Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
1.
Infect Immun ; 92(3): e0056022, 2024 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-38363133

RESUMO

The Gram-negative bacterium Coxiella burnetii is the causative agent of query fever in humans and coxiellosis in livestock. C. burnetii infects a variety of cell types, tissues, and animal species including mammals and arthropods, but there is much left to be understood about the molecular mechanisms at play during infection in distinct species. Human stimulator of interferon genes (STING) induces an innate immune response through the induction of type I interferons (IFNs), and IFN promotes or suppresses C. burnetii replication, depending on tissue type. Drosophila melanogaster contains a functional STING ortholog (Sting) which activates NF-κB signaling and autophagy. Here, we sought to address the role of D. melanogaster Sting during C. burnetii infection to uncover how Sting regulates C. burnetii infection in flies. We show that Sting-null flies exhibit higher mortality and reduced induction of antimicrobial peptides following C. burnetii infection compared to control flies. Additionally, Sting-null flies induce lower levels of oxidative stress genes during infection, but the provision of N-acetyl-cysteine (NAC) in food rescues Sting-null host survival. Lastly, we find that reactive oxygen species levels during C. burnetii infection are higher in Drosophila S2 cells knocked down for Sting compared to control cells. Our results show that at the host level, NAC provides protection against C. burnetii infection in the absence of Sting, thus establishing a role for Sting in protection against oxidative stress during C. burnetii infection.


Assuntos
Coxiella burnetii , Febre Q , Animais , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , NF-kappa B/metabolismo , Febre Q/microbiologia , Espécies Reativas de Oxigênio/metabolismo
2.
Infect Immun ; 89(12): e0013521, 2021 11 16.
Artigo em Inglês | MEDLINE | ID: mdl-34491791

RESUMO

Coxiella burnetii, the causative agent of query (Q) fever in humans, is an obligate intracellular bacterium. C. burnetii can naturally infect a broad range of host organisms (e.g., mammals and arthropods) and cell types. This amphotropic nature of C. burnetii, in combination with its ability to utilize both glycolytic and gluconeogenic carbon sources, suggests that the pathogen relies on metabolic plasticity to replicate in nutritionally diverse intracellular environments. To test the significance of metabolic plasticity in C. burnetii host cell colonization, C. burnetii intracellular replication in seven distinct cell lines was compared between a metabolically competent parental strain and a mutant, CbΔpckA, unable to undergo gluconeogenesis. Both the parental strain and CbΔpckA mutant exhibited host cell-dependent infection phenotypes, which were influenced by alterations to host glycolytic or gluconeogenic substrate availability. Because the nutritional environment directly impacts host cell physiology, our analysis was extended to investigate the response of C. burnetii replication in mammalian host cells cultivated in a novel physiological medium based on the nutrient composition of mammalian interstitial fluid, interstitial fluid-modeled medium (IFmM). An infection model based on IFmM resulted in exacerbation of a replication defect exhibited by the CbΔpckA mutant in specific cell lines. The CbΔpckA mutant was also attenuated during infection of an animal host. Overall, the study underscores that gluconeogenic capacity aids C. burnetii amphotropism and that the amphotropic nature of C. burnetii should be considered when resolving virulence mechanisms in this pathogen.


Assuntos
Coxiella burnetii/fisiologia , Metabolismo Energético , Interações Hospedeiro-Patógeno , Febre Q/metabolismo , Febre Q/microbiologia , Suscetibilidade a Doenças , Gluconeogênese , Glicólise , Humanos , Virulência/genética , Fatores de Virulência/genética
3.
Infect Immun ; 88(12)2020 11 16.
Artigo em Inglês | MEDLINE | ID: mdl-32928965

RESUMO

Coxiella burnetii is a zoonotic bacterial obligate intracellular parasite and the cause of query (Q) fever. During natural infection of female animals, C. burnetii shows tropism for the placenta and is associated with late-term abortion, at which time the pathogen titer in placental tissue can exceed one billion bacteria per gram. During later stages of pregnancy, placental trophoblasts serve as the major source of progesterone, a steroid hormone known to affect the replication of some pathogens. During infection of placenta-derived JEG-3 cells, C. burnetii showed sensitivity to progesterone but not the immediate precursor pregnenolone or estrogen, another major mammalian steroid hormone. Using host cell-free culture, progesterone was determined to have a direct inhibitory effect on C. burnetii replication. Synergy between the inhibitory effect of progesterone and the efflux pump inhibitors verapamil and 1-(1-naphthylmethyl)-piperazine is consistent with a role for efflux pumps in preventing progesterone-mediated inhibition of C. burnetii activity. The sensitivity of C. burnetii to progesterone, but not structurally related molecules, is consistent with the ability of progesterone to influence pathogen replication in progesterone-producing tissues.


Assuntos
Coxiella burnetii/efeitos dos fármacos , Coxiella burnetii/crescimento & desenvolvimento , Interações Hospedeiro-Patógeno , Placenta/microbiologia , Progesterona/farmacologia , Animais , Proteínas de Bactérias/química , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Replicação do DNA/efeitos dos fármacos , Sinergismo Farmacológico , Proteínas de Escherichia coli/química , Estrogênios/farmacologia , Etídio/química , Feminino , Humanos , Proteínas de Membrana/química , Proteínas de Membrana Transportadoras/química , Proteínas de Membrana Transportadoras/efeitos dos fármacos , Proteínas de Membrana Transportadoras/metabolismo , Piperazinas/farmacologia , Placenta/efeitos dos fármacos , Gravidez , Pregnenolona/farmacologia , Proteínas Quinases/química , Verapamil/farmacologia
4.
J Bacteriol ; 200(14)2018 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-29735758

RESUMO

Bacteria of the genus Chlamydia include the significant human pathogens Chlamydia trachomatis and C. pneumoniae All chlamydiae are obligate intracellular parasites that depend on infection of a host cell and transition through a biphasic developmental cycle. Following host cell invasion by the infectious elementary body (EB), the pathogen transitions to the replicative but noninfectious reticulate body (RB). Differentiation of the RB back to the EB is essential to generate infectious progeny. While the EB form has historically been regarded as metabolically inert, maintenance of infectivity during incubation with specific nutrients has revealed active maintenance of the infectious phenotype. Using transcriptome sequencing, we show that the transcriptome of extracellular EBs incubated under metabolically stimulating conditions does not cluster with germinating EBs but rather with the transcriptome of EBs isolated directly from infected cells. In addition, the transcriptional profile of the extracellular metabolizing EBs more closely resembled that of EB production than germination. Maintenance of infectivity of extracellular EBs was achieved by metabolizing chemically diverse compounds, including glucose 6-phosphate, ATP, and amino acids, all of which can be found in extracellular environments, including mucosal secretions. We further show that the EB cell type actively maintains infectivity in the inclusion after terminal differentiation. Overall, these findings contribute to the emerging understanding that the EB cell form is actively maintained through metabolic processes after terminal differentiation to facilitate prolonged infectivity within the inclusion and under host cell free conditions, for example, following deposition at mucosal surfaces.IMPORTANCE Chlamydiae are obligate intracellular Gram-negative bacteria that are responsible for a wide range of diseases in both animal and human hosts. According to the Centers for Disease Control and Prevention, C. trachomatis is the most frequently reported sexually transmitted infection in the United States, costing the American health care system nearly $2.4 billion annually. Every year, there are over 4 million new cases of Chlamydia infections in the United States and an estimated 100 million cases worldwide. To cause disease, Chlamydia must successfully complete its complex biphasic developmental cycle, alternating between an infectious cell form (EB) specialized for initiating entry into target cells and a replicative form (RB) specialized for creating and maintaining the intracellular replication niche. The EB cell form has historically been considered metabolically quiescent, a passive entity simply waiting for contact with a host cell to initiate the next round of infection. Recent studies and data presented here demonstrate that the EB maintains its infectious phenotype by actively metabolizing a variety of nutrients. Therefore, the EB appears to have an active role in chlamydial biology, possibly within multiple environments, such as mucosal surfaces, fomites, and inside the host cell after formation.


Assuntos
Proteínas de Bactérias/metabolismo , Chlamydia trachomatis/metabolismo , Transcriptoma , Animais , Proteínas de Bactérias/genética , Linhagem Celular , Chlamydia trachomatis/genética , Citoplasma , DNA Bacteriano , Regulação Bacteriana da Expressão Gênica/fisiologia , Genoma Bacteriano , Camundongos , RNA Bacteriano/genética , Análise de Sequência de RNA
5.
Appl Environ Microbiol ; 83(19)2017 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-28733283

RESUMO

Biofilm-associated infections are a clinical challenge, in part because a hydrated matrix protects the bacterial community from antibiotics. Herein, we evaluated how different osmotic compounds (maltodextrin, sucrose, and polyethylene glycol [PEG]) enhance antibiotic efficacy against Acinetobacter baumannii biofilm communities. Established (24-h) test tube biofilms (strain ATCC 17978) were treated with osmotic compounds in the presence or absence of 10× the MIC of different antibiotics (50 µg/ml tobramycin, 20 µg/ml ciprofloxacin, 300 µg/ml chloramphenicol, 30 µg/ml nalidixic acid, or 100 µg/ml erythromycin). Combining antibiotics with hypertonic concentrations of the osmotic compounds for 24 h reduced the number of biofilm bacteria by 5 to 7 log (P < 0.05). Increasing concentrations of osmotic compounds improved the effect, but there was a trade-off with increasing solution viscosity, whereby low-molecular-mass compounds (sucrose, 400-Da PEG) worked better than higher-mass compounds (maltodextrin, 3,350-Da PEG). Ten other A. baumannii strains were similarly treated with 400-Da PEG and tobramycin, resulting in a mean 2.7-log reduction in recoverable bacteria compared with tobramycin treatment alone. Multivariate regression models with data from different osmotic compounds and nine antibiotics demonstrated that the benefit from combining hypertonic treatments with antibiotics is a function of antibiotic mass and lipophilicity (r2 > 0.82; P < 0.002), and the relationship was generalizable for biofilms formed by A. baumannii and Escherichia coli K-12. Augmenting topical antibiotic therapies with a low-mass hypertonic treatment may enhance the efficacy of antibiotics against wound biofilms, particularly when using low-mass hydrophilic antibiotics.IMPORTANCE Biofilms form a barrier that protects bacteria from environmental insults, including exposure to antibiotics. We demonstrated that multiple osmotic compounds can enhance antibiotic efficacy against Acinetobacter baumannii biofilm communities, but viscosity is a limiting factor, and the most effective compounds have lower molecular mass. The synergism between osmotic compounds and antibiotics is also dependent on the hydrophobicity and mass of the antibiotics. The statistical models presented herein provide a basis for predicting the optimal combination of osmotic compounds and antibiotics against surface biofilms communities.


Assuntos
Acinetobacter baumannii/efeitos dos fármacos , Antibacterianos/farmacologia , Biofilmes/efeitos dos fármacos , Acinetobacter baumannii/crescimento & desenvolvimento , Acinetobacter baumannii/fisiologia , Ciprofloxacina/farmacologia , Testes de Sensibilidade Microbiana , Osmose , Polietilenoglicóis/farmacologia , Polissacarídeos/farmacologia , Sacarose/farmacologia , Tobramicina/farmacologia
6.
Annu Rev Microbiol ; 65: 111-28, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21639786

RESUMO

For over seven decades, Coxiella burnetii, the causative agent of human Q fever, has been considered a prototypical obligate intracellular bacterium that relies exclusively on a eukaryotic cell for growth. Intracellularly, the organism prospers in an acidified, phagolysosome-like vacuole. C. burnetii has evolved to replicate in this harsh compartment by a mechanism involving acid activation of metabolism. The ?2 Mb genome of C. burnetii is about twice the size of genomes of most obligate intracellular bacteria, and the organism's central metabolic pathways are largely intact. The absence of extensive genome reduction suggests the adaptation of C. burnetii to an obligate intracellular lifestyle is a recent evolutionary event. Indeed, insight from early work on C. burnetii metabolism, along with new information gained from metabolic pathway reconstructions, nutrient typing, and expression profiling, allowed the rescue of C. burnetii from its host cell to regain the axenic growth capacity of its ancestors. This advance removes the extensive experimental obstacles associated with intracellular obligatism and opens the door for a renaissance in C. burnetii research.


Assuntos
Coxiella burnetii/crescimento & desenvolvimento , Interações Hospedeiro-Patógeno , Febre Q/microbiologia , Animais , Evolução Biológica , Coxiella burnetii/genética , Humanos
7.
Proc Natl Acad Sci U S A ; 109(48): 19781-5, 2012 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-23129646

RESUMO

Chlamydia trachomatis is among the most clinically significant human pathogens, yet their obligate intracellular nature places severe restrictions upon research. Chlamydiae undergo a biphasic developmental cycle characterized by an infectious cell type known as an elementary body (EB) and an intracellular replicative form called a reticulate body (RB). EBs have historically been described as metabolically dormant. A cell-free (axenic) culture system was developed, which showed high levels of metabolic and biosynthetic activity from both EBs and RBs, although the requirements differed for each. EBs preferentially used glucose-6-phosphate as an energy source, whereas RBs required ATP. Both developmental forms showed increased activity when incubated under microaerobic conditions. Incorporation of isotopically labeled amino acids into proteins from both developmental forms indicated unique expression profiles, which were confirmed by genome-wide transcriptional analysis. The described axenic culture system will greatly enhance biochemical and physiological analyses of chlamydiae.


Assuntos
Chlamydia trachomatis/fisiologia , Transcrição Gênica/fisiologia , Chlamydia trachomatis/metabolismo , Chlamydia trachomatis/ultraestrutura , Meios de Cultura , Microscopia Eletrônica de Transmissão , Biossíntese de Proteínas
8.
Front Cell Infect Microbiol ; 14: 1284701, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38585652

RESUMO

Bacterial obligate intracellular parasites (BOIPs) represent an exclusive group of bacterial pathogens that all depend on invasion of a eukaryotic host cell to reproduce. BOIPs are characterized by extensive adaptation to their respective replication niches, regardless of whether they replicate within the host cell cytoplasm or within specialized replication vacuoles. Genome reduction is also a hallmark of BOIPs that likely reflects streamlining of metabolic processes to reduce the need for de novo biosynthesis of energetically costly metabolic intermediates. Despite shared characteristics in lifestyle, BOIPs show considerable diversity in nutrient requirements, metabolic capabilities, and general physiology. In this review, we compare metabolic and physiological processes of prominent pathogenic BOIPs with special emphasis on carbon, energy, and amino acid metabolism. Recent advances are discussed in the context of historical views and opportunities for discovery.


Assuntos
Parasitos , Animais , Bactérias/genética , Vacúolos , Células Eucarióticas
9.
Nat Commun ; 15(1): 697, 2024 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-38267444

RESUMO

Lipopolysaccharide (LPS) phase variation is a critical aspect of virulence in many Gram-negative bacteria. It is of particular importance to Coxiella burnetii, the biothreat pathogen that causes Q fever, as in vitro propagation of this organism leads to LPS truncation, which is associated with an attenuated and exempted from select agent status (Nine Mile II, NMII). Here, we demonstrate that NMII was recovered from the spleens of infected guinea pigs. Moreover, these strains exhibit a previously unrecognized form of elongated LPS and display increased virulence in comparison with the initial NMII strain. The reversion of a 3-bp mutation in the gene cbu0533 directly leads to LPS elongation. To address potential safety concerns, we introduce a modified NMII strain unable to produce elongated LPS.


Assuntos
Coxiella burnetii , Animais , Cobaias , Coxiella burnetii/genética , Lipopolissacarídeos , Mutação , Reprodução , Baço
10.
BMC Microbiol ; 13: 222, 2013 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-24093460

RESUMO

BACKGROUND: Coxiella burnetii is a Gram-negative intracellular bacterial pathogen that replicates within a phagolysosome-like parasitophorous vacuole (PV) of macrophages. PV formation requires delivery of effector proteins directly into the host cell cytoplasm by a type IVB secretion system. However, additional secretion systems are likely responsible for modification of the PV lumen microenvironment that promote pathogen replication. RESULTS: To assess the potential of C. burnetii to secrete proteins into the PV, we analyzed the protein content of modified acidified citrate cysteine medium for the presence of C. burnetii proteins following axenic (host cell-free) growth. Mass spectrometry generated a list of 105 C. burnetii proteins that could be secreted. Based on bioinformatic analysis, 55 proteins were selected for further study by expressing them in C. burnetii with a C-terminal 3xFLAG-tag. Secretion of 27 proteins by C. burnetii transformants was confirmed by immunoblotting culture supernatants. Tagged proteins expressed by C. burnetii transformants were also found in the soluble fraction of infected Vero cells, indicating secretion occurs ex vivo. All secreted proteins contained a signal sequence, and deletion of this sequence from selected proteins abolished secretion. These data indicate protein secretion initially requires translocation across the inner-membrane into the periplasm via the activity of the Sec translocase. CONCLUSIONS: C. burnetii secretes multiple proteins, in vitro and ex vivo, in a Sec-dependent manner. Possible roles for secreted proteins and secretion mechanisms are discussed.


Assuntos
Proteínas de Bactérias/metabolismo , Coxiella burnetii/metabolismo , Redes e Vias Metabólicas , Animais , Proteínas de Bactérias/genética , Chlorocebus aethiops , Biologia Computacional , Meios de Cultura/química , Espectrometria de Massas , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Sinais Direcionadores de Proteínas , Transporte Proteico , Deleção de Sequência , Células Vero
11.
Adv Exp Med Biol ; 984: 215-29, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22711634

RESUMO

Early metabolic studies of C. burnetii showed minimal metabolic activity of axenic (host cell-free) organisms in buffers adjusted to neutral pH. However, our understanding of the organism's physiology was greatly improved upon the discovery that C. burnetii requires an acidic pH for metabolic activation. Indeed, information gained from acid activation studies coupled with contemporary analyses using transcription microarrays, metabolic pathway reconstruction and metabolite typing, led to an axenic culture system that supports robust growth of C. burnetii. While axenic culture of C. burnetii can present some technical challenges, the technique is currently facilitating new lines of investigation and development of genetic tools. Axenic cultivation of C. burnetii should also prove useful in clinical settings.


Assuntos
Cultura Axênica/métodos , Coxiella burnetii/fisiologia , Soluções Tampão , Coxiella burnetii/genética , Coxiella burnetii/crescimento & desenvolvimento , Coxiella burnetii/metabolismo , Meios de Cultura , Concentração de Íons de Hidrogênio
12.
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
13.
Pathog Dis ; 80(1)2022 05 23.
Artigo em Inglês | MEDLINE | ID: mdl-35388904

RESUMO

Chlamydia trachomatis is a bacterial obligate intracellular parasite and a significant cause of human disease, including sexually transmitted infections and trachoma. The bacterial RNA polymerase-binding protein DksA is a transcription factor integral to the multicomponent bacterial stress response pathway known as the stringent response. The genome of C. trachomatis encodes a DksA ortholog (DksACt) that is maximally expressed at 15-20 h post infection, a time frame correlating with the onset of transition between the replicative reticulate body (RB) and infectious elementary body (EB) forms of the pathogen. Ectopic overexpression of DksACt in C. trachomatis prior to RB-EB transitions during infection of HeLa cells resulted in a 39.3% reduction in overall replication (yield) and a 49.6% reduction in recovered EBs. While the overall domain organization of DksACt is similar to the DksA ortholog of Escherichia coli (DksAEc), DksACt did not functionally complement DksAEc. Transcription of dksACt is regulated by tandem promoters, one of which also controls expression of nrdR, encoding a negative regulator of deoxyribonucleotide biosynthesis. The phenotype resulting from ectopic expression of DksACt and the correlation between dksACt and nrdR expression is consistent with a role for DksACt in the C. trachomatis developmental cycle.


Assuntos
Infecções por Chlamydia , Proteínas de Escherichia coli , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Chlamydia trachomatis/genética , Chlamydia trachomatis/metabolismo , Escherichia coli/metabolismo , Proteínas de Escherichia coli/genética , Regulação Bacteriana da Expressão Gênica , Células HeLa , Humanos
14.
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
15.
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
16.
Pathog Dis ; 79(6)2021 07 20.
Artigo em Inglês | MEDLINE | ID: mdl-34259815

RESUMO

Coxiella burnetii is a bacterial obligate intracellular parasite and the etiological agent of query (Q) fever. While the C. burnetii genome has been reduced to ∼2 Mb as a likely consequence of genome streamlining in response to parasitism, enzymes for a nearly complete central metabolic machinery are encoded by the genome. However, lack of a canonical hexokinase for phosphorylation of glucose and an apparent absence of the oxidative branch of the pentose phosphate pathway, a major mechanism for regeneration of the reducing equivalent nicotinamide adenine dinucleotide phosphate (NADPH), have been noted as potential metabolic limitations of C. burnetii. By complementing C. burnetii with the gene zwf encoding the glucose-6-phosphate-consuming and NADPH-producing enzyme glucose-6-phosphate dehydrogenase (G6PD), we demonstrate a severe metabolic fitness defect for C. burnetii under conditions of glucose limitation. Supplementation of the medium with the gluconeogenic carbon source glutamate did not rescue the growth defect of bacteria complemented with zwf. Absence of G6PD in C. burnetii therefore likely relates to the negative effect of its activity under conditions of glucose limitation. Coxiella burnetii central metabolism with emphasis on glucose, NAD+, NADP+ and NADPH is discussed in a broader perspective, including comparisons with other bacterial obligate intracellular parasites.


Assuntos
Coxiella burnetii/enzimologia , Coxiella burnetii/fisiologia , Glucosefosfato Desidrogenase/genética , Glucosefosfato Desidrogenase/metabolismo , NADP/metabolismo , Febre Q/microbiologia , Animais , Linhagem Celular , Chlorocebus aethiops , DNA Bacteriano , Glucose/metabolismo , Glucose-6-Fosfato/metabolismo , Interações Hospedeiro-Patógeno , Humanos , Redes e Vias Metabólicas , Aptidão Física , Células Vero
17.
Genetics ; 217(3)2021 03 31.
Artigo em Inglês | MEDLINE | ID: mdl-33789347

RESUMO

The gram-negative bacterium Coxiella burnetii is the causative agent of Query (Q) fever in humans and coxiellosis in livestock. Host genetics are associated with C. burnetii pathogenesis both in humans and animals; however, it remains unknown if specific genes are associated with severity of infection. We employed the Drosophila Genetics Reference Panel to perform a genome-wide association study to identify host genetic variants that affect host survival to C. burnetii infection. The genome-wide association study identified 64 unique variants (P < 10-5) associated with 25 candidate genes. We examined the role each candidate gene contributes to host survival during C. burnetii infection using flies carrying a null mutation or RNAi knockdown of each candidate. We validated 15 of the 25 candidate genes using at least one method. This is the first report establishing involvement of many of these genes or their homologs with C. burnetii susceptibility in any system. Among the validated genes, FER and tara play roles in the JAK/STAT, JNK, and decapentaplegic/TGF-ß signaling pathways which are components of known innate immune responses to C. burnetii infection. CG42673 and DIP-ε play roles in bacterial infection and synaptic signaling but have no previous association with C. burnetii pathogenesis. Furthermore, since the mammalian ortholog of CG13404 (PLGRKT) is an important regulator of macrophage function, CG13404 could play a role in host susceptibility to C. burnetii through hemocyte regulation. These insights provide a foundation for further investigation regarding the genetics of C. burnetii susceptibility across a wide variety of hosts.


Assuntos
Resistência à Doença , Variação Genética , Febre Q/genética , Locos de Características Quantitativas , Animais , Proteínas de Ciclo Celular/genética , Coxiella burnetii/patogenicidade , Proteínas de Drosophila/genética , Drosophila melanogaster , Proteínas do Olho/genética , Patrimônio Genético , Febre Q/microbiologia
18.
Front Cell Infect Microbiol ; 11: 692224, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34368013

RESUMO

The chlamydial small non coding RNA, IhtA, regulates the expression of both HctA and DdbA, the uncharacterized product of the C. trachomatis L2 CTL0322 gene. HctA is a small, highly basic, DNA binding protein that is expressed late in development and mediates the condensation of the genome during RB to EB differentiation. DdbA is conserved throughout the chlamydial lineage, and is predicted to express a small, basic, cytoplasmic protein. As it is common for sRNAs to regulate multiple mRNAs within the same physiological pathway, we hypothesize that DdbA, like HctA, is involved in RB to EB differentiation. Here, we show that DdbA is a DNA binding protein, however unlike HctA, DdbA does not contribute to genome condensation but instead likely has nuclease activity. Using a DdbA temperature sensitive mutant, we show that DdbAts creates inclusions indistinguishable from WT L2 in size and that early RB replication is likewise similar at the nonpermissive temperature. However, the number of DdbAts infectious progeny is dramatically lower than WT L2 overall, although production of EBs is initiated at a similar time. The expression of a late gene reporter construct followed live at 40°C indicates that late gene expression is severely compromised in the DdbAts strain. Viability assays, both in host cells and in axenic media indicate that the DdbAts strain is defective in the maintenance of EB infectivity. Additionally, using Whole Genome Sequencing we demonstrate that chromosome condensation is temporally separated from DNA replication during the RB to EB transition. Although DdbA does not appear to be directly involved in this process, our data suggest it is a DNA binding protein that is important in the production and maintenance of infectivity of the EB, perhaps by contributing to the remodeling of the EB chromosome.


Assuntos
Chlamydia trachomatis , Regulação Bacteriana da Expressão Gênica , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Chlamydia trachomatis/genética , Chlamydia trachomatis/metabolismo , Proteínas de Ligação a DNA/genética , Corpos de Inclusão/metabolismo
19.
Ticks Tick Borne Dis ; 12(1): 101584, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33059171

RESUMO

For most organisms, iron is an essential nutrient due to its role in fundamental cellular processes. Insufficient iron causes sub-optimal metabolism with potential effects on viability, while high levels of iron are toxic due to the formation of oxidative radicals, which damage cellular components. Many molecules and processes employed in iron uptake, storage, transport and metabolism are conserved, however significant knowledge gaps remain regarding these processes in ticks due to their unique physiology. In this study, we first identified and sequenced 13 genes likely to be involved in iron metabolism in Dermacentor andersoni cells. We then developed a method to reduce iron levels in D. andersoni cells using the iron chelator 2,2'-bipyridyl and measured the transcriptional response of these genes to iron reduction. The genes include a putative transferrin receptor, divalent metal transporter 1, duodenal cytochrome b, zinc/iron transporters zip7, zip13, zip14, mitoferrin, ferrochelatase, iron regulatory protein 1, ferritin1, ferritin2, transferrin and poly r(C)-binding protein. Overall, the transcriptional response of the target genes to iron reduction was modest. The most marked changes were a decrease in ferritin2, which transports iron through the tick hemolymph, the mitochondrial iron transporter mitoferrin, and the mitochondrial enzyme ferrochelatase. Iron regulatory protein1 was the only gene with an overall increase in transcript in response to reduced iron levels. This work lays the foundation for an improved understanding of iron metabolism in ticks which may provide molecular targets for the development of novel tick control methods and aid in the understanding of tick-pathogen interactions.


Assuntos
Proteínas de Artrópodes/genética , Dermacentor/genética , Ferro/metabolismo , Sequência de Aminoácidos , Animais , Proteínas de Artrópodes/química , Proteínas de Artrópodes/metabolismo , Dermacentor/metabolismo , Perfilação da Expressão Gênica , Alinhamento de Sequência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA