Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
PLoS Negl Trop Dis ; 16(7): e0010635, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35881641

RESUMEN

BACKGROUND: Human brucellosis caused by the facultative intracellular pathogen Brucella spp. is an endemic bacterial zoonosis manifesting as acute or chronic infections with high morbidity. Treatment typically involves a combination therapy of two antibiotics for several weeks to months, but despite this harsh treatment relapses occur at a rate of 5-15%. Although poor compliance and reinfection may account for a fraction of the observed relapse cases, it is apparent that the properties of the infectious agent itself may play a decisive role in this phenomenon. METHODOLOGY/PRINCIPAL FINDINGS: We used B. abortus carrying a dual reporter in a macrophage infection model to gain a better understanding of the efficacy of recommended therapies in cellulo. For this we used automated fluorescent microscopy as a prime read-out and developed specific CellProfiler pipelines to score infected macrophages at the population and the single cell level. Combining microscopy of constitutive and induced reporters with classical CFU determination, we quantified the protective nature of the Brucella intracellular lifestyle to various antibiotics and the ability of B. abortus to persist in cellulo despite harsh antibiotic treatments. CONCLUSION/SIGNIFICANCE: We demonstrate that treatment of infected macrophages with antibiotics at recommended concentrations fails to fully prevent growth and persistence of B. abortus in cellulo, which may be explained by a protective nature of the intracellular niche(s). Moreover, we show the presence of bona fide intracellular persisters upon antibiotic treatment, which are metabolically active and retain the full infectious potential, therefore constituting a plausible reservoir for reinfection and relapse. In conclusion, our results highlight the need to extend the spectrum of models to test new antimicrobial therapies for brucellosis to better reflect the in vivo infection environment, and to develop therapeutic approaches targeting the persister subpopulation.


Asunto(s)
Brucella abortus , Brucelosis , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Brucelosis/tratamiento farmacológico , Brucelosis/microbiología , Humanos , Macrófagos/microbiología , Reinfección
2.
Cell Microbiol ; 21(11): e13068, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31231937

RESUMEN

The processes underlying host adaptation by bacterial pathogens remain a fundamental question with relevant clinical, ecological, and evolutionary implications. Zoonotic pathogens of the genus Bartonella constitute an exceptional model to study these aspects. Bartonellae have undergone a spectacular diversification into multiple species resulting from adaptive radiation. Specific adaptations of a complex facultative intracellular lifestyle have enabled the colonisation of distinct mammalian reservoir hosts. This remarkable host adaptability has a multifactorial basis and is thought to be driven by horizontal gene transfer (HGT) and recombination among a limited genus-specific pan genome. Recent functional and evolutionary studies revealed that the conserved Bartonella gene transfer agent (BaGTA) mediates highly efficient HGT and could thus drive this evolution. Here, we review the recent progress made towards understanding BaGTA evolution, function, and its role in the evolution and pathogenesis of Bartonella spp. We notably discuss how BaGTA could have contributed to genome diversification through recombination of beneficial traits that underlie host adaptability. We further address how BaGTA may counter the accumulation of deleterious mutations in clonal populations (Muller's ratchet), which are expected to occur through the recurrent transmission bottlenecks during the complex infection cycle of these pathogens in their mammalian reservoir hosts and arthropod vectors.


Asunto(s)
Bartonella/genética , Bartonella/patogenicidad , Transferencia de Gen Horizontal/genética , Adaptación Fisiológica/genética , Animales , Proteínas Bacterianas/genética , Bartonella/crecimiento & desarrollo , Bartonella/metabolismo , Evolución Molecular , Transferencia de Gen Horizontal/fisiología , Interacciones Microbiota-Huesped , Mutación , Recombinación Genética/genética , Origen de Réplica/genética , Sistemas de Secreción Tipo IV/genética , Sistemas de Secreción Tipo IV/metabolismo
3.
mSphere ; 4(3)2019 06 26.
Artículo en Inglés | MEDLINE | ID: mdl-31243080

RESUMEN

Brucella, the agent causing brucellosis, is a major zoonotic pathogen with worldwide distribution. Brucella resides and replicates inside infected host cells in membrane-bound compartments called Brucella-containing vacuoles (BCVs). Following uptake, Brucella resides in endosomal BCVs (eBCVs) that gradually mature from early to late endosomal features. Through a poorly understood process that is key to the intracellular lifestyle of Brucella, the eBCV escapes fusion with lysosomes by transitioning to the replicative BCV (rBCV), a replicative niche directly connected to the endoplasmic reticulum (ER). Despite the notion that this complex intracellular lifestyle must depend on a multitude of host factors, a holistic view on which of these components control Brucella cell entry, trafficking, and replication is still missing. Here we used a systematic cell-based small interfering RNA (siRNA) knockdown screen in HeLa cells infected with Brucella abortus and identified 425 components of the human infectome for Brucella infection. These include multiple components of pathways involved in central processes such as the cell cycle, actin cytoskeleton dynamics, or vesicular trafficking. Using assays for pathogen entry, knockdown complementation, and colocalization at single-cell resolution, we identified the requirement of the VPS retromer for Brucella to escape the lysosomal degradative pathway and to establish its intracellular replicative niche. We thus validated the VPS retromer as a novel host factor critical for Brucella intracellular trafficking. Further, our genomewide data shed light on the interplay between central host processes and the biogenesis of the Brucella replicative niche.IMPORTANCE With >300,000 new cases of human brucellosis annually, Brucella is regarded as one of the most important zoonotic bacterial pathogens worldwide. The agent causing brucellosis resides inside host cells within vacuoles termed Brucella-containing vacuoles (BCVs). Although a few host components required to escape the degradative lysosomal pathway and to establish the ER-derived replicative BCV (rBCV) have already been identified, the global understanding of this highly coordinated process is still partial, and many factors remain unknown. To gain deeper insight into these fundamental questions, we performed a genomewide RNA interference (RNAi) screen aiming at discovering novel host factors involved in the Brucella intracellular cycle. We identified 425 host proteins that contribute to Brucella cellular entry, intracellular trafficking, and replication. Together, this study sheds light on previously unknown host pathways required for the Brucella infection cycle and highlights the VPS retromer components as critical factors for the establishment of the Brucella intracellular replicative niche.


Asunto(s)
Brucella abortus/genética , Complejos de Clasificación Endosomal Requeridos para el Transporte/genética , Interacciones Huésped-Patógeno , ARN Interferente Pequeño , Vacuolas/microbiología , Brucella abortus/fisiología , Replicación del ADN , Retículo Endoplásmico/microbiología , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Técnicas de Silenciamiento del Gen , Genoma Bacteriano , Células HeLa , Ensayos Analíticos de Alto Rendimiento , Humanos
4.
Genome Res ; 27(12): 2083-2095, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-29141959

RESUMEN

Accurate annotation of all protein-coding sequences (CDSs) is an essential prerequisite to fully exploit the rapidly growing repertoire of completely sequenced prokaryotic genomes. However, large discrepancies among the number of CDSs annotated by different resources, missed functional short open reading frames (sORFs), and overprediction of spurious ORFs represent serious limitations. Our strategy toward accurate and complete genome annotation consolidates CDSs from multiple reference annotation resources, ab initio gene prediction algorithms and in silico ORFs (a modified six-frame translation considering alternative start codons) in an integrated proteogenomics database (iPtgxDB) that covers the entire protein-coding potential of a prokaryotic genome. By extending the PeptideClassifier concept of unambiguous peptides for prokaryotes, close to 95% of the identifiable peptides imply one distinct protein, largely simplifying downstream analysis. Searching a comprehensive Bartonella henselae proteomics data set against such an iPtgxDB allowed us to unambiguously identify novel ORFs uniquely predicted by each resource, including lipoproteins, differentially expressed and membrane-localized proteins, novel start sites and wrongly annotated pseudogenes. Most novelties were confirmed by targeted, parallel reaction monitoring mass spectrometry, including unique ORFs and single amino acid variations (SAAVs) identified in a re-sequenced laboratory strain that are not present in its reference genome. We demonstrate the general applicability of our strategy for genomes with varying GC content and distinct taxonomic origin. We release iPtgxDBs for B. henselae, Bradyrhizobium diazoefficiens and Escherichia coli and the software to generate both proteogenomics search databases and integrated annotation files that can be viewed in a genome browser for any prokaryote.


Asunto(s)
Proteínas Bacterianas/genética , Bartonella henselae/genética , Bradyrhizobium/genética , Escherichia coli/genética , Genoma Bacteriano , Proteogenómica , Bases de Datos de Proteínas , Anotación de Secuencia Molecular , Sistemas de Lectura Abierta , Programas Informáticos
5.
PLoS Genet ; 13(10): e1007077, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29073136

RESUMEN

Host-targeting type IV secretion systems (T4SS) evolved from conjugative T4SS machineries that mediate interbacterial plasmid transfer. However, the origins of effectors secreted by these virulence devices have remained largely elusive. Previous work showed that some effectors exhibit homology to toxins of bacterial toxin-antitoxin modules, but the evolutionary trajectories underlying these ties had not been resolved. We previously reported that FicT toxins of FicTA toxin-antitoxin modules disrupt cellular DNA topology via their enzymatic FIC (filamentation induced by cAMP) domain. Intriguingly, the FIC domain of the FicT toxin VbhT of Bartonella schoenbuchensis is fused to a type IV secretion signal-the BID (Bep intracellular delivery) domain-similar to the Bartonella effector proteins (Beps) that are secreted into eukaryotic host cells via the host-targeting VirB T4SS. In this study, we show that the VbhT toxin is an interbacterial effector protein secreted via the conjugative Vbh T4SS that is closely related to the VirB T4SS and encoded by plasmid pVbh of B. schoenbuchensis. We therefore propose that the Vbh T4SS together with its effector VbhT represent an evolutionary missing link on a path that leads from a regular conjugation system and FicTA toxin-antitoxin modules to the VirB T4SS and the Beps. Intriguingly, phylogenetic analyses revealed that the fusion of FIC and BID domains has probably occurred independently in VbhT and the common ancestor of the Beps, suggesting parallel evolutionary paths. Moreover, several other examples of TA module toxins that are bona fide substrates of conjugative T4SS indicate that their recruitment as interbacterial effectors is prevalent and serves yet unknown biological functions in the context of bacterial conjugation. We propose that the adaptation for interbacterial transfer favors the exaptation of FicT and other TA module toxins as inter-kingdom effectors and may thus constitute an important stepping stone in the evolution of host-targeted effector proteins.


Asunto(s)
Antitoxinas/metabolismo , Sistemas de Secreción Bacterianos/genética , Sistemas de Secreción Bacterianos/metabolismo , Toxinas Bacterianas/metabolismo , Bartonella/genética , Bartonella/patogenicidad , Secuencia de Aminoácidos , Antitoxinas/genética , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Toxinas Bacterianas/genética , Infecciones por Bartonella/microbiología , Conjugación Genética , Regulación Bacteriana de la Expresión Génica , Interacciones Huésped-Patógeno , Plásmidos , Homología de Secuencia , Virulencia
6.
Curr Opin Microbiol ; 39: 34-41, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28858701

RESUMEN

Bacterial pathogen-host cell interactions involve an intricate interplay of multiple components from both partners. Systems level surveys have been used widely to profile host requirements for pathogen infection. Functional genomics, and more specifically genome-wide perturbation screens, constitute attractive methodologies to assess such host infectomes. Although these strategies have successfully identified numerous critical host factors, they may have failed in generating the high-quality data required for systems level analysis. This is the case for most RNA interference (RNAi) setups with their high propensity to off-target effects (OTE). However, recent efforts to circumvent OTE in RNAi-based experiments as well as the emergence of alternative strategies will likely soon allow significant breakthrough in the systems level understanding of bacterial pathogen-host interactions.


Asunto(s)
Bacterias , Sistemas CRISPR-Cas , Interacciones Huésped-Patógeno , Interferencia de ARN , Biología de Sistemas/métodos , Animales , Bacterias/genética , Bacterias/patogenicidad , Infecciones Bacterianas , Humanos
7.
Cell Syst ; 4(6): 611-621.e6, 2017 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-28624614

RESUMEN

The Bartonella gene transfer agent (BaGTA) is an archetypical example for domestication of a phage-derived element to permit high-frequency genetic exchange in bacterial populations. Here we used multiplexed transposon sequencing (TnSeq) and single-cell reporters to globally define the core components and transfer dynamics of BaGTA. Our systems-level analysis has identified inner- and outer-circle components of the BaGTA system, including 55 regulatory components, as well as an additional 74 and 107 components mediating donor transfer and recipient uptake functions. We show that the stringent response signal guanosine-tetraphosphate (ppGpp) restricts BaGTA induction to a subset of fast-growing cells, whereas BaGTA particle uptake depends on a functional Tol-Pal trans-envelope complex that mediates outer-membrane invagination upon cell division. Our findings suggest that Bartonella evolved an efficient strategy to promote genetic exchange within the fittest subpopulation while disfavoring exchange of deleterious genetic information, thereby facilitating genome integrity and rapid host adaptation.


Asunto(s)
Bartonella/genética , Transferencia de Gen Horizontal/genética , Proteínas Bacterianas/genética , Bacteriófagos/genética , División Celular/genética , Genética , Guanosina Tetrafosfato/genética
8.
Genome Biol Evol ; 9(3): 761-776, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-28338931

RESUMEN

The α-proteobacterial genus Bartonella comprises a group of ubiquitous mammalian pathogens that are studied as a model for the evolution of bacterial pathogenesis. Vast abundance of two particular phylogenetic lineages of Bartonella had been linked to enhanced host adaptability enabled by lineage-specific acquisition of a VirB/D4 type IV secretion system (T4SS) and parallel evolution of complex effector repertoires. However, the limited availability of genome sequences from one of those lineages as well as other, remote branches of Bartonella has so far hampered comprehensive understanding of how the VirB/D4 T4SS and its effectors called Beps have shaped Bartonella evolution. Here, we report the discovery of a third repertoire of Beps associated with the VirB/D4 T4SS of B. ancashensis, a novel human pathogen that lacks any signs of host adaptability and is only distantly related to the two species-rich lineages encoding a VirB/D4 T4SS. Furthermore, sequencing of ten new Bartonella isolates from under-sampled lineages enabled combined in silico analyses and wet lab experiments that suggest several parallel layers of functional diversification during evolution of the three Bep repertoires from a single ancestral effector. Our analyses show that the Beps of B. ancashensis share many features with the two other repertoires, but may represent a more ancestral state that has not yet unleashed the adaptive potential of such an effector set. We anticipate that the effectors of B. ancashensis will enable future studies to dissect the evolutionary history of Bartonella effectors and help unraveling the evolutionary forces underlying bacterial host adaptation.


Asunto(s)
Sistemas de Secreción Bacterianos/genética , Infecciones por Bartonella/genética , Bartonella/genética , Sistemas de Secreción Tipo IV/genética , Proteínas Bacterianas/genética , Bartonella/patogenicidad , Infecciones por Bartonella/microbiología , Infecciones por Bartonella/patología , Evolución Molecular , Interacciones Huésped-Patógeno/genética , Humanos , Filogenia , Factores de Virulencia/genética
9.
J Proteomics ; 99: 123-37, 2014 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-24486812

RESUMEN

Proteomics data provide unique insights into biological systems, including the predominant subcellular localization (SCL) of proteins, which can reveal important clues about their functions. Here we analyzed data of a complete prokaryotic proteome expressed under two conditions mimicking interaction of the emerging pathogen Bartonella henselae with its mammalian host. Normalized spectral count data from cytoplasmic, total membrane, inner and outer membrane fractions allowed us to identify the predominant SCL for 82% of the identified proteins. The spectral count proportion of total membrane versus cytoplasmic fractions indicated the propensity of cytoplasmic proteins to co-fractionate with the inner membrane, and enabled us to distinguish cytoplasmic, peripheral inner membrane and bona fide inner membrane proteins. Principal component analysis and k-nearest neighbor classification training on selected marker proteins or predominantly localized proteins, allowed us to determine an extensive catalog of at least 74 expressed outer membrane proteins, and to extend the SCL assignment to 94% of the identified proteins, including 18% where in silico methods gave no prediction. Suitable experimental proteomics data combined with straightforward computational approaches can thus identify the predominant SCL on a proteome-wide scale. Finally, we present a conceptual approach to identify proteins potentially changing their SCL in a condition-dependent fashion. BIOLOGICAL SIGNIFICANCE: The work presented here describes the first prokaryotic proteome-wide subcellular localization (SCL) dataset for the emerging pathogen B. henselae (Bhen). The study indicates that suitable subcellular fractionation experiments combined with straight-forward computational analysis approaches assessing the proportion of spectral counts observed in different subcellular fractions are powerful for determining the predominant SCL of a large percentage of the experimentally observed proteins. This includes numerous cases where in silico prediction methods do not provide any prediction. Avoiding a treatment with harsh conditions, cytoplasmic proteins tend to co-fractionate with proteins of the inner membrane fraction, indicative of close functional interactions. The spectral count proportion (SCP) of total membrane versus cytoplasmic fractions allowed us to obtain a good indication about the relative proximity of individual protein complex members to the inner membrane. Using principal component analysis and k-nearest neighbor approaches, we were able to extend the percentage of proteins with a predominant experimental localization to over 90% of all expressed proteins and identified a set of at least 74 outer membrane (OM) proteins. In general, OM proteins represent a rich source of candidates for the development of urgently needed new therapeutics in combat of resurgence of infectious disease and multi-drug resistant bacteria. Finally, by comparing the data from two infection biology relevant conditions, we conceptually explore methods to identify and visualize potential candidates that may partially change their SCL in these different conditions. The data are made available to researchers as a SCL compendium for Bhen and as an assistance in further improving in silico SCL prediction algorithms.


Asunto(s)
Proteínas Bacterianas/metabolismo , Bartonella henselae/metabolismo , Modelos Biológicos , Proteoma/metabolismo , Proteómica/métodos
10.
Mol Microbiol ; 90(4): 756-75, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24033511

RESUMEN

The co-ordinated expression of virulence factors is a critical process for any bacterial pathogen to colonize its host. Here we investigated the mechanisms of niche adaptation of the zoonotic pathogen Bartonella henselae by combining genetic approaches and shotgun proteomics. We demonstrated that expression of the VirB/D4 type IV secretion system (T4SS) and its secreted effector proteins require the alternative sigma factor RpoH1, which levels are controlled by the stringent response (SR) components DksA and SpoT. The RpoH1-dependent activation requires an active BatR/BatS two-component system (TCS) while BatR expression is controlled by RpoH1 and the SR components. Deletion of spoT results in a strong attenuation of VirB/D4 T4SS expression whereas dksA, rpoH1 or batR deletion fully abolishes its activity. In contrast to their activating effect on the VirB/D4 T4SS, which is critical at the early stage of host infection, SpoT and DksA negatively regulate the Trw T4SS, which mediates host-specific erythrocyte infection at a later stage of the colonization process. Our findings support a model where the SR signalling and the physiological pH-induced BatR/BatS TCS conjointly control the spatiotemporal expression of B. henselae adaptation factors during host infection.


Asunto(s)
Proteínas Bacterianas/metabolismo , Sistemas de Secreción Bacterianos , Bartonella henselae/metabolismo , Factor sigma/metabolismo , Factores de Virulencia/metabolismo , Proteínas Bacterianas/genética , Bartonella henselae/genética , Bartonella henselae/patogenicidad , Regulación Bacteriana de la Expresión Génica , Interacciones Huésped-Patógeno , Mutagénesis , Proteómica , Factor sigma/genética , Transducción de Señal , Factores de Virulencia/genética
11.
Genome Res ; 23(11): 1916-27, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23878158

RESUMEN

Prokaryotes, due to their moderate complexity, are particularly amenable to the comprehensive identification of the protein repertoire expressed under different conditions. We applied a generic strategy to identify a complete expressed prokaryotic proteome, which is based on the analysis of RNA and proteins extracted from matched samples. Saturated transcriptome profiling by RNA-seq provided an endpoint estimate of the protein-coding genes expressed under two conditions which mimic the interaction of Bartonella henselae with its mammalian host. Directed shotgun proteomics experiments were carried out on four subcellular fractions. By specifically targeting proteins which are short, basic, low abundant, and membrane localized, we could eliminate their initial underrepresentation compared to the estimated endpoint. A total of 1250 proteins were identified with an estimated false discovery rate below 1%. This represents 85% of all distinct annotated proteins and ∼90% of the expressed protein-coding genes. Genes that were detected at the transcript but not protein level, were found to be highly enriched in several genomic islands. Furthermore, genes that lacked an ortholog and a functional annotation were not detected at the protein level; these may represent examples of overprediction in genome annotations. A dramatic membrane proteome reorganization was observed, including differential regulation of autotransporters, adhesins, and hemin binding proteins. Particularly noteworthy was the complete membrane proteome coverage, which included expression of all members of the VirB/D4 type IV secretion system, a key virulence factor.


Asunto(s)
Bartonella henselae/genética , Secuencia de Bases , Proteoma/genética , Proteómica/métodos , Factores de Virulencia/genética , Animales , Proteínas Bacterianas/genética , Perfilación de la Expresión Génica , Regulación Bacteriana de la Expresión Génica , Genoma Bacteriano , Modelos Biológicos , Anotación de Secuencia Molecular , Proteoma/metabolismo , Factores de Virulencia/metabolismo
12.
Proc Natl Acad Sci U S A ; 109(24): 9581-6, 2012 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-22635269

RESUMEN

Subversion of host organism cAMP signaling is an efficient and widespread mechanism of microbial pathogenesis. Bartonella effector protein A (BepA) of vasculotumorigenic Bartonella henselae protects the infected human endothelial cells against apoptotic stimuli by elevation of cellular cAMP levels by an as yet unknown mechanism. Here, adenylyl cyclase (AC) and the α-subunit of the AC-stimulating G protein (Gαs) were identified as potential cellular target proteins for BepA by gel-free proteomics. Results of the proteomics screen were evaluated for physical and functional interaction by: (i) a heterologous in vivo coexpression system, where human AC activity was reconstituted under the regulation of Gαs and BepA in Escherichia coli; (ii) in vitro AC assays with membrane-anchored full-length human AC and recombinant BepA and Gαs; (iii) surface plasmon resonance experiments; and (iv) an in vivo fluorescence bimolecular complementation-analysis. The data demonstrate that BepA directly binds host cell AC to potentiate the Gαs-dependent cAMP production. As opposed to the known microbial mechanisms, such as ADP ribosylation of G protein α-subunits by cholera and pertussis toxins, the fundamentally different BepA-mediated elevation of host cell cAMP concentration appears subtle and is dependent on the stimulus of a G protein-coupled receptor-released Gαs. We propose that this mechanism contributes to the persistence of Bartonella henselae in the chronically infected vascular endothelium.


Asunto(s)
Adenilil Ciclasas/metabolismo , Bartonella/metabolismo , AMP Cíclico/biosíntesis , Subunidades alfa de la Proteína de Unión al GTP Gs/metabolismo , Dominio Catalítico
13.
Proc Natl Acad Sci U S A ; 108(35): 14643-8, 2011 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-21844337

RESUMEN

Bacterial type IV secretion systems (T4SS) mediate interbacterial conjugative DNA transfer and transkingdom protein transfer into eukaryotic host cells in bacterial pathogenesis. The sole bacterium known to naturally transfer DNA into eukaryotic host cells via a T4SS is the plant pathogen Agrobacterium tumefaciens. Here we demonstrate T4SS-mediated DNA transfer from a human bacterial pathogen into human cells. We show that the zoonotic pathogen Bartonella henselae can transfer a cryptic plasmid occurring in the bartonellae into the human endothelial cell line EA.hy926 via its T4SS VirB/VirD4. DNA transfer into EA.hy926 cells was demonstrated by using a reporter derivative of this Bartonella-specific mobilizable plasmid generated by insertion of a eukaryotic egfp-expression cassette. Fusion of the C-terminal secretion signal of the endogenous VirB/VirD4 protein substrate BepD with the plasmid-encoded DNA-transport protein Mob resulted in a 100-fold increased DNA transfer rate. Expression of the delivered egfp gene in EA.hy926 cells required cell division, suggesting that nuclear envelope breakdown may facilitate passive entry of the transferred ssDNA into the nucleus as prerequisite for complementary strand synthesis and transcription of the egfp gene. Addition of an eukaryotic neomycin phosphotransferase expression cassette to the reporter plasmid facilitated selection of stable transgenic EA.hy926 cell lines that display chromosomal integration of the transferred plasmid DNA. Our data suggest that T4SS-dependent DNA transfer into host cells may occur naturally during human infection with Bartonella and that these chronically infecting pathogens have potential for the engineering of in vivo gene-delivery vectors with applications in DNA vaccination and therapeutic gene therapy.


Asunto(s)
Proteínas Bacterianas/fisiología , Bartonella henselae/genética , Conjugación Genética , Transferencia de Gen Horizontal , Secuencia de Bases , División Celular , Proteínas Fluorescentes Verdes/genética , Humanos , Datos de Secuencia Molecular , Plásmidos
14.
Cell Microbiol ; 13(3): 419-31, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21044238

RESUMEN

The vasculotropic pathogen Bartonella henselae (Bh) intimately interacts with human endothelial cells (ECs) and subverts multiple cellular functions. Here we report that Bh specifically interferes with vascular endothelial growth factor (VEGF) signalling in ECs. Bh infection abrogated VEGF-induced proliferation and wound closure of EC monolayers as well as the capillary-like sprouting of EC spheroids. On the molecular level, Bh infection did not alter VEGF receptor 2 (VEGFR2) expression or cell surface localization, but impeded VEGF-stimulated phosphorylation of VEGFR2 at tyrosine(1175) . Consistently, we observed that Bh infection diminished downstream events of the tyrosine(1175) -dependent VEGFR2-signalling pathway leading to EC proliferation, i.e. phospholipase-Cγ activation, cytosolic calcium fluxes and mitogen-activated protein kinase ERK1/2 phosphorylation. Pervanadate treatment neutralized the inhibitory activity of Bh on VEGF signalling, suggesting that Bh infection may activate a phosphatase that alleviates VEGFR2 phosphorylation. Inhibition of VEGFR2 signalling by Bh infection was strictly dependent on a functional VirB type IV secretion system and thereby translocated Bep effector proteins. The data presented in this study underscore the role of the VirB/Bep system as important factor controlling EC proliferation in response to Bh infection; not only as previously reported by counter-acting an intrinsic bacterial mitogenic stimulus, but also by restricting the exogenous angiogenic stimulation by Bh-induced VEGF.


Asunto(s)
Bartonella henselae/patogenicidad , Células Endoteliales/metabolismo , Células Endoteliales/microbiología , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/metabolismo , Proteínas Bacterianas/metabolismo , Bartonella henselae/efectos de los fármacos , Bartonella henselae/inmunología , Bartonella henselae/metabolismo , Western Blotting , Calcio , Electroforesis en Gel de Poliacrilamida , Células Endoteliales/citología , Humanos , Immunoblotting , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Fosfolipasa C gamma/metabolismo , Fosforilación , Reacción en Cadena de la Polimerasa , Vanadatos/farmacología , Factor A de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
15.
PLoS Pathog ; 6(6): e1000946, 2010 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-20548954

RESUMEN

Bacterial pathogens typically infect only a limited range of hosts; however, the genetic mechanisms governing host-specificity are poorly understood. The alpha-proteobacterial genus Bartonella comprises 21 species that cause host-specific intraerythrocytic bacteremia as hallmark of infection in their respective mammalian reservoirs, including the human-specific pathogens Bartonella quintana and Bartonella bacilliformis that cause trench fever and Oroya fever, respectively. Here, we have identified bacterial factors that mediate host-specific erythrocyte colonization in the mammalian reservoirs. Using mouse-specific Bartonella birtlesii, human-specific Bartonella quintana, cat-specific Bartonella henselae and rat-specific Bartonella tribocorum, we established in vitro adhesion and invasion assays with isolated erythrocytes that fully reproduce the host-specificity of erythrocyte infection as observed in vivo. By signature-tagged mutagenesis of B. birtlesii and mutant selection in a mouse infection model we identified mutants impaired in establishing intraerythrocytic bacteremia. Among 45 abacteremic mutants, five failed to adhere to and invade mouse erythrocytes in vitro. The corresponding genes encode components of the type IV secretion system (T4SS) Trw, demonstrating that this virulence factor laterally acquired by the Bartonella lineage is directly involved in adherence to erythrocytes. Strikingly, ectopic expression of Trw of rat-specific B. tribocorum in cat-specific B. henselae or human-specific B. quintana expanded their host range for erythrocyte infection to rat, demonstrating that Trw mediates host-specific erythrocyte infection. A molecular evolutionary analysis of the trw locus further indicated that the variable, surface-located TrwL and TrwJ might represent the T4SS components that determine host-specificity of erythrocyte parasitism. In conclusion, we show that the laterally acquired Trw T4SS diversified in the Bartonella lineage to facilitate host-restricted adhesion to erythrocytes in a wide range of mammals.


Asunto(s)
Bacteriemia/microbiología , Proteínas Bacterianas/metabolismo , Infecciones por Bartonella/microbiología , Bartonella/metabolismo , Adhesión Celular , Eritrocitos/microbiología , Factores de Virulencia/metabolismo , Animales , Gatos , Eritrocitos/metabolismo , Eritrocitos/patología , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Ratas
16.
J Bacteriol ; 192(13): 3352-67, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20418395

RESUMEN

Here, we report the first comprehensive study of Bartonella henselae gene expression during infection of human endothelial cells. Expression of the main cluster of upregulated genes, comprising the VirB type IV secretion system and its secreted protein substrates, is shown to be under the positive control of the transcriptional regulator BatR. We demonstrate binding of BatR to the promoters of the virB operon and a substrate-encoding gene and provide biochemical evidence that BatR and BatS constitute a functional two-component regulatory system. Moreover, in contrast to the acid-inducible (pH 5.5) homologs ChvG/ChvI of Agrobacterium tumefaciens, BatR/BatS are optimally activated at the physiological pH of blood (pH 7.4). By conservation analysis of the BatR regulon, we show that BatR/BatS are uniquely adapted to upregulate a genus-specific virulence regulon during hemotropic infection in mammals. Thus, we propose that BatR/BatS two-component system homologs represent vertically inherited pH sensors that control the expression of horizontally transmitted gene sets critical for the diverse host-associated life styles of the alphaproteobacteria.


Asunto(s)
Proteínas Bacterianas/metabolismo , Bartonella henselae/metabolismo , Proteínas Bacterianas/genética , Línea Celular , Línea Celular Tumoral , Electroforesis en Gel de Poliacrilamida , Ensayo de Cambio de Movilidad Electroforética , Citometría de Flujo , Regulación Bacteriana de la Expresión Génica/fisiología , Prueba de Complementación Genética , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Concentración de Iones de Hidrógeno , Immunoblotting , Operón/genética , Filogenia , Regiones Promotoras Genéticas/genética , Regiones Promotoras Genéticas/fisiología , Unión Proteica/genética , Unión Proteica/fisiología , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
17.
Thromb Haemost ; 94(2): 347-61, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16113825

RESUMEN

The bacterial pathogen Bartonella henselae (Bh) is responsible for a broad range of clinical manifestations, including the formation of vascular tumours as the result of pathogen-triggered vasoproliferation. In vitro, the interaction of Bh with human umbilical vein endothelial cells (Huvec) involves (i) cytoskeletal rearrangements in conjunction with bacterial internalization, (ii) nuclear factor kappaB (NFkappaB)-dependent proinflammatory activation, (iii) the inhibition of apoptosis, and (iv) the modulation of angiogenic properties such as proliferation, migration, and tubular differentiation. To study the transcriptional signature of these pathogen-triggered changes of Huvec, we performed transcriptional profiling with Affymetrix U133 GeneChips. At 6 h or 30 h of infection, a total of 706 genes displayed a clear and statistically significant change of expression (>2.5-fold, t-test p-value<0.05). These included 314 up-regulated genes dominated by the innate immune response. The gene list comprises subsets of tumour necrosis factor alpha (TNFalpha, 99 genes) and interferon alpha (IFNalpha, 30 genes) inducible genes, which encode components of the NF-kappaB-dependent proinflammatory response and the type I IFN-dependent anti-infective response, respectively. The remaining set of 197 up-regulated genes mirrors other cellular changes induced by Bh, in particular proliferation and proangiogenic activation. The set of 362 down-regulated genes includes 41TNFalpha - or IFNalpha-suppressible genes, and 52 genes involved in cell cycle control or progression. This comprehensive analysis of Bh-triggered changes of the Huvec transcriptome identified candidate genes putatively involved in controlling innate immune responses, cell cycle, and vascular remodelling, and may thus provide the basis for functional studies of the molecular mechanisms underlying these pathogen-induced cellular processes.


Asunto(s)
Bartonella henselae/metabolismo , Endotelio Vascular/citología , Endotelio Vascular/microbiología , Regulación de la Expresión Génica , Sistema Inmunológico , Transcripción Genética , Ciclo Celular , Proliferación Celular , Análisis por Conglomerados , Cartilla de ADN/química , Regulación hacia Abajo , Endotelio Vascular/patología , Humanos , Interferón-alfa/metabolismo , Familia de Multigenes , Mutación , FN-kappa B/metabolismo , Neovascularización Patológica , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Estadística como Asunto , Factores de Tiempo , Factor de Necrosis Tumoral alfa/metabolismo , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA