Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 62
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Proc Natl Acad Sci U S A ; 119(1)2022 01 04.
Article in English | MEDLINE | ID: mdl-34930823

ABSTRACT

Coxiella burnetii is a bacterial pathogen that replicates within host cells by establishing a membrane-bound niche called the Coxiella-containing vacuole. Biogenesis of this compartment requires effectors of its Dot/Icm type IV secretion system. A large cohort of such effectors has been identified, but the function of most of them remain elusive. Here, by a cell-based functional screening, we identified the effector Cbu0513 (designated as CinF) as an inhibitor of NF-κB signaling. CinF is highly similar to a fructose-1,6-bisphosphate (FBP) aldolase/phosphatase present in diverse bacteria. Further study reveals that unlike its ortholog from Sulfolobus tokodaii, CinF does not exhibit FBP phosphatase activity. Instead, it functions as a protein phosphatase that specifically dephosphorylates and stabilizes IκBα. The IκBα phosphatase activity is essential for the role of CinF in C. burnetii virulence. Our results establish that C. burnetii utilizes a protein adapted from sugar metabolism to subvert host immunity.


Subject(s)
Bacterial Proteins , Coxiella burnetii , Phosphoprotein Phosphatases , Q Fever , Signal Transduction , Virulence Factors , Animals , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Chlorocebus aethiops , Coxiella burnetii/genetics , Coxiella burnetii/immunology , Coxiella burnetii/pathogenicity , HEK293 Cells , HeLa Cells , Humans , NF-kappa B/genetics , NF-kappa B/immunology , Phosphoprotein Phosphatases/genetics , Phosphoprotein Phosphatases/immunology , Q Fever/genetics , Q Fever/immunology , Signal Transduction/genetics , Signal Transduction/immunology , Vero Cells , Virulence Factors/genetics , Virulence Factors/immunology
2.
Proc Natl Acad Sci U S A ; 119(14): e2112886119, 2022 04 05.
Article in English | MEDLINE | ID: mdl-35363569

ABSTRACT

Bacterial pathogen identification, which is critical for human health, has historically relied on culturing organisms from clinical specimens. More recently, the application of machine learning (ML) to whole-genome sequences (WGSs) has facilitated pathogen identification. However, relying solely on genetic information to identify emerging or new pathogens is fundamentally constrained, especially if novel virulence factors exist. In addition, even WGSs with ML pipelines are unable to discern phenotypes associated with cryptic genetic loci linked to virulence. Here, we set out to determine if ML using phenotypic hallmarks of pathogenesis could assess potential pathogenic threat without using any sequence-based analysis. This approach successfully classified potential pathogenetic threat associated with previously machine-observed and unobserved bacteria with 99% and 85% accuracy, respectively. This work establishes a phenotype-based pipeline for potential pathogenic threat assessment, which we term PathEngine, and offers strategies for the identification of bacterial pathogens.


Subject(s)
Bacteria , Genome, Bacterial , Machine Learning , Virulence Factors , Whole Genome Sequencing , Bacteria/genetics , Bacteria/pathogenicity , Phenotype , Virulence/genetics , Virulence Factors/genetics
3.
Infect Immun ; 88(10)2020 09 18.
Article in English | MEDLINE | ID: mdl-32690632

ABSTRACT

Reproductive failure is the hallmark of brucellosis in animals. An uncommon but important complication in pregnant women who become acutely infected with Brucella melitensis is spontaneous pregnancy loss or vertical transmission to the fetus. Unfortunately, the mechanism behind reproductive failure is still obscure, partially due to the lack of a proper study model. Recently, it was demonstrated that intratracheal (IT) inoculation of nonpregnant guinea pigs would replicate features of clinical disease in humans. To determine if IT inoculation would induce reproductive disease, guinea pigs were infected at mid-gestation and monitored daily for fever and abortions. Fever developed between day 14 to 18 postinoculation, and by 3 weeks postinoculation, 75% of pregnant guinea pigs experienced stillbirths or spontaneous abortions mimicking natural disease. Next, to investigate the guinea pig as a model for evaluating vaccine efficacy during pregnancy, nonpregnant guinea pigs were vaccinated with S19, 16MΔvjbR + Quil-A, or 100 µl PBS + Quil-A (as control). Guinea pigs were bred and vaccinated guinea pigs were challenged at mid-gestation with B. melitensis IT inoculation and monitored for fever and abortions. Vaccination with both vaccines prevented fever and protected against abortion. Together, this study indicates that pregnant guinea pigs are an appropriate animal model to study reproductive disease and offer an improved model to evaluate the ability of vaccine candidates to protect against a serious manifestation of disease.


Subject(s)
Brucella Vaccine/administration & dosage , Brucella melitensis/immunology , Brucellosis/prevention & control , Disease Models, Animal , Pregnancy Complications, Infectious/prevention & control , Animals , Antibodies, Bacterial/blood , Brucella melitensis/pathogenicity , Brucellosis/microbiology , Brucellosis/pathology , Female , Guinea Pigs , Humans , Mammary Glands, Animal/microbiology , Mammary Glands, Animal/pathology , Placenta/microbiology , Placenta/pathology , Pregnancy , Pregnancy Complications, Infectious/microbiology , Pregnancy Complications, Infectious/pathology , Vaccination
4.
J Infect Dis ; 220(4): 624-634, 2019 07 19.
Article in English | MEDLINE | ID: mdl-30938819

ABSTRACT

BACKGROUND: Protective immunity against Coxiella burnetii infection is conferred by vaccination with virulent (PI-WCV), but not avirulent (PII-WCV) whole-cell inactivated bacterium. The only well-characterized antigenic difference between virulent and avirulent C. burnetii is they have smooth and rough lipopolysaccharide (LPS), respectively. METHODS: Mice were vaccinated with PI-WCV and PII-WCV. Humoral and cellular responses were evaluated using protein chip microarrays and ELISpots, respectively. Dendritic cell (DC) maturation after stimulation with PI-WVC and PII-WVC was evaluated using flow cytometry. Vaccine-challenge studies were performed to validate the importance of the receptor CCR7. RESULTS: Other than specific antibody response to PI-LPS, similar antibody profiles were observed but IgG titers were significantly higher after vaccination with PI-WCV. Furthermore, higher frequency of antigen-specific CD4+ T cells was detected in mice immunized with PI-WCV. PI-WCV-stimulated DCs displayed significantly higher levels of CCR7 and migratory ability to secondary lymphoid organs. Challenge-protection studies in wild-type and CCR7-deficient mice confirmed that CCR7 is critical for PI-WCV-induced cellular immunity. CONCLUSIONS: PI-WVC stimulates protective immunity to C. burnetii in mice through stimulation of migratory behavior in DCs for protective cellular immunity. Additionally, the humoral immune response to LPS is an important component of protective immunity.


Subject(s)
Antibodies, Bacterial/immunology , Antigens, Bacterial/immunology , Bacterial Vaccines/immunology , Coxiella burnetii/immunology , Immunity, Cellular , Q Fever/immunology , Receptors, Chemokine/immunology , Animals , Antibody Formation , Dendritic Cells/immunology , Female , Humans , Lipopolysaccharides/immunology , Mice , Q Fever/microbiology , Q Fever/prevention & control , Vaccination
5.
Traffic ; 17(5): 461-74, 2016 May.
Article in English | MEDLINE | ID: mdl-26842840

ABSTRACT

There is a fundamental gap in our understanding of how a eukaryotic cell apportions the limited space within its cell membrane. Upon infection, a cell competes with intracellular pathogens for control of this same precious resource. The struggle between pathogen and host provides us with an opportunity to uncover the mechanisms regulating subcellular space by understanding how pathogens modulate vesicular traffic and membrane fusion events to create a specialized compartment for replication. By comparing several important intracellular pathogens, we review the molecular mechanisms and trafficking pathways that drive two space allocation strategies, the formation of tight and spacious pathogen-containing vacuoles. Additionally, we discuss the potential advantages of each pathogenic lifestyle, the broader implications these lifestyles might have for cellular biology and outline exciting opportunities for future investigation.


Subject(s)
Host-Pathogen Interactions , Vacuoles/microbiology , Membrane Fusion
6.
J Infect Dis ; 215(10): 1580-1589, 2017 05 15.
Article in English | MEDLINE | ID: mdl-27703037

ABSTRACT

Coxiella burnetii is a gram-negative bacterium that causes acute and chronic Q fever. Because of the severe adverse effect of whole-cell vaccination, identification of immunodominant antigens of C. burnetii has become a major focus of Q fever vaccine development. We hypothesized that secreted C. burnetii type IV secretion system (T4SS) effectors may represent a major class of CD8+ T-cell antigens, owing to their cytosolic localization. Twenty-nine peptides were identified that elicited robust CD8+ T-cell interferon γ (IFN-γ) recall responses from mice infected with C. burnetii. Interestingly, 22 of 29 epitopes were derived from 17 T4SS-related proteins, none of which were identified as immunodominant antigens by using previous antibody-guided approaches. These epitopes were expressed in an attenuated Listeria monocytogenes vaccine strain. Immunization with recombinant L. monocytogenes vaccines induced a robust CD8+ T-cell response and conferred measurable protection against C. burnetii infection in mice. These data suggested that T4SS effectors represent an important class of C. burnetii antigens that can induce CD8+ T-cell responses. We also showed that attenuated L. monocytogenes vaccine vectors are an efficient antigen-delivery platform that can be used to induce robust protective CD8+ T-cell immune responses against C. burnetii infection.


Subject(s)
Antigen Presentation/immunology , Bacterial Vaccines/immunology , CD8-Positive T-Lymphocytes/immunology , Coxiella burnetii/immunology , Epitopes, T-Lymphocyte/immunology , Vaccines, Attenuated/immunology , Animals , Antibodies, Bacterial/immunology , Antigens, Bacterial/immunology , Bacterial Proteins/chemistry , Bacterial Proteins/immunology , Bacterial Vaccines/chemistry , Coxiella burnetii/chemistry , Epitopes, T-Lymphocyte/chemistry , Female , Interferon-gamma Release Tests , Listeria monocytogenes/immunology , Mice , Mice, Inbred C57BL , Peptide Fragments/chemistry , Peptide Fragments/immunology , Q Fever/immunology , Q Fever/microbiology , Type IV Secretion Systems/chemistry , Type IV Secretion Systems/immunology , Vaccines, Attenuated/chemistry
7.
Infect Immun ; 84(9): 2524-33, 2016 09.
Article in English | MEDLINE | ID: mdl-27324482

ABSTRACT

Coxiella burnetii, the etiological agent of Q fever in humans, is an intracellular pathogen that replicates in an acidified parasitophorous vacuole derived from host lysosomes. Generation of this replicative compartment requires effectors delivered into the host cell by the Dot/Icm type IVb secretion system. Several effectors crucial for C. burnetii intracellular replication have been identified, but the host pathways coopted by these essential effectors are poorly defined, and very little is known about how spacious vacuoles are formed and maintained. Here we demonstrate that the essential type IVb effector, CirA, stimulates GTPase activity of RhoA. Overexpression of CirA in mammalian cells results in cell rounding and stress fiber disruption, a phenotype that is rescued by overexpression of wild-type or constitutively active RhoA. Unlike other effector proteins that subvert Rho GTPases to modulate uptake, CirA is the first effector identified that is dispensable for uptake and instead recruits Rho GTPase to promote biogenesis of the bacterial vacuole. Collectively our results highlight the importance of CirA in coopting host Rho GTPases for establishment of Coxiella burnetii infection and virulence in mammalian cell culture and mouse models of infection.


Subject(s)
Bacterial Proteins/metabolism , Coxiella burnetii/metabolism , GTP Phosphohydrolases/metabolism , Q Fever/metabolism , Type IV Secretion Systems/metabolism , Virulence/physiology , rhoA GTP-Binding Protein/metabolism , Animals , Cell Line, Tumor , HeLa Cells , Host-Pathogen Interactions/physiology , Humans , Lysosomes/metabolism , Mice , Protein Transport/physiology , Q Fever/microbiology , Vacuoles/metabolism , Vacuoles/microbiology
8.
BMC Microbiol ; 15: 99, 2015 May 12.
Article in English | MEDLINE | ID: mdl-25962997

ABSTRACT

BACKGROUND: Periplasmically localized copper-zinc co-factored superoxide dismutase (SodC) enzymes have been identified in a wide range of Gram-negative bacteria and are proposed to protect bacteria from exogenously produced toxic oxygen radicals, which indicates the potential significance of a Coxiella burnetii SodC. RESULTS: Assays for SOD activity demonstrated that the cloned C. burnetii insert codes for a SOD that was active over a wide range of pH and inhibitable with 5 mM H2O2 and 1 mM sodium diethyldithiocarbamate, a characteristic of Cu/ZnSODs that distinguishes them from Fe or Mn SODs. The sodC was expressed by C. burnetii, has a molecular weight of approximately 18 kDa, which is consistent with the predicted molecular weight, and localized towards the periphery of C. burnetii. Over expression of the C. burnetii sodC in an E. coli sodC mutant restored resistance to H2O2 killing to wild type levels. CONCLUSIONS: We have demonstrated that C. burnetii does express a Cu/ZnSOD that is functional at low pH, appears to be excreted, and was able to restore H2O2 resistance in an E. coli sodC mutant. Taken together, these results indicate that the C. burnetii Cu/ZnSOD is a potentially important virulence factor.


Subject(s)
Coxiella burnetii/enzymology , Superoxide Dismutase/metabolism , Cloning, Molecular , Coxiella burnetii/genetics , Enzyme Inhibitors/analysis , Enzyme Stability , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Deletion , Gene Expression , Genetic Complementation Test , Hydrogen Peroxide/toxicity , Hydrogen-Ion Concentration , Molecular Weight , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Superoxide Dismutase/chemistry , Superoxide Dismutase/genetics
9.
Syst Biol ; 62(5): 752-62, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23736103

ABSTRACT

Rooting phylogenies is critical for understanding evolution, yet the importance, intricacies and difficulties of rooting are often overlooked. For rooting, polymorphic characters among the group of interest (ingroup) must be compared to those of a relative (outgroup) that diverged before the last common ancestor (LCA) of the ingroup. Problems arise if an outgroup does not exist, is unknown, or is so distant that few characters are shared, in which case duplicated genes originating before the LCA can be used as proxy outgroups to root diverse phylogenies. Here, we describe a genome-wide expansion of this technique that can be used to solve problems at the other end of the evolutionary scale: where ingroup individuals are all very closely related to each other, but the next closest relative is very distant. We used shared orthologous single nucleotide polymorphisms (SNPs) from 10 whole genome sequences of Coxiella burnetii, the causative agent of Q fever in humans, to create a robust, but unrooted phylogeny. To maximize the number of characters informative about the rooting, we searched entire genomes for polymorphic duplicated regions where orthologs of each paralog could be identified so that the paralogs could be used to root the tree. Recent radiations, such as those of emerging pathogens, often pose rooting challenges due to a lack of ingroup variation and large genomic differences with known outgroups. Using a phylogenomic approach, we created a robust, rooted phylogeny for C. burnetii. [Coxiella burnetii; paralog SNPs; pathogen evolution; phylogeny; recent radiation; root; rooting using duplicated genes.].


Subject(s)
Classification/methods , Coxiella burnetii/classification , Coxiella burnetii/genetics , Genomics , Phylogeny , Genome, Bacterial/genetics , Genomics/standards
10.
J Bacteriol ; 195(17): 3914-24, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23813730

ABSTRACT

Coxiella burnetii, the etiological agent of acute and chronic Q fever in humans, is a naturally intracellular pathogen that directs the formation of an acidic Coxiella-containing vacuole (CCV) derived from the host lysosomal network. Central to its pathogenesis is a specialized type IVB secretion system (T4SS) that delivers effectors essential for intracellular replication and CCV formation. Using a bioinformatics-guided approach, 234 T4SS candidate substrates were identified. Expression of each candidate as a TEM-1 ß-lactamase fusion protein led to the identification of 53 substrates that were translocated in a Dot/Icm-dependent manner. Ectopic expression in HeLa cells revealed that these substrates trafficked to distinct subcellular sites, including the endoplasmic reticulum, mitochondrion, and nucleus. Expression in Saccharomyces cerevisiae identified several substrates that were capable of interfering with yeast growth, suggesting that these substrates target crucial host processes. To determine if any of these T4SS substrates are necessary for intracellular replication, we isolated 20 clonal T4SS substrate mutants using the Himar1 transposon and transposase. Among these, 10 mutants exhibited defects in intracellular growth and CCV formation in HeLa and J774A.1 cells but displayed normal growth in bacteriological medium. Collectively, these results indicate that C. burnetii encodes a large repertoire of T4SS substrates that play integral roles in host cell subversion and CCV formation and suggest less redundancy in effector function than has been found in the comparative Legionella Dot/Icm model.


Subject(s)
Bacterial Proteins/metabolism , Coxiella burnetii/growth & development , Coxiella burnetii/metabolism , Vacuoles/microbiology , Virulence Factors/metabolism , Animals , Bacterial Proteins/genetics , Cell Line , Computational Biology , Coxiella burnetii/genetics , DNA, Bacterial/genetics , Epithelial Cells/microbiology , Humans , Macrophages/microbiology , Mice , Mutagenesis, Insertional , Protein Transport , Saccharomyces cerevisiae/growth & development , Virulence Factors/genetics
11.
Mol Cell Proteomics ; 10(10): M110.006304, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21817167

ABSTRACT

Antigen profiling using comprehensive protein microarrays is a powerful tool for characterizing the humoral immune response to infectious pathogens. Coxiella burnetii is a CDC category B bioterrorist infectious agent with worldwide distribution. In order to assess the antibody repertoire of acute and chronic Q fever patients we have constructed a protein microarray containing 93% of the proteome of Coxiella burnetii, the causative agent of Q fever. Here we report the profile of the IgG and IgM seroreactivity in 25 acute Q fever patients in longitudinal samples. We found that both early and late time points of infection have a very consistent repertoire of IgM and IgG response, with a limited number of proteins undergoing increasing or decreasing seroreactivity. We also probed a large collection of acute and chronic Q fever patient samples and identified serological markers that can differentiate between the two disease states. In this comparative analysis we confirmed the identity of numerous IgG biomarkers of acute infection, identified novel IgG biomarkers for acute and chronic infections, and profiled for the first time the IgM antibody repertoire for both acute and chronic Q fever. Using these results we were able to devise a test that can distinguish acute from chronic Q fever. These results also provide a unique perspective on isotype switch and demonstrate the utility of protein microarrays for simultaneously examining the dynamic humoral immune response against thousands of proteins from a large number of patients. The results presented here identify novel seroreactive antigens for the development of recombinant protein-based diagnostics and subunit vaccines, and provide insight into the development of the antibody response.


Subject(s)
Antigens, Bacterial/analysis , Coxiella burnetii/metabolism , Protein Array Analysis/methods , Proteome/analysis , Q Fever/immunology , Antibodies, Bacterial/genetics , Antibodies, Bacterial/immunology , Antibodies, Bacterial/metabolism , Biomarkers , Bioterrorism , Coxiella burnetii/genetics , Coxiella burnetii/immunology , Gene Expression Profiling , Humans , Immunity, Humoral/genetics , Immunoglobulin G/immunology , Immunoglobulin G/metabolism , Immunoglobulin M/immunology , Immunoglobulin M/metabolism , Proteome/immunology
12.
Proc Natl Acad Sci U S A ; 107(50): 21755-60, 2010 Dec 14.
Article in English | MEDLINE | ID: mdl-21098666

ABSTRACT

Coxiella burnetii is an obligate intracellular bacterial pathogen responsible for acute and chronic Q fever. This bacterium harbors a type IV secretion system (T4SS) highly similar to the Dot/Icm of Legionella pneumophila that is believed to be essential for its infectivity. Protein substrates of the Coxiella T4SS are predicted to facilitate the biogenesis of a phagosome permissive for its intracellular growth. However, due to the lack of genetic systems, protein transfer by the C. burnetii Dot/Icm has not been demonstrated. In this study, we report the identification of 32 substrates of the C. burnetii Dot/Icm system using a fluorescence-based ß-lactamase (TEM1) translocation assay as well as the calmodulin-dependent adenylate cyclase (CyaA) assay in the surrogate host L. pneumophila. Notably, 26 identified T4SS substrates are hypothetical proteins without predicted function. Candidate secretion substrates were obtained by using (i) a genetic screen to identify C. burnetii proteins interacting with DotF, a component of the T4SS, and (ii) bioinformatic approaches to retrieve candidate genes that harbor characteristics associated with previously reported substrates of the Dot/Icm system from both C. burnetii and L. pneumophila. Moreover, we have developed a shuttle plasmid that allows the expression of recombinant proteins in C. burnetii as TEM fusion products. Using this system, we demonstrated that a Dot/Icm substrate identified with L. pneumophila was also translocated by C. burnetii in a process that requires its C terminus, providing direct genetic evidence of a functional T4SS in C. burnetii.


Subject(s)
Bacterial Proteins/metabolism , Bacterial Secretion Systems/physiology , Coxiella burnetii/metabolism , Protein Transport/physiology , Bacterial Proteins/genetics , Computational Biology , Coxiella burnetii/genetics , Coxiella burnetii/pathogenicity , Genome, Bacterial , Legionella pneumophila/genetics , Legionella pneumophila/metabolism , Legionella pneumophila/pathogenicity , Two-Hybrid System Techniques
13.
Front Cell Infect Microbiol ; 13: 1160198, 2023.
Article in English | MEDLINE | ID: mdl-37153158

ABSTRACT

The long non-coding RNAs (lncRNAs) are evolutionarily conserved classes of non-coding regulatory transcripts of > 200 nucleotides in length. They modulate several transcriptional and post-transcriptional events in the organism. Depending on their cellular localization and interactions, they regulate chromatin function and assembly; and alter the stability and translation of cytoplasmic mRNAs. Although their proposed range of functionality remains controversial, there is increasing research evidence that lncRNAs play a regulatory role in the activation, differentiation and development of immune signaling cascades; microbiome development; and in diseases such as neuronal and cardiovascular disorders; cancer; and pathogenic infections. This review discusses the functional roles of different lncRNAs in regulation of host immune responses, signaling pathways during host-microbe interaction and infection caused by obligate intracellular bacterial pathogens. The study of lncRNAs is assuming significance as it could be exploited for development of alternative therapeutic strategies for the treatment of severe and chronic pathogenic infections caused by Mycobacterium, Chlamydia and Rickettsia infections, as well as commensal colonization. Finally, this review summarizes the translational potential of lncRNA research in development of diagnostic and prognostic tools for human diseases.


Subject(s)
Bacterial Infections , Neoplasms , RNA, Long Noncoding , Humans , RNA, Long Noncoding/metabolism , Bacterial Infections/genetics , Bacterial Infections/microbiology , Immunity
14.
Adv Exp Med Biol ; 984: 39-63, 2012.
Article in English | MEDLINE | ID: mdl-22711626

ABSTRACT

Survival of intracellular pathogenic bacteria depends on the ability to resist host-mediated degradation and to generate a replicative niche within the host. Usually, after internalization by professional phagocytic cells, the bacteria containing vacuole or phagosome traffics through the endocytic pathway, progressively acidifies and develops into a degradative mature phagolysosome. In this environment bacteria are exposed to a wide variety of anti-microbial agents, such as defensins, proteases, and reactive oxygen species (ROS) and reactive nitrogen species (RNS). Most parasitizing bacteria have evolved strategies to interfere with this maturation process and to direct the development of an environment that supports survival and replication. C. burnetii also follows this paradigm, but directs the biogenesis of a unique parasitophorous vacuole (PV), which resembles, yet is distinct from a terminal phagolysosome. Within the environment of the PV, C. burnetii is exposed to varying levels of ROS and RNS, which represent the primary defense mechanism of the host cell against this invading microorganism. Major mediators for ROS and RNS are superoxide (O (2) (-) ) and nitric oxide (NO(*)), generated by the cellular NADPH oxidase (phox) and inducible nitric oxide synthase (iNOS), respectively. C. burnetii employs several strategies to evade oxidative stress; on the host side (i) delaying phagolysosome fusion and (ii) inhibiting cellular NADPH oxidase. On the bacterial side, maintaining genome stability by (iii) evolving a preference for a low iron environment, (iv) expressing a minimal and likely crucial set of DNA repair genes and (v) detoxifying the PV by ROS and RNS degrading enzymes. Overall defense mechanisms in C. burnetii against oxidative and nitrosative stress and the regulation thereof are not fully defined and our knowledge is mainly based on genome sequence information. Comparison with E. coli as a model bacterium reveals that defense strategies of C. burnetii differ significantly and emphasize a highly adaptive evolution to this harsh and unique niche.


Subject(s)
Coxiella burnetii/physiology , Oxidative Stress/physiology , Adaptation, Physiological/genetics , Adaptation, Physiological/physiology , Animals , Coxiella burnetii/genetics , Coxiella burnetii/metabolism , DNA Damage , DNA Repair , Humans , Oxidative Stress/genetics , Phagosomes/genetics , Phagosomes/metabolism , Phagosomes/microbiology , Phagosomes/physiology
15.
Adv Exp Med Biol ; 984: 13-38, 2012.
Article in English | MEDLINE | ID: mdl-22711625

ABSTRACT

Coxiella burnetii, the causative agent of Q fever, has remained a public health concern since the identification of this organism in 1935 by E. H. Derrick in Australia and at the Rocky Mountain Laboratory in the USA by H.R. Cox and G. Davis. Human Q fever has been described in most countries where C. burnetii is ubiquitous in the environment except in New Zealand where no cases have been described. Most human infections are acquired through inhalation of contaminated aerosols that can lead to acute self-limiting febrile illness or more severe chronic cases of hepatitis or endocarditis. It is estimated that the actual incidence of human infection is under-reported as a result of imprecise tools for differential diagnosis. An intracellular lifestyle, low infectious dose, and ease of transmission have resulted in the classification of C. burnetii as a category B bio-warfare agent. The recent outbreaks in Europe are a reminder that there is much to learn about this unique intracellular pathogen, especially with the speculation of a hyper-virulent strain contributing to an outbreak in the Netherlands where over 4,000 human cases were reported. A new era in C. burnetii research has begun with the recent description of an axenic media making this an exciting time to study this bacterial pathogen.


Subject(s)
Coxiella burnetii/genetics , Coxiella burnetii/pathogenicity , Q Fever/microbiology , Disease Outbreaks , Genome, Bacterial , Genomics , Humans , Phylogeny , Public Health , Q Fever/epidemiology , Virulence/genetics
16.
Adv Exp Med Biol ; 984: 91-104, 2012.
Article in English | MEDLINE | ID: mdl-22711628

ABSTRACT

Coxiella burnetii is an obligate intracellular bacterium that causes a worldwide zoonotic disease, Q fever. Since C. burnetii infection could develop into severe chronic disease in humans, vaccination is the logical approach to prevent individuals at risk of natural and deliberate exposure. Although formalin-inactivated C. burnetii phase I vaccine (PIV) is effective in protecting vaccinated host against the infection in humans, widespread use of this vaccine is limited by its high incidence of adverse reactions, especially in individuals with prior immunity to the agent. Creation of a safe and effective vaccine to prevent Q fever remains an important goal for public health and international biosecurity. It is critical to clearly understand the mechanisms that involved in development of protective immunity against C. burnetii infection and to identify the key protective antigens for developing a safe and effective new generation vaccine against Q fever. This chapter describes new information related to the characterization of acquired immunity to C. burnetii vaccination and infection that will provide a fundamental understanding of the development of protective immunity against Q fever.


Subject(s)
Bacterial Vaccines/administration & dosage , Bacterial Vaccines/immunology , Coxiella burnetii/immunology , Q Fever/immunology , Q Fever/prevention & control , Animals , Humans , Q Fever/microbiology , Vaccination/methods
17.
Front Immunol ; 13: 886810, 2022.
Article in English | MEDLINE | ID: mdl-35693783

ABSTRACT

Coxiella burnetii is an obligate intracellular bacterium which, in humans, causes the disease Q fever. Although Q fever is most often a mild, self-limiting respiratory disease, it can cause a range of severe syndromes including hepatitis, myocarditis, spontaneous abortion, chronic valvular endocarditis, and Q fever fatigue syndrome. This agent is endemic worldwide, except for New Zealand and Antarctica, transmitted via aerosols, persists in the environment for long periods, and is maintained through persistent infections in domestic livestock. Because of this, elimination of this bacterium is extremely challenging and vaccination is considered the best strategy for prevention of infection in humans. Many vaccines against C. burnetii have been developed, however, only a formalin-inactivated, whole cell vaccine derived from virulent C. burnetii is currently licensed for use in humans. Unfortunately, widespread use of this whole cell vaccine is impaired due to the severity of reactogenic responses associated with it. This reactogenicity continues to be a major barrier to access to preventative vaccines against C. burnetii and the pathogenesis of this remains only partially understood. This review provides an overview of past and current research on C. burnetii vaccines, our knowledge of immunogenicity and reactogenicity in C. burnetii vaccines, and future strategies to improve the safety of vaccines against C. burnetii.


Subject(s)
Coxiella burnetii , Q Fever , Bacterial Vaccines , Female , Humans , Pregnancy , Vaccination/adverse effects , Vaccine Development
18.
Microbiol Spectr ; 10(4): e0248421, 2022 08 31.
Article in English | MEDLINE | ID: mdl-35913176

ABSTRACT

Coxiella burnetii requires a type IVB secretion system (T4SS) to promote intracellular replication and virulence. We hypothesized that Coxiella employs its T4SS to secrete effectors that enable stealthy colonization of immune cells. To address this, we used RNA sequencing to compare the transcriptional response of murine bone marrow-derived macrophages (BMDM) infected with those of wild-type Coxiella and a T4SS-null mutant at 8 and 24 h postinfection. We found a T4SS-independent upregulation of proinflammatory transcripts which was consistent with a proinflammatory polarization phenotype. Despite this, infected BMDM failed to completely polarize, as evidenced by modest surface expression of CD38 and CD11c, nitrate production, and reduced proinflammatory cytokine and chemokine secretion compared to positive controls. As these BMDM permitted replication of C. burnetii, we employed them to identify T4SS effectors that are essential in the specific cellular context of a primary macrophage. We found five Himar1 transposon mutants in T4SS effectors that had a replication defect in BMDM but not J774A.1 cells. The mutants were also attenuated in a SCID mouse model of infection. Among these candidate virulence factors, we found that CBU1639 contributed to the inhibition of macrophage proinflammatory responses to Coxiella infection. These data demonstrate that while T4SS is dispensable for the stealthy invasion of primary macrophages, Coxiella has evolved multiple T4SS effectors that specifically target macrophage function to proliferate within that specific cellular context. IMPORTANCE Coxiella burnetii, the causative agent of Q fever, preferentially infects macrophages of the respiratory tract when causing human disease. This work describes how primary macrophages respond to C. burnetii at the earliest stages of infection, before bacterial replication. We found that while infected macrophages increase expression of proinflammatory genes after bacterial entry, they fail to activate the accompanying antibacterial functions that might ultimately control the infection. This disconnect between initial response and downstream function was not mediated by the bacterium's type IVB secretion system, suggesting that Coxiella has other virulence factors that dampen host responses early in the infection process. Nevertheless, we were able to identify several type IVB secreted effectors that were specifically required for survival in macrophages and mice. This work is the first to identify type IVB secretion effectors that are specifically required for infection and replication within primary macrophages.


Subject(s)
Coxiella burnetii , Q Fever , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Coxiella burnetii/genetics , Host-Pathogen Interactions/physiology , Humans , Macrophages/microbiology , Mice , Mice, SCID , Q Fever/metabolism , Q Fever/microbiology , Virulence Factors/genetics , Virulence Factors/metabolism
19.
Microbiology (Reading) ; 157(Pt 2): 526-542, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21030434

ABSTRACT

Coxiella burnetii is a Gram-negative obligate intracellular pathogen and the causative agent of Q fever in humans. Q fever causes acute flu-like symptoms and may develop into a chronic disease leading to endocarditis. Its potential as a bioweapon has led to its classification as a category B select agent. An effective inactivated whole-cell vaccine (WCV) currently exists but causes severe granulomatous/necrotizing reactions in individuals with prior exposure, and is not licensed for use in most countries. Current efforts to reduce or eliminate the deleterious reactions associated with WCVs have focused on identifying potential subunit vaccine candidates. Both humoral and T cell-mediated responses are required for protection in animal models. In this study, nine novel immunogenic C. burnetii proteins were identified in extracted whole-cell lysates using 2D electrophoresis, immunoblotting with immune guinea pig sera, and tandem MS. The immunogenic C. burnetii proteins elicited antigen-specific IgG in guinea pigs vaccinated with whole-cell killed Nine Mile phase I vaccine, suggesting a T cell-dependent response. Eleven additional proteins previously shown to react with immune human sera were also antigenic in guinea pigs, showing the relevance of the guinea pig immunization model for antigen discovery. The antigens described here warrant further investigation to validate their potential use as subunit vaccine candidates.


Subject(s)
Antibody Formation , Antigens, Bacterial/immunology , Coxiella burnetii/immunology , Animals , Antibodies, Bacterial/blood , Antigens, Bacterial/metabolism , Bacterial Vaccines/immunology , Coxiella burnetii/metabolism , Electrophoresis, Gel, Two-Dimensional , Female , Guinea Pigs , Immunoglobulin G/blood , Q Fever/immunology , Q Fever/prevention & control , T-Lymphocytes/immunology , Tandem Mass Spectrometry
20.
Appl Environ Microbiol ; 77(11): 3720-5, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21478315

ABSTRACT

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.


Subject(s)
Bacteriological Techniques/methods , Coxiella burnetii/genetics , Coxiella burnetii/isolation & purification , Culture Media/chemistry , Genetics, Microbial/methods , Transformation, Bacterial , Animals , Coxiella burnetii/growth & development , Coxiella burnetii/ultrastructure , Mice , Microscopy, Electron , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL