Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Microbes Infect ; 25(7): 105172, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37343664

RESUMO

Human pathogenic bacteria circulating in the bloodstream need to find a way to interact with endothelial cells (ECs) lining the blood vessels to infect and colonise the host. The extracellular matrix (ECM) of ECs might represent an attractive initial target for bacterial interaction, as many bacterial adhesins have reported affinities to ECM proteins, in particular to fibronectin (Fn). Here, we analysed the general role of EC-expressed Fn for bacterial adhesion. For this, we evaluated the expression levels of ECM coding genes in different ECs, revealing that Fn is the highest expressed gene and thereby, it is highly abundant in the ECM environment of ECs. The role of Fn as a mediator in bacterial cell-host adhesion was evaluated in adhesion assays of Acinetobacter baumannii, Bartonella henselae, Borrelia burgdorferi, and Staphylococcus aureus to ECs. The assays demonstrated that bacteria colocalised with Fn fibres, as observed by confocal laser scanning microscopy. Fn removal from the ECM environment (FN1 knockout ECs) diminished bacterial adherence to ECs in both static and dynamic adhesion assays to varying extents, as evaluated via absolute quantification using qPCR. Interactions between adhesins and Fn might represent the crucial step for the adhesion of human-pathogenic Gram-negative and Gram-positive bacteria targeting the ECs as a niche of infection.


Assuntos
Bartonella henselae , Fibronectinas , Humanos , Adesinas Bacterianas/genética , Adesinas Bacterianas/metabolismo , Aderência Bacteriana , Bartonella henselae/genética , Bartonella henselae/metabolismo , Células Endoteliais/microbiologia , Fibronectinas/metabolismo
2.
Microbiol Spectr ; 10(5): e0211722, 2022 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-36165788

RESUMO

Adhesion to host cells is the first and most crucial step in infections with pathogenic Gram-negative bacteria and is often mediated by trimeric autotransporter adhesins (TAAs). Bartonella henselae targets the extracellular matrix glycoprotein fibronectin (Fn) via the Bartonella adhesin A (BadA) attaching the bacteria to the host cell. The TAA BadA is characterized by a highly repetitive passenger domain consisting of 30 neck/stalk domains with various degrees of similarity. To elucidate the motif sequences mediating Fn binding, we generated 10 modified BadA constructs and verified their expression via Western blotting, confocal laser scanning, and electron microscopy. We analyzed their ability to bind human plasma Fn using quantitative whole-cell enzyme-linked immunosorbent assays (ELISAs) and fluorescence microscopy. Polyclonal antibodies targeting a 15-mer amino acid motif sequence proved to reduce Fn binding. We suggest that BadA adheres to Fn in a cumulative effort with quick saturation primarily via unpaired ß-strands appearing in motifs repeatedly present throughout the neck/stalk region. In addition, we demonstrated that the length of truncated BadA constructs correlates with the immunoreactivity of human patient sera. The identification of BadA-Fn binding regions will support the development of new "antiadhesive" compounds inhibiting the initial adherence of B. henselae and other TAA-expressing pathogens to host cells. IMPORTANCE Trimeric autotransporter adhesins (TAAs) are important virulence factors and are widely present in various pathogenic Gram-negative bacteria. TAA-expressing bacteria cause a wide spectrum of human diseases, such as cat scratch disease (Bartonella henselae), enterocolitis (Yersinia enterocolitica), meningitis (Neisseria meningitis), and bloodstream infections (multidrug-resistant Acinetobacter baumannii). TAA-targeted antiadhesive strategies (against, e.g., Bartonella adhesin A [BadA], Yersinia adhesin A [YadA], Neisseria adhesin A [NadA], and Acinetobacter trimeric autotransporter [Ata]) might represent a universal strategy to counteract such bacterial infections. BadA is one of the best characterized TAAs, and because of its high number of (sub)domains, it serves as an attractive adhesin to study the domain-function relationship of TAAs in the infection process. The identification of common binding motifs between TAAs (here, BadA) and their major binding partner (here, fibronectin) provides a basis toward the design of novel "antiadhesive" compounds preventing the initial adherence of Gram-negative bacteria in infections.


Assuntos
Bartonella henselae , Bartonella , Humanos , Bartonella henselae/genética , Bartonella henselae/metabolismo , Fibronectinas/metabolismo , Sistemas de Secreção Tipo V/metabolismo , Aderência Bacteriana , Adesinas Bacterianas/genética , Adesinas Bacterianas/metabolismo , Fatores de Virulência/metabolismo
3.
Microbiol Spectr ; 10(3): e0059822, 2022 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-35435766

RESUMO

Bacterial adhesion to the host is the most decisive step in infections. Trimeric autotransporter adhesins (TAA) are important pathogenicity factors of Gram-negative bacteria. The prototypic TAA Bartonella adhesin A (BadA) from human-pathogenic Bartonella henselae mediates bacterial adherence to endothelial cells (ECs) and extracellular matrix proteins. Here, we determined the interaction between BadA and fibronectin (Fn) to be essential for bacterial host cell adhesion. BadA interactions occur within the heparin-binding domains of Fn. The exact binding sites were revealed by mass spectrometry analysis of chemically cross-linked whole-cell bacteria and Fn. Specific BadA interactions with defined Fn regions represent the molecular basis for bacterial adhesion to ECs and these data were confirmed by BadA-deficient bacteria and CRISPR-Cas knockout Fn host cells. Interactions between TAAs and the extracellular matrix might represent the key step for adherence of human-pathogenic Gram-negative bacteria to the host. IMPORTANCE Deciphering the mechanisms of bacterial host cell adhesion is a clue for preventing infections. We describe the underestimated role that the extracellular matrix protein fibronectin plays in the adhesion of human-pathogenic Bartonella henselae to host cells. Fibronectin-binding is mediated by a trimeric autotransporter adhesin (TAA) also present in many other human-pathogenic Gram-negative bacteria. We demonstrate that both TAA and host-fibronectin contribute significantly to bacterial adhesion, and we present the exact sequence of interacting amino acids from both proteins. Our work shows the domain-specific pattern of interaction between the TAA and fibronectin to adhere to host cells and opens the perspective to fight bacterial infections by inhibiting bacterial adhesion which represents generally the first step in infections.


Assuntos
Bartonella henselae , Bartonella , Adesinas Bacterianas/metabolismo , Aderência Bacteriana , Bartonella henselae/genética , Bartonella henselae/metabolismo , Adesão Celular , Células Endoteliais/metabolismo , Células Endoteliais/microbiologia , Fibronectinas/metabolismo , Humanos , Sistemas de Secreção Tipo V/metabolismo
4.
Front Microbiol ; 13: 838267, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35197960

RESUMO

Bartonella henselae is the causative agent of cat scratch disease and other clinical entities such as endocarditis and bacillary angiomatosis. The life cycle of this pathogen, with alternating host conditions, drives evolutionary and host-specific adaptations. Human, feline, and laboratory adapted B. henselae isolates often display genomic and phenotypic differences that are related to the expression of outer membrane proteins, for example the Bartonella adhesin A (BadA). This modularly-structured trimeric autotransporter adhesin is a major virulence factor of B. henselae and is crucial for the initial binding to the host via the extracellular matrix proteins fibronectin and collagen. By using next-generation long-read sequencing we demonstrate a conserved genome among eight B. henselae isolates and identify a variable genomic badA island with a diversified and highly repetitive badA gene flanked by badA pseudogenes. Two of the eight tested B. henselae strains lack BadA expression because of frameshift mutations. We suggest that active recombination mechanisms, possibly via phase variation (i.e., slipped-strand mispairing and site-specific recombination) within the repetitive badA island facilitate reshuffling of homologous domain arrays. The resulting variations among the different BadA proteins might contribute to host immune evasion and enhance long-term and efficient colonisation in the differing host environments. Considering the role of BadA as a key virulence factor, it remains important to check consistently and regularly for BadA surface expression during experimental infection procedures.

5.
Lancet Microbe ; 2(12): e685-e694, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-35544109

RESUMO

BACKGROUND: Bartonella bacilliformis is the aetiological agent of Carrión's disease, a biphasic and highly lethal illness formerly restricted to the South American Andes that is now spreading to adjacent areas. Reliable serodiagnostic approaches and vaccines are urgently needed. In this study, we aimed to identify immunodominant proteins of B bacilliformis and to establish novel and reliable serodiagnostic tools. METHODS: We used a reverse vaccinology approach in combination with an analysis of heterologous genomic expression libraries to identify immunodominant proteins, on the basis of the genome sequences of B bacilliformis strains KC583 and KC584. Antigens were screened with serum samples collected from Peruvian patients with B bacilliformis infections and from German healthy blood donors without history of travel to South America. We further analysed immunoreactive proteins of B bacilliformis with immunoblotting and line blots. We used selected target proteins to develop a diagnostic ELISA. To assess the performance of this ELISA, we did receiver operating characteristic analyses to assess the area under the curve, cutoff values, sensitivities, and specificities with 95% CIs. FINDINGS: We used serum samples obtained between Dec 23, 1990, and May 5, 2018, from 26 Peruvian patients with B bacilliformis infections and serum samples taken between Aug 28 and Aug 31, 2020, from 96 healthy German blood donors. 21 potentially immunodominant proteins were identified and recombinantly expressed, and their reactivity was assessed with immunoblotting and line blots. Of these 21 antigens, 14 were found to be immunoreactive. By using serum samples of Peruvian patients with Carrión's disease and of healthy German blood donors, we identified three antigens (porin B, autotransporter E, and hypothetical protein B) as suitable immunodominant antigens, and we applied them in a diagnostic ELISA using two different antigen combinations (porin B plus autotransporter E and porin B plus autotransporter E plus hypothetical protein B). For the combination of porin B and autotransporter E, with optical density measured at 450 nm (OD450) cutoff value of 0·29, sensitivity was 80·8% (95% CI 60·7-93·5) and specificity was 94·8% (88·3-98·3) for all Peruvian patient samples. For a combination of porin B, autotransporter E, and hypothetical protein B, with an OD450 cutoff of 0·34, sensitivity was 76·9% (56·4-91·0) and specificity was 93·8% (86·9-97·7) for all Peruvian patient samples. INTERPRETATION: This novel ELISA could represent a useful serodiagnostic tool for future epidemiological studies of B bacilliformis in endemic areas. Additionally, the immunodominant antigens we have identified could provide a first basis for future vaccine development to prevent the highly lethal Carrión's disease. FUNDING: DRUID (Novel Drug Targets against Poverty-Related and Neglected Tropical Infectious Diseases) Initiative and Robert Koch Institute. TRANSLATIONS: For the Spanish and Quechua translations of the abstract see Supplementary Materials section.


Assuntos
Infecções por Bartonella , Bartonella bacilliformis , Infecções por Bartonella/diagnóstico , Bartonella bacilliformis/genética , Humanos , Epitopos Imunodominantes/metabolismo , Porinas/metabolismo , Sistemas de Secreção Tipo V/metabolismo
6.
Microbiol Resour Announc ; 9(34)2020 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-32816983

RESUMO

Bartonella alsatica causes bacteremia in rabbits and, rarely, human infections. Here, we announce the complete and closed genome of B. alsatica IBS 382 (CIP 105477), generated by long-read Pacific Biosciences single-molecule real-time (SMRT) sequencing. The availability of this genome sequence allows future work on understanding the zoonotic potential of this pathogen.

7.
Acta Neuropathol ; 140(2): 183-208, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32529267

RESUMO

Bacterial meningitis is a deadly disease most commonly caused by Streptococcus pneumoniae, leading to severe neurological sequelae including cerebral edema, seizures, stroke, and mortality when untreated. Meningitis is initiated by the transfer of S. pneumoniae from blood to the brain across the blood-cerebrospinal fluid barrier or the blood-brain barrier (BBB). The underlying mechanisms are still poorly understood. Current treatment strategies include adjuvant dexamethasone for inflammation and cerebral edema, followed by antibiotics. The success of dexamethasone is however inconclusive, necessitating new therapies for controlling edema, the primary reason for neurological complications. Since we have previously shown a general activation of hypoxia inducible factor (HIF-1α) in bacterial infections, we hypothesized that HIF-1α, via induction of vascular endothelial growth factor (VEGF) is involved in transmigration of pathogens across the BBB. In human, murine meningitis brain samples, HIF-1α activation was observed by immunohistochemistry. S. pneumoniae infection in brain endothelial cells (EC) resulted in in vitro upregulation of HIF-1α/VEGF (Western blotting/qRT-PCR) associated with increased paracellular permeability (fluorometry, impedance measurements). This was supported by bacterial localization at cell-cell junctions in vitro and in vivo in brain ECs from mouse and humans (confocal, super-resolution, electron microscopy, live-cell imaging). Hematogenously infected mice showed increased permeability, S. pneumoniae deposition in the brain, along with upregulation of genes in the HIF-1α/VEGF pathway (RNA sequencing of brain microvessels). Inhibition of HIF-1α with echinomycin, siRNA in bEnd5 cells or using primary brain ECs from HIF-1α knock-out mice revealed reduced endothelial permeability and transmigration of S. pneumoniae. Therapeutic rescue using the HIF-1α inhibitor echinomycin resulted in increased survival and improvement of BBB function in S. pneumoniae-infected mice. We thus demonstrate paracellular migration of bacteria across BBB and a critical role for HIF-1α/VEGF therein and hence propose targeting this pathway to prevent BBB dysfunction and ensuing brain damage in infections.


Assuntos
Barreira Hematoencefálica , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Meningite Pneumocócica , Streptococcus pneumoniae , Migração Transendotelial e Transepitelial/fisiologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Barreira Hematoencefálica/metabolismo , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Fator A de Crescimento do Endotélio Vascular/metabolismo
8.
J Clin Microbiol ; 56(12)2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30257897

RESUMO

Bartonella henselae causes cat scratch disease and several other clinical entities. Infections with B. henselae are frequently occurring; however, the infection is only rarely diagnosed, mainly due to a lack of knowledge in the medical community. Microscopic immunofluorescence assays (IFA) are widely used for the serodiagnosis of B. henselae infections but are laborious and time-consuming, and interpretation is subjective. An easy and reliable method for the serological diagnosis of B. henselae infections is needed to overcome the shortcomings of the current IFA. Here, we report the development of an ELISA detecting human anti-B. henselae antibodies from serum samples. By separating the water-insoluble fraction of B. henselae Houston-1 via ion-exchange chromatography, 16 subfractions were generated and tested for immunoreactivity via line blotting. One particular fraction (fraction 24) was selected and spotted on ELISA plates using an industrial production platform. By use of well-characterized human sera from the strictly quality-controlled serum library of the German National Consiliary Laboratory for Bartonella infections, the sensitivity of this ELISA was 100% for PCR-proven infections and 76% for clinically suspected infections at a specificity of 93%. This ELISA is therefore a reliable high-throughput method allowing the serodiagnosis of B. henselae infections.


Assuntos
Anticorpos Antibacterianos/sangue , Infecções por Bartonella/diagnóstico , Bartonella henselae/imunologia , Ensaio de Imunoadsorção Enzimática , Testes Sorológicos/métodos , Infecções por Bartonella/sangue , Bartonella henselae/isolamento & purificação , Doença da Arranhadura de Gato/sangue , Doença da Arranhadura de Gato/diagnóstico , Imunofluorescência/normas , Humanos , Sensibilidade e Especificidade
9.
Parasit Vectors ; 10(1): 105, 2017 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-28285589

RESUMO

BACKGROUND: Bartonella henselae is a highly prevalent, vector-borne pathogen. Transmission to humans and animals by ticks is discussed controversially. Here, we present a case report, where eleven Ixodes ricinus ticks all harbouring B. henselae DNA were removed from one single cat. RESULTS: The first feeding tick was tested positive for B. henselae DNA. The cat was also found to be seropositive for anti-B. henselae IgG antibodies (titer 1:640). Bartonella henselae was not cultivatable from cat blood. Ten more feeding ticks removed 7 months later contained also B. henselae DNA. Sequence analysis of the 16SrDNA and the 16S-23S internal transcribed spacer (ITS) region revealed 100% sequence homology between all ticks. Bartonella adhesin A (badA) and VirB/VirD4 type IV secretion system (virB) DNA were also detected in all ticks. CONCLUSIONS: Our results indicate that cats may serve as a reservoir for adult ticks to acquire B. henselae. Whether this observation implies an increased threat for human and animal health needs to be resolved.


Assuntos
Vetores Aracnídeos/microbiologia , Infecções por Bartonella/veterinária , Bartonella henselae/isolamento & purificação , Doenças do Gato/parasitologia , Ixodes/microbiologia , Infestações por Carrapato/veterinária , Animais , Infecções por Bartonella/microbiologia , Bartonella henselae/genética , Doenças do Gato/microbiologia , Gatos , DNA Bacteriano/genética , DNA Bacteriano/isolamento & purificação , Reservatórios de Doenças , Imunoglobulina G/sangue , Reação em Cadeia da Polimerase
10.
Infect Immun ; 84(3): 711-22, 2015 Dec 28.
Artigo em Inglês | MEDLINE | ID: mdl-26712205

RESUMO

Bacterial adherence determines the virulence of many human-pathogenic bacteria. Experimental approaches elucidating this early infection event in greater detail have been performed using mainly methods of cellular microbiology. However, in vitro infections of cell monolayers reflect the in vivo situation only partially, and animal infection models are not available for many human-pathogenic bacteria. Therefore, ex vivo infection of human organs might represent an attractive method to overcome these limitations. We infected whole human umbilical cords ex vivo with Bartonella henselae or Acinetobacter baumannii under dynamic flow conditions mimicking the in vivo infection situation of human endothelium. For this purpose, methods for quantifying endothelium-adherent wild-type and trimeric autotransporter adhesin (TAA)-deficient bacteria were set up. Data revealed that (i) A. baumannii binds in a TAA-dependent manner to endothelial cells, (ii) this organ infection model led to highly reproducible adherence rates, and furthermore, (iii) this model allowed to dissect the biological function of TAAs in the natural course of human infections. These findings indicate that infection models using ex vivo human tissue samples ("organ microbiology") might be a valuable tool in analyzing bacterial pathogenicity with the capacity to replace animal infection models at least partially.


Assuntos
Infecções por Acinetobacter/microbiologia , Acinetobacter baumannii/fisiologia , Angiomatose Bacilar/microbiologia , Aderência Bacteriana , Bartonella henselae/fisiologia , Células Endoteliais/microbiologia , Cordão Umbilical/microbiologia , Acinetobacter baumannii/genética , Animais , Bartonella henselae/genética , Humanos , Técnicas In Vitro
11.
Cell Microbiol ; 17(10): 1447-63, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25857345

RESUMO

The contribution of myeloid cells to tumour microenvironments is a decisive factor in cancer progression. Tumour-associated macrophages (TAMs) mediate tumour invasion and angiogenesis through matrix remodelling, immune modulation and release of pro-angiogenic cytokines. Nothing is known about how pathogenic bacteria affect myeloid cells in these processes. Here we show that Bartonella henselae, a bacterial pathogen causing vasculoproliferative diseases (bacillary angiomatosis), reprogrammes human myeloid angiogenic cells (MACs), a pro-angiogenic subset of circulating progenitor cells, towards a TAM-like phenotype with increased pro-angiogenic capacity. B. henselae infection resulted in inhibition of cell death, activation of angiogenic cellular programmes and induction of M2 macrophage polarization. MACs infected with B. henselae incorporated into endothelial sprouts and increased angiogenic growth. Infected MACs developed a vascular mimicry phenotype in vitro, and expression of B. henselae adhesin A was essential in inducing these angiogenic effects. Secretome analysis revealed that increased pro-angiogenic activities were associated with the creation of a tumour-like microenvironment dominated by angiogenic inflammatory cytokines and matrix remodelling compounds. Our results demonstrate that manipulation of myeloid cells by pathogenic bacteria can contribute to microenvironmental regulation of pathological tissue growth and suggest parallels underlying both bacterial infections and cancer.


Assuntos
Bartonella henselae/fisiologia , Interações Hospedeiro-Patógeno , Células Progenitoras Mieloides/fisiologia , Neovascularização Patológica , Diferenciação Celular , Células Endoteliais/microbiologia , Células Endoteliais/fisiologia , Humanos , Macrófagos/microbiologia , Macrófagos/fisiologia
12.
J Bacteriol ; 196(12): 2155-65, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24682330

RESUMO

Human-pathogenic Bartonella henselae causes cat scratch disease and vasculoproliferative disorders. An important pathogenicity factor of B. henselae is the trimeric autotransporter adhesin (TAA) Bartonella adhesin A (BadA), which is modularly constructed, consisting of a head, a long and repetitive neck-stalk module, and a membrane anchor. BadA is involved in bacterial autoagglutination, binding to extracellular matrix proteins and host cells, and in proangiogenic reprogramming. The slow growth of B. henselae and limited tools for genetic manipulation are obstacles for detailed examination of BadA and its domains. Here, we established a recombinant expression system for BadA mutants in Escherichia coli allowing functional analysis of particular BadA domains. Using a BadA mutant lacking 21 neck-stalk repeats (BadA HN23), the BadA HN23 signal sequence was exchanged with that of E. coli OmpA, and the BadA membrane anchor was additionally replaced with that of Yersinia adhesin A (YadA). Constructs were cloned in E. coli, and hybrid protein expression was detected by immunoblotting, fluorescence microscopy, and flow cytometry. Functional analysis revealed that BadA hybrid proteins mediate autoagglutination and binding to collagen and endothelial cells. In vivo, expression of this BadA construct correlated with higher pathogenicity of E. coli in a Galleria mellonella infection model.


Assuntos
Adesinas Bacterianas/metabolismo , Bartonella henselae/metabolismo , Escherichia coli/metabolismo , Regulação Bacteriana da Expressão Gênica/fisiologia , Adesinas Bacterianas/genética , Animais , Aderência Bacteriana/fisiologia , Clonagem Molecular , Colágeno/química , Colágeno/metabolismo , Escherichia coli/patogenicidade , Larva/microbiologia , Mariposas/microbiologia , Mutação , Estrutura Terciária de Proteína , Proteínas Recombinantes
13.
J Leukoc Biol ; 93(6): 921-32, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23543768

RESUMO

Leishmania are intracellular parasites adapted to surviving in macrophages, whose primary function is elimination of invading pathogens. Leishmania entry into host cells is receptor-mediated. These parasites are able to engage multiple host cell-surface receptors, including MR, TLRs, CR3, and FcγRs. Here, we investigated the role of CR3 and FcγR engagement on the maturation of Leishmania-containing phagosomes using CD11b-/- and FcγR-/- macrophages, and assessing EEA1 and lysosome-associated proteins is necessary for the phagosome maturation delay, characteristic of Leishmania infection. Leishmania-containing phagosomes do not fuse with lyosomes until 5 h postinfection in WT mice. Phagolysosome fusion occurs by 1 h in CD11b and FcγR common chain KO macrophages, although receptor deficiency does not influence Leishmania entry or viability. We also investigated the influence of serum components and their effects on phagosome maturation progression. Opsonization with normal mouse serum, complement-deficient serum, or serum from Leishmania-infected mice all influenced phagosome maturation progression. Our results indicate that opsonophagocytosis influences phagosomal trafficking of Leishmania without altering the intracellular fate.


Assuntos
Leishmaniose/imunologia , Antígeno de Macrófago 1/imunologia , Fagossomos/imunologia , Receptores de IgG/imunologia , Animais , Feminino , Citometria de Fluxo , Imunofluorescência , Leishmania/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
14.
Gastroenterology ; 136(7): 2247-57, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19272387

RESUMO

BACKGROUND & AIMS: Helicobacter pylori infects half of the world's population, thereby causing significant human morbidity and mortality. The mechanisms by which professional antigen-presenting cells recognize the microbe are poorly understood. METHODS: Using dendritic cells (DCs) from TRIF, MyD88, TLR 2/4/7/9(-/-), and multiple double/triple/quadruple mutant mice, we characterized receptors and pathways mediating innate immune recognition of H pylori. RESULTS: We identified a MyD88-dependent component of the DC activation program, which was induced by surface TLRs, with TLR2 and to a minor extent also TLR4 being the exclusive surface receptors recognizing H pylori. A second MyD88-dependent component could be blocked in TLR2/4(-/-) DCs by inhibitors of endosomal acidification and depended on intracellular TLRs. We identified TLR9-mediated recognition of H pylori DNA as a principal H pylori-induced intracellular TLR pathway and further showed that H pylori RNA induces proinflammatory cytokines in a TLR-dependent manner. Microarray analysis showed complementary, redundant, and synergistic interactions between TLRs and additionally revealed gene expression patterns specific for individual TLRs, including a TLR2-dependent anti-inflammatory signature. A third component of the DC activation program was primarily composed of type I interferon-stimulated genes. This response was MyD88 and TRIF independent but was inducible by RIG-I-dependent recognition of H pylori RNA. CONCLUSIONS: These results provide novel comprehensive insights into the mechanisms of H pylori recognition by DCs. Understanding these processes provides a basis for the rational design of new vaccination strategies.


Assuntos
Helicobacter pylori/imunologia , Imunidade Inata/fisiologia , Receptores de Reconhecimento de Padrão/imunologia , Transdução de Sinais/fisiologia , Animais , Células Apresentadoras de Antígenos/imunologia , Células Apresentadoras de Antígenos/metabolismo , Células Cultivadas , DNA Bacteriano/metabolismo , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Infecções por Helicobacter/diagnóstico , Helicobacter pylori/genética , Imunidade Inata/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Animais , RNA Bacteriano/metabolismo , Receptores de Reconhecimento de Padrão/genética , Receptores de Reconhecimento de Padrão/metabolismo , Sensibilidade e Especificidade , Transdução de Sinais/genética , Receptores Toll-Like/deficiência
15.
Gastroenterology ; 133(1): 150-163.e3, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17631139

RESUMO

BACKGROUND & AIMS: Recognition of infection leads to induction of adaptive immunity through activation of antigen-presenting cells (APCs). Among APCs, dendritic cells (DCs) have the unique capacity to deliver antigens from the periphery to T cells in secondary lymphoid organs. METHODS: We analyzed molecular mechanisms of the Helicobacter pylori-induced APC activation in vitro and investigated the influence of Myd88 signaling on the phenotype of adaptive immunity to H pylori in a murine infection model. RESULTS: The adaptor protein Myd88 mediates Toll-like receptor (TLR), interleukin (IL)-1, and IL-18 signaling. DCs from wild-type, IL-1R(-/-), and IL-18(-/-) mice responded to H pylori with secretion of proinflammatory cytokines and up-regulation of major histocompatibility complex II and costimulatory molecules. In Myd88(-/-) DCs these processes were impaired profoundly, showing that TLR-dependent H pylori-sensing affects DC activation. Analysis of the H pylori-specific DC transcriptome revealed that large parts of the bacteria-induced transcriptional changes depended on Myd88 signaling, comprising numerous genes involved in crucial steps of immune regulation, such as DC maturation/differentiation, antigen uptake/presentation, and effector cell recruitment/activation. The impaired ability of Myd88(-/-) DCs, B cells, and macrophages to mount a proinflammatory response to H pylori in vitro was reflected in vivo by reduced gastric inflammation and increased bacterial colonization in Myd88-deficient mice. Furthermore, Helicobacter-specific IgG2c/IgG1 ratios were reduced in Myd88(-/-) animals, suggesting the involvement of the Myd88-dependent pathway in the instruction of adaptive immunity toward a T helper cell type 1 phenotype. CONCLUSIONS: A principal pathway by which DCs sense H pylori and become activated is the TLR-dependent signaling cascade. In vivo, Myd88 signaling affects adaptive immunity to the bacterium.


Assuntos
Células Apresentadoras de Antígenos/metabolismo , Gastrite/imunologia , Infecções por Helicobacter/imunologia , Helicobacter pylori/imunologia , Receptores Toll-Like/metabolismo , Animais , Células Apresentadoras de Antígenos/imunologia , Células Apresentadoras de Antígenos/microbiologia , Linfócitos B/imunologia , Linfócitos B/metabolismo , Linfócitos B/microbiologia , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Células Dendríticas/microbiologia , Gastrite/microbiologia , Perfilação da Expressão Gênica , Imunoglobulina G/sangue , Macrófagos Peritoneais/imunologia , Macrófagos Peritoneais/metabolismo , Macrófagos Peritoneais/microbiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Fator 88 de Diferenciação Mieloide/genética , Transdução de Sinais/imunologia , Células Th1/imunologia , Células Th1/metabolismo , Células Th1/microbiologia , Células Th2/imunologia , Células Th2/metabolismo , Células Th2/microbiologia , Receptores Toll-Like/imunologia , Ativação Transcricional/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...