Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Trends Microbiol ; 2024 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-38443279

RESUMO

Many pathogens are hard to eradicate, even in the absence of genetically detectable antimicrobial resistance mechanisms and despite proven antibiotic susceptibility. The fraction of clonal bacteria that temporarily elude effective antibiotic treatments is commonly known as 'antibiotic persisters.' Over the past decade, there has been a growing body of research highlighting the pivotal role played by the cellular host in the development of persisters. In parallel, this research has also sought to elucidate the molecular mechanisms underlying the formation of intracellular antibiotic persisters and has demonstrated a prominent role for the bacterial stress response. However, questions remain regarding the conditions leading to the formation of stress-induced persisters among a clonal population of intracellular bacteria and despite an ostensibly uniform environment. In this opinion, following a brief review of the current state of knowledge regarding intracellular antibiotic persisters, we explore the ways in which macrophage functional heterogeneity and bacterial phenotypic heterogeneity may contribute to the emergence of these persisters. We propose that the degree of mismatch between the macrophage permissiveness and the bacterial preparedness to invade and thrive intracellularly may explain the formation of stress-induced nonreplicating intracellular persisters.

2.
Front Cell Infect Microbiol ; 13: 1141868, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37065203

RESUMO

The bulk of bacteria transiently evading appropriate antibiotic regimes and recovered from non-resolutive infections are commonly refer to as persisters. In this mini-review, we discuss how antibiotic persisters stem from the interplay between the pathogen and the cellular defenses mechanisms and its underlying heterogeneity.


Assuntos
Antibacterianos , Bactérias , Antibacterianos/farmacologia , Antibacterianos/uso terapêutico
3.
EMBO Rep ; 22(9): e52972, 2021 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-34314090

RESUMO

The Gram-negative bacterium Legionella pneumophila is the causative agent of Legionnaires' disease and replicates in amoebae and macrophages within a distinct compartment, the Legionella-containing vacuole (LCV). The facultative intracellular pathogen switches between a replicative, non-virulent and a non-replicating, virulent/transmissive phase. Here, we show on a single-cell level that at late stages of infection, individual motile (PflaA -GFP-positive) and virulent (PralF - and PsidC -GFP-positive) L. pneumophila emerge in the cluster of non-growing bacteria within an LCV. Comparative proteomics of PflaA -GFP-positive and PflaA -GFP-negative L. pneumophila subpopulations reveals distinct proteomes with flagellar proteins or cell division proteins being preferentially produced by the former or the latter, respectively. Toward the end of an infection cycle (˜ 48 h), the PflaA -GFP-positive L. pneumophila subpopulation emerges at the cluster periphery, predominantly escapes the LCV, and spreads from the bursting host cell. These processes are mediated by the Legionella quorum sensing (Lqs) system. Thus, quorum sensing regulates the emergence of a subpopulation of transmissive L. pneumophila at the LCV periphery, and phenotypic heterogeneity underlies the intravacuolar bi-phasic life cycle of L. pneumophila.


Assuntos
Legionella pneumophila , Legionella , Doença dos Legionários , Proteínas de Bactérias/genética , Humanos , Legionella/genética , Legionella pneumophila/genética , Percepção de Quorum , Vacúolos
4.
Infect Drug Resist ; 14: 1319-1324, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33854344

RESUMO

Bacteremia by Pandoraea spp. has rarely been described before. We report the first case of a P. pnomenusa possible prosthetic valve endocarditis, according to the modified Duke criteria, in a 37-year old male injecting drug user suffering from recurrent endocarditis. Furthermore, we demonstrate biofilm formation by the P. pnomenusa isolates of this patient and investigate antibiotic resistance.

5.
ISME J ; 15(1): 196-210, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32951019

RESUMO

The water-borne bacterium Legionella pneumophila is the causative agent of Legionnaires' disease. In the environment, the opportunistic pathogen colonizes different niches, including free-living protozoa and biofilms. The physiological state(s) of sessile Legionella in biofilms and their functional consequences are not well understood. Using single-cell techniques and fluorescent growth rate probes as well as promoter reporters, we show here that sessile L. pneumophila exhibits phenotypic heterogeneity and adopts growing and nongrowing ("dormant") states in biofilms and microcolonies. Phenotypic heterogeneity is controlled by the Legionella quorum sensing (Lqs) system, the transcription factor LvbR, and the temperature. The Lqs system and LvbR determine the ratio between growing and nongrowing sessile subpopulations, as well as the frequency of growth resumption ("resuscitation") and microcolony formation of individual bacteria. Nongrowing L. pneumophila cells are metabolically active, express virulence genes and show tolerance toward antibiotics. Therefore, these sessile nongrowers are persisters. Taken together, the Lqs system, LvbR and the temperature control the phenotypic heterogeneity of sessile L. pneumophila, and these factors regulate the formation of a distinct subpopulation of nongrowing, antibiotic tolerant, virulent persisters. Hence, the biofilm niche of L. pneumophila has a profound impact on the ecology and virulence of this opportunistic pathogen.


Assuntos
Legionella pneumophila , Legionella , Biofilmes , Legionella/genética , Legionella pneumophila/genética , Percepção de Quorum , Virulência
6.
Redox Biol ; 34: 101572, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32487458

RESUMO

The breach of proteostasis, leading to the accumulation of protein aggregates, is a hallmark of ageing and age-associated disorders, up to now well-established in neurodegeneration. Few studies have addressed the issue of dysfunctional cell response to protein deposition also for the cardiovascular system. However, the molecular basis of proteostasis decline in vascular cells, as well as its relation to ageing, are not understood. Recent studies have indicated the associations of Nrf2 transcription factor, the critical modulator of cellular stress-response, with ageing and premature senescence. In this report, we outline the significance of protein aggregation in physiological and premature ageing of murine and human endothelial cells (ECs). Our study shows that aged donor-derived and prematurely senescent Nrf2-deficient primary human ECs, but not those overexpressing dominant-negative Nrf2, exhibit increased accumulation of protein aggregates. Such phenotype is also found in the aortas of aged mice and young Nrf2 tKO mice. Ageing-related loss of proteostasis in ECs depends on Keap1, well-known repressor of Nrf2, recently perceived as a key independent regulator of EC function and protein S-nitrosation (SNO). Deposition of protein aggregates in ECs is associated with impaired autophagy. It can be counteracted by Keap1 depletion, S-nitrosothiol reductant or rapamycin treatment. Our results show that Keap1:Nrf2 protein balance and Keap1-dependent SNO predominate Nrf2 transcriptional activity-driven mechanisms in governing proteostasis in ageing ECs.


Assuntos
Fator 2 Relacionado a NF-E2 , Agregados Proteicos , Envelhecimento/genética , Animais , Células Endoteliais/metabolismo , Proteína 1 Associada a ECH Semelhante a Kelch/genética , Proteína 1 Associada a ECH Semelhante a Kelch/metabolismo , Camundongos , Fator 2 Relacionado a NF-E2/genética , Fator 2 Relacionado a NF-E2/metabolismo , Estresse Oxidativo
7.
Cell Microbiol ; 22(5): e13163, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-31945239

RESUMO

Mycobacterium marinum is a model organism for pathogenic Mycobacterium species, including Mycobacterium tuberculosis, the causative agent of tuberculosis. These pathogens enter phagocytes and replicate within the Mycobacterium-containing vacuole, possibly followed by vacuole exit and growth in the host cell cytosol. Mycobacteria release siderophores called mycobactins to scavenge iron, an essential yet poorly soluble and available micronutrient. To investigate the role of M. marinum mycobactins, we purified by organic solvent extraction and identified by mass spectrometry the lipid-bound mycobactin (MBT) and the water-soluble variant carboxymycobactin (cMBT). Moreover, we generated by specialised phage transduction a defined M. marinum ΔmbtB deletion mutant predicted to be defective for mycobactin production. The M. marinum ΔmbtB mutant strain showed a severe growth defect in broth and phagocytes, which was partially complemented by supplying the mbtB gene on a plasmid. Furthermore, purified Fe-MBT or Fe-cMBT improved the growth of wild type as well as ΔmbtB mutant bacteria on minimal plates, but only Fe-cMBT promoted the growth of wild-type M. marinum during phagocyte infection. Finally, the intracellular growth of M. marinum ΔmbtB in Acanthamoeba castellanii amoebae was restored by coinfection with wild-type bacteria. Our study identifies and characterises the M. marinum MBT and cMBT siderophores and reveals the requirement of mycobactins for extra- and intracellular growth of the pathogen.


Assuntos
Mycobacterium marinum/metabolismo , Oxazóis/metabolismo , Fagócitos/metabolismo , Sideróforos/biossíntese , Acanthamoeba castellanii/metabolismo , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Ferro/metabolismo , Espectrometria de Massas , Camundongos , Mycobacterium marinum/genética , Mycobacterium tuberculosis , Peptídeo Sintases/genética , Peptídeo Sintases/metabolismo , Células RAW 264.7 , Sideróforos/genética , Transcriptoma , Vacúolos/metabolismo
8.
Redox Biol ; 28: 101304, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31491600

RESUMO

Premature senescence, a death escaping pathway for cells experiencing stress, is conducive to aging and cardiovascular diseases. The molecular switch between senescent and apoptotic fate remains, however, poorly recognized. Nrf2 is an important transcription factor orchestrating adaptive response to cellular stress. Here, we show that both human primary endothelial cells (ECs) and murine aortas lacking Nrf2 signaling are senescent but unexpectedly do not encounter damaging oxidative stress. Instead, they exhibit markedly increased S-nitrosation of proteins. A functional role of S-nitrosation is protection of ECs from death by inhibition of NOX4-mediated oxidative damage and redirection of ECs to premature senescence. S-nitrosation and senescence are mediated by Keap1, a direct binding partner of Nrf2, which colocalizes and precipitates with nitric oxide synthase (NOS) and transnitrosating protein GAPDH in ECs devoid of Nrf2. We conclude that the overabundance of this "unrestrained" Keap1 determines the fate of ECs by regulation of S-nitrosation and propose that Keap1/GAPDH/NOS complex may serve as an enzymatic machinery for S-nitrosation in mammalian cells.


Assuntos
Aorta/citologia , Proteína 1 Associada a ECH Semelhante a Kelch/genética , Fator 2 Relacionado a NF-E2/genética , Animais , Aorta/metabolismo , Apoptose , Linhagem Celular , Senescência Celular , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Feminino , Técnicas de Inativação de Genes , Gliceraldeído-3-Fosfato Desidrogenase (Fosforiladora)/metabolismo , Humanos , Proteína 1 Associada a ECH Semelhante a Kelch/metabolismo , Masculino , Camundongos , Óxido Nítrico/metabolismo , Nitrosação , Transdução de Sinais , Adulto Jovem
9.
Nat Commun ; 10(1): 5216, 2019 11 18.
Artigo em Inglês | MEDLINE | ID: mdl-31740681

RESUMO

The facultative intracellular bacterium Legionella pneumophila replicates in environmental amoebae and in lung macrophages, and causes Legionnaires' disease. Here we show that L. pneumophila reversibly forms replicating and nonreplicating subpopulations of similar size within amoebae. The nonreplicating bacteria are viable and metabolically active, display increased antibiotic tolerance and a distinct proteome, and show high virulence as well as the capacity to form a degradation-resistant compartment. Upon infection of naïve or interferon-γ-activated macrophages, the nonreplicating subpopulation comprises ca. 10% or 50%, respectively, of the total intracellular bacteria; hence, the nonreplicating subpopulation is of similar size in amoebae and activated macrophages. The numbers of nonreplicating bacteria within amoebae are reduced in the absence of the autoinducer synthase LqsA or other components of the Lqs quorum-sensing system. Our results indicate that virulent, antibiotic-tolerant subpopulations of L. pneumophila are formed during infection of evolutionarily distant phagocytes, in a process controlled by the Lqs system.


Assuntos
Legionella/patogenicidade , Legionelose/microbiologia , Macrófagos/microbiologia , Percepção de Quorum , Amoeba/microbiologia , Animais , Proteínas de Bactérias/metabolismo , Espaço Intracelular/microbiologia , Legionella/crescimento & desenvolvimento , Camundongos , Viabilidade Microbiana , Proteoma/metabolismo , Vacúolos/microbiologia , Virulência
10.
PLoS Pathog ; 15(2): e1007551, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30730983

RESUMO

By engulfing potentially harmful microbes, professional phagocytes are continually at risk from intracellular pathogens. To avoid becoming infected, the host must kill pathogens in the phagosome before they can escape or establish a survival niche. Here, we analyse the role of the phosphoinositide (PI) 5-kinase PIKfyve in phagosome maturation and killing, using the amoeba and model phagocyte Dictyostelium discoideum. PIKfyve plays important but poorly understood roles in vesicular trafficking by catalysing formation of the lipids phosphatidylinositol (3,5)-bisphosphate (PI(3,5)2) and phosphatidylinositol-5-phosphate (PI(5)P). Here we show that its activity is essential during early phagosome maturation in Dictyostelium. Disruption of PIKfyve inhibited delivery of both the vacuolar V-ATPase and proteases, dramatically reducing the ability of cells to acidify newly formed phagosomes and digest their contents. Consequently, PIKfyve- cells were unable to generate an effective antimicrobial environment and efficiently kill captured bacteria. Moreover, we demonstrate that cells lacking PIKfyve are more susceptible to infection by the intracellular pathogen Legionella pneumophila. We conclude that PIKfyve-catalysed phosphoinositide production plays a crucial and general role in ensuring early phagosomal maturation, protecting host cells from diverse pathogenic microbes.


Assuntos
Dictyostelium/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Adenosina Trifosfatases , Animais , Linhagem Celular , Dictyostelium/patogenicidade , Humanos , Hidrolases/metabolismo , Legionella pneumophila/patogenicidade , Legionelose/metabolismo , Macrófagos , Fagocitose , Fagossomos , Fosfatidilinositol 3-Quinases/fisiologia , Fosfatidilinositóis , Transporte Proteico , Infecções por Protozoários/metabolismo
11.
Methods Mol Biol ; 1921: 191-204, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30694493

RESUMO

Legionella pneumophila resides in multispecies biofilms, where it infects and replicates in environmental protozoa such as Acanthamoeba castellanii. Studies on L. pneumophila physiology and host-pathogen interactions are frequently conducted using clonal bacterial populations and population level analysis, overlooking the remarkable differences in single cell behavior. The fastidious nutrient requirements of extracellular L. pneumophila and the extraordinary motility of Acanthamoeba castellanii hamper an analysis at single cell resolution. In this chapter, we describe a method to study L. pneumophila and its natural host A. castellanii at single cell level by using an agarose embedment assay. Agarose-embedded bacteria and infected cells can be monitored over several hours up to several days. Using properly adapted flow chambers, agarose-embedded specimens can be subjected to a wide range of fluctuating conditions.


Assuntos
Acanthamoeba castellanii/microbiologia , Interações Hospedeiro-Patógeno , Legionella/fisiologia , Análise de Célula Única/métodos , Microscopia de Fluorescência , Fagócitos/microbiologia , Fagocitose
12.
Curr Opin Microbiol ; 41: 29-35, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29190490

RESUMO

Legionella pneumophila is a water-borne opportunistic pathogen causing a life-threatening pneumonia called 'Legionnaires' disease'. The Legionella quorum sensing (Lqs) system produces and responds to the α-hydroxyketone signaling molecule 3-hydroxypentadecane-4-one (Legionella autoinducer-1, LAI-1). The Lqs system controls the switch between the replicative/non-virulent and the transmissive/virulent phase of L. pneumophila, and it is a major regulator of natural competence, motility and virulence of the pathogen. Yet, beyond gene regulation, LAI-1 also directly affects pathogen-host interactions, since the signaling molecule modulates the migration of eukaryotic cells. Genes encoding Lqs homologues are present in many environmental bacteria, suggesting that α-hydroxyketone signaling is widely used for inter-bacterial as well as inter-kingdom signaling. In this review we summarize recent advances on the characterization of the Lqs system and its role in L. pneumophila-host cell interactions.


Assuntos
Regulação Bacteriana da Expressão Gênica , Interações Hospedeiro-Patógeno/fisiologia , Legionella pneumophila/patogenicidade , Percepção de Quorum , 4-Butirolactona/análogos & derivados , 4-Butirolactona/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Movimento Celular , Interações Hospedeiro-Patógeno/genética , Humanos , Legionella , Legionella pneumophila/genética , Legionella pneumophila/metabolismo , Doença dos Legionários/microbiologia , Transdução de Sinais , Virulência/genética
13.
EMBO Rep ; 18(10): 1817-1836, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28835546

RESUMO

The pathogenic bacterium Legionella pneumophila replicates in host cells within a distinct ER-associated compartment termed the Legionella-containing vacuole (LCV). How the dynamic ER network contributes to pathogen proliferation within the nascent LCV remains elusive. A proteomic analysis of purified LCVs identified the ER tubule-resident large GTPase atlastin3 (Atl3, yeast Sey1p) and the reticulon protein Rtn4 as conserved LCV host components. Here, we report that Sey1/Atl3 and Rtn4 localize to early LCVs and are critical for pathogen vacuole formation. Sey1 overproduction promotes intracellular growth of L. pneumophila, whereas a catalytically inactive, dominant-negative GTPase mutant protein, or Atl3 depletion, restricts pathogen replication and impairs LCV maturation. Sey1 is not required for initial recruitment of ER to PtdIns(4)P-positive LCVs but for subsequent pathogen vacuole expansion. GTP (but not GDP) catalyzes the Sey1-dependent aggregation of purified, ER-positive LCVs in vitro Thus, Sey1/Atl3-dependent ER remodeling contributes to LCV maturation and intracellular replication of L. pneumophila.


Assuntos
Retículo Endoplasmático/fisiologia , Proteínas de Ligação ao GTP/metabolismo , Legionella pneumophila/crescimento & desenvolvimento , Proteínas de Membrana/metabolismo , Vacúolos/metabolismo , Vacúolos/microbiologia , Células A549 , Dictyostelium/microbiologia , Retículo Endoplasmático/microbiologia , Proteínas de Ligação ao GTP/genética , Humanos , Legionella pneumophila/patogenicidade , Macrófagos/microbiologia , Proteínas de Membrana/genética , Proteínas Nogo/genética , Proteínas Nogo/metabolismo , Proteômica , Sistemas de Secreção Tipo IV
14.
Trends Microbiol ; 24(6): 450-462, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26924068

RESUMO

Intracellular bacterial pathogens subvert the endocytic bactericidal pathway to form specific replication-permissive compartments termed pathogen vacuoles or inclusions. To this end, the pathogens employ type III or type IV secretion systems, which translocate dozens, if not hundreds, of different effector proteins into their host cells, where they manipulate vesicle trafficking and signaling pathways in favor of the intruders. While the distinct cocktail of effectors defines the specific processes by which a pathogen vacuole is formed, the different pathogens commonly target certain vesicle trafficking routes, including the endocytic or secretory pathway. Recently, the retrograde transport pathway from endosomal compartments to the trans-Golgi network emerged as an important route affecting pathogen vacuole formation. Here, we review current insight into the host cell's retrograde trafficking pathway and how vacuolar pathogens of the genera Legionella, Coxiella, Salmonella, Chlamydia, and Simkania employ mechanistically distinct strategies to subvert this pathway, thus promoting intracellular survival and replication.


Assuntos
Sistemas de Secreção Bacterianos/fisiologia , Interações Hospedeiro-Patógeno/fisiologia , Transporte Proteico/fisiologia , Proteínas de Bactérias/metabolismo , Chlamydia/patogenicidade , Chlamydia/fisiologia , Chlamydiales/patogenicidade , Chlamydiales/fisiologia , Coxiella/patogenicidade , Coxiella/fisiologia , Endocitose , Endossomos/metabolismo , Complexo de Golgi/fisiologia , Legionella/patogenicidade , Legionella/fisiologia , Salmonella/patogenicidade , Salmonella/fisiologia , Sistemas de Secreção Tipo III , Sistemas de Secreção Tipo IV , Vacúolos/microbiologia
15.
Cell ; 158(4): 722-733, 2014 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-25126781

RESUMO

Antibiotic therapy often fails to eliminate a fraction of transiently refractory bacteria, causing relapses and chronic infections. Multiple mechanisms can induce such persisters with high antimicrobial tolerance in vitro, but their in vivo relevance remains unclear. Using a fluorescent growth rate reporter, we detected extensive phenotypic variation of Salmonella in host tissues. This included slow-growing subsets as well as well-nourished fast-growing subsets driving disease progression. Monitoring of Salmonella growth and survival during chemotherapy revealed that antibiotic killing correlated with single-cell division rates. Nondividing Salmonella survived best but were rare, limiting their impact. Instead, most survivors originated from abundant moderately growing, partially tolerant Salmonella. These data demonstrate that host tissues diversify pathogen physiology, with major consequences for disease progression and control.


Assuntos
Antibacterianos/administração & dosagem , Fluoroquinolonas/administração & dosagem , Imagem Óptica/métodos , Salmonella typhimurium/efeitos dos fármacos , Febre Tifoide/tratamento farmacológico , Febre Tifoide/microbiologia , Animais , Proteínas de Bactérias/análise , Enrofloxacina , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos BALB C , Proteoma/análise , Salmonella typhimurium/citologia , Salmonella typhimurium/crescimento & desenvolvimento , Baço/microbiologia , Baço/patologia
16.
FEBS J ; 281(13): 2977-89, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24814574

RESUMO

Ral proteins are small GTPases that play critical roles in normal physiology and in oncogenesis. There is little information on the GTPase-activating proteins (GAPs) that downregulate their activity. Here, we provide evidence that the noncatalytic ß subunit of RalGAPα1/2 ß complexes is involved in mitotic control. RalGAPß localizes to the Golgi and nucleus during interphase, and relocalizes to the mitotic spindle and cytokinetic intercellular bridge during mitosis. Depletion of RalGAPß causes chromosome misalignment and decreases the amount of mitotic cyclin B1, disturbing the metaphase-to-anaphase transition. Overexpression of RalGAPß interferes with cell division, leading to binucleation and multinucleation, and cell death. We propose that RalGAPß plays an essential role in the sequential progression of mitosis by controlling the spatial and temporal activation of Ral GTPases in the spindle assembly checkpoint (SAC) and cytokinesis. Deregulation of RalGAPß might cause genomic instability, leading to human carcinogenesis.


Assuntos
Anáfase , Proteínas Ativadoras de GTPase/fisiologia , Metáfase , Morte Celular , Linhagem Celular Tumoral , Cromatografia de Afinidade , Segregação de Cromossomos , Proteínas Ativadoras de GTPase/metabolismo , Proteínas de Choque Térmico HSP70/metabolismo , Células Endoteliais da Veia Umbilical Humana , Humanos , Mitose , Proteínas do Tecido Nervoso/metabolismo , Transporte Proteico , Fuso Acromático/metabolismo , Tubulina (Proteína)/metabolismo , Proteínas ral de Ligação ao GTP/metabolismo
17.
J Bacteriol ; 193(17): 4425-37, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21725001

RESUMO

Many virulence factors of Gram-positive bacterial pathogens are covalently anchored to the peptidoglycan (PG) by sortase enzymes. However, for rod-shaped bacteria little is known about the spatiotemporal organization of these surface proteins in the cell wall. Here we report the three-dimensional (3D) localization of the PG-bound virulence factors InlA, InlH, InlJ, and SvpA in the envelope of Listeria monocytogenes under different growth conditions. We found that all PG-anchored proteins are positioned along the lateral cell wall in nonoverlapping helices. However, these surface proteins can also become localized at the pole and asymmetrically distributed when specific regulatory pathways are activated. InlA and InlJ are enriched at poles when expressed at high levels in exponential-phase bacteria. InlA and InlH, which are σ(B)dependent, specifically relocalize to the septal cell wall and subsequently to the new pole in cells entering stationary phase. The accumulation of InlA and InlH in the septal region also occurs when oxidative stress impairs bacterial growth. In contrast, the iron-dependent protein SvpA is present at the old pole and is excluded from the septum and new pole of bacteria grown under low-iron conditions. We conclude that L. monocytogenes rapidly reorganizes the spatial localization of its PG proteins in response to changes in environmental conditions such as nutrient deprivation or other stresses. This dynamic control would distribute virulence factors at specific sites during the infectious process.


Assuntos
Proteínas de Bactérias/metabolismo , Parede Celular/metabolismo , Listeria monocytogenes/crescimento & desenvolvimento , Peptidoglicano/metabolismo , Proteínas de Bactérias/genética , Parede Celular/genética , Regulação Bacteriana da Expressão Gênica , Genes Bacterianos , Immunoblotting , Listeria monocytogenes/genética , Listeria monocytogenes/metabolismo , Proteínas de Membrana/metabolismo , Conformação Molecular , Estresse Oxidativo , Fatores de Virulência/metabolismo
18.
Infect Immun ; 78(5): 1979-89, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20176794

RESUMO

The genome of the pathogenic bacterium Listeria monocytogenes contains a family of genes encoding proteins with a leucine-rich repeat domain. One of these genes, inlH, is a sigma(B)-dependent virulence gene of unknown function. Previously, inlH was proposed to be coexpressed with two adjacent internalin genes, inlG and inlE. Using tiling arrays, we showed that inlH expression is monocistronic and specifically induced in stationary phase as well as in the intestinal lumen of mice, independent of inlG and inlE expression. Consistent with inlH sigma(B)-dependent regulation, surface expression of the InlH protein is induced when bacteria are subjected to thermal, acidic, osmotic, or oxidative stress. Disruption of inlH increases the amount of the invasion protein InlA without changing inlA transcript level, suggesting that there is a link between inlH expression and inlA posttranscriptional regulation. However, in contrast to InlA, InlH does not contribute to bacterial invasion of cultured cells in vitro or of intestinal cells in vivo. Strikingly, the reduced virulence of inlH-deficient L. monocytogenes strains is accompanied by enhanced production of interleukin-6 (IL-6) in infected tissues during the systemic phase of murine listeriosis but not by enhanced production of any other inflammatory cytokine tested. Since InlH does not modulate IL-6 secretion in macrophages at least in vitro, it may play a role in other immune cells or contribute to a pathway that modulates survival or activation of IL-6-secreting cells. These results strongly suggest that InlH is a stress-induced surface protein that facilitates pathogen survival in tissues by tempering the inflammatory response.


Assuntos
Proteínas de Bactérias/fisiologia , Interleucina-6/antagonistas & inibidores , Listeria monocytogenes/patogenicidade , Listeriose/imunologia , Fatores de Virulência/fisiologia , Animais , Proteínas de Bactérias/genética , Sangue/microbiologia , Linhagem Celular , Contagem de Colônia Microbiana , Feminino , Perfilação da Expressão Gênica , Técnicas de Inativação de Genes , Humanos , Interleucina-6/imunologia , Listeria monocytogenes/imunologia , Fígado/microbiologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Baço/microbiologia , Estresse Fisiológico , Virulência , Fatores de Virulência/biossíntese , Fatores de Virulência/genética
19.
Infect Immun ; 76(4): 1368-78, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18227172

RESUMO

The food-borne pathogen Listeria monocytogenes is adapted to a diversity of environments, such as soil, food, body fluids, and the cytosol of eukaryotic cells. The transition between saprophytic and pathogenic life is mediated through complex regulatory pathways that modulate the expression of virulence factors. Here we examined the expression of inlJ, a recently identified gene encoding a protein of the LPXTG-internalin family and involved in pathogenesis. We show that inlJ expression is controlled neither by the major listerial regulator of virulence genes, PrfA, nor by AxyR, a putative AraC regulator encoded by a gene adjacent to inlJ and divergently transcribed. The InlJ protein is not produced by bacteria grown in vitro in brain heart infusion medium or replicating in the cytosol of tissue-cultured cells. In contrast, it is efficiently produced and localized at the surface of bacteria present in the liver and blood of infected animals. Strikingly, the expression of inlJ by a heterologous promoter in L. monocytogenes or L. innocua promotes bacterial adherence to human cells in vitro. Taken together, these results strongly suggest that InlJ acts as a novel L. monocytogenes sortase-anchored adhesin specifically expressed during infection in vivo.


Assuntos
Adesinas Bacterianas/metabolismo , Regulação Bacteriana da Expressão Gênica , Listeria monocytogenes/metabolismo , Fatores de Virulência/metabolismo , Adesinas Bacterianas/genética , Animais , Linhagem Celular , Meios de Cultura/química , Humanos , Listeria monocytogenes/patogenicidade , Listeriose/sangue , Listeriose/microbiologia , Macrófagos/microbiologia , Camundongos , Transcrição Gênica , Fatores de Virulência/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA