[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.
 
 

Streptococcus suis

A special issue of Pathogens (ISSN 2076-0817).

Deadline for manuscript submissions: closed (31 December 2016) | Viewed by 82955

Special Issue Editor


E-Mail Website1 Website2
Guest Editor
Faculty of Veterinary Medicine, University of Montreal, Montreal, QC H3T 1J4, Canada
Interests: study the cellular and molecular basis of innate and adaptive immunity to capsular polysaccharides of pathogenic bacteria and the role of dendritic cells (DCs) in orchestrating these responses; special interest on Streptococcus suis research and vaccine development
Special Issues, Collections and Topics in MDPI journals

Special Issue Information

Dear Colleagues,

Streptococcus suis is a swine pathogen responsible for important economic losses in the swine industry worldwide. This pathogen has gained more attention since recent recognition of its high prevalence in human meningitis cases in South East and East Asia, and reports of outbreaks which resulted in high mortality rates. Despite the increased incidence and severity of the S. suis infection and the changing epidemiology of this bacterial disease, an effective vaccine to control disease in swine is not really available. Studies in the past few years have identified new potential virulence factors through proteomic and genomic approaches, yet the pathogenesis of the disease is still poorly understood. Identification of new serotypes or reclassification of old ones is also a matter of controversy. For this Special Issue of Pathogens, we invite you to submit research articles, review articles, short notes as well as communications related to S. suis molecular and epidemiological aspects, bacterial–host interactions, the immune response and vaccine development. We look forward to your contribution.

Dr. Mariela Segura
Guest Editor

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. Pathogens is an international peer-reviewed open access monthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. The Article Processing Charge (APC) for publication in this open access journal is 2200 CHF (Swiss Francs). Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • Streptococcus suis
  • serotype
  • virulence factor
  • innate immunity
  • adaptive immunity
  • vaccine
  • animal models

Benefits of Publishing in a Special Issue

  • Ease of navigation: Grouping papers by topic helps scholars navigate broad scope journals more efficiently.
  • Greater discoverability: Special Issues support the reach and impact of scientific research. Articles in Special Issues are more discoverable and cited more frequently.
  • Expansion of research network: Special Issues facilitate connections among authors, fostering scientific collaborations.
  • External promotion: Articles in Special Issues are often promoted through the journal's social media, increasing their visibility.
  • e-Book format: Special Issues with more than 10 articles can be published as dedicated e-books, ensuring wide and rapid dissemination.

Further information on MDPI's Special Issue polices can be found here.

Published Papers (12 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Research

Jump to: Review

3219 KiB  
Article
Evaluation of the Immunomodulatory Properties of Streptococcus suis and Group B Streptococcus Capsular Polysaccharides on the Humoral Response
by Cynthia Calzas, Morgan Taillardet, Insaf Salem Fourati, David Roy, Marcelo Gottschalk, Hugo Soudeyns, Thierry Defrance and Mariela Segura
Pathogens 2017, 6(2), 16; https://doi.org/10.3390/pathogens6020016 - 20 Apr 2017
Cited by 11 | Viewed by 4809
Abstract
Streptococcus suis and group B Streptococcus (GBS) are encapsulated streptococci causing septicemia and meningitis. Antibodies (Abs) against capsular polysaccharides (CPSs) have a crucial protective role, but the structure/composition of the CPS, including the presence of sialic acid, may interfere with the generation of [...] Read more.
Streptococcus suis and group B Streptococcus (GBS) are encapsulated streptococci causing septicemia and meningitis. Antibodies (Abs) against capsular polysaccharides (CPSs) have a crucial protective role, but the structure/composition of the CPS, including the presence of sialic acid, may interfere with the generation of anti-CPS Ab responses. We investigated the features of the CPS-specific Ab response directed against S. suis serotypes 2 and 14 and GBS serotypes III and V after infection or immunization with purified native or desialylated CPSs in mice. Whereas S. suis-infected mice developed a very low/undetectable CPS-specific IgM response, significant anti-CPS IgM titers were measured in GBS-infected animals (especially for type III GBS). No isotype switching was detected in S. suis- or GBS-infected mice. While the expression of sialic acid was essential for the immunogenicity of purified GBS type III CPS, this sugar was not responsible for the inability of purified S. suis types 2, 14 and GBS type V CPSs to induce a specific Ab response. Thus, other biochemical criteria unrelated to the presence of sialic acid may be responsible for the inaptitude of the host immune system to mount an effective response against certain S. suis and GBS CPS types. Full article
(This article belongs to the Special Issue Streptococcus suis)
Show Figures

Figure 1

Figure 1
<p>Titration of capsular polysaccharide (CPS)-specific antibodies in mice after infection with <span class="html-italic">S. suis</span> serotype 2, <span class="html-italic">S. suis</span> serotype 14, group B <span class="html-italic">Streptococcus</span> (GBS) serotype III or GBS serotype V. Mice were infected with 2 × 10<sup>7</sup> CFU of live <span class="html-italic">S. suis</span> serotype 2 strain P1/7 (<span class="html-italic">n</span> = 60) (<b>A</b>); 5 × 10<sup>6</sup> CFU of live <span class="html-italic">S. suis</span> serotype 14 strain DAN13730 (<span class="html-italic">n</span> = 40) (<b>B</b>); 2 × 10<sup>6</sup> CFU of live GBS serotype III strain COH-1 (<span class="html-italic">n</span> = 40) (<b>C</b>); or 10<sup>4</sup> CFU of live GBS serotype V strain CJB111 (<span class="html-italic">n</span> = 40) (<b>D</b>). Total Ig (IgG plus IgM) anti-CPS titers were determined by ELISA on Days 7 (in blue), 14 (in green) and 21 (in red) in surviving mice. IgM and IgG anti-CPS titers were determined on Day 21 (in red). For comparative purpose, total Ig (IgG plus IgM), IgM and IgG anti-protein (‘prot’) titers were also determined on Day 21. Data are presented in a box-and-whiskers diagram with the ends of whiskers representing the minimum and the maximum value. “C<sup>−</sup>” represents a pool of control mice (<span class="html-italic">n</span> = 3) injected with vehicle solution, whose titers were evaluated on Days 7, 14 and 21. * Statistically significant difference (<span class="html-italic">p</span> &lt; 0.05) in comparison to the respective C<sup>−</sup> group.</p>
Full article ">Figure 1 Cont.
<p>Titration of capsular polysaccharide (CPS)-specific antibodies in mice after infection with <span class="html-italic">S. suis</span> serotype 2, <span class="html-italic">S. suis</span> serotype 14, group B <span class="html-italic">Streptococcus</span> (GBS) serotype III or GBS serotype V. Mice were infected with 2 × 10<sup>7</sup> CFU of live <span class="html-italic">S. suis</span> serotype 2 strain P1/7 (<span class="html-italic">n</span> = 60) (<b>A</b>); 5 × 10<sup>6</sup> CFU of live <span class="html-italic">S. suis</span> serotype 14 strain DAN13730 (<span class="html-italic">n</span> = 40) (<b>B</b>); 2 × 10<sup>6</sup> CFU of live GBS serotype III strain COH-1 (<span class="html-italic">n</span> = 40) (<b>C</b>); or 10<sup>4</sup> CFU of live GBS serotype V strain CJB111 (<span class="html-italic">n</span> = 40) (<b>D</b>). Total Ig (IgG plus IgM) anti-CPS titers were determined by ELISA on Days 7 (in blue), 14 (in green) and 21 (in red) in surviving mice. IgM and IgG anti-CPS titers were determined on Day 21 (in red). For comparative purpose, total Ig (IgG plus IgM), IgM and IgG anti-protein (‘prot’) titers were also determined on Day 21. Data are presented in a box-and-whiskers diagram with the ends of whiskers representing the minimum and the maximum value. “C<sup>−</sup>” represents a pool of control mice (<span class="html-italic">n</span> = 3) injected with vehicle solution, whose titers were evaluated on Days 7, 14 and 21. * Statistically significant difference (<span class="html-italic">p</span> &lt; 0.05) in comparison to the respective C<sup>−</sup> group.</p>
Full article ">Figure 2
<p>Titration of CPS-specific antibodies in mice after immunization with purified native or desialylated <span class="html-italic">S. suis</span> serotype 2, <span class="html-italic">S. suis</span> serotype 14, GBS serotype III or GBS serotype V CPS. Mice (<span class="html-italic">n</span> = 8) were immunized with 2 µg of purified native or desialylated <span class="html-italic">S. suis</span> serotype 2 (<b>A</b>); <span class="html-italic">S. suis</span> serotype 14 (<b>B</b>); GBS serotype III (<b>C</b>); or GBS serotype V (<b>D</b>) CPS emulsified with STIMUNE<sup>®</sup>. Total Ig (IgG plus IgM) anti-native CPS titers were determined by ELISA on Days 7, 14 and 21. “C<sup>−</sup>” represents a pool of control mice (<span class="html-italic">n</span> = 3) injected with STIMUNE<sup>®</sup> only, whose titers were evaluated on Days 7, 14 and 21. Data from individual mice are presented, including the geometric mean with 95% confidence interval. * Statistically significant difference (<span class="html-italic">p</span> &lt; 0.05) in comparison to the C<sup>−</sup> group.</p>
Full article ">Figure 3
<p>Adjuvant effect of CpG oligodeoxynucleotides (ODNs) on the CPS-specific humoral response in mice immunized with purified native or desialylated <span class="html-italic">S. suis</span> serotype 2 CPS or purified <span class="html-italic">S. pneumoniae</span> serotype 3 CPS (PS3). (<b>A</b>) In a first set of experiments, mice (<span class="html-italic">n</span> = 8) were immunized with 2 µg of purified native (n) or desialylated (dS) <span class="html-italic">S. suis</span> serotype 2 CPS (CPS <span class="html-italic">S. suis</span>) or PS3 in PBS on Day 0 and 80 µg of CpG ODNs two days after. The control group (<span class="html-italic">n</span> = 8) received CPS or PS3 on Day 0 and PBS two days later. The placebo group (<span class="html-italic">n</span> = 3) received PBS or CpG ODNs only. Total Ig (IgG plus IgM) anti-native <span class="html-italic">S. suis</span> type 2 CPS or anti-PS3 titers were determined by ELISA on Days 7, 14 and 21. Data are presented in a box-and-whiskers diagram with the ends of whiskers representing the minimum and the maximum value. (<b>B</b>) In a second set of experiments, mice (<span class="html-italic">n</span> = 5) were immunized as described in (A), but splenocytes were collected on Day 5 after immunization, and anti-CPS antibody-secreting cells (ASCs) were enumerated by ELISpot as described in the Materials and Methods section. Data are expressed as arithmetic means with SEM. (<b>C</b>) Visualization of PS3 (top) or native <span class="html-italic">S. suis</span> type 2 CPS (bottom) specific ASCs in ELISpot wells from splenocytes of mice obtained in (B). * <span class="html-italic">p</span> &lt; 0.05 between “PS3” and “PS3 + CpG ODNs” groups.</p>
Full article ">Figure 3 Cont.
<p>Adjuvant effect of CpG oligodeoxynucleotides (ODNs) on the CPS-specific humoral response in mice immunized with purified native or desialylated <span class="html-italic">S. suis</span> serotype 2 CPS or purified <span class="html-italic">S. pneumoniae</span> serotype 3 CPS (PS3). (<b>A</b>) In a first set of experiments, mice (<span class="html-italic">n</span> = 8) were immunized with 2 µg of purified native (n) or desialylated (dS) <span class="html-italic">S. suis</span> serotype 2 CPS (CPS <span class="html-italic">S. suis</span>) or PS3 in PBS on Day 0 and 80 µg of CpG ODNs two days after. The control group (<span class="html-italic">n</span> = 8) received CPS or PS3 on Day 0 and PBS two days later. The placebo group (<span class="html-italic">n</span> = 3) received PBS or CpG ODNs only. Total Ig (IgG plus IgM) anti-native <span class="html-italic">S. suis</span> type 2 CPS or anti-PS3 titers were determined by ELISA on Days 7, 14 and 21. Data are presented in a box-and-whiskers diagram with the ends of whiskers representing the minimum and the maximum value. (<b>B</b>) In a second set of experiments, mice (<span class="html-italic">n</span> = 5) were immunized as described in (A), but splenocytes were collected on Day 5 after immunization, and anti-CPS antibody-secreting cells (ASCs) were enumerated by ELISpot as described in the Materials and Methods section. Data are expressed as arithmetic means with SEM. (<b>C</b>) Visualization of PS3 (top) or native <span class="html-italic">S. suis</span> type 2 CPS (bottom) specific ASCs in ELISpot wells from splenocytes of mice obtained in (B). * <span class="html-italic">p</span> &lt; 0.05 between “PS3” and “PS3 + CpG ODNs” groups.</p>
Full article ">Figure 4
<p>In vitro immunomodulatory effect of purified native or desialylated <span class="html-italic">S. suis</span> serotype 2, <span class="html-italic">S. suis</span> serotype 14, GBS serotype III or GBS serotype V CPS on BAFF/IL-4-induced Ig secretion by naive B cells. (<b>A</b>,<b>B</b>) Mouse splenic B cells (10<sup>6</sup> cells/mL) were incubated with purified native <span class="html-italic">S. suis</span> serotype 2, <span class="html-italic">S. suis</span> serotype 14, GBS serotype III or GBS serotype V CPS (each at 20 µg/mL) simultaneously with (central panel) or 24 h before the addition of (right panel) BAFF (1 µg/mL) along with IL-4 (50 ng/mL); (<b>C</b>,<b>D</b>) in order to evaluate the influence of sialic acid, cells were also incubated with desialylated (dS) <span class="html-italic">S. suis</span> serotype 2, <span class="html-italic">S. suis</span> serotype 14, GBS serotype III or GBS serotype V CPS in parallel to cells incubated with the respective native CPS (n) as described in (<b>A</b>,<b>B</b>). After seven days of incubation, supernatants were collected, and total IgM (<b>A</b>,<b>C</b>) and IgG (<b>B</b>,<b>D</b>) was quantified by ELISA. Cells stimulated with BAFF/IL-4 alone (black bars) served as a positive control. In the left panel, cells stimulated with medium (white bars) or each purified CPS alone are represented as an indication of the basal Ig secretion level of cells in absence of BAFF/IL-4 stimulation. Data are expressed as arithmetic means with the SEM of three (central and right panels) or four (left panels) experiments. * <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 5
<p>In vitro immunomodulatory effect of purified native <span class="html-italic">S. suis</span> serotype 2, <span class="html-italic">S. suis</span> serotype 14, GBS serotype III or GBS serotype V CPS on the Ig secretion by naive B cells induced by CpG ODNs. Mouse splenic B cells (10<sup>6</sup> cells/mL) were incubated with purified native <span class="html-italic">S. suis</span> serotype 2, <span class="html-italic">S. suis</span> serotype 14, GBS serotype III or GBS serotype V CPS (each at 20 µg/mL) simultaneously with (central panel) or 24 h before the addition of (right panel) CpG ODNs (1 µg/mL). After seven days of incubation, supernatants were collected and total IgM (<b>A</b>) and IgG (<b>B</b>) were quantified by ELISA. Cells stimulated with CpG ODNs alone (black bars) served as the positive control. In the left panel, cells stimulated with medium (white bars) or each purified CPS alone are represented as an indication of the basal Ig secretion level of cells in absence of stimulation by CpG ODNs. Data are expressed as arithmetic means with the SEM of three (central and right panels) or four (left panel) experiments.</p>
Full article ">Figure 6
<p>In vivo immunomodulatory effect of purified native <span class="html-italic">S. suis</span> serotype 2, <span class="html-italic">S. suis</span> serotype 14, GBS serotype III or GBS serotype V CPS on ovalbumin (OVA)-specific antibody response. Mice (<span class="html-italic">n</span> = 8) were co-immunized intraperitoneally with 10 µg of OVA and 2 µg of purified native <span class="html-italic">S. suis</span> serotype 2 (<b>A</b>), <span class="html-italic">S. suis</span> serotype 14 (<b>B</b>), GBS serotype III (<b>C</b>) or GBS serotype V (<b>D</b>) CPS in PBS on Days 0 and 21. Control group (<span class="html-italic">n</span> = 8) received OVA only on Days 0 and 21. Total Ig (IgG plus IgM) anti-OVA titers were determined by ELISA on Days 7, 14, 21 and 35. “C<sup>−</sup>” represents a pool of placebo mice (<span class="html-italic">n</span> = 3) injected with PBS, whose titers were evaluated on Days 7, 14, 21 and 35. Data from individual mice are presented, including the geometric mean with 95% confidence interval. An arrow indicates secondary immunization. * <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 7
<p>Titration of CPS-specific antibodies in mice after primary immunization with (α-2,3) or (α-2,6) GBS serotype III CPS. Mice (<span class="html-italic">n</span> = 8) were immunized with 2 µg of (α-2,3) or (α-2,6) GBS serotype III CPS emulsified with STIMUNE<sup>®</sup> on Day 0. Total Ig (IgG plus IgM) anti-(α-2,3) (left) or anti-(α-2,6) (right) GBS serotype III CPS titers were determined by ELISA on Days 7 and 21. “C<sup>−</sup>” represents a pool of control mice (<span class="html-italic">n</span> = 3) injected with STIMUNE<sup>®</sup> only, whose anti-(α-2,3) or anti-(α-2,6) GBS serotype III CPS titers were evaluated on Days 7 and 21. Data from individual mice are presented, including the geometric mean with 95% confidence interval. * Statistically significant difference (<span class="html-italic">p</span> &lt; 0.05) in comparison to the C<sup>−</sup> group.</p>
Full article ">
1771 KiB  
Article
Porcine Dendritic Cells as an In Vitro Model to Assess the Immunological Behaviour of Streptococcus suis Subunit Vaccine Formulations and the Polarizing Effect of Adjuvants
by Léa Martelet, Sonia Lacouture, Guillaume Goyette-Desjardins, Guy Beauchamp, Charles Surprenant, Marcelo Gottschalk and Mariela Segura
Pathogens 2017, 6(1), 13; https://doi.org/10.3390/pathogens6010013 - 22 Mar 2017
Cited by 15 | Viewed by 5124
Abstract
An in vitro porcine bone marrow-derived dendritic cell (DC) culture was developed as a model for evaluating immune polarization induced by adjuvants when administered with immunogens that may become vaccine candidates if appropriately formulated. The swine pathogen Streptococcus suis was chosen as a [...] Read more.
An in vitro porcine bone marrow-derived dendritic cell (DC) culture was developed as a model for evaluating immune polarization induced by adjuvants when administered with immunogens that may become vaccine candidates if appropriately formulated. The swine pathogen Streptococcus suis was chosen as a prototype to evaluate proposed S. suis vaccine candidates in combination with the adjuvants Poly I:C, Quil A ®, Alhydrogel ®, TiterMax Gold ® and Stimune ®. The toll-like receptor ligand Poly I:C and the saponin Quil A ® polarized swine DC cytokines towards a type 1 phenotype, with preferential production of IL-12 and TNF-α. The water-in-oil adjuvants TiterMax Gold ® and Stimune ® favoured a type 2 profile as suggested by a marked IL-6 release. In contrast, Alhydrogel ® induced a type 1/type 2 mixed cytokine profile. The antigen type differently modified the magnitude of the adjuvant effect, but overall polarization was preserved. This is the first comparative report on swine DC immune activation by different adjuvants. Although further swine immunization studies would be required to better characterize the induced responses, the herein proposed in vitro model is a promising approach that helps assessing behaviour of the vaccine formulation rapidly at the pre-screening stage and will certainly reduce numbers of animals used while advancing vaccinology science. Full article
(This article belongs to the Special Issue Streptococcus suis)
Show Figures

Figure 1

Figure 1
<p>Cytokine production by bmDCs is differentially modified by enolase in combination with different adjuvants. BmDCs derived from 10 different animals were incubated with enolase (50 μg/mL – final concentration for all conditions), alone or in combination with the adjuvants Poly I:C (50 μg/mL), Quil A ® (5 μg/mL), Alhydrogel ® (50 μg/mL), TiterMax Gold ® (100 μL/well of adjuvant–antigen emulsion) or Stimune ® (100 μL/well of adjuvant–antigen emulsion). Adjuvants alone were also evaluated. Cells incubated with medium served as negative controls (-). Cytokine levels (at 24 h of incubation) were evaluated by ELISA. Data of individuals are presented including mean ± SEM in ng/mL (<span class="html-italic">n</span> = 10). <sup>#</sup> <span class="html-italic">p</span> &lt; 0.0001–0.0005, denotes values that are significantly higher than control (-). * <span class="html-italic">p</span> &lt; 0.0001, denotes values obtained with enolase in combination with each adjuvant that are significantly higher or lower than enolase alone. <sup>a</sup> <span class="html-italic">p</span> &lt; 0.0001–0.001 denotes values obtained with enolase in combination with each adjuvant that are significantly higher than the respective adjuvant alone.</p>
Full article ">Figure 2
<p>Type 1/type 2 cytokine profiles of bmDCs stimulated with enolase in combination with different adjuvants. Data obtained in <a href="#pathogens-06-00013-f001" class="html-fig">Figure 1</a> using bmDCs derived from 10 different animals and incubated with enolase (50 μg/mL – final concentration for all conditions), alone or in combination with Poly I:C (50 μg/mL), Quil A ® (5 μg/mL), Alhydrogel ® (50 μg/mL), TiterMax Gold ® (100 μL/well of adjuvant–antigen emulsion) or Stimune ® (100 μL/well of adjuvant–antigen emulsion) were analyzed using a linear mixed model to determine the polarizing effect of adjuvants. Data are expressed as mean ± SEM in ng/mL (<span class="html-italic">n</span> = 10). * <span class="html-italic">p</span> &lt; 0.0001 denotes values obtained with enolase in combination with each adjuvant that are significantly higher or lower than enolase alone. Letters indicate differences between adjuvants in their capacity to induce the different cytokines (<span class="html-italic">p</span> &lt; 0.0001–0.0005).</p>
Full article ">Figure 3
<p>Cytokine production by bmDCs in response to stimulation by capsular polysaccharide (CPS), tetanus toxoid protein (TT) or CPS-TT conjugate. BmDCs (from three different animals) were incubated with CPS, TT, or CPS-TT conjugate at 25 μg/mL. Cells incubated with medium served as negative controls (-). Cytokine levels (at 24 h of incubation) were evaluated by ELISA. Data are expressed as mean ± SEM in ng/mL (<span class="html-italic">n</span> = 3). <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 denotes values that are significantly higher than control (-).* <span class="html-italic">p</span> &lt; 0.01, denotes values obtained with CPS-TT conjugate that are significantly higher than CPS alone. <sup>a</sup> <span class="html-italic">p</span> &lt; 0.05 denotes values obtained with CPS-TT conjugate that are significantly lower than TT alone.</p>
Full article ">Figure 4
<p>Type 1/type 2 cytokine profiles of bmDCs stimulated with antigens of diverse chemical natures in combination with different adjuvants. BmDCs (from 3 different animals) were incubated with capsular polysaccharide (CPS), tetanus toxoid protein (TT), or CPS-TT conjugate (25 μg/mL – final concentration for all conditions), alone or in combination with the adjuvants Poly I:C (50 μg/mL), Quil A ® (5 μg/mL), Alhydrogel ® (50 μg/mL), TiterMax Gold ® (100 μL/well of adjuvant–antigen emulsion) or Stimune ® (100 μL/well of adjuvant–antigen emulsion). Cytokine levels (at 24 h of incubation) were evaluated by ELISA. Data are expressed as mean ± SEM in ng/mL (<span class="html-italic">n</span> = 3). * <span class="html-italic">p</span> &lt; 0.05 denotes values obtained with CPS, TT or conjugate in combination with each adjuvant that are significantly higher or lower than the antigen alone.</p>
Full article ">
947 KiB  
Communication
Transcriptomic Analysis Reveals Selective Metabolic Adaptation of Streptococcus suis to Porcine Blood and Cerebrospinal Fluid
by Anna Koczula, Michael Jarek, Christian Visscher, Peter Valentin-Weigand, Ralph Goethe and Jörg Willenborg
Pathogens 2017, 6(1), 7; https://doi.org/10.3390/pathogens6010007 - 15 Feb 2017
Cited by 10 | Viewed by 6318
Abstract
Streptococcus suis is a zoonotic pathogen that can cause severe pathologies such as septicemia and meningitis in its natural porcine host as well as in humans. Establishment of disease requires not only virulence of the infecting strain but also an appropriate metabolic activity [...] Read more.
Streptococcus suis is a zoonotic pathogen that can cause severe pathologies such as septicemia and meningitis in its natural porcine host as well as in humans. Establishment of disease requires not only virulence of the infecting strain but also an appropriate metabolic activity of the pathogen in its host environment. However, it is yet largely unknown how the streptococcal metabolism adapts to the different host niches encountered during infection. Our previous isotopologue profiling studies on S. suis grown in porcine blood and cerebrospinal fluid (CSF) revealed conserved activities of central carbon metabolism in both body fluids. On the other hand, they suggested differences in the de novo amino acid biosynthesis. This prompted us to further dissect S. suis adaptation to porcine blood and CSF by RNA deep sequencing (RNA-seq). In blood, the majority of differentially expressed genes were associated with transport of alternative carbohydrate sources and the carbohydrate metabolism (pentose phosphate pathway, glycogen metabolism). In CSF, predominantly genes involved in the biosynthesis of branched-chain and aromatic amino acids were differentially expressed. Especially, isoleucine biosynthesis seems to be of major importance for S. suis in CSF because several related biosynthetic genes were more highly expressed. In conclusion, our data revealed niche-specific metabolic gene activity which emphasizes a selective adaptation of S. suis to host environments. Full article
(This article belongs to the Special Issue Streptococcus suis)
Show Figures

Figure 1

Figure 1
<p>Schematic metabolic model showing important pathways after proliferation of <span class="html-italic">Streptococcus suis</span> serotype 2 in porcine blood (<b>A</b>) and cerebrospinal fluid (CSF) (<b>B</b>). This model is based on the genome annotation of <span class="html-italic">S. suis</span> serotype 2 strain P1/7. Important differentially expressed genes found in our study are depicted by colored boxes. Red boxes and green boxes indicate lower- and higher-expressed genes compared to growth in Todd-Hewitt broth (THB), respectively; black boxes indicate that no differential expression was observed. Metabolic pathways are highlighted in different colors and are simplified: dark green, Embden–Meyerhof–Parnas (EMP) pathway (glycolysis); blue, pentose phosphate pathway (PPP); light purple, glycogen metabolism; red, pyruvate metabolism; brown, incomplete tricarboxylic acid (TCA) cycle; orange, Leloir pathway; yellow, pyrimidine metabolism; light green, purine metabolism; pink, amino acid metabolism. For additional information on gene annotations and metabolite abbreviations used, see <a href="#app1-pathogens-06-00007" class="html-app">Supplementary Table S5</a>.</p>
Full article ">
736 KiB  
Communication
Distribution of Type I Restriction–Modification Systems in Streptococcus suis: An Outlook
by Niels Willemse and Constance Schultsz
Pathogens 2016, 5(4), 62; https://doi.org/10.3390/pathogens5040062 - 18 Nov 2016
Cited by 22 | Viewed by 5380
Abstract
Streptococcus suis is a porcine commensal and pathogen with zoonotic potential. We recently identified a novel Type I restriction–modification (R–M) system in a zoonotic S. suis clone which has emerged in the Netherlands. Here, we describe the DNA inversions in the specificity subunit [...] Read more.
Streptococcus suis is a porcine commensal and pathogen with zoonotic potential. We recently identified a novel Type I restriction–modification (R–M) system in a zoonotic S. suis clone which has emerged in the Netherlands. Here, we describe the DNA inversions in the specificity subunit of this system in S. suis serotype 2, clonal complex 20 and explain the absence of domain movement by the absence of repeats. In addition, we identified a core Type I R–M system present in 95% of the isolates and found an association of the distribution of Type I R–M systems in the S. suis genome with population structure. We speculate on the potential role of Type I R–M systems in S. suis given the recently described associations of Type I R–M systems with virulence and propose future research directions. Full article
(This article belongs to the Special Issue Streptococcus suis)
Show Figures

Figure 1

Figure 1
<p>(<b>A</b>) Schematic representation of a Type I R–M system inspired by Loenen et al. [<a href="#B6-pathogens-05-00062" class="html-bibr">6</a>]. A typical Type I R–M system is a pentameric protein complex consisting of two R subunits, two M subunits and an S subunit encoded by <span class="html-italic">hsdR</span>, <span class="html-italic">hsdM</span> and <span class="html-italic">hsdS</span> genes, respectively. The S subunit consists of two target recognition domains (TRDs), each of which can recognize a different DNA sequence. The two TRDs of the S subunit here are indicated by S1 and S2, separated by a bar which represents a separating amino acids sequence. The two TRD domains are covalently linked as illustrated with a black linker sequence; (<b>B</b>) Alignment of the prophage loci of four representative isolates containing four variants of the SsuCC20P Type I R–M system. DNA sequences are aligned using the Artemis Comparison Tool [<a href="#B18-pathogens-05-00062" class="html-bibr">18</a>] which illustrates the DNA inversions that have taken place and resulted in four different <span class="html-italic">hsdS</span> genes. A complete <span class="html-italic">hsdS</span> gene consists of two Methylase_S (pfam01420) target recognition domains (TRDs), which are colored in the figure to illustrate the rearrangements. The putative four complete S subunits for each of the isolates are respectively; i: green/pink in isolate 2001171, ii: green/purple in isolate 2012092, iii: orange/pink in isolate 931260 and iv: orange/purple in isolate YS12. The two genes indicated in blue are the <span class="html-italic">hsdM</span> and the <span class="html-italic">hsdR</span> gene, respectively. The gene highlighted in cyan is a Type III restriction endonuclease. An integrase (tyrosine recombinase), which likely facilitates the demonstrated DNA inversions is indicated in grey. The complete annotation from 5′ to 3′ is presented in <a href="#app1-pathogens-05-00062" class="html-app">Supplementary Table S1</a>.</p>
Full article ">Figure 2
<p>Phylogenetic tree of all TRDs from SsuCC20P as present in all CC20 isolates. Tips are labelled as isolates with the location of the TRD (first [TRD1] or second [TRD2] location in the S subunit) and are colored according to the position of the TRD in the genetic map in <a href="#pathogens-05-00062-f001" class="html-fig">Figure 1</a>B. TRD1s cluster together before clustering with TRD2s, suggesting that the TRDs have a fixed position in the gene, which limits the possible S subunits variants in SsuCC20P to the observed four variants. Bootstrap values &gt;50% are indicated as percentages on the branches of the maximum likelihood tree, which was constructed with RAxML and are based on 1050 bootstraps.</p>
Full article ">
658 KiB  
Article
Clearance of Streptococcus suis in Stomach Contents of Differently Fed Growing Pigs
by Franziska Warneboldt, Saara J. Sander, Andreas Beineke, Peter Valentin-Weigand, Josef Kamphues and Christoph Georg Baums
Pathogens 2016, 5(3), 56; https://doi.org/10.3390/pathogens5030056 - 6 Aug 2016
Cited by 10 | Viewed by 5154
Abstract
Streptococcus (S.) suis translocates across the intestinal barrier of piglets after intraintestinal application. Based on these findings, an oro-gastrointestinal infection route has been proposed. Thus, the objective of this study was to investigate the survival of S. suis in the porcine [...] Read more.
Streptococcus (S.) suis translocates across the intestinal barrier of piglets after intraintestinal application. Based on these findings, an oro-gastrointestinal infection route has been proposed. Thus, the objective of this study was to investigate the survival of S. suis in the porcine stomach. Whereas surviving bacteria of S. suis serotypes 2 and 9 were not detectable after 60 min of incubation in stomach contents with a comparatively high gastric pH of 5 due to feeding of fine pellets, the number of Salmonella Derby bacteria increased under these conditions. Further experiments confirmed the clearance of S. suis serotypes 2 and 9 within 30 min in stomach contents with a pH of 4.7 independently of the bacterial growth phase. Finally, an oral infection experiment was conducted, feeding each of 18 piglets a diet mixed with 1010 CFU of S. suis serotype 2 or 9. Thorough bacteriological screenings of various mesenteric-intestinal lymph nodes and internal organs after different times of exposure did not lead to any detection of the orally applied challenge strains. In conclusion, the porcine stomach constitutes a very efficient barrier against oro-gastrointenstinal S. suis infections. Conditions leading to the passage of S. suis through the stomach remain to be identified. Full article
(This article belongs to the Special Issue Streptococcus suis)
Show Figures

Figure 1

Figure 1
<p>Mean survival factors of <span class="html-italic">S. suis</span> ST 2 strain 10, ST 9 strain A3286/94 and <span class="html-italic">Salmonella</span> Derby A147/85 as well as pH values in stomach contents ex vivo of piglets fed either a finely ground and pelleted (<b>A</b>) (<span class="html-italic">n</span> = 5) or coarsely ground meal diet (<b>B</b>) (<span class="html-italic">n</span> = 5). Stomach contents were mixed with bacteria and incubated for the indicated time points in air-tight sealed bags at 37 °C in a water bath. Standard deviations (SDs) are not included for reasons of clarity. At <span class="html-italic">t</span> = 3 min SDs were 0.192, 0.267 and 0.049 for <span class="html-italic">S. suis</span> ST2, ST9 and <span class="html-italic">Salmonella</span> Derby in (A), respectively. All other SDs were below 0.02 except for the values in (A) for <span class="html-italic">Salmonella</span> Derby at 60, 120 and 240 min with SD = 0.135; 0.191 and 1.32, respectively. The survival factor of <span class="html-italic">Salmonella</span> Derby was significantly higher at 240 min in comparison to the values at 120, 60 and 3 min (<span class="html-italic">p</span> &lt; 0.05). Differences between survival factors at 120 and 3 min were also significant. The survival factor was calculated by dividing the specific bacterial content at a specific time point (CFU/g) by the inoculation dose.</p>
Full article ">Figure 2
<p>Mean specific bacterial loads of <span class="html-italic">S. suis</span> serotype 2 strain 10 and serotype 9 strain A3286/94 grown either to exponential (exp., OD<sub>600</sub> = 0.6) or to stationary phase (stat., OD<sub>600</sub> = 1.2) as well as pH values in stomach contents ex vivo of piglets (<span class="html-italic">n</span> = 6) fed a finely ground and pelleted diet. Stomach contents were mixed and incubated for the indicated time points in air-tight sealed bags at 37 °C in a water bath. At <span class="html-italic">t</span> = 3 min SDs were 1.3, 24.6, 4.8 and 19.0 for <span class="html-italic">S. suis</span> serotype (ST) 2 (exp. phase), ST2 (stat. phase), ST9 (exp. Phase) and ST9 (stat. phase), respectively. All other SDs were below 0.01. The differences of the specific bacterial loads at t = 3 min compared to the respective values of any other time point of analysis were significant (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 3
<p>Survival factors of <span class="html-italic">Salmonella</span> Derby A147/85, <span class="html-italic">S. suis</span> serotype (ST) 2 strain 10 and <span class="html-italic">S. suis</span> ST 9 strain A3286/94 in compound feed (either in fine pellets without formic acid or as crumb feed including formic acid). Feeds were mixed either with 1.9 × 10<sup>7</sup> CFU <span class="html-italic">Salmonella</span> Derby A147/85, 7.5 × 10<sup>8</sup> CFU <span class="html-italic">S. suis</span> ST 2 strain 10 or 6.8 × 10<sup>8</sup> CFU <span class="html-italic">S. suis</span> ST 9 strain A3286/94 per g feed and incubated for the indicated time points at room temperature (20 to 24 °C). The survival factor was calculated by dividing the specific bacterial content at a specific time point (CFU/g) by the inoculation dose.</p>
Full article ">
2459 KiB  
Article
FlpS, the FNR-Like Protein of Streptococcus suis Is an Essential, Oxygen-Sensing Activator of the Arginine Deiminase System
by Jörg Willenborg, Anna Koczula, Marcus Fulde, Astrid De Greeff, Andreas Beineke, Wolfgang Eisenreich, Claudia Huber, Maren Seitz, Peter Valentin-Weigand and Ralph Goethe
Pathogens 2016, 5(3), 51; https://doi.org/10.3390/pathogens5030051 - 21 Jul 2016
Cited by 12 | Viewed by 11548
Abstract
Streptococcus (S.) suis is a zoonotic pathogen causing septicemia and meningitis in pigs and humans. During infection S. suis must metabolically adapt to extremely diverse environments of the host. CcpA and the FNR family of bacterial transcriptional regulators are important for metabolic gene [...] Read more.
Streptococcus (S.) suis is a zoonotic pathogen causing septicemia and meningitis in pigs and humans. During infection S. suis must metabolically adapt to extremely diverse environments of the host. CcpA and the FNR family of bacterial transcriptional regulators are important for metabolic gene regulation in various bacteria. The role of CcpA in S. suis is well defined, but the function of the FNR-like protein of S. suis, FlpS, is yet unknown. Transcriptome analyses of wild-type S. suis and a flpS mutant strain suggested that FlpS is involved in the regulation of the central carbon, arginine degradation and nucleotide metabolism. However, isotopologue profiling revealed no substantial changes in the core carbon and amino acid de novo biosynthesis. FlpS was essential for the induction of the arcABC operon of the arginine degrading pathway under aerobic and anaerobic conditions. The arcABC-inducing activity of FlpS could be associated with the level of free oxygen in the culture medium. FlpS was necessary for arcABC-dependent intracellular bacterial survival but redundant in a mice infection model. Based on these results, we propose that the core function of S. suis FlpS is the oxygen-dependent activation of the arginine deiminase system. Full article
(This article belongs to the Special Issue Streptococcus suis)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Influence of FlpS knock-out on global gene expression during growth of <span class="html-italic">S. suis</span>. (<b>A</b>) Summary of significantly differentially expressed genes during exp and stat growth of <span class="html-italic">S. suis</span> strain 10Δ<span class="html-italic">flpS</span> and classification of clusters of orthologous groups (COG). C, energy production and conversion; D, cell cycle control, cell division; E, amino acid transport and metabolism; F, nucleotide transport and metabolism; G, carbohydrate transport and metabolism; H, coenzyme transport and metabolism; I, lipid transport and metabolism; J, translation; K, transcription; L, replication, recombination and repair; M, cell wall/membrane biogenesis; O, post-translational modification, protein turnover, chaperones; P, inorganic ion transport and metabolism; R, general function prediction only; S, function unknown; T, signal transduction mechanisms; V, defense mechanisms; [−], no prediction; (<b>B</b>) Venn diagram illustration of the number of significant differentially expressed genes during exp and stat growth of <span class="html-italic">S. suis</span> strain 10Δ<span class="html-italic">flpS</span>; (<b>C</b>) Venn diagram illustration of the number of significant differentially expressed genes during exp and stat growth of <span class="html-italic">S. suis</span> strains 10Δ<span class="html-italic">flpS</span> and 10Δ<span class="html-italic">ccpA</span> [<a href="#B34-pathogens-05-00051" class="html-bibr">34</a>].</p>
Full article ">Figure 2
<p>Metabolic characterization of <span class="html-italic">S. suis</span> strain 10Δ<span class="html-italic">flpS</span>. (<b>A</b>) The wild-type strain 10 and strain 10Δ<span class="html-italic">flpS</span> were grown in CDM medium containing 40 mM of monosaccharides (<b>left</b> panel) or di-/trisaccharides (<b>right</b> panel) indicated, and OD<sub>630</sub> values were recorded at one-hour intervals automatically in a thermostatic 96-well microplate reader. Results and standard deviations are shown for three biological replicates. Carbohydrate substrates that could not be used for streptococcal growth in CDM are marked by asterisks; (<b>B</b>) Color map for the overall <sup>13</sup>C excess (mol %) of labeled amino acids after growth of <span class="html-italic">S. suis</span> strains in the presence of [U-<sup>13</sup>C<sub>6</sub>]glucose in THB media. Notably, only overall <sup>13</sup>C excesses above 0.5 mol % were considered as sufficient labeling rates. The results are shown for exp and stat grown bacteria. Mean values of two biological replicates for which MS measurements were performed in triplicate are given.</p>
Full article ">Figure 2 Cont.
<p>Metabolic characterization of <span class="html-italic">S. suis</span> strain 10Δ<span class="html-italic">flpS</span>. (<b>A</b>) The wild-type strain 10 and strain 10Δ<span class="html-italic">flpS</span> were grown in CDM medium containing 40 mM of monosaccharides (<b>left</b> panel) or di-/trisaccharides (<b>right</b> panel) indicated, and OD<sub>630</sub> values were recorded at one-hour intervals automatically in a thermostatic 96-well microplate reader. Results and standard deviations are shown for three biological replicates. Carbohydrate substrates that could not be used for streptococcal growth in CDM are marked by asterisks; (<b>B</b>) Color map for the overall <sup>13</sup>C excess (mol %) of labeled amino acids after growth of <span class="html-italic">S. suis</span> strains in the presence of [U-<sup>13</sup>C<sub>6</sub>]glucose in THB media. Notably, only overall <sup>13</sup>C excesses above 0.5 mol % were considered as sufficient labeling rates. The results are shown for exp and stat grown bacteria. Mean values of two biological replicates for which MS measurements were performed in triplicate are given.</p>
Full article ">Figure 3
<p>Mice infection with different <span class="html-italic">S. suis</span> regulator mutant strains. (<b>A</b>) Intranasal infection of mice. Specific bacterial loads of tracheonasal lavage (TNL) and indicated inner organs of mice (3 d.p.i.) intranasally infected with indicated <span class="html-italic">S. suis</span> strains. Each symbol represents one individual animal and medians are indicated by horizontal lines. Statistical testing was done for each TNL or organ by a Kruskal-Wallis test with a post hoc Dunn’s multiple comparisons test; (<b>B</b>) Intravenous infection of mice. The upper panel shows specific bacterial loads of blood samples and indicated inner organs of mice intravenously infected with indicated <span class="html-italic">S. suis</span> strains. Statistical testing was done for each blood sample or organ by a Kruskal-Wallis test with a post hoc Dunn’s multiple comparisons test. In the lower panel the respective Kaplan-Meier diagram for mortality of mice is shown. Significant difference is indicated by * with <span class="html-italic">p</span> &lt; 0.05 (log-rank test).</p>
Full article ">Figure 4
<p>FlpS is essential for <span class="html-italic">arcABC</span> operon expression and fitness of <span class="html-italic">S. suis</span>. (<b>A</b>) Immunoblot analyses of whole-cell lysates of <span class="html-italic">S. suis</span> strains grown to stat phase in THB medium under anaerobic conditions. Immunoblot for wild-type strain 10, strain 10Δ<span class="html-italic">flpS</span> and complemented mutant strain 10Δ<span class="html-italic">flpScomp</span> probed with polyclonal antisera raised against recombinant ArcB; (<b>B</b>) Real-time qRT-PCR experiments of <span class="html-italic">S. suis</span> strain 10 and strain 10Δ<span class="html-italic">flpS</span> grown under standard batch and anaerobic conditions. Fold changes in relative <span class="html-italic">arcB</span> transcript levels were shown for a time kinetic as described in Materials and Methods. Results for two independent experiments are depicted; (<b>C</b>) Real-time qRT-PCR experiments of <span class="html-italic">S. suis</span> strain 10, strain 10Δ<span class="html-italic">flpS</span> and strain 10Δ<span class="html-italic">flpScomp</span> grown under batch and shaking (shake) conditions. Fold changes in relative <span class="html-italic">arcB</span> transcript levels were shown for a time kinetic as described in Materials and Methods. Data from three biological replicates and are shown as means ± SEM. Statistical analysis was performed using one-way ANOVA followed by a post-Tukey test (**, <span class="html-italic">p</span> &lt; 0.01); (<b>D</b>) GFP reporter assay. Reporter plasmids carrying the GFP under control of the <span class="html-italic">arcABC</span> promoter were transformed in <span class="html-italic">S. suis</span> wild-type strain 10 and strain 10Δ<span class="html-italic">flpS</span>. Bars represent the relative fluorescence units (RFU) after normalization to the values obtained for strain 10 carrying the promoterless <span class="html-italic">gfp</span> construct (10::<span class="html-italic">gfp</span>). Experiments were carried out in triplicate and repeated twice; (<b>E</b>) Intracellular survival of the unencapsulated strain 10∆<span class="html-italic">cpsEF</span> (<b>black</b> bars) and its <span class="html-italic">flpS</span> mutant strain 10∆<span class="html-italic">cpsEF</span>∆<span class="html-italic">flpS</span> (<b>white</b> bars) in HEp-2 cells. HEp-2 cells were either treated with 200 nM bafilomycin (+Baf) for 1 h before infection to inhibit endosomal acidification or left untreated (−Baf). Results are given as percentage of intracellular bacterial survival after 2 h. Data represent means and standard deviation of two independent experiments performed in duplicates. Results were considered statistically significant with <span class="html-italic">p</span> &lt; 0.05 in a two-tailed <span class="html-italic">t</span>-test, as indicated by asterisks.</p>
Full article ">
3098 KiB  
Article
Virulence Studies of Different Sequence Types and Geographical Origins of Streptococcus suis Serotype 2 in a Mouse Model of Infection
by Jean-Philippe Auger, Nahuel Fittipaldi, Marie-Odile Benoit-Biancamano, Mariela Segura and Marcelo Gottschalk
Pathogens 2016, 5(3), 48; https://doi.org/10.3390/pathogens5030048 - 11 Jul 2016
Cited by 45 | Viewed by 6337
Abstract
Multilocus sequence typing previously identified three predominant sequence types (STs) of Streptococcus suis serotype 2: ST1 strains predominate in Eurasia while North American (NA) strains are generally ST25 and ST28. However, ST25/ST28 and ST1 strains have also been isolated in Asia and NA, [...] Read more.
Multilocus sequence typing previously identified three predominant sequence types (STs) of Streptococcus suis serotype 2: ST1 strains predominate in Eurasia while North American (NA) strains are generally ST25 and ST28. However, ST25/ST28 and ST1 strains have also been isolated in Asia and NA, respectively. Using a well-standardized mouse model of infection, the virulence of strains belonging to different STs and different geographical origins was evaluated. Results demonstrated that although a certain tendency may be observed, S. suis serotype 2 virulence is difficult to predict based on ST and geographical origin alone; strains belonging to the same ST presented important differences of virulence and did not always correlate with origin. The only exception appears to be NA ST28 strains, which were generally less virulent in both systemic and central nervous system (CNS) infection models. Persistent and high levels of bacteremia accompanied by elevated CNS inflammation are required to cause meningitis. Although widely used, in vitro tests such as phagocytosis and killing assays require further standardization in order to be used as predictive tests for evaluating virulence of strains. The use of strains other than archetypal strains has increased our knowledge and understanding of the S. suis serotype 2 population dynamics. Full article
(This article belongs to the Special Issue Streptococcus suis)
Show Figures

Figure 1

Figure 1
<p>Timeline summary of the intraperitoneal and intracisternal mouse models of infection used throughout this study. C57BL/6 mice were infected with the different <span class="html-italic">S. suis</span> serotype 2 strains using the intraperitoneal route of infection (<b>A</b>) to evaluate the systemic and subsequent central nervous system infection; or the transcutaneal intracisternal route of infection (<b>B</b>) to directly evaluate the central nervous system infection.</p>
Full article ">Figure 2
<p>Blood bacterial burden is lower in North American ST28-infected mice but similar in ST1, ST25 or Eurasian ST28-infected mice during the systemic infection. C57BL/6 mice were inoculated by intraperitoneal injection with 5 × 10<sup>7</sup> CFU and blood bacterial titers evaluated 24 h (<b>A</b>); 36 h (<b>B</b>) and 48 h (<b>C</b>) post-infection (p.i.). Data of individual mice are presented as log<sub>10</sub> CFU/mL with the geometric mean. Significance between groups is indicated by different letters (<span class="html-italic">p</span> &lt; 0.001). Only strains for which five or more mice survived at the indicated time point are presented.</p>
Full article ">Figure 3
<p>Plasma cytokine production is lowest in North American ST28-infected mice, intermediate in Eurasian ST28-infected mice, and highest in ST1- and ST25-infected mice during systemic infection. Plasma cytokine levels 12 h post-intraperitoneal inoculation of mock- (vehicle) or 5 × 10<sup>7</sup> CFU of <span class="html-italic">S. suis</span>-infected C57BL/6 mice, as determined by Luminex<sup>®</sup> for TNF-α (<b>A</b>); IL-6 (<b>B</b>); IL-12p70 (<b>C</b>); and IFN-γ (<b>D</b>). Data of individual mice are presented as pg/mL with the mean. Significance between groups is indicated by different letters (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 4
<p>Plasma chemokine production is lowest in North American ST28-infected mice, intermediate in Eurasian ST28-infected mice and highest in ST1- and ST25-infected mice during the systemic infection. Plasma chemokine levels 12 h post-intraperitoneal inoculation of mock- (vehicle) or 5 × 10<sup>7</sup> CFU of <span class="html-italic">S. suis</span>-infected C57BL/6 mice, as determined by Luminex<sup>®</sup> for CCL2 (<b>A</b>); CCL3 (<b>B</b>); CCL4 (<b>C</b>); CCL5 (<b>D</b>), and CXCL1 (<b>E</b>). Data of individual mice are presented as pg/mL with the mean. Significance between groups is indicated by different letters (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 5
<p>Histopathological studies of the brains of C57BL/6 mice infected by intraperitoneal inoculation during central nervous system infection. Presence or absence of histopathological lesions of meningitis as determined in the brains of mock-infected (vehicle) and infected mice. Micrographs of the meninges or ventricular choroid plexus of mock-infected mice (<b>A</b>); NA ST28 strain 1088563- (<b>B</b>); EA ST28 strain MNCM43- (<b>C</b>); NA ST25 strain 89-1591- (<b>D</b>); and EA ST1 strain P1/7- (<b>E</b>) infected mice. Black arrowheads indicate lesions typical of <span class="html-italic">S. suis</span> meningitis. HPS staining, 100× magnification. NA = North America; EA = Eurasia.</p>
Full article ">Figure 6
<p>Lower virulence North American ST28 strains do not induce meningitis in C57BL/6 mice following intracisternal inoculation. Presence or absence of histopathological lesions of meningitis in the brains of mock-infected (vehicle) and <span class="html-italic">S. suis</span>-infected C57BL/6 mice following intracisternal inoculation. Micrographs of the meninges or ventricular choroid plexus of mock-infected (<b>A</b>); NA ST28 strain 1054471- (<b>B</b>); NA ST28 strain 1088563- (<b>C</b>); and EA ST1 strain P1/7- (<b>D</b>) infected mice. Black arrowheads indicate lesions typical of <span class="html-italic">S. suis</span> meningitis. HPS staining, 100× magnification. NA = North America; EA = Eurasia.</p>
Full article ">Figure 7
<p>Brain and blood bacterial titers of lower virulence North American ST28 strains are transient, while those of the ST1 strain are persistent, following intracisternal inoculation. C57BL/6 mice were inoculated by intracisternal injection with 2 × 10<sup>5</sup> CFU and brain and blood bacterial titers evaluated 12 h (<b>A</b> &amp; <b>C</b>) and 24 h (<b>B</b> &amp; <b>D</b>) post-infection (p.i.). Data of individual mice are presented as log<sub>10</sub> CFU/g or CFU/mL with the geometric mean. * Indicates a significant difference between the Eurasian ST1 strain P1/7 and both North American ST28 strains (1054471 and 1088563) (<span class="html-italic">p</span> &lt; 0.01).</p>
Full article ">Figure 8
<p>Plasma (P) and brain (B) homogenate cytokine and chemokines levels of mock-infected (vehicle) or C57BL/6 mice inoculated by intraperitoneal injection with <span class="html-italic">S. suis</span> serotype 2 European ST1 strain P/7, upon presentation of clinical signs of septic shock or meningitis, by ELISA for IL-1β (<b>A</b>); IL-6 (<b>B</b>); CCL2 (<b>C</b>); CCL3 (<b>D</b>); CXCL1 (<b>E</b>); and CXCL10 (<b>F</b>). Data are presented as mean ± SEM pg/mL.</p>
Full article ">Figure 9
<p>Production of brain cytokines and chemokines following <span class="html-italic">S. suis</span> serotype 2 infection only occurs in the presence of meningitis. Brain cytokine and chemokine levels of mock-infected (vehicle) or <span class="html-italic">S. suis</span>-infected C57BL/6 mice inoculated by intraperitoneal or intracisternal injection as determined by ELISA. Intraperitoneally injected mice were euthanized upon presentation of clinical signs of meningitis or at the end of the study (14 days post-infection) and intracisternally injected mice 24 h post-infection or at the end of the study (72 h post-infection). Brain levels of IL-1β (<b>A</b>); IL-6 (<b>B</b>); CCL2 (<b>C</b>); CCL3 (<b>D</b>); CXCL1 (<b>E</b>); and CXCL10 (<b>F</b>) following infection with EA ST1 strain P1/7, NA ST25 strain 89-1591, NA ST28 strain 1088563, or EA ST28 strain MNCM43. Data are presented as mean ± SEM pg/mL. * Indicates a significant difference with mock-infected mice (<span class="html-italic">p</span> &lt; 0.05). EA = Eurasian; NA = North American.</p>
Full article ">
4480 KiB  
Article
Recruitment of Factor H to the Streptococcus suis Cell Surface is Multifactorial
by David Roy, Daniel Grenier, Mariela Segura, Annabelle Mathieu-Denoncourt and Marcelo Gottschalk
Pathogens 2016, 5(3), 47; https://doi.org/10.3390/pathogens5030047 - 7 Jul 2016
Cited by 23 | Viewed by 5063
Abstract
Streptococcus suis is an important bacterial swine pathogen and a zoonotic agent. Recently, two surface proteins of S. suis, Fhb and Fhbp, have been described for their capacity to bind factor H—a soluble complement regulatory protein that protects host cells from complement-mediated [...] Read more.
Streptococcus suis is an important bacterial swine pathogen and a zoonotic agent. Recently, two surface proteins of S. suis, Fhb and Fhbp, have been described for their capacity to bind factor H—a soluble complement regulatory protein that protects host cells from complement-mediated damages. Results obtained in this study showed an important role of host factor H in the adhesion of S. suis to epithelial and endothelial cells. Both Fhb and Fhbp play, to a certain extent, a role in such increased factor H-dependent adhesion. The capsular polysaccharide (CPS) of S. suis, independently of the presence of its sialic acid moiety, was also shown to be involved in the recruitment of factor H. However, a triple mutant lacking Fhb, Fhbp and CPS was still able to recruit factor H resulting in the degradation of C3b in the presence of factor I. In the presence of complement factors, the double mutant lacking Fhb and Fhbp was similarly phagocytosed by human macrophages and killed by pig blood when compared to the wild-type strain. In conclusion, this study suggests that recruitment of factor H to the S. suis cell surface is multifactorial and redundant. Full article
(This article belongs to the Special Issue Streptococcus suis)
Show Figures

Figure 1

Figure 1
<p>Western blot showing factor H binding protein (Fhbp) expression in <span class="html-italic">S. suis</span> wild-type strain P1/7 and complemented ∆<span class="html-italic">fhbp</span> mutant but not in the isogenic ∆<span class="html-italic">fhbp</span> mutant. Whole bacteria of <span class="html-italic">S. suis</span> wild-type strain P1/7 (lane 2), ∆<span class="html-italic">fhbp</span> mutant (lane 3) and complemented ∆<span class="html-italic">fhbp</span> mutant (lane 4) were tested for Fhbp expression. Samples were separated by SDS-polyacrylamide gel electrophoresis and transferred to a nitrocellulose membrane. Fhbp protein was detected with a monospecific rabbit polyclonal antiserum against Fhbp. Fhbp protein was not detected in ∆<span class="html-italic">fhbp</span> mutant, whereas a clear positive reaction was obtained for the wild-type strain and the complemented mutant. Molecular weights in kDa are indicated on the left side of the figure.</p>
Full article ">Figure 2
<p>Effect of factor H on cell adhesion and invasion by <span class="html-italic">S. suis</span> wild-type strain P1/7. <span class="html-italic">S. suis</span> adhesion to (<b>A</b>) human lung epithelial cells A549 and (<b>B</b>) human brain microvascular endothelial cells (hBMEC). Results were determined after 1 h exposure of A549 and hBMEC cells to <span class="html-italic">S. suis</span>, followed by extensive washing of non-adherent bacteria and cell lysis to obtain <span class="html-italic">S. suis</span> viable counts. Results are expressed as recovered CFU/mL. Significant differences between the wild-type strain P1/7 preincubated with factor H and the same strain preincubated in phosphate buffered saline (PBS) were observed for both A549 and hBMEC cells (** <span class="html-italic">p</span> = 0.006 for A549 and * <span class="html-italic">p</span> = 0.04 for hBMEC), as determined by one-way ANOVA. <span class="html-italic">S. suis</span> invasion of (<b>C</b>) human lung epithelial cells A549 and (<b>D</b>) hBMEC. Results were determined after 1 h exposure of cells to <span class="html-italic">S. suis</span>, followed by antibiotic treatment to kill extracellular bacteria and by cell lysis to obtain <span class="html-italic">S. suis</span> viable counts. No significant differences were observed. Data are expressed as mean ± standard error of mean (SEM) of at least four independent experiments.</p>
Full article ">Figure 2 Cont.
<p>Effect of factor H on cell adhesion and invasion by <span class="html-italic">S. suis</span> wild-type strain P1/7. <span class="html-italic">S. suis</span> adhesion to (<b>A</b>) human lung epithelial cells A549 and (<b>B</b>) human brain microvascular endothelial cells (hBMEC). Results were determined after 1 h exposure of A549 and hBMEC cells to <span class="html-italic">S. suis</span>, followed by extensive washing of non-adherent bacteria and cell lysis to obtain <span class="html-italic">S. suis</span> viable counts. Results are expressed as recovered CFU/mL. Significant differences between the wild-type strain P1/7 preincubated with factor H and the same strain preincubated in phosphate buffered saline (PBS) were observed for both A549 and hBMEC cells (** <span class="html-italic">p</span> = 0.006 for A549 and * <span class="html-italic">p</span> = 0.04 for hBMEC), as determined by one-way ANOVA. <span class="html-italic">S. suis</span> invasion of (<b>C</b>) human lung epithelial cells A549 and (<b>D</b>) hBMEC. Results were determined after 1 h exposure of cells to <span class="html-italic">S. suis</span>, followed by antibiotic treatment to kill extracellular bacteria and by cell lysis to obtain <span class="html-italic">S. suis</span> viable counts. No significant differences were observed. Data are expressed as mean ± standard error of mean (SEM) of at least four independent experiments.</p>
Full article ">Figure 3
<p>Effect of the deletion of <span class="html-italic">fhb</span> and <span class="html-italic">fhbp</span> on the <span class="html-italic">S. suis</span> adhesion to A549 and hBMEC cells in the presence or absence of factor H. Adhesion of <span class="html-italic">S. suis</span> factor H binding (Fhb) and Fhbp deficient mutants to (<b>A</b>,<b>C</b>) human lung epithelial cells A549 and to (<b>B</b>,<b>D</b>) hBMEC in presence (<b>A,B</b>) or absence (<b>C,D</b>) of human factor H. Experiments were performed as described in <a href="#pathogens-05-00047-f002" class="html-fig">Figure 2</a>. Results are expressed as recovered CFU/mL. Significant differences between the double knock-out ∆<span class="html-italic">fhb/</span>∆<span class="html-italic">fhbp</span> mutant and wild-type strain P1/7 as well as single mutants were observed in presence of factor H for both A549 (* <span class="html-italic">p</span> = 0.0279) and hBMEC cells (* <span class="html-italic">p</span> = 0.0214), as determined by one-way ANOVA. No significant differences were observed between the wild-type strain P1/7 and single deletion mutants (∆<span class="html-italic">fhb</span> and ∆<span class="html-italic">fhbp</span>). Data are expressed as mean ± SEM of at least four independent experiments.</p>
Full article ">Figure 3 Cont.
<p>Effect of the deletion of <span class="html-italic">fhb</span> and <span class="html-italic">fhbp</span> on the <span class="html-italic">S. suis</span> adhesion to A549 and hBMEC cells in the presence or absence of factor H. Adhesion of <span class="html-italic">S. suis</span> factor H binding (Fhb) and Fhbp deficient mutants to (<b>A</b>,<b>C</b>) human lung epithelial cells A549 and to (<b>B</b>,<b>D</b>) hBMEC in presence (<b>A,B</b>) or absence (<b>C,D</b>) of human factor H. Experiments were performed as described in <a href="#pathogens-05-00047-f002" class="html-fig">Figure 2</a>. Results are expressed as recovered CFU/mL. Significant differences between the double knock-out ∆<span class="html-italic">fhb/</span>∆<span class="html-italic">fhbp</span> mutant and wild-type strain P1/7 as well as single mutants were observed in presence of factor H for both A549 (* <span class="html-italic">p</span> = 0.0279) and hBMEC cells (* <span class="html-italic">p</span> = 0.0214), as determined by one-way ANOVA. No significant differences were observed between the wild-type strain P1/7 and single deletion mutants (∆<span class="html-italic">fhb</span> and ∆<span class="html-italic">fhbp</span>). Data are expressed as mean ± SEM of at least four independent experiments.</p>
Full article ">Figure 4
<p>Deposition of factor H to the <span class="html-italic">S. suis</span> cell surface: role of Fhb and Fhbp. Deposition of factor H to the bacterial cell surface was detected using an ELISA assay. <span class="html-italic">Streptococcus mutans</span> was included as a negative control for factor H binding. There were statistically significant differences between all <span class="html-italic">S. suis</span> strains and <span class="html-italic">S. mutans</span> as determined by one-way ANOVA (** <span class="html-italic">p</span> &lt; 0.01). No significant differences were observed between the <span class="html-italic">S. suis</span> wild-type strain P1/7 and isogenic mutants ∆<span class="html-italic">fhb</span>, ∆<span class="html-italic">fhbp</span> and ∆<span class="html-italic">fhb</span>/∆<span class="html-italic">fhbp</span>.</p>
Full article ">Figure 5
<p>Deposition of factor H to the <span class="html-italic">S. suis</span> cell surface: role of capsular polysaccharide (CPS) and its sialic acid moiety. Results of ELISA showing binding of factor H to (<b>A</b>) non-encapsulated <span class="html-italic">S. suis</span> and to (<b>B</b>) <span class="html-italic">S. suis</span> purified CPS. There were statistically significant differences between groups for <a href="#pathogens-05-00047-f005" class="html-fig">Figure 5</a>A,B as determined by one-way ANOVA. In <a href="#pathogens-05-00047-f005" class="html-fig">Figure 5</a>A, significant differences with the wild-type strain are depicted with asterisks (* <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01). Data are expressed as mean ± SEM of at least three independent experiments. In <a href="#pathogens-05-00047-f005" class="html-fig">Figure 5</a>B, different concentrations (0.1 and 1 μg/mL) of precoated purified <span class="html-italic">S. suis</span> native and desialylated CPS were incubated with factor H (10 μg/mL). Significant differences were observed with factor H incubated with native and desialylated CPS at 1 μg/mL vs. control incubated without CPS (** <span class="html-italic">p</span> = 0.0006 and ** <span class="html-italic">p</span> = 0.0012, respectively). No significant differences were observed between native and desialylated CPS in their capacity to bind factor H (<span class="html-italic">p</span> &gt; 0.05).</p>
Full article ">Figure 6
<p>Factor-I cofactor assay showing C3b degradation by <span class="html-italic">S. suis</span> strains. Immunoblot shows that factor H bound to <span class="html-italic">S. suis</span> strains serves as cofactor for factor I (FI)-mediated cleavage of C3b, resulting in formation of an α’68 kDa chain. Lane: 1, molecular mass marker; 2, Wild-type strain P1/7 alone; 3, Wild-type strain P1/7 + FI + C3b; 4, Wild-type strain P1/7 + FI + FH; 5, Wild-type strain P1/7 + FH + C3b; 6, Wild-type strain P1/7 + FH + FI + C3b; 7, Δ<span class="html-italic">fhb</span> mutant strain + FH + FI + C3b; 8, Δ<span class="html-italic">fhbp</span> mutant strain + FH + FI + C3b; 9, Δ<span class="html-italic">fhb/</span>Δ<span class="html-italic">fhbp</span> mutant strain + FH + FI + C3b; 10, Δ<span class="html-italic">cps2F</span> mutant strain + FH + FI + C3b; 11, Δ<span class="html-italic">fhb</span>/Δ<span class="html-italic">fhbp</span>/Δ<span class="html-italic">cps2F</span> mutant strain + FH + FI + C3b; and 12, molecular mass marker. All strains retained the capacity to bound factor H in a way that serves as cofactor for factor I-mediated cleavage.</p>
Full article ">Figure 7
<p>Phagocytosis of <span class="html-italic">S. suis</span> strains by THP-1 human macrophages in presence of complement-rich serum. Bacteria (1 × 10<sup>7</sup> CFU/mL) were incubated for 90 min with cells (MOI = 100) in presence of human serum, followed by gentamicin/penicillin G treatment to kill any remaining extracellular bacteria after incubation. Intracellular counts were done after three washes and cell lysis with water. Results represent the mean (CFU/mL) ± SEM of four independent experiments. There were no statistical differences between the <span class="html-italic">S. suis</span> wild-type and any of the factor H-binding protein mutants. The non-encapsulated mutant (positive control) was significantly more phagocytosed as determined by one-way ANOVA (** <span class="html-italic">p</span> &lt; 0.01).</p>
Full article ">Figure 8
<p>Killing of <span class="html-italic">S. suis</span> by swine whole blood cells. Bacteria (5 × 10<sup>5</sup> CFU) were incubated for 120 min with swine whole blood or with blood serum (bacteria alone). The percentage of killed bacteria was calculated as follows: 1 – (Bacteria recovered in blood/bacteria recovered in serum) × 100%. Data are expressed as mean ± SEM of at least three independent experiments. There were not statistical differences between the <span class="html-italic">S. suis</span> wild-type and any of the factor H-binding protein mutants. The non-encapsulated mutant (positive control) was significantly more killed as determined by one-way ANOVA (** <span class="html-italic">p</span> &lt; 0.01).</p>
Full article ">
749 KiB  
Article
Simultaneous Quantification and Differentiation of Streptococcus suis Serotypes 2 and 9 by Quantitative Real-Time PCR, Evaluated in Tonsillar and Nasal Samples of Pigs
by Niels Dekker, Ineke Daemen, Koen Verstappen, Astrid De Greeff, Hilde Smith and Birgitta Duim
Pathogens 2016, 5(3), 46; https://doi.org/10.3390/pathogens5030046 - 30 Jun 2016
Cited by 12 | Viewed by 12179
Abstract
Invasive Streptococcus suis (S. suis) infections in pigs are often associated with serotypes 2 and 9. Mucosal sites of healthy pigs can be colonized with these serotypes, often multiple serotypes per pig. To unravel the contribution of these serotypes in pathogenesis [...] Read more.
Invasive Streptococcus suis (S. suis) infections in pigs are often associated with serotypes 2 and 9. Mucosal sites of healthy pigs can be colonized with these serotypes, often multiple serotypes per pig. To unravel the contribution of these serotypes in pathogenesis and epidemiology, simultaneous quantification of serotypes is needed. A quantitative real-time PCR (qPCR) targeting cps2J (serotypes 2 and 1/2) and cps9H (serotype 9) was evaluated with nasal and tonsillar samples from S. suis exposed pigs. qPCR specifically detected serotypes in all pig samples. The serotypes loads in pig samples estimated by qPCR showed, except for serotype 9 in tonsillar samples (correlation coefficient = 0.25), moderate to strong correlation with loads detected by culture (correlation coefficient > 0.65), and also in pigs exposed to both serotypes (correlation coefficient > 0.75). This qPCR is suitable for simultaneous differentiation and quantification of important S. suis serotypes. Full article
(This article belongs to the Special Issue Streptococcus suis)
Show Figures

Figure 1

Figure 1
<p>Standard curves of <span class="html-italic">cps2J</span>-qPCR (<b>panel A</b>) and <span class="html-italic">cps9H</span>-qPCR (<b>panel B</b>) obtained by serial dilutions of bacterial suspensions. The y-intercept indicates the expected crossing point (Cp) for a sample with a quantity equal to 1 eq. CFU/PCR reaction, i.e., 1 × 10<sup>2</sup> eq. CFU/mL. The slope indicates the number of cycles between samples that differ 1.0 log10 eq. CFU /mL; a value of 3.3 is optimal. The R<sup>2</sup> value indicates the close fit between the regression line of the standard curve and the individual Cp data points; a value of 1.00 indicates a perfect fit. The efficiency indicates the increase in copies per cycle; a value of 2 is optimal.</p>
Full article ">Figure 2
<p>Correlation between the log10 of the number of colony forming units (CFU) determined by selective bacterial examination (SBE) and log 10 eq. CFU by qPCR in tonsillar samples (<b>panels A,C</b>) and in nasal samples (<b>panels B,D</b>) from pigs exposed to either <span class="html-italic">S. suis</span> serotype 2 (<b>panels A,B</b>) or serotype 9 (<b>panels C,D</b>). Pigs that were inoculated are marked by ○, and contact exposed pigs by ●. On the label of each individual point, its time point of sampling is presented (in days post inoculation).</p>
Full article ">Figure 3
<p>Correlation between the <span class="html-italic">S. suis</span> counts (log10 CFU/mL) determined by selective bacterial examination (SBE) and counts predicted by a linear mixed model with the qPCR results (log10 eq. CFU/mL) of tonsillar (<b>panel A</b>) and nasal samples (<b>panel B</b>) taken from pigs exposed to both serotypes 2 and 9 as input. The model was constructed with data of pigs colonized with either serotype 2 or 9. Pigs that were inoculated are marked by ○, and contact pigs by ●. On the label of each individual point, its time point of sampling is presented (in days post inoculation).</p>
Full article ">
1421 KiB  
Communication
Astrocytes Enhance Streptococcus suis-Glial Cell Interaction in Primary Astrocyte-Microglial Cell Co-Cultures
by Jana Seele, Roland Nau, Chittappen K. Prajeeth, Martin Stangel, Peter Valentin-Weigand and Maren Seitz
Pathogens 2016, 5(2), 43; https://doi.org/10.3390/pathogens5020043 - 13 Jun 2016
Cited by 7 | Viewed by 5522
Abstract
Streptococcus (S.) suis infections are the most common cause of meningitis in pigs. Moreover, S. suis is a zoonotic pathogen, which can lead to meningitis in humans, mainly in adults. We assume that glial cells may play a crucial role in [...] Read more.
Streptococcus (S.) suis infections are the most common cause of meningitis in pigs. Moreover, S. suis is a zoonotic pathogen, which can lead to meningitis in humans, mainly in adults. We assume that glial cells may play a crucial role in host-pathogen interactions during S. suis infection of the central nervous system. Glial cells are considered to possess important functions during inflammation and injury of the brain in bacterial meningitis. In the present study, we established primary astrocyte-microglial cell co-cultures to investigate interactions of S. suis with glial cells. For this purpose, microglial cells and astrocytes were isolated from new-born mouse brains and characterized by flow cytometry, followed by the establishment of astrocyte and microglial cell mono-cultures as well as astrocyte-microglial cell co-cultures. In addition, we prepared microglial cell mono-cultures co-incubated with uninfected astrocyte mono-culture supernatants and astrocyte mono-cultures co-incubated with uninfected microglial cell mono-culture supernatants. After infection of the different cell cultures with S. suis, bacteria-cell association was mainly observed with microglial cells and most prominently with a non-encapsulated mutant of S. suis. A time-dependent induction of NO release was found only in the co-cultures and after co-incubation of microglial cells with uninfected supernatants of astrocyte mono-cultures mainly after infection with the capsular mutant. Only moderate cytotoxic effects were found in co-cultured glial cells after infection with S. suis. Taken together, astrocytes and astrocyte supernatants increased interaction of microglial cells with S. suis. Astrocyte-microglial cell co-cultures are suitable to study S. suis infections and bacteria-cell association as well as NO release by microglial cells was enhanced in the presence of astrocytes. Full article
(This article belongs to the Special Issue Streptococcus suis)
Show Figures

Figure 1

Figure 1
<p>Phenotypical characterisation of primary mouse astrocytes and microglial cells. Flow cytometry analysis of primary astrocytes (<b>A</b>) and microglial cells (<b>B</b>) isolated from brain tissue of new-born C57BL/6 mice. Cells were stained for cell-surface antigens (CD11b, CD11c, CD45, CX3CR1, and ACSA-2) and intracellular antigens (CD68 and GFAP) as indicated. Means of two independent preparations are shown.</p>
Full article ">Figure 2
<p>Association of <span class="html-italic">S. suis</span> with primary mouse glial cells. Various glial cell culture systems: (<b>A</b>) astrocyte mono-culture, (<b>B</b>) microglial cell mono-culture, (<b>C</b>) astrocyte mono-culture pre-incubated with supernatants (SN) of uninfected microglial cell cultures, (<b>D</b>) microglial cell mono-culture pre-incubated with SN of uninfected astrocyte cultures, (<b>E</b>) astrocyte-microglial cell co-culture (low amount of microglial cells), and (<b>F</b>) astrocyte-microglial cell co-culture (high amount of microglial cells), respectively, were infected with CFSE-labeled <span class="html-italic">S. suis</span> strain 10, 10<span class="html-italic">cps</span>Δ<span class="html-italic">EF</span>, or 10Δ<span class="html-italic">sly</span> at a MOI of 10:1. Percentage of CFSE-positive cells were measured by flow cytometry. Results are expressed as means with standard deviation (SD) of three independent experiments, and statistically significant differences when compared to uninfected control cells are indicated by ** (<span class="html-italic">p</span>-value &lt; 0.01), and *** (<span class="html-italic">p</span>-value &lt; 0.001), one-way analysis of variance (ANOVA) followed by a Dunnett post-hoc test.</p>
Full article ">Figure 3
<p>Association of <span class="html-italic">S. suis</span> with primary mouse astrocytes and microglial cells. Various glial cell culture systems: (<b>A</b>) astrocyte mono-culture; (<b>B</b>) microglial cell mono-culture; (<b>C</b>) astrocyte mono-culture pre-stimulated with SN of uninfected microglial cell cultures; (<b>D</b>) microglial cell mono-culture pre-stimulated with SN of uninfected astrocyte cultures; (<b>E</b>) astrocyte-microglial cell co-culture (low amount of microglial cells); and (<b>F</b>) astrocyte-microglial cell co-culture (high amount of microglial cells), were infected with CFSE-labeled <span class="html-italic">S. suis</span> strain 10, 10<span class="html-italic">cps</span>Δ<span class="html-italic">EF</span>, or 10Δ<span class="html-italic">sly</span> at a MOI 10:1 for 2 h. Astrocytes and microglial cells were stained for the cell-surface antigens ACSA-2 and CX3CR1, respectively. Relative amounts (%) of astrocytes (white bars), microglial cells (black bars) and microglial cells/astrocytes in association with bacteria (CFSE-positive, grey bars) were determined by flow cytometry analysis. Results are expressed as means with SD of three independent experiments, and statistically significant differences when compared to uninfected control cells are indicated by * (<span class="html-italic">p</span>-value &lt; 0.05), ** (<span class="html-italic">p</span>-value &lt; 0.01), and *** (<span class="html-italic">p</span>-value &lt; 0.001), one-way-ANOVA followed by a Dunnett post-hoc test.</p>
Full article ">Figure 4
<p>NO release by primary mouse glial cell cultures after infection with <span class="html-italic">S. suis</span>. Various glial cell culture systems: (<b>A</b>) astrocyte mono-culture; (<b>B</b>) microglial cell mono-culture; (<b>C</b>) astrocyte mono-culture pre-stimulated with SN of uninfected microglial cell cultures; (<b>D</b>) microglial cell mono-culture pre-stimulated with SN of uninfected astrocyte cultures; (<b>E</b>) astrocyte-microglial cell co-culture (low amount of microglial cells); and (<b>F</b>) astrocyte-microglial cell co-culture (high amount of microglial cells), were infected with <span class="html-italic">S. suis</span> strain 10, 10<span class="html-italic">cps</span>Δ<span class="html-italic">EF</span>, or 10Δ<span class="html-italic">sly</span> at a MOI of 10:1 for 2, 7, and 24 h (hpi). Nitirc oxide (NO) release was measured using Griess reagent. All results are expressed as x-fold NO release normalized to uninfected astrocytes or microglial cells which were not pre-incubated with SN (dashed line). In the co-culture system values were normalized to uninfected astrocytes. Uninfected astrocytes or microglial cells pre-incubated with SN only or co-cultured served as further controls (Ctr, <b>C</b>–<b>F</b>). Results are expressed as means with SD of four (<b>A</b> + <b>B</b>) or six (<b>C</b>–<b>F</b>) independent experiments and statistically significant differences when compared to uninfected astrocytes and microglial cells (<b>A</b>,<b>B</b>), uninfected astrocytes and microglial cells pre-incubated with supernatants (<b>C</b>,<b>D</b>) or astrocyte-microglial cell co-cultures (<b>E</b>,<b>F</b>) are indicated by * (<span class="html-italic">p</span>-value &lt; 0.05), ** (<span class="html-italic">p</span>-value &lt; 0.01), and *** (<span class="html-italic">p</span>-value &lt; 0.001), one-way-ANOVA followed by Dunnett post-hoc test.</p>
Full article ">Figure 5
<p>Cell viability of primary mouse glial cell cultures after infection with <span class="html-italic">S. suis</span>. Various glial cell culture systems: (<b>A</b>) astrocyte mono-culture; (<b>B</b>) microglial cell mono-culture; (<b>C</b>) astrocyte mono-culture pre-stimulated with SN of uninfected microglial cell cultures; (<b>D</b>) microglial cell mono-culture pre-stimulated with SN of uninfected astrocyte cultures; (<b>E</b>) astrocyte-microglial cell co-culture (low amount of microglial cells); and (<b>F</b>) astrocyte-microglial cell co-culture (high amount of microglial cells), were infected with <span class="html-italic">S. suis</span> strain 10, 10<span class="html-italic">cps</span>Δ<span class="html-italic">EF</span>, or 10Δ<span class="html-italic">sly</span> at a MOI 10:1. 24 h post infection cell viability was determined by standard lactate dehydrogenase (LDH) release assay. Results are expressed as x-fold cytotoxicity normalized to uninfected astrocytes and microglial cells which were not pre-incubated with SN (dashed line). In the co-culture system values were normalized to uninfected astrocytes. Uninfected astrocytes or microglial cells pre-stimulated with SN only or co-cultured served as further controls (Ctr, <b>C</b>–<b>F</b>). Results are expressed as means with SD of three independent experiments. No significant differences when compared to uninfected astrocytes and microglial cells (<b>A</b>,<b>B</b>), uninfected astrocytes and microglial cells pre-incubated with supernatants (<b>C</b>,<b>D</b>) or astrocyte-microglial cell co-cultures (E,F) were found, one-way-ANOVA followed by a Dunnett post-hoc test.</p>
Full article ">
2667 KiB  
Article
Impact of Sub-Inhibitory Concentrations of Amoxicillin on Streptococcus suis Capsule Gene Expression and Inflammatory Potential
by Bruno Haas and Daniel Grenier
Pathogens 2016, 5(2), 37; https://doi.org/10.3390/pathogens5020037 - 19 Apr 2016
Cited by 10 | Viewed by 5867
Abstract
Streptococcus suis is an important swine pathogen and emerging zoonotic agent worldwide causing meningitis, endocarditis, arthritis and septicemia. Among the 29 serotypes identified to date, serotype 2 is mostly isolated from diseased pigs. Although several virulence mechanisms have been characterized in S. suis [...] Read more.
Streptococcus suis is an important swine pathogen and emerging zoonotic agent worldwide causing meningitis, endocarditis, arthritis and septicemia. Among the 29 serotypes identified to date, serotype 2 is mostly isolated from diseased pigs. Although several virulence mechanisms have been characterized in S. suis, the pathogenesis of S. suis infections remains only partially understood. This study focuses on the response of S. suis P1/7 to sub-inhibitory concentrations of amoxicillin. First, capsule expression was monitored by qRT-PCR when S. suis was cultivated in the presence of amoxicillin. Then, the pro-inflammatory potential of S. suis P1/7 culture supernatants or whole cells conditioned with amoxicillin was evaluated by monitoring the activation of the NF-κB pathway in monocytes and quantifying pro-inflammatory cytokines secreted by macrophages. It was found that amoxicillin decreased capsule expression in S. suis. Moreover, conditioning the bacterium with sub-inhibitory concentrations of amoxicillin caused an increased activation of the NF-κB pathway in monocytes following exposure to bacterial culture supernatants and to a lesser extent to whole bacterial cells. This was associated with an increased secretion of pro-inflammatory cytokines (CXCL8, IL-6, IL-1β) by macrophages. This study identified a new mechanism by which S. suis may increase its inflammatory potential in the presence of sub-inhibitory concentrations of amoxicillin, a cell wall-active antibiotic, thus challenging its use for preventive treatments or as growth factor. Full article
(This article belongs to the Special Issue Streptococcus suis)
Show Figures

Figure 1

Figure 1
<p>Relative expression of the capsule gene <span class="html-italic">cps2J</span> by <span class="html-italic">S. suis</span> P1/7 in the presence of sub-inhibitory concentrations of amoxicillin. Control: no antibiotics. *: <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 2
<p>Transmission electron microscopy of overnight cultures of <span class="html-italic">S. suis</span> P1/7 grown in THB alone (<b>A</b>) or THB supplemented with 1/2 MIC of amoxicillin (<b>B</b>).</p>
Full article ">Figure 3
<p>Activation of NF-κB signaling pathway in monocytes stimulated with culture supernatants (<b>A</b>) or <span class="html-italic">S. suis</span> P1/7 whole cells (<b>B</b>) conditioned in sub-inhibitory concentrations of amoxicillin. <span class="html-italic">Escherichia coli</span> LPS was used as a positive control. 0: no antibiotic. *: <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 4
<p>Secretion of pro-inflammatory cytokines CXCL8 (<b>A</b>); IL-6 (<b>B</b>) and IL-1β (<b>C</b>) by macrophages stimulated with culture supernatants of <span class="html-italic">S. suis</span> P1/7 conditioned with amoxicillin. Control - : no stimulation. 0: no antibiotic. *: <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 5
<p>Secretion of pro-inflammatory cytokines CXCL8 (<b>A</b>); IL-6 (<b>B</b>) and IL-1β (<b>C</b>) by macrophages stimulated with <span class="html-italic">S. suis</span> P1/7 whole cells (MOI = 50) conditioned with amoxicillin. Control - : no stimulation. 0: no antibiotic. *: <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">

Review

Jump to: Research

1027 KiB  
Review
Current Taxonomical Situation of Streptococcus suis
by Masatoshi Okura, Makoto Osaki, Ryohei Nomoto, Sakura Arai, Ro Osawa, Tsutomu Sekizaki and Daisuke Takamatsu
Pathogens 2016, 5(3), 45; https://doi.org/10.3390/pathogens5030045 - 24 Jun 2016
Cited by 118 | Viewed by 8640
Abstract
Streptococcus suis, a major porcine pathogen and an important zoonotic agent, is considered to be composed of phenotypically and genetically diverse strains. However, recent studies reported several “S. suis-like strains” that were identified as S. suis by commonly used methods [...] Read more.
Streptococcus suis, a major porcine pathogen and an important zoonotic agent, is considered to be composed of phenotypically and genetically diverse strains. However, recent studies reported several “S. suis-like strains” that were identified as S. suis by commonly used methods for the identification of this bacterium, but were regarded as distinct species from S. suis according to the standards of several taxonomic analyses. Furthermore, it has been suggested that some S. suis-like strains can be assigned to several novel species. In this review, we discuss the current taxonomical situation of S. suis with a focus on (1) the classification history of the taxon of S. suis; (2) S. suis-like strains revealed by taxonomic analyses; (3) methods for detecting and identifying this species, including a novel method that can distinguish S. suis isolates from S. suis-like strains; and (4) current topics on the reclassification of S. suis-like strains. Full article
(This article belongs to the Special Issue Streptococcus suis)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Timeline summary on the history of the taxon, serological description, identification/detection methods, and major taxonomical analyses (including the findings) of <span class="html-italic">S. suis</span>. DDH, DNA-DNA hybridization. ANI, average nucleotide identity.</p>
Full article ">
Back to TopTop