Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 143
Filtrar
1.
Virulence ; 15(1): 2350893, 2024 12.
Artículo en Inglés | MEDLINE | ID: mdl-38725096

RESUMEN

Coxiella burnetii (C. burnetii) is the causative agent of Q fever, a zoonotic disease. Intracellular replication of C. burnetii requires the maturation of a phagolysosome-like compartment known as the replication permissive Coxiella-containing vacuole (CCV). Effector proteins secreted by the Dot/Icm secretion system are indispensable for maturation of a single large CCV by facilitating the fusion of promiscuous vesicles. However, the mechanisms of CCV maintenance and evasion of host cell clearance remain to be defined. Here, we show that C. burnetii secreted Coxiella vacuolar protein E (CvpE) contributes to CCV biogenesis by inducing lysosome-like vacuole (LLV) enlargement. LLV fission by tubulation and autolysosome degradation is impaired in CvpE-expressing cells. Subsequently, we found that CvpE suppresses lysosomal Ca2+ channel transient receptor potential channel mucolipin 1 (TRPML1) activity in an indirect manner, in which CvpE binds phosphatidylinositol 3-phosphate [PI(3)P] and perturbs PIKfyve activity in lysosomes. Finally, the agonist of TRPML1, ML-SA5, inhibits CCV biogenesis and C. burnetii replication. These results provide insight into the mechanisms of CCV maintenance by CvpE and suggest that the agonist of TRPML1 can be a novel potential treatment that does not rely on antibiotics for Q fever by enhancing Coxiella-containing vacuoles (CCVs) fission.


Asunto(s)
Proteínas Bacterianas , Coxiella burnetii , Lisosomas , Fosfatidilinositol 3-Quinasas , Fosfatos de Fosfatidilinositol , Canales de Potencial de Receptor Transitorio , Vacuolas , Coxiella burnetii/metabolismo , Coxiella burnetii/crecimiento & desarrollo , Coxiella burnetii/genética , Vacuolas/microbiología , Vacuolas/metabolismo , Lisosomas/metabolismo , Lisosomas/microbiología , Fosfatos de Fosfatidilinositol/metabolismo , Humanos , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética , Canales de Potencial de Receptor Transitorio/metabolismo , Canales de Potencial de Receptor Transitorio/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Animales , Fiebre Q/microbiología , Células HeLa , Interacciones Huésped-Patógeno
2.
mSphere ; 6(4): e0044221, 2021 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-34232075

RESUMEN

Coxiella burnetii is a highly infectious, intracellular, Gram-negative bacterial pathogen that causes human Q fever, an acute flu-like illness that can progress to chronic endocarditis. C. burnetii is transmitted to humans via aerosols and has long been considered a potential biological warfare agent. Although antibiotics, such as doxycycline, effectively treat acute Q fever, a recently identified antibiotic-resistant strain demonstrates the ability of C. burnetii to resist traditional antimicrobials, and chronic disease is extremely difficult to treat with current options. These findings highlight the need for new Q fever therapeutics, and repurposed drugs that target eukaryotic functions to prevent bacterial replication are of increasing interest in infectious disease. To identify this class of anti-C. burnetii therapeutics, we screened a library of 727 FDA-approved or late-stage clinical trial compounds using a human macrophage-like cell model of infection. Eighty-eight compounds inhibited bacterial replication, including known antibiotics, antipsychotic or antidepressant treatments, antihistamines, and several additional compounds used to treat a variety of conditions. The majority of identified anti-C. burnetii compounds target host neurotransmitter system components. Serotoninergic, dopaminergic, and adrenergic components are among the most highly represented targets and potentially regulate macrophage activation, cytokine production, and autophagy. Overall, our screen identified multiple host-directed compounds that can be pursued for potential use as anti-C. burnetii drugs. IMPORTANCE Coxiella burnetii causes the debilitating disease Q fever in humans. This infection is difficult to treat with current antibiotics and can progress to long-term, potentially fatal infection in immunocompromised individuals or when treatment is delayed. Here, we identified many new potential treatment options in the form of drugs that are either FDA approved or have been used in late-stage clinical trials and target human neurotransmitter systems. These compounds are poised for future characterization as nontraditional anti-C. burnetii therapies.


Asunto(s)
Antibacterianos/farmacología , Coxiella burnetii/efectos de los fármacos , Coxiella burnetii/crecimiento & desarrollo , Interacciones Huésped-Patógeno/efectos de los fármacos , Neurotransmisores/antagonistas & inhibidores , Preparaciones Farmacéuticas/análisis , Bibliotecas de Moléculas Pequeñas/farmacología , Coxiella burnetii/genética , Ensayos Analíticos de Alto Rendimiento/métodos , Humanos , Farmacología , Fiebre Q/tratamiento farmacológico , Fiebre Q/microbiología , Células THP-1
3.
Pathog Dis ; 79(4)2021 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-33734371

RESUMEN

The human pulmonary environment is complex, containing a matrix of cells, including fibroblasts, epithelial cells, interstitial macrophages, alveolar macrophages and neutrophils. When confronted with foreign material or invading pathogens, these cells mount a robust response. Nevertheless, many bacterial pathogens with an intracellular lifecycle stage exploit this environment for replication and survival. These include, but are not limited to, Coxiella burnetii, Legionella pneumophila, Yersinia pestis, Mycobacterium tuberculosis and Staphylococcus aureus. Currently, few human disease-relevant model systems exist for studying host-pathogen interactions during these bacterial infections in the lung. Here, we present two novel infection platforms, human alveolar macrophages (hAMs) and human precision-cut lung slices (hPCLS), along with an up-to-date synopsis of research using said models. Additionally, alternative uses for these systems in the absence of pathogen involvement are presented, such as tissue banking and further characterization of the human lung environment. Overall, hAMs and hPCLS allow novel human disease-relevant investigations that other models, such as cell lines and animal models, cannot completely provide.


Asunto(s)
Infecciones Bacterianas/microbiología , Interacciones Huésped-Patógeno/inmunología , Enfermedades Pulmonares/microbiología , Pulmón/microbiología , Macrófagos Alveolares/microbiología , Modelos Biológicos , Infecciones Bacterianas/inmunología , Infecciones Bacterianas/patología , Coxiella burnetii/crecimiento & desarrollo , Coxiella burnetii/inmunología , Coxiella burnetii/patogenicidad , Humanos , Legionella pneumophila/crecimiento & desarrollo , Legionella pneumophila/inmunología , Legionella pneumophila/patogenicidad , Pulmón/inmunología , Pulmón/patología , Enfermedades Pulmonares/inmunología , Enfermedades Pulmonares/patología , Macrófagos Alveolares/inmunología , Macrófagos Alveolares/patología , Microtomía , Mycobacterium tuberculosis/crecimiento & desarrollo , Mycobacterium tuberculosis/inmunología , Mycobacterium tuberculosis/patogenicidad , Cultivo Primario de Células , Staphylococcus aureus/crecimiento & desarrollo , Staphylococcus aureus/inmunología , Staphylococcus aureus/patogenicidad , Bancos de Tejidos , Técnicas de Cultivo de Tejidos , Yersinia pestis/crecimiento & desarrollo , Yersinia pestis/inmunología , Yersinia pestis/patogenicidad
4.
Autophagy ; 17(3): 706-722, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-32116095

RESUMEN

Coxiella burnetii, the etiological agent of the zoonosis Q fever, replicates inside host cells within a large vacuole displaying autolysosomal characteristics. The development of this compartment is mediated by bacterial effectors, which interfere with a number of host membrane trafficking pathways. By screening a Coxiella transposon mutant library, we observed that transposon insertions in cbu0626 led to intracellular replication and vacuole biogenesis defects. Here, we demonstrate that CBU0626 is a novel member of the Coxiella vacuolar protein (Cvp) family of effector proteins, which is translocated by the Dot/Icm secretion system and localizes to vesicles with autolysosomal features as well as Coxiella-containing vacuoles (CCVs). We thus renamed this effector CvpF for Coxiella vacuolar protein F. CvpF specifically interacts with the host small GTPase RAB26, leading to the recruitment of the autophagosomal marker MAP1LC3B/LC3B (microtubule associated protein 1 light chain 3 beta) to CCVs. Importantly, cvpF::Tn mutants were highly attenuated compared to wild-type bacteria in the SCID mouse model of infection, highlighting the importance of CvpF for Coxiella virulence. These results suggest that CvpF manipulates endosomal trafficking and macroautophagy/autophagy induction for optimal C. burnetii vacuole biogenesis.Abbreviations: ACCM: acidified citrate cystein medium; AP: adaptor related protein complex; CCV: Coxiella-containing vacuole; Cvp: Coxiella vacuolar protein; GDI: guanosine nucleotide dissociation inhibitor; GDF: GDI dissociation factor; GEF: guanine exchange factor; LAMP1: lysosomal associated membrane protein 1; MAP1LC3B/LC3B: microtubule associated protein 1 light chain 3 beta; MTORC1: mechanistic target of rapamycin kinase MTOR complex 1; PBS: phosphate-buffered saline; PMA: phorbol myristate acetate; SQSTM1/p62: sequestosome 1; WT: wild-type.


Asunto(s)
Autofagia/fisiología , Sistemas de Secreción Bacterianos/metabolismo , Coxiella/metabolismo , Interacciones Huésped-Patógeno/inmunología , Vacuolas/microbiología , Animales , Proteínas Bacterianas/metabolismo , Coxiella burnetii/crecimiento & desarrollo , Coxiella burnetii/metabolismo , Humanos , Ratones , Vacuolas/metabolismo
5.
Infect Immun ; 88(12)2020 11 16.
Artículo en Inglés | MEDLINE | ID: mdl-32928965

RESUMEN

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.


Asunto(s)
Coxiella burnetii/efectos de los fármacos , Coxiella burnetii/crecimiento & desarrollo , Interacciones Huésped-Patógeno , Placenta/microbiología , Progesterona/farmacología , Animales , Proteínas Bacterianas/química , Línea Celular , Supervivencia Celular/efectos de los fármacos , Replicación del ADN/efectos de los fármacos , Sinergismo Farmacológico , Proteínas de Escherichia coli/química , Estrógenos/farmacología , Etidio/química , Femenino , Humanos , Proteínas de la Membrana/química , Proteínas de Transporte de Membrana/química , Proteínas de Transporte de Membrana/efectos de los fármacos , Proteínas de Transporte de Membrana/metabolismo , Piperazinas/farmacología , Placenta/efectos de los fármacos , Embarazo , Pregnenolona/farmacología , Proteínas Quinasas/química , Verapamilo/farmacología
6.
Epidemiol Infect ; 148: e21, 2020 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-32019625

RESUMEN

Coxiella burnetii, the causative agent of Q fever, is widely present in dairy products around the world. It has been isolated from unpasteurised milk and cheese and can survive for extended periods of time under typical storage conditions for these products. Although consumption of contaminated dairy products has been suggested as a potential route for transmission, it remains controversial. Given the high prevalence of C. burnetii in dairy products, we sought to examine the feasibility of transmitting the major sequence types (ST16, ST8 and ST20) of C. burnetii circulating in the United States. We delivered three strains of C. burnetii, comprising each sequence type, directly into the stomachs of immunocompetent BALB/c mice via oral gavage (OG) and assessed them for clinical symptoms, serological response and bacterial dissemination. We found that mice receiving C. burnetii by OG had notable splenomegaly only after infection with ST16. A robust immune response and persistence in the stomach and mesenteric lymph nodes were observed in mice receiving ST16 and ST20 by OG, and dissemination of C. burnetii to peripheral tissues was observed in all OG infected mice. These findings support the oral route as a mode of transmission for C. burnetii.


Asunto(s)
Coxiella burnetii/crecimiento & desarrollo , Productos Lácteos/microbiología , Transmisión de Enfermedad Infecciosa , Ingestión de Alimentos , Enfermedades Transmitidas por los Alimentos , Fiebre Q/transmisión , Animales , Coxiella burnetii/clasificación , Coxiella burnetii/genética , Modelos Animales de Enfermedad , Genotipo , Masculino , Ratones Endogámicos BALB C , Estados Unidos
7.
J Microbiol Methods ; 169: 105813, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31862458

RESUMEN

The intracellular Gram-negative bacterium, Coxiella burnetii, is a worldwide zoonotic pathogen and the causative agent of Q fever. The standard of care for C. burnetii infections involves extended periods of antibiotic treatment and the development of doxycycline-resistant strains stress the need for new treatment strategies. A previously developed axenic medium has facilitated in vitro growth of the organism. In this study, we have developed a simple culture method that is inexpensive, reliable and utilizes a modular hypoxic chamber system for either small or large scale production of bacteria without the need of a tri-gas incubator. This method provides consistent growth and yields sufficient viable bacteria within four days of culture and can be used for high-throughput screening. The viable bacteria were quantified by counting colony forming units and total bacteria were enumerated using a genomic equivalent method. The characterized bacterial inoculum was then used to optimize cell-based high-throughput immunofluorescence assays with a goal to quantify intracellular bacteria and then screen and identify compounds that inhibit early stages of C. burnetii infection in macrophages.


Asunto(s)
Cultivo Axénico/métodos , Coxiella burnetii/crecimiento & desarrollo , Ensayos Analíticos de Alto Rendimiento/métodos , Animales , Carga Bacteriana/métodos , Línea Celular , Técnica del Anticuerpo Fluorescente/métodos , Ratones , Fiebre Q/microbiología , Células RAW 264.7
8.
Cell Microbiol ; 22(5): e13154, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31872956

RESUMEN

Coxiella burnetii, the causative agent of the zoonotic disease Q fever, is a Gram-negative bacterium that replicates inside macrophages within a highly oxidative vacuole. Screening of a transposon mutant library suggested that sdrA, which encodes a putative short-chain dehydrogenase, is required for intracellular replication. Short-chain dehydrogenases are NADP(H)-dependent oxidoreductases, and SdrA contains a predicted NADP+ binding site, suggesting it may facilitate NADP(H) regeneration by C. burnetii, a key process for surviving oxidative stress. Purified recombinant 6×His-SdrA was able to convert NADP+ to NADP(H) in vitro. Mutation to alanine of a conserved glycine residue at position 12 within the predicted NADP binding site abolished significant enzymatic activity. Complementation of the sdrA mutant (sdrA::Tn) with plasmid-expressed SdrA restored intracellular replication to wild-type levels, but expressing enzymatically inactive G12A_SdrA did not. The sdrA::Tn mutant was more susceptible in vitro to oxidative stress, and treating infected host cells with L-ascorbate, an anti-oxidant, partially rescued the intracellular growth defect of sdrA::Tn. Finally, stable isotope labelling studies demonstrated a shift in flux through metabolic pathways in sdrA::Tn consistent with the presence of increased oxidative stress, and host cells infected with sdrA::Tn had elevated levels of reactive oxygen species compared with C. burnetii NMII.


Asunto(s)
Coxiella burnetii/metabolismo , NADP/metabolismo , Estrés Oxidativo , Coxiella burnetii/crecimiento & desarrollo , Citoplasma/metabolismo , Células HeLa , Humanos , Macrófagos/microbiología , Mutación , NADP/genética , Fiebre Q/metabolismo , Fiebre Q/microbiología , Regeneración , Vacuolas/microbiología
9.
Sci Rep ; 9(1): 18132, 2019 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-31792307

RESUMEN

Coxiella burnetii, the etiologic agent of Q fever, replicates in an intracellular phagolysosome with pH between 4 and 5. The impact of this low pH environment on antimicrobial treatment is not well understood. An in vitro system for testing antibiotic susceptibility of C. burnetii in axenic media was set up to evaluate the impact of pH on C. burnetii growth and survival in the presence and absence of antimicrobial agents. The data show that C. burnetii does not grow in axenic media at pH 6.0 or higher, but the organisms remain viable. At pH of 4.75, 5.25, and 5.75 moxifloxacin, doxycycline, and rifampin are effective at preventing growth of C. burnetii in axenic media, with moxifloxacin and doxycycline being bacteriostatic and rifampin having bactericidal activity. The efficacy of doxycycline and moxifloxacin improved at higher pH, whereas rifampin activity was pH independent. Hydroxychloroquine is thought to inhibit growth of C. burnetii in vivo by raising the pH of typically acidic intracellular compartments. It had no direct bactericidal or bacteriostatic activity on C. burnetii in axenic media, suggesting that raising pH of acidic intracellular compartments is its primary mechanism of action in vivo. The data suggest that doxycycline and hydroxychloroquine are primarily independent bacteriostatic agents.


Asunto(s)
Antibacterianos/farmacología , Coxiella burnetii/efectos de los fármacos , Medios de Cultivo/química , Antibacterianos/química , Cultivo Axénico/métodos , Coxiella burnetii/crecimiento & desarrollo , Doxiciclina/farmacología , Concentración de Iones de Hidrógeno , Hidroxicloroquina/farmacología , Pruebas de Sensibilidad Microbiana , Moxifloxacino/farmacología , Rifampin/farmacología
10.
PLoS Pathog ; 15(12): e1007855, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31869379

RESUMEN

Upon host cell infection, the obligate intracellular bacterium Coxiella burnetii resides and multiplies within the Coxiella-Containing Vacuole (CCV). The nascent CCV progresses through the endosomal maturation pathway into a phagolysosome, acquiring endosomal and lysosomal markers, as well as acidic pH and active proteases and hydrolases. Approximately 24-48 hours post infection, heterotypic fusion between the CCV and host endosomes/lysosomes leads to CCV expansion and bacterial replication in the mature CCV. Initial CCV acidification is required to activate C. burnetii metabolism and the Type 4B Secretion System (T4BSS), which secretes effector proteins required for CCV maturation. However, we found that the mature CCV is less acidic (pH~5.2) than lysosomes (pH~4.8). Further, inducing CCV acidification to pH~4.8 causes C. burnetii lysis, suggesting C. burnetii actively regulates pH of the mature CCV. Because heterotypic fusion with host endosomes/lysosomes may influence CCV pH, we investigated endosomal maturation in cells infected with wildtype (WT) or T4BSS mutant (ΔdotA) C. burnetii. In WT-infected cells, we observed a significant decrease in proteolytically active, LAMP1-positive endolysosomal vesicles, compared to mock or ΔdotA-infected cells. Using a ratiometric assay to measure endosomal pH, we determined that the average pH of terminal endosomes in WT-infected cells was pH~5.8, compared to pH~4.75 in mock and ΔdotA-infected cells. While endosomes progressively acidified from the periphery (pH~5.5) to the perinuclear area (pH~4.7) in both mock and ΔdotA-infected cells, endosomes did not acidify beyond pH~5.2 in WT-infected cells. Finally, increasing lysosomal biogenesis by overexpressing the transcription factor EB resulted in smaller, more proteolytically active CCVs and a significant decrease in C. burnetii growth, indicating host lysosomes are detrimental to C. burnetii. Overall, our data suggest that C. burnetii inhibits endosomal maturation to reduce the number of proteolytically active lysosomes available for heterotypic fusion with the CCV, possibly as a mechanism to regulate CCV pH.


Asunto(s)
Proteínas Bacterianas/metabolismo , Transporte Biológico/fisiología , Coxiella burnetii/crecimiento & desarrollo , Endosomas/metabolismo , Adulto , Anciano , Femenino , Interacciones Huésped-Patógeno/fisiología , Humanos , Macrófagos/metabolismo , Masculino , Persona de Mediana Edad
11.
Pathog Dis ; 77(8)2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31845968

RESUMEN

The zoonotic disease Q fever caused by the intracellular bacterium Coxiella burnetii remains a global health threat due to its high infectivity, environmental stability, the debilitating nature and the long duration of treatment. Designing new and potent drugs that target previously unexplored pathways is essential to shorten treatment time and minimise antibiotic resistance. Nicotinamide adenine dinucleotide (NAD) is an essential and ubiquitous cofactor in all living organisms. NadB, an L-aspartate oxidase catalysing the first step of the prokaryotic-specific NAD de novo biosynthetic pathway, is required for C. burnetii growth and replication inside host cells. In this study, in vitro enzyme assays utilising recombinant glutathione S-transferase tagged NadB (GST-NadB) demonstrated inhibition of the L-aspartate oxidase activity of NadB by meso-tartrate. Furthermore, meso-tartrate inhibits intracellular growth and replication of C. burnetii inside host cells in a dose-dependent manner, and has no effect on the viability of mammalian cells. Unexpectedly, meso-tartrate also inhibited growth of C. burnetii in axenic medium, and further reduces replication of the nadB mutant inside host cells, suggesting it is acting more widely than simple inhibition of NadB. Overall, these results suggest that the antibacterial activity of meso-tartrate warrants further study, including investigation of its additional target(s).


Asunto(s)
Antibacterianos/farmacología , Coxiella burnetii/efectos de los fármacos , Coxiella burnetii/crecimiento & desarrollo , Inhibidores Enzimáticos/farmacología , Tartratos/farmacología , Aminoácido Oxidorreductasas/antagonistas & inhibidores , Coxiella burnetii/enzimología , Coxiella burnetii/metabolismo , Células Epiteliales/microbiología , Células HeLa , Humanos , Viabilidad Microbiana/efectos de los fármacos , NAD/metabolismo , Células THP-1
12.
J Bacteriol ; 201(22)2019 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-31451541

RESUMEN

Coxiella burnetii is an obligate intracellular gammaproteobacterium and zoonotic agent of Q fever. We previously identified 15 small noncoding RNAs (sRNAs) of C. burnetii One of them, CbsR12 (Coxiella burnetiismall RNA 12), is highly transcribed during axenic growth and becomes more prominent during infection of cultured mammalian cells. Secondary structure predictions of CbsR12 revealed four putative CsrA-binding sites in stem loops with consensus AGGA/ANGGA motifs. We subsequently determined that CbsR12 binds to recombinant C. burnetii CsrA-2, but not CsrA-1, proteins in vitro Moreover, through a combination of in vitro and cell culture assays, we identified several in trans mRNA targets of CbsR12. Of these, we determined that CbsR12 binds and upregulates translation of carA transcripts coding for carbamoyl phosphate synthetase A, an enzyme that catalyzes the first step of pyrimidine biosynthesis. In addition, CbsR12 binds and downregulates translation of metK transcripts coding for S-adenosylmethionine synthetase, a component of the methionine cycle. Furthermore, we found that CbsR12 binds to and downregulates the quantity of cvpD transcripts, coding for a type IVB effector protein, in mammalian cell culture. Finally, we found that CbsR12 is necessary for expansion of Coxiella-containing vacuoles and affects growth rates in a dose-dependent manner in the early phase of infecting THP-1 cells. This is the first characterization of a trans-acting sRNA of C. burnetii and the first example of a bacterial sRNA that regulates both CarA and MetK synthesis. CbsR12 is one of only a few identified trans-acting sRNAs that interacts with CsrA.IMPORTANCE Regulation of metabolism and virulence in C. burnetii is not well understood. Here, we show that C. burnetii small RNA 12 (CbsR12) is highly transcribed in the metabolically active large-cell variant compared to the nonreplicative small-cell variant. We show that CbsR12 directly regulates several genes involved in metabolism, along with a type IV effector gene, in trans In addition, we demonstrate that CbsR12 binds to CsrA-2 in vitro and induces autoaggregation and biofilm formation when transcribed ectopically in Escherichia coli, consistent with other CsrA-sequestering sRNAs. These results implicate CbsR12 in the indirect regulation of a number of genes via CsrA-mediated regulatory activities. The results also support CbsR12 as a crucial regulatory component early on in a mammalian cell infection.


Asunto(s)
Coxiella burnetii/genética , Fiebre Q/microbiología , ARN Bacteriano/fisiología , ARN Pequeño no Traducido/fisiología , Proteínas de Unión al ARN/metabolismo , Vacuolas/metabolismo , Animales , Cultivo Axénico , Proteínas Bacterianas/metabolismo , Chlorocebus aethiops , Coxiella burnetii/crecimiento & desarrollo , Coxiella burnetii/metabolismo , Humanos , ARN Bacteriano/genética , ARN Bacteriano/metabolismo , ARN Pequeño no Traducido/genética , ARN Pequeño no Traducido/metabolismo , Células THP-1 , Células Vero
14.
J Bacteriol ; 201(8)2019 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-30745369

RESUMEN

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


Asunto(s)
Proteínas Bacterianas/metabolismo , Coxiella burnetii/citología , Coxiella burnetii/crecimiento & desarrollo , Regulación Bacteriana de la Expresión Génica , Factor sigma/metabolismo , Animales , Chlorocebus aethiops , Coxiella burnetii/genética , Citoplasma/microbiología , Células Epiteliales/microbiología , Eliminación de Gen , Perfilación de la Expresión Génica , Humanos , Macrófagos/microbiología , Proteómica , Factor sigma/deficiencia , Células THP-1 , Células Vero
15.
mBio ; 10(1)2019 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-30723133

RESUMEN

The Q fever agent Coxiella burnetii is a Gram-negative bacterium that invades macrophages and replicates inside a specialized lysosomal vacuole. The pathogen employs a type 4B secretion system (T4BSS) to deliver effector proteins into the host cell that modify the Coxiella-containing vacuole (CCV) into a replication-permissive niche. Mature CCVs are massive degradative organelles that acquire lysosomal proteins. Inhibition of mammalian (or mechanistic) target of rapamycin complex 1 (mTORC1) kinase by nutrient deprivation promotes autophagy and lysosome fusion, as well as activation of the transcription factors TFE3 and TFEB (TFE3/B), which upregulates expression of lysosomal genes. Here, we report that C. burnetii inhibits mTORC1 as evidenced by impaired localization of mTORC1 to endolysosomal membranes and decreased phosphorylation of elF4E-binding protein 1 (4E-BP1) and S6 kinase 1 in infected cells. Infected cells exhibit increased amounts of autophagy-related proteins protein 1A/1B-light chain 3 (LC3) and p62 as well as of activated TFE3. However, C. burnetii did not accelerate autophagy or block autophagic flux triggered by cell starvation. Activation of autophagy or transcription by TFE3/B increased CCV expansion without enhancing bacterial replication. By contrast, knockdown of tuberous sclerosis complex 1 (TSC1) or TSC2, which hyperactivates mTORC1, impaired CCV expansion and bacterial replication. Together, these data demonstrate that specific inhibition of mTORC1 by C. burnetii, but not amplified cell catabolism via autophagy, is required for optimal pathogen replication. These data reveal a complex interplay between lysosomal function and host cell metabolism that regulates C. burnetii intracellular growth.IMPORTANCECoxiella burnetii is an intracellular pathogenic bacterium that replicates within a lysosomal vacuole. Biogenesis of the Coxiella-containing vacuole (CCV) requires effector proteins delivered into the host cell cytosol by the type 4B secretion system (T4BSS). Modifications to lysosomal physiology required for pathogen replication within the CCV are poorly understood. Mammalian (or mechanistic) target of rapamycin complex 1 (mTORC1) is a master kinase that regulates lysosome structure and function. Nutrient deprivation inhibits mTORC1, which promotes cell catabolism in the form of accelerated autophagy and increased lysosome biosynthesis. Here, we report that C. burnetii growth is enhanced by T4BSS-dependent inhibition of mTORC1 that does not activate autophagy. Canonical inhibition of mTORC1 by starvation or inhibitor treatment that induces autophagic flux does not benefit C. burnetii growth. Furthermore, hyperactivation of mTORC1 impairs bacterial replication. These findings indicate that C. burnetii inhibition of mTORC1 without accelerated autophagy promotes bacterial growth.


Asunto(s)
Coxiella burnetii/crecimiento & desarrollo , Interacciones Huésped-Patógeno , Macrófagos/microbiología , Diana Mecanicista del Complejo 1 de la Rapamicina/antagonistas & inhibidores , Fagosomas/microbiología , Humanos , Células THP-1
16.
Clin Microbiol Infect ; 25(9): 1155.e1-1155.e8, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-30625413

RESUMEN

OBJECTIVE: Q fever epidemic outbreaks have been reported in French Guiana and in The Netherlands. To determine whether the C. burnetii strains involved in these epidemics had a peculiar virulence pattern, we compared the pathogenicity of the Guiana and the German strain (a clone of The Netherlands strain), in silico, in vitro, and in vivo versus the Nine Mile strain. METHOD: The pan-genomes of the Guiana (Cb175), German (Z3055), and the referent Nine Mile (RSA 493) C. burnetii strains were compared. In vitro, the growth rate and the morphological presentation were compared. In vivo (SCID and Balb/c mice), weight loss, histological lesions, C. burnetii bacterial load in deep organs, and serological response were reported according to each C. burnetii strain studied. RESULTS: The Guiana strain had 77 times more missing genes and 12 times more unique genes than the German strain. The Guiana strain presented as large cell variants (LCVs) and led to the most pronounced fatality rate in SCID mice (100% at 4 weeks). The German strain presented as small cell variants (SCVs), and had an intermediate fatality rate (75% at 4 weeks). Both the Guiana and the German strains led to a significant higher serological response at 2 and 4 weeks post infection (p <0.05). CONCLUSION: The Guiana strain was the most virulent strain, followed by the German strain and the referent Nine Mile strain. Unique and missing genes could be implicated but further investigations are necessary to specify their role.


Asunto(s)
Coxiella burnetii/patogenicidad , Brotes de Enfermedades , Fiebre Q/epidemiología , Fiebre Q/microbiología , Animales , Anticuerpos Antibacterianos/sangre , Coxiella burnetii/clasificación , Coxiella burnetii/genética , Coxiella burnetii/crecimiento & desarrollo , ADN Bacteriano/análisis , Modelos Animales de Enfermedad , Guyana Francesa/epidemiología , Variación Genética , Genoma Bacteriano/genética , Ratones Endogámicos BALB C , Ratones SCID , Países Bajos/epidemiología , Fiebre Q/sangre , Fiebre Q/patología , Análisis de Supervivencia , Virulencia
17.
J Biol Chem ; 293(48): 18636-18645, 2018 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-30315113

RESUMEN

Coxiella burnetii is an intracellular Gram-negative bacterium responsible for the important zoonotic disease Q fever. Improved genetic tools and the ability to grow this bacterium in host cell-free media has advanced the study of C. burnetii pathogenesis, but the mechanisms that allow it to survive inside the hostile phagolysosome remain incompletely understood. Previous screening of a transposon mutant library for replication within HeLa cells has suggested that nadB, encoding a putative l-aspartate oxidase required for de novo NAD synthesis, is needed for intracellular replication. Here, using genetic complementation of two independent nadB mutants and intracellular replication assays, we confirmed this finding. Untargeted metabolite analyses demonstrated key changes in metabolites in the NAD biosynthetic pathway in the nadB mutant compared with the WT, confirming the involvement of NadB in de novo NAD synthesis. Bioinformatic analysis revealed the presence of a functionally conserved arginine residue at position 275. Using site-directed mutagenesis to substitute this residue with leucine, which abolishes the activity of Escherichia coli NadB, and expression of WT and R275L GST-NadB fusion proteins in E. coli JM109, we found that purified recombinant WT GST-NadB has l-aspartate oxidase activity and that the R275L NadB variant is inactive. Complementation of the C. burnetii nadB mutant with a plasmid expressing this inactive R275L NadB failed to restore replication to WT levels, confirming the link between de novo NAD synthesis and intracellular replication of C. burnetii This suggests that targeting this prokaryotic-specific pathway could advance the development of therapeutics to combat C. burnetii infections.


Asunto(s)
Coxiella burnetii/crecimiento & desarrollo , Coxiella burnetii/metabolismo , NAD/biosíntesis , Fiebre Q/microbiología , Cromatografía de Gases , Cromatografía Liquida , Elementos Transponibles de ADN , Células HeLa , Humanos , Espectrometría de Masas , Mutagénesis Sitio-Dirigida
18.
Infect Immun ; 86(10)2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30037794

RESUMEN

Cytokine responses of chronic Q fever patients to the intracellular bacterium Coxiella burnetii have mostly been studied using ex vivo stimulation of immune cells with heat-killed C. burnetii due to the extensive measures needed to work with viable biosafety level 3 agents. Whether research with heat-killed C. burnetii can be translated to immune responses to viable C. burnetii is imperative for the interpretation of previous and future studies with heat-killed C. burnetii Peripheral blood mononuclear cells (PBMCs) of chronic Q fever patients (n = 10) and healthy controls (n = 10) were stimulated with heat-killed or viable C. burnetii of two strains, Nine Mile and the Dutch outbreak strain 3262, for 24 h, 48 h, and 7 days in the absence or presence of serum containing anti-C. burnetii antibodies. When stimulated with viable C. burnetii, PBMCs of chronic Q fever patients and controls produced fewer proinflammatory cytokines (interleukin-6 [IL-6], tumor necrosis factor alpha, and IL-1ß) after 24 h than after stimulation with heat-killed C. burnetii In the presence of Q fever seronegative serum, IL-10 production was higher after stimulation with viable rather than heat-killed C. burnetii; however, when incubating with anti-C. burnetii antibody serum, the effect on IL-10 production was reduced. Levels of adaptive, merely T-cell-derived cytokine (gamma interferon, IL-17, and IL-22) and CXCL9 production were not different between heat-killed and viable C. burnetii stimulatory conditions. Results from previous and future research with heat-killed C. burnetii should be interpreted with caution for innate cytokines, but heat-killed C. burnetii-induced adaptive cytokine production is representative of stimulation with viable bacteria.


Asunto(s)
Coxiella burnetii/inmunología , Citocinas/inmunología , Fiebre Q/inmunología , Anticuerpos Antibacterianos/inmunología , Coxiella burnetii/genética , Coxiella burnetii/crecimiento & desarrollo , Citocinas/genética , Femenino , Calor , Humanos , Interferón gamma/genética , Interferón gamma/inmunología , Interleucina-1beta/genética , Interleucina-1beta/inmunología , Leucocitos Mononucleares/inmunología , Masculino , Viabilidad Microbiana , Fiebre Q/genética , Fiebre Q/microbiología , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología
19.
Artículo en Inglés | MEDLINE | ID: mdl-29938202

RESUMEN

Lipid A is an essential basal component of lipopolysaccharide of most Gram-negative bacteria. Inhibitors targeting LpxC, a conserved enzyme in lipid A biosynthesis, are antibiotic candidates against Gram-negative pathogens. Here we report the characterization of the role of lipid A in Coxiella burnetii growth in axenic media, monkey kidney cells (BGMK and Vero), and macrophage-like THP-1 cells by using a potent LpxC inhibitor -LPC-011. We first determined the susceptibility of C. burnetii LpxC to LPC-011 in a surrogate E. coli model. In E. coli, the minimum inhibitory concentration (MIC) of LPC-011 against C. burnetii LpxC is < 0.05 µg/mL, a value lower than the inhibitor's MIC against E. coli LpxC. Considering the inhibitor's problematic pharmacokinetic properties in vivo and Coxiella's culturing time up to 7 days, the stability of LPC-011 in cell cultures was assessed. We found that regularly changing inhibitor-containing media was required for sustained inhibition of C. burnetii LpxC in cells. Under inhibitor treatment, Coxiella has reduced growth yields in axenic media and during replication in non-phagocytic cells, and has a reduced number of productive vacuoles in such cells. Inhibiting lipid A biosynthesis in C. burnetii by the inhibitor was shown in a phase II strain transformed with chlamydial kdtA. This exogenous KdtA enzyme modifies Coxiella lipid A with an α-Kdo-(2 → 8)-α-Kdo epitope that can be detected by anti-chlamydia genus antibodies. In inhibitor-treated THP-1 cells, Coxiella shows severe growth defects characterized by poor vacuole formation and low growth yields. Coxiella progenies prepared from inhibitor-treated cells retain the capability of normally infecting all tested cells in the absence of the inhibitor, which suggests a dispensable role of lipid A for infection and early vacuole development. In conclusion, our data suggest that lipid A has significance for optimal development of Coxiella-containing vacuoles, and for robust multiplication of C. burnetii in macrophage-like THP-1 cells. Unlike many bacteria, C. burnetii replication in axenic media and non-phagocytic cells was less dependent on normal lipid A biosynthesis.


Asunto(s)
Cultivo Axénico/métodos , Coxiella burnetii/crecimiento & desarrollo , Coxiella burnetii/patogenicidad , Lípido A/antagonistas & inhibidores , Macrófagos/microbiología , Amidohidrolasas/antagonistas & inhibidores , Amidohidrolasas/genética , Animales , Proteínas Bacterianas/antagonistas & inhibidores , Proteínas Bacterianas/genética , Chlorocebus aethiops , Coxiella burnetii/efectos de los fármacos , Escherichia coli/efectos de los fármacos , Escherichia coli/crecimiento & desarrollo , Escherichia coli/patogenicidad , Humanos , Ácidos Hidroxámicos/farmacología , Lípido A/genética , Macrófagos/efectos de los fármacos , Células THP-1 , Treonina/análogos & derivados , Treonina/farmacología , Vacuolas/efectos de los fármacos , Vacuolas/microbiología , Células Vero
20.
Curr Protoc Microbiol ; 50(1): e52, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29927105

RESUMEN

Coxiella burnetii is a highly infectious obligate intracellular bacterium and the etiological agent of the zoonosis Query (Q) fever. This Gram-negative gamma-proteobacterium has adapted to replicate within a specialized compartment in mammalian phagocytic cells, known as the Coxiella-containing vacuole (CCV). Knowledge of critical characteristics of the CCV microenvironment (e.g., luminal pH), analysis of the C. burnetii genome sequence, and strategic metabolic profiling have provided the basis for determining the physicochemical and nutritional conditions necessary to support axenic replication of C. burnetii. In this unit, the media currently utilized for axenic culture of C. burnetii are described, with emphasis on application. To aid in experimental reproducibility and interpretation of results, considerations and limitations are discussed. Lastly, expected results for C. burnetii axenic growth under control conditions are provided as a reference. © 2018 by John Wiley & Sons, Inc.


Asunto(s)
Cultivo Axénico/métodos , Coxiella burnetii/crecimiento & desarrollo , Fiebre Q/microbiología , Animales , Cultivo Axénico/instrumentación , Coxiella burnetii/genética , Coxiella burnetii/aislamiento & purificación , Coxiella burnetii/metabolismo , Medios de Cultivo/química , Medios de Cultivo/metabolismo , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...