Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Front Cell Infect Microbiol ; 13: 1160198, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37153158

RESUMO

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.


Assuntos
Infecções Bacterianas , Neoplasias , RNA Longo não Codificante , Humanos , RNA Longo não Codificante/metabolismo , Infecções Bacterianas/genética , Infecções Bacterianas/microbiologia , Imunidade
2.
Sci Rep ; 13(1): 6021, 2023 04 13.
Artigo em Inglês | MEDLINE | ID: mdl-37055450

RESUMO

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.


Assuntos
Bactérias , Redes Neurais de Computação , Estados Unidos
3.
Microbiol Spectr ; 10(4): e0248421, 2022 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-35913176

RESUMO

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.


Assuntos
Coxiella burnetii , Febre Q , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Coxiella burnetii/genética , Interações Hospedeiro-Patógeno/fisiologia , Humanos , Macrófagos/microbiologia , Camundongos , Camundongos SCID , Febre Q/metabolismo , Febre Q/microbiologia , Fatores de Virulência/genética , Fatores de Virulência/metabolismo
4.
Front Immunol ; 13: 886810, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35693783

RESUMO

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.


Assuntos
Coxiella burnetii , Febre Q , Vacinas Bacterianas , Feminino , Humanos , Gravidez , Vacinação/efeitos adversos , Desenvolvimento de Vacinas
5.
Proc Natl Acad Sci U S A ; 119(14): e2112886119, 2022 04 05.
Artigo em Inglês | MEDLINE | ID: mdl-35363569

RESUMO

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.


Assuntos
Bactérias , Genoma Bacteriano , Aprendizado de Máquina , Fatores de Virulência , Sequenciamento Completo do Genoma , Bactérias/genética , Bactérias/patogenicidade , Fenótipo , Virulência/genética , Fatores de Virulência/genética
6.
Proc Natl Acad Sci U S A ; 119(1)2022 01 04.
Artigo em Inglês | MEDLINE | ID: mdl-34930823

RESUMO

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.


Assuntos
Proteínas de Bactérias , Coxiella burnetii , Fosfoproteínas Fosfatases , Febre Q , Transdução de Sinais , Fatores de Virulência , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/imunologia , Chlorocebus aethiops , Coxiella burnetii/genética , Coxiella burnetii/imunologia , Coxiella burnetii/patogenicidade , Células HEK293 , Células HeLa , Humanos , NF-kappa B/genética , NF-kappa B/imunologia , Fosfoproteínas Fosfatases/genética , Fosfoproteínas Fosfatases/imunologia , Febre Q/genética , Febre Q/imunologia , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Células Vero , Fatores de Virulência/genética , Fatores de Virulência/imunologia
7.
Front Immunol ; 12: 754712, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34616410

RESUMO

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.


Assuntos
Vacinas Bacterianas/efeitos adversos , Modelos Animais de Doenças , Hipersensibilidade Tardia/imunologia , Febre Q , Células Th1/imunologia , Animais , Coxiella burnetii , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Febre Q/prevenção & controle , Vacinas de Produtos Inativados/efeitos adversos
8.
Cell Rep Med ; 2(12): 100461, 2021 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-35028605

RESUMO

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.


Assuntos
Antígenos de Bactérias/imunologia , Coxiella burnetii/imunologia , Hipersensibilidade/imunologia , Imunidade , Febre Q/imunologia , Febre Q/prevenção & controle , Aerossóis , Animais , Afinidade de Anticorpos , Variação Antigênica , Vacinas Bacterianas/imunologia , Linfócitos T CD4-Positivos/imunologia , Modelos Animais de Doenças , Feminino , Cobaias , Imunização , Lipopolissacarídeos , Pulmão/microbiologia , Pulmão/patologia , Macaca mulatta , Masculino , Camundongos Endogâmicos C57BL , Febre Q/microbiologia , Solubilidade
9.
Infect Immun ; 88(10)2020 09 18.
Artigo em Inglês | MEDLINE | ID: mdl-32690632

RESUMO

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.


Assuntos
Vacina contra Brucelose/administração & dosagem , Brucella melitensis/imunologia , Brucelose/prevenção & controle , Modelos Animais de Doenças , Complicações Infecciosas na Gravidez/prevenção & controle , Animais , Anticorpos Antibacterianos/sangue , Brucella melitensis/patogenicidade , Brucelose/microbiologia , Brucelose/patologia , Feminino , Cobaias , Humanos , Glândulas Mamárias Animais/microbiologia , Glândulas Mamárias Animais/patologia , Placenta/microbiologia , Placenta/patologia , Gravidez , Complicações Infecciosas na Gravidez/microbiologia , Complicações Infecciosas na Gravidez/patologia , Vacinação
10.
J Infect Dis ; 220(4): 624-634, 2019 07 19.
Artigo em Inglês | MEDLINE | ID: mdl-30938819

RESUMO

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.


Assuntos
Anticorpos Antibacterianos/imunologia , Antígenos de Bactérias/imunologia , Vacinas Bacterianas/imunologia , Coxiella burnetii/imunologia , Imunidade Celular , Febre Q/imunologia , Receptores de Quimiocinas/imunologia , Animais , Formação de Anticorpos , Células Dendríticas/imunologia , Feminino , Humanos , Lipopolissacarídeos/imunologia , Camundongos , Febre Q/microbiologia , Febre Q/prevenção & controle , Vacinação
11.
Microbes Infect ; 20(5): 302-307, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29331581

RESUMO

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.


Assuntos
Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Coxiella burnetii/metabolismo , Endossomos/metabolismo , Lisossomos/metabolismo , Sistemas de Secreção Tipo IV/metabolismo , Motivos de Aminoácidos/genética , Motivos de Aminoácidos/fisiologia , Proteínas de Bactérias/genética , Membrana Celular/metabolismo , Coxiella burnetii/genética , Células HeLa , Interações Hospedeiro-Patógeno , Humanos , Transporte Proteico , Febre Q/metabolismo , Febre Q/microbiologia , Saccharomyces cerevisiae/metabolismo , Sistemas de Secreção Tipo IV/genética , Vacúolos/metabolismo , Vacúolos/microbiologia , Proteínas rab de Ligação ao GTP/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo
12.
Cell Host Microbe ; 21(5): 637-649.e6, 2017 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-28494245

RESUMO

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.


Assuntos
Criptococose/imunologia , Cryptococcus neoformans/genética , Cryptococcus neoformans/patogenicidade , Interações Hospedeiro-Patógeno/imunologia , Transdução de Sinais/fisiologia , Virulência/genética , Proteínas Quinases Ativadas por AMP/genética , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Proteínas Reguladoras de Apoptose/metabolismo , Autofagia/fisiologia , Proteína Homóloga à Proteína-1 Relacionada à Autofagia/genética , Proteína Homóloga à Proteína-1 Relacionada à Autofagia/metabolismo , Transporte Biológico/fisiologia , Linhagem Celular , Coxiella burnetii/patogenicidade , Criptococose/microbiologia , Cryptococcus neoformans/crescimento & desenvolvimento , Cryptococcus neoformans/metabolismo , Modelos Animais de Doenças , Feminino , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/fisiologia , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Monócitos/metabolismo , Fagocitose , Proteínas Serina-Treonina Quinases/metabolismo , Proteômica , Células RAW 264.7 , Vacúolos/microbiologia , Virulência/fisiologia
13.
Artigo em Inglês | MEDLINE | ID: mdl-28217558

RESUMO

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.


Assuntos
Coxiella burnetii/patogenicidade , Modelos Animais de Doenças , Interações Hospedeiro-Patógeno , Febre Q/microbiologia , Febre Q/patologia , Fatores de Virulência/análise , Animais , Camundongos , Camundongos SCID , Virulência
14.
Infect Immun ; 84(9): 2524-33, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27324482

RESUMO

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.


Assuntos
Proteínas de Bactérias/metabolismo , Coxiella burnetii/metabolismo , GTP Fosfo-Hidrolases/metabolismo , Febre Q/metabolismo , Sistemas de Secreção Tipo IV/metabolismo , Virulência/fisiologia , Proteína rhoA de Ligação ao GTP/metabolismo , Animais , Linhagem Celular Tumoral , Células HeLa , Interações Hospedeiro-Patógeno/fisiologia , Humanos , Lisossomos/metabolismo , Camundongos , Transporte Proteico/fisiologia , Febre Q/microbiologia , Vacúolos/metabolismo , Vacúolos/microbiologia
15.
PLoS One ; 10(9): e0136699, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26366725

RESUMO

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.


Assuntos
Anti-Infecciosos/farmacologia , Coxiella burnetii/efeitos dos fármacos , Macrófagos/microbiologia , Proteína D Associada a Surfactante Pulmonar/farmacologia , Animais , Linhagem Celular , Coxiella burnetii/patogenicidade , Macrófagos/imunologia , Camundongos , Fagocitose
16.
Nat Rev Microbiol ; 11(8): 561-73, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23797173

RESUMO

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.


Assuntos
Coxiella burnetii/patogenicidade , Interações Hospedeiro-Patógeno , Febre Q/microbiologia , Animais , Apoptose , Aderência Bacteriana , Coxiella burnetii/genética , Humanos , Integrina alfaVbeta3/genética , Integrina alfaVbeta3/metabolismo , Membranas Intracelulares/metabolismo , Membranas Intracelulares/microbiologia , Fagocitose , Fagossomos/metabolismo , Fagossomos/microbiologia , Fatores de Tempo , Vacúolos/microbiologia
17.
Adv Exp Med Biol ; 984: 13-38, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22711625

RESUMO

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.


Assuntos
Coxiella burnetii/genética , Coxiella burnetii/patogenicidade , Febre Q/microbiologia , Surtos de Doenças , Genoma Bacteriano , Genômica , Humanos , Filogenia , Saúde Pública , Febre Q/epidemiologia , Virulência/genética
18.
Infect Immun ; 77(10): 4275-83, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19620343

RESUMO

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.


Assuntos
Antibacterianos/farmacologia , Burkholderia pseudomallei/enzimologia , Inibidores Enzimáticos/farmacologia , Isocitrato Liase/antagonistas & inibidores , Melioidose/tratamento farmacológico , Melioidose/microbiologia , Animais , Burkholderia pseudomallei/patogenicidade , Contagem de Colônia Microbiana , Isocitrato Liase/fisiologia , Dose Letal Mediana , Pulmão/microbiologia , Ratos , Análise de Sobrevida
19.
Mol Microbiol ; 59(4): 1083-96, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16430686

RESUMO

Pseudomonas aeruginosa readily binds to stainless steel and other abiotic surfaces, causing major problems in both the medical and food industries. In this study, we show that P. aeruginosa binds to abiotic surfaces in a concentration-dependent, saturable manner during the initial stages of biofilm formation. P. aeruginosa type IV pili mediate binding to stainless steel as a pilus-deficient strain does not bind to steel, purified type IV pili bound in a concentration-dependent, saturable manner, and purified pili competitively inhibited whole cell binding. PAK pili can also bind polystyrene and polyvinylchloride in a concentration-dependant and saturable manner. As an antibody specific for the C-terminal pilin receptor binding domain inhibited adherence to abiotic surfaces, the role of the C-terminal receptor binding domain in mediating binding to steel surfaces was examined. A synthetic peptide of the PAK pilin epithelial cell receptor binding domain [PAK(128-144)ox] bound directly to steel with high affinity. The interaction of pili with steel was specifically inhibited by this peptide with an apparent Ki of approximately 0.2 nM and effectively inhibited the binding of viable homologous and heterologous P. aeruginosa strains to steel with an apparent Ki of approximately 4 nM. A single point mutation (K130I) in the PAO receptor binding domain was observed to abolish binding to stainless steel while binding to human buccal epithelial cells was enhanced. Therefore, the C-terminal receptor binding domain appears to have evolved for binding a variety of surfaces.


Assuntos
Aderência Bacteriana , Biofilmes/crescimento & desenvolvimento , Proteínas de Fímbrias/metabolismo , Fímbrias Bacterianas/metabolismo , Pseudomonas aeruginosa/fisiologia , Aço Inoxidável , Adesinas Bacterianas/metabolismo , Sequência de Aminoácidos , Anticorpos Antibacterianos/farmacologia , Aderência Bacteriana/efeitos dos fármacos , Proteínas de Fímbrias/química , Proteínas de Fímbrias/genética , Fímbrias Bacterianas/efeitos dos fármacos , Fímbrias Bacterianas/genética , Flagelos/genética , Flagelos/metabolismo , Dados de Sequência Molecular , Mutação , Peptídeos/farmacologia , Pseudomonas aeruginosa/metabolismo , Propriedades de Superfície
20.
J Bacteriol ; 187(4): 1455-64, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15687210

RESUMO

The opportunistic pathogen Pseudomonas aeruginosa produces multifunctional, polar, filamentous appendages termed type IV pili. Type IV pili are involved in colonization during infection, twitching motility, biofilm formation, bacteriophage infection, and natural transformation. Electrostatic surface analysis of modeled pilus fibers generated from P. aeruginosa strain PAK, K122-4, and KB-7 pilin monomers suggested that a solvent-exposed band of positive charge may be a common feature of all type IV pili. Several functions of type IV pili, including natural transformation and biofilm formation, involve DNA. We investigated the ability of P. aeruginosa type IV pili to bind DNA. Purified PAK, K122-4, and KB-7 pili were observed to bind both bacterial plasmid and salmon sperm DNA in a concentration-dependent and saturable manner. PAK pili had the highest affinity for DNA, followed by K122-4 and KB-7 pili. DNA binding involved backbone interactions and preferential binding to pyrimidine residues even though there was no evidence of sequence-specific binding. Pilus-mediated DNA binding was a function of the intact pilus and thus required elements present in the quaternary structure. However, binding also involved the pilus tip as tip-specific, but not base-specific, antibodies inhibited DNA binding. The conservation of a Thr residue in all type IV pilin monomers examined to date, along with the electrostatic data, implies that DNA binding is a conserved function of type IV pili. Pilus-mediated DNA binding could be important for biofilm formation both in vivo during an infection and ex vivo on abiotic surfaces.


Assuntos
DNA Bacteriano/metabolismo , Proteínas de Ligação a DNA/metabolismo , DNA/metabolismo , Proteínas de Fímbrias/metabolismo , Fímbrias Bacterianas/metabolismo , Pseudomonas aeruginosa/metabolismo , Sequência de Aminoácidos , Sequência de Bases , Proteínas de Ligação a DNA/química , Proteínas de Fímbrias/química , Proteínas de Fímbrias/fisiologia , Fímbrias Bacterianas/química , Modelos Moleculares , Plasmídeos , Estrutura Quaternária de Proteína , Pseudomonas aeruginosa/ultraestrutura , Pirimidinas/metabolismo , Alinhamento de Sequência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...