Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Bacteriol ; 201(8)2019 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-30745369

RESUMEN

Coxiella burnetii, the etiological agent of Q fever, undergoes a unique biphasic developmental cycle where bacteria transition from a replicating (exponential-phase) large cell variant (LCV) form to a nonreplicating (stationary-phase) small cell variant (SCV) form. The alternative sigma factor RpoS is an essential regulator of stress responses and stationary-phase physiology in several bacterial species, including Legionella pneumophila, which has a developmental cycle superficially similar to that of C. burnetii Here, we used a C. burnetii ΔrpoS mutant to define the role of RpoS in intracellular growth and SCV development. Growth yields following infection of Vero epithelial cells or THP-1 macrophage-like cells with the rpoS mutant in the SCV form, but not the LCV form, were significantly lower than that of wild-type bacteria. RNA sequencing and whole-cell mass spectrometry of the C. burnetii ΔrpoS mutant revealed that a substantial portion of the C. burnetii genome is regulated by RpoS during SCV development. Regulated genes include those involved in stress responses, arginine transport, peptidoglycan remodeling, and synthesis of the SCV-specific protein ScvA. Genes comprising the dot/icm locus, responsible for production of the Dot/Icm type 4B secretion system, were also dysregulated in the rpoS mutant. These data were corroborated with independent assays demonstrating that the C. burnetii ΔrpoS strain has increased sensitivity to hydrogen peroxide and carbenicillin and a thinner cell wall/outer membrane complex. Collectively, these results demonstrate that RpoS is an important regulator of genes involved in C. burnetii SCV development and intracellular growth.IMPORTANCE The Q fever bacterium Coxiella burnetii has spore-like environmental stability, a characteristic that contributes to its designation as a potential bioweapon. Stability is likely conferred by a highly resistant, small cell variant (SCV) stationary-phase form that arises during a biphasic developmental cycle. Here, we define the role of the alternative sigma factor RpoS in regulating genes associated with SCV development. Genes involved in stress responses, amino acid transport, cell wall remodeling, and type 4B effector secretion were dysregulated in the rpoS mutant. Cellular impairments included defects in intracellular growth, cell wall structure, and resistance to oxidants. These results support RpoS as a central regulator of the Coxiella developmental cycle and identify developmentally regulated genes involved in morphological differentiation.


Asunto(s)
Proteínas Bacterianas/metabolismo , Coxiella burnetii/citología , Coxiella burnetii/crecimiento & desarrollo , Regulación Bacteriana de la Expresión Génica , Factor sigma/metabolismo , Animales , Chlorocebus aethiops , Coxiella burnetii/genética , Citoplasma/microbiología , Células Epiteliales/microbiología , Eliminación de Gen , Perfilación de la Expresión Génica , Humanos , Macrófagos/microbiología , Proteómica , Factor sigma/deficiencia , Células THP-1 , Células Vero
2.
Acta Virol ; 62(4): 409-414, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30472871

RESUMEN

Coxiella burnetii is an intracellular pathogenic bacterium and etiological agent of Q fever in humans. Recently, the bacterium has been set free from the strictly intracellular condition by successful cultivation in acidified citrate cysteine medium. Here, we report a bacterial cell counting method that allows rapid quantification of the absolute or relative number of live cells of C. burnetii in a high throughput manner. The method utilizes TaqMan-based quantitative polymerase chain reaction (qPCR) targeting a single dotA gene for determination of genome equivalent (GE) presented either as DNA or complementary DNA (cDNA) synthesized via reverse transcription. The assay was shown to be specific, sensitive and efficiently reproducible. The quantification was linear over a range of 30 to 3x108 copies. Since there is only one copy of the dotA gene per Coxiella chromosome, the calculated dotA copy numbers can be compared to the number of bacterial cells. Finally, we demonstrated the potential of the method to assess effects of antibiotic on cell viability and to determine the antibiotic-tolerant fraction within a cell population. Keywords: Coxiella burnetii; Q fever; real-time polymerase chain reaction; copy number; antibiotic; axenic media; dotA gene.


Asunto(s)
Coxiella burnetii , Fiebre Q , Reacción en Cadena en Tiempo Real de la Polimerasa , Coxiella burnetii/citología , Coxiella burnetii/genética , Humanos , Plásmidos , Fiebre Q/microbiología
3.
Artículo en Inglés | MEDLINE | ID: mdl-28293541

RESUMEN

Coxiella burnetii is an obligate intracellular pathogen and the causative agent of human Q fever. Replication of the bacterium within a large parasitophorous vacuole (PV) resembling a host phagolysosome is required for pathogenesis. PV biogenesis is a pathogen driven process that requires engagement of several host cell vesicular trafficking pathways to acquire vacuole components. The goal of this study was to determine if infection by C. burnetii modulates endolysosomal flux to potentially benefit PV formation. HeLa cells, infected with C. burnetii or left uninfected, were incubated with fluorescent transferrin (Tf) for 0-30 min, and the amount of Tf internalized by cells quantitated by high-content imaging. At 3 and 5 days, but not 1 day post-infection, the maximal amounts of fluorescent Tf internalized by infected cells were significantly greater than uninfected cells. The rates of Tf uptake and recycling were the same for infected and uninfected cells; however, residual Tf persisted in EEA.1 positive compartments adjacent to large PV after 30 min of recycling in the absence of labeled Tf. On average, C. burnetii-infected cells contained significantly more CD63-positive endosomes than uninfected cells. In contrast, cells containing large vacuoles generated by Chlamydia trachomatis exhibited increased rates of Tf internalization without increased CD63 expression. Our results suggest that C. burnetii infection expands the endosomal system to increase capacity for endocytic material. Furthermore, this study demonstrates the power of high-content imaging for measurement of cellular responses to infection by intracellular pathogens.


Asunto(s)
Coxiella burnetii/crecimiento & desarrollo , Coxiella burnetii/metabolismo , Endosomas/microbiología , Endosomas/ultraestructura , Vacuolas/microbiología , Infecciones por Chlamydia/microbiología , Chlamydia trachomatis/citología , Chlamydia trachomatis/fisiología , Coxiella burnetii/citología , Coxiella burnetii/patogenicidad , Endocitosis , Endosomas/metabolismo , Células HeLa , Interacciones Huésped-Patógeno , Humanos , Lisosomas , Microscopía Fluorescente , Fagosomas/microbiología , Tetraspanina 30/metabolismo , Vacuolas/metabolismo
4.
PLoS One ; 11(1): e0148032, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26821324

RESUMEN

Coxiella burnetii is a Gram-negative obligate parasitic bacterium that causes the disease Q-fever in humans. To establish its intracellular niche, it utilizes the Icm/Dot type IVB secretion system (T4BSS) to inject protein effectors into the host cell cytoplasm. The host targets of most cognate and candidate T4BSS-translocated effectors remain obscure. We used the yeast Saccharomyces cerevisiae as a model to express and study six C. burnetii effectors, namely AnkA, AnkB, AnkF, CBU0077, CaeA and CaeB, in search for clues about their role in C. burnetii virulence. When ectopically expressed in HeLa cells, these effectors displayed distinct subcellular localizations. Accordingly, GFP fusions of these proteins produced in yeast also decorated distinct compartments, and most of them altered cell growth. CaeA was ubiquitinated both in yeast and mammalian cells and, in S. cerevisiae, accumulated at juxtanuclear quality-control compartments (JUNQs) and insoluble protein deposits (IPODs), characteristic of aggregative or misfolded proteins. AnkA, which was not ubiquitinated, accumulated exclusively at the IPOD. CaeA, but not AnkA or the other effectors, caused oxidative damage in yeast. We discuss that CaeA and AnkA behavior in yeast may rather reflect misfolding than recognition of conserved targets in the heterologous system. In contrast, CBU0077 accumulated at vacuolar membranes and abnormal ER extensions, suggesting that it interferes with vesicular traffic, whereas AnkB associated with the yeast nucleolus. Both effectors shared common localization features in HeLa and yeast cells. Our results support the idea that C. burnetii T4BSS effectors manipulate multiple host cell targets, which can be conserved in higher and lower eukaryotic cells. However, the behavior of CaeA and AnkA prompt us to conclude that heterologous protein aggregation and proteostatic stress can be a limitation to be considered when using the yeast model to assess the function of bacterial effectors.


Asunto(s)
Proteínas Bacterianas/metabolismo , Coxiella burnetii/metabolismo , Agregado de Proteínas , Fiebre Q/microbiología , Saccharomyces cerevisiae/metabolismo , Apoptosis , Proteínas Bacterianas/análisis , Proteínas Bacterianas/genética , Coxiella burnetii/citología , Coxiella burnetii/genética , Expresión Génica , Células HeLa , Humanos , Estrés Oxidativo , Fiebre Q/genética , Fiebre Q/metabolismo , Saccharomyces cerevisiae/citología , Saccharomyces cerevisiae/genética , Ubiquitinación
5.
PLoS One ; 9(6): e100147, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24949863

RESUMEN

Coxiella burnetii, an obligate intracellular bacterial pathogen that causes Q fever, undergoes a biphasic developmental cycle that alternates between a metabolically-active large cell variant (LCV) and a dormant small cell variant (SCV). As such, the bacterium undoubtedly employs complex modes of regulating its lifecycle, metabolism and pathogenesis. Small RNAs (sRNAs) have been shown to play important regulatory roles in controlling metabolism and virulence in several pathogenic bacteria. We hypothesize that sRNAs are involved in regulating growth and development of C. burnetii and its infection of host cells. To address the hypothesis and identify potential sRNAs, we subjected total RNA isolated from Coxiella cultured axenically and in Vero host cells to deep-sequencing. Using this approach, we identified fifteen novel C. burnetii sRNAs (CbSRs). Fourteen CbSRs were validated by Northern blotting. Most CbSRs showed differential expression, with increased levels in LCVs. Eight CbSRs were upregulated (≥2-fold) during intracellular growth as compared to growth in axenic medium. Along with the fifteen sRNAs, we also identified three sRNAs that have been previously described from other bacteria, including RNase P RNA, tmRNA and 6S RNA. The 6S regulatory sRNA of C. burnetii was found to accumulate over log phase-growth with a maximum level attained in the SCV stage. The 6S RNA-encoding gene (ssrS) was mapped to the 5' UTR of ygfA; a highly conserved linkage in eubacteria. The predicted secondary structure of the 6S RNA possesses three highly conserved domains found in 6S RNAs of other eubacteria. We also demonstrate that Coxiella's 6S RNA interacts with RNA polymerase (RNAP) in a specific manner. Finally, transcript levels of 6S RNA were found to be at much higher levels when Coxiella was grown in host cells relative to axenic culture, indicating a potential role in regulating the bacterium's intracellular stress response by interacting with RNAP during transcription.


Asunto(s)
Coxiella burnetii/genética , ARN Bacteriano/genética , ARN Pequeño no Traducido/genética , ARN no Traducido/genética , Secuencia de Bases , Coxiella burnetii/citología , Coxiella burnetii/crecimiento & desarrollo , ARN Polimerasas Dirigidas por ADN/metabolismo , Secuenciación de Nucleótidos de Alto Rendimiento , Espacio Intracelular/metabolismo , ARN Pequeño no Traducido/metabolismo , Análisis de Secuencia de ARN , Especificidad por Sustrato , Regulación hacia Arriba
6.
J Bacteriol ; 196(11): 1925-40, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24610709

RESUMEN

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.


Asunto(s)
Proteínas Bacterianas/metabolismo , Coxiella burnetii/fisiología , Regulación Bacteriana de la Expresión Génica/fisiología , Animales , Proteínas Bacterianas/genética , Línea Celular Tumoral , Chlorocebus aethiops , Coxiella burnetii/citología , Eliminación de Gen , Humanos , ARN Bacteriano , Regulón , Células Vero
7.
J Microbiol Methods ; 96: 104-10, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24286928

RESUMEN

Coxiella burnetii undergoes a biphasic developmental cycle within its host cell that generates morphologically and physiologically distinct large cell variants (LCV) and small cell variants (SCV). During the lag phase of the C. burnetii growth cycle, non-replicating SCV differentiate into replicating LCV that in turn differentiate back into SCV during stationary phase. Nearly homogeneous SCV are observed in infected Vero cells after extended incubation (21 to 28days). In the current study, we sought to establish whether C. burnetii developmental transitions in host cells are recapitulated during host cell-free (axenic) growth in first and second generation acidified citrate cysteine media (ACCM-1 and ACCM-2, respectively). We show that ACCM-2 supported developmental transitions and viability. Although ACCM-1 also supported SCV to LCV transition, LCV to SCV transition did not occur after extended incubation (21days). Instead, C. burnetii exhibited a ghost-like appearance with bacteria containing condensed chromatin but otherwise devoid of cytoplasmic content. This phenotype correlated with a near total loss in viability between 14 and 21days of cultivation. Transcriptional profiling of C. burnetii following 14days of incubation revealed elevated expression of oxidative stress genes in ACCM-1 cultivated bacteria. ACCM-2 differs from ACCM-1 by the substitution of methyl-ß-cyclodextrin (Mß-CD) for fetal bovine serum. Addition of Mß-CD to ACCM-1 at 7days post-inoculation rescued C. burnetii viability and lowered expression of oxidative stress genes. Thus, Mß-CD appears to alleviate oxidative stress in ACCM-2 to result in C. burnetii developmental transitions and viability that mimic host cell-cultivated organisms. Axenic cultivation of C. burnetii in ACCM-2 and new methods of genetic manipulation now allow investigation of the molecular basis of C. burnetii biphasic development.


Asunto(s)
Adaptación Fisiológica , Coxiella burnetii/citología , Coxiella burnetii/crecimiento & desarrollo , Medios de Cultivo/química , Animales , Chlorocebus aethiops , Coxiella burnetii/fisiología , Viabilidad Microbiana , Células Vero
9.
Infect Immun ; 79(1): 402-13, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20937765

RESUMEN

Coxiella burnetii is a Gram-negative obligate intracellular bacterium. After internalization, this bacterium replicates in a large parasitophorous vacuole that has features of both phagolysosomes and autophagosomal compartments. We have previously demonstrated that early after internalization Coxiella phagosomes interact with both the endocytic and the autophagic pathways. In this report, we present evidence that the Coxiella-replicative vacuoles (CRVs) also interact with the secretory pathway. Rab1b is a small GTPase responsible for the anterograde transport between the endoplasmic reticulum and the Golgi apparatus. We present evidence that Rab1b is recruited to the CRV at later infection times (i.e., after 6 h of infection). Interestingly, knockdown of Rab1b altered vacuole growth, indicating that this protein was required for the proper biogenesis of the CRV. In addition, overexpression of the active GTPase-defective mutant (GFP-Rab1b Q67L) affected the development of the Coxiella-replicative compartment inhibiting bacterial growth. On the other hand, disruption of the secretory pathway by brefeldin A treatment or by overexpression of Sar1 T39N, a defective dominant-negative mutant of Sar1, affected the typical spaciousness of the CRVs. Taken together, our results show for the first time that the Coxiella-replicative niche also intercepts the early secretory pathway.


Asunto(s)
Proteínas Bacterianas/metabolismo , Coxiella burnetii/fisiología , Animales , Proteínas Bacterianas/genética , División Celular , Línea Celular , Chlorocebus aethiops , Coxiella burnetii/citología , Cricetinae , Regulación de la Expresión Génica , Humanos , Ratones , Interferencia de ARN , ARN Interferente Pequeño , Vacuolas/microbiología , Proteínas de Unión al GTP rab1/metabolismo
10.
J Microbiol Methods ; 72(3): 321-5, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18242746

RESUMEN

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.


Asunto(s)
Técnicas Bacteriológicas/métodos , Coxiella burnetii/aislamiento & purificación , Digitonina/metabolismo , Animales , Membrana Celular/efectos de los fármacos , Chlorocebus aethiops , Coxiella burnetii/citología , Coxiella burnetii/ultraestructura , Microscopía , Microscopía Electrónica de Transmisión , Células Vero
11.
J Bacteriol ; 186(21): 7344-52, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15489446

RESUMEN

Coxiella burnetii undergoes a poorly defined developmental cycle that generates morphologically distinct small-cell variants (SCV) and large-cell variants (LCV). We developed a model to study C. burnetii morphogenesis that uses Vero cells synchronously infected with homogeneous SCV (Nine Mile strain in phase II) harvested from aged infected cell cultures. A time course transmission electron microscopic analysis over 8 days of intracellular growth was evaluated in conjunction with one-step growth curves to correlate morphological differentiations with growth cycle phase. Lag phase occurred during the first 2 days postinfection (p.i.) and was primarily composed of SCV-to-LCV morphogenesis. LCV forms predominated over the next 4 days, during which exponential growth was observed. Calculated generation times during exponential phase were 10.2 h (by quantitative PCR assay) and 11.7 h (by replating fluorescent focus-forming unit assay). Stationary phase began at approximately 6 days p.i. and coincided with the reappearance of SCV, which increased in number at 8 days p.i. Quantitative reverse transcriptase-PCR demonstrated maximal expression of scvA, which encodes an SCV-specific protein, at 8 days p.i., while immunogold transmission electron microscopy revealed degradation of ScvA throughout lag and exponential phases, with increased expression observed at the onset of stationary phase. Collectively, these results indicate that the overall growth cycle of C. burnetii is characteristic of a closed bacterial system and that the replicative form of the organism is the LCV. The experimental model described in this report will allow a global transcriptome and proteome analysis of C. burnetii developmental forms.


Asunto(s)
Proteínas Bacterianas/metabolismo , Coxiella burnetii/crecimiento & desarrollo , Coxiella burnetii/ultraestructura , Proteínas de Unión al ADN/metabolismo , Regulación Bacteriana de la Expresión Génica , Animales , Proteínas Bacterianas/genética , División Celular , Chlorocebus aethiops , Coxiella burnetii/citología , Coxiella burnetii/patogenicidad , Proteínas de Unión al ADN/genética , Humanos , Microscopía Electrónica de Transmisión , Células Vero/microbiología
12.
Ann N Y Acad Sci ; 990: 460-7, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12860674

RESUMEN

During human Coxiella burnetii (C. burnetii) infections, high IL-10 levels favor replication of C. burnetii in monocytes and development of chronic Q fever, whereas IFN-gamma promotes intracellular killing. Sheep are a common source for human C. burnetii infections, but in contrast to man become transiently infected only. In a first approach to unravel the role of cytokines during ovine C. burnetii infections, we investigated by semiquantitative RT-PCR whether heat-inactivated C. burnetii affects the transcription of genes coding for IL-2, IL-4, IL-10, and INF-gamma in vitro in PBMC from sheep seropositive or seronegative for C. burnetii. By computer-assisted evaluation of band intensities the transcription rate of the cytokine genes was quantified in relation to transcription in Concanavalin A-stimulated and nonstimulated controls. Transcription rates in PBMC from seropositive animals after incubation with C. burnetii for 4 hours strongly resembled those found in PBMC from seronegative sheep. However, upon prolonged incubation (24 h) C. burnetii induced an increased IL-10 transcription in PBMC from 2 of 5 seronegative, but in PBMC from 5 of 5 seropositive animals. The data suggest that natural C. burnetii infections prime the ovine immune system towards a T(H)2-like pattern and this action thereby represents the first clue for the involvement of ovine immune cells in the response to C. burnetii infections.


Asunto(s)
Coxiella burnetii/crecimiento & desarrollo , Citocinas/genética , Linfocitos/inmunología , Linfocitos/microbiología , Animales , División Celular , Coxiella burnetii/citología , Coxiella burnetii/inmunología , Femenino , Calor , Humanos , Técnicas In Vitro , Interleucinas/genética , Reacción en Cadena de la Polimerasa , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Ovinos , Transcripción Genética
13.
Ann N Y Acad Sci ; 990: 450-9, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12860673

RESUMEN

Coxiella burnetii, an obligate intracellular bacterium, is the agent of Q fever. The chronic disease is characterized by impaired cell-mediated immune response and microbicidal activity of monocytes. We hypothesized that interleukin(IL)-4, a Th2 cytokine, interferes with the fate of C. burnetii inside monocytes. C. burnetii survived without multiplication in resting monocytes, but replicated in IL-4-treated monocytes. The effect of IL-4 is specific for monocytes since IL-4 did not stimulate C. burnetii replication in monocyte-derived macrophages. The effects of IL-4 on bacterial replication and on tumor necrosis factor (TNF) production in monocytes were apparently not related. Although IL-4 inhibited C. burnetii-stimulated release of TNF, the addition of recombinant TNF to IL-4-treated monocytes did not prevent the IL-4 effect. These results suggest that IL-4 enables monocytes to support C. burnetii replication and a Th2 polarization of immune response that may interfere with immune control of Q fever.


Asunto(s)
Coxiella burnetii/fisiología , Interleucina-4/farmacología , Leucocitos Mononucleares/microbiología , Macrófagos/microbiología , División Celular/efectos de los fármacos , Coxiella burnetii/citología , Coxiella burnetii/efectos de los fármacos , Humanos , Técnicas In Vitro , Cinética , Factor de Necrosis Tumoral alfa/farmacología
14.
Acta Virol ; 46(2): 121-4, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-12387505

RESUMEN

Phospholipids extracted from the Coxiella burnetii strain Nine Mile virulent phase I and low-virulent phase II cells were directly analyzed by fast atom bombardment mass spectrometry (FAB-MS). Constant neutral loss (CNL) scanning mass spectra (MS) were acquired to identify various phospholipids within phospholipid classes. Phospholipids from the phase I C. burnetii cells were much more complex than those from the phase II cells. Moreover, in the latter, the absence of phospholipids of the phosphatidylinositol class could be noticed. The results indicate that CNL scanning of phospholipid samples provides a rapid and simple method for identification of the phase state of the bacterium.


Asunto(s)
Coxiella burnetii/química , Coxiella burnetii/patogenicidad , Fosfolípidos/análisis , Espectrometría de Masa Bombardeada por Átomos Veloces/métodos , Coxiella burnetii/citología , Modelos Moleculares , Fosfolípidos/química , Fiebre Q/metabolismo , Fiebre Q/virología , Virulencia , Factores de Virulencia
15.
Acta Virol ; 35(6): 545-56, 1991 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-1687638

RESUMEN

An in vitro ultrastructural study was carried out on tissue cultures (J774, murine macrophage-like tumour cell line, and BHK-21, baby hamster kidney cell line) persistently infected with C. burnetii to investigate whether the events of cellular differentiation could be visualized. At a given stage of the developmental cycle, a proportion of the cells within the affected phagolysosomes clearly underwent cellular differentiation. The cells initially showed asymmetrical septation, the primary stage of cellular differentiation, and ended with the formation of the differentiated product, a precursor to the small cell. The results verified our initial observation that the events occurring during growth in a phagolysosome represent stages of a complex developmental cycle consisting not only of i) vegetative growth by typical transverse binary fission, but also ii) cellular differentiation.


Asunto(s)
Coxiella burnetii/ultraestructura , Animales , Ciclo Celular/fisiología , Diferenciación Celular/fisiología , Coxiella burnetii/citología , Técnicas de Cultivo , Microscopía Electrónica , Fagosomas/microbiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...