[go: up one dir, main page]
More Web Proxy on the site http://driver.im/
You seem to have javascript disabled. Please note that many of the page functionalities won't work as expected without javascript enabled.
 
 
Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (175)

Search Parameters:
Keywords = Streptococcus suis

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
18 pages, 452 KiB  
Article
Specialized Feed-Additive Blends of Short- and Medium-Chain Fatty Acids Improve Sow and Pig Performance During Nursery and Post-Weaning Phase
by Sandra Villagómez-Estrada, Diego Melo-Durán, Sandra van Kuijk, José F. Pérez and David Solà-Oriol
Animals 2024, 14(24), 3692; https://doi.org/10.3390/ani14243692 (registering DOI) - 20 Dec 2024
Abstract
The present study investigates the impact of supplementing diets with a synergistic blend of short- and medium-chain fatty acids (SCFAs-MCFAs) during the peripartum and lactation phases on early microbial colonization and the subsequent growth performance of newborn pigs. The experiment involved 72 sows [...] Read more.
The present study investigates the impact of supplementing diets with a synergistic blend of short- and medium-chain fatty acids (SCFAs-MCFAs) during the peripartum and lactation phases on early microbial colonization and the subsequent growth performance of newborn pigs. The experiment involved 72 sows and their litters, with a follow-up on 528 weaned pigs. Sows were fed either a control diet or a diet supplemented with SCFAs-MCFAs and the pigs were monitored for their growth performance and microbial populations. Subsequently, selected weaned pigs were allotted to an SCFAs-MCFAs diet according to the maternal diet. Results showed that SCFAs-MCFAs supplementation led to reduced backfat loss in sows and improved pig weight and uniformity at weaning (p < 0.05). Additionally, suckling pigs exhibited significant shifts in gut microbiota, including increased lactic acid bacteria and reduced Streptococcus suis populations (p < 0.05). Although there was no influence of maternal diet on pig growth after weaning, there was a modulation on bacterial populations at 7 and 35 days post-weaning. Pigs fed SCFAs-MCFAs demonstrated improved feed efficiency with notable reductions in E. coli and Streptococcus suis counts. The findings suggest that maternal dietary supplementation with SCFAs-MCFAs can positively influence both sow and pig performance, offering a potential strategy to enhance productivity and health in the commercial swine production. Full article
(This article belongs to the Section Pigs)
10 pages, 1989 KiB  
Article
Three New Dipeptide and Two New Polyketide Derivatives from the Mangrove-Derived Fungus Talaromyces sp.: Antioxidant Activity of Two Isolated Substances
by Zhihao Zeng, Jian Cai, Yi Chen, Xinlong Li, Chunmei Chen, Yonghong Liu, Lalith Jayasinghe and Xuefeng Zhou
Mar. Drugs 2024, 22(12), 559; https://doi.org/10.3390/md22120559 - 14 Dec 2024
Viewed by 450
Abstract
Five new metabolites, including three cyclic dipeptide derivatives (13) and two new polyketides (1011), together with nine known ones (49 and 1215), were isolated from the mangrove-sediments-derived fungus Talaromyces [...] Read more.
Five new metabolites, including three cyclic dipeptide derivatives (13) and two new polyketides (1011), together with nine known ones (49 and 1215), were isolated from the mangrove-sediments-derived fungus Talaromyces sp. SCSIO 41431. Their structures were determined using detailed NMR, MS spectroscopic analyses, and quantum chemical calculations. X-ray single-crystal diffraction analysis of 1 was described. Compounds 1315 demonstrated activity against Staphylococcus aureus, with MIC values ranging from 25 to 50 µg/mL. Compound 9 showed activity against Escherichia coli, Streptococcus suis, and Erysipelothrix rhusiopathiae, with an MIC value of 100 µg/mL. In addition, compounds 1 and 12 showed DPPH radical scavenging activity, with the EC50 of 27.62 and 29.34 µg/mL, compared to the positive control (ascorbic acid, EC50, 12.74 µg/mL). Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Structures of compounds <b>1</b>–<b>15</b>.</p>
Full article ">Figure 2
<p>Key HMBC, <sup>1</sup>H-<sup>1</sup>H COSY, and NOESY correlations of <b>1</b>–<b>3</b> and <b>10</b>–<b>11</b>.</p>
Full article ">Figure 3
<p>Experimental and calculated ECD spectra of compounds <b>1</b>–<b>3</b>.</p>
Full article ">Figure 4
<p>X-ray single-crystal diffraction of compound <b>1</b>.</p>
Full article ">Figure 5
<p>(<b>A</b>) DPPH radical scavenging activity of the compounds <b>1</b>–<b>15</b> (50 µg/mL). All experiments were performed at least three times. The data are presented as the mean ± SD of representative experiments. Statistical significance was determined with one-way ANOVA. * <span class="html-italic">p</span> &lt; 0.05, *** <span class="html-italic">p</span> &lt; 0.001, and **** <span class="html-italic">p</span> &lt; 0.0001 were considered statistically significant. (<b>B</b>) Compounds <b>1</b> and <b>12</b> of DPPH scavenging activity of EC<sub>50</sub>.</p>
Full article ">
19 pages, 7755 KiB  
Article
Study on the Effect of Phillyrin on Streptococcus suis In Vivo and In Vitro
by Fangyan Yuan, Lihan Zheng, Mengzhe Wang, Wei Liu, Xiaoyue Li, Ting Gao, Rui Guo, Zewen Liu, Keli Yang, Chang Li, Qiong Wu, Jiajia Zhu, Yongxiang Tian and Danna Zhou
Biomolecules 2024, 14(12), 1542; https://doi.org/10.3390/biom14121542 - 1 Dec 2024
Viewed by 473
Abstract
As a zoonotic pathogen, S. suis serotype 2 (SS2) can cause severe diseases in both pigs and humans, and develop resistance to antibiotics. Plant natural compounds are regarded as promising alternatives to conventional antibiotics. Phillyrin is the major bioactive components of Chinese herbal [...] Read more.
As a zoonotic pathogen, S. suis serotype 2 (SS2) can cause severe diseases in both pigs and humans, and develop resistance to antibiotics. Plant natural compounds are regarded as promising alternatives to conventional antibiotics. Phillyrin is the major bioactive components of Chinese herbal medicine Forsythia suspensa. In this study, we explored the activity and action mechanism of phillyrin against SS2. The results showed that phillyrin could disrupt membrane integrity, destroy intracellular structures, and increase the exosmosis of DNA. Results of PCR revealed that phillyrin affected bacterial-virulence-related genes’ expression levels. Meanwhile, phillyrin significantly decreased the adhesion activity, inhibited lactate dehydrogenase (LDH) secretion, and reduced biofilm formation of SS2 in Newborn pig trachea epithelial (NPTr) cells. Furthermore, phillyrin protected tight junction protein of NPTr cells from SS2. We reported that phillyrin (0.1 mg/kg) treatment after bacterial challenge significantly improved the survival rate, ameliorated pulmonary inflammation, and inhibited the accumulation of multiple cytokines (IL-1, IL-6, IL-8, and TNF-α). Molecular docking showed that phillyrin had a good binding activity with the Ala88 and Asp111 of suilysin (SLY), one of the most important virulence factors of SS2. Collectively, phillyrin possesses antibacterial and anti-inflammatory activities, and is a promising candidate for preventing SS2 infection. Full article
(This article belongs to the Section Natural and Bio-derived Molecules)
Show Figures

Figure 1

Figure 1
<p>Antibacterial activity of phillyrin against SC19 in vitro. (<b>A</b>) Chemical structure of phillyrin. (<b>B</b>) Analysis of phillyrin by Mass Spectrum. (<b>C</b>) The growth curve of SC19 affected by phillyrin was determined by OD600 nm at the indicated times. (<b>D</b>) The growth curve of SC19 affected by phillyrin was determined by CFU counts at the indicated times. (ns, <span class="html-italic">p</span> &gt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.001; **** <span class="html-italic">p</span>&lt; 0.0001).</p>
Full article ">Figure 2
<p>Scanning electron microscopy (SEM) and transmission electron microscopy (TEM) of SS2 after treatment with phillyrin. (<b>A</b>) TEM observation of phillyrin-untreated SC19; the bar at the bottom right means 1 µm. (<b>B</b>) TEM observation of phillyrin-treated SC19; the bar at the bottom right means 1 µm. (<b>C</b>) SEM observation of phillyrin-untreated SC19; the bar at the bottom right means 300 nm. (<b>D</b>) SEM observation of phillyrin-treated SC19; the bar at the bottom right means 300 nm. Bacterial cell presented shrinkage, cell size reduction, and perforation of the cell surface. Control cells without treatment appeared with normal shape.</p>
Full article ">Figure 3
<p>Phillyrin signifcantly improves the DNA exosmosis of SS2. SC19 was cultured to the logarithmic stage, blended to 107 CFU/mL and treated with phillyrin at 64 µg/mL and 128 µg/mL respectively.SC19 without phillyrin were used as a negative control. Supernatants were collected after co-incubated for 0, 1, 2, 4, 6 and 8 h, and the DNA content was determinated with a microspectrophotometer.</p>
Full article ">Figure 4
<p>Phillyrin inhibits the secretion of hemolysin and biofilm information of SS2. (<b>A</b>,<b>B</b>) Hemolytic activity analysis of SC19 affected by PHI. SC19 was cultured to the logarithmic stage, treated with different concentrations (1/4 MIC, 1/2 MIC, MIC, 1/4 MBC and 1/2 MBC) of phillyrin. After incubation with defibrillated sheep blood, the hemolysin was collected and detected by a spectrophotometer. (<b>C</b>,<b>D</b>) Biofilm formation analysis of SC19 affected by phillyrin. The biofilms of the SC19 with phillyrin at MIC and MBC (512 µg/mL) were stained by the crystal violet method and OD600 was determined after it dissolved. ns, <span class="html-italic">p</span>&gt;0.05; * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.001; **** <span class="html-italic">p</span> &lt; 0.0001 compared to the respective control.</p>
Full article ">Figure 5
<p>Effect of phillyrin on NPTr cells. (<b>A</b>) Analysis of cytotoxicity of phillyrin by double fluorescence staining of NPTr cells. (<b>B</b>) Cell viability was determined by a CCK8 assay. (<b>C</b>) Analysis of the cytotoxic activity of phillyrin by standard LDH release assay. ns, <span class="html-italic">p</span> &gt; 0.05. (<b>D</b>) Results were expressed as the percentage of LDH release compared to the non-phillyrin cells. Error bars represent the standard deviation of three independent experiments performed in triplicate. **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 6
<p>Adhesion ability analysis of SS2 affected by phillyrin. NPTr cells were treated by SC19 with phillyrin, and then the number of adherent bacteria was measured. The experiment was performed in triplicate and repeated at least three times. * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 7
<p>Phillyrin reduces damage of cell tight junction protein by SS2. (<b>A</b>) Expression of cell tight junction protein ZO-1 was detected by Western blot. (<b>B</b>) Western blot analysis of phillyrin treatment on the tight junction protein ZO-1 of NPTr cells infected by SC19 with or without 64 µg/mL phillyrin. The height of the bars indicates the mean values for the relative expression data ± SEM (**** <span class="html-italic">p</span> &lt; 0.0001). Original images of (<b>A</b>) can be found in <a href="#app1-biomolecules-14-01542" class="html-app">supplementary materials</a>.</p>
Full article ">Figure 8
<p>Phillyrin suppresses genes expression of SS2.The real-time PCR results of virulence factor genes (<span class="html-italic">mrp, epf, sly, stk</span>), cell adhesion-related genes (<span class="html-italic">ccpA, fbps</span>), and cell division-related genes (<span class="html-italic">gor</span>) of SC19. ** <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.001 compared to the respective control.</p>
Full article ">Figure 9
<p>Phillyrin reduced the virulence of SS2 in mice. (<b>A</b>) Virulence assay of SC19, SC19+phillyrin, and normal saline by comparing the survival of posttreatment mice. Bacterial load in the blood (<b>B</b>) and lung (<b>C</b>) tissues of the SC19-infected mice. Secretion of IL-1ß (<b>D</b>), IL-6 (<b>E</b>), IL-8 (<b>F</b>), and TNF-α (<b>G</b>) in the serum of mice infected with SC19. The height of the bars indicates the mean values for the relative expression data ± SEM (ns, <span class="html-italic">p</span> &gt; 0.05; * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.001; **** <span class="html-italic">p</span> &lt; 0.0001).</p>
Full article ">Figure 10
<p>Molecular docking of phillyrin and SLY. (<b>A</b>,<b>B</b>) Three-dimensional structure of SLY docked with phillyrin, revealing the amino acids Ala88 and Asp111 react with phillyrin. (<b>C</b>) Predictive interaction between phillyrin and SLY protein. Two key bonds indicated with the dotted arrows were established.</p>
Full article ">
13 pages, 5966 KiB  
Article
Development of a Triplex qPCR Assay Based on the TaqMan Probe for the Detection of Haemophilus parasuis, Streptococcus suis Serotype 2 and Pasteurella multocida
by Kaili Li, Yu Zhang, Tingyu Luo, Changwen Li, Haibo Yu, Wei Wang, He Zhang, Hongyan Chen, Changyou Xia and Caixia Gao
Microorganisms 2024, 12(10), 2017; https://doi.org/10.3390/microorganisms12102017 - 5 Oct 2024
Viewed by 815
Abstract
Porcine respiratory disease is a significant economic problem for the global swine industry. Haemophilus parasuis (H. parasuis), Streptococcus suis (S. suis), and Pasteurella multocida (P. multocida) are three important pathogenic bacteria of the swine respiratory tract. Notably, [...] Read more.
Porcine respiratory disease is a significant economic problem for the global swine industry. Haemophilus parasuis (H. parasuis), Streptococcus suis (S. suis), and Pasteurella multocida (P. multocida) are three important pathogenic bacteria of the swine respiratory tract. Notably, the three pathogens not only frequently manifest as mixed infections, but their striking clinical similarities also present difficulties for pig populations in terms of disease prevention and treatment. Thus, we developed a triplex real-time quantitative polymerase chain reaction (qPCR) assay based on a TaqMan probe for the detection of H. parasuis, S. suis serotype 2, and P. multocida. Primers and probes were designed to target the conserved regions of the H. parasuis OmpP2 gene, the S. suis serotype 2 gdh gene, and the P. multocida Kmt1 gene. By optimizing the reaction system and conditions, a triplex qPCR method for simultaneous detection of H. parasuis, S. suis serotype 2, and P. multocida was successfully established. The amplification efficiencies of the standard curves for all three pathogens were found to be highly similar, with values of 102.105% for H. parasuis, 105.297% for S. suis serotype 2, and 104.829% for P. multocida, and all R2 values achieving 0.999. The specificity analysis results showed that the triplex qPCR method had a strong specificity. The sensitivity test results indicated that the limit of detection can reach 50 copies/μL for all three pathogens. Both intra- and inter-assay coefficients of variation for repeatability were below 1%. This triplex qPCR method was shown to have good specificity, sensitivity, and reproducibility. Finally, the triplex qPCR method established in this study was compared with the nested PCR as recommended by the Chinese national standard (GB/T34750-2017) for H. parasuis, the PCR as recommended by the Chinese national standard (GB/T 19915.9-2005) for S. suis serotype 2, and the PCR as recommended by the Chinese agricultural industry standard (NY/T 564-2016) for P. multocida by detecting the same clinical samples. Both methods are reasonably consistent, while the triplex qPCR assay was more sensitive. In summary, triplex qPCR serves not only as a rapid and accurate detection and early prevention method for these pathogens but also constitutes a robust tool for microbial quality control in specific pathogen-free pigs. Full article
(This article belongs to the Section Microbial Biotechnology)
Show Figures

Figure 1

Figure 1
<p>A recombinant plasmid was constructed, containing three conserved gene fragments: <span class="html-italic">H. parasuis OmpP2</span> (314 bp), <span class="html-italic">S. suis</span> serotype 2 <span class="html-italic">gdh</span> (860 bp), and <span class="html-italic">P. multocida Kmt1</span> (457 bp). Specific restriction enzyme cutting sites (XhoI and BamHI for <span class="html-italic">H. parasuis OmpP2</span>, SacI and SalI for <span class="html-italic">S. suis</span> serotype 2 <span class="html-italic">gdh</span>, KpnI and NotI for <span class="html-italic">P. multocida Kmt1</span>) were placed on both ends of each fragment.</p>
Full article ">Figure 2
<p>Optimization of the triplex qPCR reaction system and procedure. (<b>A</b>) Optimization of primers in the reaction system. The optimal volumes of primers were 0.3 µL for <span class="html-italic">H. parasuis</span> and 0.6 µL for <span class="html-italic">S. suis</span> serotype 2 and <span class="html-italic">P. multocida</span>. (<b>B</b>) Optimization of probes in the reaction system. The optimal volumes of probes were 0.6 µL for <span class="html-italic">H. parasuis</span> and 0.8 µL for <span class="html-italic">P. multocida</span> and <span class="html-italic">S. suis</span> serotype 2. (<b>C</b>) Optimization of annealing temperature in the reaction procedure was performed. The optimal annealing temperature was 58 °C.</p>
Full article ">Figure 3
<p>Standard curve for the triplex qPCR, the copy number of the standard plasmid ranged from 10<sup>8</sup> to 10<sup>2</sup> copies/µL. (<b>A</b>) The standard curve for the <span class="html-italic">H. parasuis OmpP2</span> gene. (<b>B</b>) The standard curve for the <span class="html-italic">S. suis</span> serotype 2 <span class="html-italic">gdh</span> gene. (<b>C</b>) The standard curve for the <span class="html-italic">P. multocida Kmt1</span> gene.</p>
Full article ">Figure 4
<p>Specific analysis for the triplex qPCR assay. Ten pathogens (<span class="html-italic">H. parasuis</span>, <span class="html-italic">M. hyopneumoniae</span>, <span class="html-italic">M. hyorhinis</span>, <span class="html-italic">A. pleuropneumoniae</span>, <span class="html-italic">S. suis</span> serotype 2, <span class="html-italic">P. multocida</span>, PCV2, PRV, PRRSV, and SIV) were tested by the triplex qPCR assay. 1 × 10<sup>7</sup> copies/µL standard plasmid as a positive control (PC). A nuclease-free H<sub>2</sub>O control was used as a negative control (NC).</p>
Full article ">Figure 5
<p>The sensitivity of the triplex qPCR assay. (<b>A</b>) Sensitivity for the <span class="html-italic">H. parasuis OmpP2</span> gene. (<b>B</b>) Sensitivity for the <span class="html-italic">S. suis</span> serotype 2 <span class="html-italic">gdh</span> gene. (<b>C</b>) Sensitivity for the <span class="html-italic">P. multocida Kmt1</span> gene.</p>
Full article ">
13 pages, 2348 KiB  
Article
Characterization Studies on the sugC Gene of Streptococcus suis Serotype 2 in Adhesion, Invasion, and Virulence in Mice
by Zhimin Dong, Cheng Li, Xiangxue Tian, Xiaoran Guo, Xiuli Li, Weike Ren, Jingjing Chi, Li Zhang, Fuqiang Li, Yao Zhu, Wanjiang Zhang and Minghua Yan
Vet. Sci. 2024, 11(9), 447; https://doi.org/10.3390/vetsci11090447 - 21 Sep 2024
Viewed by 992
Abstract
The sugC gene of Streptococcus suis (S. suis) is a coding gene for the ATP-binding transporter-associated protein with strong pathogenicity. In order to reveal the effect of the sugC gene on the virulence of S. suis serotype 2, a wild-type strain [...] Read more.
The sugC gene of Streptococcus suis (S. suis) is a coding gene for the ATP-binding transporter-associated protein with strong pathogenicity. In order to reveal the effect of the sugC gene on the virulence of S. suis serotype 2, a wild-type strain of TJS75, isolated from fattening pigs’ brain tissue samples, was used as a parent strain, and a knockout sugC gene (ΔsugC) and complementary strain (CΔsugC) were successfully constructed via homologous recombination technology. The biological characteristics of TJS75, ΔsugC and CΔsugC were compared and analyzed through growth curves, biochemical characteristics, hemolysis characteristics, cell infection tests and pathogenicity tests on BALB/c mice. The results of the growth characteristic experiments in vitro showed that the plateau stage growth period of ΔsugC was delayed compared to the TJS75 strain, but there was no difference in the total number of bacteria. The biochemical characteristics and hemolysis ability of ΔsugC in sheep blood had no difference compared with TJS75, but its adhesion and invasion abilities in PK-15 cells were decreased. Knockout of the sugC gene had no impact on the expression levels of adhesion-related genes in TJS75 in real-time PCR analysis. In addition, the LD50 of ΔsugC in BALB/c mice was 1.47 × 108 CFU, seven times higher than that of TJS75 (LD50 = 2.15 × 107 CFU). These results illustrate that the deletion of sugC reduced the virulence of TJS75 to BALB/c mice, but its role in the adhesion and invasion of PK-15 cells in this strain needs to be further explored. In summary, this study provides evidence that the sugC gene is a virulence-related gene in the S. suis serotype 2 strain and plays a crucial role in the adhesion and invasion of S. suis. This study lays a foundation for the further exploration of the potential virulence factors and pathogenesis of S. suis. Full article
Show Figures

Figure 1

Figure 1
<p>Construction and confirmation of Δ<span class="html-italic">sugC</span> and CΔ<span class="html-italic">sugC</span>. (<b>A</b>) Construction and confirmation of Δ<span class="html-italic">sugC.</span> M = DL2000 marker; 1 = ddH<sub>2</sub>O; 2 and 4 = randomly chosen clone strains; 3 = pSET4s-Δ<span class="html-italic">sugC</span>; 5 = TJS75. (<b>B</b>) Construction and confirmation of CΔ<span class="html-italic">sugC.</span> M = DL2000 marker; 1 = ddH<sub>2</sub>O; 2 = randomly chosen clone strain; 3 = pSET2-<span class="html-italic">sugC</span>; 4 = TJS75.</p>
Full article ">Figure 2
<p>Amino acid sequence analysis of PCR products from multiple target fragments with TJS75, Δ<span class="html-italic">sugC</span> and CΔ<span class="html-italic">sugC</span>.</p>
Full article ">Figure 3
<p>Transcription of target molecules in Δ<span class="html-italic">sugC</span>, CΔ<span class="html-italic">sugC</span> and TJS75. Δ<span class="html-italic">sugC</span> vs. TJS75: <span class="html-italic">p<sub>sugC</sub></span> = 0, <span class="html-italic">p<sub>16S rRNA</sub></span> = 0.8839; CΔ<span class="html-italic">sugC</span> vs. TJS75: <span class="html-italic">p<sub>sugC</sub></span> = 0.7962, <span class="html-italic">p<sub>16S rRNA</sub></span> = 0.6983. ***: <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 4
<p>In vitro growth curves of different generations of TJS75, Δ<span class="html-italic">sugC</span> and CΔ<span class="html-italic">sugC</span>. Δ<span class="html-italic">sugC</span> vs. TJS75: <span class="html-italic">p</span><sub>6h</sub> = 0.0064, <span class="html-italic">p</span><sub>8h</sub> = 0.0035, <span class="html-italic">p</span><sub>10h</sub> = 0.0051, <span class="html-italic">p</span><sub>12h</sub> = 0.0063, <span class="html-italic">p</span><sub>14h</sub> = 0.0912, <span class="html-italic">p</span><sub>16h</sub> = 0.2954, <span class="html-italic">p</span><sub>20h</sub> = 0.2149, <span class="html-italic">p</span><sub>24h</sub> = 0.0506; CΔ<span class="html-italic">sugC</span> vs. TJS75: <span class="html-italic">p</span><sub>6h</sub> = 0.1460; <span class="html-italic">p</span><sub>8h</sub> = 0.0116; <span class="html-italic">p</span><sub>10h</sub> = 0.0092, <span class="html-italic">p</span><sub>12h</sub> = 0.1077, <span class="html-italic">p</span><sub>14h</sub> = 0.2254, <span class="html-italic">p</span><sub>16h</sub> = 0.0965, <span class="html-italic">p</span><sub>20h</sub> = 0.5040, <span class="html-italic">p</span><sub>24h</sub> = 0.1165.</p>
Full article ">Figure 5
<p>Hemolytic characteristic results of detection of <span class="html-italic">sugC</span> in <span class="html-italic">S. suis</span>. OD<sub>630</sub> values of solutions in which Δ<span class="html-italic">sugC</span>, CΔ<span class="html-italic">sugC</span> and TJS75 were interacted with sheep red blood cells. Mean<sub>TJS75</sub> = 0.3180, Mean<sub>Δ<span class="html-italic">sugC</span></sub> = 0.3050, Mean<sub>CΔ<span class="html-italic">sugC</span></sub> = 0.3110, <span class="html-italic">p</span><sub>Δ<span class="html-italic">sugC</span></sub> vs. <sub>TJS75</sub> = 0.1421, <span class="html-italic">p</span><sub>C</sub><sub>Δ<span class="html-italic">sugC</span></sub> vs. <sub>TJS75</sub> = 0.3714.</p>
Full article ">Figure 6
<p>Analysis of adhesion and invasiveness. (<b>A</b>) The MBCs for 1 h of Δ<span class="html-italic">sugC</span>, CΔ<span class="html-italic">sugC</span>, and TJS75. (<b>B</b>) The effect of GEN on the viability of PK-15 cells by MTT. Mean<sub>0 μg/mL</sub> = 0.2320, Mean<sub>16 μg/mL</sub> = 0.2224, Mean<sub>32 μg/mL</sub> = 0.2229, Mean<sub>64 μg/mL</sub> = 0.2296, Mean<sub>128 μg/mL</sub> = 0.2173, Mean<sub>256 μg/mL</sub> = 0.1021; <span class="html-italic">p</span><sub>0 μg/mL</sub> vs. <sub>16 μg/mL</sub>= 0.5014, <span class="html-italic">p</span><sub>0 μg/mL</sub> vs. <sub>32 μg/mL</sub>= 0.5805, <span class="html-italic">p</span><sub>0 μg/mL</sub> vs. <sub>64 μg/mL</sub> = 0.8622, <span class="html-italic">p</span><sub>0 μg/mL</sub> vs. <sub>128 μg/mL</sub> = 0.1952, <span class="html-italic">p</span><sub>0 μg/mL</sub> vs. <sub>256 μg/mL</sub>= 0.0102. (<b>C</b>) Results of cell adhesion test of Δ<span class="html-italic">sugC</span>, CΔ<span class="html-italic">sugC</span> and TJS75. Mean<sub>TJS75</sub> = 1.76 × 10<sup>7</sup> CFU/mL, Mean<sub>Δ<span class="html-italic">sugC</span></sub> = 1.16 × 10<sup>7</sup> CFU/mL, Mean<sub>CΔ<span class="html-italic">sugC</span></sub> = 1.62 × 10<sup>7</sup> CFU/mL, <span class="html-italic">p</span><sub>Δ<span class="html-italic">sugC</span></sub> vs. <sub>TJS75</sub> = 0.0001, <span class="html-italic">p<sub>C</sub></span><sub>Δ<span class="html-italic">sugC</span></sub> vs. <sub>TJS75</sub> = 0.0918, <span class="html-italic">p</span><sub>Δ<span class="html-italic">sugC</span></sub> vs. <sub>CΔ<span class="html-italic">sugC</span></sub> = 0.0001. (<b>D</b>) Results of cell invasion test of Δ<span class="html-italic">sugC</span>, CΔ<span class="html-italic">sugC</span>, and TJS75. Mean<sub>TJS75</sub> = 3.63 × 10<sup>4</sup> CFU/mL, Mean<sub>Δ<span class="html-italic">sugC</span></sub>= 3.14 × 10<sup>4</sup> CFU/mL, Mean<sub>CΔ<span class="html-italic">sugC</span></sub> = 3.60 × 10<sup>4</sup> CFU/mL, <span class="html-italic">p</span><sub>Δ<span class="html-italic">sugC</span></sub> vs. <sub>TJS75</sub> = 0.0307, <span class="html-italic">p<sub>C</sub></span><sub>Δ<span class="html-italic">sugC</span></sub> vs. <sub>TJS75</sub> = 0.8327, <span class="html-italic">p</span><sub>Δ<span class="html-italic">sugC</span></sub> vs. <sub>CΔ<span class="html-italic">sugC</span></sub> = 0.0165. (<b>E</b>) The relative expression of <span class="html-italic">fbps</span>, <span class="html-italic">cps2J</span>, <span class="html-italic">gdh</span> and <span class="html-italic">gapdh</span>. Δ<span class="html-italic">sugC</span> vs. TJS75: <span class="html-italic">p<sub>fbps</sub></span> = 0.0624, <span class="html-italic">p cps2J</span> = 0.1124, <span class="html-italic">p gdh</span> = 0.2038, <span class="html-italic">p gapdh</span> = 0.0528, <span class="html-italic">p<sub>16S rRNA</sub></span> = 0.6622; CΔ<span class="html-italic">sugC</span> vs. TJS75: <span class="html-italic">p<sub>fbps</sub></span> = 0.8320, <span class="html-italic">p cps2J</span> = 0.2907, <span class="html-italic">p gdh</span> = 0.2851, <span class="html-italic">p gapdh</span> = 0.6814, <span class="html-italic">p<sub>16S rRNA</sub></span> = 0.8105. *: <span class="html-italic">p</span> &lt; 0.05; ***: <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">
12 pages, 4259 KiB  
Article
Streptococcus suis Induces Macrophage M1 Polarization and Pyroptosis
by Siqi Li, Tianfeng Chen, Kexin Gao, Yong-Bo Yang, Baojie Qi, Chunsheng Wang, Tongqing An, Xuehui Cai and Shujie Wang
Microorganisms 2024, 12(9), 1879; https://doi.org/10.3390/microorganisms12091879 - 12 Sep 2024
Viewed by 980
Abstract
Streptococcus suis is an important bacterial pathogen that affects the global pig industry. The immunosuppressive nature of S. suis infection is recognized, and our previous research has confirmed thymus atrophy with a large number of necrotic cells. In this current work, we aimed to [...] Read more.
Streptococcus suis is an important bacterial pathogen that affects the global pig industry. The immunosuppressive nature of S. suis infection is recognized, and our previous research has confirmed thymus atrophy with a large number of necrotic cells. In this current work, we aimed to uncover the role of pyroptosis in cellular necrosis in thymic cells of S. suis-infected mice. Confocal microscopy revealed that S. suis activated the M1 phenotype and primed pyroptosis in the macrophages of atrophied thymus. Live cell imaging further confirmed that S. suis could induce porcine alveolar macrophage (PAM) pyroptosis in vitro, displaying cell swelling and forming large bubbles on the plasma membrane. Meanwhile, the levels of p-p38, p-extracellular signal-regulated kinase (ERK) and protein kinase B (AKT) were increased, which indicated the mitogen-activated protein kinase (MAPK) and AKT pathways were also involved in the inflammation of S. suis-infected PAMs. Furthermore, RT-PCR revealed significant mRNA expression of pro-inflammatory mediators, including interleukin (IL)-1β, IL-6, IL-18, tumor necrosis factor (TNF)-α and chemokine CXCL8. The data indicated that the inflammation induced by S. suis was in parallel with pro-inflammatory activities of M1 macrophages, pyroptosis and MAPK and AKT pathways. Pyroptosis contributes to necrotic cells and thymocyte reduction in the atrophied thymus of mice. Full article
(This article belongs to the Special Issue The Pathogenic Epidemiology of Important Swine Diseases)
Show Figures

Figure 1

Figure 1
<p>Thymic macrophage polarization in <span class="html-italic">S. suis</span> infection. Using confocal laser scanning microscopy, appropriate FITC-conjugated antibodies were used to label cells in sections from thymus of infected mice; M1 macrophage marker IL-1β (green); M2 macrophage marker CD206 (green). Macrophages were stained with F4/80 antibody (red) and cell nuclei (blue) were stained with DAPI; BF: Bright-field. Scale bars, 5 μm.</p>
Full article ">Figure 2
<p><span class="html-italic">S. suis</span> induces pyroptosis in macrophages of the thymus of infected mice. Confocal laser scanning immunostaining was performed to visualize thymic macrophages (F4/80 antibody; purple) and pyroptosis (GSDMD-N; red) in atrophied and control thymus of mice, and nuclei were stained with DAPI (blue). BF: Bright-field. Scale bars, 5 μm.</p>
Full article ">Figure 3
<p><span class="html-italic">S. suis</span> induces pro-inflammatory mediators in thymus of infected mice. (<b>A</b>) Total tissue RNA was extracted at 12 hpi and 1, 2 and 4 dpi, and the levels of IL-6, (<b>B</b>) CXCL8, (<b>C</b>) IL-1β, (<b>D</b>) IL-18, (<b>E</b>) and TNF-α mRNA expression were analyzed using real-time RT-PCR. Experiments were repeated three times. Differences were analyzed using unpaired Student’s <span class="html-italic">t</span>-test; data are mean ± SD; *: <span class="html-italic">p</span> &lt; 0.05; **: <span class="html-italic">p</span> &lt; 0.01; ***: <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 4
<p><span class="html-italic">S. suis</span>-induced LDH release and damage to primary PAMs. Primary PAMs were infected with <span class="html-italic">S. suis</span> 700794 at an MOI = 1 for 1, 2, 3, 8, 12 or 24 h, with a negative control included as 0 h. (<b>A</b>) LDH release induced by <span class="html-italic">S. suis</span> in primary PAMs was quantified by a standard LDH-release assay. (<b>B</b>) Electron micrographs of infected primary PAMs showing <span class="html-italic">S. suis</span>-induced cellular damage at 8 hpi, and yellow circle showed vacuoles of PAMs. Experiments were repeated three times, and results are expressed as means ± S.D., as determined by one-way ANOVA; *: <span class="html-italic">p</span> &lt; 0.05; scale bars, 2 μm.</p>
Full article ">Figure 5
<p>Live-cell imaging of pyroptosis after in vitro infection with <span class="html-italic">S. suis</span>. PAMs were cultured in glass-bottom cell culture dishes and treated with <span class="html-italic">S. suis</span> (MOI = 1). (<b>A</b>) Microscopy images of iPAMs treated with <span class="html-italic">S. suis</span> incubated in medium containing Hoechst 33342 (cell nuclei; blue) and propidium iodide (PI; red), the yellow arrows indicate pyrolyzed cells. (<b>B</b>) Microscopy images of primary PAMs treated with <span class="html-italic">S. suis</span>, and rectangle indicates pyrolyzed cells. Numbers at upper left indicate time post-infection (h:min:s).</p>
Full article ">Figure 6
<p>The MAPK and AKT pathways were activated by <span class="html-italic">S. suis</span> in porcine macrophages. Cells were infected with <span class="html-italic">S. suis</span> at an MOI = 1 for 15, 30, 60 and 120 min. The cell lysates were collected, and levels of proteins related to inflammatory functional indexes were determined by Western blot (<b>A</b>). Representative Western blots show the expression level of (<b>B</b>) p-p38, (<b>C</b>) p38, (<b>D</b>) p-ERK1/2, (<b>E</b>) ERK1/2 and (<b>F</b>) AKT. The mean ± SD of experiments are shown; *: <span class="html-italic">p</span> &lt; 0.05; **: <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">
12 pages, 1615 KiB  
Article
The Mycotoxins T-2 and Deoxynivalenol Facilitate the Translocation of Streptococcus suis across Porcine Ileal Organoid Monolayers
by Xiaonan Guan, Arabela R. Martinez, Marcela Fernandez, Francesc Molist, Jerry M. Wells and Regiane R. Santos
Toxins 2024, 16(9), 382; https://doi.org/10.3390/toxins16090382 - 1 Sep 2024
Viewed by 1540
Abstract
Mycotoxins have the potential to increase the risk of airway or intestinal infection due to their effects on epithelial integrity and function. The bacterium Streptococcus suis (S. suis) is often carried in pigs and can cause outbreaks of invasive disease, leading to sepsis [...] Read more.
Mycotoxins have the potential to increase the risk of airway or intestinal infection due to their effects on epithelial integrity and function. The bacterium Streptococcus suis (S. suis) is often carried in pigs and can cause outbreaks of invasive disease, leading to sepsis and meningitis in postweaning piglets. In this study, we tested the effect of two Fusarium mycotoxins (deoxynivalenol (DON) and T-2) on the integrity of the intestinal epithelium and their interaction with S. suis. Porcine ileal organoids were exposed to DON and T-2 individually or in combination and co-cultured with or without S. suis. Both DON and T-2 were toxic for ileal organoid monolayers at a concentration of 1 µM but not S. suis, even at a higher concentration of 4 µM. To mimic sub-clinical exposures on farms, DON was tested at a concentration of 0.1 µM and T-2 at a concentration of 0.01 µM. The mycotoxins alone did not affect cell permeability, but in combination with S. suis there was an increase in epithelial permeability. Furthermore, DON and T-2 together decreased the transepithelial electrical resistance and increased bacterial translocation. Full article
(This article belongs to the Section Mycotoxins)
Show Figures

Figure 1

Figure 1
<p>Mean (± SD) viability of porcine 2D monolayers of ileal organoids exposed to DON or T-2 toxin. Organoids were exposed to 0.1–10 µM DON (<b>A</b>) or T-2 (<b>B</b>) for 24 h. At the end of incubation, the ATP was quantified using the CellTiter-Glo<sup>®</sup> 2.0 assay. ATP measurements for each treatment are expressed as the percentage of the control value (untreated). a–c Different letters indicate a significant difference among treatments within each tested mycotoxin (<span class="html-italic">p</span> ≤ 0.05). The experiment was carried out in triplicate with four independent repetitions.</p>
Full article ">Figure 2
<p>(<b>A</b>): Representative ileal organoid monolayers were stained with Hoechst (nuclei in blue) and phalloidin (actin stain in green) (<b>B</b>): Mean (± SD) permeability to 4 kDa FITC-dextran was quantified by measurement of fluorescence in the basal compartment after 18 h and indicated as a percentage of the mean for the untreated control. The different letters a–d significant differences among treatments (<span class="html-italic">p</span> ≤ 0.05). The experiment was carried out in triplicate with four independent repetitions.</p>
Full article ">Figure 3
<p>Mean (± SD) transepithelial electric resistance (TER) of a 2D organoid culture continuously measured for 18 h post-seeding. (<b>A</b>) Treatments without <span class="html-italic">S. suis</span> (Control, DON, T2, and DON + T2); (<b>B</b>) Control group without <span class="html-italic">S. suis</span> (Control) and mycotoxins combined with <span class="html-italic">S. suis</span> exposure (Control + <span class="html-italic">S. suis</span>, DON + <span class="html-italic">S. suis</span>, T2 + <span class="html-italic">S. suis</span>, and DON + T2 + <span class="html-italic">S. suis</span>). The experiment was carried out in triplicate with four independent repetitions.</p>
Full article ">Figure 4
<p>Mean (± SD) <span class="html-italic">S. suis</span> colony forming units/mL (CFU/mL) in the apical (<b>A</b>) and basal (<b>B</b>) compartments of the culture wells. a,b Different letters indicate significant differences among treatments within the basal part (<span class="html-italic">p</span> ≤ 0.05). DON: 0.1 µM; T-2: 0.01 µM; DON + T-2: 0.1 µM + 0.01 µM, respectively. The experiment was carried out in triplicate with four independent repetitions.</p>
Full article ">
13 pages, 4545 KiB  
Article
Identifying Cell-Penetrating Peptides for Effectively Delivering Antimicrobial Molecules into Streptococcus suis
by Jinlu Zhu, Zijing Liang, Huochun Yao and Zongfu Wu
Antibiotics 2024, 13(8), 725; https://doi.org/10.3390/antibiotics13080725 - 2 Aug 2024
Viewed by 1325
Abstract
Cell-penetrating peptides (CPPs) are promising carriers to effectively transport antisense oligonucleotides (ASOs), including peptide nucleic acids (PNAs), into bacterial cells to combat multidrug-resistant bacterial infections, demonstrating significant therapeutic potential. Streptococcus suis, a Gram-positive bacterium, is a major bacterial pathogen in pigs and [...] Read more.
Cell-penetrating peptides (CPPs) are promising carriers to effectively transport antisense oligonucleotides (ASOs), including peptide nucleic acids (PNAs), into bacterial cells to combat multidrug-resistant bacterial infections, demonstrating significant therapeutic potential. Streptococcus suis, a Gram-positive bacterium, is a major bacterial pathogen in pigs and an emerging zoonotic pathogen. In this study, through the combination of super-resolution structured illumination microscopy (SR-SIM), flow cytometry analysis, and toxicity analysis assays, we investigated the suitability of four CPPs for delivering PNAs into S. suis cells: HIV-1 TAT efficiently penetrated S. suis cells with low toxicity against S. suis; (RXR)4XB had high penetration efficiency with inherent toxicity against S. suis; (KFF)3K showed lower penetration efficiency than HIV-1 TAT and (RXR)4XB; K8 failed to penetrate S. suis cells. HIV-1 TAT-conjugated PNA specific for the essential gyrase A subunit gene (TAT-anti-gyrA PNA) effectively inhibited the growth of S. suis. TAT-anti-gyrA PNA exhibited a significant bactericidal effect on serotypes 2, 4, 5, 7, and 9 strains of S. suis, which are known to cause human infections. Our study demonstrates the potential of CPP-ASO conjugates as new antimicrobial compounds for combating S. suis infections. Furthermore, our findings demonstrate that applying SR-SIM and flow cytometry analysis provides a convenient, intuitive, and cost-effective approach to identifying suitable CPPs for delivering cargo molecules into bacterial cells. Full article
Show Figures

Figure 1

Figure 1
<p>Representative images were used for analyzing the internalization efficiency of FITC-labeled CPPs in <span class="html-italic">S. suis</span> by SR-SIM. The negative control received an equivalent volume of water, while an equivalent concentration of FITC single molecules was also included as a control. The <span class="html-italic">S. suis</span> cell membrane was counterstained with Alexa Fluor 633-WGA (wheat germ agglutinin) and observed with a laser at a wavelength of 640 nm (WGA, red), while the fluorescence signal of FITC was observed using a laser at a wavelength of 488 nm (FITC, green).</p>
Full article ">Figure 2
<p>Cellular uptake of CPPs in <span class="html-italic">S. suis</span> was analyzed by flow cytometry. <span class="html-italic">S. suis</span> cells were exposed to 10 µM FITC-labeled CPPs, with fluorescence measured one hour post-treatment. The negative control (NC) received an equivalent volume of water, while an equivalent concentration of FITC single molecules was also included as a control. A total of 50,000 events were collected during the flow cytometry analysis. Dot plots (<b>A</b>) and histograms (<b>B</b>) were analyzed using FlowJo™ v10 software, with quadrant 3 representing the negative confidence region. SSC, or cell count, was plotted on the <span class="html-italic">y</span>-axis, and FITC fluorescence intensity was plotted on the <span class="html-italic">x</span>-axis. The MFI of FITC was analyzed using FlowJo™ v10 software (<b>C</b>). The unpaired <span class="html-italic">t</span>-test was used to compare the MFI of <span class="html-italic">S. suis</span>. * indicates <span class="html-italic">p</span> &lt; 0.05, ** indicates <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 3
<p>Growth kinetics and MIC determination for <span class="html-italic">S. suis</span> serotype 2 strain SC070731 in various concentrations of CPPs. (<b>A</b>) HIV-1 TAT at concentrations ranging from 128 to 8 µM. (<b>B</b>) (RXR)<sub>4</sub>XB at concentrations ranging from 32 to 2 µM. An equivalent volume of water was included as a control. The MIC value is shown and was determined as the lowest concentration inhibiting visible growth in the wells (OD<sub>595nm</sub> &lt; 0.1).</p>
Full article ">Figure 4
<p>Schematic illustration of the PNA target region of gene <span class="html-italic">gyrA</span>. (<b>A</b>) Region of <span class="html-italic">gyrA</span> mRNA in <span class="html-italic">S. suis</span>, with the start codon (AUG) shown in bold type. For the location relative to the start site, ‘A’ of AUG is defined as +1 in this study. The Shine–Dalgarno and PNA target sequences are shaded in green and blue, respectively. Below, the PNA sequence is shown (blue box) with the conjugated CPP HIV-1 TAT for delivery into <span class="html-italic">S. suis</span>. (<b>B</b>) Multiple sequence alignments of the gene <span class="html-italic">gyrA</span> in different serotypes of <span class="html-italic">S. suis</span>, including serotype 2 strain SC070731, serotype 4 strain ND90, serotype 5 strain GX169, serotype 7 strain WUSS013, and serotype 9 strain GZ0565. A defined section (−26 to +60 nt), including the region around the PNA binding site (blue dashed border), is shown.</p>
Full article ">Figure 5
<p>An antisense <span class="html-italic">gyrA</span>-specific PNA coupled to HIV-1 TAT (TAT-anti-<span class="html-italic">gyrA</span> PNA) exhibits antibacterial activity against <span class="html-italic">S. suis</span> strains. (<b>A</b>) Growth kinetics and MIC determination of SC070731 in various concentrations of free <span class="html-italic">gyrA</span>-specific PNA, free HIV-1 TAT, or TAT-anti-<span class="html-italic">gyrA</span> PNA at concentrations ranging from 32 to 2 µM. The MIC value is shown and was determined as the lowest concentration inhibiting visible growth in the wells (OD<sub>595nm</sub> &lt; 0.1). (<b>B</b>) Bactericidal effects of TAT-anti-<span class="html-italic">gyrA</span> PNA were determined at 1 × MIC (4 µM) and 2 × MIC (8 µM) against <span class="html-italic">S. suis</span> serotype 2 strain SC070731 during a 4-h time course. After the indicated time points, aliquots of each condition were subjected to spot assays or CFU determination on THA plates to investigate the number of viable cells. (<b>C</b>) Concentration-dependent reduction in the bacterial counts following treatment of <span class="html-italic">S. suis</span> strains of various serotypes with TAT-anti-<span class="html-italic">gyrA</span> PNA for 2 h. The unpaired <span class="html-italic">t</span>-test was used to compare the number of viable bacterial cells. ** indicates <span class="html-italic">p</span> &lt; 0.01, *** indicates <span class="html-italic">p</span> &lt; 0.001, **** indicates <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">
11 pages, 3196 KiB  
Article
Contamination of Streptococcus suis and S. suis Serotype 2 in Raw Pork and Edible Pig Organs: A Public Health Concern in Chiang Mai, Thailand
by Ratchadakorn Guntala, Likhitphorn Khamai, Nattawara Srisai, Sakaewan Ounjaijean, Woottichai Khamduang and Sayamon Hongjaisee
Foods 2024, 13(13), 2119; https://doi.org/10.3390/foods13132119 - 3 Jul 2024
Cited by 2 | Viewed by 1680
Abstract
Streptococcus suis is one of the most important zoonotic pathogens causing serious diseases in both pigs and humans, especially serotype 2. In northern Thailand, there is a notable prevalence of S. suis infection in humans and transmission has occurred mainly through the consumption [...] Read more.
Streptococcus suis is one of the most important zoonotic pathogens causing serious diseases in both pigs and humans, especially serotype 2. In northern Thailand, there is a notable prevalence of S. suis infection in humans and transmission has occurred mainly through the consumption of raw pork products. Despite the continued practice of consuming raw pork in this region, limited data exist regarding S. suis contamination in such products. Therefore, this study aimed to assess the prevalence of S. suis and S. suis serotype 2 in retail raw pork meat and edible pig organs sold in Chiang Mai city, Thailand. A total of 200 samples, comprising raw pork meat and edible pig organs, were collected from nine fresh markets in Chiang Mai city between May and July 2023. Samples were prepared and cultured in Todd-Hewitt broth. Bacterial DNA was extracted and tested for any serotypes of S. suis and serotype 2 using loop-mediated isothermal amplification (LAMP) techniques. The study revealed contaminations of S. suis and serotype 2 at rates of 84% and 34%, respectively, with a higher prevalence observed in pig organs compared to raw pork. Both S. suis and serotype 2 were detected across all nine fresh markets investigated. The prevalence of S. suis remained consistently high throughout the study period, whereas serotype 2 showed peaks in May and July. These high rates of contamination indicate that people who consume or work in close contact with raw pork or edible pig organs are at a high risk of S. suis infection. Urgent implementation and maintenance of food safety campaigns and public health interventions are crucial for disease prevention and control. Full article
(This article belongs to the Special Issue Detection and Control of Food-Borne Pathogens)
Show Figures

Figure 1

Figure 1
<p>Locations of the sites. The black stars on the map indicate locations of the 9 fresh markets (Market A–I) in Chiang Mai city.</p>
Full article ">Figure 2
<p>LAMP results for <span class="html-italic">Streptococcus suis</span> and <span class="html-italic">Streptococcus suis</span> serotype 2 detection in the first 11 representative raw pork samples. Lanes 1–11 represent the raw pork samples. (<b>a</b>) Results from LAMP-SS targeting <span class="html-italic">S. suis</span>, three samples were positive (lanes 1, 2, and 5) and two samples were negative (lanes 3 and 4). (<b>b</b>) Results from LAMP-SS2 targeting <span class="html-italic">S. suis</span> serotype 2, three samples were positive (lanes 6, 9, and 10) and three samples were negative (lanes 7, 8, and 11). DNA extracted from <span class="html-italic">S. suis</span> serotype 2 was used as positive control in both LAMP-SS and LAMP-SS2. The pattern of the LAMP products on gel electrophoresis corresponded with color change in the reaction tube. The maker (M) was a 1 kb DNA ladder. PC, positive control; NTC, no-template control.</p>
Full article ">Figure 3
<p>Prevalence of <span class="html-italic">S. suis</span> and <span class="html-italic">S. suis</span> serotype 2 contamination based on sample type. * indicates a statistically significant result (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 4
<p>Pie charts represent the prevalences of <span class="html-italic">S. suis</span> (<b>a</b>) and <span class="html-italic">S. suis</span> serotype 2 (<b>b</b>) contamination among 9 fresh markets located in Chiang Mai city, Thailand. Black area in the pie chart represents the percent of positive samples. The black stars on the map indicate locations of the 9 fresh markets (Market A–I) in Chiang Mai city.</p>
Full article ">Figure 5
<p>The prevalence rates of <span class="html-italic">S. suis</span> and <span class="html-italic">S. suis</span> serotype 2 contamination in raw pork/edible pig organs collected from 9 fresh markets according to the month of sample collection. * indicates a statistically significant result (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">
20 pages, 11978 KiB  
Article
Identification of Causal Relationships between Gut Microbiota and Influenza a Virus Infection in Chinese by Mendelian Randomization
by Qijun Liao, Fuxiang Wang, Wudi Zhou, Guancheng Liao, Haoyang Zhang, Yuelong Shu and Yongkun Chen
Microorganisms 2024, 12(6), 1170; https://doi.org/10.3390/microorganisms12061170 - 8 Jun 2024
Viewed by 1371
Abstract
Numerous studies have reported a correlation between gut microbiota and influenza A virus (IAV) infection and disease severity. However, the causal relationship between these factors remains inadequately explored. This investigation aimed to assess the influence of gut microbiota on susceptibility to human infection [...] Read more.
Numerous studies have reported a correlation between gut microbiota and influenza A virus (IAV) infection and disease severity. However, the causal relationship between these factors remains inadequately explored. This investigation aimed to assess the influence of gut microbiota on susceptibility to human infection with H7N9 avian IAV and the severity of influenza A (H1N1)pdm09 infection. A two-sample Mendelian randomization analysis was conducted, integrating our in-house genome-wide association study (GWAS) on H7N9 susceptibility and H1N1pdm09 severity with a metagenomics GWAS dataset from a Chinese population. Twelve and fifteen gut microbiotas were causally associated with H7N9 susceptibility or H1N1pdm09 severity, separately. Notably, Clostridium hylemonae and Faecalibacterium prausnitzii were negative associated with H7N9 susceptibility and H1N1pdm09 severity, respectively. Moreover, Streptococcus peroris and Streptococcus sanguinis were associated with H7N9 susceptibility, while Streptococcus parasanguini and Streptococcus suis were correlated with H1N1pdm09 severity. These results provide novel insights into the interplay between gut microbiota and IAV pathogenesis as well as new clues for mechanism research regarding therapeutic interventions or IAV infections. Future studies should concentrate on clarifying the regulatory mechanisms of gut microbiota and developing efficacious approaches to reduce the incidence of IAV infections, which could improve strategy for preventing and treating IAV infection worldwide. Full article
Show Figures

Figure 1

Figure 1
<p>Overview of the process of Mendelian randomization analysis and major assumptions.</p>
Full article ">Figure 2
<p>Causal effects of gut microbiota on H7N9 susceptibility. Summary of Mendelian randomization (MR) estimates derived from inverse variance weighted (IVW), weighted median (WM), and MR–Egger analyses. CI denotes confidence interval; OR, odds ratio; SNPs, single nucleotide polymorphisms. “s_”, “g_”, “f_” are species, genus, and family, respectively.</p>
Full article ">Figure 3
<p>Causal effects of gut microbiota on H1N1 severity. Summary of Mendelian randomization (MR) estimates obtained from inverse variance weighted (IVW), weighted median (WM), and MR–Egger analyses. CI, confidence interval; SNPs, single nucleotide polymorphisms. “s_”, “g_”, “f_” are species, genus, and family, respectively.</p>
Full article ">Figure 4
<p>Visualization of connected protein-protein interaction networks using Cytoscape. (<b>A</b>) The network of overlapped genes between H7N9 susceptibility and microbiota features. (<b>B</b>) The network of overlapped genes between H1N1 severity and microbiota features. Node size and color correspond to the respective degrees, while edge weight and color are proportional to the STRING confidence score.</p>
Full article ">Figure 5
<p>Gene Ontology pathway enrichment analysis performed on (<b>A</b>) 216 genes annotated from instrumental variables for pairs of microbial features and H7N9 susceptibility with potential causal relationships in Mendelian randomization (MR), and (<b>B</b>) 314 genes annotated from IVs for pairs of microbial features and H1N1 severity with potential causal relationships in MR. BP: biological process, CC: cellular component, MF: molecular function.</p>
Full article ">
13 pages, 1851 KiB  
Article
Immunogenicity and Protective Capacity of Sugar ABC Transporter Substrate-Binding Protein against Streptococcus suis Serotype 2, 7 and 9 Infection in Mice
by Zujie Yan, Ruyi Pan, Junjie Zhang, Jianhe Sun, Xiaochun Ma, Nihua Dong, Xiaohui Yao, Jianchao Wei, Ke Liu, Yafeng Qiu, Katie Sealey, Hester Nichols, Michael A. Jarvis, Mathew Upton, Xiangdong Li, Zhiyong Ma, Juxiang Liu and Beibei Li
Vaccines 2024, 12(5), 544; https://doi.org/10.3390/vaccines12050544 - 15 May 2024
Viewed by 1685
Abstract
Background: Streptococcus suis (S. suis) is a Gram-positive bacterium that causes substantial disease in pigs. S. suis is also an emerging zoonoses in humans, primarily in Asia, through the consumption of undercooked pork and the handling of infected pig meat [...] Read more.
Background: Streptococcus suis (S. suis) is a Gram-positive bacterium that causes substantial disease in pigs. S. suis is also an emerging zoonoses in humans, primarily in Asia, through the consumption of undercooked pork and the handling of infected pig meat as well as carcasses. The complexity of S. suis epidemiology, characterized by the presence of multiple bacterial serotypes and strains with diverse sequence types, identifies a critical need for a universal vaccine with the ability to confer cross-protective immunity. Highly conserved immunogenic proteins are generally considered good candidate antigens for subunit universal vaccines. Methods: In this study, the cross-protection of the sugar ABC transporter substrate-binding protein (S-ABC), a surface-associated immunogenic protein of S. suis, was examined in mice for evaluation as a universal vaccine candidate. Results: S-ABC was shown to be highly conserved, with 97% amino acid sequence identity across 31 S. suis strains deposited in GenBank. Recombinantly expressed S-ABC (rS-ABC) was recognized via rabbit sera specific to S. suis serotype 2. The immunization of mice with rS-ABC induced antigen-specific antibody responses, as well as IFN-γ and IL-4, in multiple organs, including the lungs. rS-ABC immunization conferred high (87.5% and 100%) protection against challenges with S. suis serotypes 2 and 9, demonstrating high cross-protection against these serotypes. Protection, albeit lower (50%), was also observed in mice challenged with S. suis serotype 7. Conclusions: These data identify S-ABC as a promising antigenic target within a universal subunit vaccine against S. suis. Full article
(This article belongs to the Section Veterinary Vaccines)
Show Figures

Figure 1

Figure 1
<p>Predicted linear B cell epitopes of S-ABC. (<b>A</b>) Linear B cell epitopes of S-ABC were predicted using the bioinformatics software ABCpred and IEDB. The predicted B cell epitope sequences are boxed. (<b>B</b>) Screening for immunodominant linear B cell epitopes. The predicted B cell epitope peptides (EP1-5) were coated onto ELISA plates and incubated with rabbit sera specific to <span class="html-italic">S. suis</span> serotype 2. The OD<sub>450nm</sub> value of each well coated with peptide was normalized to the value of control wells and plotted. A relative OD<sub>450nm</sub> value of &gt;2.1 was considered positive.</p>
Full article ">Figure 2
<p>Construction and expression of rS-ABC and rS-ABC-epitope proteins. (<b>A</b>) Schematic representation of the construction of rS-ABC-epitope. EP1 and EP5 were tandemly connected by GGGG linkers to be expressed as a multiple-epitope protein. (<b>B</b>) Expression and purification of rS-ABC and rS-ABC-epitope proteins. The rS-ABC and rS-ABC-epitope proteins were expressed from <span class="html-italic">E. coli</span>, induced with IPTG, and purified via their His-tag by using a Ni–NTA column. The resulting recombinant proteins were then examined using SDS-PAGE. Lanes: 1, protein marker; 2, uninduced bacterial cells; 3, bacterial cells induced with IPTG; and 4, purified recombinant protein. (<b>C</b>) Antigenic analysis of rS-ABC and rS-ABC-epitope proteins. The rS-ABC and rS-ABC-epitope purified proteins were examined by a Western blot using anti-His antibodies and positive rabbit sera containing antibodies specific to <span class="html-italic">S. suis</span> serotype 2.</p>
Full article ">Figure 3
<p>Levels of antibodies and cytokines in immunized mice. (<b>A</b>) Schematic representation of vaccination and sample collection. Mice were vaccinated with rS-ABC or rS-ABC-epitope proteins and then boosted at 14 days post-primary vaccination. (<b>B</b>) Levels of antibodies specific to rS-ABC or rS-ABC-epitope were examined with an ELISA in sera collected prior to primary vaccination, boost vaccination, and challenge. (<b>C</b>–<b>E</b>) Levels of IL-4 and IFN-γ in sera (B), lungs (<b>C</b>), and spleen (<b>D</b>) collected at 24 days post-primary vaccination were examined with an ELISA. Statistically significant differences between groups are indicated by asterisks (** <span class="html-italic">p</span> &lt; 0.01).</p>
Full article ">Figure 4
<p>Protective efficacy of rS-ABC and rS-ABC-epitope in mice. Three-week-old BALB/c strain mice (8 mice per group) were vaccinated with rS-ABC or rS-ABC-epitope proteins and boosted at 14 days post-primary vaccination. Mice were challenged with a lethal dose of <span class="html-italic">S. suis</span> serotype 2, 7, or 9 at 10 days post-boost vaccination and monitored for 7 days. Animals succumbed to infection between 12 and 48 h post-challenge, and the survival curves were plotted.</p>
Full article ">
13 pages, 2867 KiB  
Article
Isolation, Identification, Genomic Diversity, and Antimicrobial Resistance Analysis of Streptococcus suis in Hubei Province of China from 2021 to 2023
by Yingjun Xia, Zhaoyang Wang, Yanli Hu, Pengfei Zhao, Jianhai Li, Li Zhang, Rui Fang and Junlong Zhao
Microorganisms 2024, 12(5), 917; https://doi.org/10.3390/microorganisms12050917 - 30 Apr 2024
Cited by 1 | Viewed by 1445
Abstract
Streptococcus suis (S. suis) is a zoonotic pathogen capable of causing severe diseases in humans and pigs, including meningitis, sepsis, polyserositis, arthritis, and endocarditis. This study aimed to investigate the biological characteristics of 19 strains of S. suis isolated from diseased [...] Read more.
Streptococcus suis (S. suis) is a zoonotic pathogen capable of causing severe diseases in humans and pigs, including meningitis, sepsis, polyserositis, arthritis, and endocarditis. This study aimed to investigate the biological characteristics of 19 strains of S. suis isolated from diseased pigs in Hubei Province between 2021 and 2023. Through bioinformatics analysis, we investigated the serotype, MLST, pan-genome characteristics, SNP, AMR, and ICE of the 19 S. suis isolates. Among the 19 S. suis strains, ten serotypes were identified, and serotype 9 was the most prevalent (21.05%). Ten new alleles and nine new sequence types (STs) were discovered, with ST28 and ST243 emerging as the predominant STs. The results of the pan-genomic analysis of S. suis indicate that there are 943 core genes, 2259 shell genes, and 5663 cloud genes. Through SNP evolutionary analysis, we identified a strong genetic similarity between SS31 and the reference genome P1/7. The analysis of antibiotic resistance genes revealed widespread presence of erm(B) and tet(O) genes among 19 strains of S. suis. This association may be linked to the high resistance of S. suis to lincosamides, macrolides, and tetracyclines. Integrative and conjugative elements (ICEs) and integrative and mobilizable elements (IMEs) were identified in 16 strains, with a carriage rate of 84.21%, and resistance genes were identified within the ICE/IME elements of 8 strains. Antimicrobial susceptibility testing revealed that all strains showed sensitivity to vancomycin and lincomycin but resistance to tilmicosin, tiamulin, amoxicillin, and doxycycline. This study contributes to our understanding of the genomic diversity of S. suis in Hubei Province of China, providing essential data for the comprehensive prevention and control of S. suis infections in China. Full article
(This article belongs to the Section Veterinary Microbiology)
Show Figures

Figure 1

Figure 1
<p>Proportion of <span class="html-italic">S. suis</span> isolates in each region.</p>
Full article ">Figure 2
<p>The prevalence of serotypes of <span class="html-italic">S. suis</span> isolates from Hubei in 2021–2023. NT = Nontypeable isolates.</p>
Full article ">Figure 3
<p><span class="html-italic">S. suis</span> pan-genome. (<b>a</b>) The number of genes belonging to the core, the soft core, the shell, and the cloud of the <span class="html-italic">S. suis</span> species is pictured as a pie chart; (<b>b</b>) The relationship between the number of unique genes and the number of genomes; (<b>c</b>) The construction of the phylogenetic tree of the core genes of <span class="html-italic">S. suis</span>; Note: The tree on the left is a neighbor-joining phylogenetic tree constructed based on the core genes. The matrix plot on the right shows the presence and absence of genes on each strain in blue and white; (<b>d</b>) The SNP phylogenetic tree of 20 <span class="html-italic">S. suis</span> strains.</p>
Full article ">Figure 3 Cont.
<p><span class="html-italic">S. suis</span> pan-genome. (<b>a</b>) The number of genes belonging to the core, the soft core, the shell, and the cloud of the <span class="html-italic">S. suis</span> species is pictured as a pie chart; (<b>b</b>) The relationship between the number of unique genes and the number of genomes; (<b>c</b>) The construction of the phylogenetic tree of the core genes of <span class="html-italic">S. suis</span>; Note: The tree on the left is a neighbor-joining phylogenetic tree constructed based on the core genes. The matrix plot on the right shows the presence and absence of genes on each strain in blue and white; (<b>d</b>) The SNP phylogenetic tree of 20 <span class="html-italic">S. suis</span> strains.</p>
Full article ">Figure 4
<p>Heat map showing antimicrobial susceptibility profiles of <span class="html-italic">S. suis</span> strains. Rows represent bacterial strains, and columns represent antibiotics, where blue blocks indicate antibiotic susceptibility (S), yellow blocks indicate intermediacy (I), and red blocks indicate resistance (R) to action of antibiotics. ENR = Enrofloxacin; OFX = Ofloxacin; LNZ = Linezolid; FLR = Florfenicol; AMX = Amoxicillin; LIN = Lincomycin; DOX = Doxycycline; TIA = Tiamulin; VAN = Vancomycin; TYL = Tylosin; TIL = Tilmicosin; STR = Streptomycin.</p>
Full article ">Figure 5
<p>Comparison of ICEs carrying AMR genes. Light blue arrows indicate genes coding for Type IV Secretion System; green arrows indicate genes coding for relaxases; yellow arrows indicate genes coding for integrases; red arrows indicate <span class="html-italic">tet</span>(O) genes; and dark blue arrows indicate <span class="html-italic">erm</span>(B) genes.</p>
Full article ">
15 pages, 9339 KiB  
Article
Tackling Antibiotic Resistance: Exploring 5-Fluorouracil as a Promising Antimicrobial Strategy for the Treatment of Streptococcus suis Infection
by Jing Zuo, Yingying Quan, Jinpeng Li, Yue Li, Dong Song, Xingping Li, Yuxin Wang, Li Yi and Yang Wang
Animals 2024, 14(9), 1286; https://doi.org/10.3390/ani14091286 - 24 Apr 2024
Cited by 1 | Viewed by 1747
Abstract
Streptococcus suis (S. suis) is a zoonotic pathogen with a global distribution, which causes serious diseases in both humans and animals and economic losses in the swine industry. As antibiotic resistance increases, there is an urgent imperative to explore novel antibacterial [...] Read more.
Streptococcus suis (S. suis) is a zoonotic pathogen with a global distribution, which causes serious diseases in both humans and animals and economic losses in the swine industry. As antibiotic resistance increases, there is an urgent imperative to explore novel antibacterial alternatives. In the present study, we selected the anticancer drug 5-fluorouracil (5-FU) approved by the Food and Drug Administration (FDA) as a candidate drug to treat S. suis infections. The results showed that various pathogens, especially S. suis, are more sensitive to 5-FU. Moreover, the cytotoxicity of 5-FU is relatively low. Extensive in vitro assays demonstrated the pronounced bacteriostatic and bactericidal efficacy of 5-FU against susceptible and multidrug-resistant S. suis strains. Its mechanisms of action include damage to the bacterial cell walls and membranes, resulting in the leakage of intracellular components, and the inhibition of thymidylate synthase (TS), leading to a depletion of deoxythymidine triphosphate (dTTP) pools, ultimately causing thymine-less death and lethal DNA damage in bacteria. Gene-knockout experiments further showed that 5-FU played a role by inhibiting the thyA gene-encoding thymidine synthase. Finally, we determined that S. suis infections can be alleviated by 5-FU in the mouse infection model. This study emphasizes the antibacterial potential of 5-FU against S. suis and provides evidence for its targeting of bacterial membrane damage and DNA damage. In summary, 5-FU can control S. suis infection and is expected to become a new alternative to antibiotics. Full article
(This article belongs to the Section Veterinary Clinical Studies)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>5-FU has significant antibacterial and bactericidal effects, a low frequency of drug resistance, and low cytotoxicity. (<b>A</b>) The MIC of 5-FU against Gram-negative (red) and Gram-positive (purple) bacteria. See also <a href="#animals-14-01286-t001" class="html-table">Table 1</a>. (<b>B</b>,<b>C</b>) The bacterial-growth curve of <span class="html-italic">S. suis</span> HA9801 after treatment with 5-FU. The bacterial-growth curves were measured by the optical density at 600 nm and colony-forming units every 2 h following inoculation. (<b>D</b>) Colony-forming units (CFUs mL<sup>−1</sup>) after a 4 h treatment of <span class="html-italic">S. suis</span> HA9801 with PBS (solvent control), 20 µg/mL 5-FU (4 × MIC), 2.5 µg/mL amoxicillin (4 × MIC), or 2 mg/mL Polymyxin B (4 × MIC). The data points of 1 × 10<sup>2</sup> CFU mL<sup>−1</sup> were below the detected level. The colony-forming units of two clinically isolated antibiotic-resistant strains SS-1 and SS-2 of <span class="html-italic">S. suis</span> are shown in <a href="#app1-animals-14-01286" class="html-app">Figure S1</a>. Each datum represents three biological replicates. The mean ± SD is shown. (<b>E</b>,<b>F</b>) The resistance to multiple changes of <span class="html-italic">S. suis</span> HA9801 to 5-FU and TMP after 30 consecutive passages of each drug, and the cross-resistance of 5-FU<sup>R</sup> to commonly used clinical antimicrobials. (<b>G</b>,<b>H</b>) Cytotoxicity of 5-FU at different concentrations on HEP-2 and PK-15 cells.</p>
Full article ">Figure 2
<p>5-FU disrupts the cell wall and bacterial-membrane morphology and induces DNA damage in <span class="html-italic">S. suis</span>. (<b>A</b>) SEM images of <span class="html-italic">S. suis</span> treated with different concentrations of 5-FU. The red arrow indicates swelling and deformation of the bacterial membrane, while the green arrow indicates the bacterial fragments. (<b>B</b>) TEM images of <span class="html-italic">S. suis</span> treated with different concentrations of 5-FU. The green arrow indicates that the bacterial chromatin is not obvious, while the red arrow indicates that the bacterial capsule layer was dissolved, the cell membrane structure of <span class="html-italic">S. suis</span> ruptured, and the cellular contents leaked. (<b>C</b>) LDH release of <span class="html-italic">S. suis</span> under different concentrations of 5-FU treatment. Data are shown as the means ± SDs. ** <span class="html-italic">p</span> &lt; 0.01, **** <span class="html-italic">p</span> &lt; 0.0001, <sup>ns</sup> <span class="html-italic">p</span> &lt; 0.05. (<b>D</b>,<b>E</b>) DNA electrophoretogram and expression levels of <span class="html-italic">recA</span> and <span class="html-italic">radA</span> genes in <span class="html-italic">S. suis</span> under different concentrations of 5-FU treatment. (<b>F</b>) Light-microscopic observation of <span class="html-italic">S. suis</span> treated or untreated with 5-FU.</p>
Full article ">Figure 3
<p>Antibacterial mechanisms of 5-FU in <span class="html-italic">S. suis</span> HA9801. (<b>A</b>) A partial representation of 5-FU metabolic pathway. 5-FU uses the same metabolic mechanisms as uracil to enter the cell, metabolizing into FdUMP, FdUTP, and FUTP to exert antibacterial effects. (<b>B</b>) DNA electrophoretogram in <span class="html-italic">S. suis</span> WT and <span class="html-italic">S. suis</span> Δ<span class="html-italic">thyA</span> treated or untreated with 5-FU. M, marker; 1, WT; 2, WT + 5-FU; 3, Δ<span class="html-italic">thyA</span>; 4, Δ<span class="html-italic">thyA</span> + 5-FU; 5, WT + 5-FU + T. (<b>C</b>) Expression levels of <span class="html-italic">recA</span> and <span class="html-italic">radA</span> genes in <span class="html-italic">S. suis</span> WT and <span class="html-italic">S. suis</span> Δ<span class="html-italic">thyA</span> treated or untreated with 5-FU.</p>
Full article ">Figure 4
<p>Antibacterial experiment in mice. (<b>A</b>) Schematic diagram of mouse infection model. (<b>B</b>) Bacterial load in the lung, liver, and spleen of <span class="html-italic">S. suis</span> WT-infected or <span class="html-italic">S. suis</span> Δ<span class="html-italic">thyA</span>-infected BALB/c mice treated with 5-FU (20 µg/g) and AMX (2 mg/g). (<b>C</b>) Histopathologic changes of <span class="html-italic">S. suis</span> infections caused (black scale bar = 50 µm, magnification = 200×). The black arrows in figure showed the histopathological changes in the lung, liver, and spleen.</p>
Full article ">
16 pages, 1865 KiB  
Article
Antiviral Drug Candidate Repositioning for Streptococcus suis Infection in Non-Tumorigenic Cell Models
by Ashley Anzet van Niekerk, Sara Maluck, Patrik Mag, Csaba Kővágó, Ádám Kerek, Ákos Jerzsele, Torsten Steinmetzer and Erzsébet Pászti-Gere
Biomedicines 2024, 12(4), 783; https://doi.org/10.3390/biomedicines12040783 - 2 Apr 2024
Viewed by 1290
Abstract
The increasing prevalence of antimicrobial resistance against zoonotic bacteria, including Streptococcus (S.) suis, highlights the need for new therapeutical strategies, including the repositioning of drugs. In this study, susceptibilities of bacterial isolates were tested toward ten different 3-amidinophenyalanine (Phe(3-Am)) derivatives via determination [...] Read more.
The increasing prevalence of antimicrobial resistance against zoonotic bacteria, including Streptococcus (S.) suis, highlights the need for new therapeutical strategies, including the repositioning of drugs. In this study, susceptibilities of bacterial isolates were tested toward ten different 3-amidinophenyalanine (Phe(3-Am)) derivatives via determination of minimum inhibitory concentration (MIC) values. Some of these protease inhibitors, like compounds MI-432, MI-471, and MI-476, showed excellent antibacterial effects against S. suis. Their drug interaction potential was investigated using human liver microsomal cytochrome P450 (CYP450) measurements. In our work, non-tumorigenic IPEC-J2 cells and primary porcine hepatocytes were infected with S. suis, and the putative beneficial impact of these inhibitors was investigated on cell viability (Neutral red assay), on interleukin (IL)-6 levels (ELISA technique), and on redox balance (Amplex red method). The antibacterial inhibitors prevented S. suis-induced cell death (except MI-432) and decreased proinflammatory IL-6 levels. It was also found that MI-432 and MI-476 had antioxidant effects in an intestinal cell model upon S. suis infection. Concentration-dependent suppression of CYP3A4 function was found via application of all three inhibitors. In conclusion, our study suggests that the potential antiviral Phe(3-Am) derivatives with 2′,4′ dichloro-biphenyl moieties can be considered as effective drug candidates against S. suis infection due to their antibacterial effects. Full article
(This article belongs to the Special Issue Recent Advances in Drug Repositioning)
Show Figures

Figure 1

Figure 1
<p>Chemical structures of the applied inhibitors.</p>
Full article ">Figure 2
<p>The effect of the protease inhibitors at 10 µM, 25 µM, and 50 µM on human hepatic microsomal CYP3A4 isoenzyme function. The microsomal preparations were treated by the inhibitors for 15 min at 37 °C. The reference inhibitor ketoconazole (INH) was used at a concentration of 5 µM and significantly suppressed CYP3A4 activities in humans (*** <span class="html-italic">p</span> &lt; 0.001). The expressed data are the mean relative fluorescence intensities (RFUs) shown as a percentage of the untreated control fluorescence values ± SD (n = 3); *** signifies <span class="html-italic">p</span> &lt; 0.001 compared to controls.</p>
Full article ">Figure 3
<p>The effect of the protease inhibitors at 50 µM on human hepatic microsomal CYP1A2 (<b>A</b>), 2D6 (<b>B</b>), 2C9 (<b>C</b>), and 2C19 (<b>D</b>) isoenzyme activities. The microsomal preparations were treated by the inhibitors for 15 min at 37 °C. The reference inhibitor α-naphthoflavone (α-NF) used at a concentration of 6 µM significantly suppressed CYP1A2 activities in humans (*** <span class="html-italic">p</span> &lt; 0.001); however, neither of the protease inhibitors at 50 µM changed CYP1A2 activity. The reference inhibitor, quinidine, used at 3 µM, significantly suppressed CYP2D6 activities (* <span class="html-italic">p</span> &lt; 0.05). Furthermore, the reference inhibitors tienilic acid (TA) at 60 µM and (+)-N-3 benzylnirvanol (BN) at 30 µM significantly suppressed CYP2C9 and CYP2C19 function (*** <span class="html-italic">p</span> &lt; 0.001). The expressed data are the mean relative fluorescence intensities (RFUs) shown as a percentage of the untreated control fluorescence values ± SD (n = 3–4); * signifies <span class="html-italic">p</span> &lt; 0.05, and *** signifies <span class="html-italic">p</span> &lt; 0.001 compared to controls.</p>
Full article ">Figure 4
<p>Determination of cell viability in IPEC-J2 cells (<b>A</b>) and swine hepatocytes (<b>B</b>). Changes in cell viability determined by absorbance measurements after 24 h of administration of 10<sup>4</sup> CFU/mL <span class="html-italic">S. suis</span>, (MI-)432, (MI-)471, and (MI-)476 at 50 µM and their combinations. Data are represented as cell viability % expressed in control % with standard errors of mean (SEM), n = 3–4 samples per group. ** indicates <span class="html-italic">p</span> &lt; 0.01 and *** signifies <span class="html-italic">p</span> &lt; 0.001 compared to controls.</p>
Full article ">Figure 5
<p>Measurement of EC H₂O₂ production after treatment with <span class="html-italic">S. suis</span> (10<sup>4</sup> CFU/mL) alone or in combination with the indicated inhibitors (MI-)432, (MI-)471, and (MI-)476 at a concentration of 50 µM in IPEC-J2 cells (<b>A</b>) and in hepatocytes (<b>B</b>) using the Amplex red method. The EC H<sub>2</sub>O<sub>2</sub> levels were determined by measurements of the relative fluorescence intensities compared to the control (100%) with standard errors of mean (SEM), n = 3 samples per group. ** signifies <span class="html-italic">p</span> &lt; 0.01, and *** signifies <span class="html-italic">p</span> &lt; 0.001 compared to controls.</p>
Full article ">Figure 6
<p>Measurement of interleukin-6 production after 24 h treatment with <span class="html-italic">S. suis</span> (10<sup>4</sup> CFU/mL) alone or in combination with the indicated inhibitors (MI-)432, (MI-)471, and (MI-)476 or with the inhibitors alone in IPEC-J2 cells (<b>A</b>) and in hepatocytes (<b>B</b>). Data are represented as IL-6 levels % expressed in control with standard errors of mean (SEM), n = 3–4 samples per group. * signifies <span class="html-italic">p</span> &lt; 0.05 and *** indicates <span class="html-italic">p</span> &lt; 0.001 compared to controls.</p>
Full article ">
10 pages, 1267 KiB  
Communication
Phylogeny of Transferable Oxazolidinone Resistance Genes and Homologs
by Gábor Kardos, Levente Laczkó, Eszter Kaszab, Bálint Timmer, Krisztina Szarka, Eszter Prépost and Krisztián Bányai
Antibiotics 2024, 13(4), 311; https://doi.org/10.3390/antibiotics13040311 - 28 Mar 2024
Viewed by 1398
Abstract
Oxazolidinone resistance, especially transmissible resistance, is a major public health concern, and the origin of this resistance mechanism is not yet resolved. This study aims to delve into the phylogenetic origin of the transmissible oxazolidinone resistance mechanisms conferring cross-resistance to other drugs of [...] Read more.
Oxazolidinone resistance, especially transmissible resistance, is a major public health concern, and the origin of this resistance mechanism is not yet resolved. This study aims to delve into the phylogenetic origin of the transmissible oxazolidinone resistance mechanisms conferring cross-resistance to other drugs of human and veterinary importance. The amino acid sequences of the five cfr ribosomal methylases and optrA and poxtA were used as queries in searches against 219,549 bacterial proteomes in the NCBI RefSeq database. Hits with >40% amino acid identity and >80% query coverage were aligned, and phylogenetic trees were reconstructed. All five cfr genes yielded highly similar trees, with rlmN housekeeping ribosomal methylases located basal to the sister groups of S-adenosyl-methionine-dependent methyltransferases from various Deltaproteobacteria and Actinomycetia, including antibiotic-producing Streptomyces species, and the monophyletic group of cfr genes. The basal branches of the latter contained paenibacilli and other soil bacteria; they then could be split into the clades [cfr(C):cfr(E)] and [[cfr:cfr(B)]:cfr(D)], always with different Bacillaceae in their stems. Lachnospiraceae were encountered in the basal branches of both optrA and poxtA trees. The ultimate origin of the cfr genes is the rlmN housekeeping ribosomal methylases, which evolved into a suicide-avoiding methylase in antibiotic producers; a soil organism (Lachnospiraceae, Paenibacilli) probably acted as a transfer organism into pathogenic bacteria. In the case of optrA, the porcine pathogenic Streptococcus suis was present in all branches, while the proteins closest to poxtA originated from Clostridia. Full article
(This article belongs to the Special Issue The Evolution of Plasmid-Mediated Antimicrobial Resistance)
Show Figures

Figure 1

Figure 1
<p>Phylogeny of genes with cfr (NG070225.1) as a query reconstructed with FastTree using the amino acid hits of the reference gene sequence with matches of &gt;40% amino acid identity and &gt;85% query coverage. The numbers above represent SH-like aLRT support values.</p>
Full article ">Figure 2
<p>Phylogeny of genes with optrA (MF805732.1:8426-10393) as a query reconstructed with FastTree using the amino acid hits of the reference gene sequence with matches of &gt;40% amino acid identity and &gt;85% query coverage. The numbers above represent SH-like aLRT support values.</p>
Full article ">Figure 3
<p>Phylogeny of genes with poxtA (NG_063824.1) as a query reconstructed with FastTree using the amino acid hits of the reference gene sequence with matches of &gt;40% amino acid identity and &gt;85% query coverage. The numbers above represent SH-like aLRT support values.</p>
Full article ">
Back to TopTop