Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 3.165
Filtrer
1.
Methods Mol Biol ; 2815: 115-119, 2024.
Article de Anglais | MEDLINE | ID: mdl-38884914

RÉSUMÉ

Streptococcus suis is a swine bacterial pathogen that predominantly causes disease in weaned piglets characterized by swelling of joints, arthritis, septicemia, meningitis, and sudden death. Intravenous, intramuscular, intraperitoneal, and intranasal infection models were developed to study the bacterial pathogenicity and efficacy of vaccines and various therapeutics. The selection of the appropriate infection model is a critical step in any study, as it may impact the outcomes of the study. Here we describe a method for infecting weaned piglets with S. suis using intraperitoneal route as a reliable, consistent, and reproducible animal model to evaluate vaccine protection against systemic bacterial infection.


Sujet(s)
Modèles animaux de maladie humaine , Infections à streptocoques , Streptococcus suis , Maladies des porcs , Animaux , Suidae , Streptococcus suis/pathogénicité , Infections à streptocoques/médecine vétérinaire , Infections à streptocoques/microbiologie , Infections à streptocoques/immunologie , Maladies des porcs/microbiologie , Maladies des porcs/immunologie , Injections péritoneales
2.
Methods Mol Biol ; 2815: 131-142, 2024.
Article de Anglais | MEDLINE | ID: mdl-38884916

RÉSUMÉ

Streptococcus suis is a bacterial pathogen that can cause significant economic losses in the swine industry due to high morbidity and mortality rates in infected animals. Vaccination with bacterins, which consist of inactivated bacteria and adjuvants to enhance the pig's immune response, is an effective approach to control S. suis infections in piglets. Here we provide a description of S. suis bacterins and the methods for vaccine preparation. Moreover, this chapter also describes the addition of recombinant Sao (rSao-L) protein to the S. suis bacterin, aiming to enhance the efficacy of the bacterins against S. suis in piglets. Furthermore, the methods for evaluating the immune response elicited by the bacterins are also covered in this chapter.


Sujet(s)
Streptococcus suis , Animaux , Suidae , Streptococcus suis/immunologie , Infections à streptocoques/immunologie , Infections à streptocoques/microbiologie , Infections à streptocoques/prévention et contrôle , Infections à streptocoques/médecine vétérinaire , Maladies des porcs/microbiologie , Maladies des porcs/prévention et contrôle , Maladies des porcs/immunologie , Vaccination/méthodes , Vaccins antibactériens/immunologie , Adjuvants immunologiques/pharmacologie , Anticorps antibactériens/immunologie , Protéines recombinantes/immunologie , Protéines recombinantes/génétique , Vaccins antistreptococciques/immunologie , Vaccins antistreptococciques/administration et posologie
3.
Vet Res ; 55(1): 80, 2024 Jun 17.
Article de Anglais | MEDLINE | ID: mdl-38886823

RÉSUMÉ

Bacteria utilize intercellular communication to orchestrate essential cellular processes, adapt to environmental changes, develop antibiotic tolerance, and enhance virulence. This communication, known as quorum sensing (QS), is mediated by the exchange of small signalling molecules called autoinducers. AI-2 QS, regulated by the metabolic enzyme LuxS (S-ribosylhomocysteine lyase), acts as a universal intercellular communication mechanism across gram-positive and gram-negative bacteria and is crucial for diverse bacterial processes. In this study, we demonstrated that in Streptococcus suis (S. suis), a notable zoonotic pathogen, AI-2 QS enhances galactose utilization, upregulates the Leloir pathway for capsular polysaccharide (CPS) precursor production, and boosts CPS synthesis, leading to increased resistance to macrophage phagocytosis. Additionally, our molecular docking and dynamics simulations suggest that, similar to S. pneumoniae, FruA, a fructose-specific phosphoenolpyruvate phosphotransferase system prevalent in gram-positive pathogens, may also function as an AI-2 membrane surface receptor in S. suis. In conclusion, our study demonstrated the significance of AI-2 in the synthesis of galactose metabolism-dependent CPS in S. suis. Additionally, we conducted a preliminary analysis of the potential role of FruA as a membrane surface receptor for S. suis AI-2.


Sujet(s)
Galactose , Détection du quorum , Streptococcus suis , Streptococcus suis/physiologie , Galactose/métabolisme , Détection du quorum/physiologie , Virulence , Animaux , Capsules bactériennes/métabolisme , Lactones/métabolisme , Infections à streptocoques/médecine vétérinaire , Infections à streptocoques/microbiologie , Infections à streptocoques/immunologie , Homosérine/analogues et dérivés , Homosérine/métabolisme , Polyosides bactériens/métabolisme
4.
Front Immunol ; 15: 1409378, 2024.
Article de Anglais | MEDLINE | ID: mdl-38855112

RÉSUMÉ

Introduction: Rupture of the gestational membranes often precedes major pregnancy complications, including preterm labor and preterm birth. One major cause of inflammation in the gestational membranes, chorioamnionitis (CAM) is often a result of bacterial infection. The commensal bacterium Streptococcus agalactiae, or Group B Streptococcus (GBS) is a leading infectious cause of CAM. Obesity is on the rise worldwide and roughly 1 in 4 pregnancy complications is related to obesity, and individuals with obesity are also more likely to be colonized by GBS. The gestational membranes are comprised of several distinct cell layers which are, from outermost to innermost: maternally-derived decidual stromal cells (DSCs), fetal cytotrophoblasts (CTBs), fetal mesenchymal cells, and fetal amnion epithelial cells (AECs). In addition, the gestational membranes have several immune cell populations; macrophages are the most common phagocyte. Here we characterize the effects of palmitate, the most common long-chain saturated fatty acid, on the inflammatory response of each layer of the gestational membranes when infected with GBS, using human cell lines and primary human tissue. Results: Palmitate itself slightly but significantly augments GBS proliferation. Palmitate and GBS co-stimulation synergized to induce many inflammatory proteins and cytokines, particularly IL-1ß and matrix metalloproteinase 9 from DSCs, CTBs, and macrophages, but not from AECs. Many of these findings are recapitulated when treating cells with palmitate and a TLR2 or TLR4 agonist, suggesting broad applicability of palmitate-pathogen synergy. Co-culture of macrophages with DSCs or CTBs, upon co-stimulation with GBS and palmitate, resulted in increased inflammatory responses, contrary to previous work in the absence of palmitate. In whole gestational membrane biopsies, the amnion layer appeared to dampen immune responses from the DSC and CTB layers (the choriodecidua) to GBS and palmitate co-stimulation. Addition of the monounsaturated fatty acid oleate, the most abundant monounsaturated fatty acid in circulation, dampened the proinflammatory effect of palmitate. Discussion: These studies reveal a complex interplay between the immunological response of the distinct layers of the gestational membrane to GBS infection and that such responses can be altered by exposure to long-chain saturated fatty acids. These data provide insight into how metabolic syndromes such as obesity might contribute to an increased risk for GBS disease during pregnancy.


Sujet(s)
Chorioamnionite , Interleukine-1 bêta , Palmitates , Infections à streptocoques , Streptococcus agalactiae , Humains , Femelle , Grossesse , Interleukine-1 bêta/métabolisme , Infections à streptocoques/immunologie , Chorioamnionite/immunologie , Chorioamnionite/microbiologie , Chorioamnionite/métabolisme , Palmitates/pharmacologie , Membranes extraembryonnaires/métabolisme , Membranes extraembryonnaires/microbiologie , Membranes extraembryonnaires/immunologie , Récepteur de type Toll-2/métabolisme
5.
Immunohorizons ; 8(5): 384-396, 2024 May 01.
Article de Anglais | MEDLINE | ID: mdl-38809232

RÉSUMÉ

The mammalian Siglec receptor sialoadhesin (Siglec1, CD169) confers innate immunity against the encapsulated pathogen group B Streptococcus (GBS). Newborn lung macrophages have lower expression levels of sialoadhesin at birth compared with the postnatal period, increasing their susceptibility to GBS infection. In this study, we investigate the mechanisms regulating sialoadhesin expression in the newborn mouse lung. In both neonatal and adult mice, GBS lung infection reduced Siglec1 expression, potentially delaying acquisition of immunity in neonates. Suppression of Siglec1 expression required interactions between sialic acid on the GBS capsule and the inhibitory host receptor Siglec-E. The Siglec1 gene contains multiple STAT binding motifs, which could regulate expression of sialoadhesin downstream of innate immune signals. Although GBS infection reduced STAT1 expression in the lungs of wild-type newborn mice, we observed increased numbers of STAT1+ cells in Siglece-/- lungs. To test if innate immune activation could increase sialoadhesin at birth, we first demonstrated that treatment of neonatal lung macrophages ex vivo with inflammatory activators increased sialoadhesin expression. However, overcoming the low sialoadhesin expression at birth using in vivo prenatal exposures or treatments with inflammatory stimuli were not successful. The suppression of sialoadhesin expression by GBS-Siglec-E engagement may therefore contribute to disease pathogenesis in newborns and represent a challenging but potentially appealing therapeutic opportunity to augment immunity at birth.


Sujet(s)
Animaux nouveau-nés , Souris knockout , Acide N-acétyl-neuraminique , Facteur de transcription STAT-1 , Lectine-1 de type Ig liant l'acide sialique , Infections à streptocoques , Streptococcus agalactiae , Animaux , Souris , Streptococcus agalactiae/immunologie , Acide N-acétyl-neuraminique/métabolisme , Lectine-1 de type Ig liant l'acide sialique/métabolisme , Infections à streptocoques/immunologie , Infections à streptocoques/microbiologie , Facteur de transcription STAT-1/métabolisme , Facteur de transcription STAT-1/génétique , Immunité innée , Souris de lignée C57BL , Poumon/immunologie , Poumon/microbiologie , Poumon/métabolisme , Macrophages alvéolaires/immunologie , Macrophages alvéolaires/métabolisme , Femelle , Macrophages/immunologie , Macrophages/métabolisme , Lectines/métabolisme , Lectines/génétique , Lectines liant l'acide sialique apparentées aux immunoglobulines/métabolisme , Lectines liant l'acide sialique apparentées aux immunoglobulines/génétique , Antigènes CD/métabolisme , Antigènes CD/génétique , Antigènes de différenciation des lymphocytes B
6.
Sci Adv ; 10(22): eadn7848, 2024 May 31.
Article de Anglais | MEDLINE | ID: mdl-38809989

RÉSUMÉ

Streptococcus agalactiae [group B Streptococcus (GBS)] is a leading cause of neonatal meningitis, with late-onset disease (LOD) occurring after gastrointestinal tract colonization in infants. Bacterial membrane lipids are essential for host-pathogen interactions, and the functions of glycolipids are yet to be fully elucidated. GBS synthesizes three major glycolipids: glucosyl-diacylglycerol (Glc-DAG), diglucosyl-DAG (Glc2-DAG), and lysyl-Glc-DAG (Lys-Glc-DAG). Here, we identify the enzyme, IagB, as responsible for biosynthesis of Glc-DAG, the precursor for the two other glycolipids in GBS. To examine the collective role of glycolipids to GBS virulence, we adapted a murine model of neonatal meningitis to simulate LOD. The GBS∆iagB mutant traversed the gut-epithelial barrier comparable to wild type but was severely attenuated in bloodstream survival, resulting in decreased bacterial loads in the brain. The GBS∆iagB mutant was more susceptible to neutrophil killing and membrane targeting by host antimicrobial peptides. This work reveals an unexplored function of GBS glycolipids with their ability to protect the bacterial cell from host antimicrobial killing.


Sujet(s)
Glycolipides , Infections à streptocoques , Streptococcus agalactiae , Streptococcus agalactiae/pathogénicité , Streptococcus agalactiae/immunologie , Streptococcus agalactiae/métabolisme , Animaux , Glycolipides/métabolisme , Glycolipides/immunologie , Souris , Virulence , Infections à streptocoques/immunologie , Infections à streptocoques/microbiologie , Humains , Modèles animaux de maladie humaine , Interactions hôte-pathogène/immunologie , Granulocytes neutrophiles/immunologie , Granulocytes neutrophiles/métabolisme , Mutation
7.
Fish Shellfish Immunol ; 149: 109594, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38697376

RÉSUMÉ

Non-specific cytotoxic cells (NCCs) are vital immune cells involved in teleost's non-specific immunity. As a receptor molecule on the NCCs' surface, the non-specific cytotoxic cell receptor protein 1 (NCCRP-1) is known to play a crucial role in mediating their activity. Nevertheless, there have been limited studies on the signal molecule that transmits signals via NCCRP-1. In this study, a yeast two-hybrid (Y2H) library of tilapia liver and head kidney was constructed and subsequently screened with the bait vector NCCRP-1 of Oreochromis niloticus (On-NCCRP-1) to obtain a C-type lectin (On-CTL) with an interacting protein sequence. Consequently, the full-length sequence of On-CTL was cloned and analyzed. The expression analysis revealed that On-CTL is highly expressed in the liver and is widely distributed in other tissues. Furthermore, On-CTL expression was significantly up-regulated in the brain, intestine, and head kidney following a challenge with Streptococcus agalactiae. A point-to-point Y2H method was also used to confirm the binding between On-NCCRP-1 and On-CTL. The recombinant On-CTL (rOn-CTL) protein was purified. In vitro experiments demonstrated that rOn-CTL can up-regulate the expression of killer effector molecules in NCCs via its interaction with On-NCCRP-1. Moreover, activation of NCCs by rOn-CTL resulted in a remarkable enhancement in their ability to eliminate fathead minnow cells, indicating that rOn-CTL effectively modulates the killing activity of NCCs through the NCC receptor molecule On-NCCRP-1. These findings significantly contribute to our comprehension of the regulatory mechanisms governing NCC activity, paving the way for future research in this field.


Sujet(s)
Cichlides , Maladies des poissons , Protéines de poisson , Lectines de type C , Streptococcus agalactiae , Animaux , Cichlides/immunologie , Cichlides/génétique , Lectines de type C/génétique , Lectines de type C/immunologie , Lectines de type C/composition chimique , Protéines de poisson/génétique , Protéines de poisson/immunologie , Maladies des poissons/immunologie , Streptococcus agalactiae/physiologie , Infections à streptocoques/immunologie , Infections à streptocoques/médecine vétérinaire , Régulation de l'expression des gènes/immunologie , Séquence d'acides aminés , Immunité innée/génétique , Alignement de séquences/médecine vétérinaire , Phylogenèse , Analyse de profil d'expression de gènes/médecine vétérinaire
8.
Hum Vaccin Immunother ; 20(1): 2345943, 2024 Dec 31.
Article de Anglais | MEDLINE | ID: mdl-38757492

RÉSUMÉ

Dental caries is a prevalent oral disease that mainly results from Streptococcus mutans. Susceptibility to S. mutans decreased rapidly after weaning in a well-known rat model. However, owing to the lack of time to establish protective immunity ahead of challenge, the weaning rat model is suboptimal for assessing prophylactic vaccines against S. mutans infection. In this study, we found that, in adult rats, S. mutans cultured under air-restricted conditions showed dramatically increased colonization efficacy and accelerated development of dental caries compared with those cultured under air-unrestricted conditions. We propose that S. mutans cultured under air-restricted conditions can be used to develop an optimal caries model, especially for the evaluation of prophylactic efficacy against S. mutans. Therefore, we used the anti-caries vaccine, KFD2-rPAc, to reevaluate the protection against the challenge of S. mutans. In immunized rats, rPAc-specific protective antibodies were robustly elicited by KFD2-rPAc before the challenge. In addition to inhibiting the initial and long-term colonization of S. mutans in vivo, KFD2-rPAc immunization showed an 83% inhibitory efficacy against the development of caries, similar to that previously evaluated in a weaning rat model. These results demonstrate that culturing under air-restricted conditions can promote S. mutans infection in adult rats, thereby helping establish a rat infection model to evaluate the prophylactic efficacy of vaccines and anti-caries drugs.


Sujet(s)
Anticorps antibactériens , Caries dentaires , Modèles animaux de maladie humaine , Streptococcus mutans , Animaux , Caries dentaires/prévention et contrôle , Caries dentaires/microbiologie , Caries dentaires/immunologie , Streptococcus mutans/immunologie , Rats , Anticorps antibactériens/immunologie , Anticorps antibactériens/sang , Vaccins antistreptococciques/immunologie , Vaccins antistreptococciques/administration et posologie , Infections à streptocoques/prévention et contrôle , Infections à streptocoques/immunologie , Infections à streptocoques/microbiologie , Femelle , Rat Sprague-Dawley
9.
Microb Pathog ; 191: 106675, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38705216

RÉSUMÉ

Bovine mastitis, caused by Streptococcus agalactiae (Group B Streptococcus; GBS), poses significant economic challenges to the global dairy industry. Mouse models serves as valuable tools for assessing GBS-induced infections as an alternative to large animals. This study aimed to investigate the LD50 dose, organ bacterial load, and quantification of peritoneal leukocyte populations for GBS serotypes Ia and II isolates from China and Pakistan. Additionally, we measured indicators such as lactoferrin, albumin, and myeloperoxidase (MPO) activity. Pro-inflammatory cytokines (TNF-α, IL-1ß, IL-6, and IL-2) and anti-inflammatory cytokines (IL-10 and TGF-ß) in serum and tissue samples were evaluated using ELISA and qPCR, respectively. BALB/c mice (4 mice per group) received individual intraperitoneal injections of 100 µl containing specific bacterial inoculum concentrations (ranging from 105 to 109 CFU per mouse) of Chinese and Pakistani GBS isolates (serotypes Ia and II). Control groups received 100 µL of sterile PBS. Results revealed that the LD50 bacterial dose causing 50 % mortality in mice was 107 CFU. The highest bacterial load in all experimental groups was quantified in the peritoneum, followed by blood, mammary gland, liver, spleen, lungs, and brain. The most significant bacterial dissemination was observed in mice inoculated with Pakistani serotype Ia at 24 h, with a subsequent notable decline in bacterial counts at day 3. Notably, infection with Pakistani serotype Ia showed a trend of increased total leukocyte counts, significantly higher than Pakistani serotype II, Chinese Serotype Ia, and Chinese serotype II. A substantial influx of neutrophils and lymphocytes was observed in response to all tested serotypes, with Pakistani serotype Ia inducing a significantly higher influx compared to other groups (Pakistani serotype II, Chinese serotype Ia, and Chinese serotype II). Furthermore, TNF-α, IL-1ß, IL-2, and IL-6 expressions were significantly increased in mice one day after infection with the Pakistani serotype Ia. Compared to mice infected with the Pakistani serotype II, Chinese Serotype Ia, and Chinese serotype II, those infected with the Pakistani serotype Ia isolate exhibited the highest production of IL-10 and TGF-ß, along with significantly increased concentrations of lactoferrin, albumin, and MPO. These findings suggest that the persistence and severity of infection caused by the Pakistani serotype Ia may be linked to its ability to spread to deeper tissues. This study enhances our understanding of the clinical characteristics of bovine mastitis caused by S. agalactiae in China and Pakistan.


Sujet(s)
Cytokines , Modèles animaux de maladie humaine , Souris de lignée BALB C , Sérogroupe , Infections à streptocoques , Streptococcus agalactiae , Animaux , Streptococcus agalactiae/pathogénicité , Streptococcus agalactiae/classification , Streptococcus agalactiae/immunologie , Streptococcus agalactiae/génétique , Souris , Infections à streptocoques/microbiologie , Infections à streptocoques/immunologie , Chine , Cytokines/métabolisme , Cytokines/sang , Femelle , Pakistan , Charge bactérienne , Bovins , Dose létale 50 , Mammite bovine/microbiologie
10.
Fish Shellfish Immunol ; 150: 109596, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38692380

RÉSUMÉ

Streptococcosis, the most common bacterial disease of fish in recent years, is highly infectious and lethal, and has become an important factor hindering the healthy and sustainable development of aquaculture. Chicken egg yolk antibody (IgY) has the advantages of high antigen specificity, inexpensive and easy to obtain, simple preparation, no toxic side effects, and in line with animal welfare, which is a green and safe alternative to antibiotics. In this study, the potential of specific IgY in the treatment of gastrointestinal pathogens was explored by observing the effects of specific IgY on intestinal flora, pathological tissue, apoptosis, oxidative stress, and inflammatory response of tilapia. We used the specific IgY prepared in the early stage to feed tilapia for 10 days, and then the tilapia was challenged with Streptococcus agalactiae. The results showed that feeding IgY before challenge had a small effect on the intestinal flora, and after challenge specific IgY decreased the proportion of Streptococcus and increased the diversity of the intestinal flora; in histopathology, specific IgY decreased tissue damage and maintained the integrity of tissue structure. Further study found that specific IgY can reduce intestinal epithelial cell apoptosis and reduce caspase activity; at the same time, the content of MDA was decreased, and the activities of SOD, CAT, GSH-Px and GR were increased. In addition, specific IgY can down-regulate the expression levels of IL-8 and TNF-α genes and up-regulate the expression levels of IL-10 and TGF-ß. The results of this study showed that specific IgY could improve the intestinal flora of tilapia infected with Streptococcus agalactiae, reduce intestinal cell apoptosis, oxidative stress injury and inflammatory response, thereby reducing tissue damage and protecting the health of tilapia. Overall, specific IgY can be further explored as a potential antibiotic alternative for gastrointestinal pathogen infections.


Sujet(s)
Aliment pour animaux , Apoptose , Poulets , Cichlides , Maladies des poissons , Microbiome gastro-intestinal , Immunoglobulines , Intestins , Stress oxydatif , Infections à streptocoques , Streptococcus agalactiae , Animaux , Streptococcus agalactiae/physiologie , Infections à streptocoques/médecine vétérinaire , Infections à streptocoques/immunologie , Stress oxydatif/effets des médicaments et des substances chimiques , Apoptose/effets des médicaments et des substances chimiques , Immunoglobulines/immunologie , Cichlides/immunologie , Poulets/immunologie , Maladies des poissons/immunologie , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Aliment pour animaux/analyse , Intestins/immunologie , Régime alimentaire/médecine vétérinaire , Jaune d'œuf/immunologie , Jaune d'œuf/composition chimique
11.
Infect Immun ; 92(6): e0014124, 2024 Jun 11.
Article de Anglais | MEDLINE | ID: mdl-38722166

RÉSUMÉ

The human-specific bacterial pathogen group A Streptococcus (GAS) is a significant cause of morbidity and mortality. Macrophages are important to control GAS infection, but previous data indicate that GAS can persist in macrophages. In this study, we detail the molecular mechanisms by which GAS survives in THP-1 macrophages. Our fluorescence microscopy studies demonstrate that GAS is readily phagocytosed by macrophages, but persists within phagolysosomes. These phagolysosomes are not acidified, which is in agreement with our findings that GAS cannot survive in low pH environments. We find that the secreted pore-forming toxin Streptolysin O (SLO) perforates the phagolysosomal membrane, allowing leakage of not only protons but also large proteins including the lysosomal protease cathepsin B. Additionally, GAS recruits CD63/LAMP-3, which may contribute to lysosomal permeabilization, especially in the absence of SLO. Thus, although GAS does not inhibit fusion of the lysosome with the phagosome, it has multiple mechanisms to prevent proper phagolysosome function, allowing for persistence of the bacteria within the macrophage. This has important implications for not only the initial response but also the overall functionality of the macrophages, which may lead to the resulting pathologies in GAS infection. Our data suggest that therapies aimed at improving macrophage function may positively impact patient outcomes in GAS infection.


Sujet(s)
Protéines bactériennes , Lysosomes , Macrophages , Streptococcus pyogenes , Streptolysines , Streptococcus pyogenes/immunologie , Humains , Macrophages/microbiologie , Macrophages/immunologie , Macrophages/métabolisme , Lysosomes/métabolisme , Lysosomes/microbiologie , Streptolysines/métabolisme , Protéines bactériennes/métabolisme , Protéines bactériennes/génétique , Phagosomes/microbiologie , Phagosomes/métabolisme , Cellules THP-1 , Phagocytose , Infections à streptocoques/immunologie , Infections à streptocoques/microbiologie , Infections à streptocoques/métabolisme , Cathepsine B/métabolisme , Concentration en ions d'hydrogène
12.
Microb Pathog ; 192: 106703, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38763315

RÉSUMÉ

Streptococcus equissp.zooepidemicus (SEZ) is a crucial pathogen and contributes to various infections in numerous animal species. Swine streptococcicosis outbreak caused by SEZ has been reported in several countries in recent years. SzM protein is a cell membrane-anchored protein, which exhibits as an important virulence factor of SEZ. Effects of SzM protein on host innate immune need further study. Here, recombinant SzM (rSzM) protein of the SEZ was obtained, and mice were intraperitoneally injected with rSzM protein. We discovered that rSzM protein can recruit neutrophils into the injected site. In further study, neutrophils were isolated and treated with rSzM protein, NETs release were triggered by rSzM protein independently, and GSDMD protein was promoted-expressed and activated. In order to investigate the role of GSDMD in NETs formation, neutrophils isolated from WT mice and GSDMD-/- mice were treated with rSzM protein. The results showed that GSDMD deficiency suppressed the NETs release. In conclusion, SzM protein of SEZ can trigger the NETs release in a GSDMD-depending manner.


Sujet(s)
Protéines bactériennes , Pièges extracellulaires , Granulocytes neutrophiles , Infections à streptocoques , Streptococcus equi , Facteurs de virulence , Animaux , Souris , Granulocytes neutrophiles/immunologie , Protéines bactériennes/génétique , Protéines bactériennes/métabolisme , Pièges extracellulaires/métabolisme , Pièges extracellulaires/immunologie , Streptococcus equi/génétique , Streptococcus equi/immunologie , Infections à streptocoques/immunologie , Infections à streptocoques/microbiologie , Facteurs de virulence/génétique , Facteurs de virulence/métabolisme , Souris knockout , Protéines recombinantes/génétique , Immunité innée , Souris de lignée C57BL , Gasdermines , Protéines de liaison aux phosphates
13.
Microb Pathog ; 192: 106683, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38735447

RÉSUMÉ

Bacteria possess the ability to develop diverse and ingenious strategies to outwit the host immune system, and proteases are one of the many weapons employed by bacteria. This study sought to identify S. agalactiae additional serine protease and determine its role in virulence. The S. agalactiae THN0901 genome features one S8 family serine peptidase B (SfpB), acting as a secreted and externally exposed entity. A S8 family serine peptidase mutant strain (ΔsfpB) and complement strain (CΔsfpB) were generated through homologous recombination. Compared to the wild-type strain THN0901, the absorption of EtBr dyes was significantly reduced (P < 0.01) in ΔsfpB, implying an altered cell membrane permeability. In addition, the ΔsfpB strain had a significantly lower survival rate in macrophages (P < 0.01) and a 61.85 % lower adhesion ability to the EPC cells (P < 0.01) compared to THN0901. In the in vivo colonization experiment using tilapia as a model, 210 fish were selected and injected with different bacterial strains at a concentration of 3 × 106 CFU/tail. At 6, 12, 24, 48, 72 and 96 h post-injection, three fish were randomly selected from each group and their brain, liver, spleen, and kidney tissues were isolated. Subsequently, it was demonstrated that the ΔsfpB strain exhibited a markedly diminished capacity for colonization in tilapia. Additionally, the cumulative mortality of ΔsfpB in fish after intraperitoneal injection was reduced by 19.92-23.85 %. In conclusion, the findings in this study have demonstrated that the SfpB plays a significant role in S. agalactiae cell membrane stability and immune evasion. The immune evasion is fundamental for the development and transmission of invasive diseases, the serine protease SfpB may be a promising candidate for the development of antimicrobial agents to reduce the transmission of S. agalactiae.


Sujet(s)
Membrane cellulaire , Maladies des poissons , Échappement immunitaire , Infections à streptocoques , Streptococcus agalactiae , Streptococcus agalactiae/génétique , Streptococcus agalactiae/pathogénicité , Streptococcus agalactiae/enzymologie , Streptococcus agalactiae/immunologie , Animaux , Virulence , Infections à streptocoques/microbiologie , Infections à streptocoques/immunologie , Membrane cellulaire/métabolisme , Maladies des poissons/microbiologie , Maladies des poissons/immunologie , Adhérence bactérienne , Macrophages/microbiologie , Macrophages/immunologie , Protéines bactériennes/génétique , Protéines bactériennes/métabolisme , Protéases à sérine/génétique , Protéases à sérine/métabolisme , Facteurs de virulence/génétique , Facteurs de virulence/métabolisme , Souris
14.
BMC Vet Res ; 20(1): 173, 2024 May 03.
Article de Anglais | MEDLINE | ID: mdl-38702665

RÉSUMÉ

Strangles is a highly contagious disease of the equine upper respiratory tract caused by Streptococcus equi subspecies. Streptococcus equi subsp. equi (S. equi) and Streptococcus equi subsp. zooepidemicus (S. zooepidemicus) was isolated, as local, hot, and field strains, from horses clinically suffering from respiratory distress. The isolated Streptococci were identified using bacteriological and molecular techniques. Four formulations of inactivated S. equi vaccines were developed and evaluated. The first formulation was prepared using the S. equi isolates, adjuvanted with MONTANIDE GEL adjuvant, while the second formulation was adjuvanted with MONTANIDE ISA-70 adjuvant. The other 2 formulations were inactivated combined vaccines prepared from both S. equi and S. zooepidemicus isolates. The 3rd formulation was the combined isolates adjuvanted with MONTANIDE GEL while the 4th formulation was the combined isolates adjuvanted with MONTANIDE ISA-70. The developed vaccines' physical properties, purity, sterility, safety, and potency were ensured. The immunizing efficacy was determined in isogenic BALB/c mice and white New Zealand rabbits using the passive hemagglutination test. Also, the antibodies' titer of the combined S. equi and S. zooepidemicus vaccine adjuvanted with MONTANIDE ISA-70 in foals was tracked using an indirect enzyme-linked immunosorbent assay. The protective efficacy of the developed vaccines was determined using a challenge test in both laboratory and field animal models, where a 75% protection rate was achieved. The combined vaccine proved to be more efficacious than the monovalent vaccine. Also, the MONTANIDE ISA-70 adjuvant provided significant protective efficacy than the MONTANIDE GEL. The current work is introducing a very promising mitigative and strategic controlling solution for strangles.


Sujet(s)
Maladies des chevaux , Souris de lignée BALB C , Infections à streptocoques , Vaccins antistreptococciques , Streptococcus equi , Streptococcus , Animaux , Streptococcus equi/immunologie , Equus caballus , Lapins , Infections à streptocoques/médecine vétérinaire , Infections à streptocoques/prévention et contrôle , Infections à streptocoques/microbiologie , Infections à streptocoques/immunologie , Souris , Maladies des chevaux/prévention et contrôle , Maladies des chevaux/microbiologie , Maladies des chevaux/immunologie , Vaccins antistreptococciques/immunologie , Vaccins antistreptococciques/administration et posologie , Femelle , Anticorps antibactériens/sang , Adjuvants immunologiques/administration et posologie , Vaccins inactivés/immunologie
15.
Front Immunol ; 15: 1392456, 2024.
Article de Anglais | MEDLINE | ID: mdl-38779673

RÉSUMÉ

In response to the global threat posed by bacterial pathogens, which are the second leading cause of death worldwide, vaccine development is challenged by the diversity of bacterial serotypes and the lack of immunoprotection across serotypes. To address this, we introduce BacScan, a novel genome-wide technology for the rapid discovery of conserved highly immunogenic proteins (HIPs) across serotypes. Using bacterial-specific serum, BacScan combines phage display, immunoprecipitation, and next-generation sequencing to comprehensively identify all the HIPs in a single assay, thereby paving the way for the development of universally protective vaccines. Our validation of this technique with Streptococcus suis, a major pathogenic threat, led to the identification of 19 HIPs, eight of which conferred 20-100% protection against S. suis challenge in animal models. Remarkably, HIP 8455 induced complete immunity, making it an exemplary vaccine target. BacScan's adaptability to any bacterial pathogen positions it as a revolutionary tool that can expedite the development of vaccines with broad efficacy, thus playing a critical role in curbing bacterial transmission and slowing the march of antimicrobial resistance.


Sujet(s)
Protéines bactériennes , Animaux , Souris , Protéines bactériennes/immunologie , Protéines bactériennes/génétique , Infections à streptocoques/immunologie , Infections à streptocoques/prévention et contrôle , Streptococcus suis/immunologie , Streptococcus suis/génétique , Génome bactérien , Séquençage nucléotidique à haut débit , Femelle , Antigènes bactériens/immunologie , Antigènes bactériens/génétique , Humains , Vaccins antibactériens/immunologie
16.
Vet Res ; 55(1): 57, 2024 May 07.
Article de Anglais | MEDLINE | ID: mdl-38715138

RÉSUMÉ

Streptococcus suis is a bacterial pathogen that causes important economic losses to the swine industry worldwide. Since there are no current commercial vaccines, the use of autogenous vaccines applied to gilts/sows to enhance transfer of passive immunity is an attractive alternative to protect weaned piglets. However, there is no universal standardization in the production of autogenous vaccines and the vaccine formulation may be highly different among licenced manufacturing laboratories. In the present study, an autogenous vaccine that included S. suis serotypes 2, 1/2, 5, 7 and 14 was prepared by a licensed laboratory and administrated to gilts using a three-dose program prior to farrowing. The antibody response in gilts as well as the passive transfer of antibodies to piglets was then evaluated. In divergence with previously published data with an autogenous vaccine produced by a different company, the increased response seen in gilts was sufficient to improve maternal antibody transfer to piglets up to 5 weeks of age. However, piglets would still remain susceptible to S. suis disease which often appears during the second part of the nursery period. Vaccination did not affect the shedding of S. suis (as well as that of the specific S. suis serotypes included in the vaccine) by either gilts or piglets. Although all antibiotic treatments were absent during the trial, the clinical protective effect of the vaccination program with the autogenous vaccine could not be evaluated, since limited S. suis cases were present during the trial, confirming the need for a complete evaluation of the clinical protection that must include laboratory confirmation of the aetiological agent involved in the presence of S. suis-associated clinical signs. Further studies to evaluate the usefulness of gilt/sow vaccination with autogenous vaccines to protect nursery piglets should be done.


Sujet(s)
Autovaccins , Infections à streptocoques , Streptococcus suis , Maladies des porcs , Animaux , Streptococcus suis/immunologie , Suidae , Maladies des porcs/prévention et contrôle , Maladies des porcs/microbiologie , Maladies des porcs/immunologie , Infections à streptocoques/médecine vétérinaire , Infections à streptocoques/prévention et contrôle , Infections à streptocoques/immunologie , Femelle , Immunité acquise d'origine maternelle , Vaccins antistreptococciques/immunologie , Vaccins antistreptococciques/administration et posologie , Sérogroupe , Vaccination/médecine vétérinaire
17.
Dev Comp Immunol ; 157: 105188, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38677664

RÉSUMÉ

Emerging and re-emerging diseases in fish cause drastic economic losses in the aquaculture sector. To combat the impact of disease outbreaks and prevent the emergence of infections in culture systems, understanding the advanced strategies for protecting fish against infections is inevitable in fish health research. Therefore, the present study aimed to evaluate the induction of trained immunity and its protective efficacy against Streptococcus agalactiae in tilapia. For this, Nile tilapia and the Tilapia head kidney macrophage primary culture were primed using ß-glucan @200 µg/10 g body weight and 10 µg/mL respectively. Expression profiles of the markers of trained immunity and production of metabolites were monitored at different time points, post-priming and training, which depicted enhanced responsiveness. Higher lactate and lactate dehydrogenase (LDH) production in vitro suggests heightened glycolysis induced by priming of the cells using ß-glucan. A survival rate of 60% was observed in ß-glucan trained fish post challenge with virulent S. agalactiae at an LD50 of 2.6 × 107 cfu/ml, providing valuable insights into promising strategies of trained immunity for combating infections in fish.


Sujet(s)
Cichlides , Maladies des poissons , Macrophages , Infections à streptocoques , Streptococcus agalactiae , bêta-Glucanes , Animaux , bêta-Glucanes/métabolisme , Streptococcus agalactiae/immunologie , Cichlides/immunologie , Maladies des poissons/immunologie , Maladies des poissons/prévention et contrôle , Maladies des poissons/microbiologie , Infections à streptocoques/immunologie , Infections à streptocoques/médecine vétérinaire , Macrophages/immunologie , Cellules cultivées , Rein céphalique/immunologie , Aquaculture , Immunité innée , Glycolyse , L-Lactate dehydrogenase/métabolisme , Mémoire immunologique , Immunité entraînée
18.
Nat Commun ; 15(1): 3600, 2024 Apr 27.
Article de Anglais | MEDLINE | ID: mdl-38678029

RÉSUMÉ

Streptococcus pyogenes can cause invasive disease with high mortality despite adequate antibiotic treatments. To address this unmet need, we have previously generated an opsonic IgG1 monoclonal antibody, Ab25, targeting the bacterial M protein. Here, we engineer the IgG2-4 subclasses of Ab25. Despite having reduced binding, the IgG3 version promotes stronger phagocytosis of bacteria. Using atomic simulations, we show that IgG3's Fc tail has extensive movement in 3D space due to its extended hinge region, possibly facilitating interactions with immune cells. We replaced the hinge of IgG1 with four different IgG3-hinge segment subclasses, IgGhxx. Hinge-engineering does not diminish binding as with IgG3 but enhances opsonic function, where a 47 amino acid hinge is comparable to IgG3 in function. IgGh47 shows improved protection against S. pyogenes in a systemic infection mouse model, suggesting that IgGh47 has promise as a preclinical therapeutic candidate. Importantly, the enhanced opsonic function of IgGh47 is generalizable to diverse S. pyogenes strains from clinical isolates. We generated IgGh47 versions of anti-SARS-CoV-2 mAbs to broaden the biological applicability, and these also exhibit strongly enhanced opsonic function compared to the IgG1 subclass. The improved function of the IgGh47 subclass in two distant biological systems provides new insights into antibody function.


Sujet(s)
COVID-19 , Fragments Fc des immunoglobulines , Immunoglobuline G , SARS-CoV-2 , Streptococcus pyogenes , Animaux , Humains , Souris , Anticorps antibactériens/immunologie , Anticorps monoclonaux/immunologie , Anticorps antiviraux/immunologie , COVID-19/immunologie , COVID-19/virologie , Fragments Fc des immunoglobulines/immunologie , Fragments Fc des immunoglobulines/génétique , Fragments Fc des immunoglobulines/composition chimique , Immunoglobuline G/composition chimique , Immunoglobuline G/génétique , Immunoglobuline G/immunologie , Souris de lignée BALB C , Phagocytose , Ingénierie des protéines/méthodes , SARS-CoV-2/immunologie , Infections à streptocoques/immunologie , Infections à streptocoques/microbiologie , Streptococcus pyogenes/immunologie
19.
Fish Shellfish Immunol ; 149: 109566, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38636735

RÉSUMÉ

Fish rely on innate immune system for immunity, and nucleotide-binding oligomerization domain-like receptors (NLRs) are a vital group of receptor for recognition. In the present study, NOD1 gene was cloned and characterized from golden pompano Trachinotus ovatus, a commercially important aquaculture fish species. The ORF of T. ovatus NOD1 was 2820 bp long, encoding 939 amino acid residues with a highly conserved domains containing CARD-NACHT-LRRs. Phylogenetic analysis revealed that the T. ovatus NOD1 clustered with those of fish and separated from those of birds and mammals. T. ovatus NOD1 has wide tissue distribution with the highest expression in gills. Bacterial challenges (Streptococcus agalactiae and Vibrio alginolyticus) significantly up-regulated the expression of NOD1 with different response time. The results of T. ovatus NOD1 ligand recognition and signaling pathway analysis revealed that T. ovatus NOD1 could recognize iE-DAP at the concentration of ≧ 100 ng/mL and able to activate NF-κB signaling pathway. This study confirmed that NOD1 play a crucial role in the innate immunity of T. ovatus. The findings of this study improve our understanding on the immune function of NOD1 in teleost, especially T. ovatus.


Sujet(s)
Séquence d'acides aminés , Maladies des poissons , Protéines de poisson , Immunité innée , Protéine adaptatrice de signalisation NOD1 , Phylogenèse , Alignement de séquences , Vibrio alginolyticus , Animaux , Protéine adaptatrice de signalisation NOD1/génétique , Protéine adaptatrice de signalisation NOD1/immunologie , Protéine adaptatrice de signalisation NOD1/composition chimique , Protéines de poisson/génétique , Protéines de poisson/immunologie , Protéines de poisson/composition chimique , Immunité innée/génétique , Maladies des poissons/immunologie , Alignement de séquences/médecine vétérinaire , Vibrio alginolyticus/physiologie , Infections à streptocoques/immunologie , Infections à streptocoques/médecine vétérinaire , Streptococcus agalactiae/physiologie , Régulation de l'expression des gènes/immunologie , Analyse de profil d'expression de gènes/médecine vétérinaire , Infections à Vibrio/immunologie , Infections à Vibrio/médecine vétérinaire , Acide diamino-pimélique/composition chimique , Acide diamino-pimélique/analogues et dérivés , Perciformes/immunologie , Perciformes/génétique , Poissons/immunologie , Poissons/génétique
20.
Fish Shellfish Immunol ; 149: 109534, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38575040

RÉSUMÉ

Zinc is one of the essential microelements for the metabolism of animals. Zinc nanoparticles may have higher bioavailability due to their low specific surface area, facilitating absorption by fish. The present study aimed to evaluate the effects of supplementation with different zinc-based products on the growth and health of Nile tilapia Oreochromis niloticus. Zinc, in different sizes (nanoparticles or bulk) and forms (inorganic or organic), were used as a supplement in the tilapia diet at a dose of 15 mg kg feed-1 for 60 days. At the end of the feeding trial, production performance, hemato-immunological parameters, activity of antioxidant system enzymes, exposure to Streptococcus agalactiae and zinc concentration in the muscle were examined. After the bacterial challenge, the mean corpuscular hemoglobin concentration (MCHC) significantly increased in the fish treated with organic zinc, inorganic nano zinc, and organic nano zinc, while in the control group (inorganic zinc), MCHC remained unchanged. Regarding defense cells, dietary inorganic nano zinc increased the number of basophils (1.50 ± 1.10) compared to organic zinc (0.80 ± 0.90). Lymphocyte count increased after the challenge only in the organic zinc treatments (bulk and nanoparticles). Neutrophils decreased in the control (inorganic zinc) (2.20 ± 1.70) and inorganic nano zinc (2.60 ± 2.70) treatments after the challenge. When compared before and after the bacterial challenge, the plasma antimicrobial titer significantly increased after the bacterial challenge in all treatments. No significant differences were observed for total proteins, enzymes (SOD and CAT), cumulative survival and zinc deposition on fillet. In conclusion, organic zinc in nanoparticles or bulk size increased Nile tilapia innate defense during bacterial infection. However, the other parameters evaluated were not affected by zinc particle size or form (organic or inorganic), indicating that further evaluations should be conducted with organic zinc in nanoparticles or bulk size in the tilapia diet.


Sujet(s)
Aliment pour animaux , Cichlides , Régime alimentaire , Compléments alimentaires , Maladies des poissons , Infections à streptocoques , Streptococcus agalactiae , Zinc , Animaux , Cichlides/immunologie , Cichlides/croissance et développement , Compléments alimentaires/analyse , Zinc/administration et posologie , Aliment pour animaux/analyse , Régime alimentaire/médecine vétérinaire , Infections à streptocoques/médecine vétérinaire , Infections à streptocoques/immunologie , Streptococcus agalactiae/physiologie , Maladies des poissons/immunologie , Répartition aléatoire , Immunité innée/effets des médicaments et des substances chimiques
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...