Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Nat Ecol Evol ; 8(2): 315-324, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38177692

RESUMO

Reversing the evolution of traits harmful to humans, such as antimicrobial resistance, is a key ambition of applied evolutionary biology. A major impediment to reverse evolution is the relatively low spontaneous mutation rates that revert evolved genotypes back to their ancestral state. However, the repeated re-introduction of ancestral alleles by horizontal gene transfer (HGT) could make reverse evolution likely. Here we evolve populations of an antibiotic-resistant strain of Helicobacter pylori in growth conditions without antibiotics while introducing an ancestral antibiotic-sensitive allele by HGT. We evaluate reverse evolution using DNA sequencing and find that HGT facilitates the molecular reverse evolution of the antibiotic resistance allele, and that selection for high rates of HGT drives the evolution of increased HGT rates in low-HGT treatment populations. Finally, we use a theoretical model and carry out simulations to infer how the fitness costs of antibiotic resistance, rates of HGT and effects of genetic drift interact to determine the probability and predictability of reverse evolution.


Assuntos
Transferência Genética Horizontal , Helicobacter pylori , Humanos , Antibacterianos/farmacologia , Helicobacter pylori/genética , Evolução Molecular , Modelos Teóricos
2.
Proc Natl Acad Sci U S A ; 119(12): e2119010119, 2022 03 22.
Artigo em Inglês | MEDLINE | ID: mdl-35298339

RESUMO

Horizontal gene transfer (HGT) is important for microbial evolution, yet we know little about the fitness effects and dynamics of horizontally transferred genetic variants. In this study, we evolve laboratory populations of Helicobacter pylori, which take up DNA from their environment by natural transformation, and measure the fitness effects of thousands of transferred genetic variants. We find that natural transformation increases the rate of adaptation but comes at the cost of significant genetic load. We show that this cost is circumvented by recombination, which increases the efficiency of selection by decoupling deleterious and beneficial genetic variants. Our results show that adaptation with HGT, pervasive in natural microbial populations, is shaped by a combination of selection, recombination, and genetic drift not accounted for in existing models of evolution.


Assuntos
Transferência Genética Horizontal , Helicobacter pylori , Transferência Genética Horizontal/genética , Helicobacter pylori/genética
3.
Proc Natl Acad Sci U S A ; 117(43): 26868-26875, 2020 10 27.
Artigo em Inglês | MEDLINE | ID: mdl-33055207

RESUMO

Horizontal gene transfer (HGT) confers the rapid acquisition of novel traits and is pervasive throughout microbial evolution. Despite the central role of HGT, the evolutionary forces that drive the dynamics of HGT alleles in evolving populations are poorly understood. Here, we show that HGT alters the evolutionary dynamics of genetic variation, so that deleterious genetic variants, including antibiotic resistance genes, can establish in populations without selection. We evolve antibiotic-sensitive populations of the human pathogen Helicobacter pylori in an environment without antibiotic but with HGT from an antibiotic-resistant isolate of H. pylori We find that HGT increases the rate of adaptation, with most horizontally transferred genetic variants establishing at a low frequency in the population. When challenged with antibiotic, this low-level variation potentiates adaptation, with HGT populations flourishing in conditions where nonpotentiated populations go extinct. By extending previous models of evolution under HGT, we evaluated the conditions for the establishment and spread of HGT-acquired alleles into recipient populations. We then used our model to estimate parameters of HGT and selection from our experimental evolution data. Together, our findings show how HGT can act as an evolutionary force that facilitates the spread of nonselected genetic variation and expands the adaptive potential of microbial populations.


Assuntos
Adaptação Fisiológica/genética , Evolução Biológica , Farmacorresistência Bacteriana/genética , Transferência Genética Horizontal , Helicobacter pylori/genética , Antibacterianos , Fluxo Gênico , Aptidão Genética , Variação Genética , Metronidazol , Seleção Genética
4.
J Enzyme Inhib Med Chem ; 34(1): 1660-1667, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31530039

RESUMO

Ethoxzolamide (EZA), acetazolamide, and methazolamide are clinically used sulphonamide drugs designed to treat non-bacteria-related illnesses (e.g. glaucoma), but they also show antimicrobial activity against the gastric pathogen Helicobacter pylori. EZA showed the highest activity, and was effective against clinical isolates resistant to metronidazole, clarithromycin, and/or amoxicillin, suggesting that EZA kills H. pylori via mechanisms different from that of these antibiotics. The frequency of single-step spontaneous resistance acquisition by H. pylori was less than 5 × 10-9, showing that resistance to EZA does not develop easily. Resistance was associated with mutations in three genes, including the one that encodes undecaprenyl pyrophosphate synthase, a known target of sulphonamides. The data indicate that EZA impacts multiple targets in killing H. pylori. Our findings suggest that developing the approved anti-glaucoma drug EZA into a more effective anti-H. pylori agent may offer a faster and cost-effective route towards new antimicrobials with a novel mechanism of action.


Assuntos
Antibacterianos/farmacologia , Etoxzolamida/farmacologia , Helicobacter pylori/efeitos dos fármacos , Antibacterianos/síntese química , Antibacterianos/química , Relação Dose-Resposta a Droga , Etoxzolamida/síntese química , Etoxzolamida/química , Helicobacter pylori/crescimento & desenvolvimento , Testes de Sensibilidade Microbiana , Estrutura Molecular , Relação Estrutura-Atividade
5.
Artigo em Inglês | MEDLINE | ID: mdl-29468142

RESUMO

The Gram-negative bacterium, Helicobacter pylori, causes chronic gastritis, peptic ulcers, and gastric cancer in humans. Although the gastric epithelium is the primary site of H. pylori colonization, H. pylori can gain access to deeper tissues. Concurring with this notion, H. pylori has been found in the vicinity of endothelial cells in gastric submucosa. Endothelial cells play crucial roles in innate immune response, wound healing and tumorigenesis. This study examines the molecular mechanisms by which H. pylori interacts with and triggers inflammatory responses in endothelial cells. We observed that H. pylori infection of primary human endothelial cells stimulated secretion of the key inflammatory cytokines, interleukin-6 (IL-6) and interleukin-8 (IL-8). In particular, IL-8, a potent chemokine and angiogenic factor, was secreted by H. pylori-infected endothelial cells to levels ~10- to 20-fold higher than that typically observed in H. pylori-infected gastric epithelial cells. These inflammatory responses were triggered by the H. pylori type IV secretion system (T4SS) and the T4SS-associated adhesin CagL, but not the translocation substrate CagA. Moreover, in contrast to integrin α5ß1 playing an essential role in IL-8 induction by H. pylori upon infection of gastric epithelial cells, both integrin α5ß1 and integrin αvß3 were dispensable for IL-8 induction in H. pylori-infected endothelial cells. However, epidermal growth factor receptor (EGFR) is crucial for mediating the potent H. pylori-induced IL-8 response in endothelial cells. This study reveals a novel mechanism by which the H. pylori T4SS and its adhesin subunit, CagL, may contribute to H. pylori pathogenesis by stimulating the endothelial innate immune responses, while highlighting EGFR as a potential therapeutic target for controlling H. pylori-induced inflammation.


Assuntos
Proteínas de Bactérias/metabolismo , Infecções por Helicobacter/microbiologia , Helicobacter pylori/fisiologia , Interações Hospedeiro-Patógeno , Sistemas de Secreção Tipo IV/metabolismo , Adesinas Bacterianas/metabolismo , Biomarcadores , Linhagem Celular , Citocinas/metabolismo , Células Epiteliais/metabolismo , Células Epiteliais/microbiologia , Receptores ErbB/metabolismo , Infecções por Helicobacter/metabolismo , Humanos , Mediadores da Inflamação/metabolismo , NF-kappa B/metabolismo , Fosforilação , Transporte Proteico
6.
Sci Rep ; 7(1): 16140, 2017 11 23.
Artigo em Inglês | MEDLINE | ID: mdl-29170397

RESUMO

The Helicobacter pylori phase variable gene modH, typified by gene HP1522 in strain 26695, encodes a N6-adenosine type III DNA methyltransferase. Our previous studies identified multiple strain-specific modH variants (modH1 - modH19) and showed that phase variation of modH5 in H. pylori P12 influenced expression of motility-associated genes and outer membrane protein gene hopG. However, the ModH5 DNA recognition motif and the mechanism by which ModH5 controls gene expression were unknown. Here, using comparative single molecule real-time sequencing, we identify the DNA site methylated by ModH5 as 5'-Gm6ACC-3'. This motif is vastly underrepresented in H. pylori genomes, but overrepresented in a number of virulence genes, including motility-associated genes, and outer membrane protein genes. Motility and the number of flagella of H. pylori P12 wild-type were significantly higher than that of isogenic modH5 OFF or ΔmodH5 mutants, indicating that phase variable switching of modH5 expression plays a role in regulating H. pylori motility phenotypes. Using the flagellin A (flaA) gene as a model, we show that ModH5 modulates flaA promoter activity in a GACC methylation-dependent manner. These findings provide novel insights into the role of ModH5 in gene regulation and how it mediates epigenetic regulation of H. pylori motility.


Assuntos
Proteínas de Bactérias/metabolismo , Helicobacter pylori/metabolismo , Proteínas de Bactérias/genética , Epigênese Genética/genética , Regulação Bacteriana da Expressão Gênica/genética , Regulação Bacteriana da Expressão Gênica/fisiologia , Helicobacter pylori/genética
7.
Microbiologyopen ; 6(6)2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-29055967

RESUMO

Helicobacter pylori is a gram-negative bacterial pathogen that chronically inhabits the human stomach. To survive and maintain advantage, it has evolved unique host-pathogen interactions mediated by Helicobacter-specific proteins in the bacterial outer membrane. These outer membrane proteins (OMPs) are anchored to the cell surface via a C-terminal ß-barrel domain, which requires their assembly by the ß-barrel assembly machinery (BAM). Here we have assessed the complexity of the OMP C-terminal ß-barrel domains employed by H. pylori, and characterized the H. pyloriBAM complex. Around 50 Helicobacter-specific OMPs were assessed with predictive structural algorithms. The data suggest that H. pylori utilizes a unique ß-barrel architecture that might constitute H. pylori-specific Type V secretions system. The structural and functional diversity in these proteins is encompassed by their extramembrane domains. Bioinformatic and biochemical characterization suggests that the low ß-barrel-complexity requires only minimalist assembly machinery. The H. pylori proteins BamA and BamD associate to form a BAM complex, with features of BamA enabling an oligomerization that might represent a mechanism by which a minimalist BAM complex forms a larger, sophisticated machinery capable of servicing the outer membrane proteome of H. pylori.


Assuntos
Proteínas da Membrana Bacteriana Externa/química , Helicobacter pylori/metabolismo , Proteínas da Membrana Bacteriana Externa/genética , Proteínas da Membrana Bacteriana Externa/metabolismo , Membrana Celular/química , Membrana Celular/genética , Membrana Celular/metabolismo , Cristalografia por Raios X , Helicobacter pylori/química , Helicobacter pylori/genética , Modelos Moleculares , Conformação Proteica , Domínios Proteicos , Dobramento de Proteína
8.
Toxins (Basel) ; 9(8)2017 07 28.
Artigo em Inglês | MEDLINE | ID: mdl-28788072

RESUMO

Cytotoxin-associated gene product A (CagA) is a major virulence factor secreted by Helicobacter pylori. CagA activity in the gastric epithelium is associated with higher risk of gastric cancer development. Bacterial type IV secretion system (T4SS)-mediated translocation of CagA into the cytosol of human epithelial cells occurs via a poorly understood mechanism that requires CagA interaction with the host membrane lipid phosphatidylserine (PS) and host cell receptor integrin α5ß1. Here we have characterized the isolated recombinant middle fragment of CagA (CagA-M) that contains the positively-charged PS-binding region (aa 613-636) and a putative ß1 integrin binding site, but lacks the EPIYA region, secretion signal peptide and the CagA multimerization motif. We show that CagA-M, when immobilized on latex beads, is capable of binding to, and triggering its own uptake into, gastric epithelial cells in the absence of infection with cagA-positive H. pylori. Using site-directed mutagenesis, fluorescent and electron microscopy, and highly-specific inhibitors, we demonstrate that the cell-binding and endocytosis-like internalization of CagA-M are dependent on (1) binding to PS; (2) ß1 integrin activity; and (3) actin dynamics. Interaction of CagA-M with the host cells is accompanied by the development of long filopodia-like protrusions (macrospikes). This novel morphology is different from the hummingbird phenotype induced by the translocation of full-length CagA. The determinants within CagA-M and within the host that are important for endocytosis-like internalization into host cells are very similar to those observed for T4SS-mediated internalization of full-length CagA, suggesting that the latter may involve an endocytic pathway.


Assuntos
Actinas/metabolismo , Antígenos de Bactérias/metabolismo , Proteínas de Bactérias/metabolismo , Células Epiteliais/metabolismo , Células Epiteliais/microbiologia , Helicobacter pylori/fisiologia , Interações Hospedeiro-Patógeno , Antígenos de Bactérias/química , Proteínas de Bactérias/química , Células Cultivadas , Endocitose , Mucosa Gástrica/citologia , Humanos , Proteínas Imobilizadas/metabolismo , Proteínas Recombinantes/metabolismo
9.
Curr Top Microbiol Immunol ; 400: 105-127, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28124151

RESUMO

The methylome is defined as a map of DNA methylation patterns at single-base resolution. DNA methylation in bacteria was first discovered as a function of restriction-modification (R-M) systems. R-M systems in Helicobacter pylori, like those in other bacteria, are important host-specificity determinants that provide protection against foreign DNA. Moreover, the gene regulatory role of the methyltransferase (Mtase) unit of various Helicobacter pylori R-M systems is being increasingly recognized. Recent advances in the application of single-molecule real-time (SMRT) DNA sequencing to analyse DNA methylation have revealed for the first time comprehensive pictures of the genome-wide distribution of methylation sites in various strains of H. pylori. The methylomic data published so far have not only confirmed the significant inter-strain diversity of H. pylori Mtases and their DNA methylation profiles, but also identified numerous novel Mtase target recognition sites. The precise knowledge of the nucleotide sequence of Mtase recognition sites and their distribution within the H. pylori genome will in turn enable researchers to more readily test hypotheses on how H. pylori Mtases function to orchestrate gene regulation and/or modulate virulence. Methylomic studies hold promise for providing a deeper understanding into the roles of H. pylori Mtase and R-M systems in the physiology, epigenetics and possibly also pathogenesis of this important human pathogen. Consequently, the knowledge gained will provide crucial insights into the potential application of H. pylori methylomes as novel biomarkers for the prediction of disease outcome and/or antibiotic susceptibility.


Assuntos
Proteínas de Bactérias/genética , DNA Bacteriano/genética , Regulação Bacteriana da Expressão Gênica , Genoma Bacteriano , Infecções por Helicobacter/microbiologia , Helicobacter pylori/genética , Helicobacter pylori/patogenicidade , Proteínas de Bactérias/metabolismo , Metilação de DNA , DNA Bacteriano/metabolismo , Helicobacter pylori/metabolismo , Humanos , Virulência
10.
Cell Stress Chaperones ; 21(3): 523-33, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26928021

RESUMO

Bacterial infections typically elicit a strong Heat Shock Response (HSR) in host cells. However, the gastric pathogen Helicobacter pylori has the unique ability to repress this response, the mechanism of which has yet to be elucidated. This study sought to characterize the underlying mechanisms by which H. pylori down-modulates host HSP expression upon infection. Examination of isogenic mutant strains of H. pylori defective in components of the type IV secretion system (T4SS), identified the secretion substrate, CagA, to be essential for down-modulation of the HSPs HSPH1 (HSP105), HSPA1A (HSP72), and HSPD1 (HSP60) upon infection of the AGS gastric adenocarcinoma cell line. Ectopic expression of CagA by transient transfection was insufficient to repress HSP expression in AGS or HEK293T cells, suggesting that additional H. pylori factors are required for HSP repression. RT-qPCR analysis of HSP gene expression in AGS cells infected with wild-type H. pylori or isogenic cagA-deletion mutant found no significant change to account for reduced HSP levels. In summary, this study identified CagA to be an essential bacterial factor for H. pylori-mediated suppression of host HSP expression. The novel finding that HSPH1 is down-modulated by H. pylori further highlights the unique ability of H. pylori to repress the HSR within host cells. Elucidation of the mechanism by which H. pylori achieves HSP repression may prove to be beneficial in the identification of novel mechanisms to inhibit the HSR pathway and provide further insight into the interactions between H. pylori and the host gastric epithelium.


Assuntos
Antígenos de Bactérias/metabolismo , Proteínas de Bactérias/metabolismo , Proteínas de Choque Térmico/biossíntese , Helicobacter pylori/genética , Interações Hospedeiro-Patógeno/genética , Antígenos de Bactérias/genética , Proteínas de Bactérias/genética , Regulação da Expressão Gênica/genética , Células HEK293 , Proteínas de Choque Térmico/genética , Resposta ao Choque Térmico/genética , Infecções por Helicobacter/complicações , Infecções por Helicobacter/genética , Infecções por Helicobacter/microbiologia , Helicobacter pylori/patogenicidade , Humanos , Neoplasias Gástricas/complicações , Neoplasias Gástricas/genética , Neoplasias Gástricas/microbiologia
11.
J Infect Dis ; 213(12): 1927-31, 2016 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-26908724

RESUMO

Previous studies suggest overrepresentation of particular polymorphisms within the Helicobacter pylori CagL hypervariable motif (CagLHM) in gastric cancer-associated isolates. However, these disease correlations were geographically variable and ambiguous. We compared the disease correlation of several hundred geographically diverse CagL sequences and identified 33 CagLHM sequence combinations with disparate geographical distribution, revealing substantial worldwide CagLHM diversity, particularly within Asian countries. Notably, polymorphisms E59 and I60 were significantly overrepresented, whereas D58 and E62 were underrepresented, in gastric cancer-associated H. pylori isolates worldwide. Thus, CagLHM regional diversity may contribute to the varied prevalence of H. pylori-related gastric cancer observed in diverse populations.


Assuntos
Proteínas de Bactérias/genética , Infecções por Helicobacter/microbiologia , Helicobacter pylori/genética , Polimorfismo Genético/genética , Neoplasias Gástricas/microbiologia , Motivos de Aminoácidos , Sequência de Aminoácidos , Antígenos de Bactérias/genética , Ásia/epidemiologia , Saúde Global , Infecções por Helicobacter/epidemiologia , Helicobacter pylori/isolamento & purificação , Interações Hospedeiro-Patógeno , Humanos , Prevalência , Neoplasias Gástricas/epidemiologia
12.
PLoS One ; 10(7): e0133531, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26196862

RESUMO

Carriage of the CagA oncoprotein by the human gastric cancer-associated pathogen Helicobacter pylori is significantly associated with this typically benign chronic infection advancing to a potentially fatal outcome. However it remains to be elucidated why only a small subset of individuals infected with H. pylori CagA-positive strains develops gastric cancer. H. pylori translocates CagA into host cells using a type IV secretion apparatus that interacts with host integrin receptors via a three-amino-acid-residue RGD motif on the H. pylori protein CagL. The RGD motif of CagL also plays a major role in the induction of proinflammatory responses. Upstream of this motif is a conserved glycine flanked by four hypervariable amino acid residues (residues 58, 59, 61 and 62). Certain amino acid polymorphisms at 58 and 59 are significantly prevalent in strains from gastric cancer patients in particular geographic regions; Y58E59 is seen in Taiwan and D58K59 in India. In light of the seemingly contradictory findings of recent CagL mutagenesis studies, we have examined the contribution of sequence promiscuity specifically at CagL residues 58 and 59 to CagA translocation and H. pylori-mediated proinflammatory responses of gastric epithelial cells. Using isogenic mutants of H. pylori strains P12 and 26695 with amino acid substitutions at CagL residues 58 and 59, we determined that carriage of the polymorphisms Y58E59, D58K59, D58E59, N58E59 or N58K59 did not significantly alter the capacity of H. pylori to translocate CagA into, or induce IL-8 secretion in, host cells. Our findings, together with other recently published data, suggest that the variation at CagL residues 58 and 59 does not influence type IV secretion system function in isolation, but rather may work in concert with particular polymorphisms elsewhere in CagL to modulate disease progression.


Assuntos
Substituição de Aminoácidos , Antígenos de Bactérias/genética , Proteínas de Bactérias/genética , Helicobacter pylori/genética , Translocação Genética , Linhagem Celular Tumoral , Células Epiteliais/metabolismo , Células Epiteliais/microbiologia , Helicobacter pylori/patogenicidade , Humanos , Inflamação/metabolismo , Inflamação/microbiologia , Interleucina-8/metabolismo , Polimorfismo de Nucleotídeo Único , Sistemas de Secreção Tipo IV/metabolismo
13.
Infect Immun ; 81(10): 3880-93, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23918779

RESUMO

The natural immune response to Helicobacter pylori neither clears infection nor prevents reinfection. However, the ability of secretory antibodies to influence the course of H. pylori infection has not been determined. We compared the natural progression of H. pylori infection in wild-type C57BL/6 mice with that in mice lacking the polymeric immunoglobulin receptor (pIgR) that is essential for the secretion of polymeric antibody across mucosal surfaces. H. pylori SS1-infected wild-type and pIgR knockout (KO) mice were sampled longitudinally for gastrointestinal bacterial load, antibody response, and histological changes. The gastric bacterial loads of wild-type and pIgR KO mice remained constant and comparable at up to 3 months postinfection (mpi) despite SS1-reactive secretory IgA in the intestinal contents of wild-type mice at that time. Conversely, abundant duodenal colonization of pIgR KO animals contrasted with the near-total eradication of H. pylori from the intestine of wild-type animals by 3 mpi. H. pylori was cultured only from the duodenum of those animals in which colonization in the distal gastric antrum was of sufficient density for immunohistological detection. By 6 mpi, the gastric load of H. pylori in wild-type mice was significantly lower than in pIgR KO animals. While there was no corresponding difference between the two mouse strains in gastric pathology results at 6 mpi, reductions in gastric bacterial load correlated with increased gastric inflammation together with an intestinal secretory antibody response in wild-type mice. Together, these results suggest that naturally produced secretory antibodies can modulate the progress of H. pylori infection, particularly in the duodenum.


Assuntos
Anticorpos Antibacterianos/metabolismo , Infecções por Helicobacter/imunologia , Helicobacter pylori , Imunidade nas Mucosas/fisiologia , Mucosa Intestinal/metabolismo , Animais , Western Blotting , Regulação da Expressão Gênica/imunologia , Imunoglobulina A/sangue , Imunoglobulina A/metabolismo , Imunoglobulina G/sangue , Imunoglobulina G/metabolismo , Mucosa Intestinal/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de Imunoglobulina Polimérica/genética , Receptores de Imunoglobulina Polimérica/metabolismo
14.
Cell Microbiol ; 15(4): 554-70, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23107019

RESUMO

The type IV secretion system (T4SS) of Helicobacter pylori triggers massive inflammatory responses during gastric infection by mechanisms that are poorly understood. Here we provide evidence for a novel pathway by which the T4SS structural component, CagL, induces secretion of interleukin-8 (IL-8) independently of CagA translocation and peptidoglycan-sensing nucleotide-binding oligomerization domain 1 (NOD1) signalling. Recombinant CagL was sufficient to trigger IL-8 secretion, requiring activation of α5 ß1 integrin and the arginine-glycine-aspartate (RGD) motif in CagL. Mutation of the encoded RGD motif to arginine-glycine-alanine (RGA) in the cagL gene of H. pylori abrogated its ability to induce IL-8. Comparison of IL-8 induction between H. pylori ΔvirD4 strains bearing wild-type or mutant cagL indicates that CagL-dependent IL-8 induction can occur independently of CagA translocation. In line with this notion, exogenous CagL complemented H. pylori ΔcagL mutant in activating NF-κB and inducing IL-8 without restoring CagA translocation. The CagA translocation-independent, CagL-dependent IL-8 induction involved host signalling via integrin α5 ß1 , Src kinase, the mitogen-activated protein kinase (MAPK) pathway and NF-κB but was independent of NOD1. Our findings reveal a novel pathway whereby CagL, via interaction with host integrins, can trigger pro-inflammatory responses independently of CagA translocation or NOD1 signalling.


Assuntos
Antígenos de Bactérias/metabolismo , Proteínas de Bactérias/imunologia , Proteínas de Bactérias/metabolismo , Sistemas de Secreção Bacterianos , Helicobacter pylori/imunologia , Interleucina-8/metabolismo , Proteína Adaptadora de Sinalização NOD1/metabolismo , Proteínas de Bactérias/genética , Linhagem Celular , Interações Hospedeiro-Patógeno , Humanos , Integrina alfa5beta1/metabolismo , Proteínas Mutantes/genética , Proteínas Mutantes/imunologia , Proteínas Mutantes/metabolismo , Mutação , NF-kappa B/metabolismo , Transdução de Sinais
15.
PLoS One ; 7(8): e43245, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22905239

RESUMO

Autotransporters are secreted proteins that are assembled into the outer membrane of bacterial cells. The passenger domains of autotransporters are crucial for bacterial pathogenesis, with some remaining attached to the bacterial surface while others are released by proteolysis. An enigma remains as to whether autotransporters should be considered a class of secretion system, or simply a class of substrate with peculiar requirements for their secretion. We sought to establish a sensitive search protocol that could identify and characterize diverse autotransporters from bacterial genome sequence data. The new sequence analysis pipeline identified more than 1500 autotransporter sequences from diverse bacteria, including numerous species of Chlamydiales and Fusobacteria as well as all classes of Proteobacteria. Interrogation of the proteins revealed that there are numerous classes of passenger domains beyond the known proteases, adhesins and esterases. In addition the barrel-domain-a characteristic feature of autotransporters-was found to be composed from seven conserved sequence segments that can be arranged in multiple ways in the tertiary structure of the assembled autotransporter. One of these conserved motifs overlays the targeting information required for autotransporters to reach the outer membrane. Another conserved and diagnostic motif maps to the linker region between the passenger domain and barrel-domain, indicating it as an important feature in the assembly of autotransporters.


Assuntos
Biologia Computacional/métodos , Adesinas Bacterianas/metabolismo , Motivos de Aminoácidos , Proteínas de Bactérias/química , Transporte Biológico , Chlamydiales/metabolismo , Escherichia coli/metabolismo , Fusobactérias/metabolismo , Humanos , Cadeias de Markov , Filogenia , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Ralstonia/metabolismo , Software
16.
PLoS One ; 6(12): e27569, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22162751

RESUMO

Many host-adapted bacterial pathogens contain DNA methyltransferases (mod genes) that are subject to phase-variable expression (high-frequency reversible ON/OFF switching of gene expression). In Haemophilus influenzae and pathogenic Neisseria, the random switching of the modA gene, associated with a phase-variable type III restriction modification (R-M) system, controls expression of a phase-variable regulon of genes (a "phasevarion"), via differential methylation of the genome in the modA ON and OFF states. Phase-variable type III R-M systems are also found in Helicobacter pylori, suggesting that phasevarions may also exist in this key human pathogen. Phylogenetic studies on the phase-variable type III modH gene revealed that there are 17 distinct alleles in H. pylori, which differ only in their DNA recognition domain. One of the most commonly found alleles was modH5 (16% of isolates). Microarray analysis comparing the wild-type P12modH5 ON strain to a P12ΔmodH5 mutant revealed that six genes were either up- or down-regulated, and some were virulence-associated. These included flaA, which encodes a flagella protein important in motility and hopG, an outer membrane protein essential for colonization and associated with gastric cancer. This study provides the first evidence of this epigenetic mechanism of gene expression in H. pylori. Characterisation of H. pylori modH phasevarions to define stable immunological targets will be essential for vaccine development and may also contribute to understanding H. pylori pathogenesis.


Assuntos
Epigênese Genética , Regulação Bacteriana da Expressão Gênica , Helicobacter pylori/genética , Algoritmos , Alelos , Proteínas de Bactérias/genética , DNA/genética , Metilação de DNA , Metilases de Modificação do DNA/genética , Epigenômica , Variação Genética , Modelos Genéticos , Reação em Cadeia da Polimerase , Estrutura Terciária de Proteína , Regulon , Análise de Sequência de DNA , Software
17.
Infect Immun ; 77(11): 5116-29, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19737910

RESUMO

Studies of active immunization against Helicobacter pylori indicate that antibodies play a minor role in immunity. There is also evidence, however, that the translocation of antibodies in the stomach may be insufficient to achieve functional antibody levels in the gastric lumen. We have used a suckling mouse model of passive immunity to determine if perorally delivered antibodies can protect against infection with H. pylori. Female C57BL/6 mice were immunized parenterally with formalin-fixed cells of three clinical isolates of H. pylori (3HP) or the mouse-adapted H. pylori strain SS1 before mating. Their pups were challenged with the SS1 strain at 4 days of age and left to suckle before determination of bacterial loads 14 days later. Compared to age-matched controls, pups suckled by 3HP-vaccinated dams were significantly protected against infection (>95% reduction in median bacterial load; P<0.0001). Pups suckled by SS1-vaccinated dams were also significantly protected in terms of both median bacterial load (>99.5% reduction; P<0.0001) and the number of culture-negative pups (28% versus 2% for immune and nonimmune cohorts, respectively; P<0.0001). Similar results were obtained with pups suckled by dams immunized with a urease-deficient mutant of SS1. Fostering experiments demonstrated that protection was entirely attributable to suckling from an immunized dam, and antibody isotype analysis suggested that protection was mediated by the immunoglobulin G fraction of immune milk. Analysis of the bacterial loads in pups sampled before and after weaning confirmed that infection had been prevented in culture-negative animals. These data indicate that antibodies can prevent colonization by H. pylori and suppress the bacterial loads in animals that are colonized.


Assuntos
Anticorpos Antibacterianos/administração & dosagem , Anticorpos Antibacterianos/imunologia , Infecções por Helicobacter/prevenção & controle , Imunidade Materno-Adquirida/imunologia , Leite/imunologia , Animais , Animais Lactentes , Feminino , Infecções por Helicobacter/sangue , Infecções por Helicobacter/imunologia , Helicobacter pylori/imunologia , Imunoglobulina A/análise , Imunoglobulina A/imunologia , Imunoglobulina G/análise , Imunoglobulina G/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Leite/química
18.
FEMS Immunol Med Microbiol ; 44(2): 213-9, 2005 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-15866218

RESUMO

Helicobacter pylori populations recovered from the human stomach display extensive recombination and quasispecies development, and this suggests frequent exchange of DNA between different strains in vivo. In vitro, however, most H. pylori strains display restriction to the uptake of non-self DNA, as measured using selectable markers, regardless of their competency for transformation with self DNA. We have examined the effect of different selectable markers on double-crossover recombination efficiencies in three reference strains (1061, 26695 & SS1) and one clinical isolate (CHP1) of H. pylori. All strains were efficiently transformable to kanamycin or chloramphenicol resistance by using self-genomic DNA from isogenic mutants bearing the aphA3 or cat cassettes, respectively. However, strains 26695 and CHP1 showed a 3-5-log reduction in transformation efficiency by non-self recombinant DNA containing aphA3, when compared to cat. Strain 1061 readily accepted either cassette, and strain SS1 was poorly tolerant of any non-self DNA. Genome-wide random mutagenesis of these strains was only achievable with a selectable marker that allowed high transformation efficiency. Digestion of 32P-labelled cassettes by H. pylori lysates mirrored the transformation results and indicated that in some strains these cassettes are the targets of enzymatic restriction.


Assuntos
Proteínas de Bactérias/metabolismo , Enzimas de Restrição do DNA/metabolismo , DNA Bacteriano/metabolismo , Farmacorresistência Bacteriana/genética , Helicobacter pylori/genética , Transformação Bacteriana , Proteínas de Bactérias/genética , Cloranfenicol/farmacologia , DNA Bacteriano/genética , Marcadores Genéticos , Helicobacter pylori/efeitos dos fármacos , Helicobacter pylori/metabolismo , Humanos , Canamicina/farmacologia , Mutagênese , Recombinação Genética
19.
J Virol ; 76(21): 11113-22, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12368353

RESUMO

Picornavirus RNA replication requires the formation of replication complexes (RCs) consisting of virus-induced vesicles associated with viral nonstructural proteins and RNA. Brefeldin A (BFA) has been shown to strongly inhibit RNA replication of poliovirus but not of encephalomyocarditis virus (EMCV). Here, we demonstrate that the replication of parechovirus 1 (ParV1) is partly resistant to BFA, whereas echovirus 11 (EV11) replication is strongly inhibited. Since BFA inhibits COPI-dependent steps in endoplasmic reticulum (ER)-Golgi transport, we tested a hypothesis that different picornaviruses may have differential requirements for COPI in the formation of their RCs. Using immunofluorescence and cryo-immunoelectron microscopy we examined the association of a COPI component, beta-COP, with the RCs of EMCV, ParV1, and EV11. EMCV RCs did not contain beta-COP. In contrast, beta-COP appeared to be specifically distributed to the RCs of EV11. In ParV1-infected cells beta-COP was largely dispersed throughout the cytoplasm, with some being present in the RCs. These results suggest that there are differences in the involvement of COPI in the formation of the RCs of various picornaviruses, corresponding to their differential sensitivity to BFA. EMCV RCs are likely to be formed immediately after vesicle budding from the ER, prior to COPI association with membranes. ParV1 RCs are formed from COPI-containing membranes but COPI is unlikely to be directly involved in their formation, whereas formation of EV11 RCs appears to be dependent on COPI association with membranes.


Assuntos
Vesículas Revestidas pelo Complexo de Proteína do Envoltório/metabolismo , Complexo I de Proteína do Envoltório/metabolismo , Vírus da Encefalomiocardite/genética , Enterovirus Humano B/genética , Parechovirus/genética , RNA de Cadeia Dupla/metabolismo , RNA Viral/metabolismo , Replicação Viral , Animais , Antivirais/farmacologia , Brefeldina A/farmacologia , Linhagem Celular , Chlorocebus aethiops , Proteína Coatomer/metabolismo , Vírus da Encefalomiocardite/efeitos dos fármacos , Vírus da Encefalomiocardite/fisiologia , Vírus da Encefalomiocardite/ultraestrutura , Enterovirus Humano B/efeitos dos fármacos , Enterovirus Humano B/fisiologia , Enterovirus Humano B/ultraestrutura , Humanos , Parechovirus/efeitos dos fármacos , Parechovirus/fisiologia , Parechovirus/ultraestrutura
20.
Proc Natl Acad Sci U S A ; 99(10): 7066-71, 2002 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-12011463

RESUMO

Enterotoxigenic Escherichia coli (ETEC) is an enteric pathogen that causes cholera-like diarrhea in humans and animals. ETEC secretes a heat-labile enterotoxin (LT), which resembles cholera toxin, but the actual mechanism of LT secretion is presently unknown. We have identified a previously unrecognized type II protein secretion pathway in the prototypic human ETEC strain, H10407 (serotype O78:H11). The genes for this pathway are absent from E. coli K-12, although examination of the K-12 genome suggests that it probably once possessed them. The secretory pathway bears significant homology at the amino acid level to the type II protein secretory pathway required by Vibrio cholerae for the secretion of cholera toxin. With this in mind, we determined whether the homologous pathway of E. coli H10407 played a role in the secretion of LT. To this end, we inactivated the pathway by inserting a kanamycin-resistance gene into one of the genes (gspD) of the type II secretion pathway by homologous recombination. LT secretion by E. coli H10407 and the gspD mutant was assayed by enzyme immunoassay, and its biological activity was assessed by using Y-1 adrenal cells. This investigation showed that the protein secretory pathway is functional and necessary for the secretion of LT by ETEC. Our findings have revealed the mechanism for the secretion of LT by ETEC, which previously was unknown, and provide further evidence of close biological similarities of the LT and cholera toxin.


Assuntos
Proteínas de Bactérias/metabolismo , Toxinas Bacterianas/metabolismo , Enterotoxinas/metabolismo , Proteínas de Escherichia coli , Escherichia coli/metabolismo , Proteínas de Bactérias/genética , Toxinas Bacterianas/genética , Sequência de Bases , DNA Bacteriano , Enterotoxinas/genética , Escherichia coli/genética , Escherichia coli/crescimento & desenvolvimento , Genes Bacterianos , Dados de Sequência Molecular , Análise de Sequência de DNA
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...