Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Annu Rev Microbiol ; 74: 221-245, 2020 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-32660389

RESUMEN

Microbial pathogens have evolved complex mechanisms to interface with host cells in order to evade host defenses and replicate. However, mammalian innate immune receptors detect the presence of molecules unique to the microbial world or sense the activity of virulence factors, activating antimicrobial and inflammatory pathways. We focus on how studies of the major virulence factor of one group of microbial pathogens, the type III secretion system (T3SS) of human pathogenic Yersinia, have shed light on these important innate immune responses. Yersinia are largely extracellular pathogens, yet they insert T3SS cargo into target host cells that modulate the activity of cytosolic innate immune receptors. This review covers both the host pathways that detect the Yersinia T3SS and the effector proteins used by Yersinia to manipulate innate immune signaling.


Asunto(s)
Citosol/inmunología , Interacciones Huésped-Patógeno/inmunología , Inmunidad Innata , Sistemas de Secreción Tipo III/inmunología , Yersinia/inmunología , Proteínas Bacterianas/inmunología , Proteínas Bacterianas/metabolismo , Citosol/microbiología , Humanos , Inflamasomas , Piroptosis , Transducción de Señal , Factores de Virulencia/metabolismo , Yersinia/metabolismo , Yersinia/patogenicidad
2.
PLoS Genet ; 18(7): e1010321, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35901167

RESUMEN

The type III secretion system (T3SS) is an appendage used by many bacterial pathogens, such as pathogenic Yersinia, to subvert host defenses. However, because the T3SS is energetically costly and immunogenic, it must be tightly regulated in response to environmental cues to enable survival in the host. Here we show that expression of the Yersinia Ysc T3SS master regulator, LcrF, is orchestrated by the opposing activities of the repressive H-NS/YmoA histone-like protein complex and induction by the iron and oxygen-regulated IscR transcription factor. While deletion of iscR or ymoA has been shown to decrease and increase LcrF expression and type III secretion, respectively, the role of H-NS in this system has not been definitively established because hns is an essential gene in Yersinia. Using CRISPRi knockdown of hns, we show that hns depletion causes derepression of lcrF. Furthermore, we find that while YmoA is dispensable for H-NS binding to the lcrF promoter, YmoA binding to H-NS is important for H-NS repressive activity. We bioinformatically identified three H-NS binding regions within the lcrF promoter and demonstrate binding of H-NS to these sites in vivo using chromatin immunoprecipitation. Using promoter truncation and binding site mutation analysis, we show that two of these H-NS binding regions are important for H-NS/YmoA-mediated repression of the lcrF promoter. Surprisingly, we find that IscR is dispensable for lcrF transcription in the absence of H-NS/YmoA. Indeed, IscR-dependent regulation of LcrF and type III secretion in response to changes in oxygen, such as those Yersinia is predicted to experience during host infection, only occurs in the presence of an H-NS/YmoA complex. These data suggest that, in the presence of host tissue cues that drive sufficient IscR expression, IscR can act as a roadblock to H-NS/YmoA-dependent repression of RNA polymerase at the lcrF promoter to turn on T3SS expression.


Asunto(s)
Regulación Bacteriana de la Expresión Génica , Yersinia , Proteínas Bacterianas/metabolismo , Histonas/genética , Oxígeno/metabolismo , Yersinia/genética , Yersinia/metabolismo
3.
Antimicrob Agents Chemother ; 65(7): e0169020, 2021 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-33875435

RESUMEN

Antibiotic-resistant bacteria are an emerging global health threat. New antimicrobials are urgently needed. The injectisome type III secretion system (T3SS), required by dozens of Gram-negative bacteria for virulence but largely absent from nonpathogenic bacteria, is an attractive antimicrobial target. We previously identified synthetic cyclic peptomers, inspired by the natural product phepropeptin D, that inhibit protein secretion through the Yersinia Ysc and Pseudomonas aeruginosa Psc T3SSs but do not inhibit bacterial growth. Here, we describe the identification of an isomer, 4EpDN, that is 2-fold more potent (50% inhibitory concentration [IC50] of 4 µM) than its parental compound. Furthermore, 4EpDN inhibited the Yersinia Ysa and the Salmonella SPI-1 T3SSs, suggesting that this cyclic peptomer has broad efficacy against evolutionarily distant injectisome T3SSs. Indeed, 4EpDN strongly inhibited intracellular growth of Chlamydia trachomatis in HeLa cells, which requires the T3SS. 4EpDN did not inhibit the unrelated twin arginine translocation (Tat) system, nor did it impact T3SS gene transcription. Moreover, although the injectisome and flagellar T3SSs are evolutionarily and structurally related, the 4EpDN cyclic peptomer did not inhibit secretion of substrates through the Salmonella flagellar T3SS, indicating that cyclic peptomers broadly but specifically target the injectisome T3SS. 4EpDN reduced the number of T3SS needles detected on the surface of Yersinia pseudotuberculosis as detected by microscopy. Collectively, these data suggest that cyclic peptomers specifically inhibit the injectisome T3SS from a variety of Gram-negative bacteria, possibly by preventing complete T3SS assembly.


Asunto(s)
Sistemas de Secreción Tipo III , Yersinia pseudotuberculosis , Proteínas Bacterianas/genética , Células HeLa , Humanos , Pseudomonas aeruginosa , Sistemas de Secreción Tipo III/genética , Virulencia , Yersinia pseudotuberculosis/genética
4.
PLoS Pathog ; 15(12): e1008001, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31869388

RESUMEN

The enteropathogen Yersinia pseudotuberculosis and the related plague agent Y. pestis require the Ysc type III secretion system (T3SS) to subvert phagocyte defense mechanisms and cause disease. Yet type III secretion (T3S) in Yersinia induces growth arrest and innate immune recognition, necessitating tight regulation of the T3SS. Here we show that Y. pseudotuberculosis T3SS expression is kept low under anaerobic, iron-rich conditions, such as those found in the intestinal lumen where the Yersinia T3SS is not required for growth. In contrast, the Yersinia T3SS is expressed under aerobic or anaerobic, iron-poor conditions, such as those encountered by Yersinia once they cross the epithelial barrier and encounter phagocytic cells. We further show that the [2Fe-2S] containing transcription factor, IscR, mediates this oxygen and iron regulation of the T3SS by controlling transcription of the T3SS master regulator LcrF. IscR binds directly to the lcrF promoter and, importantly, a mutation that prevents this binding leads to decreased disseminated infection of Y. pseudotuberculosis but does not perturb intestinal colonization. Similar to E. coli, Y. pseudotuberculosis uses the Fe-S cluster occupancy of IscR as a readout of oxygen and iron conditions that impact cellular Fe-S cluster homeostasis. We propose that Y. pseudotuberculosis has coopted this system to sense entry into deeper tissues and induce T3S where it is required for virulence. The IscR binding site in the lcrF promoter is completely conserved between Y. pseudotuberculosis and Y. pestis. Deletion of iscR in Y. pestis leads to drastic disruption of T3S, suggesting that IscR control of the T3SS evolved before Y. pestis split from Y. pseudotuberculosis.


Asunto(s)
Hierro/metabolismo , Oxígeno/metabolismo , Sistemas de Secreción Tipo III/metabolismo , Infecciones por Yersinia pseudotuberculosis/inmunología , Animales , Proteínas Bacterianas/metabolismo , Sistemas de Secreción Bacterianos/genética , Escherichia coli/metabolismo , Proteínas de Escherichia coli/metabolismo , Regulación Bacteriana de la Expresión Génica/genética , Regiones Promotoras Genéticas/genética , Transcripción Genética , Yersinia/metabolismo , Yersinia pseudotuberculosis/patogenicidad , Infecciones por Yersinia pseudotuberculosis/metabolismo
5.
Artículo en Inglés | MEDLINE | ID: mdl-28652236

RESUMEN

Antibiotic-resistant bacteria are an emerging threat to global public health. New classes of antibiotics and tools for antimicrobial discovery are urgently needed. Type III secretion systems (T3SS), which are required by dozens of Gram-negative bacteria for virulence but largely absent from nonpathogenic bacteria, are promising virulence blocker targets. The ability of mammalian cells to recognize the presence of a functional T3SS and trigger NF-κB activation provides a rapid and sensitive method for identifying chemical inhibitors of T3SS activity. In this study, we generated a HEK293 stable cell line expressing green fluorescent protein (GFP) driven by a promoter containing NF-κB enhancer elements to serve as a readout of T3SS function. We identified a family of synthetic cyclic peptide-peptoid hybrid molecules (peptomers) that exhibited dose-dependent inhibition of T3SS effector secretion in Yersinia pseudotuberculosis and Pseudomonas aeruginosa without affecting bacterial growth or motility. Among these inhibitors, EpD-3'N, EpD-1,2N, EpD-1,3'N, EpD-1,2,3'N, and EpD-1,2,4'N exhibited strong inhibitory effects on translocation of the Yersinia YopM effector protein into mammalian cells (>40% translocation inhibition at 7.5 µM) and showed no toxicity to mammalian cells at 240 µM. In addition, EpD-3'N and EpD-1,2,4'N reduced the rounding of HeLa cells caused by the activity of Yersinia effector proteins that target the actin cytoskeleton. In summary, we have discovered a family of novel cyclic peptomers that inhibit the injectisome T3SS but not the flagellar T3SS.


Asunto(s)
Antibacterianos/farmacología , Péptidos Cíclicos/farmacología , Sistemas de Secreción Tipo III/efectos de los fármacos , Proteínas Bacterianas/genética , Línea Celular , Línea Celular Tumoral , Proteínas Fluorescentes Verdes , Células HEK293 , Células HeLa , Humanos , FN-kappa B/genética , Regiones Promotoras Genéticas/efectos de los fármacos , Regiones Promotoras Genéticas/genética , Transporte de Proteínas/efectos de los fármacos , Transporte de Proteínas/genética , Pseudomonas aeruginosa/efectos de los fármacos , Pseudomonas aeruginosa/genética , Sistemas de Secreción Tipo III/genética , Virulencia/efectos de los fármacos , Virulencia/genética , Yersinia pseudotuberculosis/efectos de los fármacos , Yersinia pseudotuberculosis/genética
6.
J Nat Prod ; 80(12): 3255-3266, 2017 12 22.
Artículo en Inglés | MEDLINE | ID: mdl-29144750

RESUMEN

This research set out to identify compounds from marine sponges that can act as bacterial virulence blockers. Extracts from a total of 80 sponges collected from throughout Indonesia were screened in a high-throughput NF-κB-based screen that identifies compounds capable of inhibiting the bacterial type III secretion system (T3SS) in Yersinia pseudotuberculosis. An extract that was shown to inhibit T3SS-driven NF-κB expression was obtained from an Iotrochota cf. iota sponge and was the source of seven new bromo- and iodo-containing compounds, all of which contain a 2-(4-oxyphenyl)ethan-1-amine core. Five were determined to be new compounds and named enisorines A-E (1-5). The remaining two were determined to be new hemibastadinol analogues named (+)-1-O-methylhemibastadinol 2 (6) and (+)-1-O-methylhemibastadinol 4 (7). All seven compounds inhibited T3SS-dependent YopE secretion and did not affect the growth or metabolic activity of Y. pseudotuberculosis. The most potent inhibitors of T3SS activity were enisorine C (3), enisorine E (5), and (+)-1-O-methylhemibastadinol 2 (6), all of which inhibited YopE secretion by >50% at 30 µM.


Asunto(s)
Productos Biológicos/química , Productos Biológicos/farmacología , Poríferos/química , Animales , Línea Celular Tumoral , Humanos , Indonesia , Células MCF-7 , FN-kappa B/metabolismo , Yersinia pseudotuberculosis/efectos de los fármacos
7.
Mol Microbiol ; 96(4): 764-78, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25684661

RESUMEN

The Yersinia type III secretion system (T3SS) translocates Yop effector proteins into host cells to manipulate immune defenses such as phagocytosis and reactive oxygen species (ROS) production. The T3SS translocator proteins YopB and YopD form pores in host membranes, facilitating Yop translocation. While the YopD amino and carboxy termini participate in pore formation, the role of the YopD central region between amino acids 150-227 remains unknown. We assessed the contribution of this region by generating Y. pseudotuberculosis yopD(Δ150-170) and yopD(Δ207-227) mutants and analyzing their T3SS functions. These strains exhibited wild-type levels of Yop secretion in vitro and enabled robust pore formation in macrophages. However, the yopDΔ150-170 and yopD(Δ207-227) mutants were defective in Yop translocation into CHO cells and splenocyte-derived neutrophils and macrophages. These data suggest that YopD-mediated host membrane disruption and effector Yop translocation are genetically separable activities requiring distinct protein domains. Importantly, the yopD(Δ150-170) and yopD(Δ207-227) mutants were defective in Yop-mediated inhibition of macrophage cell death and ROS production in neutrophil-like cells, and were attenuated in disseminated Yersinia infection. Therefore, the ability of the YopD central region to facilitate optimal effector protein delivery into phagocytes, and therefore robust effector Yop function, is important for Yersinia virulence.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/genética , Proteínas de la Membrana Bacteriana Externa/metabolismo , Proteínas Bacterianas/genética , Membrana Celular/metabolismo , Yersinia pseudotuberculosis/genética , Yersinia pseudotuberculosis/metabolismo , Animales , Proteínas Bacterianas/metabolismo , Células CHO , Células Cultivadas , Cricetulus , Células HL-60 , Humanos , Macrófagos/metabolismo , Macrófagos/microbiología , Ratones Endogámicos C57BL , Mutación , Estructura Terciaria de Proteína , Transporte de Proteínas , Especies Reactivas de Oxígeno/metabolismo , Sistemas de Secreción Tipo III/genética , Sistemas de Secreción Tipo III/fisiología , Yersinia pseudotuberculosis/crecimiento & desarrollo , Yersinia pseudotuberculosis/patogenicidad
8.
PLoS Pathog ; 10(6): e1004194, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24945271

RESUMEN

Type III secretion systems (T3SS) are essential for virulence in dozens of pathogens, but are not required for growth outside the host. Therefore, the T3SS of many bacterial species are under tight regulatory control. To increase our understanding of the molecular mechanisms behind T3SS regulation, we performed a transposon screen to identify genes important for T3SS function in the food-borne pathogen Yersinia pseudotuberculosis. We identified two unique transposon insertions in YPTB2860, a gene that displays 79% identity with the E. coli iron-sulfur cluster regulator, IscR. A Y. pseudotuberculosis iscR in-frame deletion mutant (ΔiscR) was deficient in secretion of Ysc T3SS effector proteins and in targeting macrophages through the T3SS. To determine the mechanism behind IscR control of the Ysc T3SS, we carried out transcriptome and bioinformatic analysis to identify Y. pseudotuberculosis genes regulated by IscR. We discovered a putative IscR binding motif upstream of the Y. pseudotuberculosis yscW-lcrF operon. As LcrF controls transcription of a number of critical T3SS genes in Yersinia, we hypothesized that Yersinia IscR may control the Ysc T3SS through LcrF. Indeed, purified IscR bound to the identified yscW-lcrF promoter motif and mRNA levels of lcrF and 24 other T3SS genes were reduced in Y. pseudotuberculosis in the absence of IscR. Importantly, mice orally infected with the Y. pseudotuberculosis ΔiscR mutant displayed decreased bacterial burden in Peyer's patches, mesenteric lymph nodes, spleens, and livers, indicating an essential role for IscR in Y. pseudotuberculosis virulence. This study presents the first characterization of Yersinia IscR and provides evidence that IscR is critical for virulence and type III secretion through direct regulation of the T3SS master regulator, LcrF.


Asunto(s)
Sistemas de Secreción Bacterianos/genética , Proteínas de Escherichia coli/genética , Factores de Transcripción/genética , Factores de Virulencia/genética , Yersinia pseudotuberculosis/genética , Yersinia pseudotuberculosis/patogenicidad , Secuencia de Aminoácidos , Animales , Sitios de Unión/genética , Elementos Transponibles de ADN/genética , Escherichia coli/genética , Perfilación de la Expresión Génica , Regulación Bacteriana de la Expresión Génica , Hígado/inmunología , Hígado/microbiología , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/microbiología , Ratones , Datos de Secuencia Molecular , Ganglios Linfáticos Agregados/inmunología , Ganglios Linfáticos Agregados/microbiología , Regiones Promotoras Genéticas/genética , Unión Proteica , Alineación de Secuencia , Bazo/inmunología , Bazo/microbiología , Transcripción Genética , Transcriptoma/genética , Infecciones por Yersinia pseudotuberculosis/inmunología , Infecciones por Yersinia pseudotuberculosis/patología
9.
J Bacteriol ; 198(4): 604-14, 2015 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-26644429

RESUMEN

Many Gram-negative pathogens express a type III secretion (T3SS) system to enable growth and survival within a host. The three human-pathogenic Yersinia species, Y. pestis, Y. pseudotuberculosis, and Y. enterocolitica, encode the Ysc T3SS, whose expression is controlled by an AraC-like master regulator called LcrF. In this review, we discuss LcrF structure and function as well as the environmental cues and pathways known to regulate LcrF expression. Similarities and differences in binding motifs and modes of action between LcrF and the Pseudomonas aeruginosa homolog ExsA are summarized. In addition, we present a new bioinformatics analysis that identifies putative LcrF binding sites within Yersinia target gene promoters.


Asunto(s)
Proteínas Bacterianas/metabolismo , Regulación Bacteriana de la Expresión Génica , Transactivadores/metabolismo , Sistemas de Secreción Tipo III/genética , Yersinia/metabolismo , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Estructura Terciaria de Proteína , Transactivadores/química , Transactivadores/genética , Sistemas de Secreción Tipo III/metabolismo , Yersinia/química , Yersinia/genética , Yersinia/crecimiento & desarrollo
10.
Antimicrob Agents Chemother ; 58(2): 1118-26, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24295981

RESUMEN

The type III secretion system (T3SS) is a bacterial appendage used by dozens of Gram-negative pathogens to subvert host defenses and cause disease, making it an ideal target for pathogen-specific antimicrobials. Here, we report the discovery and initial characterization of two related natural products with T3SS-inhibitory activity that were derived from a marine actinobacterium. Bacterial extracts containing piericidin A1 and the piericidin derivative Mer-A 2026B inhibited Yersinia pseudotuberculosis from triggering T3SS-dependent activation of the host transcription factor NF-κB in HEK293T cells but were not toxic to mammalian cells. As the Yersinia T3SS must be functional in order to trigger NF-κB activation, these data indicate that piericidin A1 and Mer-A 2026B block T3SS function. Consistent with this, purified piericidin A1 and Mer-A 2026B dose-dependently inhibited translocation of the Y. pseudotuberculosis T3SS effector protein YopM inside CHO cells. In contrast, neither compound perturbed bacterial growth in vitro, indicating that piericidin A1 and Mer-A 2026B do not function as general antibiotics in Yersinia. In addition, when Yersinia was incubated under T3SS-inducing culture conditions in the absence of host cells, Mer-A 2026B and piericidin A1 inhibited secretion of T3SS cargo as effectively as or better than several previously described T3SS inhibitors, such as MBX-1641 and aurodox. This suggests that Mer-A 2026B and piericidin A1 do not block type III secretion by blocking the bacterium-host cell interaction, but rather inhibit an earlier stage, such as T3SS needle assembly. In summary, the marine-derived natural products Mer-A 2026B and piericidin A1 possess previously uncharacterized activity against the bacterial T3SS.


Asunto(s)
Antibacterianos/farmacología , Sistemas de Secreción Bacterianos/efectos de los fármacos , FN-kappa B/genética , Piridinas/farmacología , Yersinia pseudotuberculosis/efectos de los fármacos , Actinomycetales/química , Animales , Antibacterianos/aislamiento & purificación , Aurodox/farmacología , Proteínas de la Membrana Bacteriana Externa/antagonistas & inhibidores , Proteínas de la Membrana Bacteriana Externa/metabolismo , Células CHO , Cricetulus , Relación Dosis-Respuesta a Droga , Regulación de la Expresión Génica , Células HEK293 , Ensayos Analíticos de Alto Rendimiento , Humanos , FN-kappa B/metabolismo , Transporte de Proteínas/efectos de los fármacos , Piridinas/aislamiento & purificación , Yersinia pseudotuberculosis/metabolismo
11.
J Vis Exp ; (207)2024 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-38884494

RESUMEN

A key virulence mechanism for many Gram-negative pathogens is the type III secretion system (T3SS), a needle-like appendage that translocates cytotoxic or immunomodulatory effector proteins into host cells. The T3SS is a target for antimicrobial discovery campaigns since it is accessible extracellularly and largely absent from non-pathogenic bacteria. Recent studies demonstrated that the T3SS of Yersinia and Salmonella are regulated by factors responsive to iron and oxygen, which are important niche-specific signals encountered during mammalian infection. Described here is a method for iron starvation of Yersinia pseudotuberculosis, with subsequent optional supplementation of inorganic iron. To assess the impact of oxygen availability, this iron starvation process is demonstrated under both aerobic and anaerobic conditions. Finally, incubating the cultures at the mammalian host temperature of 37 °C induces T3SS expression and allows quantification of Yersinia T3SS activity by visualizing effector proteins released into the supernatant. The steps detailed here offer an advantage over the use of iron chelators in the absence of iron starvation, which is insufficient for inducing robust iron starvation, presumably due to efficient Yersinia iron uptake and scavenging systems. Likewise, acid-washing laboratory glassware is detailed to ensure the removal of residual iron, which is essential for inducing robust iron starvation. Additionally, using a chelating agent is described to remove residual iron from media, and culturing the bacteria for several generations in the absence of iron to deplete bacterial iron stores. By incorporating standard protocols of trichloroacetic acid-induced protein precipitation, SDS-PAGE, and silver staining, this procedure demonstrates accessible ways to measure T3SS activity. While this procedure is optimized for Y. pseudotuberculosis, it offers a framework for studies in pathogens with similar robust iron uptake systems. In the age of antibiotic resistance, these methods can be expanded to assess the efficacy of antimicrobial compounds targeting the T3SS under host-relevant conditions.


Asunto(s)
Hierro , Sistemas de Secreción Tipo III , Yersinia pseudotuberculosis , Yersinia pseudotuberculosis/metabolismo , Hierro/metabolismo , Sistemas de Secreción Tipo III/metabolismo , Anaerobiosis
12.
Infect Immun ; 81(3): 905-14, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23297383

RESUMEN

Type III secretion systems (T3SSs) are used by Gram-negative pathogens to form pores in host membranes and deliver virulence-associated effector proteins inside host cells. In pathogenic Yersinia, the T3SS pore-forming proteins are YopB and YopD. Mammalian cells recognize the Yersinia T3SS, leading to a host response that includes secretion of the inflammatory cytokine interleukin-1ß (IL-1ß), Toll-like receptor (TLR)-independent expression of the stress-associated transcription factor Egr1 and the inflammatory cytokine tumor necrosis factor alpha (TNF-α), and host cell death. The known Yersinia T3SS effector proteins are dispensable for eliciting these responses, but YopB is essential. Three models describe how the Yersinia T3SS might trigger inflammation: (i) mammalian cells sense YopBD-mediated pore formation, (ii) innate immune stimuli gain access to the host cytoplasm through the YopBD pore, and/or (iii) the YopB-YopD translocon itself or its membrane insertion is proinflammatory. To test these models, we constructed a Yersinia pseudotuberculosis mutant expressing YopD devoid of its predicted transmembrane domain (YopD(ΔTM)) and lacking the T3SS cargo proteins YopHEMOJTN. This mutant formed pores in macrophages, but it could not mediate translocation of effector proteins inside host cells. Importantly, this mutant did not elicit rapid host cell death, IL-1ß secretion, or TLR-independent Egr1 and TNF-α expression. These data suggest that YopBD-mediated translocation of unknown T3SS cargo leads to activation of host pathways influencing inflammation, cell death, and response to stress. As the YopD(ΔTM) Y. pseudotuberculosis mutant formed somewhat smaller pores with delayed kinetics, an alternative model is that the wild-type YopB-YopD translocon is specifically sensed by host cells.


Asunto(s)
Macrófagos/microbiología , Yersinia pseudotuberculosis/metabolismo , Animales , Muerte Celular , Línea Celular , Proteína 1 de la Respuesta de Crecimiento Precoz/genética , Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Eliminación de Gen , Regulación de la Expresión Génica/inmunología , Regulación de la Expresión Génica/fisiología , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Ratones , Ratones Endogámicos C57BL , Reacción en Cadena de la Polimerasa/métodos , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo , Yersinia pseudotuberculosis/genética
13.
Antimicrob Agents Chemother ; 56(11): 5433-41, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22850518

RESUMEN

The recent and dramatic rise of antibiotic resistance among bacterial pathogens underlies the fear that standard treatments for infectious disease will soon be largely ineffective. Resistance has evolved against nearly every clinically used antibiotic, and in the near future, we may be hard-pressed to treat bacterial infections previously conquered by "magic bullet" drugs. While traditional antibiotics kill or slow bacterial growth, an important emerging strategy to combat pathogens seeks to block the ability of bacteria to harm the host by inhibiting bacterial virulence factors. One such virulence factor, the type three secretion system (T3SS), is found in over two dozen Gram-negative pathogens and functions by injecting effector proteins directly into the cytosol of host cells. Without T3SSs, many pathogenic bacteria are unable to cause disease, making the T3SS an attractive target for novel antimicrobial drugs. Interdisciplinary efforts between chemists and microbiologists have yielded several T3SS inhibitors, including the relatively well-studied salicylidene acylhydrazides. This review highlights the discovery and characterization of T3SS inhibitors in the primary literature over the past 10 years and discusses the future of these drugs as both research tools and a new class of therapeutic agents.


Asunto(s)
Proteínas Bacterianas/antagonistas & inhibidores , Farmacorresistencia Bacteriana Múltiple , Bacterias Gramnegativas/patogenicidad , Infecciones por Bacterias Gramnegativas/tratamiento farmacológico , Hidrazinas/farmacología , Bases de Schiff/farmacología , Factores de Virulencia/antagonistas & inhibidores , Antibacterianos/uso terapéutico , Bacterias Gramnegativas/efectos de los fármacos , Bacterias Gramnegativas/fisiología , Infecciones por Bacterias Gramnegativas/microbiología , Humanos , Hidrazinas/síntesis química , Bases de Schiff/síntesis química , Virulencia
15.
PLoS Pathog ; 5(12): e1000686, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19997504

RESUMEN

Specialized protein translocation systems are used by many bacterial pathogens to deliver effector proteins into host cells that interfere with normal cellular functions. How the host immune system recognizes and responds to this intrusive event is not understood. To address these questions, we determined the mammalian cellular response to the virulence-associated type III secretion system (T3SS) of the human pathogen Yersinia pseudotuberculosis. We found that macrophages devoid of Toll-like receptor (TLR) signaling regulate expression of 266 genes following recognition of the Y. pseudotuberculosis T3SS. This analysis revealed two temporally distinct responses that could be separated into activation of NFkappaB- and type I IFN-regulated genes. Extracellular bacteria were capable of triggering these signaling events, as inhibition of bacterial uptake had no effect on the ensuing innate immune response. The cytosolic peptidoglycan sensors Nod1 and Nod2 and the inflammasome component caspase-1 were not involved in NFkappaB activation following recognition of the Y. pseudotuberculosis T3SS. However, caspase-1 was required for secretion of the inflammatory cytokine IL-1beta in response to T3SS-positive Y. pseudotuberculosis. In order to characterize the bacterial requirements for induction of this novel TLR-, Nod1/2-, and caspase-1-independent response, we used Y. pseudotuberculosis strains lacking specific components of the T3SS. Formation of a functional T3SS pore was required, as bacteria expressing a secretion needle, but lacking the pore-forming proteins YopB or YopD, did not trigger these signaling events. However, nonspecific membrane disruption could not recapitulate the NFkappaB signaling triggered by Y. pseudotuberculosis expressing a functional T3SS pore. Although host cell recognition of the T3SS did not require known translocated substrates, the ensuing response could be modulated by effectors such as YopJ and YopT, as YopT amplified the response, while YopJ dampened it. Collectively, these data suggest that combined recognition of the T3SS pore and YopBD-mediated delivery of immune activating ligands into the host cytosol informs the host cell of pathogenic challenge. This leads to a unique, multifactorial response distinct from the canonical immune response to a bacterium lacking a T3SS.


Asunto(s)
Citocinas/inmunología , Infecciones por Yersinia pseudotuberculosis/inmunología , Yersinia pseudotuberculosis/fisiología , Animales , Proteínas Bacterianas/inmunología , Proteínas Bacterianas/metabolismo , Línea Celular Tumoral , Expresión Génica/inmunología , Humanos , Inmunidad Innata , Cinética , Macrófagos/inmunología , Ratones , Proteína Adaptadora de Señalización NOD1/genética , Proteína Adaptadora de Señalización NOD1/inmunología , Proteína Adaptadora de Señalización NOD2/genética , Proteína Adaptadora de Señalización NOD2/inmunología , Vías Secretoras/genética , Vías Secretoras/fisiología , Yersinia pseudotuberculosis/inmunología
16.
mBio ; 12(3): e0063321, 2021 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-34060331

RESUMEN

The iron-sulfur cluster coordinating transcription factor IscR is important for the virulence of Yersinia pseudotuberculosis and a number of other bacterial pathogens. However, the IscR regulon has not yet been defined in any organism. To determine the Yersinia IscR regulon and identify IscR-dependent functions important for virulence, we employed chromatin immunoprecipitation sequencing (ChIP-Seq) and RNA sequencing (RNA-Seq) of Y. pseudotuberculosis expressing or lacking iscR following iron starvation conditions, such as those encountered during infection. We found that IscR binds to the promoters of genes involved in iron homeostasis, reactive oxygen species metabolism, and cell envelope remodeling and regulates expression of these genes in response to iron depletion. Consistent with our previous work, we also found that IscR binds in vivo to the promoter of the Ysc type III secretion system (T3SS) master regulator LcrF, leading to regulation of T3SS genes. Interestingly, comparative genomic analysis suggested over 93% of IscR binding sites were conserved between Y. pseudotuberculosis and the related plague agent Yersinia pestis. Surprisingly, we found that the IscR positively regulated sufABCDSE Fe-S cluster biogenesis pathway was required for T3SS activity. These data suggest that IscR regulates the T3SS in Yersinia through maturation of an Fe-S cluster protein critical for type III secretion, in addition to its known role in activating T3SS genes through LcrF. Altogether, our study shows that iron starvation triggers IscR to coregulate multiple, distinct pathways relevant to promoting bacterial survival during infection. IMPORTANCE How bacteria adapt to the changing environment within the host is critical for their ability to survive and cause disease. For example, the mammalian host severely restricts iron availability to limit bacterial growth, referred to as nutritional immunity. Here, we show that pathogenic Yersinia use the iron-sulfur (Fe-S) cluster regulator IscR, a factor critical for pathogenesis, to sense iron availability and regulate multiple pathways known or predicted to contribute to virulence. Under low iron conditions that mimic those Yersinia encounter during infection, IscR levels increase, leading to modulation of genes involved in iron metabolism, stress resistance, cell envelope remodeling, and subversion of host defenses. These data suggest that IscR senses nutritional immunity to coordinate processes important for bacterial survival within the mammalian host.


Asunto(s)
Proteínas Bacterianas/genética , Regulación Bacteriana de la Expresión Génica/genética , Genoma Bacteriano , Genómica/métodos , Factores de Virulencia/genética , Yersinia pseudotuberculosis/genética , Yersinia pseudotuberculosis/patogenicidad , Proteínas Bacterianas/metabolismo , Sitios de Unión , Humanos , Hierro/metabolismo , Regiones Promotoras Genéticas , Virulencia , Yersinia pestis/genética , Yersinia pseudotuberculosis/metabolismo , Infecciones por Yersinia pseudotuberculosis/microbiología
17.
J Exp Med ; 200(4): 527-33, 2004 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-15302899

RESUMEN

Listeria monocytogenes is a facultative intracellular pathogen that induces a cytosolic signaling cascade resulting in expression of interferon (IFN)-beta. Although type I IFNs are critical in viral defense, their role in immunity to bacterial pathogens is much less clear. In this study, we addressed the role of type I IFNs by examining the infection of L. monocytogenes in BALB/c mice lacking the type I IFN receptor (IFN-alpha/betaR-/-). During the first 24 h of infection in vivo, IFN-alpha/betaR-/- and wild-type mice were similar in terms of L. monocytogenes survival. In addition, the intracellular fate of L. monocytogenes in macrophages cultured from IFN-alpha/betaR-/- and wild-type mice was indistinguishable. However, by 72 h after inoculation in vivo, IFN-alpha/betaR-/- mice were approximately 1,000-fold more resistant to a high dose L. monocytogenes infection. Resistance was correlated with elevated levels of interleukin 12p70 in the blood and increased numbers of CD11b+ macrophages producing tumor necrosis factor alpha in the spleen of IFN-alpha/betaR-/- mice. The results of this study suggest that L. monocytogenes might be exploiting an innate antiviral response to promote its pathogenesis.


Asunto(s)
Inmunidad Innata/inmunología , Interferón Tipo I/inmunología , Listeriosis/inmunología , Ratones/inmunología , Receptores de Interferón/deficiencia , Animales , Quimiocina CCL2/sangre , Cartilla de ADN , Ensayo de Inmunoadsorción Enzimática , Interferón gamma/sangre , Interleucina-12/sangre , Macrófagos/inmunología , Proteínas de la Membrana , Ratones Endogámicos BALB C , Ratones Mutantes , Reacción en Cadena de la Polimerasa/métodos , Receptor de Interferón alfa y beta , Bazo/inmunología , Factor de Necrosis Tumoral alfa/metabolismo
18.
PLoS Pathog ; 3(3): e51, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17397264

RESUMEN

Macrophages are permissive hosts to intracellular pathogens, but upon activation become microbiocidal effectors of innate and cell-mediated immunity. How the fate of internalized microorganisms is monitored by macrophages, and how that information is integrated to stimulate specific immune responses is not understood. Activation of macrophages with interferon (IFN)-gamma leads to rapid killing and degradation of Listeria monocytogenes in a phagosome, thus preventing escape of bacteria to the cytosol. Here, we show that activated macrophages induce a specific gene expression program to L. monocytogenes degraded in the phago-lysosome. In addition to activation of Toll-like receptor (TLR) signaling pathways, degraded bacteria also activated a TLR-independent transcriptional response that was similar to the response induced by cytosolic L. monocytogenes. More specifically, degraded bacteria induced a TLR-independent IFN-beta response that was previously shown to be specific to cytosolic bacteria and not to intact bacteria localized to the phagosome. This response required the generation of bacterial ligands in the phago-lysosome and was largely dependent on nucleotide-binding oligomerization domain 2 (NOD2), a cytosolic receptor known to respond to bacterial peptidoglycan fragments. The NOD2-dependent response to degraded bacteria required the phagosomal membrane potential and the activity of lysosomal proteases. The NOD2-dependent IFN-beta production resulted from synergism with other cytosolic microbial sensors. This study supports the hypothesis that in activated macrophages, cytosolic innate immune receptors are activated by bacterial ligands generated in the phagosome and transported to the cytosol.


Asunto(s)
Proteínas Bacterianas/metabolismo , Citosol/inmunología , Inmunidad Innata/inmunología , Listeria monocytogenes/metabolismo , Activación de Macrófagos/inmunología , Fagosomas/metabolismo , Animales , Células Cultivadas , Citosol/metabolismo , Citosol/microbiología , Regulación Bacteriana de la Expresión Génica , Interferón beta/genética , Interferón beta/metabolismo , Ligandos , Listeria monocytogenes/patogenicidad , Listeriosis/fisiopatología , Macrófagos/metabolismo , Macrófagos/microbiología , Potenciales de la Membrana/fisiología , Ratones , Ratones Endogámicos C57BL , Proteína Adaptadora de Señalización NOD2/genética , Proteína Adaptadora de Señalización NOD2/fisiología , Transducción de Señal/fisiología , Receptores Toll-Like/fisiología
19.
J Cell Biol ; 158(3): 409-14, 2002 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-12163465

RESUMEN

Listeria monocytogenes has emerged as a remarkably tractable pathogen to dissect basic aspects of cell biology, intracellular pathogenesis, and innate and acquired immunity. In order to maintain its intracellular lifestyle, L. monocytogenes has evolved a number of mechanisms to exploit host processes to grow and spread cell to cell without damaging the host cell. The pore-forming protein listeriolysin O mediates escape from host vacuoles and utilizes multiple fail-safe mechanisms to avoid causing toxicity to infected cells. Once in the cytosol, the L. monocytogenes ActA protein recruits host cell Arp2/3 complexes and enabled/vasodilator-stimulated phosphoprotein family members to mediate efficient actin-based motility, thereby propelling the bacteria into neighboring cells. Alteration in any of these processes dramatically reduces the ability of the bacteria to establish a productive infection in vivo.


Asunto(s)
Toxinas Bacterianas , Células Eucariotas/microbiología , Inmunidad Celular/fisiología , Listeria monocytogenes/metabolismo , Listeria monocytogenes/patogenicidad , Listeriosis/metabolismo , Actinas/inmunología , Actinas/metabolismo , Actinas/ultraestructura , Animales , Movimiento Celular/inmunología , Células Eucariotas/metabolismo , Células Eucariotas/ultraestructura , Proteínas de Choque Térmico/inmunología , Proteínas de Choque Térmico/metabolismo , Proteínas Hemolisinas , Interacciones Huésped-Parásitos/fisiología , Humanos , Listeria monocytogenes/ultraestructura , Listeriosis/fisiopatología , Modelos Biológicos
20.
Artículo en Inglés | MEDLINE | ID: mdl-30801065

RESUMEN

Yersinia pseudotuberculosis, closely related to Yersinia pestis, is a human pathogen and model organism for studying bacterial pathogenesis. To aid in genomic analysis and understanding bacterial virulence, we sequenced and assembled the complete genome of the human pathogen Yersinia pseudotuberculosis IP2666pIB1.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA