Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Infect Immun ; 83(2): 661-70, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25422265

RESUMO

The intracellular bacterial pathogen Coxiella burnetii directs biogenesis of a parasitophorous vacuole (PV) that acquires host endolysosomal components. Formation of a PV that supports C. burnetii replication requires a Dot/Icm type 4B secretion system (T4BSS) that delivers bacterial effector proteins into the host cell cytosol. Thus, a subset of T4BSS effectors are presumed to direct PV biogenesis. Recently, the PV-localized effector protein CvpA was found to promote C. burnetii intracellular growth and PV expansion. We predict additional C. burnetii effectors localize to the PV membrane and regulate eukaryotic vesicle trafficking events that promote pathogen growth. To identify these vacuolar effector proteins, a list of predicted C. burnetii T4BSS substrates was compiled using bioinformatic criteria, such as the presence of eukaryote-like coiled-coil domains. Adenylate cyclase translocation assays revealed 13 proteins were secreted in a Dot/Icm-dependent fashion by C. burnetii during infection of human THP-1 macrophages. Four of the Dot/Icm substrates, termed Coxiella vacuolar protein B (CvpB), CvpC, CvpD, and CvpE, labeled the PV membrane and LAMP1-positive vesicles when ectopically expressed as fluorescently tagged fusion proteins. C. burnetii ΔcvpB, ΔcvpC, ΔcvpD, and ΔcvpE mutants exhibited significant defects in intracellular replication and PV formation. Genetic complementation of the ΔcvpD and ΔcvpE mutants rescued intracellular growth and PV generation, whereas the growth of C. burnetii ΔcvpB and ΔcvpC was rescued upon cohabitation with wild-type bacteria in a common PV. Collectively, these data indicate C. burnetii encodes multiple effector proteins that target the PV membrane and benefit pathogen replication in human macrophages.


Assuntos
Proteínas de Bactérias/metabolismo , Coxiella burnetii/metabolismo , Macrófagos/microbiologia , Transporte Proteico/genética , Vacúolos/metabolismo , Proteínas de Bactérias/biossíntese , Proteínas de Bactérias/genética , Sistemas de Secreção Bacterianos/fisiologia , Linhagem Celular Tumoral , Membrana Celular , Coxiella burnetii/genética , Citosol/metabolismo , Deleção de Genes , Células HeLa , Interações Hospedeiro-Patógeno , Humanos , Febre Q/microbiologia , Febre Q/patologia , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Vacúolos/genética , Vacúolos/microbiologia
2.
J Bacteriol ; 196(11): 1925-40, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24610709

RESUMO

Successful host cell colonization by the Q fever pathogen, Coxiella burnetii, requires translocation of effector proteins into the host cytosol by a Dot/Icm type 4B secretion system (T4BSS). In Legionella pneumophila, the two-component system (TCS) PmrAB regulates the Dot/Icm T4BSS and several additional physiological processes associated with pathogenesis. Because PmrA consensus regulatory elements are associated with some dot/icm and substrate genes, a similar role for PmrA in regulation of the C. burnetii T4BSS has been proposed. Here, we constructed a C. burnetii pmrA deletion mutant to directly probe PmrA-mediated gene regulation. Compared to wild-type bacteria, C. burnetii ΔpmrA exhibited severe intracellular growth defects that coincided with failed secretion of effector proteins. Luciferase gene reporter assays demonstrated PmrA-dependent expression of 5 of 7 dot/icm operons and 9 of 11 effector-encoding genes with a predicted upstream PmrA regulatory element. Mutational analysis verified consensus sequence nucleotides required for PmrA-directed transcription. RNA sequencing and whole bacterial cell mass spectrometry of wild-type C. burnetii and the ΔpmrA mutant uncovered new components of the PmrA regulon, including several genes lacking PmrA motifs that encoded Dot/Icm substrates. Collectively, our results indicate that the PmrAB TCS is a critical virulence factor that regulates C. burnetii Dot/Icm secretion. The presence of PmrA-responsive genes lacking PmrA regulatory elements also suggests that the PmrAB TCS controls expression of regulatory systems associated with the production of additional C. burnetii proteins involved in host cell parasitism.


Assuntos
Proteínas de Bactérias/metabolismo , Coxiella burnetii/fisiologia , Regulação Bacteriana da Expressão Gênica/fisiologia , Animais , Proteínas de Bactérias/genética , Linhagem Celular Tumoral , Chlorocebus aethiops , Coxiella burnetii/citologia , Deleção de Genes , Humanos , RNA Bacteriano , Regulon , Células Vero
3.
Proc Natl Acad Sci U S A ; 110(49): E4770-9, 2013 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-24248335

RESUMO

Successful macrophage colonization by Coxiella burnetii, the cause of human Q fever, requires pathogen-directed biogenesis of a large, growth-permissive parasitophorous vacuole (PV) with phagolysosomal characteristics. The vesicular trafficking pathways co-opted by C. burnetii for PV development are poorly defined; however, it is predicted that effector proteins delivered to the cytosol by a defective in organelle trafficking/intracellular multiplication (Dot/Icm) type 4B secretion system are required for membrane recruitment. Here, we describe involvement of clathrin-mediated vesicular trafficking in PV generation and the engagement of this pathway by the C. burnetii type 4B secretion system substrate Coxiella vacuolar protein A (CvpA). CvpA contains multiple dileucine [DERQ]XXXL[LI] and tyrosine (YXXΦ)-based endocytic sorting motifs like those recognized by the clathrin adaptor protein (AP) complexes AP1, AP2, and AP3. A C. burnetii ΔcvpA mutant exhibited significant defects in replication and PV development, confirming the importance of CvpA in infection. Ectopically expressed mCherry-CvpA localized to tubular and vesicular domains of pericentrosomal recycling endosomes positive for Rab11 and transferrin receptor, and CvpA membrane interactions were lost upon mutation of endocytic sorting motifs. Consistent with CvpA engagement of the endocytic recycling system, ectopic expression reduced uptake of transferrin. In pull-down assays, peptides containing CvpA-sorting motifs and full-length CvpA interacted with AP2 subunits and clathrin heavy chain. Furthermore, depletion of AP2 or clathrin by siRNA treatment significantly inhibited C. burnetii replication. Thus, our results reveal the importance of clathrin-coated vesicle trafficking in C. burnetii infection and define a role for CvpA in subverting these transport mechanisms.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Sistemas de Secreção Bacterianos/fisiologia , Vesículas Revestidas por Clatrina/fisiologia , Coxiella burnetii/crescimento & desenvolvimento , Proteínas de Membrana Transportadoras/metabolismo , Transdução de Sinais/fisiologia , Análise de Variância , Vesículas Revestidas por Clatrina/metabolismo , Clonagem Molecular , Biologia Computacional , Coxiella burnetii/metabolismo , Células HeLa , Humanos , Microscopia de Fluorescência , Transporte Proteico/fisiologia , RNA Interferente Pequeno/genética
4.
mBio ; 2(4): e00175-11, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21862628

RESUMO

Central to Q fever pathogenesis is replication of the causative agent, Coxiella burnetii, within a phagolysosome-like parasitophorous vacuole (PV) in mononuclear phagocytes. C. burnetii modulates PV biogenesis and other host cell functions, such as apoptotic signaling, presumably via the activity of proteins delivered to the host cytosol by a Dot/Icm type IVB secretion system (T4BSS). In this study, we utilized a C. burnetii strain carrying IcmD inactivated by the Himar1 transposon to investigate the requirements for Dot/Icm function in C. burnetii parasitism of human THP-1 macrophage-like cells. The icmD::Tn mutant failed to secrete characterized T4BSS substrates, a defect that correlated with deficient replication, PV development, and apoptosis protection. Restoration of type IVB secretion and intracellular growth of the icmD::Tn mutant required complementation with icmD, -J, and -B, indicating a polar effect of the transposon insertion on downstream dot/icm genes. Induction of icmDJB expression at 1 day postinfection resulted in C. burnetii replication and PV generation. Collectively, these data prove that T4BSS function is required for productive infection of human macrophages by C. burnetii. However, illustrating the metabolic flexibility of C. burnetti, the icmD::Tn mutant could replicate intracellularly when sequestered in a PV generated by wild-type bacteria, where Dot/Icm function is provided in trans, and within a phenotypically similar PV generated by the protozoan parasite Leishmania amazonensis, where host cells are devoid of Dot/Icm T4BSS effector proteins.


Assuntos
Proteínas de Bactérias/metabolismo , Sistemas de Secreção Bacterianos , Coxiella burnetii/metabolismo , Macrófagos/microbiologia , Febre Q/microbiologia , Animais , Proteínas de Bactérias/genética , Linhagem Celular , Coxiella burnetii/genética , Coxiella burnetii/crescimento & desenvolvimento , Humanos , Macrófagos/metabolismo , Transporte Proteico , Febre Q/metabolismo , Vacúolos/metabolismo , Vacúolos/microbiologia
5.
Appl Environ Microbiol ; 77(11): 3720-5, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21478315

RESUMO

We recently described acidified citrate cysteine medium (ACCM), which supports host cell-free (axenic) growth of Coxiella burnetii. After 6 days of incubation, greater than 3 logs of growth was achieved with the avirulent Nine Mile phase II (NMII) strain. Here, we describe modified ACCM and culture conditions that support improved growth of C. burnetii and their use in genetic transformation and pathogen isolation from tissue samples. ACCM was modified by replacing fetal bovine serum with methyl-ß-cyclodextrin to generate ACCM-2. Cultivation of NMII in ACCM-2 with moderate shaking and in 2.5% oxygen yielded 4 to 5 logs of growth over 7 days. Similar growth was achieved with the virulent Nine Mile phase I and G isolates of C. burnetii. Colonies that developed after 6 days of growth in ACCM-2 agarose were approximately 0.5 mm in diameter, roughly 5-fold larger than those formed in ACCM agarose. By electron microscopy, colonies consisted primarily of the C. burnetii small cell variant morphological form. NMII was successfully cultured in ACCM-2 when medium was inoculated with as little as 10 genome equivalents contained in tissue homogenates from infected SCID mice. A completely axenic C. burnetii genetic transformation system was developed using ACCM-2 that allowed isolation of transformants in about 2 1/2 weeks. Transformation experiments demonstrated clonal populations in colonies and a transformation frequency of approximately 5 × 10(-5). Cultivation in ACCM-2 will accelerate development of C. burnetii genetic tools and provide a sensitive means of primary isolation of the pathogen from Q fever patients.


Assuntos
Técnicas Bacteriológicas/métodos , Coxiella burnetii/genética , Coxiella burnetii/isolamento & purificação , Meios de Cultura/química , Genética Microbiana/métodos , Transformação Bacteriana , Animais , Coxiella burnetii/crescimento & desenvolvimento , Coxiella burnetii/ultraestrutura , Camundongos , Microscopia Eletrônica , Fatores de Tempo
6.
J Bacteriol ; 193(7): 1493-503, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21216993

RESUMO

The intracellular bacterial pathogen Coxiella burnetii directs biogenesis of a phagolysosome-like parasitophorous vacuole (PV), in which it replicates. The organism encodes a Dot/Icm type IV secretion system (T4SS) predicted to deliver to the host cytosol effector proteins that mediate PV formation and other cellular events. All C. burnetii isolates carry a large, autonomously replicating plasmid or have chromosomally integrated plasmid-like sequences (IPS), suggesting that plasmid and IPS genes are critical for infection. Bioinformatic analyses revealed two candidate Dot/Icm substrates with eukaryotic-like motifs uniquely encoded by the QpH1 plasmid from the Nine Mile reference isolate. CpeC, containing an F-box domain, and CpeD, possessing kinesin-related and coiled-coil regions, were secreted by the closely related Legionella pneumophila Dot/Icm T4SS. An additional QpH1-specific gene, cpeE, situated in a predicted operon with cpeD, also encoded a secreted effector. Further screening revealed that three hypothetical proteins (CpeA, CpeB, and CpeF) encoded by all C. burnetii plasmids and IPS are Dot/Icm substrates. By use of new genetic tools, secretion of plasmid effectors by C. burnetii during host cell infection was confirmed using ß-lactamase and adenylate cyclase translocation assays, and a C-terminal secretion signal was identified. When ectopically expressed in HeLa cells, plasmid effectors trafficked to different subcellular sites, including autophagosomes (CpeB), ubiquitin-rich compartments (CpeC), and the endoplasmic reticulum (CpeD). Collectively, these results suggest that C. burnetii plasmid-encoded T4SS substrates play important roles in subversion of host cell functions, providing a plausible explanation for the absolute maintenance of plasmid genes by this pathogen.


Assuntos
Proteínas de Bactérias/metabolismo , Sistemas de Secreção Bacterianos/fisiologia , Coxiella burnetii/metabolismo , Regulação Bacteriana da Expressão Gênica/fisiologia , Plasmídeos/genética , Proteínas de Bactérias/genética , Sistemas de Secreção Bacterianos/genética , Sequência de Bases , Linhagem Celular , Coxiella burnetii/genética , DNA Bacteriano/genética , Células HeLa , Humanos , Transporte Proteico
7.
Microb Pathog ; 49(5): 273-84, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20600793

RESUMO

Anaplasma phagocytophilum infects neutrophils and myeloid, endothelial, and tick cell lines to reside within a host cell-derived vacuole that is indispensible for its survival. Here, we identify APH_0032 as an Anaplasma-derived protein that associates with the A. phagocytophilum-occupied vacuolar membrane (AVM). APH_0032 is a 66.1 kDa acidic protein that electrophoretically migrates with an apparent molecular weight of 130 kDa. It contains a predicted transmembrane domain and tandemly arranged direct repeats that comprise 46% of the protein. APH_0032 is undetectable on Anaplasma organisms bound to the surfaces of HL-60 cells, but is detected on the AVM and surfaces of intravacuolar bacteria beginning 24 h post-infection. APH_0032 localizes to the AVM in HL-60, THP-1, HMEC-1, and ISE6 cells. APH_0032, along with APH_1387, which encodes a confirmed AVM protein, is transcribed during A. phagocytophilum infection of tick salivary glands and murine neutrophils. APH_0032 localizes to the AVM in neutrophils recovered from infected mice. The Legionella pneumophila Dot/IcM type IV secretion system (T4SS) can heterologously secrete a CyaA-tagged version of the A. phagocytophilum VirB/D T4SS effector, AnkA, but fails to secrete CyaA-tagged APH_0032 or APH_1387. These data confirm APH_0032 as an Anaplasma-derived AVM protein and hint that neither it nor APH_1387 are T4SS effectors.


Assuntos
Anaplasma phagocytophilum/patogenicidade , Proteínas de Bactérias/metabolismo , Regulação Bacteriana da Expressão Gênica , Interações Hospedeiro-Patógeno , Membranas Intracelulares/química , Vacúolos/química , Vacúolos/microbiologia , Sequência de Aminoácidos , Animais , Proteínas de Bactérias/química , Células Cultivadas , Perfilação da Expressão Gênica , Humanos , Legionella pneumophila/genética , Legionella pneumophila/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Camundongos , Dados de Sequência Molecular , Peso Molecular , Neutrófilos/microbiologia , Estrutura Terciária de Proteína , Sequências de Repetição em Tandem , Carrapatos , Transcrição Gênica
8.
Infect Immun ; 78(8): 3465-74, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20515926

RESUMO

Coxiella burnetii infects mononuclear phagocytes, where it directs biogenesis of a vacuolar niche termed the parasitophorous vacuole (PV). Owing to its lumenal pH (approximately 5) and fusion with endolysosomal vesicles, the PV is considered phagolysosome-like. However, the degradative properties of the mature PV are unknown, and there are conflicting reports on the maturation state and growth permissiveness of PV harboring virulent phase I or avirulent phase II C. burnetii variants in human mononuclear phagocytes. Here, we employed infection of primary human monocyte-derived macrophages (HMDMs) and THP-1 cells as host cells to directly compare the PV maturation kinetics and pathogen growth in cells infected with the Nine Mile phase I variant (NMI) or phase II variant (NMII) of C. burnetii. In both cell types, phase variants replicated with similar kinetics, achieving roughly 2 to 3 log units of growth before they reached stationary phase. HMDMs infected by either phase variant secreted similar amounts of the proinflammatory cytokines interleukin-6 and tumor necrosis factor alpha. In infected THP-1 cells, equal percentages of NMI and NMII PVs decorate with the early endosomal marker Rab5, the late endosomal/lysosomal markers Rab7 and CD63, and the lysosomal marker cathepsin D at early (8 h) and late (72 h) time points postinfection (p.i.). Mature PVs (2 to 4 days p.i.) harboring NMI or NMII contained proteolytically active cathepsins and quickly degraded Escherichia coli. These data suggest that C. burnetii does not actively inhibit phagolysosome function as a survival mechanism. Instead, NMI and NMII resist degradation to replicate in indistinguishable digestive PVs that fully mature through the endolysosomal pathway.


Assuntos
Coxiella burnetii/patogenicidade , Macrófagos/microbiologia , Fagossomos/microbiologia , Catepsinas/metabolismo , Células Cultivadas , Coxiella burnetii/imunologia , Escherichia coli/imunologia , Humanos , Interleucina-6/metabolismo , Macrófagos/imunologia , Fagossomos/imunologia , Fator de Necrose Tumoral alfa/metabolismo
9.
J Bacteriol ; 191(13): 4232-42, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19411324

RESUMO

Coxiella burnetii is an obligate intracellular bacterium that directs biogenesis of a parasitophorous vacuole (PV) for replication. Effectors of PV maturation are likely translocated into the host cytosol by a type IV secretion system (T4SS) with homology to the Dot/Icm apparatus of Legionella pneumophila. Since secreted bacterial virulence factors often functionally mimic the activities of host proteins, prokaryotic proteins with eukaryotic features are considered candidate T4SS substrates. Genes encoding proteins with eukaryotic-type ankyrin repeat domains (Anks) were identified upon genome sequencing of the C. burnetii Nine Mile reference isolate, which is associated with a case of human acute Q fever. Interestingly, recent genome sequencing of the G and K isolates, derived from human chronic endocarditis patients, and of the Dugway rodent isolate revealed remarkable heterogeneity in the Ank gene family, with the Dugway isolate harboring the largest number of full-length Ank genes. Using L. pneumophila as a surrogate host, we identified 10 Dugway Anks and 1 Ank specific to the G and K endocarditis isolates translocated into the host cytosol in a Dot/Icm-dependent fashion. A 10-amino-acid C-terminal region appeared to be necessary for translocation, with some Anks also requiring the chaperone IcmS for secretion. Ectopically expressed Anks localized to a variety of subcellular regions in mammalian cells, including microtubules, mitochondria, and the PV membrane. Collectively, these data suggest that C. burnetii isolates translocate distinct subsets of the Ank protein family into the host cytosol, where they modulate diverse functions, some of which may be unique to C. burnetii pathotypes.


Assuntos
Repetição de Anquirina/fisiologia , Proteínas de Bactérias/genética , Proteínas de Bactérias/fisiologia , Transporte Biológico/fisiologia , Proteínas de Transporte/fisiologia , Coxiella burnetii , Proteínas de Membrana/fisiologia , Chaperonas Moleculares/fisiologia , Repetição de Anquirina/genética , Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Transporte Biológico/genética , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Linhagem Celular , Coxiella burnetii/genética , Coxiella burnetii/metabolismo , AMP Cíclico/metabolismo , Citosol/metabolismo , Células HeLa , Humanos , Immunoblotting , Legionella pneumophila/genética , Legionella pneumophila/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Microscopia de Fluorescência , Chaperonas Moleculares/genética
10.
Proc Natl Acad Sci U S A ; 106(11): 4430-4, 2009 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-19246385

RESUMO

The inability to propagate obligate intracellular pathogens under axenic (host cell-free) culture conditions imposes severe experimental constraints that have negatively impacted progress in understanding pathogen virulence and disease mechanisms. Coxiella burnetii, the causative agent of human Q (Query) fever, is an obligate intracellular bacterial pathogen that replicates exclusively in an acidified, lysosome-like vacuole. To define conditions that support C. burnetii growth, we systematically evaluated the organism's metabolic requirements using expression microarrays, genomic reconstruction, and metabolite typing. This led to development of a complex nutrient medium that supported substantial growth (approximately 3 log(10)) of C. burnetii in a 2.5% oxygen environment. Importantly, axenically grown C. burnetii were highly infectious for Vero cells and exhibited developmental forms characteristic of in vivo grown organisms. Axenic cultivation of C. burnetii will facilitate studies of the organism's pathogenesis and genetics and aid development of Q fever preventatives such as an effective subunit vaccine. Furthermore, the systematic approach used here may be broadly applicable to development of axenic media that support growth of other medically important obligate intracellular pathogens.


Assuntos
Coxiella burnetii/crescimento & desenvolvimento , Febre Q/microbiologia , Animais , Técnicas Bacteriológicas , Sistema Livre de Células , Chlorocebus aethiops , Coxiella burnetii/metabolismo , Meios de Cultura , Genômica , Metabolômica , Células Vero
11.
J Bacteriol ; 191(5): 1369-81, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19114492

RESUMO

Coxiella burnetii is a gram-negative obligate intracellular bacterium and the causative agent of human Q fever. The lack of methods to genetically manipulate C. burnetii significantly impedes the study of this organism. We describe here the cloning and characterization of a C. burnetii ftsZ mutant generated by mariner-based Himar1 transposon (Tn) mutagenesis. C. burnetii was coelectroporated with a plasmid encoding the Himar1 C9 transposase variant and a plasmid containing a Himar1 transposon encoding chloramphenicol acetyltransferase, mCherry fluorescent protein, and a ColE1 origin of replication. Vero cells were infected with electroporated C. burnetii and transformants scored as organisms replicating in the presence of chloramphenicol and expressing mCherry. Southern blot analysis revealed multiple transpositions in the C. burnetii genome and rescue cloning identified 30 and 5 insertions in coding and noncoding regions, respectively. Using micromanipulation, a C. burnetii clone was isolated containing a Tn insertion within the C terminus of the cell division gene ftsZ. The ftsZ mutant had a significantly lower growth rate than wild-type bacteria and frequently appeared as filamentous forms displaying incomplete cell division septa. The latter phenotype correlated with a deficiency in generating infectious foci on a per-genome basis compared to wild-type organisms. The mutant FtsZ protein was also unable to bind the essential cell division protein FtsA. This is the first description of C. burnetii harboring a defined gene mutation generated by genetic transformation.


Assuntos
Proteínas de Bactérias , Coxiella burnetii/genética , Proteínas do Citoesqueleto , Elementos de DNA Transponíveis , Mutagênese Insercional/métodos , Mutação , Sequência de Aminoácidos , Animais , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Chlorocebus aethiops , Clonagem Molecular , Coxiella burnetii/crescimento & desenvolvimento , Coxiella burnetii/patogenicidade , Coxiella burnetii/ultraestrutura , Proteínas do Citoesqueleto/química , Proteínas do Citoesqueleto/genética , Proteínas do Citoesqueleto/metabolismo , Eletroporação , Humanos , Microscopia Eletrônica de Varredura , Dados de Sequência Molecular , Plasmídeos/genética , Transformação Bacteriana , Transposases/genética , Transposases/metabolismo , Células Vero
12.
Methods Mol Biol ; 445: 389-406, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18425464

RESUMO

Coxiella burnetii is a bacterial obligate intracellular pathogen that replicates within a spacious parasitophorous vacuole (PV) with lysosomal characteristics. The pathogen actively participates in the biogenesis of its PV by synthesizing proteins that mediate vesicular interactions. Both C. burnetii and host factors that regulate PV formation are likely localized to the PV membrane, and their identification would be aided by an efficient method for isolating the C. burnetii vacuole. To this end, we developed a method to separate intact PV from host cell material that relies on fusion of the vacuole with latex bead-containing phagosomes (LBP). Sequestration of latex beads by the C. burnetii PV increases the vacuole's buoyant density and facilitates its fractionation on a sucrose step gradient. Transmission electron microscopy confirms the isolation of intact PV-containing latex beads from infected MH-S murine alveolar macrophage-like cells. Immunoblotting demonstrates that C. burnetii PV lysates are dramatically enriched for the late endosome/lysosome markers LAMP-1 and LAMP-2 when compared to total host cell lysates. Conversely, PV preparations are devoid of p62 and GM130, markers of the nucleus and Golgi apparatus, respectively, indicating effective separation of the vacuole from these host cell compartments. Two-dimensional gel electrophoresis and immunoblotting reveal distinct protein differences between C. burnetii PV and LBP. Identification of proteins unique to the PV membrane will yield important insight into C. burnetii-host interactions.


Assuntos
Coxiella burnetii/crescimento & desenvolvimento , Vacúolos/microbiologia , Animais , Coxiella burnetii/metabolismo , Eletroforese em Gel de Poliacrilamida , Immunoblotting , Focalização Isoelétrica , Lisossomos/metabolismo , Lisossomos/microbiologia , Lisossomos/ultraestrutura , Macrófagos Alveolares/microbiologia , Macrófagos Alveolares/ultraestrutura , Camundongos , Microscopia Eletrônica de Transmissão , Fagossomos/metabolismo , Fagossomos/microbiologia , Fagossomos/ultraestrutura , Proteômica/métodos , Febre Q/microbiologia , Vacúolos/ultraestrutura
13.
J Microbiol Methods ; 72(3): 321-5, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18242746

RESUMO

Purification of the obligate intracellular bacterium Coxiella burnetii requires physical disruption of infected cells. Here we describe a gentle and safe digitonin lysis procedure to release C. burnetii from infected cells. The purity, yield, and infectivity of digitonin-prepped organisms are comparable to that of organisms purified using cell lysis by sonication.


Assuntos
Técnicas Bacteriológicas/métodos , Coxiella burnetii/isolamento & purificação , Digitonina/metabolismo , Animais , Membrana Celular/efeitos dos fármacos , Chlorocebus aethiops , Coxiella burnetii/citologia , Coxiella burnetii/ultraestrutura , Microscopia , Microscopia Eletrônica de Transmissão , Células Vero
14.
Infect Immun ; 75(9): 4263-71, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17606599

RESUMO

Coxiella burnetii, the cause of human Q fever, is an aerosol-borne, obligate intracellular bacterium that targets host alveolar mononuclear phagocytic cells during infection. In all cell types examined, C. burnetii establishes a replicative niche in a lysosome-like parasitophorous vacuole where it carries out a lengthy infectious cycle with minimal cytopathic effects. The persistent and mild nature of C. burnetii infection in vitro suggests that the pathogen modulates apoptosis to sustain the host cell. In the current study, we examined the ability of C. burnetii to inhibit apoptotic cell death during infection of human THP-1 monocyte-derived macrophages and primary monkey alveolar macrophages. C. burnetii-infected cells demonstrated significant protection from death relative to uninfected cells following treatment with staurosporine, a potent inducer of intrinsic apoptosis. This protection correlated with reduced cleavage of caspase-9, caspase-3, and poly(ADP-ribose) polymerase (PARP), all proteolytic events that occur during apoptosis. Reduced PARP cleavage was also observed in cells treated with tumor necrosis factor alpha to induce extrinsic apoptosis. Apoptosis inhibition was a C. burnetii-driven process as infected cells treated with rifampin or chloramphenicol, inhibitors of bacterial RNA and protein synthesis, respectively, showed significantly reduced protection against staurosporine-induced apoptosis. C. burnetii infection affected the expression of multiple apoptosis-related genes and resulted in increased synthesis of the antiapoptotic proteins A1/Bfl-1 and c-IAP2. Collectively, these data suggest that C. burnetii modulates apoptotic pathways to inhibit host cell death, thus providing a stable, intracellular niche for the course of the pathogen's infectious cycle.


Assuntos
Apoptose/fisiologia , Coxiella burnetii/fisiologia , Macrófagos Alveolares/citologia , Macrófagos Alveolares/microbiologia , Animais , Apoptose/efeitos dos fármacos , Inibidores de Caspase , Caspases/metabolismo , Linhagem Celular Tumoral , Chlorocebus aethiops , Humanos , Macrófagos Alveolares/enzimologia , Monócitos/citologia , Monócitos/efeitos dos fármacos , Monócitos/enzimologia , Monócitos/microbiologia , Estaurosporina/farmacologia , Células Vero
15.
Appl Environ Microbiol ; 73(12): 4048-54, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17468273

RESUMO

Coxiella burnetii is an obligate intracellular bacterium that replicates in a large lysosome-like parasitophorous vacuole (PV). Current methods of cloning C. burnetii are laborious and technically demanding. We have developed an alternative cloning method that involves excision of individual C. burnetii-laden PVs from infected cell monolayers by micromanipulation. To demonstrate the cloning utility and efficiency of this procedure, we coinfected Vero cells with isogenic variants of the Nine Mile strain of C. burnetii. Coinhabited PVs harboring Nine Mile phase II (NMII) and Nine Mile phase I (NMI) or Nine Mile crazy (NMC) were demonstrated by immunofluorescence. PVs were then randomly excised from cells coinfected with NMI and NMC by micromanipulation, and PVs harboring both strains were identified by PCR. Fresh Vero cells were subsequently infected with organisms from coinhabited PVs, and the PV excision and PCR screening process was repeated. Without exception, PVs obtained from second-round excisions contained clonal populations of either NMII or NMC, demonstrating that micromanipulation is an efficient and reproducible procedure for obtaining C. burnetii clones.


Assuntos
Técnicas de Cultura de Células/métodos , Coxiella burnetii/crescimento & desenvolvimento , Vacúolos/microbiologia , Animais , Chlorocebus aethiops , Células Clonais , Coxiella burnetii/genética , Primers do DNA/genética , Técnica Indireta de Fluorescência para Anticorpo , Immunoblotting , Micromanipulação , Reação em Cadeia da Polimerase , Células Vero
16.
Traffic ; 8(3): 212-25, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17233756

RESUMO

Following invasion of non-phagocytic host cells, Salmonella enterica survives and replicates within a phagosome-like compartment known as the Salmonella-containing vacuole (SCV). It is now well established that SCV biogenesis, like phagosome biogenesis, involves sequential interactions with the endocytic pathway. However, Salmonella is believed to limit these interactions and, in particular, to avoid fusion of terminal lysosomes with the SCV. In this study, we reassessed this process using a high-resolution live-cell imaging approach and found an unanticipated level of interaction between the SCV and the endocytic pathway. Direct interactions, in which late endosomal/lysosomal content was transferred to SCVs, were detected within 30 min of invasion and continued for several hours. Mechanistically, these interactions were very similar to phagosome-lysosome fusion because they were accompanied by rapid acidification of the SCV, could be blocked by chemical perturbation of microtubules or vacuolar acidification and involved the smallGTPase Rab7. In comparison with vacuoles containing internalized Escherichia coli or heat-killed Salmonella, SCVs did show some delay of fusion and acidification, although, this appeared to be independent of either type III secretion system. These results provide compelling evidence that inhibition of SCV-lysosome fusion is not the major determinant in establishment of the Salmonella replicative niche in epithelial cells.


Assuntos
Endossomos/metabolismo , Células Epiteliais/microbiologia , Lisossomos/metabolismo , Salmonella typhimurium/metabolismo , Vacúolos/microbiologia , Biomarcadores , Linhagem Celular Tumoral , Endocitose , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Células HeLa , Humanos , Fagossomos/metabolismo , Proteínas rab de Ligação ao GTP/metabolismo , proteínas de unión al GTP Rab7
17.
Infect Immun ; 75(1): 290-8, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17088354

RESUMO

A biphasic developmental cycle whereby highly resistant small-cell variants (SCVs) are generated from large-cell variants (LCVs) is considered fundamental to the virulence of Coxiella burnetii, the causative agent of human Q fever. In this study a proteome analysis of C. burnetii developmental forms was conducted to provide insight into their unique biological and immunological properties. Silver-stained gels of SCV and LCV lysates separated by two-dimensional (2-D) gel electrophoresis resolved over 675 proteins in both developmental forms. Forty-eight proteins were greater than twofold more abundant in LCVs than in SCVs, with six proteins greater than twofold more abundant in SCVs than in LCVs. Four and 15 upregulated proteins of SCVs and LCVs, respectively, were identified by mass spectrometry, and their predicted functional roles are consistent with a metabolically active LCV and a structurally resistant SCV. One-dimensional and 2-D immunoblots of cell form lysates probed with sera from infected/vaccinated guinea pigs and convalescent-phase serum from human patients who had recovered from acute Q fever, respectively, revealed both unique SCV/LCV antigens and common SCV/LCV antigens that were often differentially synthesized. Antigens recognized during human infection were identified by mass spectroscopy and included both previously described immunodominant proteins of C. burnetii and novel immunogenic proteins that may be important in the pathophysiology of clinical Q fever and/or the induction of protective immunity.


Assuntos
Antígenos de Bactérias/análise , Proteínas de Bactérias/análise , Coxiella burnetii/fisiologia , Animais , Antígenos de Bactérias/imunologia , Proteínas de Bactérias/imunologia , Ensaio Cometa , Eletroforese em Gel Bidimensional , Cobaias , Humanos , Immunoblotting , Epitopos Imunodominantes/imunologia , Espectrometria de Massas , Microscopia Eletrônica de Transmissão , Proteoma
18.
J Bacteriol ; 188(7): 2309-24, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16547017

RESUMO

Coxiella burnetii, a gram-negative obligate intracellular bacterium, causes human Q fever and is considered a potential agent of bioterrorism. Distinct genomic groups of C. burnetii are revealed by restriction fragment-length polymorphisms (RFLP). Here we comprehensively define the genetic diversity of C. burnetii by hybridizing the genomes of 20 RFLP-grouped and four ungrouped isolates from disparate sources to a high-density custom Affymetrix GeneChip containing all open reading frames (ORFs) of the Nine Mile phase I (NMI) reference isolate. We confirmed the relatedness of RFLP-grouped isolates and showed that two ungrouped isolates represent distinct genomic groups. Isolates contained up to 20 genomic polymorphisms consisting of 1 to 18 ORFs each. These were mostly complete ORF deletions, although partial deletions, point mutations, and insertions were also identified. A total of 139 chromosomal and plasmid ORFs were polymorphic among all C. burnetii isolates, representing ca. 7% of the NMI coding capacity. Approximately 67% of all deleted ORFs were hypothetical, while 9% were annotated in NMI as nonfunctional (e.g., frameshifted). The remaining deleted ORFs were associated with diverse cellular functions. The only deletions associated with isogenic NMI variants of attenuated virulence were previously described large deletions containing genes involved in lipopolysaccharide (LPS) biosynthesis, suggesting that these polymorphisms alone are responsible for the lower virulence of these variants. Interestingly, a variant of the Australia QD isolate producing truncated LPS had no detectable deletions, indicating LPS truncation can occur via small genetic changes. Our results provide new insight into the genetic diversity and virulence potential of Coxiella species.


Assuntos
Coxiella burnetii/genética , Variação Genética , Genoma Bacteriano , Análise de Sequência com Séries de Oligonucleotídeos , Cromossomos Bacterianos/genética , DNA Bacteriano/genética , Regulação Bacteriana da Expressão Gênica , Humanos , Lipopolissacarídeos , Fases de Leitura Aberta , Filogenia , Plasmídeos/genética , Polimorfismo Genético , Febre Q/microbiologia
19.
Cell Microbiol ; 8(3): 496-507, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16469060

RESUMO

Coxiella burnetii directs the synthesis of a large parasitophorous vacuole (PV) required for replication. While some lysosomal characteristics of the PV have been described, the origin and composition of the PV membrane is largely undefined. Cholesterol is an essential component of mammalian cell membranes where it plays important regulatory and structural roles. Here we investigated the role of host cholesterol in biogenesis and maintenance of the C. burnetii PV in Vero cells. The C. burnetii PV membrane stained with filipin and was positive for the lipid raft protein flotillin-1, suggesting PV membranes are enriched in cholesterol and contain lipid raft microdomains. C. burnetii infection increased host cell cholesterol content by 1.75-fold with a coincident upregulation of host genes involved in cholesterol metabolism. Treatment with U18666A, lovastatin, or 25-hydroxycholesterol, pharmacological agents that inhibit cholesterol uptake and/or biosynthesis, altered PV morphology and partially inhibited C. burnetii replication. Complete inhibition of C. burnetii PV development and replication was observed when infected cells were treated with imipramine or ketoconazole, inhibitors of cholesterol uptake and biosynthesis respectively. We conclude that C. burnetii infection perturbs host cell cholesterol metabolism and that free access to host cholesterol stores is required for optimal C. burnetii replication.


Assuntos
Colesterol/metabolismo , Coxiella burnetii/crescimento & desenvolvimento , Coxiella burnetii/patogenicidade , Vacúolos/microbiologia , Animais , Chlorocebus aethiops , Coxiella burnetii/efeitos dos fármacos , Imipramina/farmacologia , Membranas Intracelulares/metabolismo , Cetoconazol/farmacologia , Lisossomos/microbiologia , Vacúolos/metabolismo , Células Vero
20.
Infect Immun ; 73(8): 4494-504, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16040960

RESUMO

Legionella pneumophila and Coxiella burnetii are phylogenetically related intracellular bacteria that cause aerosol-transmitted lung infections. In host cells both pathogens proliferate in vacuoles whose biogenesis displays some common features. To test the functional similarity of their respective intracellular niches, African green monkey kidney epithelial (Vero) cells, A/J mouse bone marrow-derived macrophages, human macrophages, and human dendritic cells (DC) containing mature C. burnetii replication vacuoles were superinfected with L. pneumophila, and then the acidity, lysosome-associated membrane protein (LAMP) content, and cohabitation of mature replication vacuoles was assessed. In all cell types, wild-type L. pneumophila occupied distinct vacuoles in close association with acidic, LAMP-positive C. burnetii replication vacuoles. In murine macrophages, but not primate macrophages, DC, or epithelial cells, L. pneumophila replication vacuoles were acidic and LAMP positive. Unlike wild-type L. pneumophila, type IV secretion-deficient dotA mutants trafficked to lysosome-like C. burnetii vacuoles in Vero cells where they survived but failed to replicate. In primate macrophages, DC, or epithelial cells, growth of L. pneumophila was as robust in superinfected cell cultures as in those singly infected. Thus, despite their noted similarities, L. pneumophila and C. burnetii are exquisitely adapted for replication in unique replication vacuoles, and factors that maintain the C. burnetii replication vacuole do not alter biogenesis of an adjacent L. pneumophila replication vacuole. Moreover, L. pneumophila can replicate efficiently in either lysosomal vacuoles of A/J mouse cells or in nonlysosomal vacuoles of primate cells.


Assuntos
Coxiella burnetii/metabolismo , Legionella pneumophila/metabolismo , Doença dos Legionários/microbiologia , Vacúolos/microbiologia , Animais , Antígenos de Bactérias , Proliferação de Células , Chlorocebus aethiops , Coxiella burnetii/patogenicidade , Células Dendríticas/microbiologia , Regulação Bacteriana da Expressão Gênica , Humanos , Legionella pneumophila/patogenicidade , Macrófagos/microbiologia , Camundongos , Febre Q/microbiologia , Células Vero , Virulência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...