Skip to main content

Characterization of the non-glandular gastric region microbiota in Helicobacter suis-infected versus non-infected pigs identifies a potential role for Fusobacterium gastrosuis in gastric ulceration

Abstract

Helicobacter suis has been associated with development of gastric ulcers in the non-glandular part of the porcine stomach, possibly by affecting gastric acid secretion and altering the gastric microbiota. Fusobacterium gastrosuis is highly abundant in the gastric microbiota of H. suis-infected pigs and it was hypothesized that this micro-organism could play a role in the development of gastric ulceration. The aim of this study was to obtain further insights in the influence of a naturally acquired H. suis infection on the microbiota of the non-glandular part of the porcine stomach and in the pathogenic potential of F. gastrosuis. Infection with H. suis influenced the relative abundance of several taxa at phylum, family, genus and species level. H. suis-infected pigs showed a significantly higher colonization rate of F. gastrosuis in the non-glandular gastric region compared to non-infected pigs. In vitro, viable F. gastrosuis strains as well as their lysate induced death of both gastric and oesophageal epithelial cell lines. These gastric cell death inducing bacterial components were heat-labile. Genomic analysis revealed that genes are present in the F. gastrosuis genome with sequence similarity to genes described in other Fusobacterium spp. that encode factors involved in adhesion, invasion and induction of cell death as well as in immune evasion. We hypothesize that, in a gastric environment altered by H. suis, colonization and invasion of the non-glandular porcine stomach region and production of epithelial cell death inducing metabolites by F. gastrosuis, play a role in gastric ulceration.

Introduction

Gastric ulceration is a common disease entity of pigs worldwide, with prevalences of up to 93%. It may result in decreased daily weight gain, decreased feed intake and sudden death, leading to significant economic losses and animal welfare issues. The aetiology is multifactorial. Diet particle size, management and infection with pathogens are factors that have been hypothesized to be involved [1]. The exact pathophysiological mechanism behind porcine gastric ulceration, however, is not completely clear. In marked contrast with human patients and several other animal species, gastric ulcers do not develop in the glandular part of the porcine stomach, but are almost exclusively found in the Pars oesophagea, a small area around the opening of the oesophagus which does not contain glands. Since this stomach region is not protected by mucus, it is highly susceptible to irritation by hydrochloric acid produced in the distal gland zone of the porcine stomach. Chronic irritation of the Pars oesophagea results in hyperkeratosis, erosion and finally ulceration [1, 2].

Helicobacter suis is a zoonotic bacterium that colonizes the gastric mucosa of pigs worldwide. Results of recent studies indicate that H. suis infection plays a role in porcine gastric ulcer disease, probably by affecting gastric acid secretion through alteration of the number and/or function of parietal, D- and G-cells [3, 4]. Impaired gastric acid secretion, induced by H. suis, may favor the establishment of specific gastric microbiota. Our metagenomics study of pooled samples from the different stomach regions (i.e. Pars oesophagea, cardiac, fundic and pyloric gland zones) from H. suis-positive and -negative 6ā€“8Ā months old pigs revealed that a novel Fusobacterium sp., designated F. gastrosuis, was highly abundant in the gastric microbial community of H. suis-infected animals [5].

Fusobacteria are normal constituents of the oropharyngeal, gastrointestinal and genital microbiota of a wide range of animal species, going from mammals to birds and fish. Nevertheless, they are also frequently isolated from clinical samples of both human and animal origin, especially from cases of pyonecrotic infections [6]. Infections with Fusobacterium spp. have been linked to a wide range of pathologies. Furthermore, genome analysis of Fusobacterium spp. showed the presence of a large and diverse set of virulence associated genes, such as leukotoxin and immunosuppressive protein A (fipA), which have been associated with cell death and immuno-evasion, respectively [7]. Additionally, some Fusobacterium spp. were able to adhere to and actively invade host cells without the aid of other factors, due to the presence of genes encoding adhesins and membrane-related proteins, while others only aggravated necrosis when tissue damage was initiated by other microorganisms or environmental factors [6].

We hypothesize that, in a gastric environment altered by H. suis, colonization and invasion of the Pars oesophagea and production of epithelial cell death inducing metabolites by F. gastrosuis may play a role in gastric ulceration.

The overall aim of the current study was to obtain further insights in the influence of a naturally acquired H. suis infection on the microbiota of the Pars oesophagea of the porcine stomach and in the pathogenic potential of F. gastrosuis upon co-infection. Therefore, the ability of F. gastrosuis to induce cell death in oesophageal and gastric epithelial cell lines was first determined and its genome was also analysed for the presence of genes encoding putative virulence factors which may be involved in adhesion, invasion and host cell death as well as in immuno-evasion. Secondly, the colonization rate of F. gastrosuis and its abundance in the microbiota of the Pars oesophagea was compared in H. suis-infected and non-infected pigs.

Materials and methods

Study 1: Microbiota composition of the Pars oesophagea

Sampling of porcine stomachs

Ten H. suis-positive and 10 H. suis-negative stomachs of 6ā€“8Ā months old pigs used in another study from our group were further analysed [8]. Using autoclaved tweezers and scalpels, a biopsy of 40ā€“50Ā mg consisting of mucosa and submucosa was taken from the Pars oesophagea for gastric microbiota analysis and subsequent F. gastrosuis quantification by quantitative real time (RT)-PCR was realized. In addition, a second biopsy consisting of mucosa and submucosa was taken from the Pars oesophagea to determine mRNA expression levels of genes encoding host factors (markers) involved in inflammation, hyperkeratosis and ulceration.

Gastric microbiota analysis

DNA was extracted from the gastric biopsy of the Pars oesophagea using the DNeasy Blood & Tissue Kit (Qiagen, Hilden, Germany) according to the instructions of the manufacturer. 16S rRNA gene amplicon pyrosequencing was performed using the Roche GS-Junior Genome Sequencer as described previously [9]. The obtained 16S rDNA sequence reads were processed using MOTHUR (software package v1.35), Pyronoise algorithm and UCHIME algorithm for alignment and clustering, denoising and chimera detection, respectively [10, 11]. The obtained read sets were compared to a reference dataset of aligned sequences of the corresponding region derived from the SILVA database (v1.15) of full-length rRNA sequences implemented in MOTHUR [12]. The final reads were clustered into operational taxonomic units (OTUs) using the nearest neighbor algorithm using MOTHUR with a 0.03 distance unit cut-off. A taxonomic identity was attributed to each OTU by comparison with the SILVA database using a 80% homogeneity cut-off. When a taxonomic identification lower than 80% was obtained, the taxonomic level was labelled with the first defined level from higher level followed by the label ā€œunclassifiedā€. Finally, the unique sequences for each OTU were compared with the SILVA data set using the BLASTN algorithm. For each OTU, a consensus taxonomic identification was given when less than 1% of mismatch with the aligned sequence was obtained. In the final metadata table, the following labelling was used: the population is identical to a taxonomically defined species and is labelled ā€œgenus_speciesā€; the population is identical to a reference sequence belonging to a still undefined species and is labelled ā€œgenus_NCBI accession numberā€; the sequence is not identical to any known sequence and is labelled with the corresponding OTU number.

In order to determine the effect of H. suis on the gastric microbiota composition, the H. suis-negative and H. suis-positive groups were compared.

Subsampled datasets were obtained and evaluated in MOTHUR to estimate the richness, microbial diversity and population evenness by using the Chao1 estimator, Simpsonā€™s reciprocal index and Simpsonā€™s evenness index, respectively [13, 14]. Population structure and community membership were assessed with MOTHUR using distance matrix based on Brayā€“Curtis dissimilarity index. Differences in functional profiles of gastric bacterial communities were analysed by mapping taxa into several phenotypes (i.e. metabolism, Gram staining, sporulationā€¦) using METAGENassist [15]. Only the phenotypes detected in more than 50% of the samples were included for further analysis.

Statistical analysis

Statistical differences in microbial diversity, richness and population evenness between the groups were investigated using non-parametric Kruskalā€“Wallis tests with Tukey post hoc tests using PRISM 7 (Graphpad Software). Using MOTHUR, community composition differences were investigated using Analysis of molecular variance (AMOVA) and homogeneity of molecular variance (HOMOVA). In order to highlight statistical differences in relative bacterial abundance between the groups, non-parametric Kruskalā€“Wallis tests with Tukey post hoc tests and Benjaminiā€“Hochberg false discovery rate were performed using the STAMP software. Differences were considered statistically significant at a corrected p-value of less thanā€‰0.05.

Study 2: F. gastrosuis colonization

Sampling of porcine stomachs

Sixty-eight stomachs of 6ā€“8Ā months old pigs and 60 stomachs of adult sows (1ā€“3Ā years old) were collected over a period of 10Ā months from two slaughterhouses in Flanders, Belgium. The pigs originated from different herds. Stomachs of 46, 2ā€“3Ā months old pigs were collected from two different pig herds (23 samples from each herd). The stomachs of all pigs had been previously used [4]. Based on the method of Hessing [16], mucosal lesions of the Pars oesophagea were scored as follows: score 0 for normal mucosa, score 1 for mild hyperkeratosis encompassing less than 50% of the surface, score 2 for severe hyperkeratosis encompassing more than 50% of the surface, score 3 for hyperkeratosis with few erosions, score 4 for hyperkeratosis with several erosions and score 5 for hyperkeratosis with many erosions or ulceration. Using autoclaved tweezers and scalpels, biopsies of 40ā€“50Ā mg consisting of mucosa and submucosa were taken from the Pars oesophagea as well as from the cardiac, fundic and pyloric gland zone for quantification of F. gastrosuis DNA by RT-PCR. In addition, a swab from the oral cavity was taken from each 2ā€“3Ā months old pig using a MasterAmpā„¢ Buccal Brush (Epicentre, Madison, USA), as well as biopsies of 40ā€“50Ā mg consisting of mucosa and submucosa from jejunum and colon.

Quantification of F. gastrosuis by RT-PCR

DNA was extracted from the gastric biopsies from the Pars oesophagea, cardiac, fundic and pyloric gland zone from the different age groups and from the jejunum and colon biopsies from the 2ā€“3Ā months old pigs using the DNeasy Blood & Tissue Kit (Qiagen, Hilden, Germany) according to the instructions of the manufacturer. In addition, DNA was extracted from the oral cavity swab using PrepMan Ultra Sample Preparation Reagent (Life Technologies, Carlsbad, California) according to the manufacturerā€™s instructions. The DNA extracted from the Pars oesophagea biopsies sampled for gastric microbiota analysis in study 1 was also included for F. gastrosuis quantification.

The presence of F. gastrosuis DNA was determined using a species-specific, RT-PCR based on the gyrase B (gyrB) gene [5]. The copy number of the obtained amplicons was calculated and converted to the number of F. gastrosuis bacteria per mg gastric tissue, by including tenfold dilutions of an external standard. For generating the standard, part of the gyrB gene (1212Ā bp) from F. gastrosuis strain CDW1 was amplified using UP-1 and UP-2r primers as previously described [5]. The standard consisted of tenfold dilutions, starting at 108 PCR amplicons, for each 10Ā ĀµL of reaction mixture. Two microliter of extracted DNA template was added to 10Ā ĀµL reaction mixture, consisting of 0.25Ā ĀµL of both primers (0.5Ā pmol/ĀµL) located within the 1212Ā bp fragment (to yield a 158Ā bp PCR product), 3.5Ā ĀµL HPLC water and 6Ā ĀµL SensiMixā„¢ SYBR No-ROX (Bioline Reagents Ltd, London, UK). Sense primer was GB2_F: 5ā€²-GCA GCT CAA AGA GCA AGA GAA GCA-3ā€². Anti-sense primer was GB2_R: 5ā€²-CTT CCC TGC TTT GCA GAA CCT CC-3ā€². The cycling conditions were initial denaturation at 95Ā Ā°C for 10Ā min, followed by 47 cycles of 95Ā Ā°C for 20Ā s, 60Ā Ā°C for 30Ā s and 72Ā Ā°C for 30Ā s after which the total fluorescence of the samples was measured. Both standards and samples were run in duplicate on a CFX384ā„¢ RT-PCR System with a C1000 Thermal Cycler (Bio-Rad, Hercules, California, USA).

Expression of markers for inflammation and ulceration

RNA was extracted from the gastric biopsies of the Pars oesophagea using the RNeasy Mini Kit (Qiagen, Hilden, Germany), according to the manufacturerā€™s instructions. The obtained RNA concentrations were measured using a NanoDrop spectrophotometer (Isogen Life Science, Utrecht, The Netherlands), after which the concentration of each sample was adjusted to 1Ā Ī¼g/ĀµL, followed by cDNA synthesis using the iScriptā„¢ cDNA Synthesis Kit (Bio-Rad). Expression analysis was then performed for genes encoding host factors involved in inflammation [(interleukin (Il)-1Ī² IL-6, IL-8, IL-10, CXCL13, interferon (IFN)-Ī³ and tumor necrosis factor (TNF)-Ī±] as well as in hyperkeratosis and ulceration [claudin 1, 2, 3, 4 and 18; keratin 6A; heat shock protein (Hsp) 27, 72 and 73; epidermal growth factor (EGF); basic fibroblast growth factor (BFGF); hepatocyte growth factor (HGF); transforming growth factor beta 1 (TGFB1); cyclooxygenase 2 (COX2); nitric oxide synthase 2 (NOX2); CXCL2; occludin; zonula occludens (ZO) 1 and 2]. The housekeeping genes HMBS, Cyc, RPL4 and HPRT1 were shown to have a stable mRNA expression and therefore included as reference genes [17, 18]. All primer sequences are shown in Additional file 1. The mRNA expression levels of the reference and target genes were quantified using a RT-PCR, as previously described [17]. No-template-control reaction mixtures were included and all samples were run in duplicate. The Ct-values were first normalized to the geometric mean of the Ct-values of the reference genes. Fold changes were calculated using Ī”Ī”Ct method with mean of Ct-values from the H. suis-negative group. Finally, for each target gene, the results were expressed as fold changes of the mRNA expression of the H. suis-positive group relative to mRNA expression level of the H. suis-negative group.

Statistical analysis

Statistical analysis was performed using SPSS statistics 24 (IBM, New York, USA). Differences in severity of Pars oesophageal lesions, number of colonizing F. gastrosuis bacteria and fold changes of the markers for inflammation, hyperkeratosis and ulceration were investigated using the non-parametric Kruskalā€“Wallis H test with Bonferroni correction. A p-valueā€‰ā‰¤ā€‰0.05 was considered to be significant. Correlations between mucosal lesions, fold changes and the number of F. gastrosuis bacteria were examined using the Pearson correlation coefficient. Differences were considered statistically significant at pā€‰ā‰¤ā€‰0.05.

Study 3: Effect of F. gastrosuis on epithelial neoplastic cell lines

Bacterial strains

Fusobacterium gastrosuis strains CDW1, CDW3, CDW6 and CDW8 were used for the in vitro cell experiments as previously described [5]. All strains were isolated from the Pars oesophagea of pigs suffering from gastric ulceration.

Cell lines and culture conditions

All in vitro cell experiments were performed with MKN7 (human gastric tubular adenocarcinoma cell line, Riken Cell Bank, Japan) and KYSE-450 (human oesophageal squamous cell carcinoma, ACC 387; Deutsche Sammlung von Mikroorganismen und Zellkulturen) cells. The MKN7 cells were cultured in 89% RPMI medium 1640 (supplemented with 1Ā mM l-glutamine, Invitrogen, Waltham, MA, USA), 10% fetal calf serum (FCS; HyClone, Logan, Utah, USA) and 1% penicillinā€“streptomycin (10Ā 000Ā U/mL, Invitrogen). The KYSE-450 cells were cultured in 44.5% RPMI medium 1640 (supplemented with 1Ā mMĀ l-glutamine, Invitrogen), 44.5% Hamā€™s F12 medium (supplemented with 1Ā mMĀ l-glutamine, Invitrogen), 10% FCS (HyClone) and 1% penicillinā€“streptomycin (10Ā 000Ā U/mL, Invitrogen). Penicillinā€“streptomycin was not added to the media for in vitro cell experiments with viable F. gastrosuis bacteria. The cells were seeded in plastic tissue culture flasks (VWR, Radnor, Pennsylvania, USA), maintained in a humidified incubator at 37Ā Ā°C under 5% CO2 and passaged at least twice a week using 1% trypsin solution consisting of 88% trypsin diluent (8Ā g NaCl, 0.2Ā g KCl, 0.12Ā g KH2PO4, 0.91Ā g Na2HPO4 and 4Ā mL 0.5% phenol red solution/1000Ā mL aqua dest), 10% trypsin stock solution (Invitrogen) and 2% EDTA (2Ā g/100Ā mL trypsin diluent).

Cell-death inducing effects of F. gastrosuis lysates

Both MKN7 and KYSE-450 cell lines were incubated with F. gastrosuis lysates from strains CDW1, CDW3, CDW6 and CDW8 to determine cytotoxicity. To prepare the F. gastrosuis lysate, bacteria were incubated anaerobically for 2Ā days on Columbia blood agar plates (Oxoid, Basingstoke, United Kingdom). After incubation, they were harvested by centrifugation (368Ā g, 10Ā min, 4Ā Ā°C), washed twice with Hankā€™s Balanced Salt Solution without calcium and magnesium (HBSSāˆ’) (Invitrogen) and resuspended in HBSSāˆ’ until an optical density (OD) of 2.9 at 660Ā nm was obtained (i.e. approximately 1ā€‰Ć—ā€‰108Ā bacteria/mL). The bacterial suspension was sonicated 20 times for 30Ā s and centrifuged (15Ā 000Ā g; 5Ā min, 4Ā Ā°C) to remove cellular debris. The supernatant was then filtered through a 0.22Ā Āµm pore filter (Schleicher and Schuell, Munich, Germany) and stored at āˆ’80Ā Ā°C. The resulting protein concentration was determined with the Pierce BCA protein assay kit (ThermoFisher Scientific, Waltham, Massachusetts, USA), according to the manufacturerā€™s instructions. The MKN7 and KYSE-450 cell lines were seeded at a concentration of 8ā€‰Ć—ā€‰104Ā cells/mL and 1ā€‰Ć—ā€‰105Ā cells/mL, respectively, in 6-well flat-bottom cell-culture plates (Greiner Bio One, Frickenhausen, Germany) and incubated at 37Ā Ā°C and 5% CO2 until a monolayer was obtained after 48Ā h. Each well was washed twice with Hankā€™s Balanced Salt Solution with calcium and magnesium (HBSS+) to remove dead cells caused by the trypsin treatment and/or seeding. As previously described, each F. gastrosuis lysate was then added in different concentrations [50, 200 and 500Ā Ī¼g/mL, corresponding to a calculated theoretical multiplicity of infection (MOI) of approximately 5, 20 and 50 respectively] and incubated for 0, 24, 36 and 48Ā h [19]. As a positive control, F. necrophorum subps. necrophorum lysate (50, 200 and 500Ā Ī¼g/mL) was used. Untreated cells of both cell lines were included as negative controls, as well as positive controls for necrosis and apoptosis using 0.1% Triton X-100 and 1Ā Ī¼M staurosporine, respectively. Additional controls were used to quantify and compensate for spectral bleed-through. All conditions were performed in triplicate. After incubation, the cells were trypsinized and washed 3 times with HBSSāˆ’ to remove cellular debris. Next, the cells were stained incubated for 10Ā min in the dark at room temperature with a labelling solution (10Ā mM 4-(2-hydroxyethyl)-1-piperazine ethanesulfonic acid (HEPES), 140Ā mM NaCl and 5Ā mM CaCl2) containing Annexin-V-fluorescein isothiocyanate (1/50 diluted Annexin-V-FITC; 11828681001, Sigma-Aldrich) and propidium iodide (1Ā Āµg/mL PI; P4864, Sigma-Aldrich). Acquisition and analysis was performed on a CytoFLEX flow cytometer using the CytExpert 2.0 software (Beckman Coulter, Indianapolis, Indiana, USA). Following terms were used to describe the different subpopulations: viable cells (Annexin-V-FITC negative, PI negative), early apoptotic cells (Annexin-V-FITC positive, PI negative) and late apoptotic/necrotic cells (Annexin-V-FITC positive, PI positive) [20]. A laser line of 488Ā nm was used with emission filters 525/40 and 585/42 for detection of annexin and propidium iodide, respectively. Before each measurement, calibration was performed to exclude instrument-related fluorescence intensity changes over time using CytoFLEX daily QC fluorospheres (Beckman Coulter). A minimum number of 10Ā 000 events in the single, viable cell gate was analysed for each sample. The gating strategy for both cell lines is presented in Additional files 2, 3.

Simultaneously, the presence of apoptosis and/or necrosis was verified using light microscopy. The cells were harvested and cytospins were prepared by centrifugation at 55Ā g for 5Ā min. Slides were air-dried, fixed in methanol and stained with Hemacolor (Merck, Darmstadt, Germany).

Cell-death inducing effect of viable F. gastrosuis bacteria

Prior to infection, the viability of F. gastrosuis grown and harvested as described above was determined. Approximately 5ā€‰Ć—ā€‰108Ā bacteria/mL cell medium were added to 6-well flat-bottom cell-culture plates and incubated for 0, 6, 12, 24 and 36Ā h at 37Ā Ā°C and 5% CO2. After incubation, the colony forming units (cfu) were determined by serial dilution and plating on Columbia agar plates (Oxoid, Basingstoke, UK) supplemented with 5% defibrinated sheep blood (E&O laboratories, Bonnybridge, Scotland, UK). The plates were incubated anaerobically for 3Ā days at 37Ā Ā°C.

Both MKN7 and KYSE-450 cell lines were incubated with viable F. gastrosuis bacteria from strains CDW1, CDW3, CDW6 and CDW8 to determine the effect on cell death. The bacteria were incubated and harvested until an OD of 2.9 was obtained, as described above. After obtaining a monoculture of MKN7 and KYSE-450 cell lines (Ā±ā€‰5ā€‰Ć—ā€‰105Ā cells/mL), these were inoculated with the different F. gastrosuis strains at a MOI of 5, 20 and 50 (i.e. corresponding to approximately 50, 200 and 500Ā Ī¼g/mL lysate, respectively) and incubated for 0, 2, 6 and 12Ā h. The percentages of viable, apoptotic and necrotic cells were determined using the CytoFLEX as described above.

Properties of cell death inducing F. gastrosuis component(s)

To determine whether the cell death inducing component(s) were of protein nature, F. gastrosuis bacteria were pre-treated with pronase, trypsine or proteinase K (all at 1Ā mg/mL, 2Ā h, 37Ā Ā°C) (Sigma-Aldrich) prior to incubation with MKN-7 and KYSE-450 cells. Bacteria were also pre-treated with paraformaldehyde (1%, 1Ā h, room temperature) to determine if cell death inducing agents were associated with the bacterial surface. Finally, F. gastrosuis bacteria and lysates were heat treated (100Ā Ā°C, 10Ā min) to investigate their heat-lability. The percentages of viable, apoptotic and necrotic cells were flowcytometrically determined as described above. Negative as well as positive controls (i.e. untreated F. gastrosuis bacteria and lysates, respectively) were included. The viability of F. gastrosuis bacteria after protease, paraformaldehyde and heat treatment was verified by incubation on Columbia blood agar plates (37Ā Ā°C, 48ā€“72Ā h, anaerobic) after which bacterial growth was compared with that of untreated F. gastrosuis bacteria.

Activation of the pyroptotic pathway

Similarly as shown for F. nucleatum [21], activation of the pyroptotic pathway was investigated by determining caspase-1 and IL-1Ī² expression using Western blot analysis.

Both MKN-7 and KYSE-450 cell lines were incubated either for 0, 24, 36 and 48Ā h with 50, 200 and 500Ā Ī¼g/mL F. gastrosuis lysates from strains CDW1, CDW3, CDW6, or with 5, 20 and 50 MOI F. gastrosuis bacteria from strains CDW1, CDW3, CDW6 for 0, 6, 12, 24 and 36Ā h as described. After incubation, total cell extracts were obtained using RIPA buffer supplemented with a protease inhibitor cocktail, as previously described [22]. Protein concentration was determined with the Pierce BCA protein assay kit, according to the manufacturerā€™s instructions (ThermoFisher). After the samples were boiled for 5Ā min at 95Ā Ā°C, a total of 100Ā Ī¼g was suspended in 200Ā Ī¼L loading buffer (i.e. 24Ā mL of 0.5Ā M Tris pH 6.8, 20Ā mL of 10% SDS, 10Ā mL glycerol, 5Ā mL of 0.5% bromophenol blue and 48Ā mL aqua dest) and loaded on a 10% self-prepared denaturing polyacrylamide gel. Pagerulerā„¢ plus prestained protein ladder with bands from 10 to 250Ā kDa (ThermoFisher) was used for molecular mass determination. Separation under reducing conditions was followed by blotting on a 0.45Ā Āµm nitrocellulose membrane (Bio-Rad). After blocking in Tris-buffered saline with 0.1% Tween 20 (TBS-T) and 5% milk powder for 1Ā h, overnight incubation with the primary anti-IL1Ī² (0.1Ā Ī¼g/mL; AF-201-NA, R&D systems, Minnesota, USA) and anti-caspase 1 (0.1Ā Ī¼g/mL; AB207802, Abcam, Cambridge, UK) in TBS-T with 5% milk powder was carried out. Membranes were washed with TBS-T and horseradish peroxidase-conjugated donkey-anti-goat-antibody, respectively goat-anti-rabbit-antibody (each at 1:5000 in TBS-T with 5% milk powder) were each applied for 1Ā h. The blots were washed and protein bands were immunodetected by enhanced chemiluminescence, using Supersignal West Dura Extended Duration Substrate (ThermoFisher). The recombinant human IL-1Ī²/IL-1F2 protein (201-LB, R&D Systems) and recombinant human caspase-1 protein (AB39901, Abcam) served as positive controls for the western blot analysis. Untreated MKN-7 and KYSE-450 cells served as negative control.

Statistical analysis

Statistical analysis was performed using SPSS statistics 24 (IBM, New York, USA). Differences in the percentage of viable, early apoptotic and late apoptotic/necrotic cells and cellular debris were investigated using the non-parametric Kruskalā€“Wallis H test with Bonferroni correction. A p-valueā€‰ā‰¤ā€‰0.05 was considered to be significant.

Study 4: Potential virulence associated genes in F. gastrosuis

Bacterial strain

The proteome of the F. gastrosuis type strain CDW1 has been previously determined (LT607734.1) [5]. In the current study, its proteome was further investigated for presence of genes encoding putative virulence factors which may be involved in adhesion, invasion and host cell death as well as immunoevasion.

Essential genes

The database of essential genes (DEG) version 15.2 consists of 53,885 essential protein-coding genes and 786 essential non-coding sequences from 48 prokaryotes and 26 eukaryotes. BLASTp search was performed for the proteome of F. gastrosuis against bacterial proteins of DEG with cut-off parameters of 1Eāˆ’05 e-value and bit score of 100.

Potential virulence associated genes

Screening for presence of virulence associated genes was performed using the VirulentPred tool. The proteome of F. gastrosuis was also blasted against Pfam domains associated with virulence using the Pfam search tool. In addition, the proteome of F. gastrosuis was blasted against protein sequences from the virulence factor database (VFDB) core dataset (setA) with default parameters. This database contains 1796 virulence factors and 2599 curated virulence factor-related genes obtained from 74 bacterial pathogens. The query sequences having hit with cut-off bit score >ā€‰100 were considered as potential virulence factors. Finally, data on virulence factors from other Fusobacterium spp. reported in the literature was collected [7, 23,24,25], as well as data on genes characteristic for active invasion [6]. Homology search of these proteins was performed to find close matches in F. gastrosuis using BLASTP. The query sequences having an E-valueā€‰<ā€‰1eāˆ’6 and an identity >ā€‰30% over at least 100 amino acids were hypothesized to have a similar structure and function.

Results

Study 1: Microbiota composition of the Pars oesophagea

Sufficient sequencing reads were obtained from the Pars oesophagea of all pigs (nā€‰=ā€‰20). Pyrosequencing yielded between 7479 and 7500 reads per sample. A total of 284,815 final reads were attributed to 3837 species level OTUs. Chimeric sequences represented 5ā€“10% of the total sequencing reads and were excluded from the analysis.

In general, total bacterial community analysis showed that the most dominant phylum in the porcine Pars oesophagea was Proteobacteria (56%), followed by Firmicutes (30%), Bacteroidetes (10%), Fusobacteria (3%) and Actinobacteria (1%). The relative abundance of other phyla was below 0.1%. On family level, following populations (i.e. >ā€‰1%) were dominant: Pasteurellaceae (33%), Clostridiaceae_1 (11%), Lactobacillaceae (10%), Enterobacteriaceae (9%), Prevotellaceae (7%), Comamonadaceae (4%), Veillonellaceae (3%), Moraxellaceae (3%), Campylobacteraceae (2%), Fusobacteriaceae (2%), Neisseriaceae (2%), Porphyromonadaceae (2%) and Streptococcaceae (2%). The major genera (i.e. >ā€‰1%) were Actinobacillus (29%), Clostridium_sensu_stricto_1 (10%), Lactobacillus (10%), Escherichia (7%), Pasteurellaceae_unclassified (4%), Pelomonas (4%), Moraxella (3%), Prevotella_7 (3%), Alloprevotella (2%), Campylobacter (2%), Fusobacterium (2%), Neisseriaceae_unclassified (2%), Salmonella (2%), Streptococcus (2%) and Veillonella (2%). The average gastric bacterial community composition at the phylum, family and genus level present in the H. suis-positive and -negative groups is represented in FigureĀ 1, while the bacterial community composition of the Pars oesophagea of each individual pig is shown in Additional file 4.

FigureĀ 1
figure 1

Average bacterial community compositions in the Pars oesophagea of H. suis-positive and -negative pigs. The cumulated histograms show the relative abundance of the identified taxa at phylum (A), family (B) and genus (C) level in the Pars oesophagea of H. suis-positive (nā€‰=ā€‰10) and -negative (nā€‰=ā€‰10) pigs. At family and genus level, taxa with a relative abundance <ā€‰1% are merged in the category ā€œothersā€. The unclassified populations correspond to defined groups of the genus level for which a taxonomical classification assignation to the genus could not be attributed. These populations are therefore labelled with the first defined superior hierarchical taxonomic level followed by ā€œ_unclassifiedā€ to prevent confusion.

Infection with H. suis had no effect on microbial diversity, richness and population evenness (Additional file 5). Furthermore, Unifrac-weighted analysis as well as AMOVA and HOMOVA did not reveal significant differences regarding community structure and composition of the groups. Population structure and community membership, as determined by Brayā€“Curtis dissimilarity index, were also not different between the groups.

In general, phenotypic analysis of the microbiota of the porcine Pars oesophagea revealed the presence of 19 metabolic phenotypes of which ammonia oxidizer, dehalogenation, nitrite reducer, sulfate reducer, sulfide oxidizer, chitin degradation, nitrogen fixation and xylan degrader were the most abundant, each accounting for 85%, 70%, 53%, 51%, 48%, 47%, 12% and 9% of the bacterial community, respectively. In the Pars oesophageal microbiota of all pigs, non-sporulating and Gram-negative bacteria were more abundant than sporulating and Gram-positive bacteria, respectively (38% vs 15% and 67% vs 26%, respectively). The H. suis-negative pigs showed a relative lower abundance of sporulating, nitrogen fixating and propionate metabolizing bacteria compared to the H. suis-positive pigs, although this difference was not significant (9% vs 21%, pā€‰=ā€‰0.075; 8% vs 17%, pā€‰=ā€‰0.063; 4% vs 12%, pā€‰=ā€‰0.064; respectively). Furthermore, H. suis-negative pigs showed a relative higher abundance of autotrophic bacteria compared to H. suis-positive pigs, which was borderline significant (68% vs 44%, pā€‰=ā€‰0.052).

Infection with H. suis influenced the relative abundance of several taxa at phylum, family, genus and species level, either significantly or borderline significantly. A detailed overview is presented in TableĀ 1. In general, H. suis-positive pigs showed a relative higher abundance of the following taxa with at phylum level more Fusobacteria, at family level more Fusobacteriaceae, Porphyromonadaceae, Bacteroidaceae, Clostridiaceae_1, and at genus level more Fusobacterium, Porphyromonas, Porphyromonadaceae_unclassified, Bacteroides, Alloprevotella, Clostridium_sensu_stricto_1, Leuconostoc, Pasteurella and Gammaproteobacteria_unclassified. A relative lower abundance was detected for the following taxa with at family level less Actinobacteria_unclassified, Bifidobacteriaceae, Corynebacteriaceae, Coriobacteriaceae, Streptococcaceae and Gammaproteobacteriaceae_unclassified, at genus level less Actinobacteria_unclassified, Bifidobacterium, Corynebacteriaceae_unclassified, Lachnospiraceae_UCG-007, Lachnospiraceae_XPB1014_group, Staphylococcus, Aerococcus, Selenomonas, Megasphaera, Mitsuokella and Veillonellaceae_unclassified.

TableĀ 1 Overview of the main differences in relative abundance of taxa at phylum, family, genus and species level in the Pars oesophagea of the H. suis -positive and -negative pigs

The relative abundance of F. gastrosuis was higher in the Pars oesophagea of H. suis-positive pigs compared to H. suis-negative pigs (3% vs 0.9%, pā€‰=ā€‰0.068). This was confirmed by RT-PCR, as H. suis-infected pigs showed a significantly higher colonization rate of F. gastrosuis in the Pars oesophagea compared to non-infected pigs (pā€‰<ā€‰0.05) (Additional file 6D).

Study 2: F. gastrosuis colonization in the porcine gastro-intestinal tract and impact of H. suis infection

In the 2ā€“3Ā months old pigs, the highest abundance of F. gastrosuis was found in the oral cavity, followed by the Pars oesophagea, independent from the H. suis status (pā€‰<ā€‰0.05) (Additional file 7). All pigsā€™ stomachs tested positive for presence of F. gastrosuis and the average number of bacteria per mg tissue was higher in the Pars oesophagea than in the other stomach regions (pā€‰<ā€‰0.01) (Additional file 6). The number of F. gastrosuis bacteria was lower in the 2ā€“3-compared to the 6ā€“8Ā months old pigs and adult sows for each stomach region (pā€‰<ā€‰0.01), while no significant differences were observed between the latter groups (Additional file 8).

The number of F. gastrosuis bacteria in the oral cavity was highly and positively correlated with the number in the Pars oesophagea (pā€‰<ā€‰0.001). The number of F. gastrosuis in the Pars oesophagea was highly and positively correlated with each of the numbers in the cardiac, fundic and pyloric gland zones, as well as in the jejunum (pā€‰<ā€‰0.001).

The number of F. gastrosuis bacteria did not differ between the H. suis-negative and -positive 2ā€“3Ā months old pigs, while a trend was observed towards higher number colonizing the Pars oesophagea of H. suis-positive 6ā€“8Ā months old pigs compared to the non-infected age-matched group, although this difference was not significant. The H. suis-infected adult sows showed significantly lower number of F. gastrosuis colonizing the Pars oesophagea compared to the non-infected age-matched group (pā€‰<ā€‰0.05).

Gene expression analysis of markers for inflammation and ulceration

Despite several attempts, including the use of different primer pairs, no mRNA expression was detected of the genes encoding claudin 4, NOX2, IL-6 and TNFĪ± in the porcine Pars oesophagea.

Compared to the H. suis-negative pigs, the mRNA expression of claudin 18, Hsp 72 and IL-8 was upregulated in the Pars oesophagea of H. suis-positive pigs, although either borderline or not significantly (pā€‰=ā€‰0.050, 0.211 and 0.139, respectively). In contrast, claudin 2, claudin 3 and CXCL2 gene expressions were either significantly or not significantly downregulated (pā€‰=ā€‰0.043, 0.093 and 0.065, respectively) (Additional files 9, 10).

The fold changes of claudin 18, Hsp 72, keratin 6A and ZO1 were more pronounced in pigs with a high number of F. gastrosuis bacteria per mg gastric tissue of the Pars oesophagea, since positive correlations were found between both. Conversely, the fold changes of claudin 3, BFGF, CXCL2 and IL-1Ī² were less pronounced in pigs with a high number of F. gastrosuis bacteria per mg gastric tissue due to negative correlations between both (Additional file 11).

Study 3: Cell-death inducing effects of F. gastrosuis on epithelial neoplastic cell lines

Fusobacterium gastrosuis was able to survive in an aerobic environment up to 12Ā h. After this incubation time, the viability of F. gastrosuis was significantly decreased and after 36Ā h incubation, viable F. gastrosuis bacteria could no longer be detected (data not shown).

Cell-death inducing effects on F. gastrosuis on MKN-7 cell line

Compared to the negative control, the viability of MKN-7 cells hardly decreased with an average of 3% after 24Ā h incubation with the different concentrations of F. gastrosuis lysate, but significantly decreased with an average of 32% and 65% after 36Ā h and 48Ā h incubation, respectively (pā€‰<ā€‰0.05) (FigureĀ 2A). After 2Ā h, 6Ā h and 12Ā h incubation with the different concentrations of viable F. gastrosuis bacteria, cell viability significantly decreased with an average of 23%, 24% and 34%, respectively, compared to the negative control (pā€‰<ā€‰0.05) (FigureĀ 2B).

FigureĀ 2
figure 2

Percentage of viable, early apoptotic, late apoptotic/necrotic MKN-7 cells after incubation with different concentrations of F. gastrosuis lysate (A) and viable F. gastrosuis bacteria (B). Data are shown as the average (4 strains, 3 replications per strain) percentages of viable, early apoptotic and late apoptotic/necrotic MKN-7 cells with standard deviation. A The cells were incubated for 24, 36 and 48 with 50, 200 and 500Ā Āµg F. gastrosuis lysate (4 strains). B The cells were incubated for 2, 6 and 12Ā h with 5, 20 and 50 MOI viable F. gastrosuis bacteria (4 strains).

Overall, the cell viability significantly decreased with increasing concentrations of F. gastrosuis lysate and MOI as well as with prolonged incubation time compared to the negative control (pā€‰<ā€‰0.05) (Additional files 12, 13 and 14). Although the observed decreases in cell viability were systematically caused by both an increase in apoptosis and late apoptosis/necrosis, the percentage of early apoptotic cells increased after short incubation time and/or low concentrations of F. gastrosuis lysate and MOI, while the percentage of late apoptotic/necrotic cells increased after longer incubation time and/or higher concentrations of F. gastrosuis lysate and MOI (Additional files 12, 13 and 14). Morphological features of apoptosis (i.e. plasma membrane blebbing, pyknosis) and necrosis (i.e. cell swelling, cytoplasmic vacuoles) reflecting these differences were observed on Hemacolor staining (Additional file 14).

Compared to F. gastrosuis lysate, more variation was observed between the four F. gastrosuis strains after incubation with viable F. gastrosuis bacteria. CDW1 lysate had the strongest effect on cell viability, CDW3 the weakest (pā€‰<ā€‰0.05). Conversely, viable CDW1 bacteria had the weakest effect on cell viability, CDW6 the strongest (pā€‰<ā€‰0.05).

In general, cell viability was significantly higher after incubation with the four F. gastrosuis strains compared to the positive control F. necrophorum subsp. necrophorum (pā€‰<ā€‰0.05). However, a significantly lower viability was observed for following conditions: at 50Ā Āµg lysate after 24Ā h, 36Ā h and 48Ā h for CDW1 and CDW3, and after 48Ā h also for CDW6 and CDW8; at 200Ā Āµg lysate CDW1, CDW6 and CDW8 after 48Ā h; at 50 MOI viable bacteria after 6Ā h and 12Ā h for CDW1 and after 12Ā h also for CDW6 and CDW8 (pā€‰<ā€‰0.05).

Cell-death inducing effects on F. gastrosuis on KYSE-450 cell line

The viability of KYSE-450 cells was significantly decreased with an average of 4%, 10%, 14% and 37% after 24Ā h, 36Ā h, 48Ā h and 72Ā h incubation, respectively, with the different concentrations of F. gastrosuis lysate compared to the negative control (pā€‰<ā€‰0.05) (FigureĀ 3A). Compared to the negative control, cell viability hardly decreased with an average of 3% after 2Ā h incubation with the different concentrations of viable F. gastrosuis bacteria, but significantly decreased with an average of 8% and 15% after 6Ā h and 12Ā h incubation, respectively (pā€‰<ā€‰0.05) (FigureĀ 3B).

FigureĀ 3
figure 3

Percentage of viable, early apoptotic, late apoptotic/necrotic KYSE-450 cells after incubation with different concentrations of F. gastrosuis lysate (A) and viable F. gastrosuis bacteria (B). Data are shown as the average (4 strains, 3 replications per strain) percentages of viable, early apoptotic and late apoptotic/necrotic KYSE-450 cells with standard deviation. A The cells were incubated for 24, 36, 48 and 72Ā h with 50, 200 and 500Ā Āµg F. gastrosuis lysate (4 strains). B The cells were incubated for 2, 6 and 12Ā h with 5, 20 and 50 MOI viable F. gastrosuis bacteria (4 strains). The percentage late apoptotic/necrotic cells (i.e. Annexin-V-FITC positive and PI positive) contains a small subpopulation of Annexin-V-FITC negative and PI positive cells defined as late necrotic cells. See Additional files 15, 16 for more detailed information.

Overall, the cell viability significantly decreased with increasing concentrations of F. gastrosuis lysate and MOI as well as with prolonged incubation time compared to the negative control (pā€‰<ā€‰0.05) (Additional files 15, 16 and 17). Although the observed decreases in cell viability were systematically caused by both an increase in early apoptosis and late apoptosis/necrosis, the percentage of early apoptotic cells increased after short incubation time and/or low concentrations of F. gastrosuis lysate and MOI, while the percentage of late apoptotic/necrotic cells increased after longer incubation time and/or higher concentrations of F. gastrosuis lysate and MOI. Morphological features of apoptosis (i.e. plasma membrane blebbing) and necrosis (i.e. cell swelling, cytoplasmic vacuoles) reflecting these differences were observed on Hemacolor staining (Additional file 17).

In contrast to the MKN-7 cell line, a small subpopulation of Annexin-V-FITC negative and PI positive KYSE-450 cells was detected after incubation with F. gastrosuis viable bacteria and lysates. This subpopulation was defined as late necrotic and increased with increasing concentrations of F. gastrosuis lysate and MOI as well as with prolonged incubation time compared to the negative control (pā€‰<ā€‰0.05) (Additional files 15, 16 and 17).

A similar effect on cell viability was seen after incubation with lysate obtained from the different F. gastrosuis strains. Conversely, more variance was seen after incubation with viable F. gastrosuis bacteria, where CDW1 had the weakest effect on cell viability and CDW6 the strongest (pā€‰<ā€‰0.05).

In general and in contrast to the MKN-7 cell line, the cell viability was lower after incubation with the four F. gastrosuis lysates compared to the positive control F. necrophorum subsp. necrophorum. A significantly lower viability was observed for the following conditions: at 50Ā Āµg lysate after 24Ā h and 72Ā h for CDW1, CDW3, CDW6 and CDW8, after 36Ā h and 48Ā h also for CDW6 and after 48Ā h also for CDW1 and CDW3; at 200Ā Āµg lysate after 24Ā h, 48Ā h and 72Ā h for CDW1 and CDW3 and after 72Ā h also CDW6 and CDW8; at 500Ā Āµg lysate after 24Ā h, 48Ā h and 72Ā h for CDW1 and after 48Ā h and 72Ā h also for CDW3 and CDW6 (pā€‰<ā€‰0.05).

In general and similar to the MKN-7 cell line, the cell viability was significantly higher after incubation with the four viable F. gastrosuis strains compared to the positive control F. necrophorum subsp. necrophorum bacteria (pā€‰<ā€‰0.05). However, a significantly lower viability was observed for following conditions: at five MOI viable bacteria after 6Ā h for CDW3 and CDW6 and after 12Ā h also for CDW6; at 20 MOI viable bacteria after 6Ā h and 12Ā h for CDW6; at 50 MOI after 6Ā h for CDW6 and after 12Ā h also for CDW3 (pā€‰<ā€‰0.05).

Properties of cell death inducing F. gastrosuis component(s)

Heat treatment completely abolished the cell-death inducing capacity of the four F. gastrosuis viable bacteria and lysates. Proteinase K treatment completely abolished the cell-death inducing capacity of viable F. gastrosuis bacteria, while no effects were seen for the trypsine and pronase treatments. Variable effects were seen for the paraformaldehyde treatment, namely a complete loss, partial loss (10ā€“30%) or no loss of cell-death inducing capacities, and this independent from the strains (TableĀ 2). F. gastrosuis bacteria remained viable after the different protease treatments and bacterial growth was similar to the untreated F. gastrosuis bacteria, while paraformaldehyde and heat treatment resulted in complete loss of viability.

TableĀ 2 Overview of the effect of heat, protease and formaldehyde treatment on cell-death inducing capacity of the 4 F. gastrosuis strains

Activation of the pyroptotic pathway

All samples tested negative for presence of pro- and active IL-1Ī² and caspase-1 proteins on western blot analysis, while bands were obtained for the positive controls (data not shown).

Study 4: Potential virulence associated genes in F. gastrosuis

A search for DEG homologous sequences in the proteome of F. gastrosuis yielded 1048 hits. Obtained hits have been reported to be involved in structural organization, nutrient uptake, pathogenesis, antibiotic resistance and other processes essential for the survival of F. gastrosuis (Additional file 18).

Using the VirulentPred tool, 922 genes, of which 370 encoded hypothetical proteins, were predicted to be associated with virulence. Of the entire F. gastrosuis proteome, 51 proteins showed a close match with Pfam domains associated with virulence, antibiotic resistance and immune-evasion (Additional file 18). About 162 proteins of the proteome showed significant hits against VFDB. The obtained hits have been reported to be involved in adhesion, invasion, immune-evasion, capsule formation, resistance to antibiotics and uptake of magnesium, zinc and iron (Additional file 18). Some examples of these potential virulence factors are: (i) hemolysinā€”lyses erythrocytes and creates anaerobic environment at the site of infection, stimulates the production of IL-1Ī² and TNFĪ±; (ii) ABC transportersā€”facilitates iron uptake, related with antibiotic resistance; (iii) lipooligosaccharides synthesisā€”involved in immune system evasion, attachment to epithelial tissue, mediator of the proinflammatory response; (iv) cytolysinā€”forms pores in cell membrane, induces apoptosis, promotes cellular invasion, triggers iNOS and cytokine release (Additional file 18). In total, 232 proteins showed a close match with virulence factors of other Fusobacterium spp. reported in the literature. Some examples of these matches are: (i) immunosuppressive protein (fipA)ā€”inhibits T-cell activation; (ii) haemagglutininā€”attachment to epithelial cells; (iii) outer membrane proteinsā€”mediate adherence to other pathogenic bacteria and host cells, suppress host immune system, induce cell death in lymphocytes; (iv) serine proteaseā€”degrades the extracellular matrix proteins fibrinogen and fibronectin as well as collagen I and collagen IV, contribute to damage of periodontal tissues, helps the evasion of the immune system. Finally, F. gastrosuis showed presence of genes associated with active invasion of host cells (Additional file 18).

Discussion

The gastro-intestinal microbiota plays an important role in host metabolism, nutrition, immunity and protection against pathogens [26, 27]. Disturbance of this delicate system is strongly correlated with the onset and progression of a wide range of pathologies [28]. Alterations in the gastric microbial community may play a role in porcine gastric lesion development [1]. However, due to technical limitations and the fact that the stomach was long considered inhospitable, limited information is available on the gastric microbiota composition of pigs [29]. For the first time, the microbiota of the upper, non-glandular part of the stomach of 6ā€“8Ā months old pigs was determined, as well as the impact on it of a natural H. suis infection.

Proteobacteria, Firmicutes, Bacteroidetes, Fusobacteria and Actinobacteria were the most abundant phyla present in the Pars oesophagea of 6ā€“8Ā months old pigs, irrespective of the H. suis status. These phyla have also been shown to dominate the gastric microbiota of weaned pigs [30, 31]. Nevertheless, their relative abundance may differ, as Proteobacteria was the most dominant phylum in the Pars oesophagea of 6ā€“8Ā months old pigs in the current study, while Firmicutes has been shown to dominate this stomach region in weaned pigs [31]. This apparent discrepancy between both studies might be explained by the age difference of the pigs. In suckling pigs, Lactobacillus spp., which form a major part of the Firmicutes phylum, colonize the upper, non-glandular part of the stomach in large numbers. After weaning, the diet changes from sow milk to hard food, resulting in a gradual decrease of Lactobacillus spp. [30]. Indeed, diet can greatly affect the microbial community, as bacterial enterotypes are known to cluster based on the dietary abundance of animal protein relative to carbohydrate [28]. Proteobacteria have also been shown to dominate stomach content as well as the cardiac, fundic and pyloric mucosa of weaned pigs, suggesting that other factors may affect the relative abundance of phyla, such as management and sampling sites or techniques [29]. Furthermore, the microbiota composition can also vary substantially within different herds and within pigs from the same herd, especially at the lower taxonomic levels. These factors complicate both the straightforward comparison of study results and the unequivocal definition of the gastric microbial community of healthy pigs [28, 32].

Fusobacteriaceae are part of the core microbiota of the soft palate tonsils in pigs [33, 34] and some members, such as F. necrophorum, have been associated with stomatitis and tooth root abscesses [35]. In the current study on the group of the 2ā€“3Ā months old pigs, the highest colonization rate of F. gastrosuis was found in the oral cavity, indicating that the oropharynx, and potentially its tonsils, may function as a natural reservoir for this bacterium. F. gastrosuis may enter the stomach through ingestion of food, water or saliva, after which it may opportunistically colonize the upper, non-glandular Pars oesophagea. Future studies are necessary to verify if F. gastrosuis does belong to the oral microbiota and/or if it may induce oropharyngeal pathologies. Interestingly, recent metagenomic studies have also shown presence of F. gastrosuis in the nasal and oral microbiota of dogs (Bernard Taminiau, 2018, personal communication) and in the faeces of humans [36], indicating that F. gastrosuis is able to colonize a wide range of mammalian hosts, as has been described for F. nucleatum and F. necrophorum [6]. Nevertheless, although unlikely, it might still be possible that F. gastrosuis was merely transient in these three hosts.

Recent studies indicate that H. suis infection plays an important role in porcine gastric ulceration, probably by affecting gastric acid secretion and influencing the gastric microbiota [4, 37]. In these studies, also high numbers of F. gastrosuis were detected in the gastric microbial community of H. suis-infected 6ā€“8Ā months old pigs with a downregulation of markers for gastric acid secretion [4, 5]. In the present study, 16S rRNA gene amplicon pyrosequencing revealed a higher relative abundance of F. gastrosuis in the Pars oesophageal microbiota of H. suis-infected 6ā€“8Ā months old pigs compared to non-infected pigs which was confirmed by RT-PCR. Analysis of many more pig stomachs of the same age group by RT-PCR showed a high F. gastrosuis colonization rate in the Pars oesophagea of H. suis-infected pigs as well, although the difference with non-infected pigs became less pronounced. This apparent discrepancy may have been caused by variation between herds or other factors such as diet, feeding strategy and management. Nonetheless, more stomachs should be investigated to confirm the increased F. gastrosuis colonization rate in H. suis-infected pigs. In marked contrast with our group of 6ā€“8Ā months old pigs, the numbers of F. gastrosuis bacteria did not differ between H. suis-infected and non-infected 2ā€“3Ā months old pigs. This may have been caused by the absence of gastric acid secretion alterations during the first phase of a H. suis infection i.e. in the 2ā€“3Ā months old pigs as previously reported [4]. The lower numbers of F. gastrosuis colonizing the Pars oesophagea of H. suis-infected adult sows compared to non-infected sows might be a consequence of the upregulation of gastric acid secretion during the chronic phase of infection i.e. in the H. suis-infected adult sows as again previously reported [4]. Combining the results of the current and our previous studies, we hypothesize that during the earlier phases of a H. suis infection (i.e. in the 6ā€“8Ā months old pigs), gastric acid secretion is downregulated, resulting in higher numbers of F. gastrosuis in the non-glandular part of the stomach which may facilitate the initiation of lesion development. During the more chronic phase of the infection (i.e. in the adult sows), gastric acid secretion is stimulated which may further aggravate lesion severity in the non-glandular part of the stomach which is not protected against acid due to the lack of mucus. Combined F. gastrosuis and H. suis infections in an experimental model complementing the observations obtained in naturally infected animals are, however, necessary to confirm or reject this working hypothesis.

Since few commercial porcine gastric cell line exists and as the epithelium of the oesophagus from humans and the Pars oesophagea of pigs is very similar [38], human-derived cell lines were used in the present study to further investigate in vitro the pathogenic traits of F. gastrosuis. Overall, cell death was clearly induced by both the viable F. gastrosuis bacteria and their lysate in the two selected cell lines. As lesions of the Pars oesophagea are characterized by the presence of swollen epithelial cells and necrotic debris [39, 40], these in vitro cell death data further highlight a potential role of F. gastrosuis in the development of porcine gastric ulceration. Finally, it would be interesting to evaluate the effect of F. gastrosuis on mucosal explants of the porcine Pars oesophagea, as these target species explants comprise several additional host cell and host cell-environment interactions lacking in the relatively simple gastric cell line set-up that may be important in the elucidation of porcine gastric ulceration development.

In contrast to the cell death mechanism reported for F. nucleatum in a human gingival cell line [21], the current study did not observe indications of pyroptotic pathway (i.e. pro-inflammatory programmed cell death) activation in epithelial cells incubated with F. gastrosuis, characterized by the absence of the pro-inflammatory caspase-1 and IL-1Ī² specific protein bands. It is possible that other caspases were activated through either the intrinsic or extrinsic apoptotic pathway. These complex cell-death inducing pathways needs to be further elucidated, for example by analyzing the expression of different caspase family members through flow cytometry [41].

The mechanisms by which F. gastrosuis induces host cell death are unknown. Our data suggest that heat labile proteins most likely play a role, as heat- and proteinase K-treated F. gastrosuis completely lost their cell death inducing potential. In contrast, pronase and trypsin treatments did not affect this cell death inducing capacity of F. gastrosuis, similar as described for other Fusobacterium spp. [42]. This may be explained by differences in enzymatic specificity, as trypsin specifically cleaves the peptide bonds at the carboxyl terminus of lysine and arginine residues, while proteinase K non-specifically cleaves at aromatic, aliphatic as well as hydrophobic amino acid residues [43]. The activity of pronase is also broad, yet the cell-death inducing capacity of F. gastrosuis was unaffected after the latter treatment. We hypothesize that other, currently unknown, factors may have further contributed here. For example, genes associated with capsule and biofilm formation as found in the proteome of F. gastrosuis may have contributed to this resistance against proteases such as pronase. As for the effect of formaldehyde treatment, widely used as fixation agent which slowly cross-links proteins in the bacterial cell wall, variable results were obtained. This indicates that cell-death inducing metabolites are, at least partly, associated with the bacterial surface and that their expression may be affected by environmental signals such as cell-contact. Nevertheless, this interpretation should be made with caution.

The current study identified that outer membrane proteins (OMPs) homologous to autotransporter secretion systems (type V) and/or ABC transporters are present in the proteome of F. gastrosuis. In analogy with a previous study, these OMPs might induce cell-death [44]. Indeed, bacterial secretion systems are associated with host cell death through active transport of effector metabolites [45] like hemolysin, leukotoxin and protease, all pore-forming proteins that either directly destroy host cells at high concentrations or activate apoptosis through induction of host plasma membrane changes at lower concentrations. The presence of serine protease and hemolysin homologs in the proteome of F. gastrosuis, as well as an increased percentage necrosis when epithelial cells were incubated with high F. gastrosuis concentrations support their potential role in the induction of cell death. Other putative virulence factors present in the proteome of F. gastrosuis may also have contributed to host cell death. Homologs of methionine gamma lyase, cystathionine gamma- and beta lyase were found to be associated with host tissue destruction through production of volatile sulfur products [46, 47], while gamma-glutamyl transferase [19] and genes associated with butyrate production [25] have both been associated with host cell death through the production of reactive oxygen species (ROS). Last but not least, some Fusobacterium spp. possess genes encoding a diverse set of adhesins and membrane-related proteins allowing to actively enter host cells even in the absence of tissue damage [6]. Analysis of the F. gastrosuis genome revealed the presence of such genes, supporting host colonization, host immune system evasion and deeper tissue penetration. Additional studies are necessary to determine if these putative virulence factors are expressed, secreted as well as functional. It would be interesting to create mutants lacking genes encoding these putative virulence-associated factors in order to investigate their roles in the pathogenic significance of F. gastrosuis.

In conclusion, high numbers of F. gastrosuis were demonstrated in the oropharynx and Pars oesophagea of pigs. It was also demonstrated that F. gastrosuis induces epithelial cell death and that genes are present in the genome of this bacterium with sequence similarity to genes encoding factors involved in adhesion, invasion and induction of cell death as well as immune evasion. We hypothesize that, in a gastric environment altered by H. suis, colonization and invasion of the Pars oesophagea and production of epithelial cell death inducing metabolites by F. gastrosuis cause ulceration. Experimental studies in pigs infected with H. suis and F. gastrosuis are necessary to confirm this hypothesis.

References

  1. De Witte C, Ducatelle R, Haesebrouck F (2018) The role of infectious agents in the development of porcine gastric ulceration. Vet J 236:56ā€“61

    ArticleĀ  Google ScholarĀ 

  2. Haesebrouck F, Pasmans F, Flahou B, Chiers K, Baele M, Meyns T, Decostere A, Ducatelle R (2009) Gastric helicobacters in domestic animals and nonhuman primates and their significance for human health. Clin Microbiol Rev 22:202ā€“223

    ArticleĀ  CASĀ  Google ScholarĀ 

  3. Zhang G, Ducatelle R, Mihi B, Smet A, Flahou B, Haesebrouck F (2016) Helicobacter suis affects the health and function of porcine gastric parietal cells. Vet Res 47:101

    ArticleĀ  Google ScholarĀ 

  4. De Witte C, Devriendt B, Flahou B, Bosschem I, Ducatelle R, Smet A, Haesebrouck F (2017) Helicobacter suis induces changes in gastric inflammation and acid secretion markers in pigs of different ages. Vet Res 48:34

    ArticleĀ  Google ScholarĀ 

  5. De Witte C, Flahou B, Ducatelle R, Smet A, De Bruyne E, Cnockaert M, Taminiau B, Daube G, Vandamme P, Haesebrouck F (2016) Detection, isolation and characterization of Fusobacterium gastrosuis sp. nov. colonizing the stomach of pigs. Syst Appl Microbiol 40:42ā€“50

    ArticleĀ  Google ScholarĀ 

  6. McGuire AM, Cochrane K, Griggs AD, Haas BJ, Abeel T, Zeng Q, Nice JB, Macdonald H, Birren BW, Berger BW, Allen-Vercoe E, Earl AM (2014) Evolution of invasion in a diverse set of Fusobacterium species. MBio 5:e01864ā€“e01914

    CASĀ  Google ScholarĀ 

  7. Ang MY, Heydari H, Jakubovics NS, Mahmud MI, Dutta A, Wee WY, Wong GJ, Mutha NVR, Tan SY, Choo SW (2014) FusoBase: an online Fusobacterium comparative genomic analysis platform. Database 2014:bau082

    ArticleĀ  Google ScholarĀ 

  8. Bosschem I, Flahou B, Van Deun K, De Koker S, Volf J, Smet A, Ducatelle R, Devriendt B, Haesebrouck F (2017) Species-specific immunity to Helicobacter suis. Helicobacter 22:e12375

    ArticleĀ  Google ScholarĀ 

  9. Rodriguez C, Taminiau B, BrĆ©vers B, Avesani V, Van Broeck J, Leroux A, Gallot M, Bruwier A, Amory H, DelmĆ©e M, Daube G (2015) Faecal microbiota characterisation of horses using 16 rdna barcoded pyrosequencing, and carriage rate of Clostridium difficile at hospital admission. BMC Microbiol 15:181ā€“194

    ArticleĀ  Google ScholarĀ 

  10. Haas BJ, Gevers D, Earl AM, Feldgarden M, Ward DV, Giannoukos G, Ciulla D, Tabbaa D, Highlander SK, Sodergren E, MethĆ© B, DeSantis TZ, Human Microbiome Consortium JF, Petrosino JF, Knight R, Birren BW (2011) Chimeric 16S rRNA sequence formation and detection in Sanger and 454-pyrosequenced PCR amplicons. Genome Res 21:494ā€“504

    ArticleĀ  CASĀ  Google ScholarĀ 

  11. Schloss PD, Westcott SL, Ryabin T, Hall JR, Hartmann M, Hollister EB, Lesniewski RA, Oakley BB, Parks DH, Robinson CJ, Sahl JW, Stres B, Thallinger GG, Van Horn DJ, Weber CF (2009) Introducing mothur: open-source, platform-independent, community-supported software for describing and comparing microbial communities. Appl Environ Microbiol 75:7537ā€“7541

    ArticleĀ  CASĀ  Google ScholarĀ 

  12. Pruesse E, Quast C, Knittel K, Fuchs BM, Ludwig W, Peplies J, Glƶckner FO (2007) SILVA: a comprehensive online resource for quality checked and aligned ribosomal RNA sequence data compatible with ARB. Nucleic Acids Res 35:7188ā€“7196

    ArticleĀ  CASĀ  Google ScholarĀ 

  13. Chao A, Shen T-J (2003) Nonparametric estimation of Shannonā€™s index of diversity when there are unseen species in sample. Environ Ecol Stat 10:429ā€“443

    ArticleĀ  Google ScholarĀ 

  14. Chao A, Bunge J (2002) Estimating the number of species in a stochastic abundance model. Biometrics 58:531ā€“539

    ArticleĀ  Google ScholarĀ 

  15. Arndt D, Xia J, Liu Y, Zhou Y, Guo AC, Cruz JA, Sinelnikov I, Budwill K, Nesbo CL, Wishart DS (2012) METAGENassist: a comprehensive web server for comparative metagenomics. Nucleic Acids Res 40:W88ā€“W95

    ArticleĀ  CASĀ  Google ScholarĀ 

  16. Hessing MJC, Geudeke MJ, Scheepens CJM, Tielen MJM, Schouten WGP, Wiepkema PR (1992) Mucosal lesions in the Pars oesophagea in pigsā€”prevalence and influence of stress. Tijdschr Diergeneeskd 117:445ā€“450

    CASĀ  PubMedĀ  Google ScholarĀ 

  17. Flahou B, Van Deun K, Pasmans F, Smet A, Volf J, Rychlik I, Ducatelle R, Haesebrouck F (2012) The local immune response of mice after Helicobacter suis infection: strain differences and distinction with Helicobacter pylori. Vet Res 43:75

    ArticleĀ  CASĀ  Google ScholarĀ 

  18. Nygard A-B, JĆørgensen CB, Cirera S, Fredholm M (2007) Selection of reference genes for gene expression studies in pig tissues using SYBR green qPCR. BMC Mol Biol 8:67

    ArticleĀ  Google ScholarĀ 

  19. Flahou B, Haesebrouck F, Chiers K, Van Deun K, De Smet L, Devreese B, Vandenberghe I, Favoreel H, Smet A, Pasmans F, Dā€™Herde K, Ducatelle R (2011) Gastric epithelial cell death caused by Helicobacter suis and Helicobacter pylori Ī³-glutamyl transpeptidase is mainly glutathione degradation-dependent. Cell Microbiol 13:1933ā€“1955

    ArticleĀ  CASĀ  Google ScholarĀ 

  20. Broekaert N, Devreese M, Demeyere K, Berthiller F, Michlmayr H, Varga E, Adam G, Meyer E, Croubels S (2016) Comparative in vitro cytotoxicity of modified deoxynivalenol on porcine intestinal epithelial cells. Food Chem Toxicol 95:103ā€“109

    ArticleĀ  CASĀ  Google ScholarĀ 

  21. Bui FQ, Johnson L, Roberts J, Hung S-C, Lee J, Atanasova KR, Huang P-R, Yilmaz Ɩ, Ojcius DM (2016) Fusobacterium nucleatum infection of gingival epithelial cells leads to NLRP3 inflammasome-dependent secretion of IL-1B and the danger signals ASC and HMGB1. Cell Microbiol 18:970ā€“981

    ArticleĀ  CASĀ  Google ScholarĀ 

  22. Lamote I, Demeyere K, Notebaert S, Burvenich C, Meyer E (2007) Flow cytometric assessment of estrogen receptor Ī² expression in bovine blood neutrophils. J Immunol Methods 323:88ā€“92

    ArticleĀ  CASĀ  Google ScholarĀ 

  23. Kumar A, Thotakura PL, Tiwary BK, Krishna R (2016) Target identification in Fusobacterium nucleatum by subtractive genomics approach and enrichment analysis of host-pathogen proteinā€“protein interactions. BMC Microbiol 16:84

    ArticleĀ  Google ScholarĀ 

  24. Kapatral V, Anderson I, Ivanova N, Reznik G, Los T, Lykidis A, Bhattacharyya A, Bartman A, Gardner W, Grechkin G, Zhu L, Vasieva O, Chu L, Kogan Y, Chaga O, Goltsman E, Bernal A, Larsen N, Dā€™Souza M, Walunas T, Pusch G, Haselkorn R, Fonstein M, Kyrpides N, Overbeek R (2002) Genome sequence and analysis of the oral bacterium Fusobacterium nucleatum strain ATCC 25586. J Bacteriol 184:2005ā€“2018

    ArticleĀ  CASĀ  Google ScholarĀ 

  25. Karpathy SE, Qin X, Gioia J, Jiang H, Liu Y, Petrosino JF, Yerrapragada S, Fox GE, Haake SK, Weinstock GM, Highlander SK (2007) Genome sequence of Fusobacterium nucleatum subspecies polymorphumā€”a genetically tractable Fusobacterium. PLoS One 2:e659

    ArticleĀ  Google ScholarĀ 

  26. Metzler-Zebeli BU, Mann E, Schmitz-Esser S, Wagner M, Ritzmann M, Zebeli Q (2013) Changing dietary calciumā€“phosphorus level and cereal source selectively alters abundance of bacteria and metabolites in the upper gastrointestinal tracts of weaned pigs. Appl Environ Microbiol 79:7264ā€“7272

    ArticleĀ  CASĀ  Google ScholarĀ 

  27. Castillo M, Skene G, Roca M, Anguita M, Badiola I, Duncan SH, Flint HJ, MartĆ­n-OrĆŗe SM (2007) Application of 16S rRNA gene-targetted fluorescence in situ hybridization and restriction fragment length polymorphism to study porcine microbiota along the gastrointestinal tract in response to different sources of dietary fibre. FEMS Microbiol Ecol 59:138ā€“146

    ArticleĀ  CASĀ  Google ScholarĀ 

  28. Cho I, Blaser MJ (2012) The human microbiome: at the interface of health and disease. Nat Rev Genet 13:260ā€“270

    ArticleĀ  CASĀ  Google ScholarĀ 

  29. Motta V, Trevisi P, Bertolini F, Ribani A, Schiavo G, Fontanesi L, Bosi P (2017) Exploring gastric bacterial community in young pigs. PLoS One 12:e0173029

    ArticleĀ  Google ScholarĀ 

  30. Su Y, Yao W, Perez-Gutierrez ON, Smidt H, Zhu W-Y (2008) Changes in abundance of Lactobacillus spp. and Streptococcus suis in the stomach, jejunum and ileum of piglets after weaning. FEMS Microbiol Ecol 66:546ā€“555

    ArticleĀ  CASĀ  Google ScholarĀ 

  31. Mann E, Schmitz-Esser S, Zebeli Q, Wagner M, Ritzmann M, Metzler-Zebeli BU (2014) Mucosa-associated bacterial microbiome of the gastrointestinal tract of weaned pigs and dynamics linked to dietary calciumā€“phosphorus. PLoS One 9:e86950

    ArticleĀ  Google ScholarĀ 

  32. Kernaghan S, Bujold AR, MacInnes JI (2012) The microbiome of the soft palate of swine. Anim Health Res Rev 13:110ā€“120

    ArticleĀ  Google ScholarĀ 

  33. Lowe BA, Marsh TL, Isaacs-Cosgrove N, Kirkwood RN, Kiupel M, Mulks MH (2012) Defining the core microbiome of the microbial communities in the tonsils of healthy pigs. BMC Microbiol 12:20

    ArticleĀ  CASĀ  Google ScholarĀ 

  34. Pena Cortes LC, LeVeque RM, Funk J, Marsh TL, Mulks MH (2018) Development of the tonsillar microbiome in pigs from newborn through weaning. BMC Microbiol 18:35

    ArticleĀ  Google ScholarĀ 

  35. Zimmerman JJ, Karriker LA, Ramirez A, Schwartz KJ, Stevenson GW (eds) (2012) Diseases of Swine, 10th edn. Wiley-Blackwell, Oxford, pp 255ā€“257

    Google ScholarĀ 

  36. De Paepe K, Verspreet J, Verbeke K, Raes J, Courtin CM, Van de Wiele T (2018) Insoluble wheat bran stimulates human gut microbiota resilience and propionate recovery following a clindamycin induced dysbiosis in the stimulator the human intestinal microbial ecosystem. Environ Microbiol 20:3406ā€“3426

    ArticleĀ  Google ScholarĀ 

  37. Sapierzyński R, Fabisiak M, Kizerwetter-Swida M, Cywińska A, Sapierzynski R, Fabisiak M, Kizerwetter-Swida M, Cywinska A (2007) Effect of Helicobacter sp. infection on the number of antral gastric endocrine cells in swine. Pol J Vet Sci 10:65ā€“70

    PubMedĀ  Google ScholarĀ 

  38. Ziegler A, Gonzalez L, Blikslager A (2016) Large animal models: the key to translational discovery in digestive disease research. Cell Mol Gastroenterol Hepatol 2:716ā€“724

    ArticleĀ  Google ScholarĀ 

  39. Queiroz DMD, Rocha GA, Mendes EN, DeMoura SB, DeOliveira AMR, Miranda D (1996) Association between Helicobacter and gastric ulcer disease of the pars esophagea in swine. Gastroenterology 111:19ā€“27

    ArticleĀ  CASĀ  Google ScholarĀ 

  40. Thomson JR, Friendship RM (2012) The stomach: gastric ulceration. In: Zimmerman JJ, Karriker LA, Ramirez A, Schwartz KJ, Stevenson GW (eds) Diseases of Swine, 10th edn. Wiley-Blackwell, Oxford, pp 208ā€“211

    Google ScholarĀ 

  41. Fink SL, Cookson BT (2005) Apoptosis, pyroptosis, and necrosis: mechanistic description of dead and dying eukaryotic cells. Infect Immun 73:1907ā€“1916

    ArticleĀ  CASĀ  Google ScholarĀ 

  42. Kolenbrander PE, Parrish KD, Andersen RN, Greenberg EP (1995) Intergeneric coaggregation of oral Treponema spp. with Fusobacterium spp. and intrageneric coaggregation among Fusobacterium spp. Infect Immun 63:4584ā€“4588

    CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  43. MĆ³tyĆ”n JA, TĆ³th F, TőzsĆ©r J (2013) Research applications of proteolytic enzymes in molecular biology. Biomolecules 3:923ā€“942

    ArticleĀ  Google ScholarĀ 

  44. Kaplan CW, Ma X, Paranjpe A, Jewett A, Lux R, Kinder-Haake S, Shi W (2010) Fusobacterium nucleatum outer membrane proteins Fap2 and RadD induce cell death in human lymphocytes. Infect Immun 78:4773ā€“4778

    ArticleĀ  CASĀ  Google ScholarĀ 

  45. Henderson IR, Nataro JP (2001) Virulence functions of autotransporter proteins. Infect Immun 69:1231ā€“1243

    ArticleĀ  CASĀ  Google ScholarĀ 

  46. Nagaraja TG, Chengappa MM (1998) Liver abscesses in feedlot cattle: a review. J Anim Sci 76:287ā€“298

    ArticleĀ  CASĀ  Google ScholarĀ 

  47. Chukwu EE, Nwaokorie FO, Coker AO (2014) A review of Fusobacterium necrophorum infections in humans. Br Microbiol Res J 4:480ā€“496

    ArticleĀ  Google ScholarĀ 

Download references

Acknowledgements

This work was supported by a grant from the Special Research Fund of Ghent University (BOF), Ghent, Belgium (Grant No. 01D20414).

Author information

Authors and Affiliations

Authors

Contributions

CDW, FH, KD and EM participated in the design of the study. CDW and SDB carried out the experiments, analyzed the data and drafted the manuscript. FH, RD, BT, GD, KD and EM coordinated the study and participated in the design of the study, analysis of the data and drafting of the manuscript. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Chloƫ De Witte.

Ethics declarations

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Additional files

13567_2019_656_MOESM1_ESM.docx

Additional file 1. List of primers used in quantitative RT-PCR for gene expression analysis of porcine housekeeping genes and markers for inflammation, hyperkeratosis and ulceration.

Additional file 2.

Gating strategy of the MKN-7 cell line. (A) FSC-A/SSC-A represents the distribution of cells in the light scatter based on their size and intracellular complexity, respectively. The cells of interest are gated excluding debris. (B) FSC-A/FSC-H allows discrimination between single cells and doublets, single cells are gated. (C) Gain settings and compensation matrix. (D-E) FITC-A/PE-A identifies the selective subpopulations: viable (Annexin-V-FITC negative, PI negative), early apoptotic (Annexin-V-FITC positive, PI negative) and late apoptotic/necrotic (Annexin-V-FITC positive, PI positive) cells.

Additional file 3.

Gating strategy of the KYSE-450 cell line. (A) FSC-A/SSC-A represents the distribution of cells in the light scatter based on their size and intracellular complexity, respectively. The cells of interest are gated excluding debris. (B) FSC-A/FSC-H allows discrimination between single cells and doublets, single cells are gated. (C) Gain settings and compensation matrix. (D-E) FITC-A/PE-A identifies the selective subpopulations: viable (Annexin-V-FITC negative, PI negative), early apoptotic (Annexin-V-FITC positive, PI negative), late apoptotic/necrotic (Annexin-V-FITC positive, PI positive) and late necrotic (Annexin-V-FITC negative, PI positive) cells.

Additional file 4.

Bacterial community compositions present in the Pars oesophagea of each individual pig. The cumulated histograms show the relative abundance of the identified taxa at phylum (A), family (B) and genus (C) level. At family and genus level, taxa with a relative abundance <1% are merged in the category ā€œothersā€. 1ā€“10 = H. suis-negative pigs, 11ā€“20 = H. suis-positive pigs. The unclassified populations correspond to defined groups of the genus level for which a taxonomical classification assignation to the genus cannot be attributed. These populations are therefore labelled with the first defined superior hierarchical taxonomic level followed by ā€œ_unclassifiedā€ to prevent confusion.

Additional file 5.

Overview of the bacterial richness (A), diversity (B) and evenness (C) of the Pars oesophagea of H. suis-positive and -negative pigs. The data are represented as scatter plots: each dot represents a pig, the middle line represents the median and the whiskers represent the standard error or the mean.

Additional file 6.

The number of F. gastrosuis bacteria in the different stomach regions of H. suis-positive and -negative 2ā€“3 months old pigs (A), 6ā€“8 months old pigs (B), adult sows (C) and the pigs used for the metagenomics study (D). Data are shown as log10 values of the average number of F. gastrosuis bacteria per mg tissue with standard deviation. Statistical differences were calculated using the non-parametric Kruskal-Wallis H test. *, p < 0.05.

Additional file 7.

The number of F. gastrosuis bacteria in the oral cavity and gastro-intestinal tract of 2ā€“3 months old pigs. Data are shown as log10 values of the average number of F. gastrosuis bacteria per mg tissue with standard deviation. Statistical differences were calculated using the non-parametric Kruskal-Wallis H test. *, p < 0.05; **, p < 0.001 significant differences between the regions.

Additional file 8.

The number of F. gastrosuis bacteria in the different stomach regions of 2ā€“3 months old pigs, 6ā€“8 months old pigs and adult sows. Data are shown as log10 values of the average number of F. gastrosuis bacteria per mg tissue with standard deviation. Statistical differences were calculated using the non-parametric Kruskal-Wallis H test. *, p < 0.05; **, p < 0.001 significant differences between the stomach regions. Significant differences between the age groups are indicated with brackets.

Additional file 9.

General overview of gene expression analysis of markers for inflammation and ulceration in the Pars oesophagea of H. suis-negative and -positive pigs. The data are presented as fold changes in gene expression normalized to 3 reference genes and relative to a H. suis-negative control pigs. The fold changes are shown as means with the standard error of the mean. Statistical differences were calculated using the non-parametric Kruskal-Wallis H test. *, p < 0.05; significant differences between the H. suis-positive pigs and -negative pigs.

Additional file 10.

Overview of relative fold changes of altered markers for inflammation and ulceration in the Pars oesophagea of H. suis-positive pigs. The data are presented as fold changes in gene expression normalized to 3 reference genes and relative to the H. suis-negative pigs. The fold changes are shown as means with the standard error of the mean. Statistical differences were calculated using the non-parametric Kruskal-Wallis H test. A p-value lower than 0.05 is considered to be significant.

Additional file 11.

Correlation between the number of colonizing F. gastrosuis bacteria per mg gastric tissue and the expression of markers for inflammation and ulceration. The data are presented as a scatter plot: each dot represents the individual data of a pig. The trendline shows the relationship between the relative mRNA expression of a marker for inflammation/ulceration and the log10 values of the number of F. gastrosuis bacteria per mg gastric tissue. y = equation of the trendline. r = Pearson correlation coefficient, calculated using SPSS Statistics 24. A r-value close to 1 indicates a strong, positive correlation, whereas a r-value of āˆ’1 indicates a strong, negative correlation. P-values lower than 0.05 are considered to be significant.

Additional file 12.

Percentage of viable, early apoptotic and late apoptotic/necrotic MKN-7 cells after incubation with F. gastrosuis lysate. Data are shown as the average (nĀ =Ā 3) percentages of viable (green), early apoptotic (red) and late apoptotic/necrotic (blue) MKN-7 cells with standard deviation. The cells were incubated for 24 (A), 36 (B) and 48h (C) with 50 Āµg, 200 Āµg and 500 Āµg F. gastrosuis lysate (4 strains, CDW1, 3, 6 and 8) and F. necrophorum subsp. necrophorum (Fnn) as positive control. * Significant differences between the negative control and cells incubated with each bacterial lysate (p < 0.05).

Additional file 13.

Percentage of viable, early apoptotic and late apoptotic/necrotic MKN-7 cells after incubation with viable F. gastrosuis bacteria. Data are shown as the average (nĀ =Ā 3) percentages of viable (green), early apoptotic (red) and late apoptotic/necrotic (blue) MKN-7 cells with standard deviation. The cells were incubated for 2 (A), 6 (B) and 12h (C) with 5 MOI, 20 MOI and 50 MOI viable F. gastrosuis bacteria (4 strains, CDW1, 6, 6 and 8) and F. necrophorum subsp. necrophorum (Fnn) as positive control. * Significant differences between the negative control and cells incubated with each bacterial strain (p < 0.05).

Additional file 14.

Visualization of MKN-7 cellular morphology using hemacolor staining and detection of early apoptotic and late apoptotic/necrotic cells using flow cytometry. (A-D) Hemacolor staining of MKN-7 cells incubated (A) without F. gastrosuis for 48h and (B-D) with 500 Āµg F. gastrosuis strain CDW1 incubated for (B) 24Ā h, (C) 36Ā h and (D) 48h. Following morphologic features can be seen: plasma membrane blebbing (white arrow), cell swelling (white star), pyknosis (black arrow), cytoplasmic vacuoles (black star). Original magnification x400, scale bar represents 10 Āµm. (E-L) Representative population plots displaying viable (green, Annexin-V-FITX negative, PI negative), early apoptotic (red, Annexin-V-FITX positive, PI negative), late apoptotic/necrotic (blue, Annexin-V-FITX positive, PI positive) cells of MKN-7 cells incubated (E) without F. gastrosuis lysate for 48Ā h; (F-H) with 500 Āµg F. gastrosuis strain CDW1 for (F) 24Ā h; (G) 36Ā h and (H) 48Ā h; (I) without viable F. gastrosuis bacteria for 12h; (J-L) with 50 MOI F. gastrosuis bacteria strain CDW8 for (J) 2h; (K) 6h and (L) 12h. Y-axis: propidium iodide (PE) signal intensity; X-axis: Annexin-V-fluorescein isothiocyanate (FITC) signal intensity. The percentage of population plots is presented in the corresponding gate.

Additional file 15.

Percentage of viable, early apoptotic, late apoptotic/necrotic and late necrotic KYSE-450 cells after incubation with F. gastrosuis lysate. Data are shown as the average (nĀ =Ā 3) percentages of viable (green), early apoptotic (red), late apoptotic/necrotic (blue) and late necrotic (orange) KYSE-450 cells with standard deviation. The cells were incubated for 24 (A), 36 (B), 48 (C) and 72Ā h (D) with 50 Āµg, 200 Āµg and 500 Āµg F. gastrosuis lysate (4 strains, CDW1, 3,6 and 8) and F. necrophorum subsp. necrophorum (Fnn) as positive control. * Significant differences between the negative control and cells incubated with each bacterial lysate (p < 0.05).

Additional file 16.

Percentage of viable, early apoptotic, late apoptotic/necrotic and late necrotic KYSE-450 cells after incubation with viable F. gastrosuis bacteria. Data are shown as the average (nĀ =Ā 3) percentages of viable (green), early apoptotic (red), late apoptotic/necrotic (blue) and late necrotic (orange) KYSE-450 cells with standard deviation. The cells were incubated for 2 (A), 6 (B) and 12Ā h (C) with 5 MOI, 20 MOI and 50 MOI viable F. gastrosuis bacteria (4 strains, CDW1, 3, 6 and 8) and F. necrophorum subsp. necrophorum (Fnn) as positive control. * Significant differences between the negative control and cells incubated with each bacterial strain (p < 0.05).

Additional file 17.

Visualization of KYSE-450 cellular morphology using hemacolor staining and detection of early apoptotic and late apoptotic/necrotic cells using flow cytometry. (A-D) Hemacolor staining of KYSE-450 cells incubated (A) without F. gastrosuis for 48Ā h and (B-D) with 500 Āµg F. gastrosuis strain CDW1 incubated for (B) 36Ā h, (C) 48Ā h and (D) 72Ā h. Following morphologic features can be seen: plasma membrane blebbing (white arrow), cell swelling (white star), cytoplasmic vacuoles (black star). Original magnification x400, scale bar represents 10 Āµm. (E-M) Representative population plots displaying viable (green, Annexin-V-FITC negative, PI negative), early apoptotic (red, Annexin-V-FITC positive, PI negative), late apoptotic/necrotic (blue, Annexin-V-FITC positive, PI positive) cells and late necrotic (orange, Annexin-V-FITC negative, PI positive) of KYSE-450 cells incubated (E) without F. gastrosuis lysate for 72Ā h; (F-I) with 500 Āµg F. gastrosuis strain CDW1 for (F) 24Ā h; (G) 36Ā h; (H) 48Ā h and (I) 72Ā h; (J) without viable F. gastrosuis bacteria for 12Ā h; (K-M) with 50 MOI F. gastrosuis bacteria strain CDW8 for (K) 2Ā h; (L) 6h and (M) 12Ā h. Y-axis: propidium iodide (PE) signal intensity; X-axis: Annexin-V-fluorescein isothiocyanate (FITC) signal intensity. The percentage of population plots is presented in the corresponding gate.

13567_2019_656_MOESM18_ESM.xlsx

Additional file 18. Overview of potential virulence associated genes in F. gastrosuis using DEG, VirulentPred tool, Pfam and literature.

Rights and permissions

Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

De Witte, C., Demeyere, K., De Bruyckere, S. et al. Characterization of the non-glandular gastric region microbiota in Helicobacter suis-infected versus non-infected pigs identifies a potential role for Fusobacterium gastrosuis in gastric ulceration. Vet Res 50, 39 (2019). https://doi.org/10.1186/s13567-019-0656-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13567-019-0656-9