Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.596
Filter
1.
Int J Biol Sci ; 20(10): 4007-4028, 2024.
Article in English | MEDLINE | ID: mdl-39113698

ABSTRACT

Cholesterol and Helicobacter pylori (H. pylori) are both risk factors for gastric cancer (GC). However, the relationship between cholesterol and H. pylori and their function in the progression of GC are controversial. In this study, we addressed that H. pylori could induce mitochondrial cholesterol accumulation and promote GC proliferation and protect GC cells against apoptosis via cholesterol. Metabolomic and transcriptomic sequencing were used to identify CYP11A1 responsible for H. pylori-induced cholesterol accumulation. In vitro and in vivo function experiments revealed that cholesterol could promote the proliferation of GC and inhibit apoptosis. Mechanically, the interaction of Cytotoxin-associated gene A (CagA) and CYP11A1 redistributed mitochondrial CYP11A1 outside the mitochondria and subsequently caused mitochondrial cholesterol accumulation. The CYP11A1-knockdown upregulated cholesterol accumulation and reproduced the effect of cholesterol on GC in a cholesterol-dependent manner. Moreover, CYP11A1-knockdown or H. pylori infection inhibited mitophagy and maintained the mitochondria homeostasis. H. pylori could contribute to the progression of GC through the CagA/CYP11A1-mitoCHO axis. This study demonstrates that H. pylori can contribute to the progression of GC via cholesterol, and eradicating H. pylori is still prognostically beneficial to GC patients.


Subject(s)
Cholesterol , Helicobacter pylori , Mitochondria , Stomach Neoplasms , Helicobacter pylori/metabolism , Stomach Neoplasms/microbiology , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Cholesterol/metabolism , Humans , Mitochondria/metabolism , Cholesterol Side-Chain Cleavage Enzyme/metabolism , Cholesterol Side-Chain Cleavage Enzyme/genetics , Helicobacter Infections/metabolism , Helicobacter Infections/microbiology , Animals , Antigens, Bacterial/metabolism , Antigens, Bacterial/genetics , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Cell Line, Tumor , Mice , Apoptosis , Male , Cell Proliferation
2.
Commun Biol ; 7(1): 984, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39138305

ABSTRACT

Heme trafficking is essential for cellular function, yet mechanisms of transport and/or heme interaction are not well defined. The System I and System II bacterial cytochrome c biogenesis pathways are developing into model systems for heme trafficking due to their functions in heme transport, heme stereospecific positioning, and mediation of heme attachment to apocytochrome c. Here we focus on the System II pathway, CcsBA, that is proposed to be a bi-functional heme transporter and holocytochrome c synthase. An extensive structure-function analysis of recombinantly expressed Helicobacter pylori and Campylobacter jejuni CcsBAs revealed key residues required for heme interaction and holocytochrome c synthase activity. Homologous residues were previously identified to be required for heme interaction in Helicobacter hepaticus CcsBA. This study provides direct, biochemical evidence that mechanisms of heme interaction are conserved, leading to the proposal that the CcsBA WWD heme-handling domain represents a novel target for therapeutics.


Subject(s)
Bacterial Proteins , Campylobacter jejuni , Helicobacter pylori , Heme , Heme/metabolism , Helicobacter pylori/enzymology , Helicobacter pylori/genetics , Helicobacter pylori/metabolism , Campylobacter jejuni/enzymology , Campylobacter jejuni/genetics , Campylobacter jejuni/metabolism , Structure-Activity Relationship , Bacterial Proteins/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Protein Binding , Models, Molecular , Lyases
3.
Infect Immun ; 92(8): e0022424, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-38975764

ABSTRACT

Colonization of the human stomach with Helicobacter pylori strains producing active forms of the secreted toxin VacA is associated with an increased risk of peptic ulcer disease and gastric cancer, compared with colonization with strains producing hypoactive forms of VacA. Previous studies have shown that active s1m1 forms of VacA cause cell vacuolation and mitochondrial dysfunction. In this study, we sought to define the cellular metabolic consequences of VacA intoxication. Untargeted metabolomic analyses revealed that several hundred metabolites were significantly altered in VacA-treated gastroduodenal cells (AGS and AZ-521) compared with control cells. Pathway analysis suggested that VacA caused alterations in taurine and hypotaurine metabolism. Treatment of cells with the purified active s1m1 form of VacA, but not hypoactive s2m1 or Δ6-27 VacA-mutant proteins (defective in membrane channel formation), caused reductions in intracellular taurine and hypotaurine concentrations. Supplementation of the tissue culture medium with taurine or hypotaurine protected AZ-521 cells against VacA-induced cell death. Untargeted global metabolomics of VacA-treated AZ-521 cells or AGS cells in the presence or absence of extracellular taurine showed that taurine was the main intracellular metabolite significantly altered by extracellular taurine supplementation. These results indicate that VacA causes alterations in cellular taurine metabolism and that repletion of taurine is sufficient to attenuate VacA-induced cell death. We discuss these results in the context of previous literature showing the important role of taurine in cell physiology and the pathophysiology or treatment of multiple pathologic conditions, including gastric ulcers, cardiovascular disease, malignancy, inflammatory diseases, and other aging-related disorders.


Subject(s)
Bacterial Proteins , Helicobacter pylori , Taurine , Taurine/metabolism , Taurine/analogs & derivatives , Humans , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Helicobacter pylori/metabolism , Helicobacter Infections/microbiology , Helicobacter Infections/metabolism , Cell Line , Host-Pathogen Interactions , Metabolomics
4.
Gut Microbes ; 16(1): 2379439, 2024.
Article in English | MEDLINE | ID: mdl-39052777

ABSTRACT

Efflux pumps play a crucial role in the development of antibiotic resistance. The aim of this study was to investigate the relationship between efflux pump gene expression and resistance gene mutations in Helicobacter pylori. Twenty-six clinical strains with varying resistance characteristics were selected for further experiment. Seven susceptible strains were induced to become resistant, and the expression of efflux pump genes and point mutations were recorded. Four susceptible strains were selected to undergo candidate mutation construction, and changes in efflux pump gene expression were detected. Efflux pump knockout strains were constructed, and their effects on preventing and reversing antibiotic resistance gene mutations were assessed. Results showed that the expression of efflux pump genes hefA and hefD was significantly higher in the multidrug-resistant group compared to other groups. During the process of antibiotic-induced resistance, efflux pump gene expression did not exhibit a steady increase or decrease. Strains with the A2143G or A2142G point mutations in 23S rRNA exhibited lower hefA gene expression. Strains with mutations at 87K/91N, 87N/91 G, 87K/91D, or 87N/91Y in gyrA and the 194insertA mutation in rdxA showed higher hefA gene expression compared to the wild-type strain. During the process of antibiotic-induced resistance, the strain with the knockout of the efflux pump gene hefA developed mutations in the 23S rRNA, gyrA, or rdxA genes later compared to the wild-type strain. Knockout of the efflux pump gene could reverse the phenotypic resistance to clarithromycin or metronidazole in some strains but had no effect on reverse resistance gene mutation. This study suggested that different resistance gene point mutations may have varying effects on efflux pump gene expression. Knockout of the efflux pump gene can delay or prevent antibiotic resistance gene mutations to some extent and can reverse phenotypic resistance to clarithromycin and metronidazole in certain strains.


Subject(s)
Anti-Bacterial Agents , Bacterial Proteins , Helicobacter Infections , Helicobacter pylori , Membrane Transport Proteins , Helicobacter pylori/genetics , Helicobacter pylori/drug effects , Helicobacter pylori/metabolism , Anti-Bacterial Agents/pharmacology , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Helicobacter Infections/microbiology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Humans , Drug Resistance, Bacterial/genetics , Point Mutation , Mutation , Microbial Sensitivity Tests , Gene Expression Regulation, Bacterial , Drug Resistance, Multiple, Bacterial/genetics , RNA, Ribosomal, 23S/genetics , DNA Gyrase/genetics , DNA Gyrase/metabolism
5.
Gut Microbes ; 16(1): 2382766, 2024.
Article in English | MEDLINE | ID: mdl-39068523

ABSTRACT

CagA, a virulence factor of Helicobacter pylori (H. pylori), is known to drive inflammation in gastric epithelial cells and is typically degraded through autophagy. However, the molecular mechanism by which CagA evades autophagy-mediated degradation remains elusive. This study found that H. pylori inhibits autophagic flux by upregulating the expression of AU-rich element RNA-binding factor 1 (AUF1). We confirmed that AUF1 does not affect autophagy initiation but instead hampers lysosomal clearance, as evidenced by treatments with 3-MA, CQ and BafA1. Upregulated AUF1 stabilizes CagA protein levels by inhibiting the autolysosomal degradation of intracellular CagA in H. pylori-infected gastric epithelial cells. Knocking down AUF1 promotes CagA degradation, an effect that can be reversed by the lysosome inhibitor BafA1 and CQ. Transcriptome analysis of AUF1-knockdown gastric epithelial cells infected with H. pylori indicated that AUF1 regulates the expression of lysosomal-associated hydrolase genes, specifically CTSD, to inhibit autolysosomal degradation. Moreover, we observed that knockdown of AUF1 enhanced the stability of CTSD mRNA and identified AUF1 binding to the 3'UTR region of CTSD mRNA. AUF1-mediated downregulation of CTSD expression contributes to CagA stability, and AUF1 overexpression leads to an increase in CagA levels in exosomes, thus promoting extracellular inflammation. In clinical gastric mucosa, the expression of AUF1 and its cytoplasmic translocation are associated with H. pylori-associated gastritis, with CagA being necessary for the translocation of AUF1 into the cytoplasm. Our findings suggest that AUF1 is a novel host-positive regulator of CagA, and dysregulation of AUF1 expression increases the risk of H. pylori-associated gastritis.


Subject(s)
Antigens, Bacterial , Autophagy , Bacterial Proteins , Epithelial Cells , Gastric Mucosa , Helicobacter Infections , Helicobacter pylori , Heterogeneous Nuclear Ribonucleoprotein D0 , Heterogeneous-Nuclear Ribonucleoprotein D , Lysosomes , Antigens, Bacterial/metabolism , Antigens, Bacterial/genetics , Heterogeneous Nuclear Ribonucleoprotein D0/metabolism , Helicobacter pylori/metabolism , Helicobacter pylori/genetics , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Humans , Lysosomes/metabolism , Lysosomes/microbiology , Helicobacter Infections/microbiology , Helicobacter Infections/metabolism , Helicobacter Infections/pathology , Heterogeneous-Nuclear Ribonucleoprotein D/metabolism , Heterogeneous-Nuclear Ribonucleoprotein D/genetics , Epithelial Cells/microbiology , Epithelial Cells/metabolism , Gastric Mucosa/microbiology , Gastric Mucosa/metabolism , Inflammation/metabolism , Inflammation/microbiology , Virulence Factors/metabolism , Virulence Factors/genetics , Cell Line
6.
Nucleic Acids Res ; 52(12): 7321-7336, 2024 Jul 08.
Article in English | MEDLINE | ID: mdl-38842933

ABSTRACT

The ParABS system, composed of ParA (an ATPase), ParB (a DNA binding protein), and parS (a centromere-like DNA), regulates bacterial chromosome partition. The ParB-parS partition complex interacts with the nucleoid-bound ParA to form the nucleoid-adaptor complex (NAC). In Helicobacter pylori, ParA and ParB homologs are encoded as HpSoj and HpSpo0J (HpParA and HpParB), respectively. We determined the crystal structures of the ATP hydrolysis deficient mutant, HpParAD41A, and the HpParAD41A-DNA complex. We assayed the CTPase activity of HpParB and identified two potential DNA binding modes of HpParB regulated by CTP, one is the specific DNA binding by the DNA binding domain and the other is the non-specific DNA binding through the C-terminal domain under the regulation of CTP. We observed an interaction between HpParAD41A and the N-terminus fragment of HpParB (residue 1-10, HpParBN10) and determined the crystal structure of the ternary complex, HpParAD41A-DNA-HpParBN10 complex which mimics the NAC formation. HpParBN10 binds near the HpParAD41A dimer interface and is clamped by flexible loops, L23 and L34, through a specific cation-π interaction between Arg9 of HpParBN10 and Phe52 of HpParAD41A. We propose a molecular mechanism model of the ParABS system providing insight into chromosome partition in bacteria.


Subject(s)
Bacterial Proteins , Chromosomes, Bacterial , DNA-Binding Proteins , Helicobacter pylori , Bacterial Proteins/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Helicobacter pylori/genetics , Helicobacter pylori/metabolism , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Chromosomes, Bacterial/metabolism , Chromosomes, Bacterial/chemistry , Chromosomes, Bacterial/genetics , Models, Molecular , Crystallography, X-Ray , Protein Binding , DNA, Bacterial/metabolism , DNA, Bacterial/chemistry , DNA, Bacterial/genetics , Adenosine Triphosphatases/metabolism , Adenosine Triphosphatases/chemistry , Adenosine Triphosphatases/genetics , Chromosome Segregation , Adenosine Triphosphate/metabolism , Binding Sites
7.
Adv Sci (Weinh) ; 11(30): e2309712, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38887155

ABSTRACT

Helicobacter pylori (H. pylori) infection is the primary risk factor for the pathogenesis of gastric cancer (GC). N6-methyladenosine (m6A) plays pivotal roles in mRNA metabolism and hnRNPA2B1 as an m6A reader is shown to exert m6A-dependent mRNA stabilization in cancer. This study aims to explore the role of hnRNPA2B1 in H. pylori-associated GC and its novel molecular mechanism. Multiple datasets and tissue microarray are utilized for assessing hnRNPA2B1 expression in response to H. pylori infection and its clinical prognosis in patients with GC. The roles of hnRNPA2B1 are investigated through a variety of techniques including glucose metabolism analysis, m6A-epitranscriptomic microarray, Ribo-seq, polysome profiling, RIP-seq. In addition, hnRNPA2B1 interaction with poly(A) binding protein cytoplasmic 1 (PABPC1) is validated using mass spectrometry and co-IP. These results show that hnRNPA2B1 is upregulated in GC and correlated with poor prognosis. H. pylori infection induces hnRNPA2B1 upregulation through recruiting NF-κB to its promoter. Intriguingly, cytoplasm-anchored hnRNPA2B1 coordinated PABPC1 to stabilize its relationship with cap-binding eIF4F complex, which facilitated the translation of CIP2A, DLAT and GPX1 independent of m6A modification. In summary, hnRNPA2B1 facilitates the non-m6A translation of epigenetic mRNAs in GC progression by interacting with PABPC1-eIF4F complex and predicts poor prognosis for patients with GC.


Subject(s)
Disease Progression , Helicobacter Infections , Helicobacter pylori , Heterogeneous-Nuclear Ribonucleoprotein Group A-B , Poly(A)-Binding Protein I , Stomach Neoplasms , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Stomach Neoplasms/microbiology , Stomach Neoplasms/pathology , Humans , Helicobacter pylori/genetics , Helicobacter pylori/metabolism , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/metabolism , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/genetics , Helicobacter Infections/genetics , Helicobacter Infections/metabolism , Poly(A)-Binding Protein I/metabolism , Poly(A)-Binding Protein I/genetics , Mice , Animals , Prognosis , Disease Models, Animal , Male , Adenosine/analogs & derivatives , Adenosine/metabolism , Adenosine/genetics
8.
BMC Genomics ; 25(1): 466, 2024 May 13.
Article in English | MEDLINE | ID: mdl-38741045

ABSTRACT

BACKGROUND: Protein-protein interactions (PPIs) hold significant importance in biology, with precise PPI prediction as a pivotal factor in comprehending cellular processes and facilitating drug design. However, experimental determination of PPIs is laborious, time-consuming, and often constrained by technical limitations. METHODS: We introduce a new node representation method based on initial information fusion, called FFANE, which amalgamates PPI networks and protein sequence data to enhance the precision of PPIs' prediction. A Gaussian kernel similarity matrix is initially established by leveraging protein structural resemblances. Concurrently, protein sequence similarities are gauged using the Levenshtein distance, enabling the capture of diverse protein attributes. Subsequently, to construct an initial information matrix, these two feature matrices are merged by employing weighted fusion to achieve an organic amalgamation of structural and sequence details. To gain a more profound understanding of the amalgamated features, a Stacked Autoencoder (SAE) is employed for encoding learning, thereby yielding more representative feature representations. Ultimately, classification models are trained to predict PPIs by using the well-learned fusion feature. RESULTS: When employing 5-fold cross-validation experiments on SVM, our proposed method achieved average accuracies of 94.28%, 97.69%, and 84.05% in terms of Saccharomyces cerevisiae, Homo sapiens, and Helicobacter pylori datasets, respectively. CONCLUSION: Experimental findings across various authentic datasets validate the efficacy and superiority of this fusion feature representation approach, underscoring its potential value in bioinformatics.


Subject(s)
Computational Biology , Protein Interaction Mapping , Protein Interaction Mapping/methods , Computational Biology/methods , Algorithms , Helicobacter pylori/metabolism , Helicobacter pylori/genetics , Support Vector Machine , Proteins/metabolism , Proteins/chemistry , Humans , Protein Interaction Maps , Databases, Protein
9.
J Cell Mol Med ; 28(9): e18358, 2024 May.
Article in English | MEDLINE | ID: mdl-38693868

ABSTRACT

Gastric cancer is considered a class 1 carcinogen that is closely linked to infection with Helicobacter pylori (H. pylori), which affects over 1 million people each year. However, the major challenge to fight against H. pylori and its associated gastric cancer due to drug resistance. This research gap had led our research team to investigate a potential drug candidate targeting the Helicobacter pylori-carcinogenic TNF-alpha-inducing protein. In this study, a total of 45 daidzein derivatives were investigated and the best 10 molecules were comprehensively investigated using in silico approaches for drug development, namely pass prediction, quantum calculations, molecular docking, molecular dynamics simulations, Lipinski rule evaluation, and prediction of pharmacokinetics. The molecular docking study was performed to evaluate the binding affinity between the target protein and the ligands. In addition, the stability of ligand-protein complexes was investigated by molecular dynamics simulations. Various parameters were analysed, including root-mean-square deviation (RMSD), root-mean-square fluctuation (RMSF), radius of gyration (Rg), hydrogen bond analysis, principal component analysis (PCA) and dynamic cross-correlation matrix (DCCM). The results has confirmed that the ligand-protein complex CID: 129661094 (07) and 129664277 (08) formed stable interactions with the target protein. It was also found that CID: 129661094 (07) has greater hydrogen bond occupancy and stability, while the ligand-protein complex CID 129664277 (08) has greater conformational flexibility. Principal component analysis revealed that the ligand-protein complex CID: 129661094 (07) is more compact and stable. Hydrogen bond analysis revealed favourable interactions with the reported amino acid residues. Overall, this study suggests that daidzein derivatives in particular show promise as potential inhibitors of H. pylori.


Subject(s)
Helicobacter pylori , Isoflavones , Molecular Docking Simulation , Molecular Dynamics Simulation , Helicobacter pylori/drug effects , Helicobacter pylori/metabolism , Isoflavones/pharmacology , Isoflavones/chemistry , Isoflavones/metabolism , Humans , Hydrogen Bonding , Ligands , Protein Binding , Principal Component Analysis , Helicobacter Infections/microbiology , Helicobacter Infections/drug therapy , Bacterial Proteins/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/antagonists & inhibitors , Stomach Neoplasms/microbiology , Stomach Neoplasms/drug therapy
10.
J Agric Food Chem ; 72(18): 10469-10476, 2024 May 08.
Article in English | MEDLINE | ID: mdl-38659344

ABSTRACT

Lacto-N-difucohexaose II (LNDFH II) is a typical fucosylated human milk oligosaccharide and can be enzymatically produced from lacto-N-tetraose (LNT) by a specific α1,3/4-fucosyltransferase from Helicobacter pylori DMS 6709, referred to as FucT14. Previously, we constructed an engineered Escherichia coli BL21(DE3) with a single plasmid for highly efficient biosynthesis of LNT. In this study, two additional plasmids harboring the de novo GDP-L-fucose pathway module and FucT14, respectively, were further introduced to construct the strain for successful biosynthesis of LNDFH II. FucT14 was actively expressed, and the engineered strain produced LNDFH II as the major product, lacto-N-fucopentaose (LNFP) V as the minor product, and a trace amount of LNFP II and 3-fucosyllactose as very minor products. Additional expression of the α1,3-fucosyltransferase FutM1 from a Bacteroidaceae bacterium from the gut metagenome could obviously enhance the LNDFH II biosynthesis. After optimization of induction conditions, the maximum titer reached 3.011 g/L by shake-flask cultivation. During the fed-batch cultivation, LNDFH II was highly efficiently produced with the highest titer of 18.062 g/L and the productivity yield of 0.301 g/L·h.


Subject(s)
Bacterial Proteins , Escherichia coli , Fucosyltransferases , Guanosine Diphosphate Fucose , Metabolic Engineering , Humans , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Biosynthetic Pathways , Escherichia coli/genetics , Escherichia coli/metabolism , Fucosyltransferases/genetics , Fucosyltransferases/metabolism , Guanosine Diphosphate Fucose/metabolism , Helicobacter pylori/genetics , Helicobacter pylori/metabolism , Helicobacter pylori/enzymology , Oligosaccharides/metabolism , Oligosaccharides/biosynthesis
11.
J Mol Biol ; 436(10): 168573, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38626867

ABSTRACT

Iron homeostasis is a critical process for living organisms because this metal is an essential co-factor for fundamental biochemical activities, like energy production and detoxification, albeit its excess quickly leads to cell intoxication. The protein Fur (ferric uptake regulator) controls iron homeostasis in bacteria by switching from its apo- to holo-form as a function of the cytoplasmic level of ferrous ions, thereby modulating gene expression. The Helicobacter pylori HpFur protein has the rare ability to operate as a transcriptional commutator; apo- and holo-HpFur function as two different repressors with distinct DNA binding recognition properties for specific sets of target genes. Although the regulation of apo- and holo-HpFur in this bacterium has been extensively investigated, we propose a genome-wide redefinition of holo-HpFur direct regulon in H. pylori by integration of RNA-seq and ChIP-seq data, and a large extension of the apo-HpFur direct regulon. We show that in response to iron availability, new coding sequences, non-coding RNAs, toxin-antitoxin systems, and transcripts within open reading frames are directly regulated by apo- or holo-HpFur. These new targets and the more thorough validation and deeper characterization of those already known provide a complete and updated picture of the direct regulons of this two-faced transcriptional regulator.


Subject(s)
Bacterial Proteins , Gene Expression Regulation, Bacterial , Helicobacter pylori , Iron , Regulon , Repressor Proteins , Helicobacter pylori/genetics , Helicobacter pylori/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Iron/metabolism , Regulon/genetics , Repressor Proteins/genetics , Repressor Proteins/metabolism
12.
J Biomed Sci ; 31(1): 44, 2024 Apr 29.
Article in English | MEDLINE | ID: mdl-38685037

ABSTRACT

BACKGROUND: Helicobacter pylori, the main cause of various gastric diseases, infects approximately half of the human population. This pathogen is auxotrophic for cholesterol which it converts to various cholesteryl α-glucoside derivatives, including cholesteryl 6'-acyl α-glucoside (CAG). Since the related biosynthetic enzymes can be translocated to the host cells, the acyl chain of CAG likely comes from its precursor phosphatidylethanolamine (PE) in the host membranes. This work aims at examining how the acyl chain of CAG and PE inhibits the membrane functions, especially bacterial adhesion. METHODS: Eleven CAGs that differ in acyl chains were used to study the membrane properties of human gastric adenocarcinoma cells (AGS cells), including lipid rafts clustering (monitored by immunofluorescence with confocal microscopy) and lateral membrane fluidity (by the fluorescence recovery after photobleaching). Cell-based and mouse models were employed to study the degree of bacterial adhesion, the analyses of which were conducted by using flow cytometry and immunofluorescence staining, respectively. The lipidomes of H. pylori, AGS cells and H. pylori-AGS co-cultures were analyzed by Ultraperformance Liquid Chromatography-Tandem Mass Spectroscopy (UPLC-MS/MS) to examine the effect of PE(10:0)2, PE(18:0)2, PE(18:3)2, or PE(22:6)2 treatments. RESULTS: CAG10:0, CAG18:3 and CAG22:6 were found to cause the most adverse effect on the bacterial adhesion. Further LC-MS analysis indicated that the treatment of PE(10:0)2 resulted in dual effects to inhibit the bacterial adhesion, including the generation of CAG10:0 and significant changes in the membrane compositions. The initial (1 h) lipidome changes involved in the incorporation of 10:0 acyl chains into dihydro- and phytosphingosine derivatives and ceramides. In contrast, after 16 h, glycerophospholipids displayed obvious increase in their very long chain fatty acids, monounsaturated and polyunsaturated fatty acids that are considered to enhance membrane fluidity. CONCLUSIONS: The PE(10:0)2 treatment significantly reduced bacterial adhesion in both AGS cells and mouse models. Our approach of membrane remodeling has thus shown great promise as a new anti-H. pylori therapy.


Subject(s)
Cholesterol/analogs & derivatives , Helicobacter pylori , Helicobacter pylori/metabolism , Helicobacter pylori/physiology , Mice , Animals , Humans , Membrane Lipids/metabolism , Cell Line, Tumor , Helicobacter Infections/drug therapy , Helicobacter Infections/microbiology , Helicobacter Infections/metabolism , Cholesterol Esters/metabolism
13.
J Biophotonics ; 17(6): e202400074, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38566476

ABSTRACT

Helicobacter pylori (H. pylori) is a type of bacteria that infects the stomach. The detection of H. pylori is an essential part of current clinical practices because this disease can cause peptic ulcers, chronic inflammation of the stomach lining but also stomach cancer. Helicobacter pylori has a naturally occurring enzyme that hydrolyzes urea into ammonium carbonate called urease. Many methods exist for the detection of H. pylori infection, but an innovative approach is to detect the ammonia in the breath (ABT, Ammonia Breath Test). In this research study, using photoacoustic spectroscopy method, the ammonia concentration in the breathing zone of people with H. pylori were measured and were compared with ammonia concentration from the respiration of healthy people. From the ABT determinations of this study, the ammonia was established to be increased with 498 ppb at people with H. pylori when we compare with ABT of healthy people.


Subject(s)
Ammonia , Breath Tests , Helicobacter Infections , Helicobacter pylori , Photoacoustic Techniques , Humans , Helicobacter pylori/metabolism , Breath Tests/methods , Ammonia/metabolism , Helicobacter Infections/microbiology , Male , Adult , Female
14.
Sci Rep ; 14(1): 7683, 2024 04 01.
Article in English | MEDLINE | ID: mdl-38561502

ABSTRACT

Helicobacter pylori (H. pylori), known for causing gastric inflammation, gastritis and gastric cancer, prompted our study to investigate the differential expression of cytokines in gastric tissues, which is crucial for understanding H. pylori infection and its potential progression to gastric cancer. Focusing on Il-1ß, IL-6, IL-8, IL-12, IL-18, and TNF-α, we analysed gene and protein levels to differentiate between H. pylori-infected and non-infected gastritis. We utilised real-time quantitative polymerase chain reaction (RT-qPCR) for gene quantification, immunohistochemical staining, and ELISA for protein measurement. Gastric samples from patients with gastritis were divided into three groups: (1) non-gastritis (N-group) group, (2) gastritis without H. pylori infection (G-group), and (3) gastritis with H. pylori infection (GH-group), each consisting of 8 samples. Our findings revealed a statistically significant variation in cytokine expression. Generally, cytokine levels were higher in gastritis, but in H. pylori-infected gastritis, IL-1ß, IL-6, and IL-8 levels were lower compared to H. pylori-independent gastritis, while IL-12, IL-18, and TNF-α levels were higher. This distinct cytokine expression pattern in H. pylori-infected gastritis underscores a unique inflammatory response, providing deeper insights into its pathogenesis.


Subject(s)
Gastritis , Helicobacter Infections , Helicobacter pylori , Helicobacter , Stomach Neoplasms , Humans , Cytokines/metabolism , Helicobacter pylori/metabolism , Interleukin-18/genetics , Interleukin-18/metabolism , Helicobacter/metabolism , Interleukin-8/metabolism , Tumor Necrosis Factor-alpha/metabolism , Interleukin-6/metabolism , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Gastritis/pathology , Interleukin-12/metabolism , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Helicobacter Infections/genetics , Helicobacter Infections/metabolism , Gastric Mucosa/metabolism
15.
Nucleic Acids Res ; 52(10): 5572-5595, 2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38499492

ABSTRACT

Adaptation to variations in pH is crucial for the ability of Helicobacter pylori to persist in the human stomach. The acid responsive two-component system ArsRS, constitutes the global regulon that responds to acidic conditions, but molecular details of how transcription is affected by the ArsR response regulator remains poorly understood. Using a combination of DNA-binding studies, in vitro transcription assays, and H. pylori mutants, we demonstrate that phosphorylated ArsR (ArsR-P) forms an active protein complex that binds DNA with high specificity in order to affect transcription. Our data showed that DNA topology is key for DNA binding. We found that AT-rich DNA sequences direct ArsR-P to specific sites and that DNA-bending proteins are important for the effect of ArsR-P on transcription regulation. The repression of sabA transcription is mediated by ArsR-P with the support of Hup and is affected by simple sequence repeats located upstream of the sabA promoter. Here stochastic events clearly contribute to the fine-tuning of pH-dependent gene regulation. Our results reveal important molecular aspects for how ArsR-P acts to repress transcription in response to acidic conditions. Such transcriptional control likely mediates shifts in bacterial positioning in the gastric mucus layer.


Subject(s)
Adhesins, Bacterial , Bacterial Proteins , Gene Expression Regulation, Bacterial , Helicobacter pylori , Adhesins, Bacterial/metabolism , Adhesins, Bacterial/genetics , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , DNA, Bacterial/metabolism , DNA, Bacterial/genetics , Helicobacter pylori/genetics , Helicobacter pylori/metabolism , Hydrogen-Ion Concentration , Phosphorylation , Promoter Regions, Genetic , Protein Binding , Transcription, Genetic/genetics , Mutation
16.
Microbiol Spectr ; 12(5): e0047024, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38501821

ABSTRACT

Bacterial lipoproteins are post-translationally modified by the addition of acyl chains that anchor the protein to bacterial membranes. This modification includes two ester-linked and one amide-linked acyl chain on lipoproteins from Gram-negative bacteria. Helicobacter pylori lipoproteins have important functions in pathogenesis (including delivering the CagA oncoprotein to mammalian cells) and are recognized by host innate and adaptive immune systems. The number and variety of acyl chains on lipoproteins impact the innate immune response through Toll-like receptor 2. The acyl chains added to lipoproteins are derived from membrane phospholipids. H. pylori membrane phospholipids have previously been shown to consist primarily of C14:0 and C19:0 cyclopropane-containing acyl chains. However, the acyl composition of H. pylori lipoproteins has not been determined. In this study, we characterized the acyl composition of two representative H. pylori lipoproteins, Lpp20 and CagT. Fatty acid methyl esters were prepared from both purified lipoproteins and analyzed by gas chromatography-mass spectrometry. For comparison, we also analyzed H. pylori phospholipids. Consistent with previous studies, we observed that the H. pylori phospholipids contain primarily C14:0 and C19:0 cyclopropane-containing fatty acids. In contrast, both the ester-linked and amide-linked fatty acids found in H. pylori lipoproteins were observed to be almost exclusively C16:0 and C18:0. A discrepancy between the acyl composition of membrane phospholipids and lipoproteins as reported here for H. pylori has been previously reported in other bacteria including Borrelia and Brucella. We discuss possible mechanisms.IMPORTANCEColonization of the stomach by Helicobacter pylori is an important risk factor in the development of gastric cancer, the third leading cause of cancer-related death worldwide. H. pylori persists in the stomach despite an immune response against the bacteria. Recognition of lipoproteins by TLR2 contributes to the innate immune response to H. pylori. However, the role of H. pylori lipoproteins in bacterial persistence is poorly understood. As the host response to lipoproteins depends on the acyl chain content, defining the acyl composition of H. pylori lipoproteins is an important step in characterizing how lipoproteins contribute to persistence.


Subject(s)
Bacterial Proteins , Fatty Acids , Helicobacter pylori , Lipoproteins , Helicobacter pylori/immunology , Helicobacter pylori/metabolism , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Fatty Acids/metabolism , Fatty Acids/chemistry , Lipoproteins/metabolism , Lipoproteins/chemistry , Phospholipids/metabolism , Phospholipids/chemistry , Humans , Helicobacter Infections/microbiology , Immunity, Innate , Gas Chromatography-Mass Spectrometry
17.
Helicobacter ; 29(2): e13069, 2024.
Article in English | MEDLINE | ID: mdl-38516860

ABSTRACT

Helicobacter pylori (H. pylori) seems to play causative roles in gastric cancers. H. pylori has also been detected in established gastric cancers. How the presence of H. pylori modulates immune response to the cancer is unclear. The cytotoxicity of natural killer (NK) cells, toward infected or malignant cells, is controlled by the repertoire of activating and inhibitory receptors expressed on their surface. Here, we studied H. pylori-induced changes in the expression of ligands, of activating and inhibitory receptors of NK cells, in the gastric adenocarcinoma AGS cells, and their impacts on NK cell responses. AGS cells lacked or had low surface expression of the class I major histocompatibility complex (MHC-I) molecules HLA-E and HLA-C-ligands of the major NK cell inhibitory receptors NKG2A and killer-cell Ig-like receptor (KIR), respectively. However, AGS cells had high surface expression of ligands of activating receptors DNAM-1 and CD2, and of the adhesion molecules LFA-1. Consistently, AGS cells were sensitive to killing by NK cells despite the expression of inhibitory KIR on NK cells. Furthermore, H. pylori enhanced HLA-C surface expression on AGS cells. H. pylori infection enhanced HLA-C protein synthesis, which could explain H. pylori-induced HLA-C surface expression. H. pylori infection enhanced HLA-C surface expression also in the hepatoma Huh7 and HepG2 cells. Furthermore, H. pylori-induced HLA-C surface expression on AGS cells promoted inhibition of NK cells by KIR, and thereby protected AGS cells from NK cell cytotoxicity. These results suggest that H. pylori enhances HLA-C expression in host cells and protects them from the cytotoxic attack of NK cells expressing HLA-C-specific inhibitory receptors.


Subject(s)
Adenocarcinoma , Helicobacter Infections , Helicobacter pylori , Stomach Neoplasms , Humans , Adenocarcinoma/genetics , Adenocarcinoma/microbiology , Adenocarcinoma/pathology , Helicobacter Infections/genetics , Helicobacter Infections/pathology , Helicobacter pylori/metabolism , Histocompatibility Antigens Class I/metabolism , HLA-C Antigens/genetics , HLA-C Antigens/metabolism , Killer Cells, Natural/metabolism , Killer Cells, Natural/pathology , Receptors, Immunologic/metabolism , Receptors, KIR/metabolism , Stomach Neoplasms/genetics , Stomach Neoplasms/microbiology , Stomach Neoplasms/pathology
18.
J Investig Med High Impact Case Rep ; 12: 23247096241240176, 2024.
Article in English | MEDLINE | ID: mdl-38504422

ABSTRACT

Gastric cancer ranks as the fifth leading cause of global cancer incidences, exhibiting varied prevalence influenced by geographical, ethnic, and lifestyle factors, as well as Helicobacter pylori infection. The ATM gene on chromosome 11q22 is vital for genomic stability as an initiator of the DNA damage response, and mutations in this gene have been associated with various cancers. Poly ADP-ribose polymerase (PARP) inhibitors, such as olaparib, have shown efficacy in cancers with homologous recombination repair deficiencies, notably in those with ATM mutations. Here, we present a case of a 66-year-old patient with germline ATM-mutated metastatic gastric cancer with very high CA 19-9 (48 000 units/mL) who demonstrated an exceptional response to the addition of olaparib to chemo-immunotherapy and subsequent olaparib maintenance monotherapy for 12 months. CA 19-9 was maintained at low level for 18 months. Despite the failure of a phase II clinical trial on olaparib in gastric cancer (NCT01063517) to meet its primary endpoint, intriguing findings emerged in the subset of ATM-mutated patients, who exhibited notable improvements in overall survival. Our case underscores the potential clinical utility of olaparib in germline ATM-mutated gastric cancer and emphasizes the need for further exploration through larger clinical trials. Ongoing research and clinical trials are essential for optimizing the use of PARP inhibitors, identifying biomarkers, and advancing personalized treatment strategies for gastric cancer.


Subject(s)
Helicobacter Infections , Helicobacter pylori , Phthalazines , Piperazines , Stomach Neoplasms , Humans , Aged , Stomach Neoplasms/drug therapy , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Helicobacter pylori/metabolism , Germ Cells/metabolism , Germ Cells/pathology , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism
19.
Neoplasia ; 50: 100981, 2024 04.
Article in English | MEDLINE | ID: mdl-38422751

ABSTRACT

PURPOSE: Helicobacter pylori (H. pylori) is a significant risk factor for development of gastric cancer (GC), one of the deadliest malignancies in the world. However, the mechanism by which H. pylori induces gastric oncogenesis remains unclear. Here, we investigated the function of IL-6 in gastric oncogenesis and macrophage-epithelial cell interactions. METHODS: We analyzed publicly available datasets to investigate the expression of IL-6 and infiltration of M2 macrophages in GC tissues, and determine the inter-cellular communication in the context of IL-6. Human gastric epithelial and macrophage cell lines (GES-1 and THP-1-derived macrophages, respectively) were used in mono- and co-culture experiments to investigate autocrine-and paracrine induction of IL-6 expression in response to H. pylori or IL-6 stimulation. RESULTS: We found that IL-6 is highly expressed in GC and modulates survival. M2 macrophage infiltration is predominant in GC and drives an IL-6 mediated communication with gastric epithelium cells. In vitro, IL-6 triggers its own expression in GES-1 and THP-1-derived macrophages cells. In addition, these cell lines are able to upregulate each other's IL-6 levels in an autocrine fashion, which is enhanced by H. pylori stimulation. CONCLUSION: This study indicates that IL-6 in the tumor microenvironment is essential for intercellular communication. We show that H. pylori enhances an IL-6-driven autocrine and paracrine positive feedback loop between macrophages and gastric epithelial cells, which may contribute to gastric carcinogenesis.


Subject(s)
Helicobacter Infections , Helicobacter pylori , Stomach Neoplasms , Humans , Helicobacter pylori/metabolism , Interleukin-6/metabolism , Epithelial Cells/metabolism , Gastric Mucosa/metabolism , Stomach Neoplasms/pathology , Macrophages/pathology , Carcinogenesis/pathology , Helicobacter Infections/complications , Helicobacter Infections/metabolism , Helicobacter Infections/pathology , Tumor Microenvironment
20.
J Transl Med ; 22(1): 148, 2024 02 13.
Article in English | MEDLINE | ID: mdl-38351014

ABSTRACT

Helicobacter pylori (H. pylori) is a major risk factor of gastric cancer (GC). The SUMO-activating enzyme SAE1(SUMO-activating enzyme subunit 1), which is indispensable for protein SUMOylation, involves in human tumorigenesis. In this study, we used the TIMER and TCGA database to explore the SAE1 expression in GC and normal tissues and Kaplan-Meier Plotter platform for survival analysis of GC patients. GC tissue microarray and gastric samples from patients who underwent endoscopic treatment were employed to detect the SAE1expression. Our results showed that SAE1 was overexpressed in GC tissues and higher SAE1 expression was associated with worse clinical characteristics of GC patients. Cell and animal models showed that H. pylori infection upregulated SAE1, SUMO1, and SUMO2/3 protein expression. Functional assays suggested that suppression of SAE1 attenuated epithelial-mesenchymal transition (EMT) biomarkers and cell proliferation abilities induced by H. pylori. Cell and animal models of ROS inhibition in H. pylori showed that ROS could mediate the H. pylori-induced upregulation of SAE1, SUMO1, and SUMO2/3 protein. RNA sequencing was performed and suggested that knockdown of SAE1 could exert an impact on IGF-1 expression. General, increased SUMOylation modification is involved in H. pylori-induced GC.


Subject(s)
Helicobacter Infections , Helicobacter pylori , Stomach Neoplasms , Animals , Humans , Up-Regulation/genetics , Stomach Neoplasms/pathology , Helicobacter pylori/genetics , Helicobacter pylori/metabolism , Reactive Oxygen Species/metabolism , Cell Transformation, Neoplastic , Helicobacter Infections/complications , Helicobacter Infections/genetics , Helicobacter Infections/metabolism , Ubiquitin-Activating Enzymes/genetics , Ubiquitin-Activating Enzymes/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL