Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
iScience ; 25(9): 104909, 2022 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-36060064

RESUMEN

Leishmania parasites use elaborate virulence mechanisms to invade and thrive in macrophages. These virulence mechanisms inhibit host cell defense responses and generate a specialized replicative niche, the parasitophorous vacuole. In this work, we performed a genome-wide RNAi screen in Drosophila macrophage-like cells to identify the host factors necessary for Leishmania amazonensis infection. This screen identified 52 conserved genes required specifically for parasite entry, including several components of the SUMOylation machinery. Further studies in mammalian macrophages found that L. amazonensis infection inhibited SUMOylation within infected macrophages and this inhibition enhanced parasitophorous vacuole growth and parasite proliferation through modulation of multiple genes especially ATP6V0D2, which in turn affects CD36 expression and cholesterol levels. Together, these data suggest that parasites actively sabotage host SUMOylation and alter host transcription to improve their intracellular niche and enhance their replication.

2.
Infect Immun ; 88(11)2020 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-32817329

RESUMEN

Enucleated cells or cytoplasts (cells whose nucleus is removed in vitro) represent an unexplored biological model for intracellular infection studies due to the abrupt interruption of nuclear processing and new RNA synthesis by the host cell in response to pathogen entry. Using enucleated fibroblasts hosting the protozoan parasite Leishmania amazonensis, we demonstrate that parasite multiplication and biogenesis of large parasitophorous vacuoles in which parasites multiply are independent of the host cell nucleus. Dual RNA sequencing of both host cytoplast and intracellular parasite transcripts identified host transcripts that are more preserved or degraded upon interaction with parasites and also parasite genes that are differentially expressed when hosted by nucleated or enucleated cells. Cytoplasts are suitable host cells, which persist in culture for more than 72 h and display functional enrichment of transcripts related to mitochondrial functions and mRNA translation. Crosstalk between nucleated host de novo gene expression in response to intracellular parasitism and the parasite gene expression to counteract or benefit from these host responses induces a parasite transcriptional profile favoring parasite multiplication and aerobic respiration, and a host-parasite transcriptional landscape enriched in host cell metabolic functions related to NAD, fatty acid, and glycolytic metabolism. Conversely, interruption of host nucleus-parasite cross talk by infection of enucleated cells generates a host-parasite transcriptional landscape in which cytoplast transcripts are enriched in phagolysosome-related pathway, prosurvival, and SerpinB-mediated immunomodulation. In addition, predictive in silico analyses indicated that parasite transcript products secreted within cytoplasts interact with host transcript products conserving the host V-ATPase proton translocation function and glutamine/proline metabolism. The collective evidence indicates parasite-mediated control of host cell transcripts half-life that is beneficial to parasite intracellular multiplication and escape from host immune responses. These findings will contribute to improved drug targeting and serve as database for L. amazonensis-host cell interactions.


Asunto(s)
Fibroblastos/parasitología , Regulación de la Expresión Génica Arqueal/fisiología , Interacciones Huésped-Parásitos/fisiología , Leishmania mexicana/parasitología , Leishmania/fisiología , Animales , Línea Celular , Ratones , Transcriptoma
3.
Infect Immun ; 84(5): 1603-1614, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26975994

RESUMEN

The trypanosomatids Leishmania amazonensis and Trypanosoma cruzi are excellent models for the study of the cell biology of intracellular protozoan infections. After their uptake by mammalian cells, the parasitic protozoan flagellates L. amazonensis and T. cruzi lodge within acidified parasitophorous vacuoles (PVs). However, whereas L. amazonensis develops in spacious, phagolysosome-like PVs that may enclose numerous parasites, T. cruzi is transiently hosted within smaller vacuoles from which it soon escapes to the host cell cytosol. To investigate if parasite-specific vacuoles are required for the survival and differentiation of T. cruzi, we constructed chimeric vacuoles by infection of L. amazonensis amastigote-infected macrophages with T. cruzi epimastigotes (EPIs) or metacyclic trypomastigotes (MTs). These chimeric vacuoles, easily observed by microscopy, allowed the entry and fate of T. cruzi in L. amazonensis PVs to be dynamically recorded by multidimensional imaging of coinfected cells. We found that although T. cruzi EPIs remained motile and conserved their morphology in chimeric vacuoles, T. cruzi MTs differentiated into amastigote-like forms capable of multiplying. These results demonstrate that the large adaptive vacuoles of L. amazonensis are permissive to T. cruzi survival and differentiation and that noninfective EPIs are spared from destruction within the chimeric PVs. We conclude that T. cruzi differentiation can take place in Leishmania-containing vacuoles, suggesting this occurs prior to their escape into the host cell cytosol.


Asunto(s)
Diferenciación Celular , Leishmania/fisiología , Macrófagos/parasitología , Trypanosoma cruzi/fisiología , Vacuolas/parasitología , Animales , Coinfección/parasitología , Leishmania/crecimiento & desarrollo , Ratones , Microscopía Confocal , Microscopía Electrónica de Rastreo , Microscopía Electrónica de Transmisión , Microscopía Fluorescente , Células RAW 264.7 , Trypanosoma cruzi/crecimiento & desarrollo
4.
Cell Microbiol ; 13(8): 1234-49, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21658173

RESUMEN

Bacterial and parasitic intracellular pathogens or their secreted products have been shown to induce host cell transcriptional responses, which may benefit the host, favour the microorganism or be unrelated to the infection. In most instances, however, it is not known if the host cell nucleus is proximately required for the development of an intracellular infection. This information can be obtained by the infection of artificially enucleated host cells (cytoplasts). This model, although rather extensively used in studies of viral infection, has only been applied to few bacterial pathogens, which do not include Mycobacterium spp. Here, we investigate the internalization, phagosome biogenesis and survival of M. smegmatis in enucleated type II alveolar epithelial cells. Cytoplasts were infected with M. smegmatis, but the percentage of infection was significantly lower than that of nucleated cells. Scanning electron microscopy indicated that in both cells and cytoplasts, bacteria were internalized by a phagocytosis-like mechanism. Interestingly, phagosome fusion with lysosomes and mycobacterial killing were both more efficient in enucleated than in nucleated cells, a finding that may be correlated with the increased number of autophagic vesicles developed in cytoplasts. We provide evidence that although quantitative changes were observed, the full development of the infection, as well as mycobacterial killing did not require the presence of the host cell nucleus.


Asunto(s)
Endocitosis , Células Epiteliales/microbiología , Lisosomas/microbiología , Mycobacterium smegmatis/patogenicidad , Fagosomas/microbiología , Línea Celular , Núcleo Celular/fisiología , Humanos , Viabilidad Microbiana , Microscopía Electrónica , Microscopía Fluorescente
5.
PLoS Negl Trop Dis ; 4(12): e905, 2010 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-21151877

RESUMEN

Protozoan parasites of the genus Leishmania alternate between flagellated, elongated extracellular promastigotes found in insect vectors, and round-shaped amastigotes enclosed in phagolysosome-like Parasitophorous Vacuoles (PVs) of infected mammalian host cells. Leishmania amazonensis amastigotes occupy large PVs which may contain many parasites; in contrast, single amastigotes of Leishmania major lodge in small, tight PVs, which undergo fission as parasites divide. To determine if PVs of these Leishmania species can fuse with each other, mouse macrophages in culture were infected with non-fluorescent L. amazonensis amastigotes and, 48 h later, superinfected with fluorescent L. major amastigotes or promastigotes. Fusion was investigated by time-lapse image acquisition of living cells and inferred from the colocalization of parasites of the two species in the same PVs. Survival, multiplication and differentiation of parasites that did or did not share the same vacuoles were also investigated. Fusion of PVs containing L. amazonensis and L. major amastigotes was not found. However, PVs containing L. major promastigotes did fuse with pre-established L. amazonensis PVs. In these chimeric vacuoles, L. major promastigotes remained motile and multiplied, but did not differentiate into amastigotes. In contrast, in doubly infected cells, within their own, unfused PVs metacyclic-enriched L. major promastigotes, but not log phase promastigotes--which were destroyed--differentiated into proliferating amastigotes. The results indicate that PVs, presumably customized by L. major amastigotes or promastigotes, differ in their ability to fuse with L. amazonensis PVs. Additionally, a species-specific PV was required for L. major destruction or differentiation--a requirement for which mechanisms remain unknown. The observations reported in this paper should be useful in further studies of the interactions between PVs to different species of Leishmania parasites, and of the mechanisms involved in the recognition and fusion of PVs.


Asunto(s)
Leishmania major/patogenicidad , Leishmania mexicana/patogenicidad , Macrófagos/parasitología , Vacuolas/parasitología , Animales , Células Cultivadas , Femenino , Leishmania major/crecimiento & desarrollo , Leishmania mexicana/crecimiento & desarrollo , Ratones , Ratones Endogámicos BALB C , Viabilidad Microbiana , Microscopía por Video
6.
Infect Immun ; 75(8): 3700-6, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17502387

RESUMEN

Cell infection with Trypanosoma cruzi, the agent of Chagas' disease, begins with the uptake of infective trypomastigotes within phagosomes and their release into the cytosol, where they transform into replicating amastigotes; the latter, in turn, differentiate into cytolytically released and infective trypomastigotes. We ask here if the T. cruzi infection program can develop in enucleated host cells. Monolayers of L929 cells, enucleated by centrifugation in the presence of cytochalasin B and kept at 34 degrees C to extend the survival of cytoplasts, were infected with parasites of the CL strain. Percent infection, morphology, stage-specific markers, and numbers of parasites per cell were evaluated in nucleated and enucleated cells, both of which were present in the same preparations. Parasite uptake, differentiation and multiplication of amastigotes, development of epimastigote- and trypomastigote-like forms, and initial cytolytic release of parasites were all documented for cytoplasts and nucleated cells. Although the doubling times were similar, parasite loads at 48 and 72 h were significantly lower in the cytoplasts than in nucleated cells. Similar results were obtained with the highly virulent strain Y as well as with strains CL-14 and G, which exhibit low virulence for mice. Cytoplasts could also be infected with the CL strain 24 or 48 h after enucleation. Thus, infection of cells by T. cruzi can take place in enucleated host cells, i.e., in the absence of modulation of chromosomal and nucleolar gene transcription and of RNA modification and processing in the nucleus.


Asunto(s)
Núcleo Celular , Citoplasma/parasitología , Trypanosoma cruzi/crecimiento & desarrollo , Animales , Línea Celular , Femenino , Fibroblastos/parasitología , Estadios del Ciclo de Vida , Ratones , Microscopía Fluorescente
7.
Cell Microbiol ; 9(4): 891-909, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17087732

RESUMEN

The etiologic agent of Q fever Coxiella burnetii, is an intracellular obligate parasite that develops large vacuoles with phagolysosomal characteristics, containing multiple replicating bacteria. We have previously shown that Phase II C. burnetii replicative vacuoles generated after 24-48 h post infection are decorated with the autophagic protein LC3. The aim of the present study was to examine, at earlier stages of infection, the distribution and roles of the small GTPases Rab5 and Rab7, markers of early and late endosomes respectively, as well as of the protein LC3 on C. burnetii trafficking. Our results indicate that: (i) Coxiella phagosomes (Cph) acquire the two Rab proteins sequentially during infection; (ii) overexpression of a dominant negative mutant form of Rab5, but not of Rab7, impaired Coxiella entry, whereas both Rab5 and Rab7 dominant negative mutants inhibited vacuole formation; (iii) Cph colocalized with the protein LC3 as early as 5 min after infection; acquisition of this protein appeared to be a bacterially driven process, because it was inhibited by the bacteriostatic antibiotic chloramphenicol and (iv) C. burnetii delayed the arrival of the typical lysosomal protease cathepsin D to the Cph, which delay is further increased by starvation-induced autophagy. Based on our results we propose that C. burnetii transits through the normal endo/phagocytic pathway but actively interacts with autophagosomes at early times after infection. This intersection with the autophagic pathway delays fusion with the lysosomal compartment possibly favouring the intracellular differentiation and survival of the bacteria.


Asunto(s)
Autofagia/fisiología , Coxiella burnetii/crecimiento & desarrollo , Transducción de Señal/fisiología , Animales , Autofagia/genética , Western Blotting , Células CHO , Catepsinas/genética , Catepsinas/metabolismo , Cricetinae , Cricetulus , Técnica del Anticuerpo Fluorescente Indirecta , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Lisosomas/metabolismo , Lisosomas/microbiología , Microscopía Fluorescente , Fagosomas/metabolismo , Transducción de Señal/genética , Vacuolas/metabolismo , Vacuolas/microbiología , Proteínas de Unión al GTP rab/genética , Proteínas de Unión al GTP rab/metabolismo , Proteínas de Unión al GTP rab5/genética , Proteínas de Unión al GTP rab5/metabolismo , Proteínas de Unión a GTP rab7
8.
Cell Microbiol ; 7(7): 981-93, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15953030

RESUMEN

Pathogens evolved mechanisms to invade host cells and to multiply in the cytosol or in compositionally and functionally customized membrane-bound compartments. Coxiella burnetii, the agent of Q fever in man is a Gram-negative gamma-proteobacterium which multiplies in large, acidified, hydrolase-rich and fusogenic vacuoles with phagolysosomal-like characteristics. We reported previously that C. burnetii phase II replicative compartments are labelled by LC3, a protein specifically localized to autophagic vesicles. We show here that autophagy in Chinese hamster ovary cells, induced by amino acid deprivation prior to infection with Coxiella increased the number of infected cells, the size of the vacuoles, and their bacterial load. Furthermore, overexpression of GFP-LC3 or of GFP-Rab24 - a protein also localized to autophagic vacuoles - likewise accelerated the development of Coxiella-vacuoles at early times after infection. However, overexpression of mutants of those proteins that cannot be targeted to autophagosomes dramatically decreased the number and size of the vacuoles in the first hours of infection, although by 48 h the infection was similar to that of non-transfected controls. Overall, the results suggest that transit through the autophagic pathway increases the infection with Coxiella by providing a niche more favourable to their initial survival and multiplication.


Asunto(s)
Autofagia , Coxiella burnetii/crecimiento & desarrollo , Vacuolas/microbiología , Animales , Células CHO , Coxiella burnetii/patogenicidad , Cricetinae , Genes Reporteros , Proteínas Fluorescentes Verdes/análisis , Humanos , Microscopía Confocal , Microscopía Fluorescente , Proteínas Asociadas a Microtúbulos/metabolismo , Modelos Biológicos , Vacuolas/química , Vacuolas/metabolismo , Proteínas de Unión al GTP rab/metabolismo
9.
Infect Immun ; 72(4): 2075-80, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15039329

RESUMEN

Coxiella burnetii, the agent of Q fever in humans and coxiellosis in other mammals, is an obligate intracellular bacterium which is sheltered and multiplies within typically large phagolysosome-like replicative vacuoles (LRVs). We have previously shown that, compared with fibroblasts, mouse resident peritoneal macrophages control the development of LRVs and bacterial multiplication within these vacuoles. Earlier experiments with the nitric oxide (NO) synthase inhibitor aminoguanidine (AG) revealed that the control is exerted by NO induced by the bacteria. We report here that phagocytosis of apoptotic-like, but not of aldehyde-killed, lymphocytes by the macrophages reduced the production of NO induced by the bacteria and increased the development of LRVs and, therefore, the total bacterial load in the cultures. Experiments with macrophages from mice deficient for inducible NO synthase (iNOS(-)/(-)) confirmed the involvement of NO in the control of infection, since neither apoptotic lymphocytes nor AG affected the development of LRVs in these phagocytes. Since macrophages are important for the clearance of apoptotic bodies and C. burnetii is able to induce apoptosis in human monocytes, the phenomenon shown here may be biologically relevant to the development of Q fever and coxiellosis.


Asunto(s)
Apoptosis , Coxiella burnetii/patogenicidad , Regulación hacia Abajo , Macrófagos/microbiología , Óxido Nítrico/biosíntesis , Fagocitosis , Animales , Células Cultivadas , Coxiella burnetii/ultraestructura , Susceptibilidad a Enfermedades , Humanos , Macrófagos/inmunología , Macrófagos/fisiología , Ratones , Microscopía Confocal , Fagosomas/microbiología , Fiebre Q/microbiología , Vacuolas/microbiología
10.
Mol Microbiol ; 49(4): 965-76, 2003 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12890021

RESUMEN

Coxiella burnetii is an obligate intracellular pathogen that replicates in large endocytic vacuoles. Genomic sequence data indicate that 21 genes encoding products that are similar to components of the Legionella pneumophila Dot/Icm type IV secretion system are located on a contiguous 35 kb region of the Coxiella chromosome. It was found that several dot/icm genes were expressed by Coxiella during host cell infection and that dot/icm gene expression preceded the formation of large replicative vacuoles. To determine whether these genes encode a functional type IV secretion system, we have amplified the Coxiella dotB, icmQ, icmS and icmW genes and produced the encoded proteins in Legionella mutants in which the native copy of each gene had been deleted. The Coxiella dotB, icmS and icmW products restored dot/icm-dependent growth of Legionella mutants in eukaryotic host cells. The Coxiella IcmQ protein and the Legionella IcmR protein did not interact, which could explain why the Coxiella icmQ gene was unable to restore growth to a Legionella icmQ mutant. Thus, Coxiella encodes functional components of a type IV secretion system expressed in vivo that is mechanistically related to the Legionella Dot/Icm apparatus. These studies suggest that a dot/icm-related secretion system could play an important role in creating the specialized vacuole that supports Coxiella replication.


Asunto(s)
Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Coxiella burnetii/genética , Coxiella burnetii/metabolismo , Legionella pneumophila/metabolismo , Chaperonas Moleculares/metabolismo , Animales , Chlorocebus aethiops , Regulación Bacteriana de la Expresión Génica , Vectores Genéticos , Humanos , Legionella pneumophila/genética , Macrófagos/citología , Macrófagos/metabolismo , Macrófagos/microbiología , Ratones , Chaperonas Moleculares/genética , Vacuolas/metabolismo , Células Vero
11.
Infect Immun ; 71(3): 1225-33, 2003 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-12595436

RESUMEN

In most primary or continuous cell cultures infected with the Q-fever agent Coxiella burnetii, bacteria are typically sheltered in phagolysosome-like, large replicative vacuoles (LRVs). We recently reported that only a small proportion of mouse peritoneal macrophages (PMPhi) infected with a nonvirulent, phase II strain of C. burnetii developed LRVs and that their relative bacterial load increased only slowly. In the majority of infected PMPhi, the bacteria were confined to the small vesicles. We show here that nitric oxide (NO) induced by the bacteria partially accounts for the restricted development of LRVs in primary macrophages. Thus, (i) PMPhi and bone marrow-derived macrophages (BMMPhi) challenged with phase II C. burnetii produced significant amounts of NO; (ii) the NO synthase inhibitors aminoguanidine and N-methyl-L-arginine reduced the production of NO and increased the frequency of LRVs (although the relative bacterial loads of individual LRVs did not change, the estimated loads per well increased appreciably); (iii) gamma interferon (IFN-gamma) or the NO donor sodium nitroprusside, added to BMMPhi prior to or after infection, reduced the development and the relative bacterial loads of LRVs and lowered the yield of viable bacteria recovered from the cultures; and (iv) these effects of IFN-gamma may not be entirely dependent on the production of NO since IFN-gamma also controlled the infection in macrophages from inducible NO synthase knockout mice. It remains to be determined whether NO reduced the development of LRVs by acting directly on the bacteria; by acting on the traffic, fusion, or fission of cell vesicles; or by a combination of these mechanisms.


Asunto(s)
Coxiella burnetii/fisiología , Macrófagos/microbiología , Óxido Nítrico/fisiología , Animales , Femenino , Interferón gamma/farmacología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Óxido Nítrico Sintasa/antagonistas & inhibidores , Óxido Nítrico Sintasa/fisiología , Óxido Nítrico Sintasa de Tipo II , Nitroprusiato/farmacología , Vacuolas/microbiología
12.
Cytokine ; 19(5): 213-7, 2002 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-12393167

RESUMEN

Dietary fish-oil (FO) supplementation has been shown to inhibit inflammation in various clinical disease states and to be beneficial in experimental models of inflammation and bacterial and plasmodial infection. In mice, FO increases macrophage production of tumour necrosis factor alpha (TNF). Production of TNF has been reported to be important in the resistance of mice against various Leishmania spp. We investigated whether dietary supplementation with FO protects susceptible Balb/c mice against infection with Leishmania amazonensis. No influence of the FO diet on the course of infection was observed, as evaluated by the increase in thickness of infected footpads over forty days. Lipopolysaccharide (LPS)-induced TNF production of peritoneal cells was however significantly increased in FO fed mice (P<0.01). When L. amazonensis was used as a stimulus, the in-vitro production of TNF by isolated peritoneal cells was minimal and did not differ between the various treatment groups. Addition of interferon gamma did not restore the effect of FO on TNF production capacity. We conclude that dietary supplementation with FO is of no benefit in Leishmaniasis in susceptible Balb/c mice, and that L. amazonensis is an insufficient trigger for TNF production in this model.


Asunto(s)
Suplementos Dietéticos , Aceites de Pescado/uso terapéutico , Leishmaniasis/tratamiento farmacológico , Animales , Peso Corporal/efectos de los fármacos , Femenino , Aceites de Pescado/farmacología , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Interferón gamma/farmacología , Leishmania/efectos de los fármacos , Leishmania/fisiología , Leishmaniasis/metabolismo , Ratones , Ratones Endogámicos BALB C , Factores de Tiempo , Factor de Necrosis Tumoral alfa/metabolismo
13.
Infect Immun ; 70(10): 5816-21, 2002 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12228312

RESUMEN

The obligate intracellular bacterium Coxiella burnetii, the agent of Q fever in humans and of coxiellosis in other animals, survives and replicates within large, acidified, phagolysosome-like vacuoles known to fuse homo- and heterotypically with other vesicles. To further characterize these vacuoles, HeLa cells were infected with C. burnetii phase II; 48 h later, bacteria-containing vacuoles were labeled by LysoTracker, a marker of acidic compartments, and accumulated monodansylcadaverine and displayed protein LC3, both markers of autophagic vacuoles. Furthermore, 3-methyladenine and wortmannin, agents known to inhibit early stages in the autophagic process, each blocked Coxiella vacuole formation. These autophagosomal features suggest that Coxiella vacuoles interact with the autophagic pathway. The localization and role of wild-type and mutated Rab5 and Rab7, markers of early and late endosomes, respectively, were also examined to determine the role of these small GTPases in the trafficking of C. burnetii phase II. Green fluorescent protein (GFP)-Rab5 and GFP-Rab7 constructs were overexpressed and visualized by fluorescence microscopy. Coxiella-containing large vacuoles were labeled with wild-type Rab7 (Rab7wt) and with GTPase-deficient mutant Rab7Q67L, whereas no colocalization was observed with the dominant-negative mutant Rab7T22N. The vacuoles were also decorated by GFP-Rab5Q79L but not by GFP-Rab5wt. These results suggest that Rab7 participates in the biogenesis of the parasitophorous vacuoles.


Asunto(s)
Coxiella burnetii/patogenicidad , Fiebre Q/metabolismo , Fiebre Q/microbiología , Vacuolas/microbiología , Proteínas de Unión al GTP rab/metabolismo , Animales , Autofagia , Compartimento Celular , Células HeLa , Humanos , Mutación , Fiebre Q/patología , Vacuolas/metabolismo , Vacuolas/patología , Proteínas de Unión al GTP rab/genética , Proteínas de Unión a GTP rab7
14.
Microbes Infect ; 4(6): 591-8, 2002 May.
Artículo en Inglés | MEDLINE | ID: mdl-12048028

RESUMEN

Coxiella burnetii, the agent of Q fever in man and of coxiellosis in other species, is a small, dimorphic, obligate intracellular bacterium, sheltered within large, acidified, and hydrolase-rich phagosomes. Although several primary and established cell lines, macrophage-like cells, and primary macrophages from other species have been infected with C. burnetii, the infection of mouse primary macrophages has not been sufficiently characterized. In this report quantification of DAPI (4', 6-diamino-2-phenylindole) fluorescence images acquired by confocal microscopy, and transmission electron microscopy were used to compare the infection of three mouse-derived cells, L929 fibroblasts, J774 macrophage-like cells, and resident peritoneal macrophages, with a phase II clone of C. burnetii known to be non-virulent for mammals. Infected peritoneal phagocytes differed from L929 or J774 cells in that: (a) large vacuoles took longer to appear (3-5 d instead of 2), and were only found in a subset (20-30%) of macrophages, as opposed to in more than 70% of the other cells; (b) total and vacuole-associated relative bacterial loads in L929 and J774 cells were several-fold higher than in peritoneal macrophages; (c) estimated doubling times of the bacteria were about 68 h in the primary macrophages, 18 h in J774 and 22 h in L929 cells. Thus, mouse resident peritoneal macrophages control both the formation of the large vacuoles and the intracellular proliferation of C. burnetii phase II.


Asunto(s)
Coxiella burnetii/inmunología , Macrófagos Peritoneales/microbiología , Animales , Coxiella burnetii/crecimiento & desarrollo , Femenino , Colorantes Fluorescentes/química , Indoles/química , Macrófagos Peritoneales/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Microscopía Confocal , Microscopía Electrónica , Microscopía de Interferencia , Fiebre Q/inmunología , Fiebre Q/microbiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA