Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Bioengineering (Basel) ; 9(10)2022 Oct 17.
Artigo em Inglês | MEDLINE | ID: mdl-36290534

RESUMO

Bacterial adhesion to dental implants is the onset for the development of pathological biofilms. Reliable characterization of this initial process is the basis towards the development of anti-biofilm strategies. In the present study, single-cell force spectroscopy (SCFS), by means of an atomic force microscope connected to a microfluidic pressure control system (FluidFM), was used to comparably measure adhesion forces of different oral bacteria within a similar experimental setup to the common implant material titanium. The bacteria selected belong to different ecological niches in oral biofilms: the commensal pioneers Streptococcus oralis and Actinomyces naeslundii; secondary colonizer Veillonella dispar; and the late colonizing pathogens Porphyromonas gingivalis as well as fimbriated and non-fimbriated Aggregatibacter actinomycetemcomitans. The results showed highest values for early colonizing pioneer species, strengthening the link between adhesion forces and bacteria's role in oral biofilm development. Additionally, the correlation between biophysical cellular characteristics and SCFS results across species was analyzed. Here, distinct correlations between electrostatically driven maximum adhesion force, bacterial surface elasticity and surface charge as well as single-molecule attachment points, stretching capability and metabolic activity, could be identified. Therefore, this study provides a step towards the detailed understanding of oral bacteria initial adhesion and could support the development of infection-resistant implant materials in future.

2.
BMC Oral Health ; 21(1): 313, 2021 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-34144677

RESUMO

BACKGROUND: Peri-implant mucositis and peri-implantitis are highly prevalent biofilm-associated diseases affecting the tissues surrounding dental implants. As antibiotic treatment is ineffective to fully cure biofilm mediated infections, antimicrobial modifications of implants to reduce or prevent bacterial colonization are called for. Preclinical in vivo evaluation of the functionality of new or modified implant materials concerning bacterial colonization and peri-implant health is needed to allow progress in this research field. For this purpose reliable animal models are needed. METHODS: Custom made endosseous dental implants were installed in female Sprague Dawley rats following a newly established three-step implantation procedure. After healing of the bone and soft tissue, the animals were assigned to two groups. Group A received a continuous antibiotic treatment for 7 weeks, while group B was repeatedly orally inoculated with human-derived strains of Streptococcus oralis, Fusobacterium nucleatum and Porphyromonas gingivalis for six weeks, followed by 1 week without inoculation. At the end of the experiment, implantation sites were clinically assessed and biofilm colonization was quantified via confocal laser scanning microscopy. Biofilm samples were tested for presence of the administered bacteria via PCR analysis. RESULTS: The inner part of the custom made implant screw could be identified as a site of reliable biofilm formation in vivo. S. oralis and F. nucleatum were detectable only in the biofilm samples from group B animals. P. gingivalis was not detectable in samples from either group. Quantification of the biofilm volume on the implant material revealed no statistically significant differences between the treatment groups. Clinical inspection of implants in group B animals showed signs of mild to moderate peri-implant mucositis (4 out of 6) whereas the mucosa of group A animals appeared healthy (8/8). The difference in the mucosa health status between the treatment groups was statistically significant (p = 0.015). CONCLUSIONS: We developed a new rodent model for the preclinical evaluation of dental implant materials with a special focus on the early biofilm colonization including human-derived oral bacteria. Reliable biofilm quantification on the implant surface and the symptoms of peri-implant mucositis of the bacterially inoculated animals will serve as a readout for experimental evaluation of biofilm-reducing modifications of implant materials.


Assuntos
Implantes Dentários , Peri-Implantite , Animais , Biofilmes , Implantes Dentários/efeitos adversos , Feminino , Porphyromonas gingivalis , Ratos , Ratos Sprague-Dawley
3.
Nucleic Acids Res ; 48(18): 10397-10412, 2020 10 09.
Artigo em Inglês | MEDLINE | ID: mdl-32946572

RESUMO

The RNA helicase RIG-I plays a key role in sensing pathogen-derived RNA. Double-stranded RNA structures bearing 5'-tri- or diphosphates are commonly referred to as activating RIG-I ligands. However, endogenous RNA fragments generated during viral infection via RNase L also activate RIG-I. Of note, RNase-digested RNA fragments bear a 5'-hydroxyl group and a 2',3'-cyclic phosphate. How endogenous RNA fragments activate RIG-I despite the lack of 5'-phosphorylation has not been elucidated. Here we describe an endogenous RIG-I ligand (eRL) that is derived from the internal transcribed spacer 2 region (ITS2) of the 45S ribosomal RNA after partial RNase A digestion in vitro, RNase A protein transfection or RNase L activation. The immunostimulatory property of the eRL is dependent on 2',3'-cyclic phosphate and its sequence is characterized by a G-quadruplex containing sequence motif mediating guanosine-5'-triphosphate (GTP) binding. In summary, RNase generated self-RNA fragments with 2',3'-cyclic phosphate function as nucleotide-5'-triphosphate binding aptamers activating RIG-I.


Assuntos
Proteína DEAD-box 58/genética , RNA Helicases/genética , RNA Ribossômico/genética , RNA/genética , Guanosina Trifosfato/genética , Humanos , Ligantes , Fosfatos/metabolismo , RNA/química , RNA Helicases/metabolismo , Receptores Imunológicos , Ribonucleases/genética
4.
Clin Oral Implants Res ; 31(9): 865-880, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32583509

RESUMO

OBJECTIVES: The aim of this study was to evaluate volume, vitality and diversity of biofilms on the abutment materials zirconia and titanium as a function of time using an in vivo model for the biofilm formation. MATERIALS AND METHODS: The development of biofilms on zirconia and titanium grade 4 test specimens in the human oral cavity over time was analysed. After pretreatment, a total of 96 titanium and 96 zirconia discs were fixed on 12 composite splints, which were worn by 12 volunteers. After 6 hr, 24 hr, 3 days and 5 days, biofilms on 48 specimens of each material were analysed with confocal laser scanning microscopy (CLSM). The microbiota composition on the other 48 test specimens was examined using full-length 16S sequence analysis. Statistical analysis was performed by SPSS and R, and level of significance was set at 0.05. RESULTS: Confocal laser scanning microscopy analysis of the biofilms revealed significant changes in volume over time on zirconia and titanium. The material did not significantly influence the volume or live/dead ratio at the individual time points. The composition of the microbiome was influenced by the age of the biofilm, but not by the material of the test specimen. The most frequently found bacteria were Streptococcus spp., followed by Neisseria spp., Rothia spp., Haemophilus spp., Gemella spp. and Abiotrophia spp. CONCLUSIONS: On both materials, the quantity and diversity of the microbiome increased over time. Apart from a slight difference in Veillonella abundance at one time point, there were no significant differences between zirconia and titanium.


Assuntos
Titânio , Zircônio , Biofilmes , Humanos , Propriedades de Superfície , Tempo
5.
Acta Physiol (Oxf) ; 230(2): e13498, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32415725

RESUMO

AIM: SLC26A3 (DRA) mediates the absorption of luminal Cl- in exchange for HCO3- in the distal intestine. Its expression is lost in congenital chloride diarrhoea (CLD) and strongly decreased in the presence of intestinal inflammation. To characterize the consequences of a loss of Slc26a3 beyond disturbed electrolyte transport, colonic mucus synthesis, surface accumulation and composition, pH microclimate, microbiome composition and development of inflammation was studied in slc26a3-/- mice. METHODS: The epithelial surface pH microclimate and the surface mucus accumulation in vivo was assessed by two photon microscopy in exteriorized mid colon of anaesthetized slc26a3-/- and wt littermates. Mucus synthesis, composition and inflammatory markers were studied by qPCR and immunohistochemistry and microbiome composition by 16S rRNA sequencing. RESULTS: Colonic pH microclimate was significantly more acidic in slc26a3-/- and to a lesser extent in cftr-/- than in wt mice. Goblet cell thecae per crypt were decreased in slc26a3-/- and increased in cftr-/- colon. Mucus accumulation in vivo was reduced, but much less so than in cftr-/- colon, which is possibly related to the different colonic fluid balance. Slc26a3-/- colonic luminal microbiome displayed strong decrease in diversity. These alterations preceded and maybe causally related to increased mucosal TNFα mRNA expression levels and leucocyte infiltration in the mid-distal colon of slc26a3-/- but not of cftr-/- mice. CONCLUSIONS: These findings may explain the strong increase in the susceptibility of slc26a3-/- mice to DSS damage, and offer insight into the mechanisms leading to an increased incidence of intestinal inflammation in CLD patients.


Assuntos
Antiporters , Microbiota , Animais , Antiportadores de Cloreto-Bicarbonato , Colo , Humanos , Concentração de Íons de Hidrogênio , Mucosa Intestinal , Camundongos , Microclima , Mucinas , RNA Ribossômico 16S , Transportadores de Sulfato/genética
6.
ISME J ; 13(10): 2500-2522, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31201356

RESUMO

Aggregatibacter and Haemophilus species are relevant human commensals and opportunistic pathogens. Consequently, their bacteriophages may have significant impact on human microbial ecology and pathologies. Our aim was to reveal the prevalence and diversity of bacteriophages infecting Aggregatibacter and Haemophilus species that colonize the human body. Genome mining with comparative genomics, screening of clinical isolates, and profiling of metagenomes allowed characterization of 346 phages grouped in 52 clusters and 18 superclusters. Less than 10% of the identified phage clusters were represented by previously characterized phages. Prophage diversity patterns varied significantly for different phage types, host clades, and environmental niches. A more diverse phage community lysogenizes Haemophilus influenzae and Haemophilus parainfluenzae strains than Aggregatibacter actinomycetemcomitans and "Haemophilus ducreyi". Co-infections occurred more often in "H. ducreyi". Phages from Aggregatibacter actinomycetemcomitans preferably lysogenized strains of specific serotype. Prophage patterns shared by subspecies clades of different bacterial species suggest similar ecoevolutionary drivers. Changes in frequencies of DNA uptake signal sequences and guanine-cytosine content reflect phage-host long-term coevolution. Aggregatibacter and Haemophilus phages were prevalent at multiple oral sites. Together, these findings should help exploring the ecoevolutionary forces shaping virus-host interactions in the human microbiome. Putative lytic phages, especially phiKZ-like, may provide new therapeutic options.


Assuntos
Aggregatibacter/virologia , Bacteriófagos/fisiologia , Haemophilus/virologia , Aggregatibacter/classificação , Bacteriófagos/classificação , Bacteriófagos/genética , Bacteriófagos/isolamento & purificação , Composição de Bases , Biodiversidade , Genoma Viral , Genômica , Haemophilus/classificação , Especificidade de Hospedeiro , Humanos , Lisogenia , Metagenoma , Filogenia , Prófagos/classificação , Prófagos/genética , Prófagos/isolamento & purificação , Prófagos/fisiologia
7.
ACS Appl Mater Interfaces ; 11(26): 23026-23038, 2019 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-31173692

RESUMO

To combat implant-associated infections, there is a need for novel materials which effectively inhibit bacterial biofilm formation. In the present study, the antiadhesive properties of titanium surface functionalization based on the "slippery liquid-infused porous surfaces" (SLIPS) principle were demonstrated and the underlying mechanism was analyzed. The immobilized liquid layer was stable over 13 days of continuous flow in an oral flow chamber system. With increasing flow rates, the surface exhibited a significant reduction in attached biofilm of both the oral initial colonizer  Streptococcus oralis and an oral multispecies biofilm composed of S. oralis, Actinomyces naeslundii, Veillonella dispar, and Porphyromonas gingivalis. Using single cell force spectroscopy, reduced S. oralis adhesion forces on the lubricant layer could be measured. Gene expression patterns in biofilms on SLIPS, on control surfaces, and expression patterns of planktonic cultures were also compared. For this purpose, the genome of S. oralis strain ATCC 9811 was sequenced using PacBio Sequel technology. Even though biofilm cells showed clear changes in gene expression compared to planktonic cells, no differences could be detected between bacteria on SLIPS and on control surfaces. Therefore, it can be concluded that the ability of liquid-infused titanium to repel S. oralis biofilms is mainly due to weakened bacterial adhesion to the underlying liquid interface.


Assuntos
Aderência Bacteriana/efeitos dos fármacos , Biofilmes/efeitos dos fármacos , Análise de Célula Única/métodos , Titânio/química , Actinomyces/efeitos dos fármacos , Actinomyces/patogenicidade , Biofilmes/crescimento & desenvolvimento , Regulação Bacteriana da Expressão Gênica/efeitos dos fármacos , Humanos , Porphyromonas gingivalis/efeitos dos fármacos , Porphyromonas gingivalis/patogenicidade , Análise Espectral , Streptococcus oralis/química , Streptococcus oralis/patogenicidade , Propriedades de Superfície , Titânio/farmacologia , Veillonella/efeitos dos fármacos , Veillonella/patogenicidade
8.
Gastroenterology ; 154(3): 612-623.e7, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29066327

RESUMO

BACKGROUND & AIMS: Helicobacter pylori is remarkable for its genetic variation; yet, little is known about its genetic changes during early stages of human infection, as the bacteria adapt to their new environment. We analyzed genome and methylome variations in a fully virulent strain of H pylori during experimental infection. METHODS: We performed a randomized Phase I/II, observer-blind, placebo-controlled study of 12 healthy, H pylori-negative adults in Germany from October 2008 through March 2010. The volunteers were given a prophylactic vaccine candidate (n = 7) or placebo (n = 5) and then challenged with H pylori strain BCM-300. Biopsy samples were collected and H pylori were isolated. Genomes of the challenge strain and 12 reisolates, obtained 12 weeks after (or in 1 case, 62 weeks after) infection were sequenced by single-molecule, real-time technology, which, in parallel, permitted determination of genome-wide methylation patterns for all strains. Functional effects of genetic changes observed in H pylori strains during human infection were assessed by measuring release of interleukin 8 from AGS cells (to detect cag pathogenicity island function), neutral red uptake (to detect vacuolating cytotoxin activity), and adhesion assays. RESULTS: The observed mutation rate was in agreement with rates previously determined from patients with chronic H pylori infections, without evidence of a mutation burst. A loss of cag pathogenicity island function was observed in 3 reisolates. In addition, 3 reisolates from the vaccine group acquired mutations in the vacuolating cytotoxin gene vacA, resulting in loss of vacuolization activity. We observed interstrain variation in methylomes due to phase variation in genes encoding methyltransferases. CONCLUSIONS: We analyzed adaptation of a fully virulent strain of H pylori to 12 different volunteers to obtain a robust estimate of the frequency of genetic and epigenetic changes in the absence of interstrain recombination. Our findings indicate that the large amount of genetic variation in H pylori poses a challenge to vaccine development. ClinicalTrials.gov no: NCT00736476.


Assuntos
Metilação de DNA , Epigênese Genética , Genoma Bacteriano , Ilhas Genômicas , Infecções por Helicobacter/microbiologia , Helicobacter pylori/genética , Antígenos de Bactérias/administração & dosagem , Antígenos de Bactérias/genética , Antígenos de Bactérias/imunologia , Aderência Bacteriana , Proteínas de Bactérias/administração & dosagem , Proteínas de Bactérias/genética , Proteínas de Bactérias/imunologia , Vacinas Bacterianas/administração & dosagem , Vacinas Bacterianas/genética , Vacinas Bacterianas/imunologia , Biópsia , Regulação Bacteriana da Expressão Gênica , Genótipo , Alemanha , Infecções por Helicobacter/diagnóstico , Infecções por Helicobacter/imunologia , Infecções por Helicobacter/prevenção & controle , Helicobacter pylori/imunologia , Helicobacter pylori/patogenicidade , Interações Hospedeiro-Patógeno , Humanos , Interleucina-8/imunologia , Interleucina-8/metabolismo , Mutação , Fenótipo , Polimorfismo de Nucleotídeo Único , Fatores de Tempo , Virulência
9.
Nat Commun ; 7: 11995, 2016 06 22.
Artigo em Inglês | MEDLINE | ID: mdl-27329939

RESUMO

Recombination plays a dominant role in the evolution of the bacterial pathogen Helicobacter pylori, but its dynamics remain incompletely understood. Here we use an in vitro transformation system combined with genome sequencing to study chromosomal integration patterns after natural transformation. A single transformation cycle results in up to 21 imports, and repeated transformations generate a maximum of 92 imports (8% sequence replacement). Import lengths show a bimodal distribution with averages of 28 and 1,645 bp. Reanalysis of paired H. pylori genomes from chronically infected people demonstrates the same bimodal import pattern in vivo. Restriction endonucleases (REases) of the recipient bacteria fail to inhibit integration of homeologous DNA, independently of methylation. In contrast, REases limit the import of heterologous DNA. We conclude that restriction-modification systems inhibit the genomic integration of novel sequences, while they pose no barrier to homeologous recombination, which reconciles the observed stability of the H. pylori gene content and its highly recombinational population structure.


Assuntos
Cromossomos Bacterianos , Infecções por Helicobacter/microbiologia , Helicobacter pylori/genética , Helicobacter pylori/metabolismo , DNA/genética , Enzimas de Restrição do DNA/metabolismo , Enzimas de Restrição-Modificação do DNA/genética , DNA Bacteriano/genética , Escherichia coli/genética , Variação Genética , Genoma Bacteriano , Genômica , Humanos , Mutagênese , Polimorfismo de Nucleotídeo Único , Recombinação Genética , Análise de Sequência de DNA
10.
mSphere ; 1(1)2016.
Artigo em Inglês | MEDLINE | ID: mdl-27303676

RESUMO

The human diarrheal pathogens Campylobacter jejuni and Campylobacter coli interfere with host innate immune signaling by different means, and their flagellins, FlaA and FlaB, have a low intrinsic property to activate the innate immune receptor Toll-like receptor 5 (TLR5). We have investigated here the hypothesis that the unusual secreted, flagellin-like molecule FlaC present in C. jejuni, C. coli, and other Campylobacterales might activate cells via TLR5 and interact with TLR5. FlaC shows striking sequence identity in its D1 domains to TLR5-activating flagellins of other bacteria, such as Salmonella, but not to nonstimulating Campylobacter flagellins. We overexpressed and purified FlaC and tested its immunostimulatory properties on cells of human and chicken origin. Treatment of cells with highly purified FlaC resulted in p38 activation. FlaC directly interacted with TLR5. Preincubation with FlaC decreased the responsiveness of chicken and human macrophage-like cells toward the bacterial TLR4 agonist lipopolysaccharide (LPS), suggesting that FlaC mediates cross-tolerance. C. jejuni flaC mutants induced an increase of cell responses in comparison to those of the wild type, which was suppressed by genetic complementation. Supplementing excess purified FlaC likewise reduced the cellular response to C. jejuni. In vivo, the administration of ultrapure FlaC led to a decrease in cecal interleukin 1ß (IL-1ß) expression and a significant change of the cecal microbiota in chickens. We propose that Campylobacter spp. have evolved a novel type of secreted immunostimulatory flagellin-like effector in order to specifically modulate host responses, for example toward other pattern recognition receptor (PRR) ligands, such as LPS. IMPORTANCE Flagellins not only are important for bacterial motility but are major bacterial proteins that can modulate host responses via Toll-like receptor 5 (TLR5) or other pattern recognition receptors. Campylobacterales colonizing the intestinal tracts of different host species harbor a gene coding for an unusual flagellin, FlaC, that is not involved in motility but is secreted and possesses a chimeric amino acid sequence composed of TLR5-activating and non-TLR5-activating flagellin sequences. Campylobacter jejuni FlaC activates cells to increase in cytokine expression in chicken and human cells, promotes cross-tolerance to TLR4 ligands, and alters chicken cecal microbiota. We propose that FlaC is a secreted effector flagellin that has specifically evolved to modulate the immune response in the intestinal tract in the presence of the resident microbiota and may contribute to bacterial persistence. The results also strengthen the role of the flagellar type III apparatus as a functional secretion system for bacterial effector proteins.

11.
Sci Rep ; 6: 18594, 2016 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-26729566

RESUMO

Inhabitants of Túquerres in the Colombian Andes have a 25-fold higher risk of gastric cancer than inhabitants of the coastal town Tumaco, despite similar H. pylori prevalences. The gastric microbiota was recently shown in animal models to accelerate the development of H. pylori-induced precancerous lesions. 20 individuals from each town, matched for age and sex, were selected, and gastric microbiota analyses were performed by deep sequencing of amplified 16S rDNA. In parallel, analyses of H. pylori status, carriage of the cag pathogenicity island and assignment of H. pylori to phylogeographic groups were performed to test for correlations between H. pylori strain properties and microbiota composition. The gastric microbiota composition was highly variable between individuals, but showed a significant correlation with the town of origin. Multiple OTUs were detected exclusively in either Tumaco or Túquerres. Two operational taxonomic units (OTUs), Leptotrichia wadei and a Veillonella sp., were significantly more abundant in Túquerres, and 16 OTUs, including a Staphylococcus sp. were significantly more abundant in Tumaco. There was no significant correlation of H. pylori phylogeographic population or carriage of the cagPAI with microbiota composition. From these data, testable hypotheses can be generated and examined in suitable animal models and prospective clinical trials.


Assuntos
Microbiota , Neoplasias Gástricas/epidemiologia , Neoplasias Gástricas/etiologia , Estômago/microbiologia , Adulto , Colômbia/epidemiologia , Feminino , Infecções por Helicobacter/complicações , Infecções por Helicobacter/microbiologia , Helicobacter pylori/genética , Humanos , Masculino , Metagenoma , Metagenômica , Pessoa de Meia-Idade , Risco , Neoplasias Gástricas/diagnóstico
12.
Nat Immunol ; 16(7): 698-707, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26006013

RESUMO

The epithelium is the main entry point for many viruses, but the processes that protect barrier surfaces against viral infections are incompletely understood. Here we identified interleukin 22 (IL-22) produced by innate lymphoid cell group 3 (ILC3) as an amplifier of signaling via interferon-λ (IFN-λ), a synergism needed to curtail the replication of rotavirus, the leading cause of childhood gastroenteritis. Cooperation between the receptor for IL-22 and the receptor for IFN-λ, both of which were 'preferentially' expressed by intestinal epithelial cells (IECs), was required for optimal activation of the transcription factor STAT1 and expression of interferon-stimulated genes (ISGs). These data suggested that epithelial cells are protected against viral replication by co-option of two evolutionarily related cytokine networks. These data may inform the design of novel immunotherapy for viral infections that are sensitive to interferons.


Assuntos
Citocinas/imunologia , Expressão Gênica/imunologia , Interleucinas/imunologia , Infecções por Rotavirus/imunologia , Animais , Células CACO-2 , Linhagem Celular , Chlorocebus aethiops , Citocinas/genética , Citocinas/farmacologia , Cães , Sinergismo Farmacológico , Células Epiteliais/imunologia , Células Epiteliais/metabolismo , Células Epiteliais/virologia , Expressão Gênica/efeitos dos fármacos , Células HT29 , Humanos , Immunoblotting , Interleucinas/genética , Interleucinas/farmacologia , Mucosa Intestinal/metabolismo , Intestinos/imunologia , Intestinos/virologia , Células Madin Darby de Rim Canino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Dados de Sequência Molecular , Receptores de Citocinas/genética , Receptores de Citocinas/imunologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Infecções por Rotavirus/genética , Infecções por Rotavirus/virologia , Fator de Transcrição STAT1/genética , Fator de Transcrição STAT1/imunologia , Fator de Transcrição STAT1/metabolismo , Células Vero , Interleucina 22
13.
Gut ; 64(2): 222-32, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24811998

RESUMO

OBJECTIVE: Antimicrobial peptides (AMP) provide protection from infection by pathogenic microorganisms and restrict bacterial growth at epithelial surfaces to maintain mucosal homeostasis. In addition, they exert a significant anti-inflammatory activity. Here we analysed the anatomical distribution and biological activity of an orally administered AMP in the context of bacterial infection and host-microbial homeostasis. DESIGN: The anatomical distribution as well as antibacterial and anti-inflammatory activity of the endogenous AMP cryptdin 2 and the synthetic peptide Pep19-2.5 at the enteric mucosal surface were analysed by immunostaining, functional viability and stimulation assays, an oral Salmonella enterica subsp. enterica sv. Typhimurium (S. Typhimurium) model and comparative microbiota analysis. RESULTS: Endogenous cryptdin 2 was found attached to bacteria of the enteric microbiota within the intestinal mucus layer. Similarly, the synthetic peptide Pep19-2.5 attached rapidly to bacterial cells, exhibited a marked affinity for the intestinal mucus layer in vivo, altered the structural organisation of endotoxin in a mucus matrix and demonstrated potent anti-inflammatory and antibacterial activity. Oral Pep19-2.5 administration induced significant changes in the composition of the enteric microbiota as determined by high-throughput 16S rDNA sequencing. This may have contributed to the only transient improvement of the clinical symptoms after oral infection with S. Typhimurium. CONCLUSIONS: Our findings demonstrate the anti-inflammatory activity and mucus affinity of the synthetic AMP Pep19-2.5 and characterise the influence on microbiota composition and enteropathogen infection after oral administration.


Assuntos
Antibacterianos/farmacocinética , Anti-Inflamatórios/farmacocinética , Mucosa Intestinal/metabolismo , Fragmentos de Peptídeos/farmacocinética , Administração Oral , Animais , Antibacterianos/administração & dosagem , Antibacterianos/uso terapêutico , Anti-Inflamatórios/administração & dosagem , Células Cultivadas , Defensinas , Avaliação Pré-Clínica de Medicamentos/métodos , Feminino , Interações Hospedeiro-Patógeno/fisiologia , Humanos , Mucosa Intestinal/microbiologia , Camundongos Endogâmicos C57BL , Microbiota/efeitos dos fármacos , Muco/metabolismo , Muco/microbiologia , Fragmentos de Peptídeos/administração & dosagem , Fragmentos de Peptídeos/uso terapêutico , Proteínas/metabolismo , Infecções por Salmonella/tratamento farmacológico , Infecções por Salmonella/metabolismo , Infecções por Salmonella/microbiologia , Salmonella typhimurium/efeitos dos fármacos , Salmonella typhimurium/metabolismo , Salmonella typhimurium/fisiologia
14.
J Immunol ; 193(8): 4223-34, 2014 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-25210121

RESUMO

Recent results indicate a significant contribution of innate immune signaling to maintain mucosal homeostasis, but the precise underlying signal transduction pathways are ill-defined. By comparative analysis of intestinal epithelial cells isolated from conventionally raised and germ-free mice, as well as animals deficient in the adaptor molecules MyD88 and TRIF, the TLR3 and TLR4, as well as the type I and III IFN receptors, we demonstrate significant TLR-mediated signaling under homeostatic conditions. Surprisingly, homeostatic expression of Reg3γ and Paneth cell enteric antimicrobial peptides critically relied on TRIF and, in part, TLR3 but was independent of IFN receptor signaling. Reduced antimicrobial peptide expression was associated with significantly lower numbers of Paneth cells and a reduced Paneth cell maturation and differentiation factor expression in TRIF mutant compared with wild-type epithelium. This phenotype was not transferred to TRIF-sufficient germ-free animals during cohousing. Low antimicrobial peptide expression in TRIF-deficient mice caused reduced immediate killing of orally administered bacteria but was not associated with significant alterations in the overall composition of the enteric microbiota. The phenotype was rapidly restored in a TRIF-independent fashion after transient epithelial damage. Our results identify TRIF signaling as a truly homeostatic pathway to maintain intestinal epithelial barrier function revealing fundamental differences in the innate immune signaling between mucosal homeostasis and tissue repair.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/imunologia , Peptídeos Catiônicos Antimicrobianos/imunologia , Imunidade Inata/imunologia , Mucosa Intestinal/imunologia , Listeria monocytogenes/imunologia , Proteínas/metabolismo , Salmonella typhimurium/imunologia , Proteínas Adaptadoras de Transporte Vesicular/genética , Animais , Peptídeos Catiônicos Antimicrobianos/biossíntese , Ciclinas/metabolismo , Mucosa Intestinal/microbiologia , Listeriose/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/genética , Proteínas Associadas a Pancreatite , Celulas de Paneth/metabolismo , Receptores de Interferon/genética , Infecções por Salmonella/imunologia , Transdução de Sinais/imunologia , Receptor 3 Toll-Like/genética , Receptor 4 Toll-Like/genética
15.
PLoS One ; 8(8): e70783, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23951007

RESUMO

The mouse pathobiont Helicobacter hepaticus can induce typhlocolitis in interleukin-10-deficient mice, and H. hepaticus infection of immunodeficient mice is widely used as a model to study the role of pathogens and commensal bacteria in the pathogenesis of inflammatory bowel disease. C57BL/6J Il10(-/-) mice kept under specific pathogen-free conditions in two different facilities (MHH and MIT), displayed strong differences with respect to their susceptibilities to H. hepaticus-induced intestinal pathology. Mice at MIT developed robust typhlocolitis after infection with H. hepaticus, while mice at MHH developed no significant pathology after infection with the same H. hepaticus strain. We hypothesized that the intestinal microbiota might be responsible for these differences and therefore performed high resolution analysis of the intestinal microbiota composition in uninfected mice from the two facilities by deep sequencing of partial 16S rRNA amplicons. The microbiota composition differed markedly between mice from both facilities. Significant differences were also detected between two groups of MHH mice born in different years. Of the 119 operational taxonomic units (OTUs) that occurred in at least half the cecum or colon samples of at least one mouse group, 24 were only found in MIT mice, and another 13 OTUs could only be found in MHH samples. While most of the MHH-specific OTUs could only be identified to class or family level, the MIT-specific set contained OTUs identified to genus or species level, including the opportunistic pathogen, Bilophila wadsworthia. The susceptibility to H. hepaticus-induced colitis differed considerably between Il10(-/-) mice originating from the two institutions. This was associated with significant differences in microbiota composition, highlighting the importance of characterizing the intestinal microbiome when studying murine models of IBD.


Assuntos
Colite/microbiologia , Suscetibilidade a Doenças/microbiologia , Infecções por Helicobacter/microbiologia , Helicobacter hepaticus/patogenicidade , Interleucina-10/imunologia , Microbiota/imunologia , Animais , Ceco/imunologia , Ceco/microbiologia , Ceco/patologia , Colite/imunologia , Colite/patologia , Colo/imunologia , Colo/microbiologia , Colo/patologia , DNA Complementar/classificação , DNA Complementar/genética , Suscetibilidade a Doenças/imunologia , Infecções por Helicobacter/imunologia , Infecções por Helicobacter/patologia , Helicobacter hepaticus/fisiologia , Interleucina-10/deficiência , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , RNA Ribossômico 16S/classificação , RNA Ribossômico 16S/genética , Análise de Sequência de DNA
16.
Proc Natl Acad Sci U S A ; 110(34): 13880-5, 2013 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-23898187

RESUMO

Helicobacter pylori infects the stomachs of one in two humans and can cause sequelae that include ulcers and cancer. Here we sequenced the genomes of 97 H. pylori isolates from 52 members of two families living in rural conditions in South Africa. From each of 45 individuals, two H. pylori strains were isolated from the antrum and corpus parts of the stomach, and comparisons of their genomes enabled us to study within-host evolution. In 5 of these 45 hosts, the two genomes were too distantly related to be derived from each other and therefore represented evidence of multiple infections. From the remaining 40 genome pairs, we estimated that the synonymous mutation rate was 1.38 × 10(-5) per site per year, with a low effective population size within host probably reflecting population bottlenecks and immune selection. Some individuals showed very little evidence for recombination, whereas in others, recombination introduced up to 100-times more substitutions than mutation. These differences may reflect unequal opportunities for recombination depending on the presence or absence of multiple infections. Comparing the genomes carried by distinct individuals enabled us to establish probable transmission links. Transmission events were found significantly more frequently between close relatives, and between individuals living in the same house. We found, however, that a majority of individuals (27/52) were not linked by transmission to other individuals. Our results suggest that transmission does not always occur within families, and that coinfection with multiple strains is frequent and evolutionarily important despite a fast turnover of the infecting strains within-host.


Assuntos
Evolução Molecular , Genoma Bacteriano/genética , Infecções por Helicobacter/transmissão , Helicobacter pylori/genética , Estômago/microbiologia , Sequência de Bases , Biologia Computacional , Infecções por Helicobacter/microbiologia , Humanos , Modelos Genéticos , Dados de Sequência Molecular , Taxa de Mutação , Densidade Demográfica , Recombinação Genética/genética , População Rural , Análise de Sequência de DNA , África do Sul
17.
FEMS Microbiol Rev ; 37(5): 736-61, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23790154

RESUMO

The human stomach is a formidable barrier to orally ingested microorganisms and was long thought to be sterile. The discovery of Helicobacter pylori, a carcinogenic bacterial pathogen that infects the stomach mucosa of more than one half of all humans globally, has started a major paradigm shift in our understanding of the stomach as an ecological niche for bacteria. The special adaptations that enable H. pylori to colonize this well-protected habitat have been intensively studied over the last three decades. In contrast, our knowledge concerning bacteria other than H. pylori in the human stomach is still quite limited. However, a substantial body of evidence documents convincingly that bacteria can regularly be sampled from the stomachs of healthy adults. Commonly detected phyla include Firmicutes, Actinobacteria, Bacteroidetes, and Proteobacteria, and characteristic genera are Lactobacillus, Streptococcus, and Propionibacterium. In this review, we summarize the available literature about the gastric microbiota in humans and selected model animals, discuss the methods used in its characterization, and identify gaps in our knowledge that need to be addressed to advance our understanding of the bacterial colonization of the different layers of the gastric mucosa and its potential role in health and disease.


Assuntos
Microbiota , Estômago/microbiologia , Animais , Biologia Computacional , Helicobacter pylori/isolamento & purificação , Humanos , Estômago/anatomia & histologia , Estômago/fisiologia
18.
PLoS One ; 6(12): e28012, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22164224

RESUMO

Many dinoflagellate species are notorious for the toxins they produce and ecological and human health consequences associated with harmful algal blooms (HABs). Dinoflagellates are particularly refractory to genomic analysis due to the enormous genome size, lack of knowledge about their DNA composition and structure, and peculiarities of gene regulation, such as spliced leader (SL) trans-splicing and mRNA transposition mechanisms. Alexandrium ostenfeldii is known to produce macrocyclic imine toxins, described as spirolides. We characterized the genome of A. ostenfeldii using a combination of transcriptomic data and random genomic clones for comparison with other dinoflagellates, particularly Alexandrium species. Examination of SL sequences revealed similar features as in other dinoflagellates, including Alexandrium species. SL sequences in decay indicate frequent retro-transposition of mRNA species. This probably contributes to overall genome complexity by generating additional gene copies. Sequencing of several thousand fosmid and bacterial artificial chromosome (BAC) ends yielded a wealth of simple repeats and tandemly repeated longer sequence stretches which we estimated to comprise more than half of the whole genome. Surprisingly, the repeats comprise a very limited set of 79-97 bp sequences; in part the genome is thus a relatively uniform sequence space interrupted by coding sequences. Our genomic sequence survey (GSS) represents the largest genomic data set of a dinoflagellate to date. Alexandrium ostenfeldii is a typical dinoflagellate with respect to its transcriptome and mRNA transposition but demonstrates Alexandrium-like stop codon usage. The large portion of repetitive sequences and the organization within the genome is in agreement with several other studies on dinoflagellates using different approaches. It remains to be determined whether this unusual composition is directly correlated to the exceptionally genome organization of dinoflagellates with a low amount of histones and histone-like proteins.


Assuntos
Dinoflagellida/genética , Dinoflagellida/fisiologia , Sequência de Aminoácidos , Cromossomos Artificiais Bacterianos/genética , Códon de Terminação , Mapeamento de Sequências Contíguas/métodos , DNA/genética , Elementos de DNA Transponíveis , DNA Complementar/metabolismo , Etiquetas de Sequências Expressas , Biblioteca Gênica , Genoma , Genômica , Histonas/química , Dados de Sequência Molecular , RNA Mensageiro/metabolismo , Análise de Sequência de DNA , Homologia de Sequência de Aminoácidos , Transcrição Gênica
19.
BMC Genomics ; 11: 248, 2010 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-20403159

RESUMO

BACKGROUND: The dinoflagellate Alexandrium minutum typically produces paralytic shellfish poisoning (PSP) toxins, which are known only from cyanobacteria and dinoflagellates. While a PSP toxin gene cluster has recently been characterized in cyanobacteria, the genetic background of PSP toxin production in dinoflagellates remains elusive. RESULTS: We constructed and analysed an expressed sequence tag (EST) library of A. minutum, which contained 15,703 read sequences yielding a total of 4,320 unique expressed clusters. Of these clusters, 72% combined the forward-and reverse reads of at least one bacterial clone. This sequence resource was then used to construct an oligonucleotide microarray. We analysed the expression of all clusters in three different strains. While the cyanobacterial PSP toxin genes were not found among the A. minutum sequences, 192 genes were differentially expressed between toxic and non-toxic strains. CONCLUSIONS: Based on this study and on the lack of identified PSP synthesis genes in the two existent Alexandrium tamarense EST libraries, we propose that the PSP toxin genes in dinoflagellates might be more different from their cyanobacterial counterparts than would be expected in the case of a recent gene transfer. As a starting point to identify possible PSP toxin-associated genes in dinoflagellates without relying on a priori sequence information, the sequences only present in mRNA pools of the toxic strain can be seen as putative candidates involved in toxin synthesis and regulation, or acclimation to intracellular PSP toxins.


Assuntos
Dinoflagellida/genética , Perfilação da Expressão Gênica , Toxinas Marinhas/genética , Dinoflagellida/metabolismo , Etiquetas de Sequências Expressas , Análise de Sequência com Séries de Oligonucleotídeos , Intoxicação por Frutos do Mar/microbiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...