Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 318
1.
Int J Mol Sci ; 24(13)2023 Jun 30.
Article En | MEDLINE | ID: mdl-37446087

Having previously shown that soluble E-cadherin (sE-cad) is found in sera of Q fever patients and that infection of BeWo cells by C. burnetii leads to modulation of the E-cad/ß-cat pathway, our purpose was to identify which sheddase(s) might catalyze the cleavage of E-cad. Here, we searched for a direct mechanism of cleavage initiated by the bacterium itself, assuming the possible synthesis of a sheddase encoded in the genome of C. burnetii or an indirect mechanism based on the activation of a human sheddase. Using a straightforward bioinformatics approach to scan the complete genomes of four laboratory strains of C. burnetii, we demonstrate that C. burnetii encodes a 451 amino acid sheddase (CbHtrA) belonging to the HtrA family that is differently expressed according to the bacterial virulence. An artificial CbHtrA gene (CoxbHtrA) was expressed, and the CoxbHtrA recombinant protein was found to have sheddase activity. We also found evidence that the C. burnetii infection triggers an over-induction of the human HuHtrA gene expression. Finally, we demonstrate that cleavage of E-cad by CoxbHtrA on macrophages-THP-1 cells leads to an M2 polarization of the target cells and the induction of their secretion of IL-10, which "disarms" the target cells and improves C. burnetii replication. Taken together, these results demonstrate that the genome of C. burnetii encodes a functional HtrA sheddase and establishes a link between the HtrA sheddase-induced cleavage of E-cad, the M2 polarization of the target cells and their secretion of IL-10, and the intracellular replication of C. burnetii.


Bacterial Proteins , Coxiella burnetii , Humans , Coxiella burnetii/enzymology , Coxiella burnetii/genetics , Coxiella burnetii/pathogenicity , Interleukin-10/metabolism , Macrophages/microbiology , Q Fever/microbiology , Q Fever/physiopathology , THP-1 Cells/microbiology , Cadherins/metabolism , Genome, Bacterial/genetics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Gene Expression Regulation, Bacterial , Recombinant Proteins/genetics , Host Microbial Interactions , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Escherichia coli/genetics
2.
Front Cell Infect Microbiol ; 13: 1336600, 2023.
Article En | MEDLINE | ID: mdl-38282619

Coxiella burnetii is an obligate intracellular bacterium that causes Q fever, a zoonotic disease typically manifests as a severe flu-illness. After invading into the host cells, C. burnetii delivers effectors to regulate the vesicle trafficking and fusion events to form a large and mature Coxiella-containing vacuole (CCV), providing sufficient space and nutrition for its intracellular growth and proliferation. Lysosomal trafficking regulator (LYST) is a member of the Beige and Chediak-Higashi syndrome (BEACH) family, which regulates the transport of vesicles to lysosomes and regulates TLR signaling pathway, but the effect of LYST on C. burnetii infection is unclear. In this study, a series of experiments has been conducted to investigate the influence of LYST on intracellular growth of C. burnetii. Our results showed that lyst transcription was up-regulated in the host cells after C. burnetii infection, but there is no significant change in lyst expression level after infection with the Dot/Icm type IV secretion system (T4SS) mutant strain, while CCVs expansion and significantly increasing load of C. burnetii appeared in the host cells with a silenced lyst gene, suggesting LYST inhibits the intracellular proliferation of C. burnetii by reducing CCVs size. Then, the size of CCVs and the load of C. burnetii in the HeLa cells pretreated with E-64d were significantly decreased. In addition, the level of iNOS was decreased significantly in LYST knockout THP-1 cells, which was conducive to the intracellular replication of C. burnetii. This data is consistent with the phenotype of L-NMMA-treated THP-1 cells infected with C. burnetii. Our results revealed that the upregulation of lyst transcription after infection is due to effector secretion of C. burnetii and LYST inhibit the intracellular replication of C. burnetii by reducing the size of CCVs and inducing nos2 expression.


Coxiella burnetii , Q Fever , Vesicular Transport Proteins , Humans , Coxiella burnetii/pathogenicity , HeLa Cells , Host-Pathogen Interactions/genetics , Lysosomes/metabolism , Q Fever/microbiology , Vacuoles/microbiology , THP-1 Cells , Vesicular Transport Proteins/genetics , Nitric Oxide Synthase Type II/metabolism
4.
Proc Natl Acad Sci U S A ; 119(1)2022 01 04.
Article En | MEDLINE | ID: mdl-34930823

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.


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.
PLoS One ; 16(4): e0250116, 2021.
Article En | MEDLINE | ID: mdl-33852632

BACKGROUND: Coxiella burnetii is the causative agent of Q fever which is a highly infectious zoonotic disease. C. burnetii has become one of the most important causes of abortion in livestock, which can lead to widespread abortions in these animals. There are very limited studies on the prevalence of C. burnetii infection in cases of animal abortion in Iran. The aim of this study was to investigate the occurrence of C. burnetii in ruminant abortion samples in Iran. METHODS: Abortion samples from cattle, sheep and goats were collected from different parts of Iran and were tested using Real-time PCR targeting the IS1111 element of C. burnetii. RESULTS: In this study, 36 samples (24.7%) of the 146 collected samples were positive for C. burnetii. The prevalence of C. burnetii was 21.3% (20 of 94 samples) in sheep samples. Also, 10 of 46 cattle samples (21.7%) were positive. All six goat abortion samples were positive for C. burnetii. CONCLUSIONS: The findings of the study demonstrate that C. burnetii plays an important role in domestic ruminant abortions in Iran, suggesting that more attention should be paid to the role of C. burnetii in domestic animal abortions by veterinary organizations. The risk of transmitting the infection to humans due to abortion of animals should also be considered.


Abortion, Veterinary/microbiology , Coxiella burnetii/isolation & purification , Q Fever/epidemiology , Abortion, Veterinary/epidemiology , Animals , Animals, Domestic/genetics , Cattle , Cattle Diseases/epidemiology , Cattle Diseases/microbiology , Coxiella burnetii/genetics , Coxiella burnetii/pathogenicity , Female , Goat Diseases/epidemiology , Goat Diseases/microbiology , Goats , Iran , Livestock/genetics , Livestock/microbiology , Pregnancy , Q Fever/diagnosis , Q Fever/genetics , Real-Time Polymerase Chain Reaction/methods , Real-Time Polymerase Chain Reaction/veterinary , Ruminants/genetics , Ruminants/microbiology , Sheep , Sheep Diseases/epidemiology , Sheep Diseases/microbiology , Zoonoses/epidemiology
6.
Genetics ; 217(3)2021 03 31.
Article En | MEDLINE | ID: mdl-33789347

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


Disease Resistance , Genetic Variation , Q Fever/genetics , Quantitative Trait Loci , Animals , Cell Cycle Proteins/genetics , Coxiella burnetii/pathogenicity , Drosophila Proteins/genetics , Drosophila melanogaster , Eye Proteins/genetics , Genetic Background , Q Fever/microbiology
7.
Cell Microbiol ; 23(6): e13331, 2021 06.
Article En | MEDLINE | ID: mdl-33774901

Coxiella burnetii is a bacterial pathogen that replicates in a specialised lysosome-derived organelle called the Coxiella-containing vacuole (CCV). Establishment of the CCV requires the Dot/Icm type IVB secretion system. A previous transposon mutagenesis screen identified the gene cbu1754 as being important for the intracellular replication of C. burnetii. To understand the function of the protein encoded by cbu1754, CCV maturation and intracellular replication phenotypes of a cbu1754 mutant were analysed. In contrast to vacuoles containing wild-type C. burnetii Nine Mile phase II, vacuoles containing the isogenic cbu1754 mutant were smaller and did not display detectible amounts of the autophagy protein LC3, which indicated a CCV biogenesis defect. The Cbu1754 protein was not efficiently delivered into the host cell cytosol during infection, which indicated this protein is not a Dot/Icm-translocated effector protein. Secondary structure predictions suggested that Cbu1754 could be similar to the Legionella pneumophila LvgA protein, which is a component of the Dot/Icm apparatus. Consistent with this hypothesis, production of Cbu1754 in an L. pneumophila ∆lvgA mutant restored LvgA-dependent activities. The L. pneumophila proteins LvgA, IcmS and IcmW are interacting partners that comprise a subassembly of the coupling protein complex that mediates Dot/Icm-dependent effector translocation. Similarly, the Cbu1754 protein was found to be a component of the chaperone complex containing the C. burnetii proteins IcmS and IcmW. Thus, the Cbu1754 protein is an LvgA-related protein important for Dot/Icm function and intracellular replication of C. burnetii.


Bacterial Proteins/genetics , Coxiella burnetii/genetics , DNA Replication , Host-Pathogen Interactions , Vacuoles/microbiology , Bacterial Proteins/metabolism , Coxiella burnetii/chemistry , Coxiella burnetii/pathogenicity , Gene Expression Regulation, Bacterial , HeLa Cells , Humans , Legionella pneumophila/genetics , Phenotype , Virulence Factors/genetics
8.
Pathog Dis ; 79(4)2021 03 31.
Article En | MEDLINE | ID: mdl-33734371

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


Bacterial Infections/microbiology , Host-Pathogen Interactions/immunology , Lung Diseases/microbiology , Lung/microbiology , Macrophages, Alveolar/microbiology , Models, Biological , Bacterial Infections/immunology , Bacterial Infections/pathology , Coxiella burnetii/growth & development , Coxiella burnetii/immunology , Coxiella burnetii/pathogenicity , Humans , Legionella pneumophila/growth & development , Legionella pneumophila/immunology , Legionella pneumophila/pathogenicity , Lung/immunology , Lung/pathology , Lung Diseases/immunology , Lung Diseases/pathology , Macrophages, Alveolar/immunology , Macrophages, Alveolar/pathology , Microtomy , Mycobacterium tuberculosis/growth & development , Mycobacterium tuberculosis/immunology , Mycobacterium tuberculosis/pathogenicity , Primary Cell Culture , Staphylococcus aureus/growth & development , Staphylococcus aureus/immunology , Staphylococcus aureus/pathogenicity , Tissue Banks , Tissue Culture Techniques , Yersinia pestis/growth & development , Yersinia pestis/immunology , Yersinia pestis/pathogenicity
9.
Adv Med Sci ; 66(1): 113-118, 2021 Mar.
Article En | MEDLINE | ID: mdl-33461099

PURPOSE: Dermacentor reticulatus is the second most common tick species in Poland after Ixodes ricinus. The aim of the study was to analyze the presence of pathogen DNA in D. reticulatus. MATERIALS AND METHODS: Ticks were collected in The Protected Landscape Area of the Bug and Nurzec Valley (52°40' N and 22°28' E) between 2016 and 2017. End-point PCR for Borrelia burgdorferi sensu lato, Anaplasma phagocytophilum, Babesia spp., Rickettsia spp., Bartonella spp. and Coxiella burnetii detection was performed. RESULTS: Tick-borne pathogens' DNA was detected in 11.3% of 301 ticks: B. burgdorferi s.l. in 3.6%, Babesia spp. in 6.3%, A. phagocytophilum in 0.7% and B. burgdorferi s.l.-Babesia spp. co-infection in 0.7%. In all 21 Babesia spp. positive samples, sequence analysis confirmed the presence of Babesia canis with an 80.3%-98.3% homology with the B. canis sequences in GenBank. C. burnetii, Bartonella spp., and Rickettsia spp. DNA were not detected. CONCLUSIONS: Dermacentor reticulatus from north-eastern Poland were found to carry three of the most common tick-borne pathogens (B. burgdorferi s.l., Babesia canis, A. phagocytophilum) which lead to single and mixed infections. Babesia canis was the most prevalent pathogen identified in D. reticulatus.


Anaplasma phagocytophilum/pathogenicity , Bartonella/pathogenicity , Borrelia burgdorferi/pathogenicity , Coxiella burnetii/pathogenicity , Dermacentor/microbiology , Rickettsia/pathogenicity , Tick-Borne Diseases/epidemiology , Anaplasma phagocytophilum/isolation & purification , Animals , Bartonella/isolation & purification , Bartonella Infections/microbiology , Borrelia burgdorferi/isolation & purification , Coxiella burnetii/isolation & purification , Ehrlichiosis/microbiology , Humans , Lyme Disease/microbiology , Poland/epidemiology , Q Fever/microbiology , Rickettsia/isolation & purification , Rickettsia Infections/microbiology , Tick-Borne Diseases/microbiology
10.
Cell Microbiol ; 23(4): e13305, 2021 04.
Article En | MEDLINE | ID: mdl-33355405

The obligate intracellular pathogen Coxiella burnetii is the causative agent of the zoonosis Q fever. C. burnetii infection can have severe outcomes due to the development of chronic infection. To establish and maintain an infection, C. burnetii depends on a functional type IVB secretion system (T4BSS) and, thus, on the translocation of effector proteins into the host cell. Here, we showed that the C. burnetii T4BSS effector protein CaeB targets the conserved endoplasmatic reticulum (ER) stress sensor IRE1 during ER stress in mammalian and plant cells. CaeB-induced upregulation of IRE1 RNase activity was essential for CaeB-mediated inhibition of ER stress-induced cell death. Our data reveal a novel role for CaeB in ER stress signalling modulation and demonstrate that CaeB is involved in pathogenicity in vivo. Furthermore, we provide evidence that C. burnetii infection leads to modulation of the ER stress sensors IRE1 and PERK, but not ATF6 during ER stress. While the upregulation of the RNase activity of IRE1 during ER stress depends on CaeB, modulation of PERK is CaeB independent, suggesting that C. burnetii encodes several factors influencing ER stress during infection.


Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Coxiella burnetii/pathogenicity , Endoplasmic Reticulum Stress , Host-Pathogen Interactions , Moths/microbiology , Signal Transduction , Animals , Cell Death , Coxiella burnetii/chemistry , Coxiella burnetii/genetics , DNA Replication , HEK293 Cells , Humans , Larva/microbiology
11.
PLoS One ; 15(10): e0240249, 2020.
Article En | MEDLINE | ID: mdl-33075103

The persistent and highly transmissible Coxiella burnetii is a neglected infection that negatively affects reproductive parameters of livestock. It is also of zoonotic importance and has been reported to cause devastating human infections globally. Domestic ruminants represent the most frequent source of human infection. Data from Nigeria are very few and outdated. There is a significant gap in up-to-date information on the exposure, spatial distribution and risk factors of infection of this important disease. The exposure to C. burnetii was determined using sensitive serological assays in cattle and small ruminants. A total of 538 animals made up of 268 cattle and 270 small ruminants were sampled from three northern Nigerian states. The proportion of cattle sampled that were seropositive from the study locations were: Kwara 14/90 (15.6%; 95% CI: 8.8-24.7); Plateau 10/106 (9.43%; 95% CI: 4.6-16.7) and Borno 4/72 (5.56%; 95% CI: 1.5-13.6) states. Lower seroprevalence was recorded among the small ruminants sampled, with positives recorded from sheep and goat sampled from only Kwara state 6/184 (3.3%; 95% CI: 1.2-7.0); while none of the small ruminants sampled from Plateau were seropositive. The results of the bivariate analysis showed that none of the tested independent variables (village, age group, sex, breed of cattle, presence of ticks, reproductive status, and management system) were statistically significant factors associated with seropositivity of cattle for antibodies to C. burnetii. Stakeholders involved in animal husbandry should be duly educated on proper disposal of birth products as well as bodily fluids in order to reduce environmental contamination, persistence and human infection.


Coxiella burnetii/pathogenicity , Q Fever/epidemiology , Animals , Cattle , Cross-Sectional Studies , Enzyme-Linked Immunosorbent Assay , Female , Goats , Male , Risk Factors , Seroepidemiologic Studies , Serologic Tests , Sheep
12.
PLoS One ; 15(10): e0241246, 2020.
Article En | MEDLINE | ID: mdl-33125388

Q fever is an important zoonosis, yet it is often neglected and can present large outbreaks, as observed in the Netherlands. In the past few years, cases of Q fever have been described in Brazil; however, the epidemiological situation of Q fever in ruminants, the main reservoir of the pathogen, is unknown in this country. Our study aimed to estimate the prevalence of C. burnetii in cattle sent to slaughterhouses using an immunofluorescence assay (IFA) and quantitative real-time PCR (qPCR). From 1515 cattle serum samples collected from nine slaughterhouses, 23.8% (360/1515) were serologically positive by IFA (cutoff titer>1:64), indicating past or recent exposure to C. burnetii infection. Among the 54 cities sampled during the study, 83.3% (45/54) had at least one seropositive animal. Subsequently, all seropositive samples were submitted to qPCR for C. burnetii DNA, and 12.2% (44/360) of the sera were qPCR positive, which indicates bacteremia and suggests active or recent infection. The results highlight the risk for abattoir workers that results from exposure to contaminated aerosols produced during slaughter procedures. Moreover, the heat maps that were construction from the positive samples demonstrate the widespread distribution of C. burnetii in the State of São Paulo, Brazil and denotes the need for surveillance and preventive measures to reduce the prevalence in cattle.


Abattoirs/statistics & numerical data , Coxiella burnetii/isolation & purification , Public Health/statistics & numerical data , Animals , Brazil/epidemiology , Cattle , Cattle Diseases/epidemiology , Coxiella burnetii/classification , Coxiella burnetii/pathogenicity , Fluorescent Antibody Technique , Geography , Phylogeny , Q Fever/epidemiology , Real-Time Polymerase Chain Reaction
13.
PLoS One ; 15(9): e0239260, 2020.
Article En | MEDLINE | ID: mdl-32931511

Coxiella burnetii is a highly infectious zoonotic pathogen infecting wide range of mammals, including humans. In the present study, a total of 711 blood samples from bovines [cattle (n = 543) and buffaloes (n = 168)] from eight farms at different geographical locations in India were screened for C. burnetii targeting the IS1111 and the com1 genes. The anti-C. burnetii antibodies in serum samples were detected using indirect-ELISA kits. Also, a total of 21 parameters pertaining to animal health and farm management were identified to assess their role as possible risk factors for coxiellosis among the targeted farms. The apparent prevalence (positive for PCR and/or ELISA) for coxiellosis was reported to be 24.5% in cattle and 8.9% in buffaloes. In cattle, the detection rate of C. burnetii employing the IS1111 gene (8.5%) was found to be significantly higher (p<0.05) as compared to the com1 (6.5%) gene. The seropositivity by ELISA was higher among cattle (17.7%) than in buffaloes (8.3%). Further, on univariable analysis of risk factors, species (cattle) (OR:3.31; 95%CI:1.88-5.82), inadequate floor spacing (OR:1.64; 95%CI:1.10-2.43), mastitis (OR:2.35, 95%CI:1.45-3.81) and reproductive disorders (OR:2.54; 95%CI:1.67-3.85) were significantly (p<0.05) having high odds for coxiellosis. The multivariable logistic regression analysis of the animal level risk factors revealed that species and age were found to be significantly associated with coxiellosis. However, since the number of screened farms is limited; further research is needed with a higher number of animals to confirm the farm level odds ratio of risk factors. Quarantine and biosecurity measures including farm hygiene operations were observed to be inadequate and also the lack of awareness about coxiellosis among the farm workers. In absence of vaccination program for coxiellosis in India, robust surveillance, farm biosecurity measures and the awareness for the disease among risk groups can play an important role in the disease prevention and subsequent transmission of the pathogen.


Antibodies, Bacterial/blood , Cattle Diseases/blood , Coxiella burnetii/genetics , Q Fever/blood , Animals , Cattle , Cattle Diseases/epidemiology , Cattle Diseases/microbiology , Coxiella burnetii/pathogenicity , Enzyme-Linked Immunosorbent Assay , Farmers , Female , Humans , India/epidemiology , Milk/microbiology , Polymerase Chain Reaction , Q Fever/genetics , Q Fever/microbiology , Zoonoses/blood , Zoonoses/genetics , Zoonoses/microbiology
14.
mSphere ; 5(4)2020 07 22.
Article En | MEDLINE | ID: mdl-32699121

Coxiella burnetii, the causative agent of Query (Q) fever in humans, is a highly infectious obligate intracellular bacterium. Following uptake into a host cell, C. burnetii replicates within a phagolysosome-derived compartment referred to as the Coxiella-containing vacuole (CCV). During infection, C. burnetii exhibits tropism for tissues related to iron storage and recycling (e.g., the liver and splenic red pulp), suggesting that pathogen physiology is linked to host iron metabolism. Iron has been described to have a limited role in C. burnetii virulence regulation, despite evidence that C. burnetii-infected host cells increase expression of transferrin receptors, thereby suggesting that active iron acquisition by the bacterium occurs upon infection. Through the use of host cell-free culture, C. burnetii was separated from the host cell in order to directly assess the role of different forms of iron in C. burnetii replication and viability, and therefore virulence. Results indicate that C. burnetii tolerates molecular iron over a broad concentration range (i.e., ∼0.001 to 1 mM) and undergoes gross loss of viability upon iron starvation. C. burnetii protein synthesis and energy metabolism, however, occur nearly uninhibited under iron concentrations not permissive to replication. Despite the apparent absence of genes related to acquisition of host-associated iron-containing proteins, C. burnetii replication is supported by hemoglobin, transferrin, and ferritin, likely due to release of iron from such proteins under acidic conditions. Moreover, chelation of host iron pools inhibited pathogen replication during infection of cultured cells.IMPORTANCE Host organisms restrict the availability of iron to invading pathogens in order to reduce pathogen replication. To counteract the host's response to infection, bacteria can rely on redundant mechanisms to obtain biologically diverse forms of iron during infection. C. burnetii appears specifically dependent on molecular iron for replication and viability and exhibits a response to iron akin to bacteria that colonize iron-rich environments. Physiological adaptation of C. burnetii to the unique acidic and degradative environment of the CCV is consistent with access of this pathogen to molecular iron.


Coxiella burnetii/physiology , Host-Pathogen Interactions , Iron/metabolism , Microbial Viability , Coxiella burnetii/pathogenicity , HeLa Cells , Humans , Phagosomes/microbiology , Q Fever/microbiology
15.
Cell Microbiol ; 22(10): e13246, 2020 10.
Article En | MEDLINE | ID: mdl-32720355

Intracellular bacterial pathogens harbour genes, the closest homologues of which are found in eukaryotes. Regulator of chromosome condensation 1 (RCC1) repeat proteins are phylogenetically widespread and implicated in protein-protein interactions, such as the activation of the small GTPase Ran by its cognate guanine nucleotide exchange factor, RCC1. Legionella pneumophila and Coxiella burnetii, the causative agents of Legionnaires' disease and Q fever, respectively, harbour RCC1 repeat coding genes. Legionella pneumophila secretes the RCC1 repeat 'effector' proteins LegG1, PpgA and PieG into eukaryotic host cells, where they promote the activation of the pleiotropic small GTPase Ran, microtubule stabilisation, pathogen vacuole motility and intracellular bacterial growth as well as host cell migration. The RCC1 repeat effectors localise to the pathogen vacuole or the host plasma membrane and target distinct components of the Ran GTPase cycle, including Ran modulators and the small GTPase itself. Coxiella burnetii translocates the RCC1 repeat effector NopA into host cells, where the protein localises to nucleoli. NopA binds to Ran GTPase and promotes the nuclear accumulation of Ran(GTP), thus pertubing the import of the transcription factor NF-κB and innate immune signalling. Hence, divergent evolution of bacterial RCC1 repeat effectors defines the range of Ran GTPase cycle targets and likely allows fine-tuning of Ran GTPase activation by the pathogens at different cellular sites.


Biological Evolution , Coxiella burnetii/metabolism , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/metabolism , Legionella pneumophila/metabolism , ran GTP-Binding Protein/metabolism , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cell Nucleolus/enzymology , Coxiella burnetii/genetics , Coxiella burnetii/pathogenicity , Enzyme Activation , Genes, Bacterial , Host-Pathogen Interactions , Humans , Legionella/genetics , Legionella/metabolism , Legionella/pathogenicity , Legionella pneumophila/genetics , Legionnaires' Disease/microbiology , Protein Transport , Q Fever/microbiology , Vacuoles/metabolism , Vacuoles/microbiology
17.
PLoS One ; 15(6): e0234360, 2020.
Article En | MEDLINE | ID: mdl-32520958

Coxiella burnetii causes Q fever in humans and coxiellosis in animals. In humans, it causes acute febrile illnesses like influenza, pneumonia, hepatitis, and chronic illnesses such as endocarditis, vascular infection, and post-infectious fatigue syndrome. It is widely distributed worldwide, and its main reservoirs are sheep, goats, and cattle. This study aimed to determine the frequency of C. burnetii infection using molecular detection and to identify the associated factors in livestock farmers and cattle from the Magdalena Medio region of Antioquia, Colombia. Using real-time polymerase chain reaction (PCR), molecular detection was performed for the IS1111 insertion sequence of C. burnetii using genomic DNA collected from the peripheral blood of 143 livestock farmers and 192 cattle from 24 farms located in Puerto Berrío, Puerto Nare, and Puerto Triunfo. To confirm the results, bidirectional amplicon sequencing of 16S rRNA was performed in four of the positive samples. Additionally, factors associated with C. burnetii were identified using a Poisson regression with cluster effect adjustment. Real-time PCR showed positive results in 25.9% and 19.5% of livestock farmer samples and cattle samples, respectively. For livestock farmers, factors associated with C. burnetii were the area where the farm was located [Puerto Berrío, adjusted prevalence ratio (aPR): 2.13, 95% confidence interval (CI): 1.10-4.11], presence of hens (aPR: 1.47, 95% CI: 1.21-1.79), horses (aPR: 1.61, 95% CI: 1.54-1.67), and ticks (aPR: 2.36, 95% CI: 1.03-5.42) in the residence, and consumption of raw milk (aPR: 1.47, 95% CI: 1.26-1.72). For cattle, the factors associated with Coxiella genus were municipality (Puerto Nare; aPR: 0.39, 95% CI: 0.37-0.41) and time of residence on the farm (≥49 months; aPR: 2.28, 95% CI: 1.03-5.20). By analyzing sequences of the 16S rRNA molecular marker, C. burnetii infection was confirmed in livestock farmers. However, in cattle, only the presence of Coxiella-type bacteria was identified. Further research is necessary to determine the potential role that these types of bacteria have as etiological agents for disease in livestock farmers and cattle from the study area.


Coxiella burnetii/genetics , Coxiella burnetii/isolation & purification , Q Fever/diagnosis , Adult , Animals , Antibodies, Bacterial/blood , Cattle , Cattle Diseases/diagnosis , Cattle Diseases/microbiology , Colombia/epidemiology , Coxiella burnetii/pathogenicity , DNA, Bacterial/genetics , Farmers , Female , Humans , Livestock/genetics , Male , Middle Aged , Prevalence , Q Fever/blood , Q Fever/epidemiology , RNA, Ribosomal, 16S/genetics , Real-Time Polymerase Chain Reaction/methods , Zoonoses/diagnosis , Zoonoses/genetics
18.
PLoS One ; 15(5): e0233100, 2020.
Article En | MEDLINE | ID: mdl-32453753

Equine abortion is a cause of severe economic loss to the equine industry. Equine herpesvirus 1 is considered a primary cause of infectious abortion in horses, however other infectious agents can also cause abortion. Abortions due to zoonotic pathogens have implications for both human and animal health. We determined the prevalence of Coxiella burnetii, Leptospira spp. and Toxoplasma gondii in 600 aborted equine foetal tissues that were submitted to our diagnostic laboratories at the University of Melbourne from 1994 to 2019. Using qPCR we found that the prevalence of C. burnetii was 4%. The highest annual incidence of C. burnetii was observed between 1997-2003 and 2016-2018. The prevalence of C. burnetii in Victoria and New South Wales was 3% and 6% respectively. All the samples tested negative for Leptospira spp. and Toxoplasma gondii DNA. Equine herpesvirus 1 DNA was detected at a prevalence of 3%. This study has provided evidence for the presence of C. burnetii in equine aborted foetal tissues in Australia, but the role of C. burnetii as potential cause of abortion in Australia requires further investigation. C. burnetii is a zoonotic disease agent that causes the disease 'Q fever' in humans. We recommend that appropriate protective measures should be considered when handling material associated with equine abortions to reduce the risk of becoming infected with C. burnetii.


Coxiella burnetii/pathogenicity , Herpesvirus 1, Equid/pathogenicity , Leptospira/pathogenicity , Toxoplasma/pathogenicity , Animals , Female , Horses , Open Reading Frames/genetics , Polymerase Chain Reaction , Pregnancy , Retrospective Studies
20.
Cell Microbiol ; 22(4): e13180, 2020 04.
Article En | MEDLINE | ID: mdl-32185905

Coxiella burnetii is an obligate intracellular bacterial pathogen responsible for severe worldwide outbreaks of the zoonosis Q fever. The remarkable resistance to environmental stress, extremely low infectious dose and ease of dissemination, contributed to the classification of C. burnetii as a class B biothreat. Unique among intracellular pathogens, C. burnetii escapes immune surveillance and replicates within large autophagolysosome-like compartments called Coxiella-containing vacuoles (CCVs). The biogenesis of these compartments depends on the subversion of several host signalling pathways. For years, the obligate intracellular nature of C. burnetii imposed significant experimental obstacles to the study of its pathogenic traits. With the development of an axenic culture medium in 2009, C. burnetii became genetically tractable, thus allowing the implementation of mutagenesis tools and screening approaches to identify its virulence determinants and investigate its complex interaction with host cells. Here, we review the key advances that have contributed to our knowledge of C. burnetii pathogenesis, leading to the rise of this once-neglected pathogen to an exceptional organism to study the intravacuolar lifestyle.


Coxiella burnetii/genetics , Coxiella burnetii/pathogenicity , Host-Pathogen Interactions , Vacuoles/microbiology , Animals , Autophagosomes/microbiology , Axenic Culture/methods , Bacterial Proteins/metabolism , Bacterial Secretion Systems/metabolism , Biotechnology , Humans , Mutation , Phenotype , Q Fever/microbiology
...