Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 50
Filtrar
1.
J Bacteriol ; 204(10): e0005422, 2022 Oct 18.
Artigo em Inglês | MEDLINE | ID: mdl-36106853

RESUMO

The outer membrane (OM) of Gram-negative bacteria efficiently protects from harmful environmental stresses such as antibiotics, disinfectants, or dryness. The main constituents of the OM are integral OM ß-barrel proteins (OMPs). In Gram-negative bacteria such as Escherichia coli, Yersinia enterocolitica, and Pseudomonas aeruginosa, the insertion of OMPs depends on a sophisticated biogenesis pathway. This comprises the SecYEG translocon, which enables inner membrane (IM) passage; the chaperones SurA, Skp, and DegP, which facilitate the passage of ß-barrel OMPs through the periplasm; and the ß-barrel assembly machinery (BAM), which facilitates insertion into the OM. In E. coli, Y. enterocolitica, and P. aeruginosa, the deletion of SurA is particularly detrimental and leads to a loss of OM integrity, sensitization to antibiotic treatment, and reduced virulence. In search of targets that could be exploited to develop compounds that interfere with OM integrity in Acinetobacter baumannii, we employed the multidrug-resistant strain AB5075 to generate single gene knockout strains lacking individual periplasmic chaperones. In contrast to E. coli, Y. enterocolitica, and P. aeruginosa, AB5075 tolerates the lack of SurA, Skp, or DegP with only weak mutant phenotypes. While the double knockout strains ΔsurAΔskp and ΔsurAΔdegP are conditionally lethal in E. coli, all double deletions were well tolerated by AB5075. Strikingly, even a triple-knockout strain of AB5075, lacking surA, skp, and degP, was viable. IMPORTANCE Acinetobacter baumannii is a major threat to human health due to its ability to persist in the hospital environment, resistance to antibiotic treatment, and ability to deploy multiple and redundant virulence factors. In a rising number of cases, infections with multidrug-resistant A. baumannii end up fatally, because all antibiotic treatment options fail. Thus, novel targets have to be identified and alternative therapeutics have to be developed. The knockout of periplasmic chaperones has previously proven to significantly reduce virulence and even break antibiotic resistance in other Gram-negative pathogens. Our study in A. baumannii demonstrates how variable the importance of the periplasmic chaperones SurA, Skp, and DegP can be and suggests the existence of mechanisms allowing A. baumannii to cope with the lack of the three periplasmic chaperones.


Assuntos
Acinetobacter baumannii , Proteínas da Membrana Bacteriana Externa , Desinfetantes , Humanos , Acinetobacter baumannii/genética , Acinetobacter baumannii/metabolismo , Antibacterianos/farmacologia , Antibacterianos/metabolismo , Proteínas da Membrana Bacteriana Externa/genética , Proteínas da Membrana Bacteriana Externa/metabolismo , Infecção Hospitalar/microbiologia , Proteínas de Ligação a DNA/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Hospitais , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Periplasma/metabolismo , Dobramento de Proteína , Canais de Translocação SEC/metabolismo , Fatores de Virulência/metabolismo , Yersinia enterocolitica , Pseudomonas aeruginosa , Farmacorresistência Bacteriana Múltipla
2.
Mol Microbiol ; 113(6): 1240-1254, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32068313

RESUMO

The elucidation of the molecular mechanisms of secretion through bacterial protein secretion systems is impeded by a shortage of assays to quantitatively assess secretion kinetics. Also the analysis of the biological role of these secretion systems as well as the identification of inhibitors targeting these systems would greatly benefit from the availability of a simple, quick and quantitative assay to monitor principle secretion and injection into host cells. Here, we present a versatile solution to this need, utilizing the small and very bright NanoLuc luciferase to assess the function of the type III secretion system encoded by Salmonella pathogenicity island 1. Type III secretion substrate-NanoLuc fusions are readily secreted into the culture supernatant, where they can be quantified by luminometry after removal of bacteria. The NanoLuc-based secretion assay features a very high signal-to-noise ratio and sensitivity down to the nanolitre scale. The assay enables monitoring of secretion kinetics and is adaptable to a high throughput screening format in 384-well microplates. We further developed a split NanoLuc-based assay that enables the real-time monitoring of type III secretion-dependent injection of effector-HiBiT fusions into host cells stably expressing the complementing NanoLuc-LgBiT.


Assuntos
Proteínas de Bactérias/metabolismo , Medições Luminescentes/métodos , Sistemas de Secreção Tipo III/metabolismo , Ensaios de Triagem em Larga Escala/métodos , Luciferases , Transporte Proteico , Salmonella/genética , Salmonella/metabolismo
3.
Artigo em Inglês | MEDLINE | ID: mdl-31818817

RESUMO

With the aim to identify potential new targets to restore antimicrobial susceptibility of multidrug-resistant (MDR) Pseudomonas aeruginosa isolates, we generated a high-density transposon (Tn) insertion mutant library in an MDR P. aeruginosa bloodstream isolate (isolate ID40). The depletion of Tn insertion mutants upon exposure to cefepime or meropenem was measured in order to determine the common resistome for these clinically important antipseudomonal ß-lactam antibiotics. The approach was validated by clean deletions of genes involved in peptidoglycan synthesis/recycling, such as the genes for the lytic transglycosylase MltG, the murein (Mur) endopeptidase MepM1, the MurNAc/GlcNAc kinase AmgK, and the uncharacterized protein YgfB, all of which were identified in our screen as playing a decisive role in survival after treatment with cefepime or meropenem. We found that the antibiotic resistance of P. aeruginosa can be overcome by targeting usually nonessential genes that turn essential in the presence of therapeutic concentrations of antibiotics. For all validated genes, we demonstrated that their deletion leads to the reduction of ampC expression, resulting in a significant decrease in ß-lactamase activity, and consequently, these mutants partly or completely lost resistance against cephalosporins, carbapenems, and acylaminopenicillins. In summary, the determined resistome may comprise promising targets for the development of drugs that may be used to restore sensitivity to existing antibiotics, specifically in MDR strains of P. aeruginosa.


Assuntos
Antibacterianos/farmacologia , Proteínas de Bactérias/genética , Elementos de DNA Transponíveis , Farmacorresistência Bacteriana Múltipla/genética , Pseudomonas aeruginosa/genética , Resistência beta-Lactâmica/genética , Proteínas de Bactérias/metabolismo , Cefepima/farmacologia , Endopeptidases/deficiência , Endopeptidases/genética , Deleção de Genes , Regulação Bacteriana da Expressão Gênica , Glicosiltransferases/deficiência , Glicosiltransferases/genética , Humanos , Meropeném/farmacologia , Testes de Sensibilidade Microbiana , Mutagênese , Fosfotransferases (Aceptor do Grupo Álcool)/deficiência , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Infecções por Pseudomonas/tratamento farmacológico , Infecções por Pseudomonas/microbiologia , Pseudomonas aeruginosa/efeitos dos fármacos , Pseudomonas aeruginosa/enzimologia , Pseudomonas aeruginosa/isolamento & purificação , beta-Lactamases/genética , beta-Lactamases/metabolismo
4.
Int J Med Microbiol ; 309(5): 344-350, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-31178419

RESUMO

Type III secretion systems (T3SS) play a crucial role for virulence in many Gram-negative bacteria. After tight bacterial contact to host cells, the T3SS injects effector proteins into the host cells, which leads to cell invasion, tissue destruction and/or immune evasion. Over the last decade several attempts were made to characterize the host-cell interactions which precede and determine effector protein injection during infection. The development of the TEM-ß-lactamase reporter was an important breakthrough to achieve this goal. By this means it was demonstrated that during infection with many Gram-negative pathogens such as Salmonella, Pseudomonas or Yersinia the main targets of T3SS are leukocytes of the myeloid lineage such as neutrophils, macrophages or dendritic cells. This is due to the recruitment of these cells to the site of infection, but also due to the specific interplay between bacterial and host cells. Comprehensive studies on Yersinia pestis, Yersinia enterocolitica and Yersinia pseudotuberculosis effector translocation show that adhesins such as Invasin (Inv), Yersinia adhesin A (YadA) and attachment and invasion locus (Ail) are critical for effector translocation. Here, mainly the complex interaction of YadA and Ail with various host cell receptor repertoires on leukocytes and the modulatory effects of serum factors direct effector translocation predominantly towards myeloid cells. The current understanding suggests that mostly protein based interactions between bacteria and host determine host cell specific effector translocation during Yersinia infection. However, for Shigella dysenteriae infection it was shown that glycan-glycan interactions can also play a critical role for the adhesion preceding effector translocation. In addition, the Shigella infection model revealed that the activation status of cells is a further criterium directing effector translocation into a distinct cell population. In this review the current understanding of the complex and species-specific interaction between bacteria and host cells leading to type III secretion is discussed.


Assuntos
Aderência Bacteriana , Interações entre Hospedeiro e Microrganismos , Transporte Proteico , Sistemas de Secreção Tipo III/metabolismo , Adesinas Bacterianas/metabolismo , Animais , Proteínas da Membrana Bacteriana Externa/metabolismo , Humanos , Shigella/imunologia , Shigella/patogenicidade , Virulência/imunologia , Fatores de Virulência/metabolismo , Yersinia/imunologia , Yersinia/patogenicidade
5.
Int J Med Microbiol ; 306(6): 357-66, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27107739

RESUMO

Enteropathogenic Yersinia enterocolitica (Ye) enters the host via contaminated food. After colonisation of the small intestine Ye invades the Peyer's patches (PPs) via M cells and disseminates to the mesenteric lymph nodes (MLNs), spleen and liver. Whether Ye uses other invasion routes and which pathogenicity factors are required remains elusive. Oral infection of lymphotoxin-ß-receptor deficient mice lacking PPs and MLNs with Ye revealed similar bacterial load in the spleen 1h post infection as wild-type mice, demonstrating a PP-independent dissemination route for Ye. Immunohistological analysis of the small intestine revealed Ye in close contact with mononuclear phagocytes (MPs), specifically CX3CR1(+) monocyte-derived cells (MCs) as well as CD103(+) dendritic cells (DCs). This finding was confirmed by flow cytometry and imaging flow cytometry analysis of lamina propria (LP) leukocytes showing CD103(+) DCs and MCs with intracellular Ye. Uptake of Ye by LP CD103(+) DCs and MCs was dependent on the pathogenicity factor invasin, whereas the adhesin YadA was dispensable as demonstrated by Ye deletion mutants. Furthermore, Ye were found exclusively associated with CD103(+) DCs in the MLNs from wild-type mice, but not from CCR7(-/-) mice, demonstrating a CCR7 dependent transport of Ye by CD103(+) DCs from LP to the MLNs. In contrast, dissemination of Ye to the spleen was dependent on MCs as significantly less Ye could be recovered from the spleen of CX3CR1(GFP/GFP) mice compared to wild-type mice. Altogether, MCs and CD103(+) DCs contribute to immediate invasion and dissemination of Ye. This together with data from other bacteria suggests MPs as general pathogenic entry site in the intestine.


Assuntos
Interações Hospedeiro-Patógeno , Intestino Delgado/patologia , Fagócitos/microbiologia , Yersiniose/patologia , Yersinia enterocolitica/imunologia , Yersinia enterocolitica/fisiologia , Animais , Carga Bacteriana , Feminino , Citometria de Fluxo , Imuno-Histoquímica , Intestino Delgado/imunologia , Intestino Delgado/microbiologia , Fígado/microbiologia , Linfonodos/imunologia , Linfonodos/microbiologia , Camundongos Endogâmicos C57BL , Nódulos Linfáticos Agregados/imunologia , Nódulos Linfáticos Agregados/microbiologia , Baço/microbiologia , Fatores de Tempo , Yersiniose/imunologia , Yersiniose/microbiologia
6.
Int J Med Microbiol ; 306(2): 77-88, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26718660

RESUMO

Injection of Yersinia outer proteins (Yops) into host cells by a type III secretion system is an important immune evasion mechanism of Yersinia enterocolitica (Ye). In this process Ye invasin (Inv) binds directly while Yersinia adhesin A (YadA) binds indirectly via extracellular matrix (ECM) proteins to ß1 integrins on host cells. Although leukocytes turned out to be an important target of Yop injection by Ye, it was unclear which Ye adhesins and which leukocyte receptors are required for Yop injection. To explain this, we investigated the role of YadA, Inv and ß1 integrins for Yop injection into leukocytes and their impact on the course of systemic Ye infection in mice. Ex vivo infection experiments revealed that adhesion of Ye via Inv or YadA is sufficient to promote Yop injection into leukocytes as revealed by a ß-lactamase reporter assay. Serum factors inhibit YadA- but not Inv-mediated Yop injection into B and T cells, shifting YadA-mediated Yop injection in the direction of neutrophils and other myeloid cells. Systemic Ye mouse infection experiments demonstrated that YadA is essential for Ye virulence and Yop injection into leukocytes, while Inv is dispensable for virulence and plays only a transient and minor role for Yop injection in the early phase of infection. Ye infection of mice with ß1 integrin-depleted leukocytes demonstrated that ß1 integrins are dispensable for YadA-mediated Yop injection into leukocytes, but contribute to Inv-mediated Yop injection. Despite reduced Yop injection into leukocytes, ß1 integrin-deficient mice exhibited an increased susceptibility for Ye infection, suggesting an important role of ß1 integrins in immune defense against Ye. This study demonstrates that Yop injection into leukocytes by Ye is largely mediated by YadA exploiting, as yet unknown, leukocyte receptors.


Assuntos
Adesinas Bacterianas/fisiologia , Proteínas da Membrana Bacteriana Externa/administração & dosagem , Integrina beta1/fisiologia , Leucócitos/metabolismo , Yersiniose/sangue , Yersinia enterocolitica , Adesinas Bacterianas/genética , Alelos , Animais , Integrina beta1/genética , Camundongos , Camundongos Endogâmicos C57BL , Plasmídeos
7.
Cell Microbiol ; 17(8): 1179-204, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25678064

RESUMO

The current paradigm suggests that Yersinia enterocolitica (Ye) adheres to host cells via the outer membrane proteins Yersinia adhesin A (YadA) or invasin (Inv) to facilitate injection of Yops by the type III secretion system. In this process Inv binds directly to ß1 integrins of host cells while YadA may bind indirectly via extracellular matrix proteins to ß1 integrins. Here we challenged this paradigm and investigated the requirements for Yop injection. We demonstrate that Inv- but not YadA-mediated adhesion depends on ß1 integrin binding and activation, and that tight adhesion is a prerequisite for Yop injection. By means of novel transgenic cell lines, shRNA approaches and RGD peptides, we found that YadA, in contrast to Inv, may use a broad host cell receptor repertoire for host cell adhesion. In the absence of ß1 integrins, YadA mediates Yop injection by interaction with αV integrins in cooperation with yet unknown cofactors expressed by epithelial cells, but not fibroblasts. Electron microscopic and flow chamber studies revealed that a defined intimate contact area between Ye and host cells resulting in adhesion forces resisting shear stress is required for Yop injection. Thus, the indirect binding of YadA to a broad extracellular matrix (ECM) binding host cell receptor repertoire of different cell types makes YadA a versatile tool to ensure Yop injection. In conclusion, given the differential expression of the outer membrane proteins Inv and YadA in the course of Ye infection and differential expression of integrins by various host cell populations, the data demonstrate that Ye is flexibly armed to accomplish Yop injection in different host cell types, a central event in its immune evasion strategy.


Assuntos
Adesinas Bacterianas/metabolismo , Aderência Bacteriana , Toxinas Bacterianas/metabolismo , Interações Hospedeiro-Patógeno , Yersinia enterocolitica/fisiologia , Células Epiteliais/microbiologia , Fibroblastos/metabolismo , Citometria de Fluxo , Integrina alfaV/metabolismo , Integrina beta1/metabolismo , Microscopia Eletrônica , Ligação Proteica , Transporte Proteico
8.
J Immunol ; 191(7): 3913-21, 2013 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-24014876

RESUMO

Type I IFN signaling amplifies the secretion of LPS-induced proinflammatory cytokines such as TNF-α or IL-6 and might thus contribute to the high mortality associated with Gram-negative septic shock in humans. The underlying molecular mechanism, however, is ill defined. In this study, we report the generation of mice deficient in IFN-induced protein with tetratricopeptide repeats 2 (Ifit2) and demonstrate that Ifit2 is a critical signaling intermediate for LPS-induced septic shock. Ifit2 expression was significantly upregulated in response to LPS challenge in an IFN-α receptor- and IFN regulatory factor (Irf)9-dependent manner. Also, LPS induced secretion of IL-6 and TNF-α by bone marrow-derived macrophages (BMDMs) was significantly enhanced in the presence of Ifit2. In accordance, Ifit2-deficient mice exhibited significantly reduced serum levels of IL-6 and TNF-α and reduced mortality in an endotoxin shock model. Investigation of the underlying signal transduction events revealed that Ifit2 upregulates Irf3 phosphorylation. In the absence of Irf3, reduced Ifn-ß mRNA expression and Ifit2 protein expression after LPS stimulation was found. Also, Tnf-α and Il-6 secretion but not Tnf-α and Il-6 mRNA expression levels were reduced. Thus, IFN-stimulated Ifit2 via enhanced Irf3 phosphorylation upregulates the secretion of proinflammatory cytokines. It thereby amplifies LPS-induced cytokine production and critically influences the outcome of endotoxin shock.


Assuntos
Interferon Tipo I/metabolismo , Lipopolissacarídeos/imunologia , Proteínas/imunologia , Choque Séptico/imunologia , Fator de Necrose Tumoral alfa/biossíntese , Animais , Proteínas Reguladoras de Apoptose , Citocinas/biossíntese , Modelos Animais de Doenças , Feminino , Regulação da Expressão Gênica , Inflamação/imunologia , Inflamação/metabolismo , Camundongos , Camundongos Knockout , Proteínas/genética , RNA Mensageiro/genética , Proteínas de Ligação a RNA , Transdução de Sinais
9.
bioRxiv ; 2024 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-38559167

RESUMO

To promote intracellular survival and infection, Legionella spp. translocate hundreds of effector proteins into eukaryotic host cells using a type IV b protein secretion system (T4bSS). T4bSS are well known to translocate soluble as well as transmembrane domain-containing effector proteins (TMD-effectors) but the mechanisms of secretion are still poorly understood. Herein we investigated the secretion of hydrophobic TMD-effectors, of which about 80 were previously reported to be encoded by L. pneumophila. A proteomic analysis of fractionated membranes revealed that TMD-effectors are targeted to and inserted into the bacterial inner membranes of L. pneumophila independent of the presence of a functional T4bSS. While the T4bSS chaperones IcmS and IcmW were critical for secretion of all tested TMD-effectors, they did not influence inner membrane targeting of these proteins. As for soluble effector proteins, translocation of TMD-effectors into host cells depended on a C-terminal secretion signal and this signal needed to be presented towards the cytoplasmic side of the inner membrane. A different secretion behavior of TMD- and soluble effectors and the need for small periplasmic loops within TMD-effectors provided strong evidence that TMD-effectors are secreted in a two-step secretion process: Initially, an inner membrane intermediate is formed, that is extracted towards the cytoplasmic side, possibly by the help of the type IV coupling protein complex and subsequently secreted into eukaryotic host cells by the T4bSS core complex. Overall, our study highlights the amazing versatility of T4bSS to secrete soluble and TMD-effectors from different subcellular locations of the bacterial cell.

10.
Commun Biol ; 6(1): 254, 2023 03 10.
Artigo em Inglês | MEDLINE | ID: mdl-36894667

RESUMO

YgfB-mediated ß-lactam resistance was recently identified in multi drug resistant Pseudomonas aeruginosa. We show that YgfB upregulates expression of the ß-lactamase AmpC by repressing the function of the regulator of the programmed cell death pathway AlpA. In response to DNA damage, the antiterminator AlpA induces expression of the alpBCDE autolysis genes and of the peptidoglycan amidase AmpDh3. YgfB interacts with AlpA and represses the ampDh3 expression. Thus, YgfB indirectly prevents AmpDh3 from reducing the levels of cell wall-derived 1,6-anhydro-N-acetylmuramyl-peptides, required to induce the transcriptional activator AmpR in promoting the ampC expression and ß-lactam resistance. Ciprofloxacin-mediated DNA damage induces AlpA-dependent production of AmpDh3 as previously shown, which should reduce ß-lactam resistance. YgfB, however, counteracts the ß-lactam enhancing activity of ciprofloxacin by repressing ampDh3 expression and lowering the benefits of this drug combination. Altogether, YgfB represents an additional player in the complex regulatory network of AmpC regulation.


Assuntos
Pseudomonas aeruginosa , Resistência beta-Lactâmica , Pseudomonas aeruginosa/genética , Resistência beta-Lactâmica/genética , Ciprofloxacina/farmacologia , beta-Lactamas/farmacologia
11.
Cell Physiol Biochem ; 30(4): 853-62, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22907484

RESUMO

TFF2 is one of the members of the trefoil factor family, known for its role in protection of gastrointestinal epithelia upon injury; however, recent studies suggest that TFF2 could also play an important role in the immune system. In the present study Tff2 deficient and wild type mice were infected by Y. enterocolitica which resulted in a lethal outcome in all Tff2 deficient mice, but not in WT animals. Yersinia invaded Peyer's patches more efficiently as shown by high bacterial titers in the KO mice while wild type mice displayed lower titers and a visible bacterial accumulation in the intestine. Bacterial accumulation in Peyer's patches of Tff2 deficient mice was accompanied by increased recruitment of macrophages. While an increased level of MAC-1 positive cells was observed in the spleens of both Tff2 deficient and WT mice at third day post infection, bacterial dissemination to liver, lung and kidneys was observed only in Tff2 knock-out mice. Analysis of the cellular composition of spleen did not reveal any substantial alteration to WT animals, suggesting possible disregulation of hemopoietic cells involved in immune response to Y. enterocolitica. These new data indicate that Tff2 plays an important role in immune response by protecting the organism from consequences of infection and that Tff2 knock-out mice react adversely to bacterial infections, in this case specifically to Y. enterocolitica.


Assuntos
Técnicas de Inativação de Genes , Intestinos/microbiologia , Mucinas/genética , Proteínas Musculares/genética , Peptídeos/genética , Nódulos Linfáticos Agregados/microbiologia , Yersiniose/genética , Yersiniose/transmissão , Yersinia enterocolitica/patogenicidade , Animais , Feminino , Imunidade Inata , Mucosa Intestinal/metabolismo , Intestinos/imunologia , Intestinos/patologia , Antígeno de Macrófago 1/análise , Antígeno de Macrófago 1/imunologia , Camundongos , Camundongos Knockout , Mucinas/imunologia , Proteínas Musculares/imunologia , Peptídeos/imunologia , Nódulos Linfáticos Agregados/imunologia , Nódulos Linfáticos Agregados/metabolismo , Nódulos Linfáticos Agregados/patologia , Baço/imunologia , Baço/metabolismo , Baço/microbiologia , Baço/patologia , Fator Trefoil-2 , Yersiniose/imunologia , Yersiniose/patologia , Yersinia enterocolitica/imunologia
12.
PLoS Pathog ; 6(11): e1001212, 2010 Nov 24.
Artigo em Inglês | MEDLINE | ID: mdl-21124820

RESUMO

CD4(+) T cells are essential for the control of Yersinia enterocolitica (Ye) infection in mice. Ye can inhibit dendritic cell (DC) antigen uptake and degradation, maturation and subsequently T-cell activation in vitro. Here we investigated the effects of Ye infection on splenic DCs and T-cell proliferation in an experimental mouse infection model. We found that OVA-specific CD4(+) T cells had a reduced potential to proliferate when stimulated with OVA after infection with Ye compared to control mice. Additionally, proliferation of OVA-specific CD4(+) T cells was markedly reduced when cultured with splenic CD8α(+) DCs from Ye infected mice in the presence of OVA. In contrast, T-cell proliferation was not impaired in cultures with CD4(+) or CD4(-)CD8α(-) DCs isolated from Ye infected mice. However, OVA uptake and degradation as well as cytokine production were impaired in CD8α(+) DCs, but not in CD4(+) and CD4(-)CD8α(-) DCs after Ye infection. Pathogenicity factors (Yops) from Ye were most frequently injected into CD8α(+) DCs, resulting in less MHC class II and CD86 expression than on non-injected CD8α(+) DCs. Three days post infection with Ye the number of splenic CD8α(+) and CD4(+) DCs was reduced by 50% and 90%, respectively. The decreased number of DC subsets, which was dependent on TLR4 and TRIF signaling, was the result of a faster proliferation and suppressed de novo DC generation. Together, we show that Ye infection negatively regulates the stimulatory capacity of some but not all splenic DC subpopulations in vivo. This leads to differential antigen uptake and degradation, cytokine production, cell loss, and cell death rates in various DC subpopulations. The data suggest that these effects might be caused directly by injection of Yops into DCs and indirectly by affecting the homeostasis of CD4(+) and CD8α(+) DCs. These events may contribute to reduced T-cell proliferation and immune evasion of Ye.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Células Dendríticas/imunologia , Células Dendríticas/microbiologia , Evasão da Resposta Imune/imunologia , Fatores de Virulência/metabolismo , Yersiniose/imunologia , Yersinia enterocolitica/imunologia , Proteínas Adaptadoras de Transporte Vesicular/fisiologia , Animais , Apresentação de Antígeno , Western Blotting , Linfócitos T CD4-Positivos/microbiologia , Citocinas/metabolismo , Células Dendríticas/metabolismo , Feminino , Citometria de Fluxo , Imunofluorescência , Técnicas Imunoenzimáticas , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Baço/citologia , Baço/imunologia , Baço/microbiologia , Receptor 4 Toll-Like/fisiologia , Fatores de Virulência/genética , Yersiniose/metabolismo , Yersiniose/microbiologia
13.
Methods Mol Biol ; 2427: 57-71, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35619025

RESUMO

Studies of bacterial protein secretion have relied on a variety of reporters that allow the tracking of secreted proteins. However, the lack of truly quantitative and highly sensitive reporters has hindered, in particular, the investigation of the kinetics of protein secretion. In this chapter, we describe a luminescence-based assay using NanoLuc luciferase to analyse secretion and injection into host cells of type III secretion (T3S) substrates encoded on Salmonella pathogenicity island-1 (SPI-1). This method has a very high sensitivity and high signal-to-noise ratio. Moreover, the simplicity of the protocol and the rapid determination and quantification of the luminescence makes it ideal for the monitoring of the kinetics of secretion but also convenient for high-throughput screenings. The protocols presented here include (1) Salmonella SPI-1 secretion assay, where the T3S substrates-NanoLuc fusions are detected by luminometry in the bacterial supernatant, and (2) Salmonella injection assays, using the split-Nanoluc (HiBiT/LgBiT) to monitor the injection of T3S substrates-HiBiT fusions into the host cells stably expressing LgBiT.


Assuntos
Ilhas Genômicas , Salmonella , Peptídeos e Proteínas de Sinalização Intercelular , Luciferases/genética , Luciferases/metabolismo , Salmonella/genética , Salmonella/metabolismo
14.
Front Microbiol ; 13: 913434, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35910598

RESUMO

Bartonella spp. are Gram-negative facultative intracellular pathogens that infect diverse mammals and cause a long-lasting intra-erythrocytic bacteremia in their natural host. These bacteria translocate Bartonella effector proteins (Beps) into host cells via their VirB/VirD4 type 4 secretion system (T4SS) in order to subvert host cellular functions, thereby leading to the downregulation of innate immune responses. Most studies on the functional analysis of the VirB/VirD4 T4SS and the Beps were performed with the major zoonotic pathogen Bartonella henselae for which efficient in vitro infection protocols have been established. However, its natural host, the cat, is unsuitable as an experimental infection model. In vivo studies were mostly confined to rodent models using rodent-specific Bartonella species, while the in vitro infection protocols devised for B. henselae are not transferable for those pathogens. The disparities of in vitro and in vivo studies in different species have hampered progress in our understanding of Bartonella pathogenesis. Here we describe the murine-specific strain Bartonella taylorii IBS296 as a new model organism facilitating the study of bacterial pathogenesis both in vitro in cell cultures and in vivo in laboratory mice. We implemented the split NanoLuc luciferase-based translocation assay to study BepD translocation through the VirB/VirD4 T4SS. We found increased effector-translocation into host cells if the bacteria were grown on tryptic soy agar (TSA) plates and experienced a temperature shift immediately before infection. The improved infectivity in vitro was correlating to an upregulation of the VirB/VirD4 T4SS. Using our adapted infection protocols, we showed BepD-dependent immunomodulatory phenotypes in vitro. In mice, the implemented growth conditions enabled infection by a massively reduced inoculum without having an impact on the course of the intra-erythrocytic bacteremia. The established model opens new avenues to study the role of the VirB/VirD4 T4SS and the translocated Bep effectors in vitro and in vivo.

15.
Biology (Basel) ; 11(2)2022 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-35205164

RESUMO

The complex interplay of a pathogen with its virulence and fitness factors, the host's immune response, and the endogenous microbiome determine the course and outcome of gastrointestinal infection. The expansion of a pathogen within the gastrointestinal tract implies an increased risk of developing severe systemic infections, especially in dysbiotic or immunocompromised individuals. We developed a mechanistic computational model that calculates and simulates such scenarios, based on an ordinary differential equation system, to explain the bacterial population dynamics during gastrointestinal infection. For implementing the model and estimating its parameters, oral mouse infection experiments with the enteropathogen, Yersinia enterocolitica (Ye), were carried out. Our model accounts for specific pathogen characteristics and is intended to reflect scenarios where colonization resistance, mediated by the endogenous microbiome, is lacking, or where the immune response is partially impaired. Fitting our data from experimental mouse infections, we can justify our model setup and deduce cues for further model improvement. The model is freely available, in SBML format, from the BioModels Database under the accession number MODEL2002070001.

16.
Lab Invest ; 91(2): 310-24, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20956974

RESUMO

Yersinia enterocolitica is a common cause of acute gastroenteritis. This study aimed to clarify the mechanisms leading to barrier dysfunction and diarrhea. Exposure of human colonic HT-29/B6 cells to Y. enterocolitica resulted in a decrease in transepithelial resistance from 404±23 to 163±21 Ω cm² (P<0.001) in parallel with an increase in mannitol (182 Da) and fluorescein (332 Da) permeability, whereas short circuit current did not change. This effect was time dependent, required the presence of living bacteria, could not be triggered by bacterial supernatants and was not due to Yersinia outer proteins. Concomitantly, Y. enterocolitica induced necrosis as indicated by an increase in lactate dehydrogenase-release, whereas epithelial apoptosis was not upregulated. Local changes in conductivity were detected by conductance scanning, indicating 'leaky regions' within the epithelium that were visualized by biotinylation and confocal microscopy. In these regions, claudin-3 and -4 and, especially claudin-8, were redistributed off the tight junction (TJ) into the cytoplasm. In addition, the expression of claudin-2, -3, -8, -10 and ZO-1 was diminished as quantified by immunoblotting. Moreover, we found claudin-8 to be regulated by the c-Jun N-terminal kinase, the inhibition of which attenuated the Y. enterocolitica-induced decrease in transepithelial resistance and restored claudin-8 protein level. In conclusion, barrier dysfunction in Y. enterocolitica infection is due to circumscribed epithelial TJ protein changes and necrotic cell loss, as a consequence of which leak flux diarrhea and antigen-uptake provoking extraintestinal arthritis may be triggered.


Assuntos
Colo/citologia , Diarreia/microbiologia , Epitélio/patologia , Necrose/patologia , Junções Íntimas/patologia , Yersinia enterocolitica/fisiologia , Cálcio/metabolismo , Linhagem Celular , Claudinas/metabolismo , Colo/microbiologia , Espectroscopia Dielétrica , Impedância Elétrica , Epitélio/microbiologia , Fluoresceína/metabolismo , Humanos , Immunoblotting , Marcação In Situ das Extremidades Cortadas , L-Lactato Desidrogenase/metabolismo , Manitol/metabolismo , Microscopia Eletrônica , Permeabilidade , Reação em Cadeia da Polimerase Via Transcriptase Reversa
17.
PLoS Pathog ; 5(8): e1000551, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19680448

RESUMO

Yersinia enterocolitica (Ye) evades the immune system of the host by injection of Yersinia outer proteins (Yops) via a type three secretion system into host cells. In this study, a reporter system comprising a YopE-beta-lactamase hybrid protein and a fluorescent staining sensitive to beta-lactamase cleavage was used to track Yop injection in cell culture and in an experimental Ye mouse infection model. Experiments with GD25, GD25-beta1A, and HeLa cells demonstrated that beta1-integrins and RhoGTPases play a role for Yop injection. As demonstrated by infection of splenocyte suspensions in vitro, injection of Yops appears to occur randomly into all types of leukocytes. In contrast, upon infection of mice, Yop injection was detected in 13% of F4/80(+), 11% of CD11c(+), 7% of CD49b(+), 5% of Gr1(+) cells, 2.3% of CD19(+), and 2.6% of CD3(+) cells. Taking the different abundance of these cell types in the spleen into account, the highest total number of Yop-injected cells represents B cells, particularly CD19(+)CD21(+)CD23(+) follicular B cells, followed by neutrophils, dendritic cells, and macrophages, suggesting a distinct cellular tropism of Ye. Yop-injected B cells displayed a significantly increased expression of CD69 compared to non-Yop-injected B cells, indicating activation of these cells by Ye. Infection of IFN-gammaR (receptor)- and TNFRp55-deficient mice resulted in increased numbers of Yop-injected spleen cells for yet unknown reasons. The YopE-beta-lactamase hybrid protein reporter system provides new insights into the modulation of host cell and immune responses by Ye Yops.


Assuntos
Proteínas da Membrana Bacteriana Externa/metabolismo , Interações Hospedeiro-Patógeno/fisiologia , Imunidade Inata/imunologia , Leucócitos/metabolismo , Yersiniose/microbiologia , Yersinia enterocolitica/patogenicidade , Animais , Linhagem Celular Transformada , Modelos Animais de Doenças , Feminino , Inativação Gênica , Células HeLa , Humanos , Imunidade Inata/genética , Leucócitos/microbiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Yersiniose/imunologia , Yersiniose/fisiopatologia
18.
Blood ; 113(6): 1326-31, 2009 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-19074007

RESUMO

Oxygen-dependent antimicrobial activity of human polymorphonuclear leukocytes (PMNs) relies on the phagocyte nicotinamide adenine dinucleotide phosphate (NADPH) oxidase to generate oxidants. As the oxidase transfers electrons from NADPH the membrane will depolarize and concomitantly terminate oxidase activity, unless there is charge translocation to compensate. Most experimental data implicate proton channels as the effectors of this charge compensation, although large-conductance Ca2+-activated K+ (BK) channels have been suggested to be essential for normal PMN antimicrobial activity. To test this latter notion, we directly assessed the role of BK channels in phagocyte function, including the NADPH oxidase. PMNs genetically lacking BK channels (BK(-/-)) had normal intracellular and extracellular NADPH oxidase activity in response to both receptor-independent and phagocytic challenges. Furthermore, NADPH oxidase activity of human PMNs and macrophages was normal after treatment with BK channel inhibitors. Although BK channel inhibitors suppressed endotoxin-mediated tumor necrosis factor-alpha secretion by bone marrow-derived macrophages (BMDMs), BMDMs of BK(-/-) and wild-type mice responded identically and exhibited the same ERK, PI3K/Akt, and nuclear factor-kappaB activation. Based on these data, we conclude that the BK channel is not required for NADPH oxidase activity in PMNs or macrophages or for endotoxin-triggered tumor necrosis factor-alpha release and signal transduction BMDMs.


Assuntos
Imunidade Inata , Canais de Potássio Ativados por Cálcio de Condutância Alta/fisiologia , Macrófagos/imunologia , Neutrófilos/imunologia , Animais , Feminino , Citometria de Fluxo , Indóis/farmacologia , Lipopolissacarídeos/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Músculo Liso Vascular/citologia , Músculo Liso Vascular/metabolismo , NADPH Oxidases/metabolismo , NF-kappa B/metabolismo , Fagócitos/fisiologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Explosão Respiratória , Transdução de Sinais , Superóxidos/metabolismo , Artérias da Tíbia/citologia , Artérias da Tíbia/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
19.
Mol Cell Biol ; 27(13): 4603-16, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17485448

RESUMO

Whereas interleukin-10 (IL-10) is an anti-inflammatory cytokine known to regulate macrophage activation, its full mechanism of action remains incompletely defined. In a screen to identify novel IL-10-induced genes, we cloned the mouse ortholog of human ABIN-3 (also termed LIND). ABIN-3 expression was induced selectively by IL-10 in both mouse and human mononuclear phagocytes coordinately undergoing proinflammatory responses. In contrast to the previously characterized ABINs, mouse ABIN-3 was incapable of inhibiting NF-kappaB activation by proinflammatory stimuli. Generation and analysis of ABIN-3-null mice demonstrated that ABIN-3 is unnecessary for the anti-inflammatory effects of IL-10 as well as for proper negative regulation of NF-kappaB. Conversely, human ABIN-3 was capable of inhibiting NF-kappaB activation in response to signaling from Toll-like receptor, IL-1, and tumor necrosis factor. Enforced expression of human ABIN-3 in human monocytic cells suppressed the cytoplasmic degradation of IkappaBalpha, the activation of NF-kappaB, and the induction of proinflammatory genes. Comparative sequence analyses revealed that mouse ABIN-3 lacks a complete ABIN homology domain, which was required for the functional activity of human ABIN-3. ABIN-3 is, thus, an IL-10-induced gene product capable of attenuating NF-kappaB in human macrophages yet is inoperative in mice and represents a basis for species-specific differences in IL-10 actions.


Assuntos
Inflamação/patologia , Interleucina-10/farmacologia , Proteínas/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Sequência de Aminoácidos , Animais , Ativação Enzimática/efeitos dos fármacos , Variação Genética , Humanos , Quinase I-kappa B/metabolismo , Mediadores da Inflamação/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular , Lipopolissacarídeos/farmacologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos , Dados de Sequência Molecular , NF-kappa B/metabolismo , Biossíntese de Proteínas/efeitos dos fármacos , Estrutura Terciária de Proteína , Proteínas/química , Homologia de Sequência de Aminoácidos , Transdução de Sinais/efeitos dos fármacos , Especificidade da Espécie , Células U937
20.
Gastroenterology ; 134(3): 756-67, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18325389

RESUMO

BACKGROUND & AIMS: Hypoxia inducible factor-1 (HIF-1) is the key transcriptional regulator during adaptation to hypoxia. Recent studies provide evidence for HIF-1 activation during bacterial infections. However, molecular details of how bacteria activate HIF-1 remain unclear. Here, we pursued the role of bacterial siderophores in HIF-1 activation during infection with Enterobacteriaceae. METHODS: In vivo, HIF-1 activation and HIF-1-dependent gene induction in Peyer's patches were analyzed after orogastric infection with Yersinia enterocolitica. The course of an orogastric Y enterocolitica infection was determined using mice with a deletion of HIF-1alpha in the intestine. In vitro, the mechanism of HIF-1 activation was analyzed in infections with Y enterocolitica, Salmonella enterica subsp enterica, and Enterobacter aerogenes. RESULTS: Infection of mice with Y enterocolitica led to functional activation of HIF-1 in Peyer's patches. Because mice with deletion of HIF-1alpha in the intestinal epithelium showed a significantly higher susceptibility to orogastric Y enterocolitica infections, bacterial HIF-1 activation appears to represent a host defense mechanism. Additional studies with Y enterocolitica, S enterica subsp enterica, or E aerogenes, and, moreover, application of their siderophores (yersiniabactin, salmochelin, aerobactin) caused a robust, dose-dependent HIF-1 response in human epithelia and endothelia, independent of cellular hypoxia. HIF-1 activation occurs most likely because of inhibition of prolylhydroxylase activity and is abolished upon infection with siderophore uptake deficient bacteria. CONCLUSIONS: Taken together, this study reveals what we believe to be a previously unrecognized role of bacterial siderophores for hypoxia-independent activation of HIF-1 during infection with human pathogenic bacteria.


Assuntos
Enterobacteriaceae/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Nódulos Linfáticos Agregados/metabolismo , Sideróforos/metabolismo , Yersiniose/metabolismo , Animais , Células CACO-2 , Hipóxia Celular , Modelos Animais de Doenças , Células Endoteliais/metabolismo , Células Endoteliais/microbiologia , Enterobacter aerogenes/metabolismo , Enterobacteriaceae/patogenicidade , Células Epiteliais/metabolismo , Células Epiteliais/microbiologia , Feminino , Regulação da Expressão Gênica , Células HeLa , Humanos , Ácidos Hidroxâmicos/metabolismo , Hidroxilação , Subunidade alfa do Fator 1 Induzível por Hipóxia/deficiência , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Oxigênio/metabolismo , Nódulos Linfáticos Agregados/microbiologia , Fenóis/metabolismo , Pró-Colágeno-Prolina Dioxigenase/metabolismo , Salmonella enterica/metabolismo , Tiazóis/metabolismo , Fatores de Tempo , Ativação Transcricional , Regulação para Cima , Yersiniose/genética , Yersiniose/microbiologia , Yersinia enterocolitica/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA