Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 139(4): 770-9, 2009 Nov 13.
Artigo em Inglês | MEDLINE | ID: mdl-19914169

RESUMO

Riboswitches are RNA elements acting in cis, controlling expression of their downstream genes through a metabolite-induced alteration of their secondary structure. Here, we demonstrate that two S-adenosylmethionine (SAM) riboswitches, SreA and SreB, can also function in trans and act as noncoding RNAs in Listeria monocytogenes. SreA and SreB control expression of the virulence regulator PrfA by binding to the 5'-untranslated region of its mRNA. Absence of the SAM riboswitches SreA and SreB increases the level of PrfA and virulence gene expression in L. monocytogenes. Thus, the impact of the SAM riboswitches on PrfA expression highlights a link between bacterial virulence and nutrient availability. Together, our results uncover an unexpected role for riboswitches and a distinct class of regulatory noncoding RNAs in bacteria.


Assuntos
Proteínas de Bactérias/genética , Listeria monocytogenes/genética , Fatores de Terminação de Peptídeos/genética , Sequências Reguladoras de Ácido Ribonucleico , Regiões 5' não Traduzidas , Sequência de Bases , Escherichia coli/genética , Escherichia coli/metabolismo , Listeria monocytogenes/metabolismo , Listeria monocytogenes/patogenicidade , Dados de Sequência Molecular , Temperatura , Virulência
2.
Proc Natl Acad Sci U S A ; 118(40)2021 10 05.
Artigo em Inglês | MEDLINE | ID: mdl-34599102

RESUMO

Listeriolysin S (LLS) is a thiazole/oxazole-modified microcin (TOMM) produced by hypervirulent clones of Listeria monocytogenes LLS targets specific gram-positive bacteria and modulates the host intestinal microbiota composition. To characterize the mechanism of LLS transfer to target bacteria and its bactericidal function, we first investigated its subcellular distribution in LLS-producer bacteria. Using subcellular fractionation assays, transmission electron microscopy, and single-molecule superresolution microscopy, we identified that LLS remains associated with the bacterial cell membrane and cytoplasm and is not secreted to the bacterial extracellular space. Only living LLS-producer bacteria (and not purified LLS-positive bacterial membranes) display bactericidal activity. Applying transwell coculture systems and microfluidic-coupled microscopy, we determined that LLS requires direct contact between LLS-producer and -target bacteria in order to display bactericidal activity, and thus behaves as a contact-dependent bacteriocin. Contact-dependent exposure to LLS leads to permeabilization/depolarization of the target bacterial cell membrane and adenosine triphosphate (ATP) release. Additionally, we show that lipoteichoic acids (LTAs) can interact with LLS and that LTA decorations influence bacterial susceptibility to LLS. Overall, our results suggest that LLS is a TOMM that displays a contact-dependent inhibition mechanism.


Assuntos
Bacteriocinas/metabolismo , Membrana Celular/metabolismo , Proteínas Hemolisinas/metabolismo , Listeria monocytogenes/metabolismo , Trifosfato de Adenosina/metabolismo , Citoplasma/metabolismo
3.
Cell Microbiol ; 22(4): e13183, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32185895

RESUMO

Decades of breakthroughs resulting from cross feeding of microbiological research and technological innovation have promoted Listeria monocytogenes to the rank of model microorganism to study host-pathogen interactions. The extraordinary capacity of this bacterium to interfere with a vast array of host cellular processes uncovered new concepts in microbiology, cell biology and infection biology. Here, we review technological advances that revealed how bacteria and host interact in space and time at the molecular, cellular, tissue and whole body scales, ultimately revolutionising our understanding of Listeria pathogenesis. With the current bloom of multidisciplinary integrative approaches, Listeria entered a new microbiology era.


Assuntos
Biotecnologia , Interações Hospedeiro-Patógeno , Listeria monocytogenes/patogenicidade , Listeriose/microbiologia , Animais , Proteínas de Bactérias , Regulação Bacteriana da Expressão Gênica , Humanos , Listeria monocytogenes/genética , Camundongos , Virulência , Fatores de Virulência
4.
Molecules ; 25(21)2020 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-33105870

RESUMO

Nicotinamide adenine dinucleotide (NAD) kinases are essential and ubiquitous enzymes involved in the tight regulation of NAD/nicotinamide adenine dinucleotide phosphate (NADP) levels in many metabolic pathways. Consequently, they represent promising therapeutic targets in cancer and antibacterial treatments. We previously reported diadenosine derivatives as NAD kinase inhibitors with bactericidal activities on Staphylococcus aureus. Among them, one compound (namely NKI1) was found effective in vivo in a mouse infection model. With the aim to gain detailed knowledge about the selectivity and mechanism of action of this lead compound, we planned to develop a chemical probe that could be used in affinity-based chemoproteomic approaches. Here, we describe the first functionalized chemical probe targeting a bacterial NAD kinase. Aminoalkyl functional groups were introduced on NKI1 for further covalent coupling to an activated SepharoseTM matrix. Inhibitory properties of functionalized NKI1 derivatives together with X-ray characterization of their complexes with the NAD kinase led to identify candidate compounds that are amenable to covalent coupling to a matrix.


Assuntos
Adenina/análogos & derivados , Adenosina/síntese química , Antibacterianos/síntese química , Proteínas de Bactérias/antagonistas & inibidores , Inibidores Enzimáticos/síntese química , Fosfotransferases (Aceptor do Grupo Álcool)/antagonistas & inibidores , Adenina/síntese química , Adenina/farmacologia , Adenosina/farmacologia , Sequência de Aminoácidos , Animais , Antibacterianos/farmacologia , Modelos Animais de Doenças , Inibidores Enzimáticos/farmacologia , Camundongos , Modelos Moleculares , NADP/química , Conformação Proteica , Sefarose/química , Staphylococcus aureus
5.
Genes Immun ; 20(5): 357-370, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30940874

RESUMO

Plague is a vector-borne disease caused by Yersinia pestis. Transmitted by fleas from rodent reservoirs, Y. pestis emerged <6000 years ago from an enteric bacterial ancestor through events of gene gain and genome reduction. It is a highly remarkable model for the understanding of pathogenic bacteria evolution, and a major concern for public health as highlighted by recent human outbreaks. A complex set of virulence determinants, including the Yersinia outer-membrane proteins (Yops), the broad-range protease Pla, pathogen-associated molecular patterns (PAMPs), and iron capture systems play critical roles in the molecular strategies that Y. pestis employs to subvert the human immune system, allowing unrestricted bacterial replication in lymph nodes (bubonic plague) and in lungs (pneumonic plague). Some of these immunogenic proteins as well as the capsular antigen F1 are exploited for diagnostic purposes, which are critical in the context of the rapid onset of death in the absence of antibiotic treatment (less than a week for bubonic plague and <48 h for pneumonic plague). Here, we review recent research advances on Y. pestis evolution, virulence factor function, bacterial strategies to subvert mammalian innate immune responses, vaccination, and problems associated with pneumonic plague diagnosis.


Assuntos
Peste/microbiologia , Yersinia pestis/patogenicidade , Animais , Evolução Molecular , Humanos , Peste/diagnóstico , Peste/epidemiologia , Peste/terapia , Fatores de Virulência/genética , Yersinia pestis/genética , Yersinia pestis/imunologia
6.
Proc Natl Acad Sci U S A ; 113(20): 5706-11, 2016 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-27140611

RESUMO

Listeria monocytogenes is responsible for gastroenteritis in healthy individuals and for a severe invasive disease in immunocompromised patients. Among the three identified L. monocytogenes evolutionary lineages, lineage I strains are overrepresented in epidemic listeriosis outbreaks, but the mechanisms underlying the higher virulence potential of strains of this lineage remain elusive. Here, we demonstrate that Listeriolysin S (LLS), a virulence factor only present in a subset of lineage I strains, is a bacteriocin highly expressed in the intestine of orally infected mice that alters the host intestinal microbiota and promotes intestinal colonization by L. monocytogenes, as well as deeper organ infection. To our knowledge, these results therefore identify LLS as the first bacteriocin described in L. monocytogenes and associate modulation of host microbiota by L. monocytogenes epidemic strains to increased virulence.


Assuntos
Bacteriocinas/metabolismo , Microbioma Gastrointestinal , Listeria monocytogenes/fisiologia , Listeriose/microbiologia , Animais , Epidemias , Feminino , Interações Hospedeiro-Patógeno , Humanos , Listeria monocytogenes/patogenicidade , Listeriose/epidemiologia , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Viabilidade Microbiana , Virulência
7.
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
8.
EMBO J ; 30(14): 2934-47, 2011 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-21685873

RESUMO

The gut is a major barrier against microbes and encloses various innate lymphoid cells (ILCs), including two subsets expressing the natural cytotoxicity receptor NKp46. A subset of NKp46(+) cells expresses retinoic acid receptor-related orphan receptor γt (RORγt) and produces IL-22, like lymphoid tissue inducer (LTi) cells. Other NKp46(+) cells lack RORγt and produce IFN-γ, like conventional Natural Killer (cNK) cells. The identity, the regulation and the in vivo functions of gut NKp46(+) ILCs largely remain to be unravelled. Using pan-genomic profiling, we showed here that small intestine (SI) NKp46(+)RORγt(-) ILCs correspond to SI NK cells. Conversely, we identified a transcriptional programme conserved in fetal LTi cells and adult SI NKp46(+)RORγt(+) and NKp46(-)RORγt(+) ILCs. We also demonstrated that the IL-1ß/IL-1R1/MyD88 pathway, but not the commensal flora, drove IL-22 production by NKp46(+)RORγt(+) ILCs. Finally, oral Listeria monocytogenes infection induced IFN-γ production in SI NK and IL-22 production in NKp46(+)RORγt(+) ILCs, but only IFN-γ contributed to control bacteria dissemination. NKp46(+) ILC heterogeneity is thus associated with subset-specific transcriptional programmes and effector functions that govern their implication in gut innate immunity.


Assuntos
Linhagem da Célula , Imunidade Inata , Linfócitos/metabolismo , Linfócitos/microbiologia , Receptor 1 Desencadeador da Citotoxicidade Natural/metabolismo , Receptores do Ácido Retinoico/metabolismo , Animais , Feminino , Citometria de Fluxo , Intestino Delgado/imunologia , Intestino Delgado/metabolismo , Intestino Delgado/microbiologia , Listeria monocytogenes/isolamento & purificação , Listeriose/metabolismo , Listeriose/microbiologia , Linfócitos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/fisiologia , Receptor 1 Desencadeador da Citotoxicidade Natural/genética , Receptores de Interleucina-1/fisiologia , Receptores do Ácido Retinoico/genética , Distribuição Tecidual , Receptor gama de Ácido Retinoico
9.
Nature ; 459(7249): 950-6, 2009 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-19448609

RESUMO

The bacterium Listeria monocytogenes is ubiquitous in the environment and can lead to severe food-borne infections. It has recently emerged as a multifaceted model in pathogenesis. However, how this bacterium switches from a saprophyte to a pathogen is largely unknown. Here, using tiling arrays and RNAs from wild-type and mutant bacteria grown in vitro, ex vivo and in vivo, we have analysed the transcription of its entire genome. We provide the complete Listeria operon map and have uncovered far more diverse types of RNAs than expected: in addition to 50 small RNAs (<500 nucleotides), at least two of which are involved in virulence in mice, we have identified antisense RNAs covering several open-reading frames and long overlapping 5' and 3' untranslated regions. We discovered that riboswitches can act as terminators for upstream genes. When Listeria reaches the host intestinal lumen, an extensive transcriptional reshaping occurs with a SigB-mediated activation of virulence genes. In contrast, in the blood, PrfA controls transcription of virulence genes. Remarkably, several non-coding RNAs absent in the non-pathogenic species Listeria innocua exhibit the same expression patterns as the virulence genes. Together, our data unravel successive and coordinated global transcriptional changes during infection and point to previously unknown regulatory mechanisms in bacteria.


Assuntos
Regulação Bacteriana da Expressão Gênica , Listeria monocytogenes/genética , Listeria monocytogenes/patogenicidade , RNA Bacteriano/genética , Transcrição Gênica/genética , Animais , Genes Bacterianos/genética , Genoma Bacteriano/genética , Intestinos/microbiologia , Camundongos , Fases de Leitura Aberta/genética , Óperon/genética , RNA Bacteriano/análise , Sequências Reguladoras de Ácido Ribonucleico/genética , Regiões não Traduzidas/genética , Virulência/genética
10.
Nature ; 455(7216): 1114-8, 2008 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-18806773

RESUMO

The ability to cross host barriers is an essential virulence determinant of invasive microbial pathogens. Listeria monocytogenes is a model microorganism that crosses human intestinal and placental barriers, and causes severe maternofetal infections by an unknown mechanism. Several studies have helped to characterize the bacterial invasion proteins InlA and InlB. However, their respective species specificity has complicated investigations on their in vivo role. Here we describe two novel and complementary animal models for human listeriosis: the gerbil, a natural host for L. monocytogenes, and a knock-in mouse line ubiquitously expressing humanized E-cadherin. Using these two models, we uncover the essential and interdependent roles of InlA and InlB in fetoplacental listeriosis, and thereby decipher the molecular mechanism underlying the ability of a microbe to target and cross the placental barrier.


Assuntos
Proteínas de Bactérias/metabolismo , Doenças Fetais/microbiologia , Listeria monocytogenes/fisiologia , Listeriose/transmissão , Troca Materno-Fetal , Proteínas de Membrana/metabolismo , Doenças Placentárias/microbiologia , Animais , Proteínas de Bactérias/genética , Caderinas/genética , Células Cultivadas , Modelos Animais de Doenças , Enterócitos/microbiologia , Células Epiteliais/microbiologia , Feminino , Gerbillinae , Humanos , Listeriose/microbiologia , Proteínas de Membrana/genética , Camundongos , Dados de Sequência Molecular , Gravidez , Complicações Infecciosas na Gravidez/metabolismo , Complicações Infecciosas na Gravidez/microbiologia , Ligação Proteica , Receptores de Fatores de Crescimento/metabolismo , Especificidade da Espécie
11.
Microbes Infect ; 26(4): 105334, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38556158

RESUMO

Global burden of infectious diseases and antimicrobial resistance are major public health issues calling for innovative control measures. Bacterial NAD kinase (NADK) is a crucial enzyme for production of NADP(H) and growth. In Staphylococcus aureus, NADK promotes pathogenesis by supporting production of key virulence determinants. Here, we find that knockdown of NADK by CRISPR interference sensitizes S. aureus to osmotic stress and to stresses induced by antibiotics targeting the envelop as well as replication, transcription and translation. Thus, NADK represents a promising target for the development of inhibitors which could be used in combination with current antibiotics.


Assuntos
Antibacterianos , Fosfotransferases (Aceptor do Grupo Álcool) , Staphylococcus aureus , Antibacterianos/farmacologia , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/genética , Regulação Bacteriana da Expressão Gênica , Técnicas de Silenciamento de Genes , Pressão Osmótica , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Staphylococcus aureus/enzimologia , Staphylococcus aureus/genética , Staphylococcus aureus/efeitos dos fármacos , Estresse Fisiológico
12.
FEMS Microbiol Rev ; 48(3)2024 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-38734892

RESUMO

Bloodstream infection is a major public health concern associated with high mortality and high healthcare costs worldwide. Bacteremia can trigger fatal sepsis whose prevention, diagnosis, and management have been recognized as a global health priority by the World Health Organization. Additionally, infection control is increasingly threatened by antimicrobial resistance, which is the focus of global action plans in the framework of a One Health response. In-depth knowledge of the infection process is needed to develop efficient preventive and therapeutic measures. The pathogenesis of bloodstream infection is a dynamic process resulting from the invasion of the vascular system by bacteria, which finely regulate their metabolic pathways and virulence factors to overcome the blood immune defenses and proliferate. In this review, we highlight our current understanding of determinants of bacterial survival and proliferation in the bloodstream and discuss their interactions with the molecular and cellular components of blood.


Assuntos
Bactérias , Humanos , Bacteriemia/microbiologia , Fatores de Virulência , Sangue/microbiologia , Viabilidade Microbiana
13.
PLoS Pathog ; 7(8): e1002168, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21829365

RESUMO

L. monocytogenes is a facultative intracellular bacterium responsible for listeriosis. It is able to invade, survive and replicate in phagocytic and non-phagocytic cells. The infectious process at the cellular level has been extensively studied and many virulence factors have been identified. Yet, the role of InlK, a member of the internalin family specific to L. monocytogenes, remains unknown. Here, we first show using deletion analysis and in vivo infection, that InlK is a bona fide virulence factor, poorly expressed in vitro and well expressed in vivo, and that it is anchored to the bacterial surface by sortase A. We then demonstrate by a yeast two hybrid screen using InlK as a bait, validated by pulldown experiments and immunofluorescence analysis that intracytosolic bacteria via an interaction with the protein InlK interact with the Major Vault Protein (MVP), the main component of cytoplasmic ribonucleoproteic particules named vaults. Although vaults have been implicated in several cellular processes, their role has remained elusive. Our analysis demonstrates that MVP recruitment disguises intracytosolic bacteria from autophagic recognition, leading to an increased survival rate of InlK over-expressing bacteria compared to InlK(-) bacteria. Together these results reveal that MVP is hijacked by L. monocytogenes in order to counteract the autophagy process, a finding that could have major implications in deciphering the cellular role of vault particles.


Assuntos
Autofagia , Proteínas de Bactérias/metabolismo , Listeria monocytogenes/metabolismo , Listeriose/metabolismo , Partículas de Ribonucleoproteínas em Forma de Abóbada/metabolismo , Fatores de Virulência/metabolismo , Aminoaciltransferases/genética , Aminoaciltransferases/metabolismo , Animais , Proteínas de Bactérias/genética , Cisteína Endopeptidases/genética , Cisteína Endopeptidases/metabolismo , Feminino , Células HEK293 , Células HeLa , Humanos , Listeria monocytogenes/genética , Listeriose/genética , Camundongos , Camundongos Endogâmicos BALB C , Ligação Proteica , Técnicas do Sistema de Duplo-Híbrido , Partículas de Ribonucleoproteínas em Forma de Abóbada/genética , Fatores de Virulência/genética
14.
Proc Natl Acad Sci U S A ; 107(40): 17333-8, 2010 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-20855622

RESUMO

Listeria monocytogenes is an intracellular pathogen responsible for severe foodborne infections. It can replicate in both phagocytic and nonphagocytic mammalian cells. The infectious process at the cellular level has been studied extensively, but how the bacterium overcomes early host innate immune responses remains largely unknown. Here we show that InlC, a member of the internalin family, is secreted intracellularly and directly interacts with IKKα, a subunit of the IκB kinase complex critical for the phosphorylation of IκB and activation of NF-κB, the major regulator of innate immune responses. Infection experiments with WT Listeria or the inlC-deletion mutant and transfection of cells with InlC reveal that InlC expression impairs phosphorylation and consequently delays IκB degradation normally induced by TNF-α, a classical NF-κB stimulator. Moreover, infection of RAW 264.7 macrophages by the inlC mutant leads to increased production of proinflammatory cytokines compared with that obtained with the WT. Finally, in a peritonitis mouse model, we show that infection with the inlC mutant induces increased production of chemokines and increased recruitment of neutrophils in the peritoneal cavity compared with infection with WT. Together, these results demonstrate that InlC, by interacting with IKKα, dampens the host innate response induced by Listeria during the infection process.


Assuntos
Proteínas de Bactérias/imunologia , Quinase I-kappa B/metabolismo , Imunidade Inata , Subunidades Proteicas/metabolismo , Animais , Linhagem Celular , Humanos , Quinase I-kappa B/genética , Listeria monocytogenes/imunologia , Listeria monocytogenes/patogenicidade , Camundongos , Regiões Promotoras Genéticas , Subunidades Proteicas/genética , Fator de Necrose Tumoral alfa/metabolismo , Técnicas do Sistema de Duplo-Híbrido
15.
Microbiol Resour Announc ; 12(3): e0002423, 2023 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-36840571

RESUMO

Here, we report complete genome sequences of two clinical isolates of Staphylococcus aureus, namely, Xen31 and Xen36, which have been genetically modified to express an optimized Photorhabdus luminescens luciferase operon. Xen31 and Xen36 are bioluminescent strains used widely for investigation of bacterial pathogenesis, drug discovery, and development of novel therapies.

16.
Microbiol Resour Announc ; 12(11): e0083823, 2023 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-37906029

RESUMO

We report the complete genome sequence of Yersinia pseudotuberculosis strain SP-1303, identified as part of lineage 8 and associated with Far East scarlet-like fever. The genome includes the chromosome, the Yersinia-virulence plasmid (pYV) encoding a type III secretion system essential for virulence, the pVM82 plasmid, and two cryptic plasmids.

17.
Microbiol Spectr ; : e0382622, 2023 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-36847572

RESUMO

The genus Yersinia includes a large variety of nonpathogenic and life-threatening pathogenic bacteria, which cause a broad spectrum of diseases in humans and animals, such as plague, enteritis, Far East scarlet-like fever (FESLF), and enteric redmouth disease. Like most clinically relevant microorganisms, Yersinia spp. are currently subjected to intense multi-omics investigations whose numbers have increased extensively in recent years, generating massive amounts of data useful for diagnostic and therapeutic developments. The lack of a simple and centralized way to exploit these data led us to design Yersiniomics, a web-based platform allowing straightforward analysis of Yersinia omics data. Yersiniomics contains a curated multi-omics database at its core, gathering 200 genomic, 317 transcriptomic, and 62 proteomic data sets for Yersinia species. It integrates genomic, transcriptomic, and proteomic browsers, a genome viewer, and a heatmap viewer to navigate within genomes and experimental conditions. For streamlined access to structural and functional properties, it directly links each gene to GenBank, the Kyoto Encyclopedia of Genes and Genomes (KEGG), UniProt, InterPro, IntAct, and the Search Tool for the Retrieval of Interacting Genes/Proteins (STRING) and each experiment to Gene Expression Omnibus (GEO), the European Nucleotide Archive (ENA), or the Proteomics Identifications Database (PRIDE). Yersiniomics provides a powerful tool for microbiologists to assist with investigations ranging from specific gene studies to systems biology studies. IMPORTANCE The expanding genus Yersinia is composed of multiple nonpathogenic species and a few pathogenic species, including the deadly etiologic agent of plague, Yersinia pestis. In 2 decades, the number of genomic, transcriptomic, and proteomic studies on Yersinia grew massively, delivering a wealth of data. We developed Yersiniomics, an interactive web-based platform, to centralize and analyze omics data sets on Yersinia species. The platform allows user-friendly navigation between genomic data, expression data, and experimental conditions. Yersiniomics will be a valuable tool to microbiologists.

18.
Eur J Med Chem ; 246: 114941, 2023 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-36455355

RESUMO

Nicotinamide adenine dinucleotide kinases (NAD kinases) are essential and ubiquitous enzymes involved in the production of NADP(H) which is an essential cofactor in many metabolic pathways. Targeting NAD kinase (NADK), a rate limiting enzyme of NADP biosynthesis pathway, represents a new promising approach to treat bacterial infections. Previously, we have produced the first NADK inhibitor active against staphylococcal infection. From this linear di-adenosine derivative, namely NKI1, we designed macrocyclic analogues. Here, we describe the synthesis and evaluation of an original series of cyclic diadenosine derivatives as NADK inhibitors of two pathogenic bacteria, Listeria monocytogenes and Staphylococcus aureus. The nature and length of the link between the two adenosine units were examined leading to sub-micromolar inhibitors of NADK1 from L. monocytogenes, including its most potent in vitro inhibitor reported so far (with a 300-fold improvement compared to NKI1).


Assuntos
Adenosina , Fosfotransferases (Aceptor do Grupo Álcool) , NADP/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Adenosina/farmacologia , Relação Estrutura-Atividade , Bactérias/metabolismo
19.
J Infect Dis ; 204(5): 731-40, 2011 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-21844299

RESUMO

Microbial pathogens have evolved mechanisms to overcome immune responses and successfully infect their host. Here, we studied how Listeria monocytogenes evades immune detection by peptidoglycan (PGN) modification. By analyzing L. monocytogenes muropeptides, we detected O-acetylated muramic acid residues. We identified an O-acetyltransferase gene, oatA, in the L. monocytogenes genome sequence. Comparison of PGN from parental and isogenic oatA mutant strains showed that the O-acetyltransferase OatA O-acetylates Listeria PGN. We also found that PGN O-acetylation confers resistance to different types of antimicrobial compounds targeting bacterial cell wall such as lysozyme, ß-lactam antibiotics, and bacteriocins and that O-acetylation is required for Listeria growth in macrophages. Moreover, oatA mutant virulence is drastically affected in mice following intravenous or oral inoculation. In addition, the oatA mutant induced early secretion of proinflammatory cytokines and chemokines in vivo. These results suggest an important role for OatA in limiting innate immune responses and promoting bacterial survival in the infected host.


Assuntos
Acetiltransferases/imunologia , Citocinas/metabolismo , Listeria monocytogenes/imunologia , Listeriose/imunologia , Peptidoglicano/imunologia , Fatores de Virulência/imunologia , Acetilação , Acetiltransferases/genética , Animais , Linhagem Celular , Feminino , Humanos , Imunidade Inata , Dose Letal Mediana , Listeria monocytogenes/genética , Listeria monocytogenes/crescimento & desenvolvimento , Listeria monocytogenes/patogenicidade , Listeriose/genética , Fígado/metabolismo , Fígado/microbiologia , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Testes de Sensibilidade Microbiana , Ácidos Murâmicos/metabolismo , Peptidoglicano/química , Baço/microbiologia , Células Th1/metabolismo , Células Th2/metabolismo , Fatores de Virulência/genética
20.
Elife ; 112022 06 20.
Artigo em Inglês | MEDLINE | ID: mdl-35723663

RESUMO

Nicotinamide adenine dinucleotide phosphate (NADPH) is the primary electron donor for reductive reactions that are essential for the biosynthesis of major cell components in all organisms. Nicotinamide adenine dinucleotide kinase (NADK) is the only enzyme that catalyzes the synthesis of NADP(H) from NAD(H). While the enzymatic properties and physiological functions of NADK have been thoroughly studied, the role of NADK in bacterial pathogenesis remains unknown. Here, we used CRISPR interference to knock down NADK gene expression to address the role of this enzyme in Staphylococcus aureus pathogenic potential. We find that NADK inhibition drastically decreases mortality of zebrafish infected with S. aureus. Furthermore, we show that NADK promotes S. aureus survival in infected macrophages by protecting bacteria from antimicrobial defense mechanisms. Proteome-wide data analysis revealed that production of major virulence-associated factors is sustained by NADK. We demonstrate that NADK is required for expression of the quorum-sensing response regulator AgrA, which controls critical S. aureus virulence determinants. These findings support a key role for NADK in bacteria survival within innate immune cells and the host during infection.


Assuntos
Staphylococcus aureus , Fatores de Virulência , Animais , NAD/metabolismo , NADP/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Staphylococcus aureus/genética , Staphylococcus aureus/metabolismo , Fatores de Virulência/genética , Peixe-Zebra/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA