Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 121(25): e2315481121, 2024 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-38870060

RESUMEN

Intracellular bacterial pathogens divert multiple cellular pathways to establish their niche and persist inside their host. Coxiella burnetii, the causative agent of Q fever, secretes bacterial effector proteins via its Type 4 secretion system to generate a Coxiella-containing vacuole (CCV). Manipulation of lipid and protein trafficking by these effectors is essential for bacterial replication and virulence. Here, we have characterized the lipid composition of CCVs and found that the effector Vice interacts with phosphoinositides and membranes enriched in phosphatidylserine and lysobisphosphatidic acid. Remarkably, eukaryotic cells ectopically expressing Vice present compartments that resemble early CCVs in both morphology and composition. We found that the biogenesis of these compartments relies on the double function of Vice. The effector protein initially localizes at the plasma membrane of eukaryotic cells where it triggers the internalization of large vacuoles by macropinocytosis. Then, Vice stabilizes these compartments by perturbing the ESCRT machinery. Collectively, our results reveal that Vice is an essential C. burnetii effector protein capable of hijacking two major cellular pathways to shape the bacterial replicative niche.


Asunto(s)
Proteínas Bacterianas , Coxiella burnetii , Complejos de Clasificación Endosomal Requeridos para el Transporte , Pinocitosis , Vacuolas , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Proteínas Bacterianas/metabolismo , Coxiella burnetii/metabolismo , Vacuolas/metabolismo , Vacuolas/microbiología , Humanos , Células HeLa , Membrana Celular/metabolismo , Animales , Fosfatidilinositoles/metabolismo
2.
Biol Cell ; 114(9): 237-253, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-35851960

RESUMEN

Ezrin protein is involved in the interaction of actin cytoskeleton with membrane receptors such as CD44. It regulates plasma membrane dynamics and intracellular signaling. Coxiella burnetii, the etiologic agent of Q fever, is internalized into host cell through a poorly characterized molecular mechanism. Here we analyzed the role of ezrin and CD44 in the C. burnetii internalization by HeLa cells. The knockdown of ezrin and CD44 inhibited the bacterial uptake. Interestingly, at early stages of C. burnetii internalization, ezrin was recruited to the cell membrane fraction and phosphorylated. Moreover, the overexpression of non-phosphorylatable and phosphomimetic ezrin mutants decreased and increased the bacterial entry, respectively. A decrease in the internalization of C. burnetii was observed by the overexpression of CD44 truncated forms containing the intracellular or the extracellular domains. Interestingly, the CD44 mutant was unable to interact with ERM proteins decreased the bacterial internalization. These findings demonstrate the participation of ezrin in the internalization process of C. burnetii in non-phagocytic cells. Additionally, we present evidence that CD44 receptor would be involved in that process.


Asunto(s)
Coxiella burnetii , Proteínas del Citoesqueleto/metabolismo , Receptores de Hialuranos/metabolismo , Citoesqueleto de Actina , Coxiella burnetii/metabolismo , Células HeLa , Humanos
3.
Proc Natl Acad Sci U S A ; 117(24): 13708-13718, 2020 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-32482853

RESUMEN

The Q fever agent Coxiella burnetii uses a defect in organelle trafficking/intracellular multiplication (Dot/Icm) type 4b secretion system (T4SS) to silence the host innate immune response during infection. By investigating C. burnetii effector proteins containing eukaryotic-like domains, here we identify NopA (nucleolar protein A), which displays four regulator of chromosome condensation (RCC) repeats, homologous to those found in the eukaryotic Ras-related nuclear protein (Ran) guanine nucleotide exchange factor (GEF) RCC1. Accordingly, NopA is found associated with the chromatin nuclear fraction of cells and uses the RCC-like domain to interact with Ran. Interestingly, NopA triggers an accumulation of Ran-GTP, which accumulates at nucleoli of transfected or infected cells, thus perturbing the nuclear import of transcription factors of the innate immune signaling pathway. Accordingly, qRT-PCR analysis on a panel of cytokines shows that cells exposed to the C. burnetii nopA::Tn or a Dot/Icm-defective dotA::Tn mutant strain present a functional innate immune response, as opposed to cells exposed to wild-type C. burnetii or the corresponding nopA complemented strain. Thus, NopA is an important regulator of the innate immune response allowing Coxiella to behave as a stealth pathogen.


Asunto(s)
Proteínas Bacterianas/metabolismo , Coxiella burnetii/metabolismo , Fiebre Q/inmunología , Animales , Proteínas Bacterianas/genética , Coxiella burnetii/genética , Femenino , Interacciones Huésped-Patógeno , Humanos , Inmunidad Innata , Ratones , Ratones Endogámicos C57BL , Ratones SCID , Fiebre Q/genética , Fiebre Q/microbiología
4.
J Biol Chem ; 295(21): 7391-7403, 2020 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-32303638

RESUMEN

The intracellular bacterial pathogen Coxiella burnetii is the etiological agent of the emerging zoonosis Q fever. Crucial to its pathogenesis is type 4b secretion system-mediated secretion of bacterial effectors into host cells that subvert host cell membrane trafficking, leading to the biogenesis of a parasitophorous vacuole for intracellular replication. The characterization of prokaryotic serine/threonine protein kinases in bacterial pathogens is emerging as an important strategy to better understand host-pathogen interactions. In this study, we investigated CstK (for Coxiella Ser/Thr kinase), a protein kinase identified in C. burnetii by in silico analysis. We demonstrate that this putative protein kinase undergoes autophosphorylation on Thr and Tyr residues and phosphorylates a classical eukaryotic protein kinase substrate in vitro This dual Thr-Tyr kinase activity is also observed for a eukaryotic dual-specificity Tyr phosphorylation-regulated kinase class. We found that CstK is translocated during infections and localizes to Coxiella-containing vacuoles (CCVs). Moreover, a CstK-overexpressing C. burnetii strain displayed a severe CCV development phenotype, suggesting that CstK fine-tunes CCV biogenesis during the infection. Protein-protein interaction experiments identified the Rab7 GTPase-activating protein TBC1D5 as a candidate CstK-specific target, suggesting a role for this host GTPase-activating protein in Coxiella infections. Indeed, CstK co-localized with TBC1D5 in noninfected cells, and TBC1D5 was recruited to CCVs in infected cells. Accordingly, TBC1D5 depletion from infected cells significantly affected CCV development. Our results indicate that CstK functions as a bacterial effector protein that interacts with the host protein TBC1D5 during vacuole biogenesis and intracellular replication.


Asunto(s)
Proteínas Bacterianas/metabolismo , Coxiella burnetii/enzimología , Proteínas Activadoras de GTPasa/metabolismo , Proteínas Quinasas/metabolismo , Fiebre Q/metabolismo , Vacuolas/metabolismo , Proteínas Bacterianas/genética , Línea Celular Tumoral , Coxiella burnetii/genética , Proteínas Activadoras de GTPasa/genética , Humanos , Fosforilación , Proteínas Quinasas/genética , Fiebre Q/genética , Vacuolas/genética , Vacuolas/microbiología
5.
Cell Microbiol ; 22(4): e13180, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32185905

RESUMEN

Coxiella burnetii is an obligate intracellular bacterial pathogen responsible for severe worldwide outbreaks of the zoonosis Q fever. The remarkable resistance to environmental stress, extremely low infectious dose and ease of dissemination, contributed to the classification of C. burnetii as a class B biothreat. Unique among intracellular pathogens, C. burnetii escapes immune surveillance and replicates within large autophagolysosome-like compartments called Coxiella-containing vacuoles (CCVs). The biogenesis of these compartments depends on the subversion of several host signalling pathways. For years, the obligate intracellular nature of C. burnetii imposed significant experimental obstacles to the study of its pathogenic traits. With the development of an axenic culture medium in 2009, C. burnetii became genetically tractable, thus allowing the implementation of mutagenesis tools and screening approaches to identify its virulence determinants and investigate its complex interaction with host cells. Here, we review the key advances that have contributed to our knowledge of C. burnetii pathogenesis, leading to the rise of this once-neglected pathogen to an exceptional organism to study the intravacuolar lifestyle.


Asunto(s)
Coxiella burnetii/genética , Coxiella burnetii/patogenicidad , Interacciones Huésped-Patógeno , Vacuolas/microbiología , Animales , Autofagosomas/microbiología , Cultivo Axénico/métodos , Proteínas Bacterianas/metabolismo , Sistemas de Secreción Bacterianos/metabolismo , Biotecnología , Humanos , Mutación , Fenotipo , Fiebre Q/microbiología
6.
J Bacteriol ; 201(22)2019 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-31451541

RESUMEN

Coxiella burnetii is an obligate intracellular gammaproteobacterium and zoonotic agent of Q fever. We previously identified 15 small noncoding RNAs (sRNAs) of C. burnetii One of them, CbsR12 (Coxiella burnetiismall RNA 12), is highly transcribed during axenic growth and becomes more prominent during infection of cultured mammalian cells. Secondary structure predictions of CbsR12 revealed four putative CsrA-binding sites in stem loops with consensus AGGA/ANGGA motifs. We subsequently determined that CbsR12 binds to recombinant C. burnetii CsrA-2, but not CsrA-1, proteins in vitro Moreover, through a combination of in vitro and cell culture assays, we identified several in trans mRNA targets of CbsR12. Of these, we determined that CbsR12 binds and upregulates translation of carA transcripts coding for carbamoyl phosphate synthetase A, an enzyme that catalyzes the first step of pyrimidine biosynthesis. In addition, CbsR12 binds and downregulates translation of metK transcripts coding for S-adenosylmethionine synthetase, a component of the methionine cycle. Furthermore, we found that CbsR12 binds to and downregulates the quantity of cvpD transcripts, coding for a type IVB effector protein, in mammalian cell culture. Finally, we found that CbsR12 is necessary for expansion of Coxiella-containing vacuoles and affects growth rates in a dose-dependent manner in the early phase of infecting THP-1 cells. This is the first characterization of a trans-acting sRNA of C. burnetii and the first example of a bacterial sRNA that regulates both CarA and MetK synthesis. CbsR12 is one of only a few identified trans-acting sRNAs that interacts with CsrA.IMPORTANCE Regulation of metabolism and virulence in C. burnetii is not well understood. Here, we show that C. burnetii small RNA 12 (CbsR12) is highly transcribed in the metabolically active large-cell variant compared to the nonreplicative small-cell variant. We show that CbsR12 directly regulates several genes involved in metabolism, along with a type IV effector gene, in trans In addition, we demonstrate that CbsR12 binds to CsrA-2 in vitro and induces autoaggregation and biofilm formation when transcribed ectopically in Escherichia coli, consistent with other CsrA-sequestering sRNAs. These results implicate CbsR12 in the indirect regulation of a number of genes via CsrA-mediated regulatory activities. The results also support CbsR12 as a crucial regulatory component early on in a mammalian cell infection.


Asunto(s)
Coxiella burnetii/genética , Fiebre Q/microbiología , ARN Bacteriano/fisiología , ARN Pequeño no Traducido/fisiología , Proteínas de Unión al ARN/metabolismo , Vacuolas/metabolismo , Animales , Cultivo Axénico , Proteínas Bacterianas/metabolismo , Chlorocebus aethiops , Coxiella burnetii/crecimiento & desarrollo , Coxiella burnetii/metabolismo , Humanos , ARN Bacteriano/genética , ARN Bacteriano/metabolismo , ARN Pequeño no Traducido/genética , ARN Pequeño no Traducido/metabolismo , Células THP-1 , Células Vero
7.
Proc Natl Acad Sci U S A ; 113(23): E3260-9, 2016 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-27226300

RESUMEN

The Q fever bacterium Coxiella burnetii replicates inside host cells within a large Coxiella-containing vacuole (CCV) whose biogenesis relies on the Dot/Icm-dependent secretion of bacterial effectors. Several membrane trafficking pathways contribute membranes, proteins, and lipids for CCV biogenesis. These include the endocytic and autophagy pathways, which are characterized by phosphatidylinositol 3-phosphate [PI(3)P]-positive membranes. Here we show that the C. burnetii secreted effector Coxiella vacuolar protein B (CvpB) binds PI(3)P and phosphatidylserine (PS) on CCVs and early endosomal compartments and perturbs the activity of the phosphatidylinositol 5-kinase PIKfyve to manipulate PI(3)P metabolism. CvpB association to early endosome triggers vacuolation and clustering, leading to the channeling of large PI(3)P-positive membranes to CCVs for vacuole expansion. At CCVs, CvpB binding to early endosome- and autophagy-derived PI(3)P and the concomitant inhibition of PIKfyve favor the association of the autophagosomal machinery to CCVs for optimal homotypic fusion of the Coxiella-containing compartments. The importance of manipulating PI(3)P metabolism is highlighted by mutations in cvpB resulting in a multivacuolar phenotype, rescuable by gene complementation, indicative of a defect in CCV biogenesis. Using the insect model Galleria mellonella, we demonstrate the in vivo relevance of defective CCV biogenesis by highlighting an attenuated virulence phenotype associated with cvpB mutations.


Asunto(s)
Proteínas Bacterianas/metabolismo , Sistemas de Secreción Bacterianos/metabolismo , Coxiella burnetii , Vacuolas/metabolismo , Animales , Proteínas Bacterianas/genética , Sistemas de Secreción Bacterianos/genética , Chlorocebus aethiops , Coxiella burnetii/metabolismo , Coxiella burnetii/patogenicidad , Humanos , Lepidópteros/microbiología , Mutación , Fosfatos de Fosfatidilinositol/metabolismo , Fosfatidilserinas/metabolismo , Virulencia
8.
Curr Top Microbiol Immunol ; 413: 243-268, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29536362

RESUMEN

Coxiella burnetii is the etiological agent of the zoonotic disease Q fever, which manifests in severe outbreaks and is associated with important health and economic burden. Moreover, C. burnetii belongs to the list of class B bioterrorism organisms, as it is an airborne and highly infective pathogen with remarkable resistance to environmental stresses. Detailed study of the host-pathogen interaction during C. burnetii infection has been hampered due to the obligate intracellular nature of this pathogen. However, the development of an axenic culture medium, together with the implementation of bioinformatics tools and high-content screening approaches, have significantly progressed C. burnetii research in the last decade. This has facilitated identification of the Dot/Icm type IV secretion system (T4SS) as an essential virulence factor. T4SS is used to deliver an arsenal of effector proteins into the cytoplasm of the host cell. These effectors mediate the survival of the host cell and the development of very large replicative compartments called Coxiella-containing vacuoles (CCVs). Biogenesis of the CCV relies on T4SS-dependent re-routing of numerous intracellular trafficking pathways to deliver membranes and nutrients that are essential for bacterial replication. This review aims to illustrate the key milestones that have contributed to ascribe C. burnetii as a model organism for the study of host/pathogen interactions as well as presenting an up-to-date description of our knowledge of the cell biology of C. burnetii infections.


Asunto(s)
Coxiella burnetii , Fiebre Q , Proteínas Bacterianas , Sistemas de Secreción Bacterianos , Interacciones Huésped-Patógeno , Humanos , Sistemas de Secreción Tipo IV
9.
PLoS Pathog ; 11(5): e1004892, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25978383

RESUMEN

Q fever is a highly infectious disease with a worldwide distribution. Its causative agent, the intracellular bacterium Coxiella burnetii, infects a variety of vertebrate species, including humans. Its evolutionary origin remains almost entirely unknown and uncertainty persists regarding the identity and lifestyle of its ancestors. A few tick species were recently found to harbor maternally-inherited Coxiella-like organisms engaged in symbiotic interactions, but their relationships to the Q fever pathogen remain unclear. Here, we extensively sampled ticks, identifying new and atypical Coxiella strains from 40 of 58 examined species, and used this data to infer the evolutionary processes leading to the emergence of C. burnetii. Phylogenetic analyses of multi-locus typing and whole-genome sequencing data revealed that Coxiella-like organisms represent an ancient and monophyletic group allied to ticks. Remarkably, all known C. burnetii strains originate within this group and are the descendants of a Coxiella-like progenitor hosted by ticks. Using both colony-reared and field-collected gravid females, we further establish the presence of highly efficient maternal transmission of these Coxiella-like organisms in four examined tick species, a pattern coherent with an endosymbiotic lifestyle. Our laboratory culture assays also showed that these Coxiella-like organisms were not amenable to culture in the vertebrate cell environment, suggesting different metabolic requirements compared to C. burnetii. Altogether, this corpus of data demonstrates that C. burnetii recently evolved from an inherited symbiont of ticks which succeeded in infecting vertebrate cells, likely by the acquisition of novel virulence factors.


Asunto(s)
Evolución Biológica , Enfermedades Transmisibles Emergentes/transmisión , Coxiella burnetii/fisiología , Salud Global , Fiebre Q/transmisión , Simbiosis , Garrapatas/microbiología , Animales , Secuencia de Bases , Conducta Animal , Línea Celular , Enfermedades Transmisibles Emergentes/epidemiología , Enfermedades Transmisibles Emergentes/microbiología , Enfermedades Transmisibles Emergentes/veterinaria , Coxiella burnetii/clasificación , Coxiella burnetii/crecimiento & desarrollo , Coxiella burnetii/aislamiento & purificación , Coxiellaceae/clasificación , Coxiellaceae/crecimiento & desarrollo , Coxiellaceae/aislamiento & purificación , Coxiellaceae/fisiología , Femenino , Genoma Bacteriano , Humanos , Masculino , Intercambio Materno-Fetal , Viabilidad Microbiana , Datos de Secuencia Molecular , Filogenia , Embarazo , Prevalencia , Fiebre Q/epidemiología , Fiebre Q/microbiología , Fiebre Q/veterinaria , Garrapatas/fisiología
10.
PLoS Pathog ; 10(3): e1004013, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24651569

RESUMEN

Coxiella burnetii is the agent of the emerging zoonosis Q fever. This pathogen invades phagocytic and non-phagocytic cells and uses a Dot/Icm secretion system to co-opt the endocytic pathway for the biogenesis of an acidic parasitophorous vacuole where Coxiella replicates in large numbers. The study of the cell biology of Coxiella infections has been severely hampered by the obligate intracellular nature of this microbe, and Coxiella factors involved in host/pathogen interactions remain to date largely uncharacterized. Here we focus on the large-scale identification of Coxiella virulence determinants using transposon mutagenesis coupled to high-content multi-phenotypic screening. We have isolated over 3000 Coxiella mutants, 1082 of which have been sequenced, annotated and screened. We have identified bacterial factors that regulate key steps of Coxiella infections: 1) internalization within host cells, 2) vacuole biogenesis/intracellular replication, and 3) protection of infected cells from apoptosis. Among these, we have investigated the role of Dot/Icm core proteins, determined the role of candidate Coxiella Dot/Icm substrates previously identified in silico and identified additional factors that play a relevant role in Coxiella pathogenesis. Importantly, we have identified CBU_1260 (OmpA) as the first Coxiella invasin. Mutations in ompA strongly decreased Coxiella internalization and replication within host cells; OmpA-coated beads adhered to and were internalized by non-phagocytic cells and the ectopic expression of OmpA in E. coli triggered its internalization within cells. Importantly, Coxiella internalization was efficiently inhibited by pretreating host cells with purified OmpA or by incubating Coxiella with a specific anti-OmpA antibody prior to host cell infection, suggesting the presence of a cognate receptor at the surface of host cells. In summary, we have developed multi-phenotypic assays for the study of host/pathogen interactions. By applying our methods to Coxiella burnetii, we have identified the first Coxiella protein involved in host cell invasion.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/genética , Coxiella burnetii/fisiología , Coxiella burnetii/patogenicidad , Interacciones Huésped-Patógeno/genética , Fiebre Q/genética , Animales , Elementos Transponibles de ADN , Técnica del Anticuerpo Fluorescente , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Microscopía Electrónica de Rastreo , Mutagénesis Sitio-Dirigida , Fenotipo , Virulencia
11.
Traffic ; 13(12): 1653-66, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22984946

RESUMEN

Invasive bacterial pathogens often target cellular proteins involved in adhesion as a first event during infection. For example, Listeria monocytogenes uses the bacterial protein InlA to interact with E-cadherin, hijack the host adherens junction (AJ) machinery and invade non-phagocytic cells by a clathrin-dependent mechanism. Here, we investigate a potential role for clathrin in cell-cell adhesion. We observed that the initial steps of AJ formation trigger the phosphorylation of clathrin, and its transient localization at forming cell-cell contacts. Furthermore, we show that clathrin serves as a hub for the recruitment of proteins that are necessary for the actin rearrangements that accompany the maturation of AJs. Using an InlA/E-cadherin chimera, we show that adherent cells expressing the chimera form AJs with cells expressing E-cadherin. We demonstrate that non-adherent cells expressing the InlA chimera, as bacteria, can be internalized by E-cadherin-expressing adherent cells. Together these results reveal that a common clathrin-mediated machinery may regulate internalization and cell adhesion and that the relative mobility of one of the interacting partners plays an important role in the commitment to either one of these processes.


Asunto(s)
Uniones Adherentes/microbiología , Clatrina/metabolismo , Endocitosis , Uniones Adherentes/metabolismo , Animales , Proteínas Bacterianas/metabolismo , Cadherinas/metabolismo , Adhesión Celular , Perros , Células HeLa , Interacciones Huésped-Patógeno , Humanos , Listeria monocytogenes/patogenicidad , Células de Riñón Canino Madin Darby , Fosforilación
12.
J Biol Chem ; 287(20): 16849-59, 2012 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-22393044

RESUMEN

Group IVα phospholipase A(2) (PLA(2)IVα) is a lipolytic enzyme that catalyzes the hydrolysis of membrane phospholipids to generate precursors of potent inflammatory lipid mediators. Here, the role of PLA(2)IVα in Fc receptor (FcR)-mediated phagocytosis was investigated, demonstrating that PLA(2)IVα is selectively activated upon FcR-mediated phagocytosis in macrophages and that it rapidly translocates to the site of the nascent phagosome. Moreover, pharmacological inhibition of PLA(2)IVα by pyrrophenone reduces particle internalization by up to 50%. In parallel, fibroblasts from PLA(2)IVα knock-out mice overexpressing FcγRIIA and able to internalize IgG-opsonized beads show 50% lower phagocytosis, compared with wild-type cells, and transfection of PLA(2)IVα fully recovers this impaired function. Interestingly, transfection of the catalytically inactive deleted PLA(2)IVα mutant (PLA(2)IVα(1-525)) and point mutant (PLA(2)IVα-S228C) also promotes recovery of this impaired function. Finally, transfection of the PLA(2)IVα C2 domain (which is directly involved in PLA(2)IVα membrane binding), but not of PLA(2)IVα-D43N (which cannot bind to membranes), rescues FcR-mediated phagocytosis. These data unveil a new mechanism of action for PLA(2)IVα, which demonstrates that the membrane binding, and not the enzymatic activity, is required for PLA(2)IVα modulation of FcR-mediated phagocytosis.


Asunto(s)
Membrana Celular/enzimología , Fosfolipasas A2 Grupo IV/metabolismo , Macrófagos/enzimología , Fagocitosis/fisiología , Fagosomas/metabolismo , Sustitución de Aminoácidos , Animales , Línea Celular Transformada , Membrana Celular/genética , Fibroblastos/citología , Fibroblastos/enzimología , Fosfolipasas A2 Grupo IV/genética , Humanos , Macrófagos/citología , Ratones , Ratones Noqueados , Mutación Missense , Fagocitosis/efectos de los fármacos , Fagosomas/genética , Estructura Terciaria de Proteína , Transporte de Proteínas/efectos de los fármacos , Transporte de Proteínas/fisiología , Pirrolidinas/farmacología , Receptores de IgG/genética , Receptores de IgG/metabolismo
13.
Cell Microbiol ; 14(7): 1097-108, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22381134

RESUMEN

The bacterial pathogens Listeria monocytogenes and enteropathogenic Escherichia coli (EPEC) generate motile actin-rich structures (comet tails and pedestals) as part of their infectious processes. Nexilin, an actin-associated protein and a component of focal adhesions, has been suggested to be involved in actin-based motility. To determine whether nexilin is commandeered during L. monocytogenes and EPEC infections, we infected cultured cells and found that nexilin is crucial for L. monocytogenes invasion as levels of internalized bacteria were significantly decreased in nexilin-targeted siRNA-treated cells. In addition, nexilin is a component of the machinery that drives the formation of L. monocytogenes comet tails and EPEC pedestals. Nexilin colocalizes with stationary bacteria and accumulates at the distal portion of comet tails and pedestals of motile bacteria. We also show that nexilin is crucial for efficient comet tail formation as cells pre-treated with nexilin siRNA generate malformed comet tails, whereas nexilin is dispensable during EPEC pedestal generation. These findings demonstrate that nexilin is required for efficient infection with invasive and adherent bacteria and is key to the actin-rich structures these microbes generate.


Asunto(s)
Actinas/metabolismo , Escherichia coli Enteropatógena/metabolismo , Escherichia coli Enteropatógena/patogenicidad , Listeria monocytogenes/metabolismo , Listeria monocytogenes/patogenicidad , Proteínas de Microfilamentos/metabolismo , Línea Celular , Técnicas de Silenciamiento del Gen , Silenciador del Gen , Humanos
14.
Commun Biol ; 6(1): 1124, 2023 11 06.
Artículo en Inglés | MEDLINE | ID: mdl-37932372

RESUMEN

The intracellular bacterial pathogen Coxiella burnetii evades the host response by secreting effector proteins that aid in establishing a replication-friendly niche. Bacterial filamentation induced by cyclic AMP (Fic) enzymes can act as effectors by covalently modifying target proteins with the posttranslational AMPylation by transferring adenosine monophosphate (AMP) from adenosine triphosphate (ATP) to a hydroxyl-containing side chain. Here we identify the gene product of C. burnetii CBU_0822, termed C. burnetii Fic 2 (CbFic2), to AMPylate host cell histone H3 at serine 10 and serine 28. We show that CbFic2 acts as a bifunctional enzyme, both capable of AMPylation as well as deAMPylation, and is regulated by the binding of DNA via a C-terminal helix-turn-helix domain. We propose that CbFic2 performs AMPylation in its monomeric state, switching to a deAMPylating dimer upon DNA binding. This study unveils reversible histone modification by a specific enzyme of a pathogenic bacterium.


Asunto(s)
Coxiella burnetii , AMP Cíclico , Histonas , ADN , Serina
15.
J Virol ; 85(6): 2980-9, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21191016

RESUMEN

The ubiquitin ligase CBLL1 (also known as HAKAI) has been proposed to be a critical cellular factor exploited by West Nile virus (WNV) for productive infection. CBLL1 has emerged as a major hit in a recent RNA interference screen designed to identify cellular factors required for the early stages of the WNV life cycle. Follow-up experiments showed that HeLa cells knocked down for CBLL1 by a small interfering RNA (siRNA) failed to internalize WNV particles and resisted infection. Furthermore, depletion of a free-ubiquitin pool by the proteasome inhibitor MG132 abolished WNV endocytosis, suggesting that CBLL1 acts in concert with the ubiquitin proteasome system to mediate virus internalization. Here, we examined the effect of CBLL1 knockdown and proteasome inhibitors on infection by WNV and other flaviviruses. We identified new siRNAs that repress the CBLL1 protein and strongly inhibit the endocytosis of Listeria monocytogenes, a bacterial pathogen known to require CBLL1 to invade host cells. Strikingly, however, we detected efficient WNV, dengue virus, and yellow fever virus infection of human cells, despite potent downregulation of CBLL1 by RNA interference. In addition, we found that the proteasome inhibitors MG132 and lactacystin did not affect WNV internalization but strongly repressed flavivirus RNA translation and replication. Together, these data do not support a requirement for CBLL1 during flavivirus entry and rather suggest an essential role of the ubiquitin/proteasome pathway for flavivirus genome amplification.


Asunto(s)
Virus del Dengue/fisiología , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitina/metabolismo , Internalización del Virus , Replicación Viral , Virus del Nilo Occidental/fisiología , Virus de la Fiebre Amarilla/fisiología , Acetilcisteína/análogos & derivados , Acetilcisteína/metabolismo , Técnicas de Silenciamiento del Gen , Células HeLa , Humanos , Leupeptinas/metabolismo , Listeria monocytogenes/patogenicidad , Complejo de la Endopetidasa Proteasomal/metabolismo , Inhibidores de Proteasoma , ARN Interferente Pequeño/metabolismo , Ubiquitina-Proteína Ligasas/antagonistas & inhibidores
16.
Nat Cell Biol ; 7(6): 570-80, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15880102

RESUMEN

Membrane fission is a fundamental step in membrane transport. So far, the only fission protein machinery that has been implicated in in vivo transport involves dynamin, and functions in several, but not all, transport pathways. Thus, other fission machineries may exist. Here, we report that carboxy-terminal binding protein 3/brefeldin A-ribosylated substrate (CtBP3/BARS) controls fission in basolateral transport from the Golgi to the plasma membrane and in fluid-phase endocytosis, whereas dynamin is not involved in these steps. Conversely, CtBP3/BARS protein is inactive in apical transport to the plasma membrane and in receptor-mediated endocytosis, both steps being controlled by dynamin. This indicates that CtBP3/BARS controls membrane fission in endocytic and exocytic transport pathways, distinct from those that require dynamin.


Asunto(s)
Proteínas Portadoras/metabolismo , Dinaminas/metabolismo , Membranas Intracelulares/metabolismo , Orgánulos/metabolismo , Factores de Transcripción/metabolismo , Vesículas Transportadoras/metabolismo , Animales , Células COS , Membrana Celular/metabolismo , Membrana Celular/ultraestructura , Chlorocebus aethiops , Perros , Endocitosis/fisiología , Exocitosis/fisiología , Aparato de Golgi/metabolismo , Aparato de Golgi/ultraestructura , Membranas Intracelulares/ultraestructura , Microscopía Electrónica de Transmisión , Orgánulos/ultraestructura , Transporte de Proteínas/fisiología , Receptores de Superficie Celular/metabolismo , Vesículas Transportadoras/ultraestructura
17.
Bioessays ; 32(6): 496-504, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20486136

RESUMEN

Clathrin and the endocytosis machinery has recently been described as being required in mammalian cells for the internalization of large particles including pathogenic bacteria, fungi, and large viruses. These apparently unexpected observations, within the framework of the classical mechanisms for the formation of clathrin-coated vesicles, are now considered as examples of a new non-classical function of clathrin, which can promote the internalization of membrane domains associated to planar clathrin lattices. The role of actin downstream of clathrin seems to be critical for this still poorly characterized process. The historical frontier between endocytosis and phagocytosis is vanishing in the light of this new role for clathrin.


Asunto(s)
Clatrina/metabolismo , Endocitosis/fisiología , Actinas/metabolismo , Animales , Interacciones Huésped-Patógeno , Humanos , Listeria/fisiología , Modelos Biológicos , Fagocitosis/fisiología
18.
Toxins (Basel) ; 13(10)2021 10 09.
Artículo en Inglés | MEDLINE | ID: mdl-34679006

RESUMEN

Intracellular bacterial pathogens establish their replicative niches within membrane-encompassed compartments, called vacuoles. A subset of these bacteria uses a nanochannel called the type 4 secretion system (T4SS) to inject effector proteins that subvert the host cell machinery and drive the biogenesis of these compartments. These bacteria have also developed sophisticated ways of altering the innate immune sensing and response of their host cells, which allow them to cause long-lasting infections and chronic diseases. This review covers the mechanisms employed by intravacuolar pathogens to escape innate immune sensing and how Type 4-secreted bacterial effectors manipulate host cell mechanisms to allow the persistence of bacteria.


Asunto(s)
Bacterias/inmunología , Inmunidad Innata , Sistemas de Secreción Tipo IV , Infecciones Bacterianas , Fenómenos Fisiológicos Bacterianos , Interacciones Huésped-Patógeno , Vacuolas
19.
PLoS Negl Trop Dis ; 15(1): e0009008, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33406079

RESUMEN

Q fever is a widespread zoonotic disease caused by the intracellular bacterium Coxiella burnetii. While transmission is primarily but not exclusively airborne, ticks are usually thought to act as vectors on the basis of early microscopy studies. However, recent observations revealed that endosymbionts of ticks have been commonly misidentified as C. burnetii, calling the importance of tick-borne transmission into question. In this study, we re-evaluated the vector competence of the African soft tick Ornithodoros moubata for an avirulent strain of C. burnetii. To this end, we used an artificial feeding system to initiate infection of ticks, specific molecular tools to monitor further infections, and culture assays in axenic and cell media to check for the viability of C. burnetii excreted by ticks. We observed typical traits associated with vector competence: The exposure to an infected blood meal resulted in viable and persistent infections in ticks, trans-stadial transmissions of infection from nymphs to adults and the ability of adult ticks to transmit infectious C. burnetii. However, in contrast to early studies, we found that infection differed substantially between tick organs. In addition, while adult female ticks were infected, we did not observe C. burnetii in eggs, suggesting that transovarial transmission is not effective. Finally, we detected only a sporadic presence of C. burnetii DNA in tick faeces, but no living bacterium was further isolated in culture assays, suggesting that excretion in faeces is not a common mode of transmission in O. moubata.


Asunto(s)
Vectores Artrópodos/microbiología , Coxiella burnetii/aislamiento & purificación , Ornithodoros/microbiología , Fiebre Q/transmisión , Animales , Heces/microbiología , Femenino , Masculino
20.
Autophagy ; 17(3): 706-722, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-32116095

RESUMEN

Coxiella burnetii, the etiological agent of the zoonosis Q fever, replicates inside host cells within a large vacuole displaying autolysosomal characteristics. The development of this compartment is mediated by bacterial effectors, which interfere with a number of host membrane trafficking pathways. By screening a Coxiella transposon mutant library, we observed that transposon insertions in cbu0626 led to intracellular replication and vacuole biogenesis defects. Here, we demonstrate that CBU0626 is a novel member of the Coxiella vacuolar protein (Cvp) family of effector proteins, which is translocated by the Dot/Icm secretion system and localizes to vesicles with autolysosomal features as well as Coxiella-containing vacuoles (CCVs). We thus renamed this effector CvpF for Coxiella vacuolar protein F. CvpF specifically interacts with the host small GTPase RAB26, leading to the recruitment of the autophagosomal marker MAP1LC3B/LC3B (microtubule associated protein 1 light chain 3 beta) to CCVs. Importantly, cvpF::Tn mutants were highly attenuated compared to wild-type bacteria in the SCID mouse model of infection, highlighting the importance of CvpF for Coxiella virulence. These results suggest that CvpF manipulates endosomal trafficking and macroautophagy/autophagy induction for optimal C. burnetii vacuole biogenesis.Abbreviations: ACCM: acidified citrate cystein medium; AP: adaptor related protein complex; CCV: Coxiella-containing vacuole; Cvp: Coxiella vacuolar protein; GDI: guanosine nucleotide dissociation inhibitor; GDF: GDI dissociation factor; GEF: guanine exchange factor; LAMP1: lysosomal associated membrane protein 1; MAP1LC3B/LC3B: microtubule associated protein 1 light chain 3 beta; MTORC1: mechanistic target of rapamycin kinase MTOR complex 1; PBS: phosphate-buffered saline; PMA: phorbol myristate acetate; SQSTM1/p62: sequestosome 1; WT: wild-type.


Asunto(s)
Autofagia/fisiología , Sistemas de Secreción Bacterianos/metabolismo , Coxiella/metabolismo , Interacciones Huésped-Patógeno/inmunología , Vacuolas/microbiología , Animales , Proteínas Bacterianas/metabolismo , Coxiella burnetii/crecimiento & desarrollo , Coxiella burnetii/metabolismo , Humanos , Ratones , Vacuolas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA