Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
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
2.
Sci Rep ; 13(1): 6021, 2023 04 13.
Article in English | MEDLINE | ID: mdl-37055450

ABSTRACT

Limited data significantly hinders our capability of biothreat assessment of novel bacterial strains. Integration of data from additional sources that can provide context about the strain can address this challenge. Datasets from different sources, however, are generated with a specific objective and which makes integration challenging. Here, we developed a deep learning-based approach called the neural network embedding model (NNEM) that integrates data from conventional assays designed to classify species with new assays that interrogate hallmarks of pathogenicity for biothreat assessment. We used a dataset of metabolic characteristics from a de-identified set of known bacterial strains that the Special Bacteriology Reference Laboratory (SBRL) of the Centers for Disease Control and Prevention (CDC) has curated for use in species identification. The NNEM transformed results from SBRL assays into vectors to supplement unrelated pathogenicity assays from de-identified microbes. The enrichment resulted in a significant improvement in accuracy of 9% for biothreat. Importantly, the dataset used in our analysis is large, but noisy. Therefore, the performance of our system is expected to improve as additional types of pathogenicity assays are developed and deployed. The proposed NNEM strategy thus provides a generalizable framework for enrichment of datasets with previously collected assays indicative of species.


Subject(s)
Bacteria , Neural Networks, Computer , United States
3.
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
4.
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
5.
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
6.
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
7.
Front Immunol ; 12: 754712, 2021.
Article in English | MEDLINE | ID: mdl-34616410

ABSTRACT

Q-VAX®, a whole cell, formalin-inactivated vaccine, is the only vaccine licensed for human use to protect against Coxiella burnetii, the cause of Q fever. Although this vaccine provides long-term protection, local and systemic reactogenic responses are common in previously sensitized individuals which prevents its use outside of Australia. Despite the importance of preventing these adverse reactions to develop widely accepted, novel vaccines against C. burnetii, little is understood about the underlying cellular mechanisms. This is mostly attributed to the use of a guinea pig reactogenicity model where complex cellular analysis is limited. To address this, we compared three different mouse strains develop a model of C. burnetii whole cell vaccine reactogenic responses. SKH1 and C57Bl/6, but not BALBc mice, develop local granulomatous reactions after either infection- or vaccine-induced sensitization. We evaluated local and systemic responses by measuring T cell populations from the vaccination site and spleen during elicitation using flow cytometry. Local reaction sites showed influx of IFNγ+ and IL17a+ CD4 T cells in sensitized mice compared with controls and a reduction in IL4+ CD4 T cells. Additionally, sensitized mice showed a systemic response to elicitation by an increase in IFNγ+ and IL17a+ CD4 T cells in the spleen. These results indicate that local and systemic C. burnetii reactogenic responses are consistent with a Th1 delayed-type hypersensitivity. Our experiments provide insights into the pathophysiology of C. burnetii whole cell vaccine reactogenicity and demonstrate that C57Bl/6 and SKH1 mice can provide a valuable model for evaluating the reactogenicity of novel C. burnetii vaccine candidates.


Subject(s)
Bacterial Vaccines/adverse effects , Disease Models, Animal , Hypersensitivity, Delayed/immunology , Q Fever , Th1 Cells/immunology , Animals , Coxiella burnetii , Female , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Q Fever/prevention & control , Vaccines, Inactivated/adverse effects
8.
J Vis Exp ; (175)2021 09 17.
Article in English | MEDLINE | ID: mdl-34605819

ABSTRACT

Identification of emerging bacterial pathogens is critical for human health and security. Bacterial adherence to host cells is an essential step in bacterial infections and constitutes a hallmark of potential threat. Therefore, examining the adherence of bacteria to host cells can be used as a component of bacterial threat assessment. A standard method for enumerating bacterial adherence to host cells is to co-incubate bacteria with host cells, harvest the adherent bacteria, plate the harvested cells on solid media, and then count the resultant colony forming units (CFU). Alternatively, bacterial adherence to host cells can be evaluated using immunofluorescence microscopy-based approaches. However, conventional strategies for implementing these approaches are time-consuming and inefficient. Here, a recently developed automated fluorescence microscopy-based imaging method is described. When combined with high-throughput image processing and statistical analysis, the method enables rapid quantification of bacteria that adhere to host cells. Two bacterial species, Gram-negative Pseudomonas aeruginosa and Gram-positive Listeria monocytogenes and corresponding negative controls, were tested to demonstrate the protocol. The results show that this approach rapidly and accurately enumerates adherent bacteria and significantly reduces experimental workloads and timelines.


Subject(s)
Bacterial Adhesion , Humans
9.
Autophagy ; 17(3): 706-722, 2021 03.
Article in English | MEDLINE | ID: mdl-32116095

ABSTRACT

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.


Subject(s)
Autophagy/physiology , Bacterial Secretion Systems/metabolism , Coxiella/metabolism , Host-Pathogen Interactions/immunology , Vacuoles/microbiology , Animals , Bacterial Proteins/metabolism , Coxiella burnetii/growth & development , Coxiella burnetii/metabolism , Humans , Mice , Vacuoles/metabolism
10.
Cell Rep Med ; 2(12): 100461, 2021 12 21.
Article in English | MEDLINE | ID: mdl-35028605

ABSTRACT

Q fever is caused by the intracellular bacterium Coxiella burnetii, for which there is no approved vaccine in the United States. A formalin-inactivated whole-cell vaccine (WCV) from virulent C. burnetii NMI provides single-dose long-lived protection, but concerns remain over vaccine reactogenicity. We therefore sought an alternate approach by purifying native C. burnetii antigens from the clonally derived avirulent NMII strain. A soluble bacterial extract, termed Sol II, elicits high-titer, high-avidity antibodies and induces a CD4 T cell response that confers protection in naive mice. In addition, Sol II protects against pulmonary C. burnetii challenge in three animal models without inducing hypersensitivity. An NMI-derived extract, Sol I, enhances protection further and outperforms the WCV gold standard. Collectively, these data represent a promising approach to design highly effective, non-reactogenic Q fever vaccines.


Subject(s)
Antigens, Bacterial/immunology , Coxiella burnetii/immunology , Hypersensitivity/immunology , Immunity , Q Fever/immunology , Q Fever/prevention & control , Aerosols , Animals , Antibody Affinity , Antigenic Variation , Bacterial Vaccines/immunology , CD4-Positive T-Lymphocytes/immunology , Disease Models, Animal , Female , Guinea Pigs , Immunization , Lipopolysaccharides , Lung/microbiology , Lung/pathology , Macaca mulatta , Male , Mice, Inbred C57BL , Q Fever/microbiology , Solubility
11.
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
12.
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
13.
Microbes Infect ; 20(5): 302-307, 2018 05.
Article in English | MEDLINE | ID: mdl-29331581

ABSTRACT

Coxiella burnetii is an obligate intracellular pathogen that replicates in an endolysosome-like compartment termed the Coxiella-containing vacuole (CCV). Formation of this unique replicative niche requires delivery of bacterial effector proteins into the host cytosol where they mediate crucial interactions with the host. We previously identified an essential Dot/Icm effector, CirA that is required for intracellular replication and CCV formation. Furthermore, CirA was shown to stimulate the GTPase activity of RhoA in vitro. In the current study, we used a bioinformatics-guided approach and identified three arginine finger-like motifs, often found in Rho GTPase-activating proteins (GAPs) and endosome-lysosome basolateral sorting signals associated with vesicle trafficking. When expressed in mammalian cells, mutation of either endosome-lysosome-basolateral sorting signals or the arginine finger-like motifs rescued stress phenotypes and decreased plasma membrane localization of ectopically expressed CirA. We further demonstrate that endosome-lysosome sorting signals are required for co-localization with Rab5 and Rab7. Collectively our data indicate that arginine finger-like motifs and endosome-lysosome-basolateral sorting signals within CirA are essential for interaction with the host cytoskeleton.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Coxiella burnetii/metabolism , Endosomes/metabolism , Lysosomes/metabolism , Type IV Secretion Systems/metabolism , Amino Acid Motifs/genetics , Amino Acid Motifs/physiology , Bacterial Proteins/genetics , Cell Membrane/metabolism , Coxiella burnetii/genetics , HeLa Cells , Host-Pathogen Interactions , Humans , Protein Transport , Q Fever/metabolism , Q Fever/microbiology , Saccharomyces cerevisiae/metabolism , Type IV Secretion Systems/genetics , Vacuoles/metabolism , Vacuoles/microbiology , rab GTP-Binding Proteins/metabolism , rhoA GTP-Binding Protein/metabolism
14.
Cell Host Microbe ; 21(5): 637-649.e6, 2017 May 10.
Article in English | MEDLINE | ID: mdl-28494245

ABSTRACT

Cryptococcus neoformans (Cn) is a deadly fungal pathogen whose intracellular lifestyle is important for virulence. Host mechanisms controlling fungal phagocytosis and replication remain obscure. Here, we perform a global phosphoproteomic analysis of the host response to Cryptococcus infection. Our analysis reveals numerous and diverse host proteins that are differentially phosphorylated following fungal ingestion by macrophages, thereby indicating global reprogramming of host kinase signaling. Notably, phagocytosis of the pathogen activates the host autophagy initiation complex (AIC) and the upstream regulatory components LKB1 and AMPKα, which regulate autophagy induction through their kinase activities. Deletion of Prkaa1, the gene encoding AMPKα1, in monocytes results in resistance to fungal colonization of mice. Finally, the recruitment of AIC components to nascent Cryptococcus-containing vacuoles (CnCVs) regulates the intracellular trafficking and replication of the pathogen. These findings demonstrate that host AIC regulatory networks confer susceptibility to infection and establish a proteomic resource for elucidating host mechanisms that regulate fungal intracellular parasitism.


Subject(s)
Cryptococcosis/immunology , Cryptococcus neoformans/genetics , Cryptococcus neoformans/pathogenicity , Host-Pathogen Interactions/immunology , Signal Transduction/physiology , Virulence/genetics , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Animals , Apoptosis Regulatory Proteins/metabolism , Autophagy/physiology , Autophagy-Related Protein-1 Homolog/genetics , Autophagy-Related Protein-1 Homolog/metabolism , Biological Transport/physiology , Cell Line , Coxiella burnetii/pathogenicity , Cryptococcosis/microbiology , Cryptococcus neoformans/growth & development , Cryptococcus neoformans/metabolism , Disease Models, Animal , Female , Fungal Proteins/genetics , Fungal Proteins/metabolism , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/physiology , Macrophages/immunology , Macrophages/metabolism , Macrophages/microbiology , Mice , Mice, Inbred C57BL , Monocytes/metabolism , Phagocytosis , Protein Serine-Threonine Kinases/metabolism , Proteomics , RAW 264.7 Cells , Vacuoles/microbiology , Virulence/physiology
15.
Article in English | MEDLINE | ID: mdl-28217558

ABSTRACT

Coxiella burnetii is an intracellular, zoonotic pathogen that is the causative agent of Q fever. Infection most frequently occurs after inhalation of contaminated aerosols, which can lead to acute, self-limiting febrile illness or more serve chronic infections such as hepatitis or endocarditis. Macrophages are the principal target cells during infection where C. burnetii resides and replicates within a unique phagolysosome-like compartment, the Coxiella-containing vacuole (CCV). The first virulence determinant described as necessary for infection was full-length lipopolysaccarride (LPS); spontaneous rough mutants (phase II) arise after passage in immuno-incompetent hosts. Phase II C. burnetii are attenuated in immuno-competent animals, but are fully capable of infecting a variety of host cells in vitro. A clonal strain of the Nine Mile isolate (RSA439, clone 4), has a 26 KDa chromosomal deletion that includes LPS biosynthetic genes and is uniquely approved for use in BL2/ABL2 conditions. With the advances of axenic media and genetic tools for C. burnetii research, the characterization of novel virulence determinants is ongoing and almost exclusively performed using this attenuated clone. A major problem with predicting essential virulence loci with RSA439 is that, although some cell-autonomous phenotypes can be assessed in tissue culture, no animal model for assessing pathogenesis has been defined. Here we describe the use of SCID mice for predicting virulence factors of C. burnetii, in either independent or competitive infections. We propose that this model allows for the identification of mutations that are competent for intracellular replication in vitro, but attenuated for growth in vivo and predict essential innate immune responses modulated by the pathogen during infection as a central pathogenic strategy.


Subject(s)
Coxiella burnetii/pathogenicity , Disease Models, Animal , Host-Pathogen Interactions , Q Fever/microbiology , Q Fever/pathology , Virulence Factors/analysis , Animals , Mice , Mice, SCID , Virulence
16.
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
17.
PLoS One ; 10(9): e0136699, 2015.
Article in English | MEDLINE | ID: mdl-26366725

ABSTRACT

Coxiella burnetii is a Gram-negative, obligate intracellular bacterium and the causative agent of Q fever. Infections are usually acquired after inhalation of contaminated particles, where C. burnetii infects its cellular target cells, alveolar macrophages. Respiratory pathogens encounter the C-type lectin surfactant protein D (SP-D) during the course of natural infection. SP-D is a component of the innate immune response in the lungs and other mucosal surfaces. Many Gram-negative pulmonary pathogens interact with SP-D, which can cause aggregation, bactericidal effects and aid in bacterial clearance. Here we show that SP-D binds to C. burnetii in a calcium-dependent manner with no detectable bacterial aggregation or bactericidal effects. Since SP-D interactions with bacteria often alter macrophage interactions, it was determined that SP-D treatment resulted in a significant decrease in C. burnetii interactions to a mouse alveolar macrophage model cell line MH-S indicating SP-D causes a significant decrease in phagocytosis. The ability of SP-D to modulate macrophage activation by C. burnetii was tested and it was determined that SP-D does not alter the correlates measured for macrophage activation. Taken together these studies support those demonstrating limited activation of alveolar macrophages with C. burnetii and demonstrate interactions with SP-D participate in reduction of phagocyte attachment and phagocytosis.


Subject(s)
Anti-Infective Agents/pharmacology , Coxiella burnetii/drug effects , Macrophages/microbiology , Pulmonary Surfactant-Associated Protein D/pharmacology , Animals , Cell Line , Coxiella burnetii/pathogenicity , Macrophages/immunology , Mice , Phagocytosis
18.
Nat Rev Microbiol ; 11(8): 561-73, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23797173

ABSTRACT

The agent of Q fever, Coxiella burnetii, is an obligate intracellular bacterium that causes acute and chronic infections. The study of C. burnetii pathogenesis has benefited from two recent fundamental advances: improved genetic tools and the ability to grow the bacterium in extracellular media. In this Review, we describe how these recent advances have improved our understanding of C. burnetii invasion and host cell modulation, including the formation of replication-permissive Coxiella-containing vacuoles. Furthermore, we describe the Dot/Icm (defect in organelle trafficking/intracellular multiplication) system, which is used by C. burnetii to secrete a range of effector proteins into the host cell, and we discuss the role of these effectors in remodelling the host cell.


Subject(s)
Coxiella burnetii/pathogenicity , Host-Pathogen Interactions , Q Fever/microbiology , Animals , Apoptosis , Bacterial Adhesion , Coxiella burnetii/genetics , Humans , Integrin alphaVbeta3/genetics , Integrin alphaVbeta3/metabolism , Intracellular Membranes/metabolism , Intracellular Membranes/microbiology , Phagocytosis , Phagosomes/metabolism , Phagosomes/microbiology , Time Factors , Vacuoles/microbiology
19.
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
20.
Infect Immun ; 77(10): 4275-83, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19620343

ABSTRACT

Burkholderia pseudomallei, the causative agent of melioidosis, has often been called the great "mimicker," and clinical disease due to this organism may include acute, chronic, and latent pulmonary infections. Interestingly, chronic pulmonary melioidosis is often mistaken for tuberculosis, and this can have significant consequences, as the treatments for these two infections are radically different. The recurrent misdiagnosis of melioidosis for tuberculosis has caused many to speculate that these two bacterial pathogens use similar pathways to produce latent infections. Here we show that isocitrate lyase is a persistence factor for B. pseudomallei, and inhibiting the activity of this enzyme during experimental chronic B. pseudomallei lung infection forces the infection into an acute state, which can then be treated with antibiotics. We found that if antibiotics are not provided in combination with isocitrate lyase inhibitors, the resulting B. pseudomallei infection overwhelms the host, resulting in death. These results suggest that the inhibition of isocitrate lyase activity does not necessarily attenuate virulence as previously observed for Mycobacterium tuberculosis infections but does force the bacteria into a replicating state where antibiotics are effective. Therefore, isocitrate lyase inhibitors could be developed for chronic B. pseudomallei infections but only for use in combination with effective antibiotics.


Subject(s)
Anti-Bacterial Agents/pharmacology , Burkholderia pseudomallei/enzymology , Enzyme Inhibitors/pharmacology , Isocitrate Lyase/antagonists & inhibitors , Melioidosis/drug therapy , Melioidosis/microbiology , Animals , Burkholderia pseudomallei/pathogenicity , Colony Count, Microbial , Isocitrate Lyase/physiology , Lethal Dose 50 , Lung/microbiology , Rats , Survival Analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...