Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Nat Struct Mol Biol ; 29(12): 1170-1177, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36456825

RESUMEN

Polysaccharides play critical roles in bacteria, including the formation of protective capsules and biofilms and establishing specific host cell interactions. Their transport across membranes is often mediated by ATP-binding cassette (ABC) transporters, which utilize ATP to translocate diverse molecules. Cyclic ß-glucans (CßGs) are critical for host interaction of the Rhizobiales, including the zoonotic pathogen Brucella. CßGs are exported into the periplasmic space by the cyclic glucan transporter (Cgt). The interaction of an ABC transporter with a polysaccharide substrate has not been visualized so far. Here we use single-particle cryoelectron microscopy to elucidate the structures of Cgt from Brucella abortus in four conformational states. The substrate-bound structure reveals an unusual binding pocket at the height of the cytoplasmic leaflet, whereas ADP-vanadate models hint at an alternative mechanism of substrate release. Our work provides insights into the translocation of large, heterogeneous substrates and sheds light on protein-polysaccharide interactions in general.


Asunto(s)
Transportadoras de Casetes de Unión a ATP , Brucella abortus , beta-Glucanos , Adenosina Trifosfato/metabolismo , Transportadoras de Casetes de Unión a ATP/metabolismo , beta-Glucanos/metabolismo , Brucella abortus/metabolismo , Microscopía por Crioelectrón , Glucanos/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Polisacáridos
2.
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
3.
Cell Host Microbe ; 27(3): 476-485.e7, 2020 03 11.
Artículo en Inglés | MEDLINE | ID: mdl-32101706

RESUMEN

Chronically infecting pathogens avoid clearance by the innate immune system by promoting premature transition from an initial pro-inflammatory response toward an anti-inflammatory tissue-repair response. STAT3, a central regulator of inflammation, controls this transition and thus is targeted by numerous chronic pathogens. Here, we show that BepD, an effector of the chronic bacterial pathogen Bartonella henselae targeted to infected host cells, establishes an exceptional pathway for canonical STAT3 activation, thereby impairing secretion of pro-inflammatory TNF-α and stimulating secretion of anti-inflammatory IL-10. Tyrosine phosphorylation of EPIYA-related motifs in BepD facilitates STAT3 binding and activation via c-Abl-dependent phosphorylation of Y705. The tyrosine-phosphorylated scaffold of BepD thus represents a signaling hub for intrinsic STAT3 activation that is independent from canonical STAT3 activation via transmembrane receptor-associated Janus kinases. We anticipate that our findings on a molecular shortcut to STAT3 activation will inspire new treatment options for chronic infections and inflammatory diseases.


Asunto(s)
Proteínas Bacterianas/metabolismo , Bartonella henselae/inmunología , Interleucina-10/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Secuencia de Aminoácidos , Animales , Línea Celular , Citocinas/inmunología , Femenino , Quinasas Janus/metabolismo , Ratones , Ratones Endogámicos C57BL , Fosforilación , Células RAW 264.7 , Factor de Necrosis Tumoral alfa/metabolismo
4.
Sci Rep ; 8(1): 5173, 2018 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-29581535

RESUMEN

Mitochondria are complex organelles that participate in many cellular functions, ranging from ATP production to immune responses against viruses and bacteria. This integration of a plethora of functions within a single organelle makes mitochondria a very attractive target to manipulate for intracellular pathogens. We characterised the crosstalk that exists between Brucella abortus, the causative agent of brucellosis, and the mitochondria of infected cells. Brucella replicates in a compartment derived from the endoplasmic reticulum (ER) and modulates ER functionality by activating the unfolded protein response. However, the impact of Brucella on the mitochondrial population of infected cells still requires a systematic study. We observed physical contacts between Brucella containing vacuoles and mitochondria. We also found that B. abortus replication is independent of mitochondrial oxidative phosphorylation and that mitochondrial reactive oxygen species do not participate to the control of B. abortus infection in vitro. We demonstrated that B. abortus and B. melitensis induce a drastic mitochondrial fragmentation at 48 hours post-infection in different cell types, including myeloid and non-myeloid cells. This fragmentation is DRP1-independent and might be caused by a deficit of mitochondrial fusion. However, mitochondrial fragmentation does not change neither Brucella replication efficiency, nor the susceptibility of infected cells to TNFα-induced apoptosis.


Asunto(s)
Brucella abortus/genética , Brucelosis/genética , Dinaminas/genética , Factor de Necrosis Tumoral alfa/genética , Animales , Apoptosis/genética , Brucella abortus/patogenicidad , Brucelosis/microbiología , Brucelosis/patología , Retículo Endoplásmico/genética , Retículo Endoplásmico/microbiología , Humanos , Macrófagos/metabolismo , Macrófagos/patología , Ratones , Mitocondrias/genética , Mitocondrias/microbiología , Células RAW 264.7 , Especies Reactivas de Oxígeno/metabolismo , Respuesta de Proteína Desplegada/genética , Vacuolas/genética
5.
Appl Environ Microbiol ; 83(12)2017 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-28411218

RESUMEN

Bacterial conjugation is a mechanism of horizontal DNA transfer. The relaxase TrwC of the conjugative plasmid R388 cleaves one strand of the transferred DNA at the oriT gene, covalently attaches to it, and leads the single-stranded DNA (ssDNA) into the recipient cell. In addition, TrwC catalyzes site-specific integration of the transferred DNA into its target sequence present in the genome of the recipient bacterium. Here, we report the analysis of the efficiency and specificity of the integrase activity of TrwC in human cells, using the type IV secretion system of the human pathogen Bartonella henselae to introduce relaxase-DNA complexes. Compared to Mob relaxase from plasmid pBGR1, we found that TrwC mediated a 10-fold increase in the rate of plasmid DNA transfer to human cells and a 100-fold increase in the rate of chromosomal integration of the transferred DNA. We used linear amplification-mediated PCR and plasmid rescue to characterize the integration pattern in the human genome. DNA sequence analysis revealed mostly reconstituted oriT sequences, indicating that TrwC is active and recircularizes transferred DNA in human cells. One TrwC-mediated site-specific integration event was detected, proving that TrwC is capable of mediating site-specific integration in the human genome, albeit with very low efficiency compared to the rate of random integration. Our results suggest that TrwC may stabilize the plasmid DNA molecules in the nucleus of the human cell, probably by recircularization of the transferred DNA strand. This stabilization would increase the opportunities for integration of the DNA by the host machinery.IMPORTANCE Different biotechnological applications, including gene therapy strategies, require permanent modification of target cells. Long-term expression is achieved either by extrachromosomal persistence or by integration of the introduced DNA. Here, we studied the utility of conjugative relaxase TrwC, a bacterial protein with site-specific integrase activity in bacteria, as an integrase in human cells. Although it is not efficient as a site-specific integrase, we found that TrwC is active in human cells and promotes random integration of the transferred DNA in the human genome, probably acting as a DNA chaperone until it is integrated by host mechanisms. TrwC-DNA complexes can be delivered to human cells through a type IV secretion system involved in pathogenesis. Thus, TrwC could be used in vivo to transfer the DNA of interest into the appropriate cell and promote its integration. If used in combination with a site-specific nuclease, it could lead to site-specific integration of the incoming DNA by homologous recombination.


Asunto(s)
Proteínas Bacterianas/metabolismo , Bartonella henselae/enzimología , Genoma Humano , Integrasas/metabolismo , Angiomatosis Bacilar/genética , Angiomatosis Bacilar/microbiología , Proteínas Bacterianas/genética , Bartonella henselae/genética , Línea Celular , Conjugación Genética , ADN Bacteriano/genética , ADN Bacteriano/metabolismo , Humanos , Integrasas/genética , Plásmidos/genética , Plásmidos/metabolismo
6.
PLoS One ; 11(9): e0163654, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27657533

RESUMEN

FIC domain proteins mediate post-translational modifications of target proteins, which typically results in their inactivation. Depending on the conservation of crucial active site residues, the FIC fold serves as structural scaffold for various enzymatic activities, mostly target adenylylation. The founding member of the vast Fic protein family, EcFicT, was identified in Escherichia coli some time ago. The G55R point mutant of EcFicT displays the "filamentation induced by cAMP" (Fic) phenotype at high 3',5'-cyclic adenosine monophosphate (cAMP) concentrations and elevated temperature, but the underlying molecular mechanism and any putative biochemical activity of EcFicT have remained unknown. EcFicT belongs to class I Fic toxin proteins that are encoded together with a small inhibitory protein (antitoxin), named EcFicA in E. coli. Here, we report the crystal structures of two mutant EcFicT/EcFicA complexes (EcFicTG55RA and EcFicTAE28G) both showing close resemblance with the structure of the AMP-transferase VbhT from Bartonella schoenbuchensis in complex with its cognate antitoxin VbhA. However, crucial differences in the active site of EcFicT compared to VbhT and other AMP-transferases rationalize the lack of evidence for adenylylation activity. Comprehensive bioinformatic analysis suggests that EcFicT has evolved from canonical AMP-transferases and has acquired a conserved binding site for a yet to be discovered novel substrate. The G55R mutation has no effect on structure or thermal stability of EcFicT, such that the molecular basis for its associated Fic phenotype remains elusive. We anticipate that this structure will inspire further bioinformatic and experimental analyses in order to characterize the enzymatic activity of EcFicT and help revealing its physiological role.

7.
Proc Natl Acad Sci U S A ; 113(5): E529-37, 2016 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-26787847

RESUMEN

Filamentation induced by cyclic AMP (FIC)-domain enzymes catalyze adenylylation or other posttranslational modifications of target proteins to control their function. Recently, we have shown that Fic enzymes are autoinhibited by an α-helix (αinh) that partly obstructs the active site. For the single-domain class III Fic proteins, the αinh is located at the C terminus and its deletion relieves autoinhibition. However, it has remained unclear how activation occurs naturally. Here, we show by structural, biophysical, and enzymatic analyses combined with in vivo data that the class III Fic protein NmFic from Neisseria meningitidis gets autoadenylylated in cis, thereby autonomously relieving autoinhibition and thus allowing subsequent adenylylation of its target, the DNA gyrase subunit GyrB. Furthermore, we show that NmFic activation is antagonized by tetramerization. The combination of autoadenylylation and tetramerization results in nonmonotonic concentration dependence of NmFic activity and a pronounced lag phase in the progress of target adenylylation. Bioinformatic analyses indicate that this elaborate dual-control mechanism is conserved throughout class III Fic proteins.


Asunto(s)
Proteínas Bacterianas/metabolismo , Biopolímeros/metabolismo , AMP Cíclico/metabolismo , Neisseria meningitidis/enzimología , Nucleotidiltransferasas/metabolismo , Girasa de ADN/metabolismo , Modelos Moleculares
8.
mBio ; 6(3): e00598-15, 2015 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-25991686

RESUMEN

UNLABELLED: Listeria monocytogenes enters nonphagocytic cells by a receptor-mediated mechanism that is dependent on a clathrin-based molecular machinery and actin rearrangements. Bacterial intra- and intercellular movements are also actin dependent and rely on the actin nucleating Arp2/3 complex, which is activated by host-derived nucleation-promoting factors downstream of the cell receptor Met during entry and by the bacterial nucleation-promoting factor ActA during comet tail formation. By genome-wide small interfering RNA (siRNA) screening for host factors involved in bacterial infection, we identified diverse cellular signaling networks and protein complexes that support or limit these processes. In addition, we could precise previously described molecular pathways involved in Listeria invasion. In particular our results show that the requirements for actin nucleators during Listeria entry and actin comet tail formation are different. Knockdown of several actin nucleators, including SPIRE2, reduced bacterial invasion while not affecting the generation of comet tails. Most interestingly, we observed that in contrast to our expectations, not all of the seven subunits of the Arp2/3 complex are required for Listeria entry into cells or actin tail formation and that the subunit requirements for each of these processes differ, highlighting a previously unsuspected versatility in Arp2/3 complex composition and function. IMPORTANCE: Listeria is a bacterial pathogen that induces its internalization within the cytoplasm of human cells and has been used for decades as a major molecular tool to manipulate cells in order to explore and discover cellular functions. We have inactivated individually, for the first time in epithelial cells, all the genes of the human genome to investigate whether each gene modifies positively or negatively the Listeria infectious process. We identified novel signaling cascades that have never been associated with Listeria infection. We have also revisited the role of the molecular complex Arp2/3 involved in the polymerization of the actin cytoskeleton, which was shown previously to be required for Listeria entry and movement inside host cells, and we demonstrate that contrary to the general dogma, some subunits of the complex are dispensable for both Listeria entry and bacterial movement.


Asunto(s)
Actinas/metabolismo , Endocitosis , Interacciones Huésped-Patógeno , Listeria monocytogenes/fisiología , Transducción de Señal , Células Epiteliales/microbiología , Silenciador del Gen , Pruebas Genéticas , Células HeLa , Humanos , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo
9.
PLoS One ; 9(9): e107289, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25202966

RESUMEN

Type II DNA topoisomerases are essential enzymes that catalyze topological rearrangement of double-stranded DNA using the free energy generated by ATP hydrolysis. Bacterial DNA gyrase is a prototype of this family and is composed of two subunits (GyrA, GyrB) that form a GyrA2GyrB2 heterotetramer. The N-terminal 43-kDa fragment of GyrB (GyrB43) from E. coli comprising the ATPase and the transducer domains has been studied extensively. The dimeric fragment is competent for ATP hydrolysis and its structure in complex with the substrate analog AMPPNP is known. Here, we have determined the remaining conformational states of the enzyme along the ATP hydrolysis reaction path by solving crystal structures of GyrB43 in complex with ADP⋅BeF3, ADP⋅Pi, and ADP. Upon hydrolysis, the enzyme undergoes an obligatory 12° domain rearrangement to accommodate the 1.5 Å increase in distance between the γ- and ß-phosphate of the nucleotide within the sealed binding site at the domain interface. Conserved residues from the QTK loop of the transducer domain (also part of the domain interface) couple the small structural change within the binding site with the rigid body motion. The domain reorientation is reflected in a significant 7 Å increase in the separation of the two transducer domains of the dimer that would embrace one of the DNA segments in full-length gyrase. The observed conformational change is likely to be relevant for the allosteric coordination of ATP hydrolysis with DNA binding, cleavage/re-ligation and/or strand passage.


Asunto(s)
Adenosina Difosfato/análogos & derivados , Adenosina Trifosfatasas/metabolismo , Girasa de ADN/metabolismo , Proteínas de Escherichia coli/metabolismo , Escherichia coli/metabolismo , Adenosina Difosfato/metabolismo , Adenosina Trifosfato/metabolismo , Sitios de Unión/genética , ADN Superhelicoidal/genética , Escherichia coli/genética , Hidrólisis , Movimiento (Física) , Estructura Terciaria de Proteína
10.
PLoS Pathog ; 10(6): e1004187, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24945914

RESUMEN

Numerous bacterial pathogens secrete multiple effectors to modulate host cellular functions. These effectors may interfere with each other to efficiently control the infection process. Bartonellae are Gram-negative, facultative intracellular bacteria using a VirB type IV secretion system to translocate a cocktail of Bartonella effector proteins (Beps) into host cells. Based on in vitro infection models we demonstrate here that BepE protects infected migratory cells from injurious effects triggered by BepC and is required for in vivo dissemination of bacteria from the dermal site of inoculation to blood. Human endothelial cells (HUVECs) infected with a ΔbepE mutant of B. henselae (Bhe) displayed a cell fragmentation phenotype resulting from Bep-dependent disturbance of rear edge detachment during migration. A ΔbepCE mutant did not show cell fragmentation, indicating that BepC is critical for triggering this deleterious phenotype. Complementation of ΔbepE with BepEBhe or its homologues from other Bartonella species abolished cell fragmentation. This cyto-protective activity is confined to the C-terminal Bartonella intracellular delivery (BID) domain of BepEBhe (BID2.EBhe). Ectopic expression of BID2.EBhe impeded the disruption of actin stress fibers by Rho Inhibitor 1, indicating that BepE restores normal cell migration via the RhoA signaling pathway, a major regulator of rear edge retraction. An intradermal (i.d.) model for B. tribocorum (Btr) infection in the rat reservoir host mimicking the natural route of infection by blood sucking arthropods allowed demonstrating a vital role for BepE in bacterial dissemination from derma to blood. While the Btr mutant ΔbepDE was abacteremic following i.d. inoculation, complementation with BepEBtr, BepEBhe or BIDs.EBhe restored bacteremia. Given that we observed a similar protective effect of BepEBhe on infected bone marrow-derived dendritic cells migrating through a monolayer of lymphatic endothelial cells we propose that infected dermal dendritic cells may be involved in disseminating Bartonella towards the blood stream in a BepE-dependent manner.


Asunto(s)
Sistemas de Secreción Bacterianos , Bartonella/patogenicidad , Citoprotección , Células Dendríticas/microbiología , Células Endoteliales de la Vena Umbilical Humana/microbiología , Citoesqueleto de Actina/metabolismo , Animales , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Bartonella/inmunología , Infecciones por Bartonella/inmunología , Infecciones por Bartonella/patología , Células de la Médula Ósea/citología , Células de la Médula Ósea/inmunología , Movimiento Celular , Células Cultivadas , Células Dendríticas/citología , Células Dendríticas/inmunología , Femenino , Interacciones Huésped-Patógeno , Células Endoteliales de la Vena Umbilical Humana/citología , Humanos , Ratones , Ratones Endogámicos BALB C , Estructura Terciaria de Proteína , Transporte de Proteínas , Ratas , Ratas Wistar , Transducción de Señal , Factores de Virulencia/genética , Factores de Virulencia/metabolismo , Proteína de Unión al GTP rhoA/metabolismo
11.
Proteomics ; 14(9): 1048-52, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24677795

RESUMEN

AMPylation is a posttranslational modification (PTM) that has recently caught much attention in the context of bacterial infections as pathogens were shown to secrete Fic proteins that AMPylate Rho GTPases and thus interfere with host cell signaling processes. Although Fic proteins are widespread and found in all kingdoms of life, only a small number of AMPylation targets are known to date. A major obstacle to target identification is the limited availability of generic strategies allowing sensitive and robust identification of AMPylation events. Here, we present an unbiased MS-based approach utilizing stable isotope-labeled ATP. The ATP isotopes are transferred onto target proteins in crude cell lysates by in vitro AMPylation introducing specific reporter ion clusters that allow detection of AMPylated peptides in complex biological samples by MS analysis. Applying this strategy on the secreted Fic protein Bep2 of Bartonella rochalimae, we identified the filamenting protein vimentin as an AMPylation target that was confirmed by independent assays. Vimentin represents a new class of target proteins and its identification emphasizes our method as a valuable tool to systematically uncover AMPylation targets. Furthermore, the approach can be generically adapted to study targets of other PTMs that allow incorporation of isotopically labeled substrates.


Asunto(s)
Adenosina Monofosfato/análisis , Adenosina Monofosfato/metabolismo , Proteínas/análisis , Proteínas/metabolismo , Proteómica/métodos , Adenosina Monofosfato/química , Marcaje Isotópico , Espectrometría de Masas , Proteínas/química
12.
PLoS One ; 8(5): e64901, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23738009

RESUMEN

The ubiquitous FIC domain is evolutionarily conserved from bacteria to human and has been shown to catalyze AMP transfer onto protein side-chain hydroxyl groups. Recently, it was predicted that most catalytically competent Fic proteins are inhibited by the presence of an inhibitory helix αinh that is provided by a cognate anti-toxin (class I), or is part of the N- or C-terminal part of the Fic protein itself (classes II and III). In vitro, inhibition is relieved by mutation of a conserved glutamate of αinh to glycine. For the class III bacterial Fic protein NmFic from Neisseria meningitidis, the inhibitory mechanism has been elucidated. Here, we extend above study by including bacterial class I and II Fic proteins VbhT from Bartonella schoenbuchensis and SoFic from Shewanella oneidensis, respectively, and the respective E->G mutants. Comparative enzymatic and crystallographic analyses show that, in all three classes, the ATP substrate binds to the wild-type FIC domains, but with the α-phosphate in disparate and non-competent orientations. In the E->G mutants, however, the tri-phosphate moiety is found reorganized to the same tightly bound structure through a unique set of hydrogen bonds with Fic signature motif residues. The γ-phosphate adopts the location that is taken by the inhibitory glutamate in wild-type resulting in an α-phosphate orientation that can be attacked in-line by a target side-chain hydroxyl group. The latter is properly registered to the Fic active center by main-chain ß-interactions with the ß-hairpin flap. These data indicate that the active site motif and the exposed edge of the flap are both required to form an adenylylation-competent Fic protein.


Asunto(s)
Adenosina Trifosfato/metabolismo , Dominio Catalítico , Secuencia Conservada , Nucleotidiltransferasas/antagonistas & inhibidores , Nucleotidiltransferasas/metabolismo , Adenilil Imidodifosfato/metabolismo , Bartonella/enzimología , Biocatálisis , Ácido Glutámico , Modelos Moleculares , Mutación , Nucleotidiltransferasas/química , Nucleotidiltransferasas/genética , Unión Proteica , Shewanella/enzimología
13.
N Biotechnol ; 30(3): 308-13, 2013 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-23395778

RESUMEN

Hand-in-hand with the availability of full genome sequences for eukaryotic model organisms and humans the demand for analysis of gene function on a system level has grown. In a process called RNA interference (RNAi) specific mRNA species can be degraded by introduction of double-stranded small interfering RNAs (siRNAs) that are complementary to the targeted transcript sequence. This enables the selective impairment of gene function. During the past decade RNAi has been exploited in many different eukaryotic cell types and model organisms. Large-scale and eventually genome-wide RNAi screens ablating gene functions in a systematic manner have delivered an overwhelming amount of data on the requirement of distinct gene products for major cellular pathways. A large part of the RNAi field is dedicated to disease states such as cancer or infection with the prospect of discovering pathways suitable for new therapeutic interventions. Here some of the major steps in the development of the RNAi technology will be outlined and exemplified with a focus on the progress made in the field of mammalian host-pathogen interactions.


Asunto(s)
Interacciones Huésped-Patógeno/genética , Interferencia de ARN , Biología de Sistemas , Animales , Humanos , ARN Interferente Pequeño/genética
14.
Cell Microbiol ; 15(5): 759-78, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23163798

RESUMEN

The Gram-negative, zoonotic pathogen Bartonella henselae is the aetiological agent of cat scratch disease, bacillary angiomatosis and peliosis hepatis in humans. Two pathogenicity factors of B. henselae - each displaying multiple functions in host cell interaction - have been characterized in greater detail: the trimeric autotransporter Bartonella adhesin A (BadA) and the type IV secretion system VirB/D4 (VirB/D4 T4SS). BadA mediates, e.g. binding to fibronectin (Fn), adherence to endothelial cells (ECs) and secretion of vascular endothelial growth factor (VEGF). VirB/D4 translocates several Bartonella effector proteins (Beps) into the cytoplasm of infected ECs, resulting, e.g. in uptake of bacterial aggregates via the invasome structure, inhibition of apoptosis and activation of a proangiogenic phenotype. Despite this knowledge of the individual activities of BadA or VirB/D4 it is unknown whether these major virulence factors affect each other in their specific activities. In this study, expression and function of BadA and VirB/D4 were analysed in a variety of clinical B. henselae isolates. Data revealed that most isolates have lost expression of either BadA or VirB/D4 during in vitro passages. However, the phenotypic effects of coexpression of both virulence factors was studied in one clinical isolate that was found to stably coexpress BadA and VirB/D4, as well as by ectopic expression of BadA in a strain expressing VirB/D4 but not BadA. BadA, which forms a dense layer on the bacterial surface, negatively affected VirB/D4-dependent Bep translocation and invasome formation by likely preventing close contact between the bacterial cell envelope and the host cell membrane. In contrast, BadA-dependent Fn binding, adhesion to ECs and VEGF secretion were not affected by a functional VirB/D4 T4SS. The obtained data imply that the essential virulence factors BadA and VirB/D4 are likely differentially expressed during different stages of the infection cycle of Bartonella.


Asunto(s)
Adhesinas Bacterianas/metabolismo , Bartonella henselae/genética , Enfermedad por Rasguño de Gato/microbiología , Gatos/microbiología , Animales , Adhesión Bacteriana/genética , Bartonella henselae/patogenicidad , Enfermedad por Rasguño de Gato/metabolismo , Línea Celular , Células Endoteliales/citología , Células Endoteliales/metabolismo , Células Endoteliales/microbiología , Regulación Bacteriana de la Expresión Génica , Interacciones Huésped-Patógeno , Unión Proteica , Virulencia/genética , Factores de Virulencia/genética , Factores de Virulencia/metabolismo
15.
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
16.
FEMS Microbiol Rev ; 36(3): 563-99, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22229763

RESUMEN

Bartonella spp. are facultative intracellular bacteria that typically cause a long-lasting intraerythrocytic bacteremia in their mammalian reservoir hosts, thereby favoring transmission by blood-sucking arthropods. In most cases, natural reservoir host infections are subclinical and the relapsing intraerythrocytic bacteremia may last weeks, months, or even years. In this review, we will follow the infection cycle of Bartonella spp. in a reservoir host, which typically starts with an intradermal inoculation of bacteria that are superficially scratched into the skin from arthropod feces and terminates with the pathogen exit by the blood-sucking arthropod. The current knowledge of bacterial countermeasures against mammalian immune response will be presented for each critical step of the pathogenesis. The prevailing models of the still-enigmatic primary niche and the anatomical location where bacteria reside, persist, and are periodically seeded into the bloodstream to cause the typical relapsing Bartonella spp. bacteremia will also be critically discussed. The review will end up with a discussion of the ability of Bartonella spp., namely Bartonella henselae, Bartonella quintana, and Bartonella bacilliformis, to induce tumor-like vascular deformations in humans having compromised immune response such as in patients with AIDS.


Asunto(s)
Infecciones por Bartonella/inmunología , Infecciones por Bartonella/microbiología , Bartonella bacilliformis/patogenicidad , Bartonella henselae/patogenicidad , Bartonella quintana/patogenicidad , Interacciones Huésped-Patógeno , Animales , Artrópodos/microbiología , Infecciones Asintomáticas , Bacteriemia/microbiología , Bacteriemia/patología , Infecciones por Bartonella/patología , Bartonella bacilliformis/inmunología , Bartonella henselae/inmunología , Bartonella quintana/inmunología , Enfermedad Crónica , Vectores de Enfermedades , Humanos , Zoonosis/microbiología , Zoonosis/transmisión
17.
Nature ; 482(7383): 107-10, 2012 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-22266942

RESUMEN

Fic proteins that are defined by the ubiquitous FIC (filamentation induced by cyclic AMP) domain are known to catalyse adenylylation (also called AMPylation); that is, the transfer of AMP onto a target protein. In mammalian cells, adenylylation of small GTPases through Fic proteins injected by pathogenic bacteria can cause collapse of the actin cytoskeleton and cell death. It is unknown how this potentially deleterious adenylylation activity is regulated in the widespread Fic proteins that are found in all domains of life and that are thought to have critical roles in intrinsic signalling processes. Here we show that FIC-domain-mediated adenylylation is controlled by a conserved mechanism of ATP-binding-site obstruction that involves an inhibitory α-helix (α(inh)) with a conserved (S/T)XXXE(G/N) motif, and that in this mechanism the invariable glutamate competes with ATP γ-phosphate binding. Consistent with this, FIC-domain-mediated growth arrest of bacteria by the VbhT toxin of Bartonella schoenbuchensis is intermolecularly repressed by the VbhA antitoxin through tight binding of its α(inh) to the FIC domain of VbhT, as shown by structure and function analysis. Furthermore, structural comparisons with other bacterial Fic proteins, such as Fic of Neisseria meningitidis and of Shewanella oneidensis, show that α(inh) frequently constitutes an amino-terminal or carboxy-terminal extension to the FIC domain, respectively, partially obstructing the ATP binding site in an intramolecular manner. After mutation of the inhibitory motif in various Fic proteins, including the human homologue FICD (also known as HYPE), adenylylation activity is considerably boosted, consistent with the anticipated relief of inhibition. Structural homology modelling of all annotated Fic proteins indicates that inhibition by α(inh) is universal and conserved through evolution, as the inhibitory motif is present in ∼90% of all putatively adenylylation-active FIC domains, including examples from all domains of life and from viruses. Future studies should reveal how intrinsic or extrinsic factors modulate adenylylation activity by weakening the interaction of α(inh) with the FIC active site.


Asunto(s)
Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , AMP Cíclico/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Bartonella , Proteínas Portadoras/química , Proteínas Portadoras/metabolismo , Catálisis , Dominio Catalítico , Escherichia coli , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Ácido Glutámico/metabolismo , Humanos , Proteínas de la Membrana/química , Proteínas de la Membrana/metabolismo , Viabilidad Microbiana , Modelos Moleculares , Peso Molecular , Neisseria meningitidis , Nucleotidiltransferasas , Estructura Terciaria de Proteína , Shewanella
18.
PLoS One ; 6(10): e25106, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22043280

RESUMEN

The gram-negative, zoonotic pathogen Bartonella henselae (Bhe) translocates seven distinct Bartonella effector proteins (Beps) via the VirB/VirD4 type IV secretion system (T4SS) into human cells, thereby interfering with host cell signaling [1], [2]. In particular, the effector protein BepG alone or the combination of effector proteins BepC and BepF trigger massive F-actin rearrangements that lead to the establishment of invasome structures eventually resulting in the internalization of entire Bhe aggregates [2], [3]. In this report, we investigate the molecular function of the effector protein BepF in the eukaryotic host cell. We show that the N-terminal [E/T]PLYAT tyrosine phosphorylation motifs of BepF get phosphorylated upon translocation but do not contribute to invasome-mediated Bhe uptake. In contrast, we found that two of the three BID domains of BepF are capable to trigger invasome formation together with BepC, while a mutation of the WxxxE motif of the BID-F1 domain inhibited its ability to contribute to the formation of invasome structures. Next, we show that BepF function during invasome formation can be replaced by the over-expression of constitutive-active Rho GTPases Rac1 or Cdc42. Finally we demonstrate that BID-F1 and BID-F2 domains promote the formation of filopodia-like extensions in NIH 3T3 and HeLa cells as well as membrane protrusions in HeLa cells, suggesting a role for BepF in Rac1 and Cdc42 activation during the process of invasome formation.


Asunto(s)
Proteínas Bacterianas/fisiología , Sistemas de Secreción Bacterianos/fisiología , Traslocación Bacteriana , Bartonella henselae/patogenicidad , Angiomatosis Bacilar , Animales , Células HeLa , Humanos , Ratones , Células 3T3 NIH
19.
J Cell Sci ; 124(Pt 21): 3591-602, 2011 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-22045736

RESUMEN

The VirB/D4 type IV secretion system (T4SS) of the bacterial pathogen Bartonella henselae (Bhe) translocates seven effector proteins (BepA-BepG) into human cells that subvert host cellular functions. Two redundant pathways dependent on BepG or the combination of BepC and BepF trigger the formation of a bacterial uptake structure termed the invasome. Invasome formation is a multi-step process consisting of bacterial adherence, effector translocation, aggregation of bacteria on the cell surface and engulfment, and eventually, complete internalization of the bacterial aggregate occurs in an F-actin-dependent manner. In the present study, we show that Bhe-triggered invasome formation depends on integrin-ß1-mediated signaling cascades that enable assembly of the F-actin invasome structure. We demonstrate that Bhe interacts with integrin ß1 in a fibronectin- and VirB/D4 T4SS-independent manner and that activated integrin ß1 is essential for both effector translocation and the actin rearrangements leading to invasome formation. Furthermore, we show that talin1, but not talin2, is required for inside-out activation of integrin ß1 during invasome formation. Finally, integrin-ß1-mediated outside-in signaling by FAK, Src, paxillin and vinculin is necessary for invasome formation. This is the first example of a bacterial entry process that fully exploits the bi-directional signaling capacity of integrin receptors in a talin1-specific manner.


Asunto(s)
Angiomatosis Bacilar/metabolismo , Angiomatosis Bacilar/microbiología , Bartonella henselae/fisiología , Integrina beta1/metabolismo , Transducción de Señal , Talina/metabolismo , Actinas/metabolismo , Angiomatosis Bacilar/genética , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Bartonella henselae/genética , Bartonella henselae/patogenicidad , Línea Celular , Membrana Celular/genética , Membrana Celular/metabolismo , Membrana Celular/microbiología , Humanos , Integrina beta1/genética , Unión Proteica , Transporte de Proteínas , Talina/genética
20.
J Bacteriol ; 193(22): 6257-65, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21908662

RESUMEN

Bacterial type IV secretion systems (T4SSs) are involved in processes such as bacterial conjugation and protein translocation to animal cells. In this work, we have switched the substrates of T4SSs involved in pathogenicity for DNA transfer. Plasmids containing part of the conjugative machinery of plasmid R388 were transferred by the T4SS of human facultative intracellular pathogen Bartonella henselae to both recipient bacteria and human vascular endothelial cells. About 2% of the human cells expressed a green fluorescent protein (GFP) gene from the plasmid. Plasmids of different sizes were transferred with similar efficiencies. B. henselae codes for two T4SSs: VirB/VirD4 and Trw. A ΔvirB mutant strain was transfer deficient, while a ΔtrwE mutant was only slightly impaired in DNA transfer. DNA transfer was in all cases dependent on protein TrwC of R388, the conjugative relaxase, implying that it occurs by a conjugation-like mechanism. A DNA helicase-deficient mutant of TrwC could not promote DNA transfer. In the absence of TrwB, the coupling protein of R388, DNA transfer efficiency dropped 1 log. The same low efficiency was obtained with a TrwB point mutation in the region involved in interaction with the T4SS. TrwB interacted with VirB10 in a bacterial two-hybrid assay, suggesting that it may act as the recruiter of the R388 substrate for the VirB/VirD4 T4SS. A TrwB ATPase mutant behaved as dominant negative, dropping DNA transfer efficiency to almost null levels. B. henselae bacteria recovered from infected human cells could transfer the mobilizable plasmid into recipient Escherichia coli under certain conditions, underscoring the versatility of T4SSs.


Asunto(s)
Angiomatosis Bacilar/microbiología , Proteínas Bacterianas/metabolismo , Sistemas de Secreción Bacterianos , Bartonella henselae/genética , Conjugación Genética , Células Endoteliales/microbiología , Plásmidos/genética , Transfección , Angiomatosis Bacilar/genética , Proteínas Bacterianas/genética , Bartonella henselae/metabolismo , Bartonella henselae/patogenicidad , Línea Celular , Escherichia coli/genética , Humanos , Plásmidos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA