Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 695
Filter
1.
PLoS One ; 19(5): e0303877, 2024.
Article in English | MEDLINE | ID: mdl-38771828

ABSTRACT

Coxiella burnetii, the causative agent of Q fever, is a zoonotic bacteria of global public health significance. The organism has a complex, diverse, and relatively poorly understood animal reservoir but there is increasing evidence that macropods play some part in the epidemiology of Q fever in Australia. The aim of this cross-sectional survey was to estimate the animal- and tissue-level prevalence of coxiellosis amongst eastern grey (Macropus giganteus) and red (Osphranter rufus) kangaroos co-grazing with domestic cattle in a Q fever endemic area in Queensland. Serum, faeces and tissue samples from a range of organs were collected from 50 kangaroos. A total of 537 tissue samples were tested by real-time PCR, of which 99 specimens from 42 kangaroos (84% of animals, 95% confidence interval [CI], 71% to 93%) were positive for the C. burnetii IS1111 gene when tested in duplicate. Twenty of these specimens from 16 kangaroos (32%, 95% CI 20% to 47%) were also positive for the com1 or htpAB genes. Serum antibodies were present in 24 (57%, 95% CI 41% to 72%) of the PCR positive animals. There was no statistically significant difference in PCR positivity between organs and no single sample type consistently identified C. burnetii positive kangaroos. The results from this study identify a high apparent prevalence of C. burnetii amongst macropods in the study area, albeit seemingly with an inconsistent distribution within tissues and in relatively small quantities, often verging on the limits of detection. We recommend Q fever surveillance in macropods should involve a combination of serosurveys and molecular testing to increase chances of detection in a population, noting that a range of tissues would likely need to be sampled to confirm the diagnosis in a suspect positive animal.


Subject(s)
Antibodies, Bacterial , Coxiella burnetii , Macropodidae , Q Fever , Animals , Coxiella burnetii/genetics , Coxiella burnetii/immunology , Macropodidae/microbiology , Queensland/epidemiology , Q Fever/epidemiology , Q Fever/veterinary , Q Fever/microbiology , Q Fever/immunology , Antibodies, Bacterial/blood , Antibodies, Bacterial/immunology , Livestock/microbiology , Cattle , Cross-Sectional Studies
2.
Front Immunol ; 13: 915244, 2022.
Article in English | MEDLINE | ID: mdl-35833118

ABSTRACT

Vγ9Vδ2 T cells have been reported to participate to the immune response against infectious diseases such as the Q fever caused by Coxiella burnetii infection. Indeed, the number and proportion of Vγ9Vδ2 T cells are increased during the acute phase of Q fever. Human Vγ9Vδ2 T cell responses are triggered by phosphoantigens (pAgs) produced by pathogens and malignant cells, that are sensed via the membrane receptors butyrophilin-3A1 (BTN3A1) and -2A1 (BTN2A1). Here, by using CRISPR-Cas9 inactivation in THP-1 cells, we show that BTN3A and BTN2A are required to Vγ9Vδ2 T cell response to C. burnetii infection, though not directly involved in the infection process. Furthermore, C. burnetii-infected monocytes display increased BTN3A and BTN2A expression and induce Vγ9Vδ2 T cell activation that can be inhibited by specific antagonist mAb. More importantly, we show that the antimicrobial functions of Vγ9Vδ2 T cells towards C. burnetii are enhanced in the presence of an BTN3A activating antibody. This supports the role of Vγ9Vδ2 T cells in the control of C. burnetii infection and argues in favor of targeting these cells as an alternative treatment strategy for infectious diseases caused by intracellular bacteria.


Subject(s)
Antigens, CD , Butyrophilins , Communicable Diseases , Coxiella burnetii , Q Fever , Antigens, CD/immunology , Butyrophilins/immunology , Humans , Q Fever/immunology , Receptors, Antigen, T-Cell, gamma-delta/immunology , T-Lymphocytes/immunology
3.
Elife ; 112022 02 09.
Article in English | MEDLINE | ID: mdl-35137689

ABSTRACT

Background: Chronic Q fever is a zoonosis caused by the bacterium Coxiella burnetii which can manifest as infection of an abdominal aortic aneurysm (AAA). Antibiotic therapy often fails, resulting in severe morbidity and high mortality. Whereas previous studies have focused on inflammatory processes in blood, the aim of this study was to investigate local inflammation in aortic tissue. Methods: Multiplex immunohistochemistry was used to investigate local inflammation in Q fever AAAs compared to atherosclerotic AAAs in aorta tissue specimen. Two six-plex panels were used to study both the innate and adaptive immune systems. Results: Q fever AAAs and atherosclerotic AAAs contained similar numbers of CD68+ macrophages and CD3+ T cells. However, in Q fever AAAs, the number of CD68+CD206+ M2 macrophages was increased, while expression of GM-CSF was decreased compared to atherosclerotic AAAs. Furthermore, Q fever AAAs showed an increase in both the number of CD8+ cytotoxic T cells and CD3+CD8-FoxP3+ regulatory T cells. Finally, Q fever AAAs did not contain any well-defined granulomas. Conclusions: These findings demonstrate that despite the presence of pro-inflammatory effector cells, persistent local infection with C. burnetii is associated with an immune-suppressed microenvironment. Funding: This work was supported by SCAN consortium: European Research Area - CardioVascualar Diseases (ERA-CVD) grant [JTC2017-044] and TTW-NWO open technology grant [STW-14716].


Subject(s)
Adaptive Immunity/immunology , Aortic Aneurysm, Abdominal/immunology , Atherosclerosis/immunology , Immunity, Innate/immunology , Q Fever/immunology , Aged , Aortic Aneurysm, Abdominal/metabolism , Aortic Aneurysm, Abdominal/microbiology , Atherosclerosis/metabolism , Atherosclerosis/microbiology , Female , Humans , Immunohistochemistry/methods , Inflammation/immunology , Inflammation/microbiology , Macrophages/metabolism , Male , Middle Aged , Q Fever/metabolism , Q Fever/microbiology , T-Lymphocytes/metabolism
4.
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
5.
Front Immunol ; 12: 754690, 2021.
Article in English | MEDLINE | ID: mdl-34795669

ABSTRACT

This study aimed to explore if viable C. burnetii avirulent Nine Mile phase II (NMII) can elicit protective immunity against virulent NM phase I (NMI) infection. Interestingly, mice immunized with viable NMII elicited significant protection against NMI infection at different time points post-immunization. Viable NMII induced a dose-dependent NMI-specific IgG response in mice, but all doses of NMII-immunized mice conferred a similar level of protection. Comparing different routes of immunization indicated that intranasally immunized mice showed significantly higher levels of protection than other immunization routes. The observation that viable NMII induced a similar level of long-term protection against NMI challenge as the formalin-inactivated NMI vaccine (PIV) suggests that viable NMII bacteria can induce a similar level of long-term protection against virulent NMI challenge as the PIV. Viable NMII also induced significant protection against challenge with virulent Priscilla and Scurry strains, suggesting that viable NMII can elicit broad protection. Immune sera and splenocytes from viable NMII-immunized mice are protective against NMI infection, but immune serum-receiving mice did not control NMI replication. Additionally, viable NMII conferred a comparable level of protection in wild-type, CD4+ T cell-deficient, and CD8+ T cell-deficient mice, and partial protection in B cell-deficient mice. However, NMII-immunized T cell-deficient mice were unable to prevent C. burnetii replication. Thus, both B cells and T cells are required for viable NMII-induced protective immunity but T cells may play a critical role. Collectively, this study demonstrates the feasibility of using avirulent NMII as a live attenuated vaccine against human Q fever.


Subject(s)
Bacterial Vaccines/immunology , Coxiella burnetii/immunology , Q Fever/immunology , Vaccines, Attenuated/immunology , Animals , B-Lymphocytes/immunology , Feasibility Studies , Mice , Q Fever/prevention & control , T-Lymphocytes/immunology
6.
Virulence ; 12(1): 2461-2473, 2021 12.
Article in English | MEDLINE | ID: mdl-34516359

ABSTRACT

Coxiella burnetii is an obligate intracellular bacterium that causes the human disease Q fever, which can manifest as an acute flu-like illness or a long-term chronic illness, such as endocarditis. Three genotypes (ST8, ST16, and ST20) of Coxiella burnetii are commonly found in the contemporary US and are associated with specific animal hosts. Although all three genotypes have been isolated from humans with Q fever, studies comparing virulence between C. burnetii sequence types have been rare. Here, groups of mice were infected via aerosol inoculation with isolates derived from cow's milk, environmental, animal, and human samples. Mice were monitored for weight loss and blood samples were takenweekly. Animals were euthanized at 2- and 12-weeks post-infection, and bacterial burden was determined for tissues by real-time PCR. The levels of anti-Coxiella antibodies and selected inflammatory cytokines were determined for serum samples. Weight loss and splenomegaly were observed in mice infected with ST20 and ST16 isolates but were absent in the mice infected with ST8 isolates. Bacterial concentrations in the tissues were lower in the ST8 isolates at 2 weeks post-infection relative to all other isolates. ST16 and ST20 isolates induced robust antibody and cytokine responses, while ST8 isolates produced significantly lower anti-C. burnetii titers early in the infection but saw increased titers in some animals several weeks post-infection. The data suggest that the ST8 isolates are less virulent in this mouse model, as they produce less robust antibody responses that are slow to develop, relative to the ST16 and ST20 isolates.


Subject(s)
Coxiella burnetii , Q Fever , Animals , Antibodies, Bacterial/immunology , Antibody Formation , Coxiella burnetii/genetics , Cytokines/immunology , Female , Genotype , Mice , Q Fever/immunology , United States , Virulence , Weight Loss
7.
Front Immunol ; 12: 701811, 2021.
Article in English | MEDLINE | ID: mdl-34394097

ABSTRACT

For the zoonotic disease Q fever, serological analysis plays a dominant role in the diagnosis of Coxiella burnetii infection and in pre-screening for past exposure prior to vaccination. A number of studies suggest that assessment of C. burnetii-specific T-cell IFNγ responses may be a more sensitive tool to assess past exposure. In this study, we assessed the performance of a whole blood C. burnetii IFNγ release assay in comparison to serological detection in an area of high Q fever incidence in 2014, up to seven years after initial exposure during the Dutch Q fever outbreak 2007-2010. In a cohort of >1500 individuals from the Dutch outbreak village of Herpen, approximately 60% had mounted IFNγ responses to C. burnetii. This proportion was independent of the Coxiella strain used for stimulation and much higher than the proportion of individuals scored sero-positive using the serological gold standard immunofluorescence assay. Moreover, C. burnetii-specific IFNγ responses were found to be more durable than antibody responses in two sub-groups of individuals known to have sero-converted as of 2007 or previously reported to the municipality as notified Q fever cases. A novel ready-to-use version of the IFNγ release assay assessed in a subgroup of pre-exposed individuals in 2021 (10-14 years post exposure) proved again to be more sensitive than serology in detecting past exposure. These data demonstrate that C. burnetii-induced IFNγ release is indeed a more sensitive and durable marker of exposure to C. burnetii than are serological responses. In combination with a simplified assay version suitable for implementation in routine diagnostic settings, this makes the assessment of IFNγ responses a valuable tool for exposure screening to obtain epidemiological data, and to identify previously exposed individuals in pre-vaccination screens.


Subject(s)
Antibodies, Bacterial/immunology , Antibody Formation/immunology , Biomarkers/blood , Coxiella burnetii/immunology , Interferon-gamma/blood , Interferon-gamma/immunology , Animals , Cross-Sectional Studies , Humans , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/microbiology , Q Fever/blood , Q Fever/immunology , Q Fever/microbiology , Zoonoses/blood , Zoonoses/immunology , Zoonoses/microbiology
8.
Front Immunol ; 12: 653092, 2021.
Article in English | MEDLINE | ID: mdl-33815413

ABSTRACT

Q fever is caused by the obligate intracellular bacterium, Coxiella burnetii, a designated potential agent of bioterrorism because of its route of transmission, resistance to disinfectants, and low infectious dose. The only vaccine licensed for human use is Q-VAX® (Seqirus, licensed in Australia), a formalin-inactivated whole-cell vaccine, which produces severe local and systemic reactogenic responses in previously sensitized individuals. Accordingly, the U.S. Food and Drug Administration and other regulatory bodies around the world, have been reluctant to approve Q-VAX for widespread use. To obviate these adverse reactions, we prepared recombinant protein subunit vaccine candidates containing purified CBU1910, CBU0307, CBU0545, CBU0612, CBU0891, and CBU1398 proteins and TLR triagonist adjuvants. TLR triagonist adjuvants combine different TLR agonists to enhance immune responses to vaccine antigens. We tested both the protective efficacy and reactogenicity of our vaccine candidates in Hartley guinea pigs using intratracheal infection with live C. burnetii. While all of our candidates showed varying degrees of protection during challenge, local reactogenic responses were significantly reduced for one of our vaccine candidates when compared with a formalin-inactivated whole-cell vaccine. Our findings show that subunit vaccines combined with novel TLR triagonist adjuvants can generate protective immunity to C. burnetii infection while reducing reactogenic responses.


Subject(s)
Adjuvants, Immunologic/pharmacology , Bacterial Vaccines/pharmacology , Coxiella burnetii/immunology , Q Fever/prevention & control , Toll-Like Receptors/antagonists & inhibitors , Adjuvants, Immunologic/therapeutic use , Animals , Antigens, Bacterial/genetics , Antigens, Bacterial/pharmacology , Antigens, Bacterial/therapeutic use , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Bacterial Vaccines/genetics , Bacterial Vaccines/therapeutic use , Disease Models, Animal , Guinea Pigs , Humans , Immunogenicity, Vaccine , Q Fever/immunology , Q Fever/microbiology , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use , Vaccines, Subunit/genetics , Vaccines, Subunit/pharmacology , Vaccines, Subunit/therapeutic use , Vaccines, Synthetic/genetics , Vaccines, Synthetic/pharmacology , Vaccines, Synthetic/therapeutic use
9.
Eur J Clin Microbiol Infect Dis ; 40(7): 1569-1572, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33566203

ABSTRACT

We evaluated the long-term serological follow-up of patients with vascular risk factors for chronic Q fever that were previously Coxiella burnetii seropositive. C. burnetii phase I IgG titers were reevaluated in patients that gave informed consent or retrospectively collected in patients already deceased or lost to follow-up. Of 107 patients, 25 (23.4%) became seronegative, 77 (72.0%) retained a profile of past resolved Q fever infection, and five (4.7%) developed chronic Q fever. We urge clinicians to stay vigilant for chronic Q fever beyond two years after primary infection and perform serological testing based on clinical presentation.


Subject(s)
Antibodies, Bacterial/blood , Coxiella burnetii , Q Fever/blood , Aged , Antibodies, Bacterial/immunology , Female , Humans , Immunoglobulin G/blood , Male , Middle Aged , Q Fever/drug therapy , Q Fever/immunology , Q Fever/microbiology , Retrospective Studies , Risk Factors
10.
Vet Pathol ; 58(4): 624-642, 2021 07.
Article in English | MEDLINE | ID: mdl-33357072

ABSTRACT

Coxiella burnetii, a highly adapted obligate intracellular bacterial pathogen and the cause of the zoonosis Q fever, is a reemerging public health threat. C. burnetii employs a Type IV secretion system (T4SS) to establish and maintain its intracellular niche and modulate host immune responses including the inhibition of apoptosis. Interactions between C. burnetii and caspase-1-mediated inflammasomes are not fully elucidated. This study confirms that C. burnetii does not activate caspase-1 during infection of mouse macrophages in vitro. C. burnetii-infected cells did not develop NLRP3 and ASC foci indicating its ability to avoid cytosolic detection. C. burnetii is unable to inhibit the pyroptosis and IL-1ß secretion that is induced by potent inflammasome stimuli but rather enhances these caspase-1-mediated effects. We found that C. burnetii upregulates pro-IL-1ß and robustly primes NLRP3 inflammasomes via TLR2 and MyD88 signaling. As for wildtype C. burnetii, T4SS-deficient mutants primed and potentiated NLRP3 inflammasomes. An in vivo model of pulmonary infection in C57BL/6 mice was developed. Mice deficient in NLRP3 or caspase-1 were like wildtype mice in the development and resolution of splenomegaly due to red pulp hyperplasia, and histologic lesions and macrophage kinetics, but had slightly higher pulmonary bacterial burdens at the greatest measured time point. Together these findings indicate that C. burnetii primes but avoids cytosolic detection by NLRP3 inflammasomes, which are not required for the clinical resistance of C57BL/6 mice. Determining mechanisms employed by C. burnetii to avoid cytosolic detection via NLRP3 inflammasomes will be beneficial to the development of preventative and interventional therapies for Q fever.


Subject(s)
Coxiella burnetii , Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , Q Fever , Animals , Mice , Mice, Inbred C57BL , Q Fever/immunology
11.
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
12.
Sci Rep ; 10(1): 20934, 2020 12 01.
Article in English | MEDLINE | ID: mdl-33262373

ABSTRACT

Serology is essential for Q fever diagnostics, a disease caused by the bacterial pathogen Coxiella burnetii. The gold standard test is an immunofluorescence assay utilizing whole cell antigens, which are both dangerous and laborious to produce. Complexities of the antigen coupled with the subjective nature of the assay lead to decreased uniformity of test results and underscore the need for improved methodologies. Thirty-three C. burnetii proteins, previously identified as immunoreactive, were screened for reactivity to naturally infected goat serum. Based on reactivity, 10 proteins were analyzed in a secondary screen against human serum from healthy donors. Assay sensitivity and specificity ranged from 21 to 71% and 90 to 100%, respectively. Three promising antigens were identified based on receiver operating characteristic curve analysis (CBU_1718, CBU_0307, and CBU_1398). Five multiplex assays failed to outperform the individual proteins, with sensitivities and specificities ranging from 29 to 57% and 90 to 100%, respectively. Truncating the top antigen, CBU_1718, had no effect on specificity (90%); yet sensitivity decreased dramatically (71% to 21%). Through this study, we have expanded the subset of C. burnetii immunoreactive proteins validated by enzyme-linked immunosorbent assay and demonstrate the effect of novel antigen combinations and protein truncations on assay performance.


Subject(s)
Q Fever/diagnosis , Recombinant Proteins/analysis , Animals , Antibodies, Bacterial/immunology , Antigens, Bacterial/blood , Antigens, Bacterial/immunology , Coxiella burnetii/immunology , Goats , Q Fever/blood , Q Fever/immunology , ROC Curve , Recombinant Proteins/immunology , Sensitivity and Specificity
13.
Infect Immun ; 88(11)2020 10 19.
Article in English | MEDLINE | ID: mdl-32817330

ABSTRACT

Natural killer (NK) cells are critically involved in the early immune response against various intracellular pathogens, including Coxiella burnetii and Chlamydia psittaciChlamydia-infected NK cells functionally mature, induce cellular immunity, and protect themselves by killing the bacteria in secreted granules. Here, we report that infected NK cells do not allow intracellular multiday growth of Coxiella, as is usually observed in other host cell types. C. burnetii-infected NK cells display maturation and gamma interferon (IFN-γ) secretion, as well as the release of Coxiella-containing lytic granules. Thus, NK cells possess a potent program to restrain and expel different types of invading bacteria via degranulation. Strikingly, though, in contrast to Chlamydia, expulsed Coxiella organisms largely retain their infectivity and, hence, escape the cell-autonomous self-defense mechanism in NK cells.


Subject(s)
Cell Degranulation/immunology , Immunity, Cellular/immunology , Killer Cells, Natural/immunology , Killer Cells, Natural/microbiology , Q Fever/immunology , Animals , Coxiella burnetii , Mice
14.
Sci Rep ; 10(1): 13311, 2020 08 07.
Article in English | MEDLINE | ID: mdl-32770104

ABSTRACT

Q-fever is a flu-like illness caused by Coxiella burnetii (Cb), a highly infectious intracellular bacterium. There is an unmet need for a safe and effective vaccine for Q-fever. Correlates of immune protection to Cb infection are limited. We proposed that analysis by longitudinal high dimensional immune (HDI) profiling using mass cytometry combined with other measures of vaccination and protection could be used to identify novel correlates of effective vaccination and control of Cb infection. Using a vaccine-challenge model in HLA-DR transgenic mice, we demonstrated significant alterations in circulating T-cell and innate immune populations that distinguished vaccinated from naïve mice within 10 days, and persisted until at least 35 days post-vaccination. Following challenge, vaccinated mice exhibited reduced bacterial burden and splenomegaly, along with distinct effector T-cell and monocyte profiles. Correlation of HDI data to serological and pathological measurements was performed. Our data indicate a Th1-biased response to Cb, consistent with previous reports, and identify Ly6C, CD73, and T-bet expression in T-cell, NK-cell, and monocytic populations as distinguishing features between vaccinated and naïve mice. This study refines the understanding of the integrated immune response to Cb vaccine and challenge, which can inform the assessment of candidate vaccines for Cb.


Subject(s)
Bacterial Vaccines/immunology , Coxiella burnetii/immunology , Immunity, Cellular , Immunity, Innate , Q Fever/prevention & control , T-Lymphocytes/immunology , Animals , Female , HLA-DR Antigens/genetics , HLA-DR Antigens/immunology , Humans , Killer Cells, Natural/immunology , Killer Cells, Natural/pathology , Mice , Mice, Inbred BALB C , Mice, Transgenic , Monocytes/immunology , Monocytes/pathology , Q Fever/genetics , Q Fever/immunology , Q Fever/pathology , T-Lymphocytes/pathology
15.
Hum Immunol ; 81(10-11): 596-605, 2020.
Article in English | MEDLINE | ID: mdl-32718721

ABSTRACT

Query fever is a zoonotic disease caused by Coxiella burnetii. There is no universal method for the prevention of this disease. Recombinant vaccine is a potent strategy that can be utilized for this purpose. The current study was conducted to develop a multi-epitope vaccine against Coxiella burnetii. Hence, OmpA, Tuf2, GroEL, Mip and sucB antigens were used for the prediction of epitopes. Then, a multi-epitope vaccine was developed based on a molecular adjuvant and fragments that contained the best MHCI, B cell, MHCII and IFN-γ epitopes. The features of the developed vaccine including physicochemical parameters, antigenicity and protein structures were assessed. Also, interaction between the developed vaccine and TLR4/MD2 receptor along with molecular dynamics of the ligand-receptor complex were investigated. Finally, the codon adaptation and cloning were conducted for the developed vaccine. According to the results, molecular weight, instability index, antigenicity and random coil percentage of the developed vaccine were 54.4 kDa, 32.84, 1.1936 and 34.92%, respectively. Besides, residues distribution in core region of the refined model was 85%. The results demonstrated that the developed vaccine could dock to its receptor with the lowest energy of -976.7 as well as RMSD value of the complex was between 0.15 and 0.22 nm. Also, the results showed that CIA index of the codon adapted sequence was 0.95. Finally, cloning results revealed that nucleotide sequence of the developed vaccine could be successfully cloned into pET-21a (+). Based on these results, it seems that the developed vaccine can be a suitable candidate to prevent Coxiella burnetii.


Subject(s)
Bacterial Vaccines/immunology , Bacterial Zoonoses/prevention & control , Coxiella burnetii/immunology , Epitopes, B-Lymphocyte/immunology , Molecular Docking Simulation/methods , Molecular Dynamics Simulation , Q Fever/prevention & control , Amino Acid Sequence , Animals , Antigens, Bacterial/immunology , Bacterial Outer Membrane Proteins/chemistry , Bacterial Outer Membrane Proteins/immunology , Bacterial Vaccines/chemistry , Chaperonin 60/chemistry , Chaperonin 60/immunology , Epitopes, B-Lymphocyte/chemistry , Histocompatibility Antigens Class I/chemistry , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class II/chemistry , Histocompatibility Antigens Class II/immunology , Humans , Ketoglutarate Dehydrogenase Complex/chemistry , Ketoglutarate Dehydrogenase Complex/immunology , Lymphocyte Antigen 96/chemistry , Protein Structure, Secondary , Protein Structure, Tertiary , Q Fever/immunology , Q Fever/microbiology , Toll-Like Receptor 4/chemistry
16.
Proc Natl Acad Sci U S A ; 117(24): 13708-13718, 2020 06 16.
Article in English | MEDLINE | ID: mdl-32482853

ABSTRACT

The Q fever agent Coxiella burnetii uses a defect in organelle trafficking/intracellular multiplication (Dot/Icm) type 4b secretion system (T4SS) to silence the host innate immune response during infection. By investigating C. burnetii effector proteins containing eukaryotic-like domains, here we identify NopA (nucleolar protein A), which displays four regulator of chromosome condensation (RCC) repeats, homologous to those found in the eukaryotic Ras-related nuclear protein (Ran) guanine nucleotide exchange factor (GEF) RCC1. Accordingly, NopA is found associated with the chromatin nuclear fraction of cells and uses the RCC-like domain to interact with Ran. Interestingly, NopA triggers an accumulation of Ran-GTP, which accumulates at nucleoli of transfected or infected cells, thus perturbing the nuclear import of transcription factors of the innate immune signaling pathway. Accordingly, qRT-PCR analysis on a panel of cytokines shows that cells exposed to the C. burnetii nopA::Tn or a Dot/Icm-defective dotA::Tn mutant strain present a functional innate immune response, as opposed to cells exposed to wild-type C. burnetii or the corresponding nopA complemented strain. Thus, NopA is an important regulator of the innate immune response allowing Coxiella to behave as a stealth pathogen.


Subject(s)
Bacterial Proteins/metabolism , Coxiella burnetii/metabolism , Q Fever/immunology , Animals , Bacterial Proteins/genetics , Coxiella burnetii/genetics , Female , Host-Pathogen Interactions , Humans , Immunity, Innate , Mice , Mice, Inbred C57BL , Mice, SCID , Q Fever/genetics , Q Fever/microbiology
17.
Comp Immunol Microbiol Infect Dis ; 68: 101402, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31812119

ABSTRACT

Q fever caused by Coxiella burentii, gram negative obligate intracellular bacterium. The disease has been reported in wide range of animals especially ruminants. The available data about the prevalence of Q fever in camels in Egypt are limited. Therefore, the present study aimed to investigate the seroprevalence of C. burnetii among camels and identify the risk factors associated with infection. A total 315 serum samples were collected from three governorates in Egypt during 2018 and examined by an indirect Enzyme linked Immunosorbent Assay (ELISA). The obtained results were subsequent analyzed by chi-square and logistic regression. Generally, the seroprevalence of C. burnetii among camels was 22 %. The results revealed that the seroprevalence of C. burnetii increased in aged female camels in comparison with young one and was higher also in female with history of abortion (OR = 4.6, 95%CI: 2.46-8.76). The infection was significantly increased during autumn season (OR = 9.3, 95%CI: 4.23-20.5). Besides, camels in contact with small ruminants showed high level of infection (OR = 1.12, 95%CI: 0.65-1.93) or camel with heavy tick infestation (OR = 1.08, 95%CI: 0.60-1.92). Our report confirms that the seroprevalence of C. burnetii among camels in Egypt and appropriate control measures should be taken to reduce the transmission of infection to other animal species or human.


Subject(s)
Antibodies, Bacterial/blood , Camelus/microbiology , Q Fever/veterinary , Seasons , Abortion, Veterinary , Age Factors , Animals , Camelus/immunology , Coxiella burnetii , Egypt/epidemiology , Female , Male , Prevalence , Q Fever/epidemiology , Q Fever/immunology , Risk Factors , Seroepidemiologic Studies
18.
J Clin Microbiol ; 58(2)2020 01 28.
Article in English | MEDLINE | ID: mdl-31748325

ABSTRACT

Several commercially available enzyme-linked immunosorbent assays (ELISAs) for the detection of phase II IgG or IgM antibodies against Coxiella burnetii were compared. In addition, an indirect immunofluorescence test was used as a confirmation test. In all, 70 serum samples for IgG and 43 serum samples for IgM were tested. The ELISAs showed large differences in sensitivity and specificity, which led to a partially high ratio of false-negative determinations. The most convincing test was PanBio from Abbott, which unfortunately can only test IgG but not IgM.


Subject(s)
Antibodies, Bacterial/blood , Coxiella burnetii/immunology , Enzyme-Linked Immunosorbent Assay , Fluorescent Antibody Technique, Indirect , Immunoglobulin G/blood , Immunoglobulin M/blood , Humans , Q Fever/blood , Q Fever/diagnosis , Q Fever/immunology , Reagent Kits, Diagnostic , Sensitivity and Specificity
19.
Zoonoses Public Health ; 67(1): 89-92, 2020 02.
Article in English | MEDLINE | ID: mdl-31705592

ABSTRACT

BACKGROUND: Q fever is a febrile illness caused by infection with the bacterium Coxiella burnetii. It is most often transmitted by inhalation of the bacteria after it is shed by infected livestock. Recent studies have found very high C. burnetii infection rates among marine mammals, but it is not known if shedding by marine mammals creates a risk of Q fever among humans. To better understand infection of humans with exposure to marine mammals, the prevalence of antibodies against C. burnetii in serum samples taken from Alaskan Native persons residing on the Pribilof Islands was evaluated. The Pribilof Islands support large populations of northern fur seals infected with C. burnetii that may increase the risk of exposure for island residents. METHODS: Serum testing for IgG antibodies against C. burnetii (phase I and phase II) was performed, and demographic data were analysed utilizing banked serum specimens drawn from island residents from 1980 to 2000. RESULTS: The overall seroprevalence rate was 11.6% (95% CI = 9.3%-14.4%; 72/621). This is higher than the previously reported 3.1% (95% CI = 2.1%-4.3%) seroprevalence for the U.S. CONCLUSIONS: These results suggest that Alaskan Native persons may be at higher risk for exposure to C. burnetii than the general US. population, possibly due to proximity to large populations of infected marine mammals.


Subject(s)
Antibodies, Bacterial/blood , Coxiella burnetii/immunology , Q Fever/blood , Seroepidemiologic Studies , Adolescent , Adult , Aged , Aged, 80 and over , Alaska/epidemiology , Child , Child, Preschool , Female , Humans , Infant , Male , Middle Aged , Q Fever/epidemiology , Q Fever/immunology , Young Adult
20.
J Immunol ; 204(3): 611-621, 2020 02 01.
Article in English | MEDLINE | ID: mdl-31871024

ABSTRACT

Coxiella burnetii is an obligate intracellular bacterium and the causative agent of Q fever. C. burnetii is considered a potential bioterrorism agent because of its low infectious dose; resistance to heat, drying, and common disinfectants; and lack of prophylactic therapies. Q-Vax, a formalin-inactivated whole-bacteria vaccine, is currently the only prophylactic measure that is protective against C. burnetii infections but is not U.S. Food and Drug Administration approved. To overcome the safety concerns associated with the whole-bacteria vaccine, we sought to generate and evaluate recombinant protein subunit vaccines against C. burnetii To accomplish this, we formulated C. burnetii Ags with a novel TLR triagonist adjuvant platform, which used combinatorial chemistry to link three different TLR agonists together to form one adjuvanting complex. We evaluated the immunomodulatory activity of a panel of TLR triagonist adjuvants and found that they elicited unique Ag-specific immune responses both in vitro and in vivo. We evaluated our top candidates in a live C. burnetii aerosol challenge model in C56BL/6 mice and found that several of our novel vaccine formulations conferred varying levels of protection to the challenged animals compared with sham immunized mice, although none of our candidates were as protective as the commercial vaccine across all protection criteria that were analyzed. Our findings characterize a novel adjuvant platform and offer an alternative approach to generating protective and effective vaccines against C. burnetii.


Subject(s)
Bacterial Vaccines/immunology , Coxiella burnetii/physiology , Q Fever/immunology , Toll-Like Receptors/agonists , Adjuvants, Immunologic , Animals , Bacterial Vaccines/chemical synthesis , Combinatorial Chemistry Techniques , Disease Models, Animal , Female , Humans , Immunity , Immunogenicity, Vaccine , Mice , Mice, Inbred C57BL , Vaccines, Subunit
SELECTION OF CITATIONS
SEARCH DETAIL
...