Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Pathog Dis ; 812023 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-37193663

RESUMEN

Coxiella burnetii (Cb) is an obligate intracellular pathogen in nature and the causative agent of acute Q fever as well as chronic diseases. In an effort to identify genes and proteins crucial to their normal intracellular growth lifestyle, we applied a 'reverse evolution' approach where the avirulent Nine Mile Phase II strain of Cb was grown for 67 passages in chemically defined ACCM-D media and gene expression patterns and genome integrity from various passages was compared to passage number one following intracellular growth. Transcriptomic analysis identified a marked downregulation of the structural components of the type 4B secretion system (T4BSS), the general secretory (Sec) pathway, as well as 14 out of 118 previously identified genes encoding effector proteins. Additional downregulated pathogenicity determinants genes included several chaperones, LPS, and peptidoglycan biosynthesis. A general marked downregulation of central metabolic pathways was also observed, which was balanced by a marked upregulation of genes encoding transporters. This pattern reflected the richness of the media and diminishing anabolic, and ATP-generation needs. Finally, genomic sequencing and comparative genomic analysis demonstrated an extremely low level of mutation across passages, despite the observed Cb gene expression changes following acclimation to axenic media.


Asunto(s)
Coxiella burnetii , Fiebre Q , Humanos , Transcriptoma , Perfilación de la Expresión Génica , Genómica
2.
bioRxiv ; 2023 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-36798183

RESUMEN

Coxiella burnetii (Cb) is an obligate intracellular pathogen in nature and the causative agent of acute Q fever as well as chronic diseases. In an effort to identify genes and proteins crucial to their normal intracellular growth lifestyle, we applied a "Reverse evolution" approach where the avirulent Nine Mile Phase II strain of Cb was grown for 67 passages in chemically defined ACCM-D media and gene expression patterns and genome integrity from various passages was compared to passage number one following intracellular growth. Transcriptomic analysis identified a marked downregulation of the structural components of the type 4B secretion system (T4BSS), the general secretory (sec) pathway, as well as 14 out of 118 previously identified genes encoding effector proteins. Additional downregulated pathogenicity determinants genes included several chaperones, LPS, and peptidoglycan biosynthesis. A general marked downregulation of central metabolic pathways was also observed, which was balanced by a marked upregulation of genes encoding transporters. This pattern reflected the richness of the media and diminishing anabolic and ATP-generation needs. Finally, genomic sequencing and comparative genomic analysis demonstrated an extremely low level of mutation across passages, despite the observed Cb gene expression changes following acclimation to axenic media.

4.
Microbiology (Reading) ; 165(1): 1-3, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30422108

RESUMEN

Coxiella burnetii is an obligate intracellular pathogen that causes acute and chronic Q fever. C. burnetii grows within a eukaryotic host cell in a vacuole highly similar to a phagolysosome. Found worldwide, this environmentally stable pathogen is maintained in nature via chronic infection of ruminants. Aerosol-mediated infection of humans results in infection and usurpation of alveolar macrophages through mechanisms using a bacterial Type 4B Secretion System and secreted effector proteins. Advances in axenic culture and genetic systems are changing our understanding of the pathogen's physiology and intimate molecular manipulations of host cells during infection.


Asunto(s)
Coxiella burnetii/metabolismo , Fiebre Q/microbiología , Ácidos/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Sistemas de Secreción Bacterianos/genética , Sistemas de Secreción Bacterianos/metabolismo , Coxiella burnetii/clasificación , Coxiella burnetii/genética , Coxiella burnetii/aislamiento & purificación , Genoma Bacteriano , Humanos , Concentración de Iones de Hidrógeno , Filogenia , Vacuolas/química , Vacuolas/microbiología
5.
Artículo en Inglés | MEDLINE | ID: mdl-30555802

RESUMEN

Chlamydia trachomatis is an obligate intracellular pathogen with global health and economic impact. Upon infection, C. trachomatis resides within a protective niche, the inclusion, wherein it replicates and usurps host cell machinery and resources. The inclusion membrane is the key host-pathogen interface that governs specific protein-protein interactions to manipulate host signaling pathways. At the conclusion of the infection cycle, C. trachomatis exits the host cell via lysis or extrusion. Extrusion depends on the phosphorylation state of myosin light chain 2 (MLC2); the extent of phosphorylation is determined by the ongoing opposing activities of myosin phosphatase (MYPT1) and myosin kinase (MLCK). Previously, it was shown that MYPT1 is recruited to the inclusion and interacts with CT228 for regulation of host cell egress. In this study, we generated a targeted chromosomal mutation of CT228 (L2-ΔCT228) using the TargeTron system and demonstrate a loss of MYPT1 recruitment and increase in extrusion production in vitro. Mutation of CT228 did not affect chlamydial growth in cell culture or recruitment of MLC2. Moreover, we document a delay in clearance of L2-ΔCT228 during murine intravaginal infection as well as a reduction in systemic humoral response, relative to L2-wild type. Taken together, the data suggest that loss of MYPT1 recruitment (as a result of CT228 disruption) regulates the degree of host cell exit via extrusion and affects the longevity of infection in vivo.


Asunto(s)
Infecciones por Chlamydia/metabolismo , Chlamydia trachomatis/genética , Chlamydia trachomatis/patogenicidad , Silenciador del Gen , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Animales , Infecciones por Chlamydia/inmunología , Infecciones por Chlamydia/patología , Modelos Animales de Enfermedad , Femenino , Células HeLa , Interacciones Huésped-Patógeno , Humanos , Cuerpos de Inclusión/metabolismo , Ratones , Ratones Endogámicos C3H , Mutación , Cadenas Ligeras de Miosina , Quinasa de Cadena Ligera de Miosina/metabolismo , Fosfatasa de Miosina de Cadena Ligera/metabolismo , Fosforilación , Dominios y Motivos de Interacción de Proteínas , Útero/patología
6.
Pathog Dis ; 75(4)2017 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-28449081

RESUMEN

Coxiella burnetii is a Gram-negative intracellular pathogen and is the causative agent of the zoonotic disease Q fever. To cause disease, C. burnetii requires a functional type IVB secretion system (T4BSS) to transfer effector proteins required for the establishment and maintenance of a membrane-bound parasitophorous vacuole (PV) and further modulation of host cell process. However, it is not clear how the T4BSS interacts with the PV membrane since neither a secretion pilus nor an extracellular pore forming apparatus has not been described. To address this, we used the acidified citrate cysteine medium (ACCM) along with cell culture infection and immunological techniques to identify the cellular and extracellular localization of T4BSS components. Interestingly, we found that DotA and IcmX were secreted/released in a T4BSS-dependent manner into the ACCM. Analysis of C. burnetii-infected cell lines revealed that DotA colocalized with the host cell marker CD63 (LAMP3) at the PV membrane. In the absence of bacterial protein synthesis, DotA also became depleted from the PV membrane. These data are the first to identify the release/secretion of C. burnetii T4BSS components during axenic growth and the interaction of a T4BSS component with the PV membrane during infection of host cells.


Asunto(s)
Proteínas Bacterianas/metabolismo , Coxiella burnetii/crecimiento & desarrollo , Coxiella burnetii/metabolismo , Interacciones Huésped-Patógeno , Sistemas de Secreción Tipo IV/metabolismo , Vacuolas/microbiología , Proteínas Bacterianas/análisis , Tetraspanina 30/análisis , Vacuolas/química
7.
Artículo en Inglés | MEDLINE | ID: mdl-28066723

RESUMEN

Coxiella burnetii is the causative agent of Q fever and an obligate intracellular pathogen in nature that survives and grows in a parasitophorous vacuole (PV) within eukaryotic host cells. C. burnetii promotes intracellular survival by subverting apoptotic and pro-inflammatory signaling pathways that are typically regulated by nuclear transcription factor-κB (NF-κB). We and others have demonstrated that C. burnetii NMII proteins inhibit expression of pro-inflammatory cytokines and induce expression of anti-apoptotic genes during infection. Here, we demonstrate that C. burnetii promotes intracellular survival by modulating NF-κB subunit p65 (RelA) phosphorylation, and thus activation, in a Type Four B Secretion System (T4BSS)-dependent manner. Immunoblot analysis of RelA phosphorylated at serine-536 demonstrated that C. burnetii increases NF-κB activation via the canonical pathway. However, RelA phosphorylation levels were even higher in infected cells where bacterial protein or mRNA synthesis was inhibited. Importantly, we demonstrate that inhibition of RelA phosphorylation impairs PV formation and C. burnetii growth. We found that a T4BSS-defective mutant (CbΔdotA) elicited phosphorylated RelA levels similar to those of wild type C. burnetii infection treated with Chloramphenicol. Moreover, cells infected with CbΔdotA or wild type C. burnetii treated with Chloramphenicol showed similar levels of GFP-RelA nuclear localization, and significantly increased localization compared to wild type C. burnetii infection. These data indicate that without de novo protein synthesis and a functional T4BSS, C. burnetii is unable to modulate NF-κB activation, which is crucial for optimal intracellular growth.


Asunto(s)
Coxiella burnetii/metabolismo , FN-kappa B/metabolismo , Fiebre Q/microbiología , Factor de Transcripción ReIA/metabolismo , Sistemas de Secreción Tipo IV/metabolismo , Proteínas Bacterianas/metabolismo , Línea Celular/microbiología , Cloranfenicol/farmacología , Coxiella burnetii/efectos de los fármacos , Coxiella burnetii/genética , Coxiella burnetii/crecimiento & desarrollo , Células Epiteliales/microbiología , Células HeLa , Interacciones Huésped-Parásitos , Humanos , Mutación , Subunidad p52 de NF-kappa B/metabolismo , Fosforilación , Fiebre Q/inmunología , ARN Mensajero/biosíntesis , Transducción de Señal , Sistemas de Secreción Tipo IV/genética , Vacuolas/microbiología , Vía de Señalización Wnt
8.
ISME J ; 8(3): 636-649, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24048226

RESUMEN

We investigated the mechanisms of osmoadaptation in the order Halobacteriales, with special emphasis on Haladaptatus paucihalophilus, known for its ability to survive in low salinities. H. paucihalophilus genome contained genes for trehalose synthesis (trehalose-6-phosphate synthase/trehalose-6-phosphatase (OtsAB pathway) and trehalose glycosyl-transferring synthase pathway), as well as for glycine betaine uptake (BCCT family of secondary transporters and QAT family of ABC transporters). H. paucihalophilus cells synthesized and accumulated ∼1.97-3.72 µmol per mg protein of trehalose in a defined medium, with its levels decreasing with increasing salinities. When exogenously supplied, glycine betaine accumulated intracellularly with its levels increasing at higher salinities. RT-PCR analysis strongly suggested that H. paucihalophilus utilizes the OtsAB pathway for trehalose synthesis. Out of 83 Halobacteriales genomes publicly available, genes encoding the OtsAB pathway and glycine betaine BCCT family transporters were identified in 38 and 60 genomes, respectively. Trehalose (or its sulfonated derivative) production and glycine betaine uptake, or lack thereof, were experimentally verified in 17 different Halobacteriales species. Phylogenetic analysis suggested that trehalose synthesis is an ancestral trait within the Halobacteriales, with its absence in specific lineages reflecting the occurrence of gene loss events during Halobacteriales evolution. Analysis of multiple culture-independent survey data sets demonstrated the preference of trehalose-producing genera to saline and low salinity habitats, and the dominance of genera lacking trehalose production capabilities in permanently hypersaline habitats. This study demonstrates that, contrary to current assumptions, compatible solutes production and uptake represent a common mechanism of osmoadaptation within the Halobacteriales.


Asunto(s)
Betaína/metabolismo , Halobacteriales/fisiología , Proteínas Bacterianas/metabolismo , Ecosistema , Perfilación de la Expresión Génica , Glucosiltransferasas/metabolismo , Halobacteriales/clasificación , Halobacteriales/genética , Proteínas de Transporte de Membrana/genética , Datos de Secuencia Molecular , Filogenia , Salinidad , Cloruro de Sodio/metabolismo , Trehalosa/biosíntesis
9.
Vector Borne Zoonotic Dis ; 11(7): 917-22, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21254834

RESUMEN

Q fever, a zoonotic disease, is caused by a gram-negative intracellular bacterium, Coxiella burnetii. Although normally transmitted during exposure to infectious aerosols, C. burnetii is also found in arthropod vectors. In the environment, ticks are thought to play a crucial role in bacterial maintenance and transmission by infecting various mammalian species. However, the nature of the pathogen-tick relationship is not well defined. To determine C. burnetii's interactions with a cultured tick cell line, we introduced purified C. burnetii NMII into Ixodes scapularis-derived IDE8 cells and assayed for bacterial presence, replication, gene expression, and subsequent infectivity for mammalian cells. Tick cells were harvested at 24 h, 72 h, 7 days, and 11 days postinfection (PI). C. burnetii uptake and subsequent replication was demonstrated by indirect immunofluorescence assay, electron microscopy, and real-time polymerase chain reaction (PCR). When a genome equivalent multiplicity of infection of 30 was used, 30%-40% of exposed cells were seen to have small, rounded, vacuoles at 72 h PI, whereas at 7 and 11 days PI, 60%-70% of cells contained enlarged vacuoles harboring large numbers of bacteria. Quantitative PCR analysis of total genomic DNA confirmed that C. burnetii genome numbers increased significantly from 24 h to 11 days PI. Expression of C. burnetii type four secretion system homologs at 7 days PI was demonstrated by reverse transcriptase PCR. Finally, indirect immunofluorescence assay demonstrated that C. burnetii propagated within IDE8 cells were infectious for mammalian cells. These studies demonstrate the utility of cultured tick cell lines as a model to investigate C. burnetii's molecular interactions with its arthropod vectors.


Asunto(s)
Coxiella burnetii/patogenicidad , Ixodes/microbiología , Animales , Vectores Arácnidos/microbiología , Chlorocebus aethiops , Coxiella burnetii/genética , Coxiella burnetii/crecimiento & desarrollo , Técnica del Anticuerpo Fluorescente Indirecta , Reacción en Cadena de la Polimerasa , Células Vero
10.
BMC Microbiol ; 10: 244, 2010 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-20854687

RESUMEN

BACKGROUND: Coxiella burnetii is an intracellular bacterial pathogen that causes acute and chronic disease in humans. Bacterial replication occurs within enlarged parasitophorous vacuoles (PV) of eukaryotic cells, the biogenesis and maintenance of which is dependent on C. burnetii protein synthesis. These observations suggest that C. burnetii actively subverts host cell processes, however little is known about the cellular biology mechanisms manipulated by the pathogen during infection. Here, we examined host cell gene expression changes specifically induced by C. burnetii proteins during infection. RESULTS: We have identified 36 host cell genes that are specifically regulated when de novo C. burnetii protein synthesis occurs during infection using comparative microarray analysis. Two parallel sets of infected and uninfected THP-1 cells were grown for 48 h followed by the addition of chloramphenicol (CAM) to 10 µg/ml in one set. Total RNA was harvested at 72 hpi from all conditions, and microarrays performed using Phalanx Human OneArray slides. A total of 784 (mock treated) and 901 (CAM treated) THP-1 genes were up or down regulated ≥2 fold in the C. burnetii infected vs. uninfected cell sets, respectively. Comparisons between the complementary data sets (using >0 fold), eliminated the common gene expression changes. A stringent comparison (≥2 fold) between the separate microarrays revealed 36 host cell genes modulated by C. burnetii protein synthesis. Ontological analysis of these genes identified the innate immune response, cell death and proliferation, vesicle trafficking and development, lipid homeostasis, and cytoskeletal organization as predominant cellular functions modulated by C. burnetii protein synthesis. CONCLUSIONS: Collectively, these data indicate that C. burnetii proteins actively regulate the expression of specific host cell genes and pathways. This is in addition to host cell genes that respond to the presence of the pathogen whether or not it is actively synthesizing proteins. These findings indicate that C. burnetii modulates the host cell gene expression to avoid the immune response, preserve the host cell from death, and direct the development and maintenance of a replicative PV by controlling vesicle formation and trafficking within the host cell during infection.


Asunto(s)
Proteínas Bacterianas/biosíntesis , Coxiella burnetii/crecimiento & desarrollo , Interacciones Huésped-Patógeno , Monocitos/metabolismo , Muerte Celular , Línea Celular , Proliferación Celular , Cloranfenicol/farmacología , Coxiella burnetii/inmunología , Perfilación de la Expresión Génica , Regulación Bacteriana de la Expresión Génica/efectos de los fármacos , Humanos , Inmunidad Innata/genética , Monocitos/microbiología , Análisis por Matrices de Proteínas , Vacuolas/microbiología , Vacuolas/fisiología
11.
FEMS Microbiol Lett ; 311(1): 61-9, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20727011

RESUMEN

Analysis of the Coxiella burnetii RSA 493 (Nine Mile phase I strain) genome revealed ORFs with significant homology to the type IVB secretion system (T4BSS) of Legionella pneumophila. The T4BSS genes exist primarily at two loci, designated regions I (RI) and II. In C. burnetii, little is known about the T4BSS regions and the role they play in establishing and/or maintaining infection. Coxiella burnetii T4BSS RI contains genes arranged in three linkage groups: (1) icmW→CBU1651→icmX, (2) icmV→dotA→CBU1647, and (3) icmT→icmS→dotD→dotC→dotB→CBU1646. We used reverse transcriptase (RT)-PCR to demonstrate transcriptional linkage within the groups, and that icmX, icmV, and icmT are transcribed de novo by 8 h post infection (hpi). We then examined the transcript levels for icmX, icmW, icmV, dotA, dotB, and icmT during the first 24 h of an infection using quantitative RT-PCR. The expression initially increased for each gene, followed by a decrease at 24 hpi. Subsequently, we analyzed IcmT protein levels during infection and determined that the expression increases significantly from 8 to 24 hpi and then remains relatively constant. These data demonstrate temporal changes in the RNA of several C. burnetii T4SS RI homologs and the IcmT protein. These changes correspond to early stages of the C. burnetii infectious cycle.


Asunto(s)
Proteínas Bacterianas/genética , Coxiella burnetii/fisiología , Regulación Bacteriana de la Expresión Génica , Fiebre Q/microbiología , Animales , Proteínas Bacterianas/metabolismo , Chlorocebus aethiops , Coxiella burnetii/genética , Interacciones Huésped-Patógeno , Humanos , Células Vero
12.
FEMS Microbiol Lett ; 305(2): 177-83, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20199576

RESUMEN

Coxiella burnetii is a Gram-negative pleomorphic bacterium and the causative agent of Q fever. During infection, the pathogen survives and replicates within a phagosome-like parasitophorous vacuole while influencing cellular functions throughout the host cell, indicating a capacity for effector protein secretion. Analysis of the C. burnetii (RSA 493 strain) genome sequence indicates that C. burnetii contains genes with homology to the Legionella pneumophila Dot/Icm type IVB secretion system (T4BSS). T4BSSs have only been described in L. pneumophila and C. burnetii, marking it a unique virulence determinate. Characterization of bacterial virulence determinants ranging from autotransporter proteins to diverse secretion systems suggests that polar localization may be a virulence mechanism hallmark. To characterize T4BSS subcellular localization in C. burnetii, we analyzed C. burnetii-infected Vero cells by indirect immunofluorescent antibody (IFA) and immunoelectron microscopy (IEM). Using antibodies against the C. burnetii T4BSS homologs IcmT, IcmV, and DotH, IFA show that these proteins are localized to the poles of the bacterium. IEM supports this finding, showing that antibodies against C. burnetii IcmT and DotH preferentially localize to the bacterial cell pole(s). Together, these data demonstrate that the C. burnetii T4BSS localizes to the pole(s) of the bacterium during infection of host cells.


Asunto(s)
Proteínas Bacterianas/análisis , Coxiella burnetii/química , Sustancias Macromoleculares/análisis , Proteínas de Transporte de Membrana/análisis , Factores de Virulencia/análisis , Animales , Chlorocebus aethiops , Microscopía Fluorescente , Microscopía Inmunoelectrónica , Células Vero
13.
Anal Chim Acta ; 583(1): 23-31, 2007 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-17386522

RESUMEN

Accurate bacterial identification is important in diagnosing disease and in microbial forensics. Coxiella burnetii, a highly infective microorganism causative of the human disease Q fever, is now considered a U.S. category B potential bioterrorism agent. We report here an approach for the confirmatory identification of C. burnetii at the strain level which involves the combined use of matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF MS) and supervised pattern recognition via Partial Least Squares-Discriminant Analysis (PLS-DA). C. burnetii isolates investigated in this study included the following prototype strains from different geographical and/or historical origins and with different antigenic properties: Nine Mile I, Australian QD, M44, KAV, PAV, Henzerling, and Ohio. After culture and purification following standard protocols, linear MALDI-TOF mass spectra of pure bacterial cultures were acquired in positive ion mode. Mass spectral data were normalized, baseline-corrected, denoised, binarized and modeled by PLS-DA under crossvalidation conditions. Robustness with respect to uncontrolled variations in the sample preparation and MALDI analysis protocol was assessed by repeating the experiment on five different days spanning a period of 6 months. The method was validated by the prediction of unknown C. burnetii samples in an independent test set with 100% sensitivity and specificity for five out of six strain classes.


Asunto(s)
Coxiella burnetii/aislamiento & purificación , Animales , Bovinos , Coxiella burnetii/clasificación , Análisis Discriminante , Geografía , Humanos , Análisis de los Mínimos Cuadrados , Análisis Multivariante , Reconocimiento de Normas Patrones Automatizadas , Fiebre Q/diagnóstico , Especificidad de la Especie , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
14.
Vector Borne Zoonotic Dis ; 5(1): 90-1, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-15815154

RESUMEN

Prevalence of antibodies to Coxiella burnetii in 24 veterinary school-associated dairy herds in the United States was assessed through laboratory testing of bulk tank milk specimens by indirect immunofluorescent antibody assay. Twenty-two herds (92%) had evidence of antibodies to C. burnetii Phase I antibodies at a titer of > or = 1:16, and nine herds (38%) had Phase I antibody titers of > or = 1:256. These results suggest that C. burnetii infection is geographically widespread among dairy herds in the United States.


Asunto(s)
Anticuerpos Antibacterianos/análisis , Enfermedades de los Bovinos/epidemiología , Coxiella burnetii/inmunología , Fiebre Q/veterinaria , Animales , Bovinos , Femenino , Técnica del Anticuerpo Fluorescente Indirecta/veterinaria , Leche/inmunología , Fiebre Q/epidemiología , Estudios Seroepidemiológicos , Estados Unidos/epidemiología
15.
Anal Chem ; 76(14): 4017-22, 2004 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-15253637

RESUMEN

Matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF MS) specific biomarkers have been shown to be an effective tool for identifying microorganisms. In this study, we demonstrate the feasibility of using this technique to detect the obligate intracellular bacterium Coxiella burnetii, a category B bioterrorism agent. Specific biomarkers were detected in C. burnetii Nine Mile phase I (NMI) strain purified from embryonated egg yolk sac preparations. Whole organisms were applied directly to the MALDI target. MALDI-TOF MS analysis of C. burnetii NMI grown and purified at different times and places revealed a group of unique, characteristic, and reproducible spectral markers in the mass range of 1000-25000 Da. Statistical analysis of the averaged centroided masses uncovered at least 24 peptides or biomarkers. Three biomarkers observed in the MALDI-TOF MS spectrum consistently matched proteins that had been previously described in C. burnetii, one of them being the small cell variant protein A. MALDI-TOF MS analysis of whole organisms represents a sensitive and specific option for characterizing C. burnetii isolates, especially when coupled with antigen capture techniques. The method also has potential for several applications in basic microbial research, including regulation of gene expression.


Asunto(s)
Coxiella burnetii/aislamiento & purificación , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción/métodos , Animales , Proteínas Bacterianas/análisis , Biomarcadores/análisis , Embrión de Pollo , Huevos/microbiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA