Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Genome Res ; 28(9): 1372-1382, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29914970

RESUMEN

Interactions mediated by cell surface receptors initiate important instructive signaling cues but can be difficult to detect in biochemical assays because they are often highly transient and membrane-embedded receptors are difficult to solubilize in their native conformation. Here, we address these biochemical challenges by using a genome-scale, cell-based genetic screening approach using CRISPR gene knockout technology to identify cellular pathways required for specific cell surface recognition events. By using high-affinity monoclonal antibodies and low-affinity ligands, we determined the necessary screening parameters, including the importance of establishing binding contributions from the glycocalyx, that permitted the unequivocal identification of genes encoding directly interacting membrane-embedded receptors with high statistical confidence. Importantly, we show that this genome-wide screening approach additionally identified receptor-specific pathways that are required for functional display of receptors on the cell surface that included chaperones, enzymes that add post-translational modifications, trafficking proteins, and transcription factors. Finally, we demonstrate the utility of the approach by identifying IGF2R (insulin like growth factor 2 receptor) as a binding partner for the R2 subunit of GABAB receptors. We show that this interaction is direct and is critically dependent on mannose-6-phosphate, providing a mechanism for the internalization and regulation of GABAB receptor signaling. We conclude that this single approach can reveal both the molecular nature and the genetic pathways required for functional cell surface display of receptors recognized by antibodies, secreted proteins, and membrane-embedded ligands without the need to make any prior assumptions regarding their biochemical properties.


Asunto(s)
Proteínas de la Membrana/metabolismo , Mapeo de Interacción de Proteínas/métodos , Proteoma/metabolismo , Transducción de Señal , Línea Celular Tumoral , Glicocálix/metabolismo , Células HEK293 , Humanos , Proteínas de la Membrana/genética , Unión Proteica , Procesamiento Proteico-Postraduccional , Transporte de Proteínas , Proteoma/genética
2.
Nature ; 480(7378): 534-7, 2011 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-22080952

RESUMEN

Erythrocyte invasion by Plasmodium falciparum is central to the pathogenesis of malaria. Invasion requires a series of extracellular recognition events between erythrocyte receptors and ligands on the merozoite, the invasive form of the parasite. None of the few known receptor-ligand interactions involved are required in all parasite strains, indicating that the parasite is able to access multiple redundant invasion pathways. Here, we show that we have identified a receptor-ligand pair that is essential for erythrocyte invasion in all tested P. falciparum strains. By systematically screening a library of erythrocyte proteins, we have found that the Ok blood group antigen, basigin, is a receptor for PfRh5, a parasite ligand that is essential for blood stage growth. Erythrocyte invasion was potently inhibited by soluble basigin or by basigin knockdown, and invasion could be completely blocked using low concentrations of anti-basigin antibodies; importantly, these effects were observed across all laboratory-adapted and field strains tested. Furthermore, Ok(a-) erythrocytes, which express a basigin variant that has a weaker binding affinity for PfRh5, had reduced invasion efficiencies. Our discovery of a cross-strain dependency on a single extracellular receptor-ligand pair for erythrocyte invasion by P. falciparum provides a focus for new anti-malarial therapies.


Asunto(s)
Basigina/metabolismo , Eritrocitos/parasitología , Interacciones Huésped-Parásitos , Plasmodium falciparum/fisiología , Basigina/química , Basigina/genética , Eritrocitos/metabolismo , Técnicas de Silenciamiento del Gen , Humanos , Modelos Moleculares , Estructura Terciaria de Proteína
3.
Malar J ; 14: 238, 2015 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-26045295

RESUMEN

BACKGROUND: Plasmodium parasites typically elicit a non-sterile but protective immune response in human host populations, suggesting that the parasites actively modulate normal immunological mechanisms. P-selectin is a cell surface receptor expressed in mammals, that is a known component of the inflammatory response against pathogens and has been previously identified as a host factor that influences malaria-associated pathology both in human patients and rodent infection models. METHODS: To better understand the molecular mechanisms underlying the involvement of P-selectin in the pathogenesis of malaria, a systematic extracellular protein interaction screen was used to identify Plasmodium falciparum merozoite surface protein 7 (MSP7) as a binding partner of human P-selectin. This interaction, and those occurring between P-selectin and Plasmodium MSP7 homologues, was characterized biochemically. RESULTS: Plasmodium falciparum MSP7 and P-selectin were shown to bind each other directly via the N-terminus of PfMSP7 and the P-selectin C-type lectin and EGF-like domains. Orthologous proteins in the murine parasite Plasmodium berghei (PbMSRP1 and PbMSRP2) and mouse P-selectin also interacted. Finally, P-selectin, when complexed with MSP7, could no longer bind to its endogenous carbohydrate ligand, Sialyl-Lewis(X). CONCLUSIONS: Novel interactions were identified between Plasmodium MSP7 protein family members and host P-selectin receptors. Since PfMSP7 could prevent interactions between P-selectin and its leukocyte ligands, these results provide a possible mechanism for the known immunomodulatory effects of both MSP7 and P-selectin in malaria infection models.


Asunto(s)
Lectinas Tipo C/metabolismo , Proteínas de la Membrana/genética , Oligosacáridos/metabolismo , Selectina-P/genética , Plasmodium falciparum/genética , Proteínas Protozoarias/genética , Animales , Humanos , Ligandos , Proteínas de la Membrana/metabolismo , Ratones , Selectina-P/metabolismo , Plasmodium falciparum/metabolismo , Proteínas Protozoarias/metabolismo , Antígeno Sialil Lewis X
4.
Malar J ; 14: 88, 2015 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-25889240

RESUMEN

BACKGROUND: Invasion of host erythrocytes by Plasmodium falciparum is central to the pathogenesis of malaria. Invasion involves recognition events between erythrocyte receptors and ligands on the merozoite, the invasive blood form of the parasite. Identifying and characterizing host-parasite interactions is impeded by the biochemical challenges of working with membrane-embedded glycoprotein receptors. For example, the interaction between P. falciparum erythrocyte binding antigen 175 (PfEBA175) and glycophorin A (GYPA) depends on post-translational modifications that are not easily added in recombinant expression systems, and the use of native GYPA is limited by the hydrophobic transmembrane region, making it difficult to biochemically manipulate. It would, therefore, be desirable to perform quantitative binding assays with receptors embedded within the membranes of intact human erythrocytes. METHODS: The extracellular region of GYPA was over-expressed as a soluble protein in HEK293E cells. This protein was characterized, sialylated and evaluated for binding to the PfEBA175 protein. The label-free and free-solution assay, backscattering interferometry (BSI), was used to perform binding assays of two well-characterized P. falciparum invasion ligands to intact unmodified human erythrocytes. RESULTS: Findings indicate that the post-translational modifications present on native GYPA are required for it to bind recombinant PfEBA175 and that these modifications cannot be recapitulated in vitro using mammalian overexpression methods. Here, BSI was used to obtain quantitative, high fidelity interaction determinations on intact, unmodified erythrocytes. Using BSI and purified recombinant proteins constituting the entire ectodomains of the P. falciparum merozoite ligands PfEBA175 and PfRH5, K Ds of 1.1 µM and 50 nM were measured for the PfRH5-BSG and PfEBA175-GYPA interactions, respectively, in good agreement with previous biophysical measurements of these interactions. CONCLUSIONS: These results demonstrate that BSI can be used to detect and quantify the interactions of two merozoite invasion ligands with their receptors on intact human erythrocytes. BSI assays were performed on unlabelled, free-solution proteins in their native environment, requiring only nanomoles of recombinant protein. This study suggests that BSI can be used to investigate host-parasite protein interactions without the limitations of other assay platforms, and therefore represents a valuable new method to investigate the molecular mechanisms involved in erythrocyte invasion by P. falciparum.


Asunto(s)
Antígenos de Protozoos/metabolismo , Proteínas Portadoras/metabolismo , Eritrocitos/parasitología , Glicoforinas/metabolismo , Interacciones Huésped-Parásitos , Plasmodium falciparum/fisiología , Procesamiento Proteico-Postraduccional , Proteínas Protozoarias/metabolismo , Antígenos de Protozoos/genética , Proteínas Portadoras/genética , Eritrocitos/metabolismo , Glicoforinas/genética , Interferometría , Malaria Falciparum/parasitología , Malaria Falciparum/fisiopatología , Plasmodium falciparum/genética , Proteínas Protozoarias/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
5.
PLoS Pathog ; 8(11): e1003031, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23166499

RESUMEN

The motility and invasion of Plasmodium parasites is believed to require a cytoplasmic actin-myosin motor associated with a cell surface ligand belonging to the TRAP (thrombospondin-related anonymous protein) family. Current models of invasion usually invoke the existence of specific receptors for the TRAP-family ligands on the surface of the host cell; however, the identities of these receptors remain largely unknown. Here, we identify the GPI-linked protein Semaphorin-7A (CD108) as an erythrocyte receptor for the P. falciparum merozoite-specific TRAP homolog (MTRAP) by using a systematic screening approach designed to detect extracellular protein interactions. The specificity of the interaction was demonstrated by showing that binding was saturable and by quantifying the equilibrium and kinetic biophysical binding parameters using surface plasmon resonance. We found that two MTRAP monomers interact via their tandem TSR domains with the Sema domains of a Semaphorin-7A homodimer. Known naturally-occurring polymorphisms in Semaphorin-7A did not quantitatively affect MTRAP binding nor did the presence of glycans on the receptor. Attempts to block the interaction during in vitro erythrocyte invasion assays using recombinant proteins and antibodies showed no significant inhibitory effect, suggesting the inaccessibility of the complex to proteinaceous blocking agents. These findings now provide important experimental evidence to support the model that parasite TRAP-family ligands interact with specific host receptors during cellular invasion.


Asunto(s)
Antígenos CD/metabolismo , Eritrocitos/metabolismo , Malaria Falciparum/metabolismo , Plasmodium falciparum/metabolismo , Proteínas Protozoarias/metabolismo , Semaforinas/metabolismo , Animales , Antígenos CD/genética , Eritrocitos/parasitología , Eritrocitos/patología , Femenino , Proteínas Ligadas a GPI/genética , Proteínas Ligadas a GPI/metabolismo , Humanos , Malaria Falciparum/genética , Malaria Falciparum/patología , Masculino , Plasmodium falciparum/genética , Plasmodium falciparum/patogenicidad , Polimorfismo Genético , Multimerización de Proteína/genética , Estructura Terciaria de Proteína , Proteínas Protozoarias/genética , Semaforinas/genética
6.
Cell Microbiol ; 15(8): 1304-12, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23617720

RESUMEN

The invasion of host erythrocytes by the parasite Plasmodium falciparum initiates the blood stage of infection responsible for the symptoms of malaria. Invasion involves extracellular protein interactions between host erythrocyte receptors and ligands on the merozoite, the invasive form of the parasite. Despite significant research effort, many merozoite surface ligands have no known erythrocyte binding partner, most likely due to the intractable biochemical nature of membrane-tethered receptor proteins and their interactions. The few receptor-ligand pairs that have been described have largely relied on sourcing erythrocytes from patients with rare blood groups, a serendipitous approach that is unsatisfactory for systematically identifying novel receptors. We have recently developed a scalable assay called AVEXIS (for AVidity-based EXtracellular Interaction Screen), designed to circumvent the technical difficulties associated with the identification of extracellular protein interactions, and applied it to identify erythrocyte receptors for orphan P. falciparum merozoite ligands. Using this approach, we have recently identified Basigin (CD147) and Semaphorin-7A (CD108) as receptors for RH5 and MTRAP respectively. In this essay, we review techniques used to identify Plasmodium receptors and discuss how they could be applied in the future to identify novel receptors both for Plasmodium parasites but also other pathogens.


Asunto(s)
Eritrocitos/parasitología , Plasmodium falciparum/metabolismo , Mapas de Interacción de Proteínas , Proteínas Protozoarias/metabolismo , Receptores de Superficie Celular/metabolismo , Animales , Basigina/metabolismo , Proteínas Portadoras/metabolismo , Humanos , Merozoítos/metabolismo , Plasmodium falciparum/aislamiento & purificación , Semaforinas/metabolismo
7.
PLoS Negl Trop Dis ; 9(12): e0004264, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26701602

RESUMEN

BACKGROUND: A vaccine targeting Plasmodium vivax will be an essential component of any comprehensive malaria elimination program, but major gaps in our understanding of P. vivax biology, including the protein-protein interactions that mediate merozoite invasion of reticulocytes, hinder the search for candidate antigens. Only one ligand-receptor interaction has been identified, that between P. vivax Duffy Binding Protein (PvDBP) and the erythrocyte Duffy Antigen Receptor for Chemokines (DARC), and strain-specific immune responses to PvDBP make it a complex vaccine target. To broaden the repertoire of potential P. vivax merozoite-stage vaccine targets, we exploited a recent breakthrough in expressing full-length ectodomains of Plasmodium proteins in a functionally-active form in mammalian cells and initiated a large-scale study of P. vivax merozoite proteins that are potentially involved in reticulocyte binding and invasion. METHODOLOGY/PRINCIPAL FINDINGS: We selected 39 P. vivax proteins that are predicted to localize to the merozoite surface or invasive secretory organelles, some of which show homology to P. falciparum vaccine candidates. Of these, we were able to express 37 full-length protein ectodomains in a mammalian expression system, which has been previously used to express P. falciparum invasion ligands such as PfRH5. To establish whether the expressed proteins were correctly folded, we assessed whether they were recognized by antibodies from Cambodian patients with acute vivax malaria. IgG from these samples showed at least a two-fold change in reactivity over naïve controls in 27 of 34 antigens tested, and the majority showed heat-labile IgG immunoreactivity, suggesting the presence of conformation-sensitive epitopes and native tertiary protein structures. Using a method specifically designed to detect low-affinity, extracellular protein-protein interactions, we confirmed a predicted interaction between P. vivax 6-cysteine proteins P12 and P41, further suggesting that the proteins are natively folded and functional. This screen also identified two novel protein-protein interactions, between P12 and PVX_110945, and between MSP3.10 and MSP7.1, the latter of which was confirmed by surface plasmon resonance. CONCLUSIONS/SIGNIFICANCE: We produced a new library of recombinant full-length P. vivax ectodomains, established that the majority of them contain tertiary structure, and used them to identify predicted and novel protein-protein interactions. As well as identifying new interactions for further biological studies, this library will be useful in identifying P. vivax proteins with vaccine potential, and studying P. vivax malaria pathogenesis and immunity. TRIAL REGISTRATION: ClinicalTrials.gov NCT00663546.


Asunto(s)
Antígenos de Protozoos/inmunología , Vacunas contra la Malaria/inmunología , Malaria Vivax/prevención & control , Plasmodium vivax/inmunología , Proteínas Protozoarias/inmunología , Factores de Edad , Animales , Anticuerpos Antiprotozoarios/inmunología , Antígenos de Protozoos/genética , Cambodia , Eritrocitos/parasitología , Biblioteca de Genes , Células HEK293 , Humanos , Malaria Vivax/parasitología , Masculino , Merozoítos/química , Merozoítos/inmunología , Plasmodium vivax/genética , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Proteínas Protozoarias/genética , Proteínas Recombinantes/inmunología , Vacunas Sintéticas/inmunología
8.
Cell Host Microbe ; 17(1): 130-9, 2015 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-25590760

RESUMEN

Antigenic diversity has posed a critical barrier to vaccine development against the pathogenic blood-stage infection of the human malaria parasite Plasmodium falciparum. To date, only strain-specific protection has been reported by trials of such vaccines in nonhuman primates. We recently showed that P. falciparum reticulocyte binding protein homolog 5 (PfRH5), a merozoite adhesin required for erythrocyte invasion, is highly susceptible to vaccine-inducible strain-transcending parasite-neutralizing antibody. In vivo efficacy of PfRH5-based vaccines has not previously been evaluated. Here, we demonstrate that PfRH5-based vaccines can protect Aotus monkeys against a virulent vaccine-heterologous P. falciparum challenge and show that such protection can be achieved by a human-compatible vaccine formulation. Protection was associated with anti-PfRH5 antibody concentration and in vitro parasite-neutralizing activity, supporting the use of this in vitro assay to predict the in vivo efficacy of future vaccine candidates. These data suggest that PfRH5-based vaccines have potential to achieve strain-transcending efficacy in humans.


Asunto(s)
Proteínas Portadoras/inmunología , Inmunidad Heteróloga , Vacunas contra la Malaria/inmunología , Malaria/prevención & control , Animales , Anticuerpos Neutralizantes/sangre , Anticuerpos Antiprotozoarios/sangre , Antígenos de Protozoos/inmunología , Aotus trivirgatus , Modelos Animales de Enfermedad , Femenino , Malaria/inmunología , Vacunas contra la Malaria/administración & dosificación , Pruebas de Neutralización
10.
PLoS One ; 8(2): e57848, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23451277

RESUMEN

We have identified a new protein domain, which we have named the SHOCT domain (Short C-terminal domain). This domain is widespread in bacteria with over a thousand examples. But we found it is missing from the most commonly studied model organisms, despite being present in closely related species. It's predominantly C-terminal location, co-occurrence with numerous other domains and short size is reminiscent of the Gram-positive anchor motif, however it is present in a much wider range of species. We suggest several hypotheses about the function of SHOCT, including oligomerisation and nucleic acid binding. Our initial experiments do not support its role as an oligomerisation domain.


Asunto(s)
Proteínas Bacterianas/química , Secuencia de Aminoácidos , Bacterias/química , Bacterias/metabolismo , Proteínas Bacterianas/metabolismo , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/metabolismo , Modelos Moleculares , Datos de Secuencia Molecular , Estructura Terciaria de Proteína , Homología de Secuencia de Aminoácido
11.
Vaccine ; 31(2): 373-9, 2013 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-23146673

RESUMEN

The lack of an effective licensed vaccine remains one of the most significant gaps in the portfolio of tools being developed to eliminate Plasmodium falciparum malaria. Vaccines targeting erythrocyte invasion - an essential step for both parasite development and malaria pathogenesis - have faced the particular challenge of genetic diversity. Immunity-driven balancing selection pressure on parasite invasion proteins often results in the presence of multiple, antigenically distinct, variants within a population, leading to variant-specific immune responses. Such variation makes it difficult to design a vaccine that covers the full range of diversity, and could potentially facilitate the evolution of vaccine-resistant parasite strains. In this study, we investigate the effect of genetic diversity on invasion inhibition by antibodies to a high priority P. falciparum invasion candidate antigen, P. falciparum Reticulocyte Binding Protein Homologue 5 (PfRH5). Previous work has shown that virally delivered PfRH5 can induce antibodies that protect against a wide range of genetic variants. Here, we show that a full-length recombinant PfRH5 protein expressed in mammalian cells is biochemically active, as judged by saturable binding to its receptor, basigin, and is able to induce antibodies that strongly inhibit P. falciparum growth and invasion. Whole genome sequencing of 290 clinical P. falciparum isolates from across the world identifies only five non-synonymous PfRH5 SNPs that are present at frequencies of 10% or more in at least one geographical region. Antibodies raised against the 3D7 variant of PfRH5 were able to inhibit nine different P. falciparum strains, which between them included all of the five most common PfRH5 SNPs in this dataset, with no evidence for strain-specific immunity. We conclude that protein-based PfRH5 vaccines are an urgent priority for human efficacy trials.


Asunto(s)
Proteínas Portadoras/genética , Proteínas Portadoras/inmunología , Vacunas contra la Malaria/inmunología , Malaria Falciparum/prevención & control , Malaria Falciparum/parasitología , Plasmodium falciparum/genética , Plasmodium falciparum/inmunología , Anticuerpos Antiprotozoarios/inmunología , Antígenos de Protozoos/genética , Antígenos de Protozoos/inmunología , Eritrocitos/inmunología , Eritrocitos/parasitología , Humanos , Vacunas contra la Malaria/genética , Malaria Falciparum/sangre , Malaria Falciparum/inmunología , Polimorfismo de Nucleótido Simple , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología
12.
PLoS One ; 7(7): e41937, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22848665

RESUMEN

The genomes of Plasmodium parasites that cause malaria in humans, other primates, birds, and rodents all encode multiple 6-cys proteins. Distinct 6-cys protein family members reside on the surface at each extracellular life cycle stage and those on the surface of liver infective and sexual stages have been shown to play important roles in hepatocyte growth and fertilization respectively. However, 6-cys proteins associated with the blood-stage forms of the parasite have no known function. Here we investigate the biochemical nature and function of two blood-stage 6-cys proteins in Plasmodium falciparum, the most pathogenic species to afflict humans. We show that native P12 and P41 form a stable heterodimer on the infective merozoite surface and are secreted following invasion, but could find no evidence that this complex mediates erythrocyte-receptor binding. That P12 and P41 do not appear to have a major role as adhesins to erythrocyte receptors was supported by the observation that antisera to these proteins did not substantially inhibit erythrocyte invasion. To investigate other functional roles for these proteins their genes were successfully disrupted in P. falciparum, however P12 and P41 knockout parasites grew at normal rates in vitro and displayed no other obvious phenotypic changes. It now appears likely that these blood-stage 6-cys proteins operate as a pair and play redundant roles either in erythrocyte invasion or in host-immune interactions.


Asunto(s)
Plasmodium falciparum/metabolismo , Proteínas Protozoarias/química , Proteínas Protozoarias/metabolismo , Anticuerpos Antiprotozoarios/inmunología , Medios de Cultivo Condicionados/metabolismo , Eritrocitos/parasitología , Escherichia coli/genética , Células HEK293 , Humanos , Merozoítos/metabolismo , Plasmodium falciparum/crecimiento & desarrollo , Plasmodium falciparum/inmunología , Plasmodium falciparum/fisiología , Multimerización de Proteína , Estructura Cuaternaria de Proteína , Transporte de Proteínas , Proteínas Protozoarias/genética , Proteínas Protozoarias/inmunología , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/metabolismo
13.
Microbiology (Reading) ; 155(Pt 6): 2029-2039, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19383677

RESUMEN

Burkholderia cenocepacia is highly resistant to antimicrobial peptides and we hypothesized that the conversion of UDP-glucose to UDP-glucuronic acid, a reaction catalysed by the enzyme UDP-glucose dehydrogenase (Ugd) would be important for this resistance. The genome of B. cenocepacia contains three predicted ugd genes: ugd(BCAL2946), ugd(BCAM0855) and ugd(BCAM2034), all of which were individually inactivated. Only inactivation of ugd(BCAL2946) resulted in increased sensitivity to polymyxin B and this sensitivity could be overcome when either ugd(BCAL2946) or ugd(BCAM0855) but not ugd(BCAM2034) was expressed from plasmids. The growth of a conditional ugd(BCAL2946) mutant, created in the Deltaugd(BCAM0855) background, was significantly impaired under non-permissive conditions. Growth could be rescued by either ugd(BCAL2946) or ugd(BCAM0855) expressed in trans, but not by ugd(BCAM2034). Biochemical analysis of the purified, recombinant forms of Ugd(BCAL2946) and Ugd(BCAM0855) revealed that they are soluble homodimers with similar in vitro Ugd activity and comparable kinetic constants for their substrates UDP-glucose and NAD(+). Purified Ugd(BCAM2034) showed no in vitro Ugd activity. Real-time PCR analysis showed that the expression of ugd(BCAL2946) was 5.4- and 135-fold greater than that of ugd(BCAM0855) and ugd(BCAM2034), respectively. Together, these data indicate that the combined activity of Ugd(BCAL2946) and Ugd(BCAM0855) is essential for the survival of B. cenocepacia but only the most highly expressed ugd gene, ugd(BCAL2946), is required for polymyxin B resistance.


Asunto(s)
Burkholderia cepacia/efectos de los fármacos , Burkholderia cepacia/enzimología , Farmacorresistencia Bacteriana , Polimixina B/farmacología , Uridina Difosfato Glucosa Deshidrogenasa/metabolismo , Secuencia de Aminoácidos , Antibacterianos/farmacología , Burkholderia cepacia/genética , Expresión Génica , Prueba de Complementación Genética , Datos de Secuencia Molecular , Mutagénesis Insercional , ARN Bacteriano/análisis , ARN Bacteriano/genética , Proteínas Recombinantes de Fusión/biosíntesis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Uridina Difosfato Glucosa Deshidrogenasa/genética
14.
Microbiology (Reading) ; 154(Pt 8): 2513-2521, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18667584

RESUMEN

The species that presently constitute the Burkholderia cepacia complex (Bcc) have multiple roles; they include soil and water saprophytes, bioremediators, and plant, animal and human pathogens. Since the first description of pathogenicity in the Bcc was based on sour skin rot of onion bulbs, this study returned to this plant host to investigate the onion-associated phenotype of the Bcc. Many Bcc isolates, which were previously considered to be non-mucoid, produced copious amounts of exopolysaccharide (EPS) when onion tissue was provided as the sole nutrient. EPS production was not species-specific, was observed in isolates from both clinical and environmental sources, and did not correlate with the ability to cause maceration of onion tissue. Chemical analysis suggested that the onion components responsible for EPS induction were primarily the carbohydrates sucrose, fructose and fructans. Additional sugars were investigated, and all alcohol sugars tested were able to induce EPS production, in particular mannitol and glucitol. To investigate the molecular basis for EPS biosynthesis, we focused on the highly conserved bce gene cluster thought to be involved in cepacian biosynthesis. We demonstrated induction of the bce gene cluster by mannitol, and found a clear correlation between the inability of representatives of the Burkholderia cenocepacia ET12 lineage to produce EPS and the presence of an 11 bp deletion within the bceB gene, which encodes a glycosyltransferase. Insertional inactivation of bceB in Burkholderia ambifaria AMMD results in loss of EPS production on sugar alcohol media. These novel and surprising insights into EPS biosynthesis highlight the metabolic potential of the Bcc and show that a potential virulence factor may not be detected by routine laboratory culture. Our results also highlight a potential hazard in the use of inhaled mannitol as an osmolyte to improve mucociliary clearance in individuals with cystic fibrosis.


Asunto(s)
Complejo Burkholderia cepacia/metabolismo , Metabolismo de los Hidratos de Carbono , Cebollas/metabolismo , Polisacáridos Bacterianos/biosíntesis , Alcoholes del Azúcar/metabolismo , Complejo Burkholderia cepacia/genética , Cebollas/química , Extractos Vegetales/química , Extractos Vegetales/metabolismo , Polisacáridos Bacterianos/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA