Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Biochim Biophys Acta Gen Subj ; 1864(1): 129423, 2020 01.
Article in English | MEDLINE | ID: mdl-31476357

ABSTRACT

BACKGROUND: We previously reported the identification of the aapA1/IsoA1 locus as part of a new family of toxin-antitoxin (TA) systems in the human pathogen Helicobacter pylori. AapA1 belongs to type I TA bacterial toxins, and both its mechanism of action towards the membrane and toxicity features are still unclear. METHODS: The biochemical characterization of the AapA1 toxic peptide was carried out using plasmid-borne expression and mutational approaches to follow its toxicity and localization. Biophysical properties of the AapA1 interaction with lipid membranes were studied by solution and solid-state NMR spectroscopy, plasmon waveguide resonance (PWR) and molecular modeling. RESULTS: We show that despite a low hydrophobic index, this toxin has a nanomolar affinity to the prokaryotic membrane. NMR spectroscopy reveals that the AapA1 toxin is structurally organized into three distinct domains: a positively charged disordered N-terminal domain (D), a single α-helix (H), and a basic C-terminal domain (R). The R domain interacts and destabilizes the membrane, while the H domain adopts a transmembrane conformation. These results were confirmed by alanine scanning of the minimal sequence required for toxicity. CONCLUSION: Our results have shown that specific amino acid residues along the H domain, as well as the R domain, are essential for the toxicity of the AapA1 toxin. GENERAL SIGNIFICANCE: Untangling and understanding the mechanism of action of small membrane-targeting toxins are difficult, but nevertheless contributes to a promising search and development of new antimicrobial drugs.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Toxins/chemistry , Cell Membrane/chemistry , Helicobacter Infections/genetics , Helicobacter pylori/chemistry , Alanine/chemistry , Alanine/genetics , Bacterial Proteins/genetics , Bacterial Proteins/ultrastructure , Bacterial Toxins/genetics , Cell Membrane/genetics , Cell Membrane/ultrastructure , Helicobacter Infections/microbiology , Helicobacter pylori/pathogenicity , Humans , Magnetic Resonance Spectroscopy , Protein Conformation, alpha-Helical/genetics , Protein Domains/genetics , Surface Plasmon Resonance , Toxin-Antitoxin Systems/genetics
2.
Elife ; 82019 08 14.
Article in English | MEDLINE | ID: mdl-31411564

ABSTRACT

Post-transcriptional regulation plays important roles to fine-tune gene expression in bacteria. In particular, regulation of type I toxin-antitoxin (TA) systems is achieved through sophisticated mechanisms involving toxin mRNA folding. Here, we set up a genetic approach to decipher the molecular underpinnings behind the regulation of a type I TA in Helicobacter pylori. We used the lethality induced by chromosomal inactivation of the antitoxin to select mutations that suppress toxicity. We found that single point mutations are sufficient to allow cell survival. Mutations located either in the 5' untranslated region or within the open reading frame of the toxin hamper its translation by stabilizing stem-loop structures that sequester the Shine-Dalgarno sequence. We propose that these short hairpins correspond to metastable structures that are transiently formed during transcription to avoid premature toxin expression. This work uncovers the co-transcriptional inhibition of translation as an additional layer of TA regulation in bacteria.


Subject(s)
Bacterial Toxins/genetics , Helicobacter pylori/metabolism , Nucleic Acid Conformation , RNA Folding , RNA, Messenger/chemistry , Toxin-Antitoxin Systems , Bacterial Toxins/biosynthesis , Helicobacter pylori/genetics , Helicobacter pylori/growth & development , Microbial Viability , Point Mutation , Protein Biosynthesis , RNA, Messenger/genetics , Selection, Genetic
3.
Nucleic Acids Res ; 47(11): 5906-5921, 2019 06 20.
Article in English | MEDLINE | ID: mdl-31006803

ABSTRACT

In most bacteria, ribosomal RNA is transcribed as a single polycistronic precursor that is first processed by RNase III. This double-stranded specific RNase cleaves two large stems flanking the 23S and 16S rRNA mature sequences, liberating three 16S, 23S and 5S rRNA precursors, which are further processed by other ribonucleases. Here, we investigate the rRNA maturation pathway of the human gastric pathogen Helicobacter pylori. This bacterium has an unusual arrangement of its rRNA genes, the 16S rRNA gene being separated from a 23S-5S rRNA cluster. We show that RNase III also initiates processing in this organism, by cleaving two typical stem structures encompassing 16S and 23S rRNAs and an atypical stem-loop located upstream of the 5S rRNA. Deletion of RNase III leads to the accumulation of a large 23S-5S precursor that is found in polysomes, suggesting that it can function in translation. Finally, we characterize a cis-encoded antisense RNA overlapping the leader of the 23S-5S rRNA precursor. We present evidence that this antisense RNA interacts with this precursor, forming an intermolecular complex that is cleaved by RNase III. This pairing induces additional specific cleavages of the rRNA precursor coupled with a rapid degradation of the antisense RNA.


Subject(s)
Helicobacter pylori/genetics , RNA Precursors/metabolism , RNA, Ribosomal, 16S/genetics , RNA, Ribosomal, 23S/metabolism , RNA, Ribosomal, 5S/metabolism , Ribonuclease III/metabolism , Escherichia coli/metabolism , Humans , Nucleic Acid Conformation , Oligonucleotides/genetics , Polyribosomes/metabolism , RNA, Antisense , RNA, Bacterial/metabolism , RNA, Ribosomal/metabolism , Stomach Diseases/microbiology
4.
Methods Enzymol ; 612: 67-100, 2018.
Article in English | MEDLINE | ID: mdl-30502958

ABSTRACT

As the number of bacterial genomes and transcriptomes increases, so does the number of newly identified toxin-antitoxin (TA) systems. However, their functional characterization remains challenging, often requiring the use of overexpression vectors that can lead to misinterpretations of in vivo results. To fill this gap, we developed a systematic approach called FASTBAC-Seq (Functional AnalysiS of Toxin-Antitoxin Systems in BACteria by Deep Sequencing). Combining life/death phenotypic selection with next-generation sequencing, FASTBAC-Seq allows the rapid identification of loss-of-function (toxicity) mutations in toxin-encoding genes belonging to TA loci with nucleotide resolution. Here, we present the setup used on the first-time application of FASBACT-Seq to characterize a member of the aapA/IsoA family of type I TA systems hosted on the chromosome of the major human gastric pathogen Helicobacter pylori. We propose FASBACT-Seq as a powerful tool for the functional characterization of TA systems that can in addition uncover key elements for the understanding of gene expression regulation in bacteria.


Subject(s)
Bacteria/genetics , Bacteria/metabolism , High-Throughput Nucleotide Sequencing/methods , Toxin-Antitoxin Systems/physiology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression Regulation, Bacterial/genetics , Gene Expression Regulation, Bacterial/physiology , Helicobacter pylori/genetics , Helicobacter pylori/metabolism , Toxin-Antitoxin Systems/genetics
5.
Nucleic Acids Res ; 45(8): 4782-4795, 2017 05 05.
Article in English | MEDLINE | ID: mdl-28077560

ABSTRACT

Type I toxin-antitoxin (TA) systems have been identified in a wide range of bacterial genomes. Here, we report the characterization of a new type I TA system present on the chromosome of the major human gastric pathogen, Helicobacter pylori. We show that the aapA1 gene encodes a 30 amino acid peptide whose artificial expression in H. pylori induces cell death. The synthesis of this toxin is prevented by the transcription of an antitoxin RNA, named IsoA1, expressed on the opposite strand of the toxin gene. We further reveal additional layers of post-transcriptional regulation that control toxin expression: (i) transcription of the aapA1 gene generates a full-length transcript whose folding impedes translation (ii) a 3΄ end processing of this message generates a shorter transcript that, after a structural rearrangement, becomes translatable (iii) but this rearrangement also leads to the formation of two stem-loop structures allowing formation of an extended duplex with IsoA1 via kissing-loop interactions. This interaction ensures both the translation inhibition of the AapA1 active message and its rapid degradation by RNase III, thus preventing toxin synthesis under normal growth conditions. Finally, a search for homologous mRNA structures identifies similar TA systems in a large number of Helicobacter and Campylobacter genomes.


Subject(s)
Antitoxins/genetics , Bacterial Proteins/genetics , Bacterial Toxins/genetics , Helicobacter pylori/genetics , RNA Folding/genetics , RNA, Bacterial/genetics , Antitoxins/biosynthesis , Antitoxins/chemistry , Bacterial Toxins/antagonists & inhibitors , Bacterial Toxins/biosynthesis , Bacterial Toxins/chemistry , Escherichia coli , Gene Expression Regulation, Bacterial , Genome, Bacterial , Helicobacter pylori/chemistry , Helicobacter pylori/pathogenicity , Humans , RNA Processing, Post-Transcriptional/genetics , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/biosynthesis , RNA, Messenger/chemistry , Ribonuclease III/chemistry , Ribonuclease III/genetics
6.
PLoS One ; 8(4): e60315, 2013.
Article in English | MEDLINE | ID: mdl-23565224

ABSTRACT

Chronic Helicobacter pylori infection provokes an inflammation of the gastric mucosa, at high risk for ulcer and cancer development. The most virulent strains harbor the cag pathogenicity island (cagPAI) encoding a type 4 secretion system, which allows delivery of bacterial effectors into gastric epithelial cells, inducing pro-inflammatory responses and phenotypic alterations reminiscent of an epithelial-to-mesenchymal transition (EMT). This study characterizes EMT features in H. pylori-infected gastric epithelial cells, and investigates their relationship with NF-κB activation. Cultured human gastric epithelial cell lines were challenged with a cagPAI+ H. pylori strain or cag isogenic mutants. Morphological changes, epithelial and mesenchymal gene expression and EMT-related microRNAs were studied. H. pylori up-regulates mesenchymal markers, including ZEB1. This transcription factor is prominently involved in the mesenchymal transition of infected cells and its up-regulation depends on cagPAI and NF-κB activation. ZEB1 expression and NF-κB activation were confirmed by immunohistochemistry in gastric mucosa from cagPAI+ H. pylori-infected patients. Gastric epithelial cell lines express high miR-200 levels, which are linked to ZEB1 in a reciprocal negative feedback loop and maintain their epithelial phenotype in non-infected conditions. However, miR-200b/c were increased upon infection, despite ZEB1 up-regulation and mesenchymal morphology. In the miR-200b-200a-429 cluster promoter, we identified a functional NF-κB binding site, recruiting NF-κB upon infection and trans-activating the microRNA cluster transcription. In conclusion, in gastric epithelial cells, cagPAI+ H. pylori activates NF-κB, which transactivates ZEB1, subsequently promoting mesenchymal transition. The unexpected N-FκB-dependent increase of miR-200 levels likely thwarts the irreversible loss of epithelial identity in that critical situation.


Subject(s)
Epithelial-Mesenchymal Transition/genetics , Gastric Mucosa/metabolism , Gastric Mucosa/microbiology , Helicobacter Infections/genetics , Helicobacter pylori/physiology , Homeodomain Proteins/genetics , Transcription Factors/genetics , Biomarkers/metabolism , Cell Line , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Gastritis/genetics , Gastritis/metabolism , Gastritis/microbiology , Gastritis/pathology , Gene Expression , Gene Expression Regulation , Homeodomain Proteins/metabolism , Humans , MicroRNAs/genetics , NF-kappa B/metabolism , Phenotype , Transcription Factors/metabolism , Zinc Finger E-box-Binding Homeobox 1
7.
Nucleic Acids Res ; 40(3): 1356-65, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22009679

ABSTRACT

Besides the fact that miR-96 and miR-182 belong to the miR-182/183 cluster, their seed region (UUGGCA, nucleotides 2-7) is identical suggesting potential common properties in mRNA target recognition and cellular functions. Here, we used the mRNA encoding Glypican-3, a heparan-sulfate proteoglycan, as a model target as its short 3' untranslated region is predicted to contain one miR-96/182 site, and assessed whether it is post-transcriptionally regulated by these two microRNAs. We found that miR-96 downregulated GPC3 expression by targeting its mRNA 3'-untranslated region and interacting with the predicted site. This downregulatory effect was due to an increased mRNA degradation and depended on Argonaute-2. Despite its seed similarity with miR-96, miR-182 was unable to regulate GPC3. This differential regulation was confirmed on two other targets, FOXO1 and FN1. By site-directed mutagenesis, we demonstrated that the miRNA nucleotide 8, immediately downstream the UUGGCA seed, plays a critical role in target recognition by miR-96 and miR-182. Our data suggest that because of a base difference at miRNA position 8, these two microRNAs control a completely different set of genes and therefore are functionally independent.


Subject(s)
Gene Expression Regulation , Glypicans/genetics , MicroRNAs/chemistry , 3' Untranslated Regions , Base Pairing , Base Sequence , Cell Line, Tumor , Cells, Cultured , Glypicans/metabolism , Humans , MicroRNAs/physiology , Molecular Sequence Data
8.
Silence ; 2(1): 7, 2011 Oct 25.
Article in English | MEDLINE | ID: mdl-22027184

ABSTRACT

BACKGROUND: MicroRNAs, post-transcriptional regulators of eukaryotic gene expression, are implicated in host defense against pathogens. Viruses and bacteria have evolved strategies that suppress microRNA functions, resulting in a sustainable infection. In this work we report that Helicobacter pylori, a human stomach-colonizing bacterium responsible for severe gastric inflammatory diseases and gastric cancers, downregulates an embryonic stem cell microRNA cluster in proliferating gastric epithelial cells to achieve cell cycle arrest. RESULTS: Using a deep sequencing approach in the AGS cell line, a widely used cell culture model to recapitulate early events of H. pylori infection of gastric mucosa, we reveal that hsa-miR-372 is the most abundant microRNA expressed in this cell line, where, together with hsa-miR-373, it promotes cell proliferation by silencing large tumor suppressor homolog 2 (LATS2) gene expression. Shortly after H. pylori infection, miR-372 and miR-373 synthesis is highly inhibited, leading to the post-transcriptional release of LATS2 expression and thus, to a cell cycle arrest at the G1/S transition. This downregulation of a specific cell-cycle-regulating microRNA is dependent on the translocation of the bacterial effector CagA into the host cells, a mechanism highly associated with the development of severe atrophic gastritis and intestinal-type gastric carcinoma. CONCLUSIONS: These data constitute a novel example of host-pathogen interplay involving microRNAs, and unveil the couple LATS2/miR-372 and miR-373 as an unexpected mechanism in infection-induced cell cycle arrest in proliferating gastric cells, which may be relevant in inhibition of gastric epithelium renewal, a major host defense mechanism against bacterial infections.

9.
Nature ; 464(7286): 250-5, 2010 Mar 11.
Article in English | MEDLINE | ID: mdl-20164839

ABSTRACT

Genome sequencing of Helicobacter pylori has revealed the potential proteins and genetic diversity of this prevalent human pathogen, yet little is known about its transcriptional organization and noncoding RNA output. Massively parallel cDNA sequencing (RNA-seq) has been revolutionizing global transcriptomic analysis. Here, using a novel differential approach (dRNA-seq) selective for the 5' end of primary transcripts, we present a genome-wide map of H. pylori transcriptional start sites and operons. We discovered hundreds of transcriptional start sites within operons, and opposite to annotated genes, indicating that complexity of gene expression from the small H. pylori genome is increased by uncoupling of polycistrons and by genome-wide antisense transcription. We also discovered an unexpected number of approximately 60 small RNAs including the epsilon-subdivision counterpart of the regulatory 6S RNA and associated RNA products, and potential regulators of cis- and trans-encoded target messenger RNAs. Our approach establishes a paradigm for mapping and annotating the primary transcriptomes of many living species.


Subject(s)
Gene Expression Profiling , Genome, Bacterial/genetics , Helicobacter Infections/microbiology , Helicobacter pylori/genetics , RNA, Bacterial/genetics , 5' Untranslated Regions/genetics , Amino Acid Sequence , Base Sequence , Cells, Cultured , Humans , Molecular Sequence Data , Nucleic Acid Conformation , Operon/genetics , RNA, Bacterial/chemistry , RNA, Bacterial/metabolism , RNA, Messenger/genetics , RNA, Untranslated , Sequence Alignment , Transcription, Genetic/genetics
10.
Nucleic Acids Res ; 36(15): e95, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18614605

ABSTRACT

We developed a functional selection system based on randomized genetic elements (GE) to identify potential regulators of hepatitis C virus (HCV) RNA translation, a process initiated by an internal ribosomal entry site (IRES). A retroviral HCV GE library was introduced into HepG2 cells, stably expressing the Herpes simplex virus thymidine kinase (HSV-TK) under the control of the HCV IRES. Cells that expressed transduced GEs inhibiting HSV-TK were selected via their resistance to ganciclovir. Six major GEs were rescued by PCR on the selected cell DNA and identified as HCV elements. We validated our strategy by further studying the activity of one of them, GE4, encoding the 5' end of the viral NS5A gene. GE4 inhibited HCV IRES-, but not cap-dependent, reporter translation in human hepatic cell lines and inhibited HCV infection at a post-entry step, decreasing by 85% the number of viral RNA copies. This method can be applied to the identification of gene expression regulators.


Subject(s)
Gene Expression Regulation, Viral , Hepacivirus/genetics , Protein Biosynthesis , Viral Nonstructural Proteins/genetics , 5' Untranslated Regions/chemistry , Cell Line , Cloning, Molecular , Gene Library , Humans , RNA, Viral/chemistry , Regulatory Sequences, Ribonucleic Acid
11.
Biol Cell ; 95(3-4): 229-38, 2003.
Article in English | MEDLINE | ID: mdl-12867086

ABSTRACT

Oligonucleotides exhibiting a strong affinity and a high specificity for RNA hairpins were obtained by in vitro selection. Such oligomers give rise to loop-loop complexes with the target hairpins: the trans-activation responsive (TAR) element of the Human Immunodeficiency virus-1 (HIV-1) or subdomains of the Hepatitis C virus (HCV) mRNA. Chemically modified derivatives of an antiTAR aptamer were shown to compete out the binding of the viral protein Tat and to selectively inhibit the in vitro TAR-dependent transcription of a reporter gene. In addition, antisense oligomers derived from sequences selected against the domain IIId of the HCV internal ribosome entry site were shown to specifically block translation both in a cell-free assay and in cultured cells.


Subject(s)
Gene Expression Regulation, Viral/genetics , Protein Biosynthesis/genetics , RNA, Viral/genetics , Viral Proteins/biosynthesis , Animals , HIV Long Terminal Repeat/genetics , Hepacivirus/genetics , Hepacivirus/metabolism , Humans , Oligonucleotides/genetics , Oligonucleotides/metabolism , Protein Structure, Tertiary/genetics , Viral Proteins/genetics
12.
Nucleic Acids Res ; 31(4): 1275-81, 2003 Feb 15.
Article in English | MEDLINE | ID: mdl-12582247

ABSTRACT

The hepatitis C virus (HCV) 5' untranslated region (UTR) has been extensively studied with regard to its internal ribosomal entry site (IRES) activity. In this work we present results suggesting the existence of a strong promoter activity carried by the DNA sequence corresponding to the HCV 5' UTR. This activity was not detected when the HCV 5' UTR sequence was replaced by HCV 3' UTR or poliovirus 5' UTR sequences. These results were further confirmed by using bicistronic constructions. We demonstrated the presence of an mRNA initiated in this 5' UTR sequence and located the initiation site by the 5' RACE method at nucleotide 67. Furthermore, northern experiments and flow cytometry analysis showed the unambiguous activity of such a promoter sequence in stably transfected cells. Our results strongly suggest that the data obtained using bicistronic DNA constructs carrying the HCV 5' UTR should be analyzed not only at the translational but also at the transcriptional level.


Subject(s)
5' Untranslated Regions/genetics , Hepacivirus/genetics , Promoter Regions, Genetic/genetics , 5' Untranslated Regions/chemistry , Base Sequence , DNA, Complementary/genetics , Gene Expression , Genome, Viral , Green Fluorescent Proteins , Humans , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Molecular Sequence Data , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Homology, Nucleic Acid , Transcription Initiation Site , Transcription, Genetic , Transfection , Tumor Cells, Cultured
13.
Nucleic Acids Res ; 31(2): 734-42, 2003 Jan 15.
Article in English | MEDLINE | ID: mdl-12527783

ABSTRACT

Initiation of protein synthesis on the hepatitis C virus (HCV) mRNA involves a structured element corresponding to the 5' untranslated region and constituting an internal ribosome entry site (IRES). The domain IIId of the HCV IRES, an imperfect RNA hairpin extending from nucleotides 253 to 279 of the viral mRNA, has been shown to be essential for translation and for the binding of the 40S ribosomal subunit. We investigated the properties of a series of antisense 2'-O-methyloligoribonucleotides targeted to various portions of the domain IIId. Several oligomers, 14-17 nt in length, selectively inhibited in vitro translation of a bicistronic RNA construct in rabbit reticulocyte lysate with IC(50)s <10 nM. The effect was restricted to the second cistron (the Renilla luciferase) located downstream of the HCV IRES; no effect was observed on the expression of the first cistron (the firefly luciferase) which was translated in a cap-dependent manner. Moreover, antisense 2'-O-methyloligoribonucleotides specifically competed with the 40S ribosomal subunit for binding to the IRES RNA in a filter- retention assay. The antisense efficiency of the oligonucleotides was nicely correlated to their affinity for the IIId subdomain and to their ability to displace 40S ribosomal subunit, making this process a likely explanation for in vitro inhibition of HCV-IRES-dependent translation.


Subject(s)
Hepacivirus/genetics , Oligonucleotides, Antisense/genetics , Protein Biosynthesis/genetics , Ribosomes/metabolism , Base Sequence , Binding Sites/genetics , Binding, Competitive , Cell-Free System , Dose-Response Relationship, Drug , Electrophoretic Mobility Shift Assay , Humans , Luciferases/genetics , Luciferases/metabolism , Oligonucleotides, Antisense/metabolism , Oligonucleotides, Antisense/pharmacology , Plasmids/genetics , Protein Biosynthesis/drug effects , RNA/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Transfection , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL