Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
Mol Oncol ; 18(6): 1437-1459, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38627210

ABSTRACT

Different molecular classifications for gastric cancer (GC) have been proposed based on multi-omics platforms with the long-term goal of improved precision treatment. However, the GC (phospho)proteome remains incompletely characterized, particularly at the level of tyrosine phosphorylation. In addition, previous multiomics-based stratification of patient cohorts has lacked identification of corresponding cell line models and comprehensive validation of broad or subgroup-selective therapeutic targets. To address these knowledge gaps, we applied a reverse approach, undertaking the most comprehensive (phospho)proteomic analysis of GC cell lines to date and cross-validating this using publicly available data. Mass spectrometry (MS)-based (phospho)proteomic and tyrosine phosphorylation datasets were subjected to individual or integrated clustering to identify subgroups that were subsequently characterized in terms of enriched molecular processes and pathways. Significant congruence was detected between cell line proteomic and specific patient-derived transcriptomic subclassifications. Many protein kinases exhibiting 'outlier' expression or phosphorylation in the cell line dataset exhibited genomic aberrations in patient samples and association with poor prognosis, with casein kinase I isoform delta/epsilon (CSNK1D/E) being experimentally validated as potential therapeutic targets. Src family kinases were predicted to be commonly hyperactivated in GC cell lines, consistent with broad sensitivity to the next-generation Src inhibitor eCF506. In addition, phosphoproteomic and integrative clustering segregated the cell lines into two subtypes, with epithelial-mesenchyme transition (EMT) and proliferation-associated processes enriched in one, designated the EMT subtype, and metabolic pathways, cell-cell junctions, and the immune response dominating the features of the other, designated the metabolism subtype. Application of kinase activity prediction algorithms and interrogation of gene dependency and drug sensitivity databases predicted that the mechanistic target of rapamycin kinase (mTOR) and dual specificity mitogen-activated protein kinase kinase 2 (MAP2K2) represented potential therapeutic targets for the EMT and metabolism subtypes, respectively, and this was confirmed using selective inhibitors. Overall, our study provides novel, in-depth insights into GC proteomics, kinomics, and molecular taxonomy and reveals potential therapeutic targets that could provide the basis for precision treatments.


Subject(s)
Proteome , Stomach Neoplasms , Stomach Neoplasms/metabolism , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Stomach Neoplasms/classification , Humans , Proteome/metabolism , Cell Line, Tumor , Proteomics/methods , Phosphorylation , Molecular Targeted Therapy
2.
Nat Ecol Evol ; 8(2): 315-324, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38177692

ABSTRACT

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.


Subject(s)
Gene Transfer, Horizontal , Helicobacter pylori , Humans , Anti-Bacterial Agents/pharmacology , Helicobacter pylori/genetics , Evolution, Molecular , Models, Theoretical
3.
Bioinformatics ; 38(17): 4206-4213, 2022 09 02.
Article in English | MEDLINE | ID: mdl-35801909

ABSTRACT

MOTIVATION: The molecular subtyping of gastric cancer (adenocarcinoma) into four main subtypes based on integrated multiomics profiles, as proposed by The Cancer Genome Atlas (TCGA) initiative, represents an effective strategy for patient stratification. However, this approach requires the use of multiple technological platforms, and is quite expensive and time-consuming to perform. A computational approach that uses histopathological image data to infer molecular subtypes could be a practical, cost- and time-efficient complementary tool for prognostic and clinical management purposes. RESULTS: Here, we propose a deep learning ensemble approach (called DEMoS) capable of predicting the four recognized molecular subtypes of gastric cancer directly from histopathological images. DEMoS achieved tile-level area under the receiver-operating characteristic curve (AUROC) values of 0.785, 0.668, 0.762 and 0.811 for the prediction of these four subtypes of gastric cancer [i.e. (i) Epstein-Barr (EBV)-infected, (ii) microsatellite instability (MSI), (iii) genomically stable (GS) and (iv) chromosomally unstable tumors (CIN)] using an independent test dataset, respectively. At the patient-level, it achieved AUROC values of 0.897, 0.764, 0.890 and 0.898, respectively. Thus, these four subtypes are well-predicted by DEMoS. Benchmarking experiments further suggest that DEMoS is able to achieve an improved classification performance for image-based subtyping and prevent model overfitting. This study highlights the feasibility of using a deep learning ensemble-based method to rapidly and reliably subtype gastric cancer (adenocarcinoma) solely using features from histopathological images. AVAILABILITY AND IMPLEMENTATION: All whole slide images used in this study was collected from the TCGA database. This study builds upon our previously published HEAL framework, with related documentation and tutorials available at http://heal.erc.monash.edu.au. The source code and related models are freely accessible at https://github.com/Docurdt/DEMoS.git. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Subject(s)
Adenocarcinoma , Deep Learning , Stomach Neoplasms , Humans , Stomach Neoplasms/diagnostic imaging , Stomach Neoplasms/genetics , Adenocarcinoma/diagnostic imaging , Adenocarcinoma/genetics , Microsatellite Instability
4.
NPJ Precis Oncol ; 6(1): 45, 2022 Jun 23.
Article in English | MEDLINE | ID: mdl-35739342

ABSTRACT

Gastric cancer is one of the deadliest cancers worldwide. An accurate prognosis is essential for effective clinical assessment and treatment. Spatial patterns in the tumor microenvironment (TME) are conceptually indicative of the staging and progression of gastric cancer patients. Using spatial patterns of the TME by integrating and transforming the multiplexed immunohistochemistry (mIHC) images as Cell-Graphs, we propose a graph neural network-based approach, termed Cell-Graph Signature or CGSignature, powered by artificial intelligence, for the digital staging of TME and precise prediction of patient survival in gastric cancer. In this study, patient survival prediction is formulated as either a binary (short-term and long-term) or ternary (short-term, medium-term, and long-term) classification task. Extensive benchmarking experiments demonstrate that the CGSignature achieves outstanding model performance, with Area Under the Receiver Operating Characteristic curve of 0.960 ± 0.01, and 0.771 ± 0.024 to 0.904 ± 0.012 for the binary- and ternary-classification, respectively. Moreover, Kaplan-Meier survival analysis indicates that the "digital grade" cancer staging produced by CGSignature provides a remarkable capability in discriminating both binary and ternary classes with statistical significance (P value < 0.0001), significantly outperforming the AJCC 8th edition Tumor Node Metastasis staging system. Using Cell-Graphs extracted from mIHC images, CGSignature improves the assessment of the link between the TME spatial patterns and patient prognosis. Our study suggests the feasibility and benefits of such an artificial intelligence-powered digital staging system in diagnostic pathology and precision oncology.

5.
Proc Natl Acad Sci U S A ; 119(12): e2119010119, 2022 03 22.
Article in English | MEDLINE | ID: mdl-35298339

ABSTRACT

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.


Subject(s)
Gene Transfer, Horizontal , Helicobacter pylori , Gene Transfer, Horizontal/genetics , Helicobacter pylori/genetics
6.
Oncol Lett ; 21(5): 402, 2021 May.
Article in English | MEDLINE | ID: mdl-33777225

ABSTRACT

Recognized as a group I carcinogen for gastric cancer (GC) and a factor involved in the development of GC, Helicobacter pylori serves a major part in GC research. However, most studies have focused on H. pylori itself, ignoring the complicated pathogenic microbiological environment of GC and neglecting the synergistic or antagonistic effects of H. pylori with other pathogenic microorganisms. Increasing evidence has revealed that the human cytomegalovirus (HCMV) is present in several types of tumors and serves an important role in the neoplastic process of certain human malignant tumors, including GC. The aim of the present study was to explore the role of HCMV and H. pylori co-infection in GC. HCMV and H. pylori infection was analyzed in paired gastric tumor and peri-tumoral tissues from 134 (98 male and 36 female) patients using PCR. The results revealed that a total of 74 (55.2%) patients had H. pylori infection, 58 patients (43.3%) had HCMV infection, and 34 (25.4%) patients had both HCMV and H. pylori infection. Univariate and multivariate analyses demonstrated that H. pylori infection was independently associated with advanced lymphatic metastasis [P=0.007; odds ratio (OR)=3.51]. Furthermore, compared with HCMV-/H. pylori -, neither HCMV+/H. pylori - nor HCMV+/H. pylori + were associated with metastasis, but HCMV-/H. pylori + co-infection status was an independent risk factor for advanced lymphatic metastasis (P=0.005; OR=6.00). In conclusion, GC co-infected with HCMV and H. pylori exhibited a low tendency of lymph node metastasis. HCMV may interact with H. pylori to inhibit the process of lymphatic metastasis, and the mechanism requires further investigation.

7.
Proc Natl Acad Sci U S A ; 117(43): 26868-26875, 2020 10 27.
Article in English | MEDLINE | ID: mdl-33055207

ABSTRACT

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.


Subject(s)
Adaptation, Physiological/genetics , Biological Evolution , Drug Resistance, Bacterial/genetics , Gene Transfer, Horizontal , Helicobacter pylori/genetics , Anti-Bacterial Agents , Gene Flow , Genetic Fitness , Genetic Variation , Metronidazole , Selection, Genetic
8.
J Enzyme Inhib Med Chem ; 34(1): 1660-1667, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31530039

ABSTRACT

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.


Subject(s)
Anti-Bacterial Agents/pharmacology , Ethoxzolamide/pharmacology , Helicobacter pylori/drug effects , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Dose-Response Relationship, Drug , Ethoxzolamide/chemical synthesis , Ethoxzolamide/chemistry , Helicobacter pylori/growth & development , Microbial Sensitivity Tests , Molecular Structure , Structure-Activity Relationship
9.
Article in English | MEDLINE | ID: mdl-29468142

ABSTRACT

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.


Subject(s)
Bacterial Proteins/metabolism , Helicobacter Infections/microbiology , Helicobacter pylori/physiology , Host-Pathogen Interactions , Type IV Secretion Systems/metabolism , Adhesins, Bacterial/metabolism , Biomarkers , Cell Line , Cytokines/metabolism , Epithelial Cells/metabolism , Epithelial Cells/microbiology , ErbB Receptors/metabolism , Helicobacter Infections/metabolism , Humans , Inflammation Mediators/metabolism , NF-kappa B/metabolism , Phosphorylation , Protein Transport
10.
Sci Rep ; 7(1): 16140, 2017 11 23.
Article in English | MEDLINE | ID: mdl-29170397

ABSTRACT

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.


Subject(s)
Bacterial Proteins/metabolism , Helicobacter pylori/metabolism , Bacterial Proteins/genetics , Epigenesis, Genetic/genetics , Gene Expression Regulation, Bacterial/genetics , Gene Expression Regulation, Bacterial/physiology , Helicobacter pylori/genetics
11.
Microbiologyopen ; 6(6)2017 12.
Article in English | MEDLINE | ID: mdl-29055967

ABSTRACT

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.


Subject(s)
Bacterial Outer Membrane Proteins/chemistry , Helicobacter pylori/metabolism , Bacterial Outer Membrane Proteins/genetics , Bacterial Outer Membrane Proteins/metabolism , Cell Membrane/chemistry , Cell Membrane/genetics , Cell Membrane/metabolism , Crystallography, X-Ray , Helicobacter pylori/chemistry , Helicobacter pylori/genetics , Models, Molecular , Protein Conformation , Protein Domains , Protein Folding
12.
Toxins (Basel) ; 9(8)2017 07 28.
Article in English | MEDLINE | ID: mdl-28788072

ABSTRACT

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.


Subject(s)
Actins/metabolism , Antigens, Bacterial/metabolism , Bacterial Proteins/metabolism , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Helicobacter pylori/physiology , Host-Pathogen Interactions , Antigens, Bacterial/chemistry , Bacterial Proteins/chemistry , Cells, Cultured , Endocytosis , Gastric Mucosa/cytology , Humans , Immobilized Proteins/metabolism , Recombinant Proteins/metabolism
13.
Curr Top Microbiol Immunol ; 400: 105-127, 2017.
Article in English | MEDLINE | ID: mdl-28124151

ABSTRACT

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.


Subject(s)
Bacterial Proteins/genetics , DNA, Bacterial/genetics , Gene Expression Regulation, Bacterial , Genome, Bacterial , Helicobacter Infections/microbiology , Helicobacter pylori/genetics , Helicobacter pylori/pathogenicity , Bacterial Proteins/metabolism , DNA Methylation , DNA, Bacterial/metabolism , Helicobacter pylori/metabolism , Humans , Virulence
14.
Cell Stress Chaperones ; 21(3): 523-33, 2016 05.
Article in English | MEDLINE | ID: mdl-26928021

ABSTRACT

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.


Subject(s)
Antigens, Bacterial/metabolism , Bacterial Proteins/metabolism , Heat-Shock Proteins/biosynthesis , Helicobacter pylori/genetics , Host-Pathogen Interactions/genetics , Antigens, Bacterial/genetics , Bacterial Proteins/genetics , Gene Expression Regulation/genetics , HEK293 Cells , Heat-Shock Proteins/genetics , Heat-Shock Response/genetics , Helicobacter Infections/complications , Helicobacter Infections/genetics , Helicobacter Infections/microbiology , Helicobacter pylori/pathogenicity , Humans , Stomach Neoplasms/complications , Stomach Neoplasms/genetics , Stomach Neoplasms/microbiology
15.
J Infect Dis ; 213(12): 1927-31, 2016 06 15.
Article in English | MEDLINE | ID: mdl-26908724

ABSTRACT

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.


Subject(s)
Bacterial Proteins/genetics , Helicobacter Infections/microbiology , Helicobacter pylori/genetics , Polymorphism, Genetic/genetics , Stomach Neoplasms/microbiology , Amino Acid Motifs , Amino Acid Sequence , Antigens, Bacterial/genetics , Asia/epidemiology , Global Health , Helicobacter Infections/epidemiology , Helicobacter pylori/isolation & purification , Host-Pathogen Interactions , Humans , Prevalence , Stomach Neoplasms/epidemiology
16.
PLoS One ; 10(7): e0133531, 2015.
Article in English | MEDLINE | ID: mdl-26196862

ABSTRACT

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.


Subject(s)
Amino Acid Substitution , Antigens, Bacterial/genetics , Bacterial Proteins/genetics , Helicobacter pylori/genetics , Translocation, Genetic , Cell Line, Tumor , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Helicobacter pylori/pathogenicity , Humans , Inflammation/metabolism , Inflammation/microbiology , Interleukin-8/metabolism , Polymorphism, Single Nucleotide , Type IV Secretion Systems/metabolism
17.
Helicobacter ; 20(6): 449-59, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25864465

ABSTRACT

BACKGROUND: Previous findings have suggested that Helicobacter pylori induces autophagic processes and subsequently takes refuge in autophagosomes, thereby contributing to persistent infection. Recently, a noncanonical form of autophagy, LC3 (microtubule-associated protein 1 light chain 3)-associated phagocytosis (LAP), has been shown to be required for efficient clearance of some intracellular bacteria. Whether H. pylori infection induces LAP had not been examined previously. In this study, we determined the extent to which H. pylori infection induces canonical autophagy or LAP in macrophages, and the involvement of the H. pylori cag pathogenicity island (cagPAI) with these processes. METHODS: Immunofluorescence confocal microscopy was used to analyze the formation of GFP-LC3 puncta and their colocalization with H. pylori. Transmission electron microscopy was used to detect the ultrastructure of H. pylori-containing compartments. RESULTS: The majority of intracellular bacteria (85-95%) were found in phagosomes that were LC3-negative, with a small proportion (4-14%) appearing "free" in the cytosol. Only a very small percentage (0.5-6%) of intracellular H. pylori was sequestered in autophagosomes. Furthermore, no statistically significant difference in the relative distribution of H. pylori in the various compartments was observed between wild-type and cagPAI-mutant bacteria. CONCLUSIONS: In macrophages, H. pylori infection does not induce LAP, but can induce canonical autophagy, which entraps a very small fraction of intracellular bacteria. We propose that this subpopulation of intracellular H. pylori might have escaped from phagosomes into the cytosol before being sequestered by autophagosomes. The cagPAI of H. pylori has only minor influence, if any, on the extent of these processes.


Subject(s)
Autophagy , Helicobacter pylori/immunology , Host-Pathogen Interactions , Macrophages/microbiology , Macrophages/physiology , Microtubule-Associated Proteins/analysis , Phagocytosis , Animals , Cells, Cultured , Green Fluorescent Proteins/analysis , Humans , Mice, Inbred C57BL , Microscopy, Confocal , Microscopy, Electron, Transmission , Microscopy, Fluorescence , Staining and Labeling
18.
World J Microbiol Biotechnol ; 30(9): 2481-90, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24854336

ABSTRACT

Helicobacter pylori infection is common in Iran as in other developing countries. Certain genotypes of H. pylori have been associated with increased occurrence of chronic gastritis, peptic ulcers, and gastric adenocarcinoma. The aim of this study was to investigate the clinical relevance of cagL gene and other virulence genotypes of H. pylori isolates with clinical outcomes in Iranian patients. Totally, 126 symptomatic patients who underwent gastroduodenal endoscopy were enrolled in the study. Sixty-one H. pylori strains were isolated from the patients studied. The presence of the cagL, cagA, vacA, iceA, babA2 and sabA genes in the corresponding H. pylori isolates were determined by polymerase chain reaction and the results were compared with clinical outcomes and histopathology. The cagL, cagA, vacA s1, vacA s2, vacA m1, vacA m2, iceA1, iceA2, babA 2 , and sabA genotypes were detected in 96.7, 85.2, 75.4, 24.6, 29.5, 70.5, 42.6, 23, 96.7, and 83.6% of the isolates, respectively. The three genotypic combinations, cagL/cagA/vacAs1m1/iceA1/babA2/sabA, cagL/cagA/vacAs1m2/iceA1/babA2/sabA, and cagL/cagA/vacAs1m2/iceA2/babA2/sabA were determined as the most prevalent combined genotypes. There was a significant correlation between the presence of cagL gene and cagA positivity (P = 0.02). No significant correlation was found between the various genotypes and clinical outcomes (P > 0.05). The present study showed a very high prevalence of cagL genotype among the H. pylori isolates from Iranian patients. Our results demonstrated that neither single genotype nor combination genotypes of virulence-associated genes was significantly helpful markers for predicting the severity of gastroduodenal disease associated with H. pylori infection in Iranian patients.


Subject(s)
Bacterial Proteins/genetics , Helicobacter Infections/microbiology , Helicobacter Infections/pathology , Helicobacter pylori/genetics , Virulence Factors/genetics , Antigens, Bacterial/genetics , Endoscopy, Gastrointestinal , Genetic Markers , Genotype , Helicobacter pylori/isolation & purification , Histocytochemistry , Humans , Iran , Treatment Outcome
19.
J Biol Chem ; 289(10): 6332-6340, 2014 Mar 07.
Article in English | MEDLINE | ID: mdl-24375407

ABSTRACT

The gastric pathogen Helicobacter pylori is a major cause of acute chronic gastritis and the development of stomach and duodenal ulcers. Chronic infection furthermore predisposes to the development of gastric cancer. Crucial to H. pylori survival within the hostile environment of the digestive system are the adhesins SabA and BabA; these molecules belong to the same protein family and permit the bacteria to bind tightly to sugar moieties Lewis(B) and sialyl-Lewis(X), respectively, on the surface of epithelial cells lining the stomach and duodenum. To date, no representative SabA/BabA structure has been determined, hampering the development of strategies to eliminate persistent H. pylori infections that fail to respond to conventional therapy. Here, using x-ray crystallography, we show that the soluble extracellular adhesin domain of SabA shares distant similarity to the tetratricopeptide repeat fold family. The molecule broadly resembles a golf putter in shape, with the head region featuring a large cavity surrounded by loops that vary in sequence between different H. pylori strains. The N-terminal and C-terminal helices protrude at right angles from the head domain and together form a shaft that connects to a predicted outer membrane protein-like ß-barrel trans-membrane domain. Using surface plasmon resonance, we were able to detect binding of the SabA adhesin domain to sialyl-Lewis(X) and Lewis(X) but not to Lewis(A), Lewis(B), or Lewis(Y). Substitution of the highly conserved glutamine residue 159 in the predicted ligand-binding pocket abrogates the binding of the SabA adhesin domain to sialyl-Lewis(X) and Lewis(X). Taken together, these data suggest that the adhesin domain of SabA is sufficient in isolation for specific ligand binding.


Subject(s)
Adhesins, Bacterial/chemistry , Helicobacter pylori/metabolism , Lewis X Antigen/chemistry , N-Acetylneuraminic Acid/chemistry , Adhesins, Bacterial/genetics , Amino Acid Sequence , Amino Acid Substitution , Binding Sites , Conserved Sequence , Crystallography, X-Ray , Glutamine/chemistry , Glutamine/genetics , Ligands , Molecular Sequence Data , Protein Structure, Secondary , Protein Structure, Tertiary , Sialyl Lewis X Antigen , Surface Plasmon Resonance
20.
PLoS One ; 8(11): e79367, 2013.
Article in English | MEDLINE | ID: mdl-24223932

ABSTRACT

The CagA protein of Helicobacter pylori is associated with increased virulence and gastric cancer risk. CagA is translocated into the host cell by a H. pylori type IV secretion system via mechanisms that are poorly understood. Translocated CagA interacts with numerous host factors, altering a variety of host signalling pathways. The recently determined crystal structure of C-terminally-truncated CagA indicated the presence of two domains: the smaller, flexible N-terminal domain and the larger, middle domain. In this study, we have investigated the conformation, oligomeric state and stability of the N-terminal, middle and glutamate-proline-isoleucine-tyrosine-alanine (EPIYA)-repeats domains. All three domains are monomeric, suggesting that the multimerisation of CagA observed in infected cells is likely to be mediated not by CagA itself but by its interacting partners. The middle and the C-terminal domains, but not the N-terminal domain, are capable of refolding spontaneously upon heat denaturation, lending support to the hypothesis that unfolded CagA is threaded C-terminus first through the type IV secretion channel with its N-terminal domain, which likely requires interactions with other domains to refold, being threaded last. Our findings also revealed that the C-terminal EPIYA-repeats domain of CagA exists in an intrinsically disordered premolten globule state with regions in PPII conformation--a feature that is shared by many scaffold proteins that bind multiple protein components of signalling pathways. Taken together, these results provide a deeper understanding of the physicochemical properties of CagA that underpin its complex cellular and oncogenic functions.


Subject(s)
Antigens, Bacterial/chemistry , Bacterial Proteins/chemistry , Amino Acid Sequence , Antigens, Bacterial/metabolism , Bacterial Proteins/metabolism , Crystallography, X-Ray , Models, Molecular , Molecular Sequence Data , Protein Denaturation , Protein Multimerization , Protein Stability , Protein Structure, Secondary , Protein Structure, Tertiary , Proteolysis , Repetitive Sequences, Amino Acid , Temperature
SELECTION OF CITATIONS
SEARCH DETAIL
...