Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
1.
J Bacteriol ; 202(15)2020 07 09.
Artigo em Inglês | MEDLINE | ID: mdl-32393522

RESUMO

Naturally occurring free fatty acids (FFAs) are recognized as potent antimicrobial agents that also affect the production of virulence factors in bacterial pathogens. In the foodborne pathogen Listeria monocytogenes, some medium- and long-chain FFAs act as antimicrobial agents as well as signaling compounds, causing a repression of transcription of virulence genes. We previously observed that the master virulence regulator PrfA is involved in both the antimicrobial and virulence-inhibitory response of L. monocytogenes to selected FFAs, but the underlying mechanisms are presently unknown. Here, we present a systematic analysis of the antimicrobial and PrfA-inhibitory activities of medium- and long-chain FFAs of various carbon chain lengths and degrees of saturation. We observed that exposure to specific antimicrobial and nonantimicrobial FFAs prevented PrfA-dependent activation of virulence gene transcription and reduced the levels of PrfA-regulated virulence factors. Thus, an antimicrobial activity was not compulsory for the PrfA-inhibitory ability of an FFA. In vitro binding experiments revealed that PrfA-inhibitory FFAs were also able to prevent the constitutively active variant PrfA* from binding to the PrfA box in the promoter region of the virulence gene hly, whereas noninhibitory FFAs did not affect its ability to bind DNA. Notably, the unsaturated FFAs inhibited the DNA binding activity of PrfA* most efficiently. Altogether, our findings support a model in which specific FFAs orchestrate a generalized reduction of the virulence potential of L. monocytogenes by directly targeting the key virulence regulator PrfA.IMPORTANCEListeria monocytogenes is a Gram-positive pathogen able to cause foodborne infections in humans and animals. Key virulence genes in L. monocytogenes are activated by the transcription regulator PrfA, a DNA binding protein belonging to the CRP/FNR family. Various signals from the environment are known to affect the activity of PrfA, either positively or negatively. Recently, we found that specific medium- and long-chain free fatty acids act as antimicrobial agents as well as signaling compounds in L. monocytogenes Here, we show that both antimicrobial and nonantimicrobial free fatty acids inhibit PrfA-dependent activation of virulence gene transcription by interfering with the DNA binding activity of PrfA. Our findings suggest that free fatty acids could be candidates for alternative therapies against L. monocytogenes.


Assuntos
Proteínas de Bactérias/metabolismo , Proteínas de Ligação a DNA/metabolismo , Ácidos Graxos não Esterificados/metabolismo , Listeria monocytogenes/metabolismo , Fatores de Terminação de Peptídeos/metabolismo , Proteínas de Bactérias/genética , Proteínas de Ligação a DNA/genética , Regulação Bacteriana da Expressão Gênica , Humanos , Listeria monocytogenes/genética , Listeria monocytogenes/patogenicidade , Listeriose/microbiologia , Fatores de Terminação de Peptídeos/genética , Regiões Promotoras Genéticas , Virulência
2.
RNA Biol ; 16(3): 270-281, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30706751

RESUMO

The bacterial pathogen Listeria monocytogenes encodes seven homologous small regulatory RNAs, named the LhrC family of sRNAs. The LhrCs are highly induced under infection-relevant conditions and are known to inhibit the expression of multiple target mRNAs encoding virulence-associated surface proteins. In all cases studied so far, the LhrCs use their CU-rich regions for base pairing to complementary AG-rich sequences of the ribosomal binding site (RBS) of specific target mRNAs. Consequently, LhrC-mRNA interaction results in inhibition of translation followed by mRNA degradation, corresponding to the canonical model for sRNA-mediated gene regulation in bacteria. Here, we demonstrate that the LhrC sRNAs employ a different regulatory mechanism when acting to down-regulate the expression of tcsA, encoding a T cell-stimulating antigen. In this case, LhrC base pairs to an AG-rich site located well upstream of the RBS in tcsA mRNA. Using an in vitro translation assay, we found that LhrC could not prevent the ribosome from translating the tcsA messenger. Rather, the LhrC sRNAs act to decrease the half-life of tcsA mRNA in vivo. Importantly, LhrC-mediated destabilization of tcsA mRNA relies on an intact LhrC binding site near the 5´-end of the tcsA mRNA and occurs independently of translation. Based on these findings, we propose an alternative mechanism for LhrC-mediated control in L. monocytogenes that relies solely on sRNA-induced degradation of a target mRNA.


Assuntos
Regulação Bacteriana da Expressão Gênica , Listeria monocytogenes/fisiologia , Listeriose/microbiologia , Interferência de RNA , RNA Bacteriano/genética , RNA Mensageiro/genética , Pequeno RNA não Traduzido/genética , Regiões 5' não Traduzidas , Sequência de Bases , Sítios de Ligação , Listeriose/imunologia , Modelos Biológicos , Conformação de Ácido Nucleico , Biossíntese de Proteínas , Processamento Pós-Transcricional do RNA , Estabilidade de RNA , RNA Bacteriano/química , Pequeno RNA não Traduzido/química
3.
RNA Biol ; 16(10): 1424-1437, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31242083

RESUMO

The facultative intracellular pathogen Listeria monocytogenes can persist and grow in a diverse range of environmental conditions, both outside and within its mammalian host. The alternative sigma factor Sigma B (σB) plays an important role in this adaptability and is critical for the transition into the host. While some of the functions of the σB regulon in facilitating this transition are understood the role of σB-dependent small regulatory RNAs (sRNAs) remain poorly characterized. In this study, we focused on elucidating the function of Rli47, a σB-dependent sRNA that is highly induced in the intestine and in macrophages. Using a combination of in silico and in vivo approaches, a binding interaction was predicted with the Shine-Dalgarno region of the ilvA mRNA, which encodes threonine deaminase, an enzyme required for branched-chain amino acid biosynthesis. Both ilvA transcript levels and threonine deaminase activity were increased in a deletion mutant lacking the rli47 gene. The Δrli47 mutant displayed a shorter growth lag in isoleucine-depleted growth media relative to the wild-type, and a similar phenotype was also observed in a mutant lacking σB. The impact of the Δrli47 on the global transcription profile of the cell was investigated using RNA-seq, and a significant role for Rli47 in modulating amino acid metabolism was uncovered. Taken together, the data point to a model where Rli47 is responsible for specifically repressing isoleucine biosynthesis as a way to restrict growth under harsh conditions, potentially contributing to the survival of L. monocytogenes in niches both outside and within the mammalian host.


Assuntos
Proteínas de Bactérias/genética , Regulação Bacteriana da Expressão Gênica , Isoleucina/biossíntese , Listeria monocytogenes/genética , Listeria monocytogenes/metabolismo , Pequeno RNA não Traduzido/genética , Fator sigma/metabolismo , Aminoácidos/metabolismo , Proteínas de Bactérias/metabolismo , Transporte Biológico , Perfilação da Expressão Gênica , Mutação , Conformação de Ácido Nucleico , Ligação Proteica , RNA Mensageiro/genética , Pequeno RNA não Traduzido/química , Transcrição Gênica
4.
New Microbiol ; 40(2): 146-147, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28255602

RESUMO

We have previously shown that the phenothiazine, thioridazine, acts in synergy with the beta-lactam antibiotic, dicloxacillin, to kill methicillin-resistant Staphylococcus aureus. In this study, we investigated whether synergy by combining these two drugs could also be observed in vancomycin intermediate susceptible S. aureus (VISA) and methicillin-resistant Staphylococcus epidermidis (MRSE). Synergy was observed in three of four tested VISA strains, suggesting that the thickening of cell wall does not interfere with the effects of thioridazine. In S. epidermidis, no synergy was observed in all tested strains, suggesting that synergy by combining thioridazine and dicloxacillin is isolated to S. aureus species.


Assuntos
Antibacterianos/uso terapêutico , Dicloxacilina/uso terapêutico , Infecções Estafilocócicas/tratamento farmacológico , Staphylococcus aureus/efeitos dos fármacos , Staphylococcus epidermidis/efeitos dos fármacos , Tioridazina/uso terapêutico , Antibacterianos/administração & dosagem , Dicloxacilina/administração & dosagem , Antagonistas de Dopamina/administração & dosagem , Antagonistas de Dopamina/uso terapêutico , Sinergismo Farmacológico , Humanos , Testes de Sensibilidade Microbiana , Infecções Estafilocócicas/microbiologia , Tioridazina/administração & dosagem
5.
RNA Biol ; 13(9): 895-915, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27400116

RESUMO

Multicopy small RNAs (sRNAs) have gained recognition as an important feature of bacterial gene regulation. In the human pathogen Listeria monocytogenes, 5 homologous sRNAs, called LhrC1-5, control gene expression by base pairing to target mRNAs though 3 conserved UCCC motifs common to all 5 LhrCs. We show here that the sRNAs Rli22 and Rli33-1 are structurally and functionally related to LhrC1-5, expanding the LhrC family to 7 members, which makes it the largest multicopy sRNA family reported so far. Rli22 and Rli33-1 both contain 2 UCCC motifs important for post-transcriptional repression of 3 LhrC target genes. One such target, oppA, encodes a virulence-associated oligo-peptide binding protein. Like LhrC1-5, Rli22 and Rli33-1 employ their UCCC motifs to recognize the Shine-Dalgarno region of oppA mRNA and prevent formation of the ribosomal complex, demonstrating that the 7 sRNAs act in a functionally redundant manner. However, differential expression profiles of the sRNAs under infection-relevant conditions suggest that they might also possess non-overlapping functions. Collectively, this makes the LhrC family a unique case for studying the purpose of sRNA multiplicity in the context of bacterial virulence.


Assuntos
Regulação Bacteriana da Expressão Gênica , Listeria monocytogenes/genética , Família Multigênica , RNA Bacteriano/genética , Pequeno RNA não Traduzido/genética , Transcriptoma , Pareamento de Bases , Sequência de Bases , Ordem dos Genes , Conformação de Ácido Nucleico , Motivos de Nucleotídeos , Ligação Proteica , Interferência de RNA , RNA Bacteriano/química , RNA Mensageiro/genética , Pequeno RNA não Traduzido/química , Ribossomos/metabolismo
6.
Nucleic Acids Res ; 42(14): 9383-98, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25034691

RESUMO

The multicopy sRNA LhrC of the intracellular pathogen Listeria monocytogenes has been shown to be induced under infection-relevant conditions, but its physiological role and mechanism of action is not understood. In an attempt to pinpoint the exact terms of LhrC expression, cell envelope stress could be defined as a specific inducer of LhrC. In this process, the two-component system LisRK was shown to be indispensable for expression of all five copies of LhrC. lapB mRNA, encoding a cell wall associated protein that was recently identified as an important virulence factor, was disclosed to be directly bound by LhrC leading to an impediment of its translation. Although LhrC binds to Hfq, it does not require the RNA chaperone for stability or lapB mRNA interaction. The mechanism of LhrC-lapB mRNA binding was shown to involve three redundant CU-rich sites and a structural rearrangement in the sRNA. This study represents an extensive depiction of a so far uncharacterized multicopy sRNA and reveals interesting new aspects concerning its regulation, virulence association and mechanism of target binding.


Assuntos
Adesinas Bacterianas/genética , Regulação Bacteriana da Expressão Gênica , Listeria monocytogenes/genética , RNA Bacteriano/metabolismo , Pequeno RNA não Traduzido/metabolismo , Adesinas Bacterianas/metabolismo , Dosagem de Genes , Listeria monocytogenes/metabolismo , Listeria monocytogenes/patogenicidade , RNA Bacteriano/genética , RNA Bacteriano/fisiologia , RNA Mensageiro/metabolismo , Pequeno RNA não Traduzido/genética , Pequeno RNA não Traduzido/fisiologia , Estresse Fisiológico , Virulência/genética
7.
RNA Biol ; 12(9): 985-97, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26176322

RESUMO

Listeria monocytogenes is the causative agent of the foodborne disease listeriosis. During infection, L. monocytogenes produces an array of non-coding RNAs, including the multicopy sRNA LhrC. These five, nearly identical sRNAs are highly induced in response to cell envelope stress and target the virulence adhesin lapB at the post-transcriptional level. Here, we demonstrate that LhrC controls expression of additional genes encoding cell envelope-associated proteins with virulence function. Using transcriptomics and proteomics, we identified a set of genes affected by LhrC in response to cell envelope stress. Three targets were significantly down-regulated by LhrC at both the RNA and protein level: lmo2349, tcsA and oppA. All three genes encode membrane-associated proteins: A putative substrate binding protein of an amino acid ABC transporter (Lmo2349); the CD4+ T cell-stimulating antigen TcsA, and the oligopeptide binding protein OppA, of which the latter 2 are required for full virulence of L. monocytogenes. For OppA, we show that LhrC acts by direct base paring to the ribosome binding site of the oppA mRNA, leading to an impediment of its translation and a decreased mRNA level. The sRNA-mRNA interaction depends on 2 of 3 CU-rich regions in LhrC allowing binding of 2 oppA mRNAs to a single LhrC molecule. Finally, we found that LhrC contributes to infection in macrophage-like cells. These findings demonstrate a central role for LhrC in controlling the level of OppA and other virulence-associated cell envelope proteins in response to cell envelope stress.


Assuntos
Proteínas de Bactérias/genética , Proteínas de Transporte/genética , Dosagem de Genes , Regulação Bacteriana da Expressão Gênica , Lipoproteínas/genética , Listeria monocytogenes/genética , Pequeno RNA não Traduzido/genética , Animais , Proteínas de Bactérias/metabolismo , Sequência de Bases , Sítios de Ligação , Proteínas de Transporte/metabolismo , Perfilação da Expressão Gênica , Lipoproteínas/metabolismo , Listeria monocytogenes/metabolismo , Macrófagos/microbiologia , Camundongos , Mutação , Conformação de Ácido Nucleico , Motivos de Nucleotídeos , Óperon , Proteoma , Proteômica/métodos , RNA Mensageiro/química , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Pequeno RNA não Traduzido/química , Pequeno RNA não Traduzido/metabolismo , Estresse Fisiológico/genética , Transcriptoma
8.
Nucleic Acids Res ; 40(4): 1818-27, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22053088

RESUMO

Expression of virulence factors in the human bacterial pathogen Listeria monocytogenes is almost exclusively regulated by the transcriptional activator PrfA. The translation of prfA is controlled by a thermosensor located in the 5'-untranslated RNA (UTR), and is high at 37°C and low at temperatures <30°C. In order to develop a thermoregulated translational expression system, the 5'-UTR and different lengths of the prfA-coding sequences were placed in front of lacZ. When expressed in Escherichia coli, the ß-galactosidase expression was directly correlated to the length of the prfA-coding mRNA lying in front of lacZ. A similar effect was detected with gfp as a reporter gene in both L. monocytogenes and E. coli, emphasizing the requirement of the prfA-coding RNA for maximal expression. In vitro transcription/translation and mutational analysis suggests a role for the first 20 codons of the native prfA-mRNA for maximal expression. By toe-print and RNA-probing analysis, a flexible hairpin-loop located immediately downstream of the start-codon was shown to be important for ribosomal binding. The present work determines the importance of an unstructured part of the 5'-coding region of the prfA-mRNA for efficient translation.


Assuntos
Proteínas de Bactérias/genética , Códon , Fatores de Terminação de Peptídeos/genética , Biossíntese de Proteínas , Proteínas de Bactérias/biossíntese , Genes Reporter , Listeria monocytogenes/genética , Listeria monocytogenes/metabolismo , Mutação , Conformação de Ácido Nucleico , Fatores de Terminação de Peptídeos/biossíntese , Estabilidade Proteica , Estabilidade de RNA , RNA Mensageiro/biossíntese , RNA Mensageiro/química , RNA Mensageiro/metabolismo , Ribossomos/metabolismo
9.
Methods Mol Biol ; 2741: 195-206, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38217655

RESUMO

Regulatory RNAs in bacteria are known to act by base pairing with other RNAs. Interactions between two partner RNAs can be investigated by electrophoretic mobility shift assays. The regions predicted to be engaged in base pairing are analyzed by introducing mutations in one RNA that prevent RNA-RNA complex formation. Next, base pairing is restored by introducing complementary mutations in its partner RNA. Here, we describe the mutational strategy and experimental methods used to validate specific base pairing between two RNA species.


Assuntos
Bactérias , RNA , RNA/genética , Ensaio de Desvio de Mobilidade Eletroforética , Pareamento de Bases , Mutação , Bactérias/genética
10.
FEMS Microbiol Rev ; 47(4)2023 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-37437907

RESUMO

Free fatty acids (FFAs) have long been acknowledged for their antimicrobial activity. More recently, long-chain FFAs (>12 carbon atoms) are receiving increased attention for their potent antivirulence activity against pathogenic bacteria. In the gastrointestinal tract, foodborne pathogens encounter a variety of long-chain FFAs derived from the diet, metabolic activities of the gut microbiota, or the host. This review highlights the role of long-chain FFAs as signaling molecules acting to inhibit the infectious potential of important foodborne pathogens, including Salmonella and Listeria monocytogenes. Various long-chain FFAs interact with sensory proteins and transcriptional regulators controlling the expression of infection-relevant genes. Consequently, long-chain FFAs may act to disarm bacterial pathogens of their virulence factors. Understanding how foodborne pathogens sense and respond to long-chain FFAs may enable the design of new anti-infective approaches.


Assuntos
Anti-Infecciosos , Ácidos Graxos , Anti-Infecciosos/farmacologia , Ácidos Graxos não Esterificados/farmacologia , Bactérias , Fatores de Virulência/genética
11.
Microb Cell ; 10(7): 145-156, 2023 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-37395997

RESUMO

Staphylococcus aureus is a major human pathogen that utilises many surface-associated and secreted proteins to form biofilms and cause disease. However, our understanding of these processes is limited by challenges of using fluorescent protein reporters in their native environment, because they must be exported and fold correctly to become fluorescent. Here, we demonstrate the feasibility of using the monomeric superfolder GFP (msfGFP) exported from S. aureus. By fusing msfGFP to signal peptides for the Secretory (Sec) and Twin Arginine Translocation (Tat) pathways, the two major secretion pathways in S. aureus, we quantified msfGFP fluorescence in bacterial cultures and cell-free supernatant from the cultures. When fused to a Tat signal peptide, we detected msfGFP fluorescence inside but not outside bacterial cells, indicating a failure to export msfGFP. However, when fused to a Sec signal peptide, msfGFP fluorescence was present outside cells, indicating successful export of the msfGFP in the unfolded state, followed by extracellular folding and maturation to the photoactive state. We applied this strategy to study coagulase (Coa), a secreted protein and a major contributor to the formation of a fibrin network in S. aureus biofilms that protects bacteria from the host immune system and increases attachment to host surfaces. We confirmed that a genomically integrated C-terminal fusion of Coa to msfGFP does not impair the activity of Coa or its localisation within the biofilm matrix. Our findings demonstrate that msfGFP is a good candidate fluorescent reporter to consider when studying proteins secreted by the Sec pathway in S. aureus.

12.
Front Microbiol ; 14: 1324062, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38239729

RESUMO

Bacteria have evolved numerous regulatory pathways to survive in changing environments. The SOS response is an inducible DNA damage repair system that plays an indispensable role in bacterial adaptation and pathogenesis. Here we report a discovery of the previously uncharacterized protein Lmo0946 as an SOS response interfering factor (Sif) in the human pathogen Listeria monocytogenes. Functional genetic studies demonstrated that sif is indispensable for normal growth of L. monocytogenes in stress-free as well as multi-stress conditions, and sif contributes to susceptibility to ß-lactam antibiotics, biofilm formation and virulence. Absence of Sif promoted the SOS response and elevated expression of mobilome genes accompanied by mobilization of the A118 prophage and ICELm-1 mobile genetic elements (MGEs). These changes were found to be associated with decreased expression of general stress response genes from the σB regulon as well as virulence genes, including the PrfA regulon. Together, this study uncovers an unexpected role of a previously uncharacterized factor, Sif, as an inhibitor of the SOS response in L. monocytogenes.

13.
Microbiology (Reading) ; 158(Pt 4): 963-974, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22282521

RESUMO

The Gram-positive bacterium Listeria monocytogenes is widely distributed in the environment and capable of causing food-borne infections in susceptible individuals. In this study, we investigated the cell envelope stress response in L. monocytogenes. Whole-genome transcriptional profiling was performed to investigate the response upon exposure to the cell wall antibiotic cefuroxime. Differential expression (at least twofold) of 558 genes was observed, corresponding to 20 % of the L. monocytogenes genome. The majority of genes that were strongly induced by cefuroxime exposure have cell-envelope-related functions, including the dlt operon and the gene encoding penicillin-binding protein PBPD2. A large overlap was observed between the cefuroxime stimulon and genes known to be induced in L. monocytogenes in blood and during intracellular infection, indicating that the cell envelope stress response is active at various stages of the infectious process. We analysed the roles of the two-component systems LisRK and CesRK in the cell envelope response, showing that activation of the most highly cefuroxime-induced genes was LisR- and CesR-dependent. In addition, multiple VirRS- and LiaSR-regulated genes were found to be induced in response to cefuroxime exposure. In total, 53 % of the genes upregulated at least fourfold by cefuroxime exposure are under positive control by one of the four two-component systems. Using genetic analyses, we showed that several genes of the cefuroxime stimulon contribute to the innate resistance of L. monocytogenes to cefuroxime and tolerance to other cell-envelope-perturbing conditions. Collectively, these findings demonstrate central roles for two-component systems in orchestrating the cell envelope stress response in L. monocytogenes.


Assuntos
Proteínas de Bactérias/metabolismo , Cefuroxima/farmacologia , Parede Celular/fisiologia , Listeria monocytogenes/fisiologia , Proteínas Quinases/metabolismo , Fatores de Transcrição/metabolismo , Ampicilina/farmacologia , Proteínas de Bactérias/genética , Sítios de Ligação , Perfilação da Expressão Gênica , Regulação Bacteriana da Expressão Gênica , Gentamicinas/farmacologia , Listeria monocytogenes/genética , Listeria monocytogenes/metabolismo , Testes de Sensibilidade Microbiana , Proteínas Quinases/genética , Deleção de Sequência , Fatores de Transcrição/genética , Transcriptoma
14.
Nucleic Acids Res ; 38(3): 907-19, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19942685

RESUMO

Small trans-encoded RNAs (sRNAs) modulate the translation and decay of mRNAs in bacteria. In Gram-negative species, antisense regulation by trans-encoded sRNAs relies on the Sm-like protein Hfq. In contrast to this, Hfq is dispensable for sRNA-mediated riboregulation in the Gram-positive species studied thus far. Here, we provide evidence for Hfq-dependent translational repression in the Gram-positive human pathogen Listeria monocytogenes, which is known to encode at least 50 sRNAs. We show that the Hfq-binding sRNA LhrA controls the translation and degradation of its target mRNA by an antisense mechanism, and that Hfq facilitates the binding of LhrA to its target. The work presented here provides the first experimental evidence for Hfq-dependent riboregulation in a Gram-positive bacterium. Our findings indicate that modulation of translation by trans-encoded sRNAs may occur by both Hfq-dependent and -independent mechanisms, thus adding another layer of complexity to sRNA-mediated riboregulation in Gram-positive species.


Assuntos
Regulação Bacteriana da Expressão Gênica , Fator Proteico 1 do Hospedeiro/fisiologia , Listeria monocytogenes/genética , RNA Antissenso/química , RNA não Traduzido/química , Pareamento de Bases , Sequência de Bases , Fator Proteico 1 do Hospedeiro/genética , Fator Proteico 1 do Hospedeiro/metabolismo , Listeria monocytogenes/metabolismo , Dados de Sequência Molecular , Mutação , Iniciação Traducional da Cadeia Peptídica , Biossíntese de Proteínas , Estabilidade de RNA , RNA Antissenso/genética , RNA Mensageiro/química , RNA Mensageiro/metabolismo , RNA não Traduzido/genética
15.
Front Microbiol ; 13: 897682, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35633716

RESUMO

Free fatty acids (FFAs) have strong antimicrobial properties against pathogenic bacteria and are known as natural protective agents against bacterial infections. Growth of the foodborne pathogen Listeria monocytogenes is highly affected by the presence of antimicrobial FFAs, however, the response of L. monocytogenes toward FFAs is not fully understood. Here, we explore how L. monocytogenes gains tolerance toward FFAs and present a novel mechanism conferring bacterial protection against FFA toxicity. Strains tolerant against the antimicrobial FFA palmitoleic acid were isolated and whole genome sequenced, and mutations were found in genes involved in wall teichoic acid (WTA) glycosylations. We show that mutation or deletion of lmo1079, which is essential for N-acetylglucosamine (GlcNAc) glycosylation of WTAs, confer tolerance against several antimicrobial FFAs. The FFA tolerant strains are lacking GlcNAc on their WTAs, which result in a more hydrophilic surface. In line with this, we observed a reduced binding of FFAs to the surface of the FFA tolerant strains. Additionally, lack of GlcNAc on WTAs confers tolerance toward acid stress. Altogether, these findings support that GlcNAc modification of WTA plays an important role in the response of L. monocytogenes toward stress conditions encountered during growth as a saprophyte and pathogen, including FFA-rich environments. Most importantly, our data revealed that L. monocytogenes strains lacking GlcNAc on their WTAs are protected against FFA toxicity, because the FFAs are repulsed from the bacterial surface of GlcNAc-deficient strains.

16.
Front Microbiol ; 13: 895942, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35591996

RESUMO

Free fatty acids (FFAs) are known to exhibit antimicrobial and anti-virulent properties against bacterial pathogens. Specific FFAs, such as lauric acid (LA; C12:0), exert both effects against the foodborne pathogen Listeria monocytogenes: at low levels, LA acts to inhibit the activity of the virulence regulator PrfA, whereas at higher levels, LA inhibits bacterial growth. Deletion of prfA is known to promote tolerance toward antimicrobial FFAs, suggesting that the response of L. monocytogenes to anti-virulent and antimicrobial FFAs could be linked. In this study, we explored the response of L. monocytogenes toward antimicrobial FFAs holding an anti-virulence activity by isolating strains that can grow at high concentrations of LA. We found that LA-tolerant isolates carry mutations in the gene encoding the global regulator CcpA. Importantly, we discovered that mutation or deletion of ccpA protect L. monocytogenes against the antimicrobial activity of FFAs, whereas the ccpA mutants remain sensitive toward FFA's PrfA inhibitory effect. A regulatory link involving CcpA, connecting the response toward the antimicrobial and anti-virulence activities of FFAs, is therefore unlikely. To further study how deletion of ccpA promotes FFA tolerance, we performed a transcriptomic analysis of the response to LA. Our data indicated that the FFA-tolerant phenotype of the ∆ccpA strain is not induced upon LA exposure but appears to be an inherent phenotypic trait of the ccpA deletion mutation. Interestingly, we found that the bacterial surface of L. monocytogenes becomes more hydrophilic upon deletion of ccpA, and we demonstrate that CcpA plays a role in the response of L. monocytogenes to other stress conditions, including low pH and antibiotics. Altogether, our study revealed that regulatory activities of CcpA lead to an increased hydrophobicity of the bacterial surface, which may confer sensitivity of L. monocytogenes against the antimicrobial activity of FFAs. Notably, CcpA is not involved in responding to the PrfA inhibitory effect of FFAs, showing that FFA-tolerant strains can still be targeted by the anti-virulent activity of FFAs.

17.
J Biol Chem ; 285(14): 10690-702, 2010 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-20075074

RESUMO

Small non-coding RNAs (sRNA) have emerged as important elements of gene regulatory circuits. In enterobacteria such as Escherichia coli and Salmonella many of these sRNAs interact with the Hfq protein, an RNA chaperone similar to mammalian Sm-like proteins and act in the post-transcriptional regulation of many genes. A number of these highly conserved ribo-regulators are stringently regulated at the level of transcription and are part of major regulons that deal with the immediate response to various stress conditions, indicating that every major transcription factor may control the expression of at least one sRNA regulator. Here, we extend this view by the identification and characterization of a highly conserved, anaerobically induced small sRNA in E. coli, whose expression is strictly dependent on the anaerobic transcriptional fumarate and nitrate reductase regulator (FNR). The sRNA, named FnrS, possesses signatures of base-pairing RNAs, and we show by employing global proteomic and transcriptomic profiling that the expression of multiple genes is negatively regulated by the sRNA. Intriguingly, many of these genes encode enzymes with "aerobic" functions or enzymes linked to oxidative stress. Furthermore, in previous work most of the potential target genes have been shown to be repressed by FNR through an undetermined mechanism. Collectively, our results provide insight into the mechanism by which FNR negatively regulates genes such as sodA, sodB, cydDC, and metE, thereby demonstrating that adaptation to anaerobic growth involves the action of a small regulatory RNA.


Assuntos
Anaerobiose/fisiologia , Escherichia coli/genética , Regulação Bacteriana da Expressão Gênica , Biossíntese de Proteínas , RNA não Traduzido/fisiologia , Aerobiose , Pareamento de Bases , Sequência de Bases , Biomarcadores/metabolismo , Northern Blotting , Western Blotting , Eletroforese em Gel Bidimensional , Ensaio de Desvio de Mobilidade Eletroforética , Escherichia coli/metabolismo , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Perfilação da Expressão Gênica , Dados de Sequência Molecular , Análise de Sequência com Séries de Oligonucleotídeos , Processamento Pós-Transcricional do RNA , RNA Bacteriano/genética , RNA Bacteriano/metabolismo , Homologia de Sequência do Ácido Nucleico , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Transcrição Gênica
18.
Arch Microbiol ; 193(1): 23-34, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20978742

RESUMO

In recent years, small RNAs (sRNAs) have been identified as important regulators of gene expression in bacteria. Most sRNAs are encoded from intergenic regions and are only expressed under highly specific growth conditions. In Staphylococcus aureus, the alternative sigma factor, σ(B), is known to contribute to the overall stress response, antibiotic resistance, and virulence. The σ(B) regulon in S. aureus is well described and comprises approximately 200 annotated genes, including several genes encoding virulence factors. In the present study, we have identified three novel σ(B)-dependent transcripts encoded from genomic regions previously annotated as intergenic. All three transcripts, named SbrA, SbrB, and SbrC, are highly conserved in S. aureus, and we confirmed their presence in four different isolates (SH1000, Newman, COL, and UAMS-1). Curiously, two of these genes (sbrA and sbrB) were found to contain open reading frames encoding small, highly basic peptides that are conserved among Staphylococci. The third transcript (SbrC) did not contain any likely open reading frame and thus constitute a genuine non-coding sRNA. The functions of these genes are currently unknown but are likely to be important for the σ(B)-mediated response of S. aureus to adverse conditions.


Assuntos
Regulação Bacteriana da Expressão Gênica , RNA Bacteriano/genética , Fator sigma/metabolismo , Staphylococcus aureus/genética , Staphylococcus aureus/metabolismo , Sequência de Aminoácidos , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Biologia Computacional , DNA Intergênico/genética , Fator Proteico 1 do Hospedeiro/genética , Fator Proteico 1 do Hospedeiro/metabolismo , Dados de Sequência Molecular , Pequeno RNA não Traduzido/genética , Alinhamento de Sequência , Fator sigma/genética , Sítio de Iniciação de Transcrição
19.
Front Microbiol ; 12: 622829, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33935989

RESUMO

Listeria monocytogenes is an intracellular pathogen that is well known for its adaptability to life in a broad spectrum of different niches. RNA-mediated regulatory mechanisms in L. monocytogenes play important roles in successful adaptation providing fast and versatile responses to a changing environment. Recent findings indicate that non-coding RNAs (ncRNAs) regulate a variety of processes in this bacterium, such as environmental sensing, metabolism and virulence, as well as immune responses in eukaryotic cells. In this review, the current knowledge on RNA-mediated regulation in L. monocytogenes is presented, with special focus on the roles and mechanisms underlying modulation of metabolism and virulence. Collectively, these findings point to ncRNAs as important gene regulatory elements in L. monocytogenes, both outside and inside an infected host. However, the involvement of regulatory ncRNAs in bacterial physiology and virulence is still underestimated and probably will be better assessed in the coming years, especially in relation to discovering the regulatory functions of 5' and 3' untranslated regions and excludons, and by exploring the role of ncRNAs in interaction with both bacterial and host proteins.

20.
Front Microbiol ; 12: 625585, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34046019

RESUMO

Three out of the seven ribosomal RNA operons in Escherichia coli end in dual terminator structures. Between the two terminators of each operon is a short sequence that we report here to be an sRNA gene, transcribed as part of the ribosomal RNA primary transcript by read-through of the first terminator. The sRNA genes (rrA, rrB and rrF) from the three operons (rrnA, rrnB and rrnD) are more than 98% identical, and pull-down experiments show that their transcripts interact with Hfq and CsrA. Deletion of rrA, B, F, as well as overexpression of rrB, only modestly affect known CsrA-regulated phenotypes like biofilm formation, pgaA translation and glgC translation, and the role of the sRNAs in vivo may not yet be fully understood. Since RrA, B, F are short-lived and transcribed along with the ribosomal RNA components, their concentration reflect growth-rate regulation at the ribosomal RNA promoters and they could function to fine-tune other growth-phase-dependent processes in the cell. The primary and secondary structure of these small RNAs are conserved among species belonging to different genera of Enterobacteriales.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA