Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
Proc Natl Acad Sci U S A ; 121(20): e2310348121, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38709922

ABSTRACT

The evolutionary conserved YopJ family comprises numerous type-III-secretion system (T3SS) effectors of diverse mammalian and plant pathogens that acetylate host proteins to dampen immune responses. Acetylation is mediated by a central acetyltransferase domain that is flanked by conserved regulatory sequences, while a nonconserved N-terminal extension encodes the T3SS-specific translocation signal. Bartonella spp. are facultative-intracellular pathogens causing intraerythrocytic bacteremia in their mammalian reservoirs and diverse disease manifestations in incidentally infected humans. Bartonellae do not encode a T3SS, but most species possess a type-IV-secretion system (T4SS) to translocate Bartonella effector proteins (Beps) into host cells. Here we report that the YopJ homologs present in Bartonellae species represent genuine T4SS effectors. Like YopJ family T3SS effectors of mammalian pathogens, the "Bartonella YopJ-like effector A" (ByeA) of Bartonella taylorii also targets MAP kinase signaling to dampen proinflammatory responses, however, translocation depends on a functional T4SS. A split NanoLuc luciferase-based translocation assay identified sequences required for T4SS-dependent translocation in conserved regulatory regions at the C-terminus and proximal to the N-terminus of ByeA. The T3SS effectors YopP from Yersinia enterocolitica and AvrA from Salmonella Typhimurium were also translocated via the Bartonella T4SS, while ByeA was not translocated via the Yersinia T3SS. Our data suggest that YopJ family T3SS effectors may have evolved from an ancestral T4SS effector, such as ByeA of Bartonella. In this evolutionary scenario, the signal for T4SS-dependent translocation encoded by N- and C-terminal sequences remained functional in the derived T3SS effectors due to the essential role these sequences coincidentally play in regulating acetyltransferase activity.


Subject(s)
Bacterial Proteins , Bartonella , Type IV Secretion Systems , Bartonella/metabolism , Bartonella/genetics , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Humans , Type IV Secretion Systems/metabolism , Type IV Secretion Systems/genetics , Protein Transport , Animals
2.
PLoS Pathog ; 17(1): e1008548, 2021 01.
Article in English | MEDLINE | ID: mdl-33508040

ABSTRACT

Bartonellae are Gram-negative facultative-intracellular pathogens that use a type-IV-secretion system (T4SS) to translocate a cocktail of Bartonella effector proteins (Beps) into host cells to modulate diverse cellular functions. BepC was initially reported to act in concert with BepF in triggering major actin cytoskeletal rearrangements that result in the internalization of a large bacterial aggregate by the so-called 'invasome'. Later, infection studies with bepC deletion mutants and ectopic expression of BepC have implicated this effector in triggering an actin-dependent cell contractility phenotype characterized by fragmentation of migrating cells due to deficient rear detachment at the trailing edge, and BepE was shown to counterbalance this remarkable phenotype. However, the molecular mechanism of how BepC triggers cytoskeletal changes and the host factors involved remained elusive. Using infection assays, we show here that T4SS-mediated transfer of BepC is sufficient to trigger stress fiber formation in non-migrating epithelial cells and additionally cell fragmentation in migrating endothelial cells. Interactomic analysis revealed binding of BepC to a complex of the Rho guanine nucleotide exchange factor GEF-H1 and the serine/threonine-protein kinase MRCKα. Knock-out cell lines revealed that only GEF-H1 is required for mediating BepC-triggered stress fiber formation and inhibitor studies implicated activation of the RhoA/ROCK pathway downstream of GEF-H1. Ectopic co-expression of tagged versions of GEF-H1 and BepC truncations revealed that the C-terminal 'Bep intracellular delivery' (BID) domain facilitated anchorage of BepC to the plasma membrane, whereas the N-terminal 'filamentation induced by cAMP' (FIC) domain facilitated binding of GEF-H1. While FIC domains typically mediate post-translational modifications, most prominently AMPylation, a mutant with quadruple amino acid exchanges in the putative active site indicated that the BepC FIC domain acts in a non-catalytic manner to activate GEF-H1. Our data support a model in which BepC activates the RhoA/ROCK pathway by re-localization of GEF-H1 from microtubules to the plasma membrane.


Subject(s)
Actins/metabolism , Bacterial Proteins/metabolism , Bartonella/metabolism , Cell Membrane/metabolism , Protein C/metabolism , Rho Guanine Nucleotide Exchange Factors/metabolism , Stress Fibers/physiology , Bacterial Proteins/genetics , Cytoskeleton/metabolism , Endothelial Cells/cytology , Endothelial Cells/metabolism , HeLa Cells , Humans , Protein C/genetics , Rho Guanine Nucleotide Exchange Factors/genetics
3.
PLoS Pathog ; 17(1): e1009065, 2021 01.
Article in English | MEDLINE | ID: mdl-33508039

ABSTRACT

Bartonella T4SS effector BepC was reported to mediate internalization of big Bartonella aggregates into host cells by modulating F-actin polymerization. After that, BepC was indicated to induce host cell fragmentation, an interesting cell phenotype that is characterized by failure of rear-end retraction during cell migration, and subsequent dragging and fragmentation of cells. Here, we found that expression of BepC resulted in significant stress fiber formation and contractile cell morphology, which depended on combination of the N-terminus FIC (filamentation induced by c-AMP) domain and C-terminus BID (Bartonella intracellular delivery) domain of BepC. The FIC domain played a key role in BepC-induced stress fiber formation and cell fragmentation because deletion of FIC signature motif or mutation of two conserved amino acid residues abolished BepC-induced cell fragmentation. Immunoprecipitation confirmed the interaction of BepC with GEF-H1 (a microtubule-associated RhoA guanosine exchange factor), and siRNA-mediated depletion of GEF-H1 prevented BepC-induced stress fiber formation. Interaction with BepC caused the dissociation of GEF-H1 from microtubules and activation of RhoA to induce formation of stress fibers. The ROCK (Rho-associated protein kinase) inhibitor Y27632 completely blocked BepC effects on stress fiber formation and cell contractility. Moreover, stress fiber formation by BepC increased the stability of focal adhesions, which consequently impeded rear-edge detachment. Overall, our study revealed that BepC-induced stress fiber formation was achieved through the GEF-H1/RhoA/ROCK pathway.


Subject(s)
Actin Cytoskeleton/metabolism , Bartonella/metabolism , Cell Membrane/metabolism , Focal Adhesions/physiology , Rho Guanine Nucleotide Exchange Factors/metabolism , Stress Fibers/physiology , Type IV Secretion Systems/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cell Movement , Endothelial Cells/cytology , Endothelial Cells/metabolism , Humans , Microtubules/metabolism , Rho Guanine Nucleotide Exchange Factors/genetics , Type IV Secretion Systems/genetics
4.
Nat Chem ; 12(8): 732-739, 2020 08.
Article in English | MEDLINE | ID: mdl-32632184

ABSTRACT

Various pathogenic bacteria use post-translational modifications to manipulate the central components of host cell functions. Many of the enzymes released by these bacteria belong to the large Fic family, which modify targets with nucleotide monophosphates. The lack of a generic method for identifying the cellular targets of Fic family enzymes hinders investigation of their role and the effect of the post-translational modification. Here, we establish an approach that uses reactive co-substrate-linked enzymes for proteome profiling. We combine synthetic thiol-reactive nucleotide derivatives with recombinantly produced Fic enzymes containing strategically placed cysteines in their active sites to yield reactive binary probes for covalent substrate capture. The binary complexes capture their targets from cell lysates and permit subsequent identification. Furthermore, we determined the structures of low-affinity ternary enzyme-nucleotide-substrate complexes by applying a covalent-linking strategy. This approach thus allows target identification of the Fic enzymes from both bacteria and eukarya.


Subject(s)
Bacterial Proteins/metabolism , Membrane Proteins/metabolism , Nucleotidyltransferases/metabolism , Adenosine Monophosphate/metabolism , Amino Acid Sequence , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bartonella/metabolism , Biocatalysis , Crystallography, X-Ray , HeLa Cells , Humans , Membrane Proteins/chemistry , Nucleotidyltransferases/chemistry , Pasteurellaceae/metabolism , Protein Processing, Post-Translational , Protein Structure, Tertiary , Sequence Alignment , Substrate Specificity , cdc42 GTP-Binding Protein/genetics , cdc42 GTP-Binding Protein/metabolism
5.
Cell Microbiol ; 21(11): e13068, 2019 11.
Article in English | MEDLINE | ID: mdl-31231937

ABSTRACT

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.


Subject(s)
Bartonella/genetics , Bartonella/pathogenicity , Gene Transfer, Horizontal/genetics , Adaptation, Physiological/genetics , Animals , Bacterial Proteins/genetics , Bartonella/growth & development , Bartonella/metabolism , Evolution, Molecular , Gene Transfer, Horizontal/physiology , Host Microbial Interactions , Mutation , Recombination, Genetic/genetics , Replication Origin/genetics , Type IV Secretion Systems/genetics , Type IV Secretion Systems/metabolism
6.
Structure ; 25(1): 203-211, 2017 01 03.
Article in English | MEDLINE | ID: mdl-27889208

ABSTRACT

The BID (Bep intracellular delivery) domain functions as secretion signal in a subfamily of protein substrates of bacterial type IV secretion (T4S) systems. It mediates transfer of (1) relaxases and the attached DNA during bacterial conjugation, and (2) numerous Bartonella effector proteins (Beps) during protein transfer into host cells infected by pathogenic Bartonella species. Furthermore, BID domains of Beps have often evolved secondary effector functions within host cells. Here, we provide crystal structures for three representative BID domains and describe a novel conserved fold characterized by a compact, antiparallel four-helix bundle topped with a hook. The conserved hydrophobic core provides a rigid scaffold to a surface that, despite a few conserved exposed residues and similarities in charge distribution, displays significant variability. We propose that the genuine function of BID domains as T4S signal may primarily depend on their rigid structure, while the plasticity of their surface may facilitate adaptation to secondary effector functions.


Subject(s)
Bartonella/metabolism , Type VI Secretion Systems/chemistry , Bartonella/chemistry , Binding Sites , Conserved Sequence , Crystallography, X-Ray , Models, Molecular , Protein Domains , Protein Structure, Secondary
7.
Nat Rev Microbiol ; 14(6): 374-84, 2016 06.
Article in English | MEDLINE | ID: mdl-27140688

ABSTRACT

The gut microbiota can have profound effects on hosts, but the study of these relationships in humans is challenging. The specialized gut microbial community of honey bees is similar to the mammalian microbiota, as both are mostly composed of host-adapted, facultatively anaerobic and microaerophilic bacteria. However, the microbial community of the bee gut is far simpler than the mammalian microbiota, being dominated by only nine bacterial species clusters that are specific to bees and that are transmitted through social interactions between individuals. Recent developments, which include the discovery of extensive strain-level variation, evidence of protective and nutritional functions, and reports of eco-physiological or disease-associated perturbations to the microbial community, have drawn attention to the role of the microbiota in bee health and its potential as a model for studying the ecology and evolution of gut symbionts.


Subject(s)
Bacteria/isolation & purification , Bacterial Physiological Phenomena , Bees/microbiology , Gastrointestinal Microbiome , Animals , Bacteria/classification , Bacteria/genetics , Bacteria/metabolism , Bartonella/genetics , Bartonella/isolation & purification , Bartonella/metabolism , Bees/anatomy & histology , Bees/growth & development , Bees/physiology , Biological Evolution , Gastrointestinal Microbiome/physiology , Host Specificity , Humans , Lactobacillus/genetics , Lactobacillus/isolation & purification , Lactobacillus/metabolism , RNA, Ribosomal, 16S , Symbiosis
8.
mBio ; 6(6): e01867-15, 2015 Dec 08.
Article in English | MEDLINE | ID: mdl-26646013

ABSTRACT

UNLABELLED: Prokaryotes use type IV secretion systems (T4SSs) to translocate substrates (e.g., nucleoprotein, DNA, and protein) and/or elaborate surface structures (i.e., pili or adhesins). Bacterial genomes may encode multiple T4SSs, e.g., there are three functionally divergent T4SSs in some Bartonella species (vir, vbh, and trw). In a unique case, most rickettsial species encode a T4SS (rvh) enriched with gene duplication. Within single genomes, the evolutionary and functional implications of cross-system interchangeability of analogous T4SS protein components remains poorly understood. To lend insight into cross-system interchangeability, we analyzed the VirB8 family of T4SS channel proteins. Crystal structures of three VirB8 and two TrwG Bartonella proteins revealed highly conserved C-terminal periplasmic domain folds and dimerization interfaces, despite tremendous sequence divergence. This implies remarkable structural constraints for VirB8 components in the assembly of a functional T4SS. VirB8/TrwG heterodimers, determined via bacterial two-hybrid assays and molecular modeling, indicate that differential expression of trw and vir systems is the likely barrier to VirB8-TrwG interchangeability. We also determined the crystal structure of Rickettsia typhi RvhB8-II and modeled its coexpressed divergent paralog RvhB8-I. Remarkably, while RvhB8-I dimerizes and is structurally similar to other VirB8 proteins, the RvhB8-II dimer interface deviates substantially from other VirB8 structures, potentially preventing RvhB8-I/RvhB8-II heterodimerization. For the rvh T4SS, the evolution of divergent VirB8 paralogs implies a functional diversification that is unknown in other T4SSs. Collectively, our data identify two different constraints (spatiotemporal for Bartonella trw and vir T4SSs and structural for rvh T4SSs) that mediate the functionality of multiple divergent T4SSs within a single bacterium. IMPORTANCE: Assembly of multiprotein complexes at the right time and at the right cellular location is a fundamentally important task for any organism. In this respect, bacteria that express multiple analogous type IV secretion systems (T4SSs), each composed of around 12 different components, face an overwhelming complexity. Our work here presents the first structural investigation on factors regulating the maintenance of multiple T4SSs within a single bacterium. The structural data imply that the T4SS-expressing bacteria rely on two strategies to prevent cross-system interchangeability: (i) tight temporal regulation of expression or (ii) rapid diversification of the T4SS components. T4SSs are ideal drug targets provided that no analogous counterparts are known from eukaryotes. Drugs targeting the barriers to cross-system interchangeability (i.e., regulators) could dysregulate the structural and functional independence of discrete systems, potentially creating interference that prevents their efficient coordination throughout bacterial infection.


Subject(s)
Bartonella/chemistry , Bartonella/metabolism , Rickettsia typhi/chemistry , Rickettsia typhi/metabolism , Type IV Secretion Systems/chemistry , Type IV Secretion Systems/metabolism , Crystallography, X-Ray , Models, Molecular , Protein Binding , Protein Conformation , Protein Multimerization , Substrate Specificity , Two-Hybrid System Techniques
9.
Curr Opin Microbiol ; 23: 80-5, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25461577

ABSTRACT

The facultative intracellular bacteria Bartonella spp. share a common infection strategy to invade and colonize mammals in a host-specific manner. Following transmission by blood-sucking arthropods, Bartonella are inoculated in the derma and then spread, via two sequential enigmatic niches, to the blood stream where they cause a long-lasting intra-erythrocytic bacteraemia. The VirB/VirD4 type IV secretion system (VirB/D4 T4SS) is essential for the pathogenicity of most Bartonella species by injecting an arsenal of effector proteins into host cells. These bacterial effector proteins share a modular architecture, comprising domains and/or motifs that confer an array of functions. Here, we review recent advances in understanding the function and evolutionary origin of this fascinating repertoire of host-targeted bacterial effectors.


Subject(s)
Bacterial Proteins/metabolism , Bartonella/metabolism , Cell Physiological Phenomena/drug effects , Virulence Factors/metabolism , Animals , Arthropods , Bacterial Secretion Systems , Bartonella/growth & development , Blood/microbiology , Host-Pathogen Interactions , Humans , Mammals , Protein Transport
10.
Appl Environ Microbiol ; 79(16): 5082-4, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23747696

ABSTRACT

Blood samples from Apodemus agrarius from Poland yielded PCR amplicons of Bartonella species. These included B. grahamii, B. taylorii, and B. birtlesii, as is typical of European Apodemus, as well as B. elizabethae-like forms and a recombinant strain of B. taylorii, most closely related to an American isolate from Tamiasciurus hudsonicus.


Subject(s)
Bacterial Proteins/genetics , Bartonella Infections/veterinary , Bartonella/genetics , Murinae , Rodent Diseases/epidemiology , Animals , Bacterial Proteins/metabolism , Bartonella/classification , Bartonella/isolation & purification , Bartonella/metabolism , Bartonella Infections/epidemiology , Bartonella Infections/microbiology , Molecular Sequence Data , Phylogeny , Phylogeography , Poland/epidemiology , Polymerase Chain Reaction/veterinary , Rodent Diseases/microbiology , Sequence Analysis, DNA/veterinary , Sequence Homology
11.
mBio ; 4(2): e00115-13, 2013 Apr 23.
Article in English | MEDLINE | ID: mdl-23611908

ABSTRACT

UNLABELLED: Specialization of bacteria in a new niche is associated with genome repertoire changes, and speciation in bacterial specialists is associated with genome reduction. Here, we tested a signature-tagged mutant library of 3,456 Bartonella birtlesii clones to detect mutants that could grow rapidly in vitro. Overall, we found 124 mutants that grew faster than the parental wild-type strain in vitro. We sequenced the genomes of the four mutants with the most rapid growth (formed visible colonies in only 1 to 2 days compared with 5 days for the wild type) and compared them to the parental isolate genome. We found that the number of disrupted genes associated with translation in the 124 rapid-growth clones was significantly higher than the number of genes involved in translation in the full genome (P < 10(-6)). Analysis of transposon integration in the genome of the four most rapidly growing clones revealed that one clone lacked one of the two wild-type RNA ribosomal operons. Finally, one of the four clones did not induce bacteremia in our mouse model, whereas infection with the other three resulted in a significantly lower bacterial count in blood than that with the wild-type strain. IMPORTANCE: Here, we show that specialization in a specific niche could be caused by the disruption of critical genes. Most of these genes were involved in translation, and we show that evolution of obligate parasitism bacteria was specifically associated with disruption of translation system-encoding genes.


Subject(s)
Bartonella/growth & development , Bartonella/metabolism , Gene Expression Regulation, Bacterial , Protein Biosynthesis , Protein Processing, Post-Translational , Animals , Bartonella/genetics , DNA Transposable Elements , Female , Genes, Bacterial , Genome, Bacterial , Mice , Mice, Inbred BALB C , Mutagenesis, Insertional , Sequence Analysis, DNA
12.
Plasmid ; 70(1): 146-53, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23583564

ABSTRACT

The stability of components of multiprotein complexes often relies on the presence of the functional complex. To assess structural dependence among the components of the R388 Type IV secretion system (T4SS), the steady-state level of several Trw proteins was determined in the absence of other Trw components. While several Trw proteins were affected by the lack of others, we found that the coupling protein TrwB is not affected by the absence of other T4SS components, nor did its absence alter significantly the levels of integral components of the complex, underscoring the independent role of the coupling protein on the T4SS architecture. The cytoplasmic ATPases TrwK (VirB4) and TrwD (VirB11) were affected by the absence of several core complex components, while the pilus component TrwJ (VirB5) required the presence of all other Trw proteins (except for TrwB) to be detectable. Overall, the results delineate a possible assembly pathway for the T4SS of R388. We have also tested structural complementation of TrwD (VirB11) and TrwJ (VirB5) by their homologues in the highly related Trw system of Bartonella tribocorum (Bt). The results reveal a correlation with the functional complementation data previously reported.


Subject(s)
Adenosine Triphosphatases/genetics , Bacterial Proteins/genetics , Conjugation, Genetic , DNA, Bacterial/genetics , Escherichia coli/genetics , Fimbriae, Bacterial/genetics , Plasmids/genetics , Adenosine Triphosphatases/metabolism , Bacterial Proteins/metabolism , Bartonella/genetics , Bartonella/metabolism , DNA Replication , DNA, Bacterial/metabolism , Escherichia coli/metabolism , Fimbriae, Bacterial/metabolism , Genetic Complementation Test , Operon , Plasmids/metabolism
13.
PLoS One ; 7(7): e41447, 2012.
Article in English | MEDLINE | ID: mdl-22848496

ABSTRACT

Each Bartonella species appears to be highly adapted to one or a limited number of reservoir hosts, in which it establishes long-lasting intraerythrocytic bacteremia as the hallmark of infection. Recently, we identified Trw as the bacterial system involved in recognition of erythrocytes according to their animal origin. The T4SS Trw is characterized by a multiprotein complex that spans the inner and outer bacterial membranes, and possesses a hypothetical pilus structure. TrwJ, I, H and trwL are present in variable copy numbers in different species and the multiple copies of trwL and trwJ in the Bartonella trw locus are considered to encode variant forms of surface-exposed pilus components. We therefore aimed to identify which of the candidate Trw pilus components were located on the bacterial surface and involved in adhesion to erythrocytes, together with their erythrocytic receptor. Using different technologies (electron microscopy, phage display, invasion inhibition assay, far western blot), we found that only TrwJ1 and TrwJ2 were expressed and localized at the cell surface of B. birtlesii and had the ability to bind to mouse erythrocytes, and that their receptor was band3, one of the major outer-membrane glycoproteins of erythrocytes, (anion exchanger). According to these results, we propose that the interaction between TrwJ1, TrwJ2 and band 3 leads to the critical host-specific adherence of Bartonella to its host cells, erythrocytes.


Subject(s)
Anion Exchange Protein 1, Erythrocyte/metabolism , Bacterial Proteins/metabolism , Bartonella Infections/metabolism , Bartonella/metabolism , Erythrocytes/metabolism , Receptors, Cell Surface/metabolism , Animals , Anion Exchange Protein 1, Erythrocyte/genetics , Bacterial Proteins/genetics , Bartonella/genetics , Bartonella/pathogenicity , Bartonella/ultrastructure , Bartonella Infections/genetics , Erythrocytes/microbiology , Erythrocytes/ultrastructure , Female , Fimbriae, Bacterial/genetics , Fimbriae, Bacterial/metabolism , Fimbriae, Bacterial/ultrastructure , Male , Mice , Mice, Inbred BALB C , Rabbits , Receptors, Cell Surface/genetics
14.
Proc Natl Acad Sci U S A ; 109(24): 9581-6, 2012 Jun 12.
Article in English | MEDLINE | ID: mdl-22635269

ABSTRACT

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.


Subject(s)
Adenylyl Cyclases/metabolism , Bartonella/metabolism , Cyclic AMP/biosynthesis , GTP-Binding Protein alpha Subunits, Gs/metabolism , Catalytic Domain
15.
PLoS Genet ; 7(2): e1001296, 2011 Feb 10.
Article in English | MEDLINE | ID: mdl-21347280

ABSTRACT

Adaptive radiation is the rapid origination of multiple species from a single ancestor as the result of concurrent adaptation to disparate environments. This fundamental evolutionary process is considered to be responsible for the genesis of a great portion of the diversity of life. Bacteria have evolved enormous biological diversity by exploiting an exceptional range of environments, yet diversification of bacteria via adaptive radiation has been documented in a few cases only and the underlying molecular mechanisms are largely unknown. Here we show a compelling example of adaptive radiation in pathogenic bacteria and reveal their genetic basis. Our evolutionary genomic analyses of the α-proteobacterial genus Bartonella uncover two parallel adaptive radiations within these host-restricted mammalian pathogens. We identify a horizontally-acquired protein secretion system, which has evolved to target specific bacterial effector proteins into host cells as the evolutionary key innovation triggering these parallel adaptive radiations. We show that the functional versatility and adaptive potential of the VirB type IV secretion system (T4SS), and thereby translocated Bartonella effector proteins (Beps), evolved in parallel in the two lineages prior to their radiations. Independent chromosomal fixation of the virB operon and consecutive rounds of lineage-specific bep gene duplications followed by their functional diversification characterize these parallel evolutionary trajectories. Whereas most Beps maintained their ancestral domain constitution, strikingly, a novel type of effector protein emerged convergently in both lineages. This resulted in similar arrays of host cell-targeted effector proteins in the two lineages of Bartonella as the basis of their independent radiation. The parallel molecular evolution of the VirB/Bep system displays a striking example of a key innovation involved in independent adaptive processes and the emergence of bacterial pathogens. Furthermore, our study highlights the remarkable evolvability of T4SSs and their effector proteins, explaining their broad application in bacterial interactions with the environment.


Subject(s)
Bacterial Secretion Systems/genetics , Bartonella/genetics , Bartonella/metabolism , Biological Evolution , Genetic Speciation , Adaptation, Biological/genetics , Animals , Bacterial Proteins/genetics , Bartonella/classification , Computational Biology , HEK293 Cells , Host-Pathogen Interactions , Humans , Molecular Sequence Annotation , Phylogeny , Rats , Selection, Genetic , Sequence Analysis, DNA
16.
Rev. esp. quimioter ; 23(3): 109-114, sept. 2010. tab
Article in Spanish | IBECS | ID: ibc-81847

ABSTRACT

Las infecciones por Bartonella spp. incluyen un amplio espectro de enfermedades infecciosas emergentes y reemergentes. En este tipo de infecciones no existe una pauta de tratamiento universalizado, por ello, se debe ajustar a cada situación clínica. El objetivo de esta revisión es actualizar los aspectos terapéuticos de las diferentes manifestaciones clínicas provocadas por las bartonellas(AU)


Infections by Bartonella spp. include a wide spectrum of emerging and re-emerging infectious diseases. There is not a universal therapy for this infection, therefore treatment should be chosen individually. The aim of this review is to update the therapeutics aspects of this kind of infections(AU)


Subject(s)
Humans , Male , Female , Bartonella/pathogenicity , Bartonella Infections/diagnosis , Bartonella Infections/drug therapy , Endocarditis/drug therapy , Endocarditis, Bacterial/drug therapy , Cat-Scratch Disease/drug therapy , Angiomatosis, Bacillary/drug therapy , Peliosis Hepatis/drug therapy , Bacteremia/drug therapy , Gentamicins/therapeutic use , Bartonella , Bartonella/isolation & purification , Bartonella/metabolism , Bartonella Infections/epidemiology , Bartonella Infections/physiopathology , Angiomatosis, Bacillary/epidemiology , Erythromycin/therapeutic use
17.
PLoS Pathog ; 6(6): e1000946, 2010 Jun 10.
Article in English | MEDLINE | ID: mdl-20548954

ABSTRACT

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.


Subject(s)
Bacteremia/microbiology , Bacterial Proteins/metabolism , Bartonella Infections/microbiology , Bartonella/metabolism , Cell Adhesion , Erythrocytes/microbiology , Virulence Factors/metabolism , Animals , Cats , Erythrocytes/metabolism , Erythrocytes/pathology , Female , Humans , Mice , Mice, Inbred BALB C , Rats
18.
J Bacteriol ; 192(11): 2655-69, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20363945

ABSTRACT

The conjugative coupling protein TrwB is responsible for connecting the relaxosome to the type IV secretion system during conjugative DNA transfer of plasmid R388. It is directly involved in transport of the relaxase TrwC, and it displays an ATPase activity probably involved in DNA pumping. We designed a conjugation assay in which the frequency of DNA transfer is directly proportional to the amount of TrwB. A collection of point mutants was constructed in the TrwB cytoplasmic domain on the basis of the crystal structure of TrwB Delta N70, targeting the nucleotide triphosphate (NTP)-binding region, the cytoplasmic surface, or the internal channel in the hexamer. An additional set of transfer-deficient mutants was obtained by random mutagenesis. Most mutants were impaired in both DNA and protein transport. We found that the integrity of the nucleotide binding domain is absolutely required for TrwB function, which is also involved in monomer-monomer interactions. Polar residues surrounding the entrance and inside the internal channel were important for TrwB function and may be involved in interactions with the relaxosomal components. Finally, the N-terminal transmembrane domain of TrwB was subjected to random mutagenesis followed by a two-hybrid screen for mutants showing enhanced protein-protein interactions with the related TrwE protein of Bartonella tribocorum. Several point mutants were obtained with mutations in the transmembranal helices: specifically, one proline from each protein may be the key residue involved in the interaction of the coupling protein with the type IV secretion apparatus.


Subject(s)
DNA-Binding Proteins/metabolism , Escherichia coli Proteins/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bartonella/genetics , Bartonella/metabolism , Blotting, Western , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Models, Biological , Mutagenesis, Site-Directed , Point Mutation , Protein Binding/genetics , Protein Binding/physiology , Protein Structure, Secondary , Protein Structure, Tertiary , Two-Hybrid System Techniques
19.
PLoS One ; 5(3): e9765, 2010 Mar 19.
Article in English | MEDLINE | ID: mdl-20333257

ABSTRACT

Bartonella quintana is a re-emerging pathogen and the causative agent of a variety of disease manifestations in humans including trench fever. Various typing methods have been developed for B. quintana, but these tend to be limited by poor resolution and, in the case of gel-based methods, a lack of portability. Multilocus sequence typing (MLST) has been used to study the molecular epidemiology of a large number of pathogens, including B. henselae, a close relative of B. quintana. We developed a MLST scheme for B. quintana based on the 7 MLST loci employed for B. henselae with two additional loci to cover underrepresented regions of the B. quintana chromosome. A total of 16 B. quintana isolates spanning over 60 years and three continents were characterized. Allelic variation was detected in five of the nine loci. Although only 8/4270 (0.002%) of the nucleotide sites examined were variable over all loci, these polymorphisms resolved the 16 isolates into seven sequence types (STs). We also demonstrate that MLST can be applied on uncultured isolates by direct PCR from cardiac valve tissue, and suggest this method presents a promising approach for epidemiological studies in this highly clonal organism. Phylogenetic and clustering analyses suggest that two of the seven STs form a distinct lineage within the population.


Subject(s)
Bacterial Typing Techniques/methods , Bartonella/genetics , Bartonella/metabolism , Heart Valves/microbiology , Multilocus Sequence Typing , Alleles , Genetic Variation , Heart Valves/pathology , Phylogeny , Polymerase Chain Reaction , Polymorphism, Genetic , Sequence Analysis, DNA , Software
20.
PLoS Genet ; 5(7): e1000546, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19578403

ABSTRACT

The genus Bartonella comprises facultative intracellular bacteria adapted to mammals, including previously recognized and emerging human pathogens. We report the 2,341,328 bp genome sequence of Bartonella grahamii, one of the most prevalent Bartonella species in wild rodents. Comparative genomics revealed that rodent-associated Bartonella species have higher copy numbers of genes for putative host-adaptability factors than the related human-specific pathogens. Many of these gene clusters are located in a highly dynamic region of 461 kb. Using hybridization to a microarray designed for the B. grahamii genome, we observed a massive, putatively phage-derived run-off replication of this region. We also identified a novel gene transfer agent, which packages the bacterial genome, with an over-representation of the amplified DNA, in 14 kb pieces. This is the first observation associating the products of run-off replication with a gene transfer agent. Because of the high concentration of gene clusters for host-adaptation proteins in the amplified region, and since the genes encoding the gene transfer agent and the phage origin are well conserved in Bartonella, we hypothesize that these systems are driven by selection. We propose that the coupling of run-off replication with gene transfer agents promotes diversification and rapid spread of host-adaptability factors, facilitating host shifts in Bartonella.


Subject(s)
Bacteriophages/physiology , Bartonella Infections/microbiology , Bartonella/virology , Disease Reservoirs/microbiology , Gene Transfer, Horizontal , Genome, Bacterial , Mice/microbiology , Virus Replication , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacteriophages/genetics , Bartonella/classification , Bartonella/genetics , Bartonella/metabolism , Host-Pathogen Interactions , Humans , Molecular Sequence Data , Phylogeny
SELECTION OF CITATIONS
SEARCH DETAIL
...