Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Front Microbiol ; 14: 1118329, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36846801

RESUMO

Escherichia coli CspA is an RNA binding protein that accumulates during cold-shock and stimulates translation of several mRNAs-including its own. Translation in the cold of cspA mRNA involves a cis-acting thermosensor element, which enhances ribosome binding, and the trans-acting action of CspA. Using reconstituted translation systems and probing experiments we show that, at low temperature, CspA specifically promotes the translation of the cspA mRNA folded in the conformation less accessible to the ribosome, which is formed at 37°C but is retained upon cold shock. CspA interacts with its mRNA without inducing large structural rearrangements, but allowing the progression of the ribosomes during the transition from translation initiation to translation elongation. A similar structure-dependent mechanism may be responsible for the CspA-dependent translation stimulation observed with other probed mRNAs, for which the transition to the elongation phase is progressively facilitated during cold acclimation with the accumulation of CspA.

2.
Mol Microbiol ; 113(3): 570-579, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32185838

RESUMO

The acquisition process of antibiotic resistance in an otherwise susceptible organism is shaped by the ecology of the species. Unlike other relevant human pathogens, Listeria monocytogenes has maintained a high rate of susceptibility to the antibiotics used for decades to treat human and animal infections. However, L. monocytogenes can acquire antibiotic resistance genes from other organisms' plasmids and conjugative transposons. Ecological factors could account for its susceptibility. L. monocytogenes is ubiquitous in nature, most frequently including reservoirs unexposed to antibiotics, including intracellular sanctuaries. L. monocytogenes has a remarkably closed genome, reflecting limited community interactions, small population sizes and high niche specialization. The L. monocytogenes species is divided into variants that are specialized in small specific niches, which reduces the possibility of coexistence with potential donors of antibiotic resistance. Interactions with potential donors are also hampered by interspecies antagonism. However, occasional increases in population sizes (and thus the possibility of acquiring antibiotic resistance) can derive from selection of the species based on intrinsic or acquired resistance to antibiotics, biocides, heavy metals or by a natural tolerance to extreme conditions. High-quality surveillance of the emergence of resistance to the key drugs used in primary therapy is mandatory.


Assuntos
Farmacorresistência Bacteriana/genética , Listeria monocytogenes/genética , Listeria monocytogenes/metabolismo , Animais , Antibacterianos/farmacologia , Proteínas de Bactérias/genética , Resistência Microbiana a Medicamentos/genética , Humanos , Testes de Sensibilidade Microbiana , Plasmídeos/efeitos dos fármacos
4.
Proc Natl Acad Sci U S A ; 115(52): 13359-13364, 2018 12 26.
Artigo em Inglês | MEDLINE | ID: mdl-30545912

RESUMO

To overcome the action of antibiotics, bacteria have evolved a variety of different strategies, such as drug modification, target mutation, and efflux pumps. Recently, we performed a genome-wide analysis of Listeria monocytogenes gene expression after growth in the presence of antibiotics, identifying genes that are up-regulated upon antibiotic treatment. One of them, lmo0762, is a homolog of hflX, which encodes a heat shock protein that rescues stalled ribosomes by separating their two subunits. To our knowledge, ribosome splitting has never been described as an antibiotic resistance mechanism. We thus investigated the role of lmo0762 in antibiotic resistance. First, we demonstrated that lmo0762 is an antibiotic resistance gene that confers protection against lincomycin and erythromycin, and that we renamed hflXr (hflX resistance). We show that hflXr expression is regulated by a transcription attenuation mechanism relying on the presence of alternative RNA structures and a small ORF encoding a 14 amino acid peptide containing the RLR motif, characteristic of macrolide resistance genes. We also provide evidence that HflXr is involved in ribosome recycling in presence of antibiotics. Interestingly, L. monocytogenes possesses another copy of hflX, lmo1296, that is not involved in antibiotic resistance. Phylogenetic analysis shows several events of hflXr duplication in prokaryotes and widespread presence of hflXr in Firmicutes. Overall, this study reveals the Listeria hflXr as the founding member of a family of antibiotic resistance genes. The resistance conferred by this gene is probably of importance in the environment and within microbial communities.


Assuntos
Farmacorresistência Bacteriana/genética , Proteínas de Escherichia coli/metabolismo , Proteínas de Ligação ao GTP/metabolismo , Listeria monocytogenes/metabolismo , Antibacterianos/farmacologia , Proteínas de Bactérias/metabolismo , Farmacorresistência Bacteriana/fisiologia , Resistência Microbiana a Medicamentos/efeitos dos fármacos , Proteínas de Escherichia coli/genética , Evolução Molecular , Proteínas de Ligação ao GTP/genética , Listeria monocytogenes/genética , Testes de Sensibilidade Microbiana , Filogenia , Proteínas Ribossômicas/metabolismo , Ribossomos/metabolismo
5.
Nat Microbiol ; 3(8): 962, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-29941881

RESUMO

This Article contains a URL for a publically available whole-genome browser ( http://nterm.listeriomics.pasteur.fr ). However, due to technical constraint, this website has been replaced with an alternative ( https://listeriomics.pasteur.fr ).

6.
mBio ; 8(5)2017 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-29089430

RESUMO

Listeria monocytogenes is a bacterial pathogen causing severe foodborne infections in humans and animals. Listeria can enter into host cells and survive and multiply therein, due to an arsenal of virulence determinants encoded in different loci on the chromosome. Several key Listeria virulence genes are clustered in Listeria pathogenicity island 1. This important locus also contains orfX (lmo0206), a gene of unknown function. Here, we found that OrfX is a small, secreted protein whose expression is positively regulated by PrfA, the major transcriptional activator of Listeria virulence genes. We provide evidence that OrfX is a virulence factor that dampens the oxidative response of infected macrophages, which contributes to intracellular survival of bacteria. OrfX is targeted to the nucleus and interacts with the regulatory protein RybP. We show that in macrophages, the expression of OrfX decreases the level of RybP, which controls cellular infection. Collectively, these data reveal that Listeria targets RybP and evades macrophage oxidative stress for efficient infection. Altogether, OrfX is after LntA, the second virulence factor acting directly in the nucleus.IMPORTANCEListeria monocytogenes is a model bacterium that has been successfully used over the last 30 years to refine our understanding of the molecular, cellular, and tissular mechanisms of microbial pathogenesis. The major virulence factors of pathogenic Listeria species are located on a single chromosomal locus. Here, we report that the last gene of this locus encodes a small secreted nucleomodulin, OrfX, that is required for bacterial survival within macrophages and in the infected host. This work demonstrates that the production of OrfX contributes to limiting the host innate immune response by dampening the oxidative response of macrophages. We also identify a target of OrfX, RybP, which is an essential pleiotropic regulatory protein of the cell, and uncover its role in host defense. Our data reinforce the view that the secretion of nucleomodulins is an important strategy used by microbial pathogens to promote infection.


Assuntos
Proteínas de Bactérias/genética , Listeria monocytogenes/genética , Listeria monocytogenes/patogenicidade , Fatores de Virulência/genética , Células A549 , Animais , Carga Bacteriana , Proteínas de Bactérias/metabolismo , Células HEK293 , Células HeLa , Humanos , Imunidade Inata , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Listeriose/microbiologia , Fígado/microbiologia , Macrófagos/imunologia , Macrófagos/microbiologia , Camundongos , Fatores de Terminação de Peptídeos/genética , Fatores de Terminação de Peptídeos/metabolismo , Proteínas de Ligação a RNA/genética , Proteínas Repressoras , Baço/microbiologia , Virulência , Fatores de Virulência/metabolismo
7.
Curr Opin Microbiol ; 39: 81-88, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-29111488

RESUMO

Small proteins, that is, polypeptides of 50 amino acids (aa) or less, are increasingly recognized as important regulators in bacteria. Secreted or not, their small size make them versatile proteins, involved in a wide range of processes. They may allow bacteria to sense and to respond to stresses, to send signals and communicate, and to modulate infections. Bacteriophages also produce small proteins to influence lysogeny/lysis decisions. In this review, we update the present view on small proteins functions, and discuss their possible applications.


Assuntos
Bacillus subtilis , Proteínas de Bactérias , Bacteriófagos , Escherichia coli , Proteínas Virais , Transdução de Sinais
8.
BMC Genomics ; 18(1): 882, 2017 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-29145803

RESUMO

BACKGROUND: Small regulatory RNAs (sRNAs) are widely found in bacteria and play key roles in many important physiological and adaptation processes. Studying their evolution and screening for events of coevolution with other genomic features is a powerful way to better understand their origin and assess a common functional or adaptive relationship between them. However, evolution and coevolution of sRNAs with coding genes have been sparsely investigated in bacterial pathogens. RESULTS: We designed a robust and generic phylogenomics approach that detects correlated evolution between sRNAs and protein-coding genes using their observed and inferred patterns of presence-absence in a set of annotated genomes. We applied this approach on 79 complete genomes of the Listeria genus and identified fifty-two accessory sRNAs, of which most were present in the Listeria common ancestor and lost during Listeria evolution. We detected significant coevolution between 23 sRNA and 52 coding genes and inferred the Listeria sRNA-coding genes coevolution network. We characterized a main hub of 12 sRNAs that coevolved with genes encoding cell wall proteins and virulence factors. Among them, an sRNA specific to L. monocytogenes species, rli133, coevolved with genes involved either in pathogenicity or in interaction with host cells, possibly acting as a direct negative post-transcriptional regulation. CONCLUSIONS: Our approach allowed the identification of candidate sRNAs potentially involved in pathogenicity and host interaction, consistent with recent findings on known pathogenicity actors. We highlight four sRNAs coevolving with seven internalin genes, some of which being important virulence factors in Listeria.


Assuntos
Proteínas de Bactérias/genética , Evolução Molecular , Listeria/genética , Pequeno RNA não Traduzido/genética , Redes Reguladoras de Genes , Genes Bacterianos , Genoma Bacteriano , Listeria/patogenicidade
9.
Nat Microbiol ; 2: 17005, 2017 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-28191904

RESUMO

To adapt to changing environments, bacteria have evolved numerous pathways that activate stress response genes. In Gram-positive bacteria, the stressosome, a cytoplasmic complex, relays external cues and activates the sigma B regulon. The stressosome is structurally well-characterized in Bacillus, but how it senses stress remains elusive. Here, we report a genome-wide N-terminomic approach in Listeria that strikingly led to the discovery of 19 internal translation initiation sites and 6 miniproteins, among which one, Prli42, is conserved in Firmicutes. Prli42 is membrane-anchored and interacts with orthologues of Bacillus stressosome components. We reconstituted the Listeria stressosome in vitro and visualized its supramolecular structure by electron microscopy. Analysis of a series of Prli42 mutants demonstrated that Prli42 is important for sigma B activation, bacterial growth following oxidative stress and for survival in macrophages. Taken together, our N-terminonic approach unveiled Prli42 as a long-sought link between stress and the stressosome.


Assuntos
Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Firmicutes/genética , Regulação Bacteriana da Expressão Gênica , Listeria monocytogenes/genética , Estresse Fisiológico/genética , Firmicutes/metabolismo , Genoma Bacteriano , Listeria monocytogenes/metabolismo , Membranas/química , Membranas/metabolismo , Processamento de Proteína Pós-Traducional/genética , Proteômica/métodos , Regulon/genética , Fator sigma/genética , Transdução de Sinais
10.
Semin Cell Dev Biol ; 65: 11-19, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-27381344

RESUMO

Gene expression regulation is a critical question in host-pathogen interactions, and RNAs act as key players in this process. In this review, we focus on the mammalian RNA response to bacterial infection, with a special interest on microRNAs and long non-coding RNAs. We discuss the role of cellular miRNAs in immunity, the implication of circulating miRNAs as well as the influence of the microbiome on the miRNA response. We also review how pathogens counteract the host miRNA expression. Interestingly, bacterial non-coding RNAs regulate host gene expression and conversely eukaryotic miRNAs may regulate bacterial gene expression. Overall, the characterization of RNA regulatory networks represents an emerging theme in the field of host pathogen interactions.


Assuntos
Bactérias/genética , Infecções Bacterianas/genética , Interações Hospedeiro-Patógeno/genética , MicroRNAs/genética , RNA Longo não Codificante/genética , Animais , Bactérias/crescimento & desenvolvimento , Bactérias/imunologia , Infecções Bacterianas/imunologia , Infecções Bacterianas/microbiologia , Resistência à Doença/genética , Regulação da Expressão Gênica , Redes Reguladoras de Genes/imunologia , Interações Hospedeiro-Patógeno/imunologia , Humanos , Imunidade Inata , Mamíferos , MicroRNAs/imunologia , NF-kappa B/genética , NF-kappa B/imunologia , RNA Longo não Codificante/imunologia , Transdução de Sinais
11.
Methods ; 117: 48-58, 2017 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-28027957

RESUMO

RNA-protein interactions are at the bases of many biological processes, forming either tight and stable functional ribonucleoprotein (RNP) complexes (i.e. the ribosome) or transitory ones, such as the complexes involving RNA chaperone proteins. To localize the sites where a protein interacts on an RNA molecule, a common simple and inexpensive biochemical method is the footprinting technique. The protein leaves its footprint on the RNA acting as a shield to protect the regions of interaction from chemical modification or cleavages obtained with chemical or enzymatic nucleases. This method has proven its efficiency to study in vitro the organization of stable RNA-protein complexes. Nevertheless, when the protein binds the RNA very dynamically, with high off-rates, protections are very often difficult to observe. For the analysis of these transient complexes, we describe an alternative strategy adapted from the Site Directed Chemical Probing (SDCP) approach and we compare it with classical footprinting. SDCP relies on the modification of the RNA binding protein to tether an RNA probe (usually Fe-EDTA) to specific protein positions. Local cleavages on the regions of interaction can be used to localize the protein and position its domains on the RNA molecule. This method has been used in the past to monitor stable complexes; we provide here a detailed protocol and a practical example of its application to the study of Escherichia coli RNA chaperone protein S1 and its transitory complexes with mRNAs.


Assuntos
Chaperonas Moleculares/química , Impressão Molecular/métodos , Proteínas de Ligação a RNA/química , RNA/química , Coloração e Rotulagem/métodos , Sequência de Bases , Compostos de Bifenilo/química , Ácido Edético/análogos & derivados , Ácido Edético/química , Escherichia coli/genética , Escherichia coli/metabolismo , Expressão Gênica , Radical Hidroxila/metabolismo , Modelos Moleculares , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Sondas Moleculares/química , Mutagênese Sítio-Dirigida , Conformação de Ácido Nucleico , Conformação Proteica , RNA/genética , RNA/metabolismo , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
12.
Biochimie ; 114: 18-29, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25792421

RESUMO

To adapt their metabolism rapidly and constantly in response to environmental variations, bacteria often target the translation initiation process, during which the ribosome assembles on the mRNA. Here, we review different mechanisms of regulation mediated by cis-acting elements, sRNAs and proteins, showing, when possible, their intimate connection with the translational apparatus. Indeed the ribosome itself could play a direct role in several regulatory mechanisms. Different features of the regulatory signals (sequences, structures and their positions on the mRNA) are contributing to the large variety of regulatory mechanisms. Ribosome heterogeneity, variation of individual cells responses and the spatial and temporal organization of the translation process add more layers of complexity. This hampers to define manageable set of rules for bacterial translation initiation control.


Assuntos
Regulação Bacteriana da Expressão Gênica , Iniciação Traducional da Cadeia Peptídica , Proteínas de Bactérias/biossíntese , Proteínas de Bactérias/genética , Estabilidade de RNA , RNA Bacteriano/fisiologia , Ribossomos/fisiologia , Ativação Transcricional
13.
PLoS Biol ; 11(12): e1001731, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24339747

RESUMO

Regulation of translation initiation is well appropriate to adapt cell growth in response to stress and environmental changes. Many bacterial mRNAs adopt structures in their 5' untranslated regions that modulate the accessibility of the 30S ribosomal subunit. Structured mRNAs interact with the 30S in a two-step process where the docking of a folded mRNA precedes an accommodation step. Here, we used a combination of experimental approaches in vitro (kinetic of mRNA unfolding and binding experiments to analyze mRNA-protein or mRNA-ribosome complexes, toeprinting assays to follow the formation of ribosomal initiation complexes) and in vivo (genetic) to monitor the action of ribosomal protein S1 on the initiation of structured and regulated mRNAs. We demonstrate that r-protein S1 endows the 30S with an RNA chaperone activity that is essential for the docking and the unfolding of structured mRNAs, and for the correct positioning of the initiation codon inside the decoding channel. The first three OB-fold domains of S1 retain all its activities (mRNA and 30S binding, RNA melting activity) on the 30S subunit. S1 is not required for all mRNAs and acts differently on mRNAs according to the signals present at their 5' ends. This work shows that S1 confers to the ribosome dynamic properties to initiate translation of a large set of mRNAs with diverse structural features.


Assuntos
Proteínas de Escherichia coli/fisiologia , Escherichia coli/fisiologia , Biossíntese de Proteínas/fisiologia , Dobramento de RNA/fisiologia , RNA Mensageiro/fisiologia , Proteínas Ribossômicas/fisiologia , Ribossomos/fisiologia , Regulação Bacteriana da Expressão Gênica/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...