Serina Endopeptidases/genética , Fatores de Virulência/genética
9.
Infect Immun ; 84(5): 1346-1360, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26883593

RESUMO

Enteropathogenic Escherichia coli (EPEC) interferes with host cell signaling by injecting virulence effector proteins into enterocytes via a type III secretion system (T3SS). NleB1 is a novel T3SS glycosyltransferase effector from EPEC that transfers a single N-acetylglucosamine (GlcNAc) moiety in an N-glycosidic linkage to Arg(117) of the Fas-associated death domain protein (FADD). GlcNAcylation of FADD prevents the assembly of the canonical death-inducing signaling complex and inhibits Fas ligand (FasL)-induced cell death. Apart from the DXD catalytic motif of NleB1, little is known about other functional sites in the enzyme. In the present study, members of a library of 22 random transposon-based, in-frame, linker insertion mutants of NleB1 were tested for their ability to block caspase-8 activation in response to FasL during EPEC infection. Immunoblot analysis of caspase-8 cleavage showed that 17 mutant derivatives of NleB1, including the catalytic DXD mutant, did not inhibit caspase-8 activation. Regions of interest around the insertion sites with multiple or single amino acid substitutions were examined further. Coimmunoprecipitation studies of 34 site-directed mutants showed that the NleB1 derivatives with the E253A, Y219A, and PILN(63-66)AAAA (in which the PILN motif from residues 63 to 66 was changed to AAAA) mutations bound to but did not GlcNAcylate FADD. A further mutant derivative, the PDG(236-238)AAA mutant, did not bind to or GlcNAcylate FADD. Infection of mice with the EPEC-like mouse pathogen Citrobacter rodentium expressing NleBE253A and NleBY219A showed that these strains were attenuated, indicating the importance of residues E253 and Y219 in NleB1 virulence in vivo In summary, we identified new amino acid residues critical for NleB1 activity and confirmed that these are required for the virulence function of NleB1.


Assuntos
Análise Mutacional de DNA , Escherichia coli Enteropatogênica/enzimologia , Escherichia coli Enteropatogênica/patogenicidade , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Glicosiltransferases/genética , Glicosiltransferases/metabolismo , Fatores de Virulência/genética , Fatores de Virulência/metabolismo , Animais , Apoptose , Arginina/metabolismo , Citrobacter rodentium/genética , Citrobacter rodentium/patogenicidade , Elementos de DNA Transponíveis , Infecções por Enterobacteriaceae/microbiologia , Infecções por Enterobacteriaceae/patologia , Proteína Ligante Fas/metabolismo , Proteína de Domínio de Morte Associada a Fas/metabolismo , Feminino , Humanos , Camundongos Endogâmicos C57BL , Mutagênese Insercional , Mutagênese Sítio-Dirigida , Processamento de Proteína Pós-Traducional , Virulência
10.
J Biol Chem ; 290(34): 20856-20864, 2015 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-26163516

RESUMO

The translational GTPase BipA regulates the expression of virulence and pathogenicity factors in several eubacteria. BipA-dependent expression of virulence factors occurs under starvation conditions, such as encountered during infection of a host. Under these conditions, BipA associates with the small ribosomal subunit. BipA also has a second function to promote the efficiency of late steps in biogenesis of large ribosomal subunits at low temperatures, presumably while bound to the ribosome. During starvation, the cellular concentration of stress alarmone guanosine-3', 5'-bis pyrophosphate (ppGpp) is increased. This increase allows ppGpp to bind to BipA and switch its binding specificity from ribosomes to small ribosomal subunits. A conformational change of BipA upon ppGpp binding could explain the ppGpp regulation of the binding specificity of BipA. Here, we present the structures of the full-length BipA from Escherichia coli in apo, GDP-, and ppGpp-bound forms. The crystal structure and small-angle x-ray scattering data of the protein with bound nucleotides, together with a thermodynamic analysis of the binding of GDP and of ppGpp to BipA, indicate that the ppGpp-bound form of BipA adopts the structure of the GDP form. This suggests furthermore, that the switch in binding preference only occurs when both ppGpp and the small ribosomal subunit are present. This molecular mechanism would allow BipA to interact with both the ribosome and the small ribosomal subunit during stress response.


Assuntos
Apoproteínas/química , Escherichia coli Enteropatogênica/genética , Escherichia coli Enteropatogênica/patogenicidade , Proteínas de Escherichia coli/química , GTP Fosfo-Hidrolases/química , Guanosina Difosfato/química , Fosfoproteínas/química , Pirofosfatases/química , Apoproteínas/genética , Apoproteínas/metabolismo , Cristalografia por Raios X , Escherichia coli Enteropatogênica/enzimologia , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , GTP Fosfo-Hidrolases/genética , GTP Fosfo-Hidrolases/metabolismo , Expressão Gênica , Guanosina Difosfato/metabolismo , Cinética , Modelos Moleculares , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína , Pirofosfatases/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Subunidades Ribossômicas Menores/genética , Subunidades Ribossômicas Menores/metabolismo , Transdução de Sinais , Estresse Fisiológico , Termodinâmica , Virulência
11.
Cell Microbiol ; 17(12): 1766-78, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26096513

RESUMO

Upon infection of epithelial cells, enteropathogenic Escherichia coli suppresses host cell inflammatory signalling in a type III secretion system (T3SS) dependent manner. Two key T3SS effector proteins involved in this response are NleE and NleC. NleC is a zinc metalloprotease effector that degrades the p65 subunit of NF-κB. Although the site of p65 cleavage by NleC is now well described, other areas of interaction have not been precisely defined. Here we constructed overlapping truncations of p65 to identify regions required for NleC cleavage. We determined that NleC cleaved both p65 and p50 within the Rel homology domain (RHD) and that two motifs, E22IIE25 and P177VLS180 , within the RHD of p65 were important for recognition and binding by NleC. Alanine substitution of one or both of these motifs protected p65 from binding and degradation by NleC. The E22IIE25 and P177VLS180 motifs were located within the structurally distinct N-terminal subdomain of the RHD involved in DNA binding by p65 on adjacent, parallel strands. Although these motifs have not been recognized previously, both were needed for the correct localization and function of p65. In summary, this work has identified two regions of p65 within the RHD needed for binding and cleavage by NleC and provides further insight into the molecular basis of substrate recognition by a T3SS effector.


Assuntos
Escherichia coli Enteropatogênica/enzimologia , Proteínas de Escherichia coli/metabolismo , Metaloproteases/metabolismo , Fator de Transcrição RelA/metabolismo , Motivos de Aminoácidos , Análise Mutacional de DNA , Ligação Proteica , Estrutura Terciária de Proteína , Proteólise , Fator de Transcrição RelA/genética
12.
J Biol Chem ; 290(16): 10406-17, 2015 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-25678709

RESUMO

The Gram-negative bacterium enteropathogenic Escherichia coli uses a syringe-like type III secretion system (T3SS) to inject virulence or "effector" proteins into the cytoplasm of host intestinal epithelial cells. To assemble, the T3SS must traverse both bacterial membranes, as well as the peptidoglycan layer. Peptidoglycan is made of repeating N-acetylmuramic acid and N-acetylglucosamine disaccharides cross-linked by pentapeptides to form a tight mesh barrier. Assembly of many macromolecular machines requires a dedicated peptidoglycan lytic enzyme (PG-lytic enzyme) to locally clear peptidoglycan. Here we have solved the first structure of a T3SS-associated PG-lytic enzyme, EtgA from enteropathogenic E. coli. Unexpectedly, the active site of EtgA has features in common with both lytic transglycosylases and hen egg white lysozyme. Most notably, the ß-hairpin region resembles that of lysozyme and contains an aspartate that aligns with lysozyme Asp-52 (a residue critical for catalysis), a conservation not observed in other previously characterized lytic transglycosylase families to which the conserved T3SS enzymes had been presumed to belong. Mutation of the EtgA catalytic glutamate, Glu-42, conserved across lytic transglycosylases and hen egg white lysozyme, and this differentiating aspartate diminishes type III secretion in vivo, supporting its essential role in clearing the peptidoglycan for T3SS assembly. Finally, we show that EtgA forms a 1:1 complex with the building block of the polymerized T3SS inner rod component, EscI, and that this interaction enhances PG-lytic activity of EtgA in vitro, collectively providing the necessary strict localization and regulation of the lytic activity to prevent overall cell lysis.


Assuntos
Escherichia coli Enteropatogênica/genética , Proteínas de Escherichia coli/química , Regulação Bacteriana da Expressão Gênica , Glicosiltransferases/química , Peptidoglicano/química , Sequência de Aminoácidos , Sistemas de Secreção Bacterianos , Transporte Biológico , Membrana Celular/metabolismo , Parede Celular/metabolismo , Escherichia coli Enteropatogênica/enzimologia , Escherichia coli Enteropatogênica/patogenicidade , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Ácido Glutâmico/química , Ácido Glutâmico/metabolismo , Glicosiltransferases/genética , Glicosiltransferases/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Muramidase/química , Muramidase/genética , Muramidase/metabolismo , Mutação , Peptidoglicano/metabolismo , Estrutura Secundária de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Homologia de Sequência de Aminoácidos , Virulência
13.
Jpn J Infect Dis ; 68(3): 239-43, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25672408

RESUMO

The main objective of this study was to investigate the epidemiology, drug resistance and ß-lactamase genotype distribution of enteropathogenic Escherichia coli (EPEC) isolated from pediatric patients with diarrhea in southern China. The prevalence of EPEC in children with diarrhea was 3.53%. The commonest serotypes were O55:K59 and O126:K71, and the typical EPEC were more prevalent than atypical EPEC (51 vs 7). Isolates from this region were most commonly found to be resistant to ampicillin and cotrimoxazole, followed by chloramphenicol, ceftriaxone, and ceftazidime. More than 96% of the strains were susceptible to cefoperazone/sulbactam and imipenem. The most common ß-lactamase genotypes identified in 58 strains were blaCTX-M-1 (60.3%), blaTEM (56.9%), blaCTX-M-9 (27.6%), and blaSHV (15.5%). Among 58 isolates, 22 strains were found to harbor one ß-lactamase gene, and the proportions of resistance to ampicillin, cotrimoxazole, chloramphenicol, ceftriaxone, and ceftazidime, were 81.8%, 63.6%, 40.9%, 18.2%, and 9.1%, respectively. A further 30 strains carrying multiple ß-lactamase genes had increased resistance to the above antimicrobial agents (100%, 83.3%, 70.0%, 60.0%, and 30.0%, respectively). In contrast, antibiotic resistance in the last 6 strains without a detectable ß-lactamase gene was substantially reduced. Drug resistance may be associated with the ß-lactamase gene number, with a greater the number of ß-lactamase genes resulting in higher antibiotic resistance.


Assuntos
Antibacterianos/farmacologia , Diarreia/microbiologia , Escherichia coli Enteropatogênica/efeitos dos fármacos , Escherichia coli Enteropatogênica/genética , Infecções por Escherichia coli/microbiologia , beta-Lactamases/genética , Pré-Escolar , China/epidemiologia , Diarreia/epidemiologia , Farmacorresistência Bacteriana , Escherichia coli Enteropatogênica/enzimologia , Infecções por Escherichia coli/epidemiologia , Feminino , Humanos , Lactente , Recém-Nascido , Masculino , Testes de Sensibilidade Microbiana , Prevalência
14.
PLoS Pathog ; 10(11): e1004522, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25412445

RESUMO

Enteropathogenic E. coli (EPEC) and related enterobacteria rely on a type III secretion system (T3SS) effector NleE to block host NF-κB signaling. NleE is a first in class, novel S-adenosyl-L-methionine (SAM)-dependent methyltransferase that methylates a zinc-coordinating cysteine in the Npl4-like Zinc Finger (NZF) domains in TAB2/3 adaptors in the NF-κB pathway, but its mechanism of action and other human substrates are unknown. Here we solve crystal structure of NleE-SAM complex, which reveals a methyltransferase fold different from those of known ones. The SAM, cradled snugly at the bottom of a deep and narrow cavity, adopts a unique conformation ready for nucleophilic attack by the methyl acceptor. The substrate NZF domain can be well docked into the cavity, and molecular dynamic simulation indicates that Cys673 in TAB2-NZF is spatially and energetically favorable for attacking the SAM. We further identify a new NleE substrate, ZRANB3, that functions in PCNA binding and remodeling of stalled replication forks at the DNA damage sites. Specific inactivation of the NZF domain in ZRANB3 by NleE offers a unique opportunity to suggest that ZRANB3-NZF domain functions in DNA repair processes other than ZRANB3 recruitment to DNA damage sites. Our analyses suggest a novel and unexpected link between EPEC infection, virulence proteins and genome integrity.


Assuntos
DNA Helicases , Reparo do DNA , Escherichia coli Enteropatogênica , Proteínas de Escherichia coli , Simulação de Dinâmica Molecular , Proteínas Metiltransferases , Fatores de Virulência , Linhagem Celular , Cristalografia por Raios X , DNA Helicases/química , DNA Helicases/genética , DNA Helicases/metabolismo , Escherichia coli Enteropatogênica/enzimologia , Escherichia coli Enteropatogênica/genética , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Humanos , Proteínas Metiltransferases/química , Proteínas Metiltransferases/genética , Proteínas Metiltransferases/metabolismo , Estrutura Terciária de Proteína , S-Adenosilmetionina/química , S-Adenosilmetionina/genética , S-Adenosilmetionina/metabolismo , Fatores de Virulência/química , Fatores de Virulência/genética , Fatores de Virulência/metabolismo
15.
Int J Biochem Cell Biol ; 44(12): 2223-32, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22999844

RESUMO

Ubiquitylation is a widespread post-translational global regulatory system that is essential for the proper functioning of various cellular events. Recent studies have shown that certain types of Escherichia coli can exploit specific aspects of the ubiquitylation system to influence downstream targets. Despite these findings, examination of the effects pathogenic E. coli have on the overall host ubiquitylation system remain unexplored. To study the impact that pathogenic E. coli have on the ubiquitylation levels of host proteins during infections, we analyzed the entire ubiquitylation system during enteropathogenic E. coli infections of cultured cells. We found that these microbes caused a dramatic decrease in ubiquitylated host proteins during these infections. This occurred with a concomitant reduction in the expression of essential E1 activating enzymes in the host, which are integral for the initiation of the ubiquitylation cascade. Control of host E1 enzyme levels was dependent on the E. coli adherence factor plasmid which acted on host aspartyl proteases within enteropathogenic E. coli. Hijacking of the ubiquitylation system did not require the plasmid-encoded regulator or bundle forming pilus expression, as enteropathogenic E. coli mutated in those factors did not revert the ubiquitylation of host proteins or the abundance of E1 enzyme proteins to uninfected levels. Our work shows that E. coli have developed strategies to usurp post-translational systems by targeting crucial enzymes. The ability of enteropathogenic E. coli to inactivate host protein ubiquitylation could enable more efficient effector protein functionality, providing increased bacterial control of host cells during enteropathogenic E. coli pathogenesis.


Assuntos
Ácido Aspártico Proteases/metabolismo , Escherichia coli Enteropatogênica/enzimologia , Regulação Enzimológica da Expressão Gênica , Plasmídeos/fisiologia , Enzimas Ativadoras de Ubiquitina/metabolismo , Sistemas de Secreção Bacterianos , Escherichia coli Enteropatogênica/genética , Escherichia coli Enteropatogênica/fisiologia , Células Epiteliais/enzimologia , Células Epiteliais/microbiologia , Células HeLa , Interações Hospedeiro-Patógeno , Humanos , Poliubiquitina/metabolismo , Tioléster Hidrolases/metabolismo , Ubiquitina Tiolesterase/metabolismo , Enzimas Ativadoras de Ubiquitina/genética , Proteínas Ubiquitinadas/metabolismo , Ubiquitinação
16.
Infect Immun ; 80(2): 483-92, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22144482

RESUMO

Enterohemorrhagic Escherichia coli (EHEC) and enteropathogenic E. coli (EPEC) are food-borne pathogens that cause serious diarrheal diseases. To colonize the human intestine, these pathogens must overcome innate immune defenses such as antimicrobial peptides (AMPs). Bacterial pathogens have evolved various mechanisms to resist killing by AMPs, including proteolytic degradation of AMPs. To examine the ability of the EHEC and EPEC OmpT outer membrane (OM) proteases to degrade α-helical AMPs, ompT deletion mutants were generated. Determination of MICs of various AMPs revealed that both mutant strains are more susceptible than their wild-type counterparts to α-helical AMPs, although to different extents. Time course assays monitoring the degradation of LL-37 and C18G showed that EHEC cells degraded both AMPs faster than EPEC cells in an OmpT-dependent manner. Mass spectrometry analyses of proteolytic fragments showed that EHEC OmpT cleaves LL-37 at dibasic sites. The superior protection provided by EHEC OmpT compared to EPEC OmpT against α-helical AMPs was due to higher expression of the ompT gene and, in turn, higher levels of the OmpT protein in EHEC. Fusion of the EPEC ompT promoter to the EHEC ompT open reading frame resulted in decreased OmpT expression, indicating that transcriptional regulation of ompT is different in EHEC and EPEC. We hypothesize that the different contributions of EHEC and EPEC OmpT to the degradation and inactivation of LL-37 may be due to their adaptation to their respective niches within the host, the colon and small intestine, respectively, where the environmental cues and abundance of AMPs are different.


Assuntos
Peptídeos Catiônicos Antimicrobianos/metabolismo , Escherichia coli Êntero-Hemorrágica/enzimologia , Escherichia coli Enteropatogênica/enzimologia , Serina Endopeptidases/metabolismo , Peptídeos Catiônicos Antimicrobianos/farmacologia , Farmacorresistência Bacteriana , Transferência Ressonante de Energia de Fluorescência , Deleção de Genes , Regulação Bacteriana da Expressão Gênica/fisiologia , Regulação Enzimológica da Expressão Gênica/fisiologia , Humanos , Testes de Sensibilidade Microbiana , Regiões Promotoras Genéticas , Serina Endopeptidases/genética , Catelicidinas
17.
J Bacteriol ; 193(17): 4516-22, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21705596

RESUMO

Enteropathogenic Escherichia coli(EPEC) requires the tnaA-encoded enzyme tryptophanase and its substrate tryptophan to synthesize diffusible exotoxins that kill the nematode Caenorhabditis elegans. Here, we demonstrate that the RNA-binding protein CsrA and the tryptophan permease TnaB coregulate tryptophanase activity, through mutually exclusive pathways, to stimulate toxin-mediated paralysis and killing of C. elegans.


Assuntos
Sistemas de Transporte de Aminoácidos/metabolismo , Caenorhabditis elegans/microbiologia , Escherichia coli Enteropatogênica/patogenicidade , Proteínas de Escherichia coli/metabolismo , Exotoxinas/metabolismo , Proteínas de Ligação a RNA/metabolismo , Proteínas Repressoras/metabolismo , Triptofanase/genética , Sequência de Aminoácidos , Sistemas de Transporte de Aminoácidos/genética , Animais , Sequência de Bases , Escherichia coli Enteropatogênica/enzimologia , Escherichia coli Enteropatogênica/genética , Proteínas de Escherichia coli/genética , Regulação Bacteriana da Expressão Gênica , Dados de Sequência Molecular , Óperon , Proteínas de Ligação a RNA/genética , Proteínas Repressoras/genética , Triptofano/metabolismo , Triptofanase/metabolismo
18.
Mol Microbiol ; 80(1): 219-30, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21306441

RESUMO

Many bacterial pathogens utilize a type III secretion system (T3SS) to inject virulence effector proteins into host cells during infection. Previously, we found that enteropathogenic Escherichia coli (EPEC) uses the type III effector, NleE, to block the inflammatory response by inhibiting IκB degradation and nuclear translocation of the p65 subunit of NF-κB. Here we screened further effectors with unknown function for their capacity to prevent p65 nuclear translocation. We observed that ectopic expression of GFP-NleC in HeLa cells led to the degradation of p65. Delivery of NleC by the T3SS of EPEC also induced degradation of p65 in infected cells as well as other NF-κB components, c-Rel and p50. Recombinant His(6) -NleC induced p65 and p50 cleavage in HeLa cell lysates and mutation of a consensus zinc metalloprotease motif, HEIIH, abrogated NleC proteolytic activity. NleC inhibited IL-8 production during prolonged EPEC infection of HeLa cells in a protease activity-dependent manner. A double nleE/nleC mutant was further impaired for its ability to inhibit IL-8 secretion than either a single nleE or a single nleC mutant. We conclude that NleC is a type III effector protease that degrades NF-κB thereby contributing the arsenal of bacterial effectors that inhibit innate immune activation.


Assuntos
Escherichia coli Enteropatogênica/enzimologia , Escherichia coli Enteropatogênica/metabolismo , Proteínas de Escherichia coli/metabolismo , Fator de Transcrição RelA/metabolismo , Proteínas de Escherichia coli/genética , Imunofluorescência , Células HeLa , Humanos , Immunoblotting , Interleucina-8/metabolismo , Microscopia Confocal , Fator de Transcrição RelA/genética
19.
Microbiology (Reading) ; 157(Pt 4): 1145-1160, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21233160

RESUMO

Enteropathogenic Escherichia coli (EPEC) is an important cause of infectious diarrhoea. It colonizes human intestinal epithelial cells by delivering effector proteins into the host cell cytoplasm via a type III secretion system (T3SS) encoded within the chromosomal locus of enterocyte effacement (LEE). The LEE pathogenicity island also encodes a lytic transglycosylase (LT) homologue named EtgA. In the present work we investigated the significance of EtgA function in type III secretion (T3S). Purified recombinant EtgA was found to have peptidoglycan lytic activity in vitro. Consistent with this function, signal peptide processing and bacterial cell fractionation revealed that EtgA is a periplasmic protein. EtgA possesses the conserved glutamate characteristic of the LT family, and we show here that it is essential for enzymic activity. Overproduction of EtgA in EPEC inhibits bacterial growth and induces cell lysis unless the predicted catalytic glutamate is mutated. An etgA mutant is attenuated for T3S, red blood cell haemolysis and EspA filamentation. BfpH, a plasmid-encoded putative LT, was not able to functionally replace EtgA. Overall, our results indicate that the muramidase activity of EtgA is not critical but makes a significant contribution to the efficiency of the T3S process.


Assuntos
Escherichia coli Enteropatogênica/enzimologia , Escherichia coli Enteropatogênica/metabolismo , Proteínas de Escherichia coli/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Muramidase/metabolismo , Fatores de Virulência/metabolismo , Bacteriólise , Eritrócitos/efeitos dos fármacos , Expressão Gênica , Técnicas de Inativação de Genes , Hemólise , Humanos , Hidrólise , Peptidoglicano/metabolismo , Proteínas Periplásmicas/metabolismo , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Virulência
20.
J Biol Chem ; 286(7): 5100-7, 2011 Feb 18.
Artigo em Inglês | MEDLINE | ID: mdl-21148319

RESUMO

The NFκB transcription factor is a key component of immune and inflammatory signaling as its activation induces the expression of antimicrobial reagents, chemokines, cytokines, and anti-apoptotic factors. Many pathogens encode effector proteins that target factors regulating NFκB activity and can provide novel insights on regulatory mechanisms. Given the link of NFκB dysfunction with inflammatory diseases and some cancers, these effectors have therapeutic potential. Here, screening enteropathogenic Escherichia coli proteins for those implicated in suppressing NFκB function revealed that eGFP-NleC, unlike eGFP, strongly inhibited basal and TNFα-induced NFκB reporter activity to prevent secretion of the chemokine, IL-8. Work involving NleC variants, chemical inhibitors, and immunoprecipitation studies support NleC being a zinc metalloprotease that degrades NFκB-IκBα complexes. The findings are consistent with features between residues 33-65 recruiting NFκB for proteasomal-independent degradation by a mechanism inhibited by metalloprotease inhibitors or disruption of a consensus zinc metalloprotease motif spanning NleC residues 183-187. This raises the prospect that mammalian cells, or other pathogens, employ a similar mechanism to modulate NFκB activity. Moreover, NleC represents a novel tool for validating NFκB as a therapeutic target and, indeed, as a possible therapeutic reagent.


Assuntos
Escherichia coli Enteropatogênica/enzimologia , Proteínas de Escherichia coli/metabolismo , Metaloendopeptidases/metabolismo , NF-kappa B/metabolismo , Escherichia coli Enteropatogênica/genética , Proteínas de Escherichia coli/genética , Células HeLa , Humanos , Interleucina-8/metabolismo , Interleucina-8/farmacologia , Metaloendopeptidases/genética , NF-kappa B/genética , Complexo de Endopeptidases do Proteassoma , Fator de Necrose Tumoral alfa/metabolismo , Fator de Necrose Tumoral alfa/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...