Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Mol Ther Methods Clin Dev ; 30: 288-302, 2023 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-37583716

RESUMEN

Recombinant adeno-associated viruses (rAAVs) deliver DNA to numerous cell types. However, packaging of partial genomes into the rAAV capsid is of concern. Although empty rAAV capsids are studied, there is little information regarding the impact of partial DNA content on rAAV performance in controlled studies. To address this, we tested vectors containing varying levels of partial, self-complementary EGFP genomes. Density gradient cesium chloride ultracentrifugation was used to isolate three distinct rAAV populations: (1) a lighter fraction, (2) a moderate fraction, and (3) a heavy fraction. Alkaline gels, Illumina Mi-Seq, size exclusion chromatography with multi-angle light scattering (SEC-MALS), and charge detection mass spectrometry (CD-MS) were used to characterize the genome of each population and ddPCR to quantify residual DNA molecules. Live-cell imaging and EGFP ELISA assays demonstrated reduced expression following transduction with the light fraction compared with the moderate and heavy fractions. However, PCR-based assays showed that the light density delivered EGFP DNA to cells as efficiently as the moderate and heavy fractions. Mi-Seq data revealed an underrepresentation of the promoter region for EGFP, suggesting that expression of EGFP was reduced because of lack of regulatory control. This work demonstrates that rAAVs containing partial genomes contribute to the DNA signal but have reduced vector performance.

2.
Hum Gene Ther ; 34(7-8): 325-338, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36927085

RESUMEN

To provide safe recombinant adeno-associated viruses (rAAV) to patients, scalable manufacturing processes are required. However, these processes may introduce impurities that impact the performance and quality of the final drug product. Empty rAAV capsids are product-related impurities. Regulatory guidance requires that accurate analytical methods be implemented early in product development to measure the level of empty capsids. A process confirmation vector, produced from 200 L production, was used to develop and optimize a size exclusion chromatography with UV and multiangle light scattering (SEC-MALS) method. Vector produced from a 500 L production was used to assess the full-to-empty ratio using the following analytical methods: sedimentation velocity analytical ultracentrifugation (SV-AUC), droplet digital PCR (ddPCR) with capsid enzyme-linked immunosorbent assay (ELISA), bulk absorbance at 260/280 nm, cryogenic electron microscopy, and SEC-MALS. This test article was used for a 30-day, non-Good Laboratory Practices animal study that assessed biodistribution of the product (STRX-330). SEC-MALS outperformed the other methods and correlated well with SV-AUC values of full-to-empty particles. In addition, SEC-MALS agreed with ddPCR and ELISA measurements for vector genomes/mL and capsid particles/mL, respectively. SEC-MALS was linear, accurate, and precise while achieving chromatography quality control (QC) recommendations. Compared to other stability-indicating assays, SEC-MALS performed similarly to ddPCR, capsid ELISA, and infectivity assays in accelerated stress studies. In response to alkaline, but not acidic stress, SEC-MALS indicated distinct changes in the DNA content of the monomer Adeno-associated viruses (AAV) peak for STRX-330, which was supported by ddPCR data. Conversely, acidic treatment resulted in more aggregated vector, but did not impact the DNA content. This work indicates that SEC-MALS is a valuable analytical tool in the analytical development and QC testing of AAV. In addition, this work suggests that SEC-MALS can provide fundamental understanding of AAV in response to environmental stress. This may impact steps of the manufacturing process to minimize conditions that reduce performance.


Asunto(s)
ADN , Terapia Genética , Distribución Tisular , Cromatografía en Gel , Reacción en Cadena de la Polimerasa , Dependovirus/genética
3.
Vaccines (Basel) ; 8(4)2020 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-33153043

RESUMEN

Non-typhoidal Salmonella (NTS) serovars are significant health burden worldwide. Although much effort has been devoted to developing typhoid-based vaccines for humans, currently there is no NTS vaccine available. Presented here is the efficacy of a live attenuated serovar Typhimurium strain (NC983). Oral delivery of strain NC983 was capable of fully protecting C57BL/6 and BALB/c mice against challenge with virulent Typhimurium. Strain NC983 was found to elicit an anti-Typhimurium IgG response following administration of vaccine and boosting doses. Furthermore, in competition experiments with virulent S. Typhimurium (ATCC 14028), NC983 was highly defective in colonization of the murine liver and spleen. Collectively, these results indicate that strain NC983 is a potential live attenuated vaccine strain that warrants further development.

4.
J Bacteriol ; 200(22)2018 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-30181123

RESUMEN

Borrelia burgdorferi, the causative agent of Lyme disease, encounters two disparate host environments during its enzootic life cycle, Ixodes ticks and mammalian hosts. B. burgdorferi has a small genome that encodes a streamlined cyclic dimeric GMP (c-di-GMP) signaling system comprising a single diguanylate cyclase, Rrp1, and two phosphodiesterases. This system is essential for spirochete survival in ticks, in part because it controls the expression of the glp operon involved in glycerol utilization. In this study, we showed that a B. burgdorferi c-di-GMP receptor, PlzA, functions as both a positive and a negative regulator for glp expression. Deletion of plzA or mutation in plzA that impaired c-di-GMP binding abolished glp expression. On the other hand, overexpression of plzA resulted in glp repression, which could be rescued by simultaneous overexpression of rrp1. plzA overexpression in the rrp1 mutant, which is devoid of c-di-GMP, or overexpression of a plzA mutant incapable of c-di-GMP binding further enhanced glp repression. Combined results suggest that c-di-GMP-bound PlzA functions as a positive regulator, whereas ligand-free PlzA acts as a negative regulator for glp expression. Thus, PlzA of B. burgdorferi with a streamlined c-di-GMP signaling system not only controls multiple targets, as previously envisioned, but has also evolved different modes of action.IMPORTANCE The Lyme disease pathogen, Borrelia burgdorferi, has a simple cyclic dimeric GMP (c-di-GMP) signaling system essential for adaptation of the pathogen to the complicated tick environment. The c-di-GMP effector of B. burgdorferi, PlzA, has been shown to regulate multiple cellular processes, including motility, osmolality sensing, and nutrient utilization. The findings of this study demonstrate that PlzA not only controls multiple targets but also has different functional modalities, allowing it to act as both positive and negative regulator of the glp operon expression. This work highlights how bacteria with a small genome can compensate for the limited regulatory repertoire by increasing the complexity of targets and modes of action in their regulatory proteins.


Asunto(s)
Proteínas Bacterianas/metabolismo , Borrelia burgdorferi/metabolismo , Proteínas Portadoras/metabolismo , Glicerol/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Animales , Proteínas Bacterianas/genética , Borrelia burgdorferi/genética , Proteínas Portadoras/genética , Regulación Bacteriana de la Expresión Génica , Péptidos y Proteínas de Señalización Intracelular/genética , Operón , Unión Proteica , Transducción de Señal
5.
Virulence ; 9(1): 585-587, 2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-29380670

RESUMEN

Bacteria interact with their host through protein secretion systems and surface structures. Pathogenic bacteria encode protein secretion systems that promote the invasion of the host's tissue, the evasion of the host's immune response, the thwarting microbial competitors, and ultimately survival within the host. For motile bacteria, the presence of extracellular flagella provides the host with a structural motif used for activation of the immune system. Within this issue of Virulence, the article "Identification of a novel gene in ROD9 island of Salmonella Enteritidis involved in the alteration of virulence-associated protein expression" describes the contribution of a gene, SEN1005, toward host-pathogen interaction. The authors demonstrate the contribution of SEN1005 to cell culture bioassays and infection in a mouse model of colitis. In each tested scenario, deletion of SEN1005 results in a phenotypic defect that was complemented by providing the SEN1005 gene in trans. SEN1005 contributes to the expression of known virulence factors within SPI-1, flagellar and chemotaxis genes, and heat shock/chaperone genes. Although much work is needed to fully elucidate the function of SEN1005, this work contributes toward our understanding of the genetic factors used by Salmonella to cause foodborne illnesses.


Asunto(s)
Salmonella enteritidis , Sistemas de Secreción Tipo VI , Animales , Proteínas Bacterianas , Interacciones Huésped-Patógeno , Péptidos y Proteínas de Señalización Intercelular , Péptidos , Salmonella typhimurium , Virulencia
6.
Genome Announc ; 4(5)2016 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-27738027

RESUMEN

Foodborne infections caused by Salmonella enterica serovars are a significant problem worldwide. Presented here is the genome sequence of the nontyphoidal S. enterica serovar Typhimurium mutant strain NC983, a potential vaccine candidate.

7.
Poult Sci ; 95(7): 1617-1623, 2016 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-26994203

RESUMEN

Salmonella enterica serovar Typhimurium (S. Typhimurium) is a leading cause of salmonellosis. Poultry and poultry products are implicated in transmission of Salmonella to humans. In 2013, an outbreak of S Typhimurium occurred that comprised 39 states within the United States and was associated with backyard flocks of chickens. Colonization of the avian host by S Typhimurium requires numerous genetic factors encoded within the bacterium. Of particular interest are genetic factors induced by alternative sigma factors within S Typhimurium since these genetic elements are important for adaptation to different environmental stresses. The heat shock response is a dedicated change in gene regulation within bacteria in response to several stresses, specifically growth at 42°C. Because chickens have a higher body temperature than other animals (42°C) the hypothesis was tested that components of the heat shock response are important for optimal fitness within the chicken. To this end, deletion of the heat shock proteases clpPX (BTNC0022) or lon (BTNC0021) was accomplished and the bacterial fitness in vivo was compared to the "wild-type" strain (NC1040) using a competition assay. One-day-old chicks were orally gavaged with an equal mixture of NC1040 and either BTNC0022 or BTNC0021. Quantification of viable bacteria over time by using plate counts indicated that deletion of either heat shock protease resulted in significantly reduced colonization of the chicken ceca compared to the wild-type strain. To satisfy the molecular Koch's postulates, clpPX and lon mutants were complemented in trans using a low-copy number plasmid for additional in vivo experiments. Complementation studies confirmed the importance of either heat shock protease to colonization of the chicken ceca. This report demonstrated that both ClpPX and Lon were important for optimal fitness within chickens. Moreover, these results suggested that components of the heat shock may be critical factors used by S. Typhimurium for colonization of poultry. The use of feed additives or other treatments that inactivate or inhibit ClpPX or Lon may reduce the bacterial burden of S. Typhimurium in poultry.


Asunto(s)
Proteínas Bacterianas/genética , Pollos , Aptitud Genética , Enfermedades de las Aves de Corral/microbiología , Salmonelosis Animal/microbiología , Salmonella typhimurium/fisiología , Animales , Proteínas Bacterianas/metabolismo , Ciego/microbiología , Respuesta al Choque Térmico , Proteasa La/genética , Proteasa La/metabolismo , Salmonella typhimurium/genética , Factor sigma/genética , Factor sigma/metabolismo
8.
Infect Immun ; 83(12): 4848-60, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26438793

RESUMEN

Outer surface protein C (OspC) is one of the major lipoproteins expressed on the surface of Borrelia burgdorferi during tick feeding and the early phase of mammalian infection. OspC is required for B. burgdorferi to establish infection in both immunocompetent and SCID mice and has been proposed to facilitate evasion of innate immune defenses. However, the exact biological function of OspC remains elusive. In this study, we showed that the ospC-deficient spirochete could not establish infection in NOD-scid IL2rγ(null) mice that lack B cells, T cells, NK cells, and lytic complement. The ospC mutant also could not establish infection in anti-Ly6G-treated SCID and C3H/HeN mice (depletion of neutrophils). However, depletion of mononuclear phagocytes at the skin site of inoculation in SCID and C3H/HeN mice allowed the ospC mutant to establish infection in vivo. In phagocyte-depleted mice, the ospC mutant was able to colonize the joints and triggered neutrophilia during dissemination. Furthermore, we found that phagocytosis of green fluorescent protein (GFP)-expressing ospC mutant spirochetes by murine peritoneal macrophages and human THP-1 macrophage-like cells, but not in PMN-HL60, was significantly higher than parental wild-type B. burgdorferi strains, suggesting that OspC has an antiphagocytic property. In addition, overproduction of OspC in spirochetes also decreased the uptake of spirochetes by murine peritoneal macrophages. Together, our findings provide evidence that mononuclear phagocytes play a key role in clearance of the ospC mutant and that OspC promotes spirochetes' evasion of macrophages during early Lyme borreliosis.


Asunto(s)
Antígenos Bacterianos/inmunología , Proteínas de la Membrana Bacteriana Externa/inmunología , Borrelia burgdorferi/genética , Regulación Bacteriana de la Expresión Génica , Evasión Inmune , Enfermedad de Lyme/inmunología , Macrófagos Peritoneales/inmunología , Animales , Antígenos Bacterianos/genética , Linfocitos B/inmunología , Linfocitos B/microbiología , Linfocitos B/patología , Proteínas de la Membrana Bacteriana Externa/genética , Borrelia burgdorferi/inmunología , Borrelia burgdorferi/patogenicidad , Línea Celular , Femenino , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/microbiología , Células Asesinas Naturales/patología , Enfermedad de Lyme/genética , Enfermedad de Lyme/microbiología , Enfermedad de Lyme/patología , Macrófagos Peritoneales/microbiología , Macrófagos Peritoneales/patología , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Neutrófilos/inmunología , Neutrófilos/microbiología , Neutrófilos/patología , Linfocitos T/inmunología , Linfocitos T/microbiología , Linfocitos T/patología
9.
Appl Environ Microbiol ; 81(23): 8192-201, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26386070

RESUMEN

Salmonella enterica serovars Typhimurium (S. Typhimurium) and Enteritidis (S. Enteritidis) are foodborne pathogens, and outbreaks are often associated with poultry products. Chickens are typically asymptomatic when colonized by these serovars; however, the factors contributing to this observation are uncharacterized. Whereas symptomatic mammals have a body temperature between 37°C and 39°C, chickens have a body temperature of 41°C to 42°C. Here, in vivo experiments using chicks demonstrated that numbers of viable S. Typhimurium or S. Enteritidis bacteria within the liver and spleen organ sites were ≥4 orders of magnitude lower than those within the ceca. When similar doses of S. Typhimurium or S. Enteritidis were given to C3H/HeN mice, the ratio of the intestinal concentration to the liver/spleen concentration was 1:1. In the avian host, this suggested poor survival within these tissues or a reduced capacity to traverse the host epithelial layer and reach liver/spleen sites or both. Salmonella pathogenicity island 1 (SPI-1) promotes localization to liver/spleen tissues through invasion of the epithelial cell layer. Following in vitro growth at 42°C, SPI-1 genes sipC, invF, and hilA and the SPI-1 rtsA activator were downregulated compared to expression at 37°C. Overexpression of the hilA activators fur, fliZ, and hilD was capable of inducing hilA-lacZ at 37°C but not at 42°C despite the presence of similar levels of protein at the two temperatures. In contrast, overexpression of either hilC or rtsA was capable of inducing hilA and sipC at 42°C. These data indicate that physiological parameters of the poultry host, such as body temperature, have a role in modulating expression of virulence.


Asunto(s)
Pollos , Islas Genómicas , Enfermedades de las Aves de Corral/genética , Salmonelosis Animal/genética , Salmonella enteritidis/fisiología , Salmonella typhimurium/fisiología , Animales , Ciego/microbiología , Ciego/fisiología , Hígado/microbiología , Hígado/fisiología , Enfermedades de las Aves de Corral/metabolismo , Enfermedades de las Aves de Corral/microbiología , Salmonelosis Animal/metabolismo , Salmonelosis Animal/microbiología , Salmonella enteritidis/genética , Salmonella typhimurium/genética , Bazo/microbiología , Bazo/fisiología , Temperatura
10.
Emerg Microbes Infect ; 4: e54, 2015 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-26954993

RESUMEN

Borrelia burgdorferi, the etiological agent of Lyme disease, does not produce lipopolysaccharide but expresses a large number of lipoproteins on its cell surface. These outer membrane lipoproteins are highly immunogenic and have been used for serodiagnosis of Lyme disease. Recent studies have shown that highly conserved cytosolic proteins such as enolase and elongation factor Tu (EF-Tu) unexpectedly localized on the surface of bacteria including B. burgdorferi, and surface-localized enolase has shown to contribute to the enzootic cycle of B. burgdorferi. In this study, we studied the immunogenicity, surface localization, and function of B. burgdorferi EF-Tu. We found that EF-Tu is highly immunogenic in mice, and EF-Tu antibodies were readily detected in Lyme disease patients. On the other hand, active immunization studies showed that EF-Tu antibodies did not protect mice from infection when challenged with B. burgdorferi via either needle inoculation or tick bites. Borrelial mouse-tick cycle studies showed that EF-Tu antibodies also did not block B. burgdorferi migration and survival in ticks. Consistent with these findings, we found that EF-Tu primarily localizes in the protoplasmic cylinder of spirochetes and is not on the surface of B. burgdorferi. Taken together, our studies suggest that B. burgdorferi EF-Tu is not surfaced exposed, but it is highly immunogenic and is a potential serodiagnostic marker for Lyme borreliosis.


Asunto(s)
Antígenos Bacterianos/inmunología , Borrelia burgdorferi/inmunología , Enfermedad de Lyme/inmunología , Factor Tu de Elongación Peptídica/inmunología , Animales , Anticuerpos Antibacterianos/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/inmunología , Borrelia burgdorferi/genética , Humanos , Ixodes/microbiología , Larva , Lipoproteínas/genética , Lipoproteínas/inmunología , Enfermedad de Lyme/microbiología , Ratones , Ratones Endogámicos C3H , Factor Tu de Elongación Peptídica/genética , Proteínas Recombinantes
11.
Infect Immun ; 82(5): 1840-9, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24566626

RESUMEN

Cyclic di-AMP (c-di-AMP) is a recently discovered second messenger in bacteria. Most of work on c-di-AMP signaling has been done in Gram-positive bacteria, firmicutes, and actinobacteria, where c-di-AMP signaling pathways affect potassium transport, cell wall structure, and antibiotic resistance. Little is known about c-di-AMP signaling in other bacteria. Borrelia burgdorferi, the causative agent of Lyme disease, is a spirochete that has a Gram-negative dual membrane. In this study, we demonstrated that B. burgdorferi BB0619, a DHH-DHHA1 domain protein (herein designated DhhP), functions as c-di-AMP phosphodiesterase. Recombinant DhhP hydrolyzed c-di-AMP to pApA in a Mn(2+)- or Mg(2+)-dependent manner. In contrast to c-di-AMP phosphodiesterases reported thus far, DhhP appears to be essential for B. burgdorferi growth both in vitro and in the mammalian host. Inactivation of the chromosomal dhhP gene could be achieved only in the presence of a plasmid-encoded inducible dhhP gene. The conditional dhhP mutant had a dramatic increase in intracellular c-di-AMP level in comparison to the isogenic wild-type strain. Unlike what has been observed in Gram-positive bacteria, elevated cellular c-di-AMP in B. burgdorferi did not result in an increased resistance to ß-lactamase antibiotics, suggesting that c-di-AMP's functions in spirochetes differ from those in Gram-positive bacteria. In addition, the dhhP mutant was defective in induction of the σ(S) factor, RpoS, and the RpoS-dependent outer membrane virulence factor OspC, which uncovers an important role of c-di-AMP in B. burgdorferi virulence.


Asunto(s)
Proteínas Bacterianas/metabolismo , Borrelia burgdorferi/citología , Borrelia burgdorferi/enzimología , Enfermedad de Lyme/microbiología , Hidrolasas Diéster Fosfóricas/metabolismo , Animales , Antibacterianos/farmacología , Antígenos Bacterianos/genética , Antígenos Bacterianos/metabolismo , Proteínas de la Membrana Bacteriana Externa/genética , Proteínas de la Membrana Bacteriana Externa/metabolismo , Proteínas Bacterianas/genética , Técnicas Bacteriológicas , Borrelia burgdorferi/patogenicidad , Farmacorresistencia Bacteriana/genética , Electroforesis en Gel de Poliacrilamida , Eliminación de Gen , Regulación Bacteriana de la Expresión Génica , Immunoblotting , Ratones , Hidrolasas Diéster Fosfóricas/genética , Estructura Terciaria de Proteína , Transducción de Señal , Virulencia
12.
PLoS One ; 9(1): e84625, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24392147

RESUMEN

Pathogenic spirochetes cause clinically relevant diseases in humans and animals, such as Lyme disease and leptospirosis. The causative agent of Lyme disease, Borrelia burgdorferi, and the causative agent of leptospirosis, Leptospria interrogans, encounter reactive oxygen species (ROS) during their enzootic cycles. This report demonstrated that physiologically relevant concentrations of pyruvate, a potent H2O2 scavenger, and provided passive protection to B. burgdorferi and L. interrogans against H2O2. When extracellular pyruvate was absent, both spirochetes were sensitive to a low dose of H2O2 (≈0.6 µM per h) generated by glucose oxidase (GOX). Despite encoding a functional catalase, L. interrogans was more sensitive than B. burgdorferi to H2O2 generated by GOX, which may be due to the inherent resistance of B. burgdorferi because of the virtual absence of intracellular iron. In B. burgdorferi, the nucleotide excision repair (NER) and the DNA mismatch repair (MMR) pathways were important for survival during H2O2 challenge since deletion of the uvrB or the mutS genes enhanced its sensitivity to H2O2 killing; however, the presence of pyruvate fully protected ΔuvrB and ΔmutS from H2O2 killing further demonstrating the importance of pyruvate in protection. These findings demonstrated that pyruvate, in addition to its classical role in central carbon metabolism, serves as an important H2O2 scavenger for pathogenic spirochetes. Furthermore, pyruvate reduced ROS generated by human neutrophils in response to the Toll-like receptor 2 (TLR2) agonist zymosan. In addition, pyruvate reduced neutrophil-derived ROS in response to B. burgdorferi, which also activates host expression through TLR2 signaling. Thus, pathogenic spirochetes may exploit the metabolite pyruvate, present in blood and tissues, to survive H2O2 generated by the host antibacterial response generated during infection.


Asunto(s)
Peróxido de Hidrógeno/toxicidad , Ácido Pirúvico/metabolismo , Spirochaetales/efectos de los fármacos , Spirochaetales/metabolismo , Acetatos/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Borrelia burgdorferi/efectos de los fármacos , Borrelia burgdorferi/genética , Borrelia burgdorferi/metabolismo , Reparación del ADN/efectos de los fármacos , Eliminación de Gen , Glucosa Oxidasa/metabolismo , Humanos , Proteína MutS de Unión a los Apareamientos Incorrectos del ADN/genética , Proteína MutS de Unión a los Apareamientos Incorrectos del ADN/metabolismo , Neutrófilos/metabolismo , Neutrófilos/microbiología , Estrés Oxidativo/efectos de los fármacos , Ácido Pirúvico/farmacología , Especies Reactivas de Oxígeno/metabolismo , Spirochaetales/genética
13.
Infect Immun ; 82(1): 333-40, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24166960

RESUMEN

Herein we report an important role for the ferric uptake regulator (Fur) in the resistance of Salmonella enterica serovar Typhimurium to the reactive nitrogen species produced by inducible nitric oxide (NO) synthase in an NRAMP1(r) murine model of acute systemic infection. The expression of fur protected Salmonella grown under normoxic and hypoxic conditions against the bacteriostatic activity of NO. The hypersusceptibility of fur-deficient Salmonella to the cytotoxic actions of NO coincides with a marked repression of respiratory activity and the reduced ability of the bacteria to detoxify NO. A fur mutant Salmonella strain contained reduced levels of the terminal quinol oxidases of the electron transport chain. Addition of the heme precursor δ-aminolevulinic acid restored the cytochrome content, respiratory activity, NO consumption, and wild-type growth in bacteria undergoing nitrosative stress. The innate antinitrosative defenses regulated by Fur added to the adaptive response associated with the NO-detoxifying activity of the flavohemoprotein Hmp. Our investigations indicate that, in addition to playing a critical role in iron homeostasis, Fur is an important antinitrosative determinant of Salmonella pathogenesis.


Asunto(s)
Proteínas Bacterianas/fisiología , Proteínas Represoras/fisiología , Infecciones por Salmonella/microbiología , Salmonella typhimurium/patogenicidad , Análisis de Varianza , Animales , Proteínas Bacterianas/inmunología , Modelos Animales de Enfermedad , Ratones , Óxido Nítrico Sintasa de Tipo II , Estrés Oxidativo/fisiología , Proteínas Represoras/deficiencia , Proteínas Represoras/inmunología , Infecciones por Salmonella/fisiopatología , Salmonella typhimurium/inmunología , Estrés Fisiológico/fisiología
14.
Artículo en Inglés | MEDLINE | ID: mdl-24298449

RESUMEN

Borrelia burgdorferi (Bb) is the causative agent of Lyme disease transmitted to humans by ticks of the Ixodes spp. Bb is a unique bacterial pathogen because it does not require iron (Fe(2+)) for its metabolism. Bb encodes a ferritin-like Dps homolog called NapA (also called BicA), which can bind Fe or copper (Cu(2+)), and a manganese (Mn(2+)) transport protein, Borrelia metal transporter A (BmtA); both proteins are required for colonization of the tick vector, but BmtA is also required for the murine host. This demonstrates that Bb's metal homeostasis is a critical facet of the complex enzootic life cycle between the arthropod and murine hosts. Although metals are known to influence the expression of virulence determinants during infection, it is unknown how or if metals regulate virulence in Bb. Recent evidence demonstrates that Bb modulates the intracellular Mn(2+) and zinc (Zn(2+)) content and, in turn, these metals regulate gene expression through influencing the Ferric Uptake Regulator (Fur) homolog Borrelia Oxidative Stress Regulator (BosR). This mini-review focuses on the burgeoning study of metal-dependent gene regulation within Bb.


Asunto(s)
Borrelia burgdorferi/genética , Borrelia burgdorferi/metabolismo , Regulación Bacteriana de la Expresión Génica/efectos de los fármacos , Metales/metabolismo , Animales , Homeostasis , Humanos , Ixodes , Ratones
15.
Artículo en Inglés | MEDLINE | ID: mdl-24106689

RESUMEN

In the ancient anaerobic environment, ferrous iron (Fe(2+)) was one of the first metal cofactors. Oxygenation of the ancient world challenged bacteria to acquire the insoluble ferric iron (Fe(3+)) and later to defend against reactive oxygen species (ROS) generated by the Fenton chemistry. To acquire Fe(3+), bacteria produce low-molecular weight compounds, known as siderophores, which have extremely high affinity for Fe(3+). However, during infection the host restricts iron from pathogens by producing iron- and siderophore-chelating proteins, by exporting iron from intracellular pathogen-containing compartments, and by limiting absorption of dietary iron. Ferric Uptake Regulator (Fur) is a transcription factor which utilizes Fe(2+) as a corepressor and represses siderophore synthesis in pathogens. Fur, directly or indirectly, controls expression of enzymes that protect against ROS damage. Thus, the challenges of iron homeostasis and defense against ROS are addressed via Fur. Although the role of Fur as a repressor is well-documented, emerging evidence demonstrates that Fur can function as an activator. Fur activation can occur through three distinct mechanisms (1) indirectly via small RNAs, (2) binding at cis regulatory elements that enhance recruitment of the RNA polymerase holoenzyme (RNAP), and (3) functioning as an antirepressor by removing or blocking DNA binding of a repressor of transcription. In addition, Fur homologs control defense against peroxide stress (PerR) and control uptake of other metals such as zinc (Zur) and manganese (Mur) in pathogenic bacteria. Fur family members are important for virulence within bacterial pathogens since mutants of fur, perR, or zur exhibit reduced virulence within numerous animal and plant models of infection. This review focuses on the breadth of Fur regulation in pathogenic bacteria.


Asunto(s)
Bacterias/genética , Bacterias/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Regulación Bacteriana de la Expresión Génica , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Transcripción Genética , Animales , Bacterias/aislamiento & purificación , Infecciones Bacterianas/microbiología , Compuestos Férricos/metabolismo , Interacciones Huésped-Patógeno , Humanos , Estrés Oxidativo , Especies Reactivas de Oxígeno/metabolismo , Especies Reactivas de Oxígeno/toxicidad
16.
Infect Immun ; 81(8): 2743-52, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23690398

RESUMEN

Borrelia burgdorferi, the causative agent of Lyme disease, must adapt to two diverse niches, an arthropod vector and a mammalian host. RpoS, an alternative sigma factor, plays a central role in spirochetal adaptation to the mammalian host by governing expression of many genes important for mammalian infection. B. burgdorferi is known to be unique in metal utilization, and little is known of the role of biologically available metals in B. burgdorferi. Here, we identified two transition metal ions, manganese (Mn(2+)) and zinc (Zn(2+)), that influenced regulation of RpoS. The intracellular Mn(2+) level fluctuated approximately 20-fold under different conditions and inversely correlated with levels of RpoS and the major virulence factor OspC. Furthermore, an increase in intracellular Mn(2+) repressed temperature-dependent induction of RpoS and OspC; this repression was overcome by an excess of Zn(2+). Conversely, a decrease of intracellular Mn(2+) by deletion of the Mn(2+) transporter gene, bmtA, resulted in elevated levels of RpoS and OspC. Mn(2+) affected RpoS through BosR, a Fur family homolog that is required for rpoS expression: elevated intracellular Mn(2+) levels greatly reduced the level of BosR protein but not the level of bosR mRNA. Thus, Mn(2+) and Zn(2+) appeared to be important in modulation of the RpoS pathway that is essential to the life cycle of the Lyme disease spirochete. This finding supports the emerging notion that transition metals such as Mn(2+) and Zn(2+) play a critical role in regulation of virulence in bacteria.


Asunto(s)
Borrelia burgdorferi/patogenicidad , Regulación Bacteriana de la Expresión Génica/fisiología , Manganeso/metabolismo , Zinc/metabolismo , Antígenos Bacterianos/biosíntesis , Proteínas de la Membrana Bacteriana Externa/biosíntesis , Proteínas Bacterianas/biosíntesis , Borrelia burgdorferi/genética , Borrelia burgdorferi/metabolismo , Immunoblotting , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor sigma/biosíntesis , Virulencia/fisiología , Factores de Virulencia/metabolismo
17.
J Biol Chem ; 287(23): 19284-93, 2012 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-22500025

RESUMEN

Borrelia burgdorferi, the causative agent of Lyme disease, exists in nature through a complex life cycle involving ticks of the Ixodes genus and mammalian hosts. During its life cycle, B. burgdorferi experiences fluctuations in oxygen tension and may encounter reactive oxygen species (ROS). The key metalloenzyme to degrade ROS in B. burgdorferi is SodA. Although previous work suggests that B. burgdorferi SodA is an iron-dependent superoxide dismutase (SOD), later work demonstrates that B. burgdorferi is unable to transport iron and contains an extremely low intracellular concentration of iron. Consequently, the metal cofactor for SodA has been postulated to be manganese. However, experimental evidence to support this hypothesis remains lacking. In this study, we provide biochemical and genetic data showing that SodA is a manganese-dependent enzyme. First, B. burgdorferi contained SOD activity that is resistant to H(2)O(2) and NaCN, characteristics associated with Mn-SODs. Second, the addition of manganese to the Chelex-treated BSK-II enhanced SodA expression. Third, disruption of the manganese transporter gene bmtA, which significantly lowers the intracellular manganese, greatly reduced SOD activity and SodA expression, suggesting that manganese regulates the level of SodA. In addition, we show that B. burgdorferi is resistant to streptonigrin, a metal-dependent redox cycling compound that produces ROS, and that SodA plays a protective role against the streptonigrin. Taken together, our data demonstrate the Lyme disease spirochete encodes a manganese-dependent SOD that contributes to B. burgdorferi defense against intracellular superoxide.


Asunto(s)
Antibióticos Antineoplásicos/farmacología , Proteínas Bacterianas/metabolismo , Borrelia burgdorferi/enzimología , Farmacorresistencia Bacteriana/fisiología , Hierro , Estreptonigrina/farmacología , Superóxido Dismutasa/metabolismo , Proteínas Bacterianas/antagonistas & inhibidores , Proteínas Bacterianas/genética , Borrelia burgdorferi/genética , Farmacorresistencia Bacteriana/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Humanos , Transporte Iónico/efectos de los fármacos , Transporte Iónico/fisiología , Oxidantes/farmacología , Cianuro de Sodio/farmacología , Hidróxido de Sodio/farmacología , Superóxido Dismutasa/antagonistas & inhibidores , Superóxido Dismutasa/genética , Superóxidos/metabolismo
18.
BMC Microbiol ; 11: 236, 2011 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-22017966

RESUMEN

BACKGROUND: The Ferric uptake regulator (Fur) is a transcriptional regulator that controls iron homeostasis in bacteria. Although the regulatory role of Fur in Escherichia coli is well characterized, most of the studies were conducted under routine culture conditions, i.e., in ambient oxygen concentration. To reveal potentially novel aspects of the Fur regulon in Salmonella enterica serovar Typhimurium under oxygen conditions similar to that encountered in the host, we compared the transcriptional profiles of the virulent wild-type strain (ATCC 14028s) and its isogenic Δfur strain under anaerobic conditions. RESULTS: Microarray analysis of anaerobically grown Δfur S. Typhimurium identified 298 differentially expressed genes. Expression of several genes controlled by Fnr and NsrR appeared to be also dependent on Fur. Furthermore, Fur was required for the activity of the cytoplasmic superoxide disumutases (MnSOD and FeSOD). The regulation of FeSOD gene, sodB, occurred via small RNAs (i.e., the ryhB homologs, rfrA and rfrB) with the aid of the RNA chaperone Hfq. The transcription of sodA was increased in Δfur; however, the enzyme was inactive due to the incorporation of iron instead of manganese in SodA. Additionally, in Δfur, the expression of the gene coding for the ferritin-like protein (ftnB) was down-regulated, while the transcription of the gene coding for the nitric oxide (NO·) detoxifying flavohemoglobin (hmpA) was up-regulated. The promoters of ftnB and hmpA do not contain recognized Fur binding motifs, which indicated their probable indirect regulation by Fur. However, Fur activation of ftnB was independent of Fnr. In addition, the expression of the gene coding for the histone-like protein, H-NS (hns) was increased in Δfur. This may explain the observed down-regulation of the tdc operon, responsible for the anaerobic degradation of threonine, and ftnB in Δfur. CONCLUSIONS: This study determined that Fur is a positive factor in ftnB regulation, while serving to repress the expression of hmpA. Furthermore, Fur is required for the proper expression and activation of the antioxidant enzymes, FeSOD and MnSOD. Finally, this work identified twenty-six new targets of Fur regulation, and demonstrates that H-NS repressed genes are down-regulated in Δfur.


Asunto(s)
Proteínas Bacterianas/metabolismo , Regulación Bacteriana de la Expresión Génica , Oxígeno/metabolismo , Regulón , Proteínas Represoras/metabolismo , Salmonella typhimurium/genética , Salmonella typhimurium/metabolismo , Anaerobiosis , Proteínas Bacterianas/genética , Hierro/metabolismo , Proteínas Represoras/genética
19.
PLoS Pathog ; 7(6): e1002133, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21738477

RESUMEN

Cyclic dimeric GMP (c-di-GMP) is a bacterial second messenger that modulates many biological processes. Although its role in bacterial pathogenesis during mammalian infection has been documented, the role of c-di-GMP in a pathogen's life cycle within a vector host is less understood. The enzootic cycle of the Lyme disease pathogen Borrelia burgdorferi involves both a mammalian host and an Ixodes tick vector. The B. burgdorferi genome encodes a single copy of the diguanylate cyclase gene (rrp1), which is responsible for c-di-GMP synthesis. To determine the role of c-di-GMP in the life cycle of B. burgdorferi, an Rrp1-deficient B. burgdorferi strain was generated. The rrp1 mutant remains infectious in the mammalian host but cannot survive in the tick vector. Microarray analyses revealed that expression of a four-gene operon involved in glycerol transport and metabolism, bb0240-bb0243, was significantly downregulated by abrogation of Rrp1. In vitro, the rrp1 mutant is impaired in growth in the media containing glycerol as the carbon source (BSK-glycerol). To determine the contribution of the glycerol metabolic pathway to the rrp1 mutant phenotype, a glp mutant, in which the entire bb0240-bb0243 operon is not expressed, was generated. Similar to the rrp1 mutant, the glp mutant has a growth defect in BSK-glycerol medium. In vivo, the glp mutant is also infectious in mice but has reduced survival in ticks. Constitutive expression of the bb0240-bb0243 operon in the rrp1 mutant fully rescues the growth defect in BSK-glycerol medium and partially restores survival of the rrp1 mutant in ticks. Thus, c-di-GMP appears to govern a catabolic switch in B. burgdorferi and plays a vital role in the tick part of the spirochetal enzootic cycle. This work provides the first evidence that c-di-GMP is essential for a pathogen's survival in its vector host.


Asunto(s)
Grupo Borrelia Burgdorferi , GMP Cíclico/análogos & derivados , Ixodes/metabolismo , Enfermedad de Lyme/transmisión , Animales , Grupo Borrelia Burgdorferi/genética , GMP Cíclico/metabolismo , Vectores de Enfermedades , Proteínas de Escherichia coli/genética , Técnicas de Inactivación de Genes , Glicerol/metabolismo , Ixodes/microbiología , Enfermedad de Lyme/microbiología , Ratones , Ratones Endogámicos C3H , Análisis por Micromatrices , Liasas de Fósforo-Oxígeno/genética , Reacción en Cadena de la Polimerasa , Eliminación de Secuencia
20.
J Bacteriol ; 193(2): 497-505, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21075923

RESUMEN

Iron is an essential element for the survival of living cells. However, excess iron is toxic, and its uptake is exquisitely regulated by the ferric uptake regulator, Fur. In Salmonella, the Salmonella pathogenicity island 1 (SPI-1) encodes a type three secretion system, which is required for invasion of host epithelial cells in the small intestine. A major activator of SPI-1 is HilA, which is encoded within SPI-1. One known regulator of hilA is Fur. The mechanism of hilA regulation by Fur is unknown. We report here that Fur is required for virulence in Salmonella enterica serovar Typhimurium and that Fur is required for the activation of hilA, as well as of other HilA-dependent genes, invF and sipC. The Fur-dependent regulation of hilA was independent of PhoP, a known repressor of hilA. Instead, the expression of the gene coding for the histone-like protein, hns, was significantly derepressed in the fur mutant. Indeed, the activation of hilA by Fur was dependent on 28 nucleotides located upstream of hns. Moreover, we used chromatin immunoprecipitation to show that Fur bound, in vivo, to the upstream region of hns in a metal-dependent fashion. Finally, deletion of fur in an hns mutant resulted in Fur-independent activation of hilA. In conclusion, Fur activates hilA by repressing the expression of hns.


Asunto(s)
Proteínas Bacterianas/biosíntesis , Proteínas Bacterianas/metabolismo , Proteínas de Unión al ADN/metabolismo , Regulación Bacteriana de la Expresión Génica , Proteínas Represoras/metabolismo , Salmonella typhimurium/fisiología , Transactivadores/biosíntesis , Factores de Virulencia/biosíntesis , Animales , Proteínas Bacterianas/genética , Inmunoprecipitación de Cromatina , ADN Bacteriano/metabolismo , Proteínas de Unión al ADN/biosíntesis , Modelos Animales de Enfermedad , Técnicas de Inactivación de Genes , Genes Bacterianos , Islas Genómicas , Ratones , Ratones Endogámicos C3H , Operón , Unión Proteica , Proteínas Represoras/genética , Salmonelosis Animal/microbiología , Salmonella typhimurium/patogenicidad , Análisis de Supervivencia , Factores de Transcripción/biosíntesis , Virulencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...