Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mol Microbiol ; 121(3): 513-528, 2024 03.
Artigo em Inglês | MEDLINE | ID: mdl-38115201

RESUMO

Intracellular bacteria have evolved mechanisms to invade host cells, establish an intracellular niche that allows survival and replication, produce progeny, and exit the host cell after completion of the replication cycle to infect new target cells. Bacteria exit their host cell by (i) initiation of apoptosis, (ii) lytic cell death, and (iii) exocytosis. While bacterial egress is essential for bacterial spreading and, thus, pathogenesis, we currently lack information about egress mechanisms for the obligate intracellular pathogen C. burnetii, the causative agent of the zoonosis Q fever. Here, we demonstrate that C. burnetii inhibits host cell apoptosis early during infection, but induces and/or increases apoptosis at later stages of infection. Only at later stages of infection did we observe C. burnetii egress, which depends on previously established large bacteria-filled vacuoles and a functional intrinsic apoptotic cascade. The released bacteria are not enclosed by a host cell membrane and can infect and replicate in new target cells. In summary, our data argue that C. burnetii egress in a non-synchronous way at late stages of infection. Apoptosis-induction is important for C. burnetii egress, but other pathways most likely contribute.


Assuntos
Coxiella burnetii , Febre Q , Humanos , Coxiella burnetii/metabolismo , Febre Q/metabolismo , Febre Q/microbiologia , Febre Q/patologia , Apoptose/fisiologia , Transdução de Sinais , Vacúolos/metabolismo , Interações Hospedeiro-Patógeno
2.
Int J Med Microbiol ; 313(6): 151590, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-38056089

RESUMO

The Q-GAPS (Q fever GermAn interdisciplinary Program for reSearch) consortium was launched in 2017 as a German consortium of more than 20 scientists with exceptional expertise, competence, and substantial knowledge in the field of the Q fever pathogen Coxiella (C.) burnetii. C. burnetii exemplifies as a zoonotic pathogen the challenges of zoonotic disease control and prophylaxis in human, animal, and environmental settings in a One Health approach. An interdisciplinary approach to studying the pathogen is essential to address unresolved questions about the epidemiology, immunology, pathogenesis, surveillance, and control of C. burnetii. In more than five years, Q-GAPS has provided new insights into pathogenicity and interaction with host defense mechanisms. The consortium has also investigated vaccine efficacy and application in animal reservoirs and identified expanded phenotypic and genotypic characteristics of C. burnetii and their epidemiological significance. In addition, conceptual principles for controlling, surveilling, and preventing zoonotic Q fever infections were developed and prepared for specific target groups. All findings have been continuously integrated into a Web-based, interactive, freely accessible knowledge and information platform (www.q-gaps.de), which also contains Q fever guidelines to support public health institutions in controlling and preventing Q fever. In this review, we will summarize our results and show an example of how an interdisciplinary consortium provides knowledge and better tools to control a zoonotic pathogen at the national level.


Assuntos
Coxiella burnetii , Saúde Única , Febre Q , Animais , Humanos , Coxiella burnetii/genética , Febre Q/epidemiologia , Febre Q/prevenção & controle , Zoonoses/epidemiologia , Zoonoses/prevenção & controle , Estudos Interdisciplinares
3.
PLoS One ; 18(8): e0290554, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37616234

RESUMO

Reducing antibiotic use is one of the biggest challenges in pig farming, as antibiotics have been used for years to control typical problems such as newborn or post-weaning diarrhea. The pressure a one health approach has created on animal production regarding antimicrobial resistance is an opportunity to find other strategies against enterobacterial pathogens in suckling and weaned piglets. A farm-specific approach could have a good success due to the individual farm structures in Germany and other countries. In this study, non-metric multidimensional scaling, hierarchical clustering, and latent class analysis were used to determine the impact of antibiotic use on antibiotic resistance patterns and pathogen prevalence in 20 German pig farms. This may help to develop individualized health strategies. 802 fresh fecal samples were collected from sows and piglets from 20 piglet production and rearing farms at different production times (sows antepartum and postpartum, suckling piglets, weaned piglets). In addition, the use of antibiotics was recorded. DNA extracts were subjected to quantitative real-time qPCR with primers specific for antibiotic resistance genes (int1, sul1-3, dfrA1, mcr-1, blaCTX-M), and virulence factors of relevant bacteria (C. difficile, C. perfringens, Salmonella, Escherichia/Shigella/Hafnia, E. coli). Linear and logistic regression models were used to analyze the relationship between different antibiotics and the major genes contributing to the clustering of observations for the different animal groups. Clustering revealed different farm clusters for sows, suckling piglets, and weaned piglets, with the most remarkable diversity in antibiotic use among weaned piglets. Amoxicillin, lincomycin, and enrofloxacin were identified as the most probable cause of increased odds of the presence of relevant antibiotic resistance genes (mcr1, dfrA1, blaCTX-M). Still, direct effects of a specific antibiotic on its associated resistance gene were rare. Enrofloxacin and florfenicol favored the occurrence of C. difficile in sows. The E. coli fimbriae genes were less affected by antibiotic use in sows and piglets, but the F4 fimbriae gene could be associated with the integrase 1 gene in piglets. The results confirm that multidrug-resistant enterobacteria are widespread in German pig farms and give awareness of the impact of current antibiotic use while searching for alternative health strategies.


Assuntos
Antibacterianos , Clostridioides difficile , Animais , Suínos , Feminino , Antibacterianos/farmacologia , Fazendas , Enrofloxacina , Escherichia coli , Farmacorresistência Bacteriana , Enterobacteriaceae , Clostridium perfringens
4.
Front Immunol ; 14: 960927, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36793725

RESUMO

Background: Coxiella burnetii is a zoonotic pathogen, infecting humans, livestock, pets, birds and ticks. Domestic ruminants such as cattle, sheep, and goats are the main reservoir and major cause of human infection. Infected ruminants are usually asymptomatic, while in humans infection can cause significant disease. Human and bovine macrophages differ in their permissiveness for C. burnetii strains from different host species and of various genotypes and their subsequent host cell response, but the underlying mechanism(s) at the cellular level are unknown. Methods: C. burnetii infected primary human and bovine macrophages under normoxic and hypoxic conditions were analyzed for (i) bacterial replication by CFU counts and immunofluorescence; (ii) immune regulators by westernblot and qRT-PCR; cytokines by ELISA; and metabolites by gas chromatography-mass spectrometry (GC-MS). Results: Here, we confirmed that peripheral blood-derived human macrophages prevent C. burnetii replication under oxygen-limiting conditions. In contrast, oxygen content had no influence on C. burnetii replication in bovine peripheral blood-derived macrophages. In hypoxic infected bovine macrophages, STAT3 is activated, even though HIF1α is stabilized, which otherwise prevents STAT3 activation in human macrophages. In addition, the TNFα mRNA level is higher in hypoxic than normoxic human macrophages, which correlates with increased secretion of TNFα and control of C. burnetii replication. In contrast, oxygen limitation does not impact TNFα mRNA levels in C. burnetii-infected bovine macrophages and secretion of TNFα is blocked. As TNFα is also involved in the control of C. burnetii replication in bovine macrophages, this cytokine is important for cell autonomous control and its absence is partially responsible for the ability of C. burnetii to replicate in hypoxic bovine macrophages. Further unveiling the molecular basis of macrophage-mediated control of C. burnetii replication might be the first step towards the development of host directed intervention measures to mitigate the health burden of this zoonotic agent.


Assuntos
Coxiella burnetii , Febre Q , Animais , Bovinos , Citocinas/metabolismo , Hipóxia/metabolismo , Macrófagos , Oxigênio/metabolismo , Ruminantes , Fator de Necrose Tumoral alfa/metabolismo
5.
EMBO Mol Med ; 15(2): e15931, 2023 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-36479617

RESUMO

Infection with the intracellular bacterium Coxiella (C.) burnetii can cause chronic Q fever with severe complications and limited treatment options. Here, we identify the enzyme cis-aconitate decarboxylase 1 (ACOD1 or IRG1) and its product itaconate as protective host immune pathway in Q fever. Infection of mice with C. burnetii induced expression of several anti-microbial candidate genes, including Acod1. In macrophages, Acod1 was essential for restricting C. burnetii replication, while other antimicrobial pathways were dispensable. Intratracheal or intraperitoneal infection of Acod1-/- mice caused increased C. burnetii burden, weight loss and stronger inflammatory gene expression. Exogenously added itaconate restored pathogen control in Acod1-/- mouse macrophages and blocked replication in human macrophages. In axenic cultures, itaconate directly inhibited growth of C. burnetii. Finally, treatment of infected Acod1-/- mice with itaconate efficiently reduced the tissue pathogen load. Thus, ACOD1-derived itaconate is a key factor in the macrophage-mediated defense against C. burnetii and may be exploited for novel therapeutic approaches in chronic Q fever.


Assuntos
Coxiella burnetii , Febre Q , Animais , Humanos , Camundongos , Coxiella burnetii/genética , Macrófagos , Febre Q/genética , Febre Q/microbiologia
6.
Front Cell Infect Microbiol ; 12: 867689, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35755850

RESUMO

HIF1α is an important transcription factor regulating not only cellular responses to hypoxia, but also anti-infective defense responses. We recently showed that HIF1α hampers replication of the obligate intracellular pathogen Coxiella burnetii which causes the zoonotic disease Q fever. Prior to development of chronic Q fever, it is assumed that the bacteria enter a persistent state. As HIF1α and/or hypoxia might be involved in the induction of C. burnetii persistence, we analyzed the role of HIF1α and hypoxia in the interaction of macrophages with C. burnetii to understand how the bacteria manipulate HIF1α stability and activity. We demonstrate that a C. burnetii-infection initially induces HIF1α stabilization, which decreases then over the course of an infection. This reduction depends on bacterial viability and a functional type IV secretion system (T4SS). While neither the responsible T4SS effector protein(s) nor the molecular mechanism leading to this partial HIF1α destabilization have been identified, our results demonstrate that C. burnetii influences the expression of HIF1α target genes in multiple ways. Therefore, a C. burnetii infection promotes HIF1α-mediated upregulation of several metabolic target genes; affects apoptosis-regulators towards a more pro-apoptotic signature; and under hypoxic conditions, shifts the ratio of the inflammatory genes analyzed towards a pro-inflammatory profile. Taken together, C. burnetii modulates HIF1α in a still elusive manner and alters the expression of multiple HIF1α target genes.


Assuntos
Coxiella burnetii , Febre Q , Coxiella burnetii/metabolismo , Expressão Gênica , Interações Hospedeiro-Patógeno , Humanos , Hipóxia , Febre Q/microbiologia , Sistemas de Secreção Tipo IV/metabolismo
7.
PLoS Pathog ; 18(2): e1010266, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35134097

RESUMO

Inhibition of host cell apoptosis is crucial for survival and replication of several intracellular bacterial pathogens. To interfere with apoptotic pathways, some pathogens use specialized secretion systems to inject bacterial effector proteins into the host cell cytosol. One of these pathogens is the obligate intracellular bacterium Coxiella burnetii, the etiological agent of the zoonotic disease Q fever. In this study, we analyzed the molecular activity of the anti-apoptotic T4SS effector protein AnkG (CBU0781) to understand how C. burnetii manipulates host cell viability. We demonstrate by co- and RNA-immunoprecipitation that AnkG binds to the host cell DExD box RNA helicase 21 (DDX21) as well as to the host cell 7SK small nuclear ribonucleoprotein (7SK snRNP) complex, an important regulator of the positive transcription elongation factor b (P-TEFb). The co-immunoprecipitation of AnkG with DDX21 is probably mediated by salt bridges and is independent of AnkG-7SK snRNP binding, and vice versa. It is known that DDX21 facilitates the release of P-TEFb from the 7SK snRNP complex. Consistent with the documented function of released P-TEFb in RNA Pol II pause release, RNA sequencing experiments confirmed AnkG-mediated transcriptional reprogramming and showed that expression of genes involved in apoptosis, trafficking, and transcription are influenced by AnkG. Importantly, DDX21 and P-TEFb are both essential for AnkG-mediated inhibition of host cell apoptosis, emphasizing the significance of the interaction of AnkG with both, the DDX21 protein and the 7SK RNA. In line with a critical function of AnkG in pathogenesis, the AnkG deletion C. burnetii strain was severely affected in its ability to inhibit host cell apoptosis and to generate a replicative C. burnetii-containing vacuole. In conclusion, the interference with the activity of regulatory host cell RNAs mediated by a bacterial effector protein represent a novel mechanism through which C. burnetii modulates host cell transcription, thereby enhancing permissiveness to bacterial infection.


Assuntos
Proteínas de Bactérias/metabolismo , Coxiella burnetii/metabolismo , RNA Helicases DEAD-box/metabolismo , Fator B de Elongação Transcricional Positiva/metabolismo , Febre Q/metabolismo , Ribonucleoproteínas Nucleares Pequenas/metabolismo , Sistemas de Secreção Tipo IV/metabolismo , Apoptose , Sobrevivência Celular , Coxiella burnetii/genética , Células HEK293 , Células HeLa , Interações Hospedeiro-Patógeno , Humanos , Mutação , Febre Q/microbiologia , Células THP-1
8.
PLoS One ; 16(8): e0256112, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34398927

RESUMO

Strategies to combat microbiota-associated health problems are of high interest in pig production. Successful intervention strategies with beneficial long-term effects are still missing. Most studies on pig microbiota have been conducted under standardized experimental conditions, but the situation in commercial farms differs dramatically. This study describes the fecal microbiota in German commercial pig farms under practical conditions. The study is part of the larger project "Optibiom" that aims to use bacterial composition and farm metadata to formulate tailor-made solutions for farm-specific health maintenance strategies. Special consideration is given to the sow-piglet relationship. Fecal samples from sows and their piglets were collected at two time points each in 20 different farms (sows ante- and postpartum and piglets before and after weaning). The extracted DNA was sequenced with Illumina 16S rDNA sequencing. For data analysis and visualization, differential abundance analyses, as well as hierarchical clustering and nonmetric multidimensional scaling (NMDS) were performed. A new "family unit" was implemented to compare farms based on the association between the microbiota in sows and their offspring. There are distinct changes in the microbial communities in sows before and after birth as well as in suckling and post-weaning piglets. The suckling pig microbiota is particularly different from all other groups and shows a lower bacterial diversity. While dominant genera in antepartum sows further displace the abundance of non-dominant genera postpartum, the opposite was true for piglets, where non-dominant bacteria in the suckling phase became dominant after weaning. The family unit for sows and their piglets led to separate cluster formation for some farms. The results indicate that the sow-piglet relationship is one driving force for the observed differences of the pig farms. The next step in the analysis will be the combination of metadata (feeding, housing and management practices) to find farm-specific differences that can be exploited to formulate a farm-specific health maintenance strategy.


Assuntos
Bactérias/classificação , Fazendas/organização & administração , Fezes/microbiologia , Microbiota , Suínos/microbiologia , Desmame , Animais , Animais Recém-Nascidos , Bactérias/genética , Bactérias/isolamento & purificação , Fazendas/normas , Feminino , Alemanha , Lactação/fisiologia
9.
Cell Microbiol ; 23(4): e13305, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33355405

RESUMO

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.


Assuntos
Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Coxiella burnetii/patogenicidade , Estresse do Retículo Endoplasmático , Interações Hospedeiro-Patógeno , Mariposas/microbiologia , Transdução de Sinais , Animais , Morte Celular , Coxiella burnetii/química , Coxiella burnetii/genética , Replicação do DNA , Células HEK293 , Humanos , Larva/microbiologia
10.
Cell Mol Life Sci ; 78(5): 1887-1907, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33125509

RESUMO

Various factors of the tissue microenvironment such as the oxygen concentration influence the host-pathogen interaction. During the past decade, hypoxia-driven signaling via hypoxia-inducible factors (HIF) has emerged as an important factor that affects both the pathogen and the host. In this chapter, we will review the current knowledge of this complex interplay, with a particular emphasis given to the impact of hypoxia and HIF on the inflammatory and antimicrobial activity of myeloid cells, the bacterial responses to hypoxia and the containment of bacterial infections under oxygen-limited conditions. We will also summarize how low oxygen concentrations influence the metabolism of neutrophils, macrophages and dendritic cells. Finally, we will discuss the consequences of hypoxia and HIFα activation for the invading pathogen, with a focus on Pseudomonas aeruginosa, Mycobacterium tuberculosis, Coxiella burnetii, Salmonella enterica and Staphylococcus aureus. This includes a description of the mechanisms and microbial factors, which the pathogens use to sense and react to hypoxic conditions.


Assuntos
Mycobacterium tuberculosis/fisiologia , Células Mieloides/microbiologia , Pseudomonas aeruginosa/fisiologia , Staphylococcus aureus/fisiologia , Animais , Hipóxia Celular , Interações Hospedeiro-Patógeno , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Células Mieloides/metabolismo , Oxigênio/metabolismo
11.
Front Cell Infect Microbiol ; 10: 559915, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33282747

RESUMO

Coxiella burnetii is an obligate intracellular pathogen and the causative agent of the zoonotic disease Q fever. Following uptake by alveolar macrophages, the pathogen replicates in an acidic phagolysosomal vacuole, the C. burnetii-containing vacuole (CCV). Effector proteins translocated into the host cell by the type IV secretion system (T4SS) are important for the establishment of the CCV. Here we focus on the effector protein AnkF and its role in establishing the CCV. The C. burnetii AnkF knock out mutant invades host cells as efficiently as wild-type C. burnetii, but this mutant is hampered in its ability to replicate intracellularly, indicating that AnkF might be involved in the development of a replicative CCV. To unravel the underlying reason(s), we searched for AnkF interactors in host cells and identified vimentin through a yeast two-hybrid approach. While AnkF does not alter vimentin expression at the mRNA or protein levels, the presence of AnkF results in structural reorganization and vesicular co-localization with recombinant vimentin. Ectopically expressed AnkF partially accumulates around the established CCV and endogenous vimentin is recruited to the CCV in a time-dependent manner, suggesting that AnkF might attract vimentin to the CCV. However, knocking-down endogenous vimentin does not affect intracellular replication of C. burnetii. Other cytoskeletal components are recruited to the CCV and might compensate for the lack of vimentin. Taken together, AnkF is essential for the establishment of the replicative CCV, however, its mode of action is still elusive.


Assuntos
Coxiella burnetii , Febre Q , Proteínas de Bactérias/genética , Interações Hospedeiro-Patógeno , Humanos , Sistemas de Secreção Tipo IV/genética , Vacúolos
12.
Sci Rep ; 10(1): 15396, 2020 09 21.
Artigo em Inglês | MEDLINE | ID: mdl-32958854

RESUMO

The ability to inhibit host cell apoptosis is important for the intracellular replication of the obligate intracellular pathogen Coxiella burnetii, as it allows the completion of the lengthy bacterial replication cycle. Effector proteins injected into the host cell by the C. burnetii type IVB secretion system (T4BSS) are required for the inhibition of host cell apoptosis. AnkG is one of these anti-apoptotic effector proteins. The inhibitory effect of AnkG requires its nuclear localization, which depends on p32-dependent intracellular trafficking and importin-α1-mediated nuclear entry of AnkG. Here, we compared the sequences of ankG from 37 C. burnetii isolates and classified them in three groups based on the predicted protein size. The comparison of the three different groups allowed us to identify the first 28 amino acids as essential and sufficient for the anti-apoptotic activity of AnkG. Importantly, only the full-length protein from the first group is a bona fide effector protein injected into host cells during infection and has anti-apoptotic activity. Finally, using the Galleria mellonella infection model, we observed that AnkG from the first group has the ability to attenuate pathology during in vivo infection, as it allows survival of the larvae despite bacterial replication.


Assuntos
Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Proteínas de Bactérias/fisiologia , Morte Celular/efeitos dos fármacos , Coxiella burnetii/metabolismo , Interações Hospedeiro-Patógeno , Humanos , Transporte Proteico , Alinhamento de Sequência , Fatores de Virulência/metabolismo
13.
Infect Immun ; 88(11)2020 10 19.
Artigo em Inglês | MEDLINE | ID: mdl-32817330

RESUMO

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.


Assuntos
Degranulação Celular/imunologia , Imunidade Celular/imunologia , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/microbiologia , Febre Q/imunologia , Animais , Coxiella burnetii , Camundongos
14.
Yale J Biol Med ; 92(4): 619-628, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31866777

RESUMO

The obligate intracellular pathogen Coxiella burnetii is the causative agent of the worldwide zoonotic disease Q fever. This Gram-negative bacterium infects macrophages where it establishes a replicative niche in an acidic and phagolysosome-like vacuole. Establishing and maintaining the niche requires a functional type IV secretion system (T4SS) which translocates multiple effector proteins into the host cell. These effector proteins act by manipulating diverse cellular processes allowing the bacterium to establish an infection and complete its complex biphasic developmental cycle. The lengthy nature of this life cycle suggests that C. burnetii has to successfully deal with cellular defense processes. Cell death is one mechanism infected cells frequently utilize to control or to at least minimize the impact of an infection. To date, four effector proteins have been identified in C. burnetii, which interfere with the induction of cell death. Three, AnkG, CaeA, and CaeB, affect intrinsic apoptosis, CaeA additionally extrinsic apoptosis. The proteins target different steps of the apoptotic pathway and are not conserved among isolates suggesting redundancy as an important feature of cell death inhibition. The fourth effector protein, IcaA, interferes with the non-canonical pathway of pyroptosis, an important inflammatory cell death pathway for controlling infectious disease. Autophagy is relevant for the C. burnetii life-cycle, but to which extent autophagic cell death is a factor in bacterial survival and proliferation is still not clear. To convincingly understand how bacterial manipulation of autophagy affects cell death either directly or indirectly will require further experiments. Collectively, C. burnetii modulates the extrinsic and intrinsic apoptotic pathways and non-canonical pyroptosis to inhibit host cell death, thereby providing a stable, intracellular niche for the course of the pathogen's infectious cycle.


Assuntos
Coxiella burnetii/fisiologia , Interações Hospedeiro-Patógeno , Animais , Apoptose , Autofagia , Humanos , Modelos Biológicos
15.
Cell Rep ; 26(13): 3502-3510.e6, 2019 03 26.
Artigo em Inglês | MEDLINE | ID: mdl-30917307

RESUMO

In hypoxic and inflamed tissues, oxygen (O2)-dependent antimicrobial defenses are impaired due to a shortage of O2. To gain insight into the mechanisms that control bacterial infection under hypoxic conditions, we infected macrophages with the obligate intracellular pathogen Coxiella burnetii, the causative agent of Q fever. Our experiments revealed that hypoxia impeded C. burnetii replication in a hypoxia-inducible factor (HIF) 1α-dependent manner. Mechanistically, under hypoxia, HIF1α impaired the activity of STAT3, which in turn reduced the intracellular level of TCA cycle intermediates, including citrate, and impeded C. burnetii replication in macrophages. However, bacterial viability was maintained, allowing the persistence of C. burnetii, which is a prerequisite for the development of chronic Q fever. This knowledge will open future research avenues on the pathogenesis of chronic Q fever. In addition, the regulation of TCA cycle metabolites by HIF1α represents a previously unappreciated mechanism of host defense against intracellular pathogens.


Assuntos
Ciclo do Ácido Cítrico , Coxiella burnetii/imunologia , Macrófagos/imunologia , Adulto , Animais , Hipóxia Celular , Células Cultivadas , Feminino , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/fisiologia , Macrófagos/microbiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Oxigênio/metabolismo , Febre Q/imunologia , Fator de Transcrição STAT3/fisiologia
16.
Front Immunol ; 10: 165, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30800124

RESUMO

The intracellular pathogen Coxiella (C.) burnetii causes Q fever, a usually self-limiting respiratory infection that becomes chronic and severe in some patients. Innate immune recognition of C. burnetii and its role in the decision between resolution and chronicity is not understood well. However, TLR2 is important for the response to C. burnetii in mice, and genetic polymorphisms in Myd88 have been associated with chronic Q fever in humans. Here, we have employed MyD88-deficient mice in infection models with the attenuated C. burnetii Nine Mile phase II strain (NMII). Myd88-/- macrophages failed to restrict the growth of NMII in vitro, and to upregulate production of the cytokines TNF, IL-6, and IL-10. Following intraperitoneal infection, NMII bacterial burden was significantly higher on day 5 and 20 in organs of Myd88-/- mice. After infection via the natural route by intratracheal injection, a higher bacterial load in the lung and increased dissemination of NMII to other organs was observed in MyD88-deficient mice. While wild-type mice essentially cleared NMII on day 27 after intratracheal infection, it was still readily detectable on day 42 in multiple organs in the absence of MyD88. Despite the elevated bacterial load, Myd88-/- mice had less granulomatous inflammation and expressed significantly lower levels of chemoattractants, inflammatory cytokines, and of several IFNγ-induced genes relevant for control of intracellular pathogens. Together, our results show that MyD88-dependent signaling is essential for early control of C. burnetii replication and to prevent systemic spreading. The continued presence of NMII in the organs of Myd88-/- mice constitutes a new mouse model to study determinants of chronicity and resolution in Q fever.


Assuntos
Coxiella burnetii/genética , Fator 88 de Diferenciação Mieloide/genética , Fator 88 de Diferenciação Mieloide/metabolismo , Febre Q/microbiologia , Animais , Citocinas/metabolismo , Modelos Animais de Doenças , Técnicas de Inativação de Genes , Genoma Bacteriano , Fígado/microbiologia , Pulmão/microbiologia , Macrófagos/imunologia , Macrófagos/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Óxido Nítrico Sintase Tipo II/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Transdução de Sinais/imunologia , Baço/microbiologia
17.
Curr Opin Microbiol ; 47: 59-65, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30640035

RESUMO

Intracellular bacterial pathogens intimately interact with the infected host cell to prevent elimination and to ensure survival. One group of intracellular pathogens, including Coxiella burnetii, Legionella pneumophila, Brucella spp., Anaplasma phagocytophilum, and Ehrlichia chaffeensis, utilizes a type IV secretion system (T4SS) that injects effectors to modulate host cell signalling, vesicular trafficking, autophagy, cell death and transcription to ensure survival [1]. So far, little emphasis has been directed towards understanding how these bacteria manipulate host cell metabolism. This manipulation is not only important for gaining access to nutrients, but also for regulating specific virulence programs [2,3]. Here, we will summarize recent progress made in characterizing the manipulation of host cell metabolism by C. burnetii and other intracellular pathogens utilizing a T4SS.


Assuntos
Bactérias Gram-Negativas/crescimento & desenvolvimento , Bactérias Gram-Negativas/metabolismo , Interações Hospedeiro-Patógeno , Sistemas de Secreção Tipo IV/metabolismo , Fatores de Virulência/metabolismo , Animais , Bactérias Gram-Negativas/patogenicidade , Humanos , Virulência
18.
Nat Commun ; 9(1): 3467, 2018 08 27.
Artigo em Inglês | MEDLINE | ID: mdl-30150748

RESUMO

Many bacteria export effector proteins fulfilling their function in membranes of a eukaryotic host. These effector membrane proteins appear to contain signals for two incompatible bacterial secretion pathways in the same protein: a specific export signal, as well as transmembrane segments that one would expect to mediate targeting to the bacterial inner membrane. Here, we show that the transmembrane segments of effector proteins of type III and type IV secretion systems indeed integrate in the membrane as required in the eukaryotic host, but that their hydrophobicity in most instances is just below the threshold required for mediating targeting to the bacterial inner membrane. Furthermore, we show that binding of type III secretion chaperones to both the effector's chaperone-binding domain and adjacent hydrophobic transmembrane segments also prevents erroneous targeting. These results highlight the evolution of a fine discrimination between targeting pathways that is critical for the virulence of many bacterial pathogens.


Assuntos
Proteínas de Bactérias/metabolismo , Sistemas de Secreção Bacterianos/metabolismo , Proteínas de Membrana/metabolismo , Interações Hidrofóbicas e Hidrofílicas , Ligação Proteica , Virulência
19.
J Microbiol Methods ; 151: 39-43, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29857017

RESUMO

Flow cytometry enables the analysis of cells by labeling them with fluorescent probes. We describe a novel flow cytometric approach permitting reliable analysis of Coxiella (C.) burnetii-infected cells. The method quantifies infection-forming units (IFUs) in a dose-dependent manner and allows for the specific detection of infection/replication-competent coxiella in cell cultures.


Assuntos
Coxiella burnetii/isolamento & purificação , Citometria de Fluxo , Animais , Técnicas de Cultura de Células , Células Cultivadas , Contagem de Colônia Microbiana , Coxiella burnetii/metabolismo , Interações Hospedeiro-Patógeno , Camundongos
20.
Int J Med Microbiol ; 308(1): 77-83, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-28935173

RESUMO

Coxiella burnetii is an obligate intracellular and airborne pathogen which can cause the zoonotic disease Q fever. After inhalation of contaminated aerosols alveolar macrophages are taking up C. burnetii into a phagosome. This phagosome matures to a very large vacuole called the C. burnetii-containing vacuole (CCV). Host endogenous and bacterial driven processes lead to the biogenesis of this unusual compartment, which resembles partially a phagolysosome. However, there are several important differences to the classical phagolysosome, which are crucial for the ability of C. burnetii to replicate intracellularly and depend on a functional type IV secretion system (T4SS). The T4SS delivers effector proteins into the host cell cytoplasm to redirect intracellular processes, leading to the establishment of a microenvironment allowing bacterial replication. This article summarizes the current knowledge of the microenvironment permissive for C. burnetii replication.


Assuntos
Coxiella burnetii/fisiologia , Interações Hospedeiro-Patógeno , Macrófagos Alveolares/microbiologia , Sistemas de Secreção Tipo IV/metabolismo , Vacúolos/microbiologia , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Coxiella burnetii/crescimento & desenvolvimento , Coxiella burnetii/metabolismo , Humanos , Modelos Biológicos , Fagossomos/metabolismo , Fagossomos/microbiologia , Febre Q/microbiologia , Sistemas de Secreção Tipo IV/genética , Vacúolos/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...