Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.151
Filter
1.
Vet Res ; 55(1): 57, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38715138

ABSTRACT

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.


Subject(s)
Autovaccines , Streptococcal Infections , Streptococcus suis , Swine Diseases , Animals , Streptococcus suis/immunology , Swine , Swine Diseases/prevention & control , Swine Diseases/microbiology , Swine Diseases/immunology , Streptococcal Infections/veterinary , Streptococcal Infections/prevention & control , Streptococcal Infections/immunology , Female , Immunity, Maternally-Acquired , Streptococcal Vaccines/immunology , Streptococcal Vaccines/administration & dosage , Serogroup , Vaccination/veterinary
2.
Hum Vaccin Immunother ; 20(1): 2345943, 2024 Dec 31.
Article in English | MEDLINE | ID: mdl-38757492

ABSTRACT

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.


Subject(s)
Antibodies, Bacterial , Dental Caries , Disease Models, Animal , Streptococcus mutans , Animals , Dental Caries/prevention & control , Dental Caries/microbiology , Dental Caries/immunology , Streptococcus mutans/immunology , Rats , Antibodies, Bacterial/immunology , Antibodies, Bacterial/blood , Streptococcal Vaccines/immunology , Streptococcal Vaccines/administration & dosage , Streptococcal Infections/prevention & control , Streptococcal Infections/immunology , Streptococcal Infections/microbiology , Female , Rats, Sprague-Dawley
3.
Fish Shellfish Immunol ; 149: 109594, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38697376

ABSTRACT

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.


Subject(s)
Cichlids , Fish Diseases , Fish Proteins , Lectins, C-Type , Streptococcus agalactiae , Animals , Cichlids/immunology , Cichlids/genetics , Lectins, C-Type/genetics , Lectins, C-Type/immunology , Lectins, C-Type/chemistry , Fish Proteins/genetics , Fish Proteins/immunology , Fish Diseases/immunology , Streptococcus agalactiae/physiology , Streptococcal Infections/immunology , Streptococcal Infections/veterinary , Gene Expression Regulation/immunology , Amino Acid Sequence , Immunity, Innate/genetics , Sequence Alignment/veterinary , Phylogeny , Gene Expression Profiling/veterinary
4.
BMC Vet Res ; 20(1): 173, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38702665

ABSTRACT

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.


Subject(s)
Horse Diseases , Mice, Inbred BALB C , Streptococcal Infections , Streptococcal Vaccines , Streptococcus equi , Streptococcus , Animals , Streptococcus equi/immunology , Horses , Rabbits , Streptococcal Infections/veterinary , Streptococcal Infections/prevention & control , Streptococcal Infections/microbiology , Streptococcal Infections/immunology , Mice , Horse Diseases/prevention & control , Horse Diseases/microbiology , Horse Diseases/immunology , Streptococcal Vaccines/immunology , Streptococcal Vaccines/administration & dosage , Female , Antibodies, Bacterial/blood , Adjuvants, Immunologic/administration & dosage , Vaccines, Inactivated/immunology
5.
FEMS Microbiol Rev ; 48(3)2024 May 08.
Article in English | MEDLINE | ID: mdl-38678005

ABSTRACT

Group B Streptococcus (GBS; also known as Streptococcus agalactiae) is an opportunistic bacterial pathogen that causes sepsis, meningitis, pneumonia, and skin and soft tissue infections in neonates and healthy or immunocompromised adults. GBS is well-adapted to survive in humans due to a plethora of virulence mechanisms that afford responses to support bacterial survival in dynamic host environments. These mechanisms and responses include counteraction of cell death from exposure to excess metal ions that can cause mismetallation and cytotoxicity, and strategies to combat molecules such as reactive oxygen and nitrogen species that are generated as part of innate host defence. Cytotoxicity from reactive molecules can stem from damage to proteins, DNA, and membrane lipids, potentially leading to bacterial cell death inside phagocytic cells or within extracellular spaces within the host. Deciphering the ways in which GBS responds to the stress of cytotoxic reactive molecules within the host will benefit the development of novel therapeutic and preventative strategies to manage the burden of GBS disease. This review summarizes knowledge of GBS carriage in humans and the mechanisms used by the bacteria to circumvent killing by these important elements of host immune defence: oxidative stress, nitrosative stress, and stress from metal ion intoxication/mismetallation.


Subject(s)
Metals , Streptococcal Infections , Streptococcus agalactiae , Streptococcus agalactiae/physiology , Streptococcus agalactiae/pathogenicity , Humans , Streptococcal Infections/microbiology , Streptococcal Infections/immunology , Metals/metabolism , Metals/toxicity , Oxidative Stress , Reactive Oxygen Species/metabolism , Stress, Physiological , Virulence , Opportunistic Infections/microbiology
6.
Fish Shellfish Immunol ; 149: 109572, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38636739

ABSTRACT

Streptococcosis outbreaks caused by Streptococcus agalactiae infection in tilapia aquaculture have been consistently reported and associated with high mortality and morbidity leading to significant economic losses. Existing vaccine candidates against Streptococcus spp. are designed for intraperitoneal injections that are not practical and labor-intensive which have prompted farmers to protect aquatic animals with antibiotics, thus encouraging the emergence of multidrug resistant bacteria. In this study, a live recombinant L. lactis vaccine expressing a 1403 bp surface immunogenic protein (SIP) and a 1100 bp truncated SIP (tSIP) gene was developed and evaluated against S. agalactiae infection in tilapia. Both SIP and tSIP sequences were cloned and transformed into L. lactis. The recombinant L.lactis vaccine was orally administered to juvenile tilapia for a month. Detection of SIP-specific serum IgM in vaccinated groups compared to control groups indicated that recombinant proteins expressed from L. lactis could elicit immunogenic reactions in tilapia. Fish immunized with the tSIP vaccine also showed the highest level of protection compared to other test groups, and the mortality rate was significantly reduced compared to both control groups. The relative percentage of survival (RPS) against S. agalactiae for both SIP and tSIP-vaccinated groups was 50 % and 89 %, respectively, at 14 days post-challenge. Significant up-regulation of IgM, IL-1ß, IL-10, TNF-α and IFN-γ were observed at day 34 between the vaccinated and control groups. These results indicated that the recombinant lactococcal tSIP vaccine can elicit both cell-mediated and humoral responses and is recommended as a potential oral vaccine against S. agalactiae infection. Future work will include further in vivo challenge assessments of this vaccine candidate fused with adjuvants to boost immunogenicity levels in tilapia.


Subject(s)
Cichlids , Fish Diseases , Streptococcal Infections , Streptococcus agalactiae , Animals , Streptococcus agalactiae/immunology , Streptococcal Infections/veterinary , Streptococcal Infections/prevention & control , Streptococcal Infections/immunology , Fish Diseases/prevention & control , Fish Diseases/immunology , Cichlids/immunology , Administration, Oral , Vaccines, Synthetic/immunology , Vaccines, Synthetic/administration & dosage , Streptococcal Vaccines/immunology , Streptococcal Vaccines/administration & dosage , Bacterial Vaccines/immunology , Bacterial Vaccines/administration & dosage , Lactococcus lactis/genetics , Lactococcus lactis/immunology , Bacterial Proteins/immunology , Bacterial Proteins/genetics
7.
Fish Shellfish Immunol ; 149: 109566, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38636735

ABSTRACT

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.


Subject(s)
Amino Acid Sequence , Fish Diseases , Fish Proteins , Immunity, Innate , Nod1 Signaling Adaptor Protein , Phylogeny , Sequence Alignment , Vibrio alginolyticus , Animals , Nod1 Signaling Adaptor Protein/genetics , Nod1 Signaling Adaptor Protein/immunology , Nod1 Signaling Adaptor Protein/chemistry , Fish Proteins/genetics , Fish Proteins/immunology , Fish Proteins/chemistry , Immunity, Innate/genetics , Fish Diseases/immunology , Sequence Alignment/veterinary , Vibrio alginolyticus/physiology , Streptococcal Infections/immunology , Streptococcal Infections/veterinary , Streptococcus agalactiae/physiology , Gene Expression Regulation/immunology , Gene Expression Profiling/veterinary , Vibrio Infections/immunology , Vibrio Infections/veterinary , Diaminopimelic Acid/chemistry , Diaminopimelic Acid/analogs & derivatives , Perciformes/immunology , Perciformes/genetics , Fishes/immunology , Fishes/genetics
8.
Fish Shellfish Immunol ; 149: 109567, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38641215

ABSTRACT

Streptococcosis, an emerging infectious disease caused by Streptococcus agalactiae, has had adverse effects on farmed tilapia. Several vaccines have been developed to prevent this disease and induce a specific immune response against S. agalactiae infection. In this study the use of MONTANIDE™ GR01, a new adjuvant for oral vaccination, was optimized for use in tilapia under laboratory and field studies. In the laboratory trial the immune response and protective efficacy of two doses of MONTANIDE™ GR01, 20 % (w/w) and 2 % (w/w), included into the feed-based adjuvanted vaccines were assessed comparatively. Following immunization, the innate immune parameters studied in serum, including lysozyme, myeloperoxidase, catalase and glutathione peroxidase activity, were all increased significantly. Furthermore, specific IgM antibodies against S. agalactiae were induced significantly in serum post-vaccination, with higher levels observed in both groups that received the feed-based adjuvanted vaccine. Under both injection and immersion challenge conditions, the relative percent survival for the feed-based adjuvanted vaccine groups ranged from 78 % to 84 %. Following use of the low dose concentration of MONTANIDE™ GR01 for oral vaccination of tilapia in cage culture systems, several innate immune parameters were effectively enhanced in the immunized fish. Similarly, the levels of specific IgM antibodies in the serum of feed-based vaccinated fish were significantly enhanced, reaching their highest levels 2-5 months post-vaccination. Cytokines associated with innate and adaptive immunity were also examined, and the expression levels of several genes showed significant up-regulation. This indicates that both cellular and humoral immune responses were induced by the feed-based adjuvanted vaccine. The economic impact of a feed-based adjuvanted vaccine was examined following vaccination, considering the growth performance and feed utilization of the fish. It was found that the Economic Performance Index and Economic Conversion Ratio were unaffected by vaccination, further demonstrating that there are no negative impacts associated with administering a feed-based vaccine to fish. In conclusion, the data from this study indicate that MONTANIDE™ GR01 is a highly valuable adjuvant for oral vaccination, as demonstrated by its ability to induce a strong immune response and effectively prevent streptococcal disease in Nile tilapia.


Subject(s)
Adjuvants, Immunologic , Cichlids , Fish Diseases , Immunity, Innate , Streptococcal Infections , Streptococcus agalactiae , Animals , Streptococcus agalactiae/immunology , Streptococcal Infections/veterinary , Streptococcal Infections/prevention & control , Streptococcal Infections/immunology , Fish Diseases/prevention & control , Fish Diseases/immunology , Cichlids/immunology , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/pharmacology , Administration, Oral , Animal Feed/analysis , Streptococcal Vaccines/immunology , Streptococcal Vaccines/administration & dosage , Vaccination/veterinary
9.
Fish Shellfish Immunol ; 149: 109534, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38575040

ABSTRACT

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.


Subject(s)
Animal Feed , Cichlids , Diet , Dietary Supplements , Fish Diseases , Streptococcal Infections , Streptococcus agalactiae , Zinc , Animals , Cichlids/immunology , Cichlids/growth & development , Dietary Supplements/analysis , Zinc/administration & dosage , Animal Feed/analysis , Diet/veterinary , Streptococcal Infections/veterinary , Streptococcal Infections/immunology , Streptococcus agalactiae/physiology , Fish Diseases/immunology , Random Allocation , Immunity, Innate/drug effects
10.
Dev Comp Immunol ; 156: 105167, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38574830

ABSTRACT

IRF9 can play an antibacterial role by regulating the type I interferon (IFN) pathway. Streptococcus iniae can cause many deaths of yellowfin seabream, Acanthopagrus latus in pond farming. Nevertheless, the regulatory mechanism of type I IFN signalling by A. latus IRF9 (AlIRF9) against S. iniae remains elucidated. In our study, AlIRF9 has a total cDNA length of 3200 bp and contains a 1311 bp ORF encoding a presumed 436 amino acids (aa). The genomic DNA sequence of AlIRF9 has nine exons and eight introns, and AlIRF9 was expressed in various tissues, containing the stomach, spleen, brain, skin, and liver, among which the highest expression was in the spleen. Moreover, AlIRF9 transcriptions in the spleen, liver, kidney, and brain were increased by S. iniae infection. By overexpression of AlIRF9, AlIRF9 is shown as a whole-cell distribution, mainly concentrated in the nucleus. Moreover, the promoter fragments of -415 to +192 bp and -311 to +196 bp were regarded as core sequences from two AlIFNa3s. The point mutation analyses verified that AlIFNa3 and AlIFNa3-like transcriptions are dependent on both M3 sites with AlIRF9. In addition, AlIRF9 could greatly reduce two AlIFNa3s and interferon signalling factors expressions. These results showed that in A. latus, both AlIFNa3 and AlIFNa3-like can mediate the regulation of AlIRF9 in the process of infection with S. iniae.


Subject(s)
Fish Diseases , Fish Proteins , Interferon-Stimulated Gene Factor 3, gamma Subunit , Sea Bream , Streptococcal Infections , Streptococcus iniae , Animals , Fish Proteins/genetics , Fish Proteins/metabolism , Streptococcal Infections/immunology , Fish Diseases/immunology , Fish Diseases/microbiology , Interferon-Stimulated Gene Factor 3, gamma Subunit/genetics , Interferon-Stimulated Gene Factor 3, gamma Subunit/metabolism , Sea Bream/genetics , Sea Bream/immunology , Sea Bream/microbiology , Streptococcus iniae/physiology , Promoter Regions, Genetic/genetics , Signal Transduction , Gene Expression Regulation , Immunity, Innate/genetics
11.
Vaccine ; 42(13): 3230-3238, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38627147

ABSTRACT

INTRODUCTION: Capsular polysaccharide (CPS) serotype-specific Immunoglobulin G (IgG) in cord blood has been proposed as a correlate of protection against invasive Group B Streptococcus (iGBS) disease. Although protective levels are required in infants throughout the window of vulnerability up to 3 months of age, little is known regarding the kinetics of GBS-specific IgG over this period. METHODS: We enrolled 33 healthy infants born to mothers colonized with GBS. We collected cord blood and infant blood samples either at one (21-35 days), two (49-63 days), or three months of age (77-91 days). We measured GBS serotype-specific CPS IgG concentrations and calculated the decay rate using a mixed-effects model. We further explored whether the antibody kinetics were affected by common maternal and infant factors and estimated the correlation between IgG concentration at birth and one, two, and three months of age. RESULTS: The half-life estimate of IgG concentration for homologous and non-homologous GBS serotypes in paired samples with detectable IgG levels at both time points was 27.4 (95 % CI: 23.5-32.9) days. The decay rate did not vary by maternal age (p = 0.7), ethnicity (p = 0.1), gravida (p = 0.1), gestation (p = 0.7), and infant sex (p = 0.1). Predicted IgG titres above the assay lower limit of quantification on day 30 strongly correlated with titres at birth (Spearman correlation coefficient 0.71 [95 % CI: 0.60-0.80]). CONCLUSION: Our results provide a basis for future investigations into the use of antibody kinetics in defining a serocorrelate of protection against late-onset iGBS disease.


Subject(s)
Antibodies, Bacterial , Immunoglobulin G , Streptococcal Infections , Streptococcus agalactiae , Humans , Antibodies, Bacterial/blood , Antibodies, Bacterial/immunology , Streptococcus agalactiae/immunology , Immunoglobulin G/blood , Infant , Female , Infant, Newborn , Streptococcal Infections/immunology , Male , United Kingdom , Fetal Blood/immunology , Cohort Studies , Pregnancy , Adult , Serogroup , Immunity, Maternally-Acquired
12.
Nat Commun ; 15(1): 3600, 2024 Apr 27.
Article in English | MEDLINE | ID: mdl-38678029

ABSTRACT

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.


Subject(s)
Antibodies, Viral , COVID-19 , Immunoglobulin Fc Fragments , Immunoglobulin G , SARS-CoV-2 , Streptococcus pyogenes , Animals , Immunoglobulin G/immunology , Streptococcus pyogenes/immunology , SARS-CoV-2/immunology , Mice , Humans , Antibodies, Viral/immunology , COVID-19/immunology , COVID-19/virology , Immunoglobulin Fc Fragments/immunology , Immunoglobulin Fc Fragments/genetics , Immunoglobulin Fc Fragments/chemistry , Streptococcal Infections/immunology , Streptococcal Infections/microbiology , Antibodies, Monoclonal/immunology , Antibodies, Bacterial/immunology , Phagocytosis , Female , Protein Engineering/methods , Mice, Inbred BALB C
13.
J Immunotoxicol ; 21(1): 2345152, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38659406

ABSTRACT

The recent global resurgence of severe infections caused by the Group A streptococcus (GAS) pathogen, Streptococcus pyogenes, has focused attention on this microbial pathogen, which produces an array of virulence factors, such as the pore-forming toxin, streptolysin O (SOT). Importantly, the interactions of SOT with human neutrophils (PMN), are not well understood. The current study was designed to investigate the effects of pretreatment of isolated human PMN with purified SOT on several pro-inflammatory activities, including generation of reactive oxygen species (ROS), degranulation (elastase release), influx of extracellular calcium (Ca2+) and release of extracellular DNA (NETosis), using chemiluminescence, spectrophotometric and fluorimetric procedures, respectively. Exposure of PMN to SOT alone caused modest production of ROS and elastase release, while pretreatment with the toxin caused significant augmentation of chemoattractant (fMLP)-activated ROS generation and release of elastase by activated PMN. These effects of treatment of PMN with SOT were associated with both a marked and sustained elevation of cytosolic Ca2+concentrations and significant increases in the concentrations of extracellular DNA, indicative of NETosis. The current study has identified a potential role for SOT in augmenting the Ca2+-dependent pro-inflammatory interactions of PMN, which, if operative in a clinical setting, may contribute to hyper-activation of PMN and GAS-mediated tissue injury.


Subject(s)
Extracellular Traps , Neutrophils , Streptococcus pyogenes , Streptolysins , Humans , Bacterial Proteins/metabolism , Calcium/metabolism , Cell Degranulation/drug effects , Cells, Cultured , Extracellular Traps/immunology , Extracellular Traps/metabolism , Inflammation/immunology , Neutrophil Activation/drug effects , Neutrophils/immunology , Neutrophils/metabolism , Neutrophils/drug effects , Pancreatic Elastase/metabolism , Reactive Oxygen Species/metabolism , Streptococcal Infections/immunology , Streptococcus pyogenes/immunology , Streptolysins/metabolism
14.
Clin Exp Nephrol ; 28(5): 359-374, 2024 May.
Article in English | MEDLINE | ID: mdl-38170299

ABSTRACT

BACKGROUND: Post-streptococcal glomerulonephritis (PSGN) is a consequence of the infection by group A beta-hemolytic streptococcus. During this infection, various immunological processes generated by streptococcal antigens are triggered, such as the induction of antibodies and immune complexes. This activation of the immune system involves both innate and acquired immunity. The immunological events that occur at the renal level lead to kidney damage with chronic renal failure as well as resolution of the pathological process (in most cases). Angiotensin II (Ang II) is a molecule with vasopressor and pro-inflammatory capacities, being an important factor in various inflammatory processes. During PSGN some events are defined that make Ang II conceivable as a molecule involved in the inflammatory processes during the disease. CONCLUSION: This review is focused on defining which reported events would be related to the presence of this hormone in PSGN.


Subject(s)
Angiotensin II , Glomerulonephritis , Streptococcal Infections , Streptococcus pyogenes , Humans , Glomerulonephritis/immunology , Glomerulonephritis/microbiology , Glomerulonephritis/etiology , Streptococcal Infections/immunology , Streptococcal Infections/complications , Streptococcal Infections/microbiology , Streptococcus pyogenes/immunology , Animals , Kidney/immunology , Kidney/pathology
15.
N Engl J Med ; 389(3): 215-227, 2023 Jul 20.
Article in English | MEDLINE | ID: mdl-37467497

ABSTRACT

BACKGROUND: Natural history studies have correlated serotype-specific anti-capsular polysaccharide (CPS) IgG in newborns with a reduced risk of group B streptococcal disease. A hexavalent CPS-cross-reactive material 197 glycoconjugate vaccine (GBS6) is being developed as a maternal vaccine to prevent invasive group B streptococcus in young infants. METHODS: In an ongoing phase 2, placebo-controlled trial involving pregnant women, we assessed the safety and immunogenicity of a single dose of various GBS6 formulations and analyzed maternally transferred anti-CPS antibodies. In a parallel seroepidemiologic study that was conducted in the same population, we assessed serotype-specific anti-CPS IgG concentrations that were associated with a reduced risk of invasive disease among newborns through 89 days of age to define putative protective thresholds. RESULTS: Naturally acquired anti-CPS IgG concentrations were associated with a reduced risk of disease among infants in the seroepidemiologic study. IgG thresholds that were determined to be associated with 75 to 95% reductions in the risk of disease were 0.184 to 0.827 µg per milliliter. No GBS6-associated safety signals were observed among the mothers or infants. The incidence of adverse events and of serious adverse events were similar across the trial groups for both mothers and infants; more local reactions were observed in the groups that received GBS6 containing aluminum phosphate. Among the infants, the most common serious adverse events were minor congenital anomalies (umbilical hernia and congenital dermal melanocytosis). GBS6 induced maternal antibody responses to all serotypes, with maternal-to-infant antibody ratios of approximately 0.4 to 1.3, depending on the dose. The percentage of infants with anti-CPS IgG concentrations above 0.184 µg per milliliter varied according to serotype and formulation, with 57 to 97% of the infants having a seroresponse to the most immunogenic formulation. CONCLUSIONS: GBS6 elicited anti-CPS antibodies against group B streptococcus in pregnant women that were transferred to infants at levels associated with a reduced risk of invasive group B streptococcal disease. (Funded by Pfizer and the Bill and Melinda Gates Foundation; C1091002 ClinicalTrials.gov number, NCT03765073.).


Subject(s)
Streptococcal Infections , Streptococcal Vaccines , Streptococcus agalactiae , Female , Humans , Infant , Infant, Newborn , Pregnancy , Antibodies, Bacterial , Immunoglobulin G , Seroepidemiologic Studies , Streptococcal Infections/epidemiology , Streptococcal Infections/immunology , Streptococcal Infections/prevention & control , Vaccines, Combined/administration & dosage , Vaccines, Combined/adverse effects , Vaccines, Combined/immunology , Vaccines, Combined/therapeutic use , Vaccines, Conjugate/administration & dosage , Vaccines, Conjugate/adverse effects , Vaccines, Conjugate/immunology , Vaccines, Conjugate/therapeutic use , Streptococcal Vaccines/administration & dosage , Streptococcal Vaccines/adverse effects , Streptococcal Vaccines/immunology , Streptococcal Vaccines/therapeutic use , Immunity, Maternally-Acquired/immunology
16.
J Immunol ; 208(5): 1232-1247, 2022 03 01.
Article in English | MEDLINE | ID: mdl-35110419

ABSTRACT

The ß protein from group B Streptococcus (GBS) is a ∼132-kDa, cell-surface exposed molecule that binds to multiple host-derived ligands, including complement factor H (FH). Many details regarding this interaction and its significance to immune evasion by GBS remain unclear. In this study, we identified a three-helix bundle domain within the C-terminal half of the B75KN region of ß as the major FH-binding determinant and determined its crystal structure at 2.5 Å resolution. Analysis of this structure suggested a role in FH binding for a loop region connecting helices α1 and α2, which we confirmed by mutagenesis and direct binding studies. Using a combination of protein cross-linking and mass spectrometry, we observed that B75KN bound to complement control protein (CCP)3 and CCP4 domains of FH. Although this binding site lies within a complement regulatory region of FH, we determined that FH bound by ß retained its decay acceleration and cofactor activities. Heterologous expression of ß by Lactococcus lactis resulted in recruitment of FH to the bacterial surface and a significant reduction of C3b deposition following exposure to human serum. Surprisingly, we found that FH binding by ß was not required for bacterial resistance to phagocytosis by neutrophils or killing of bacteria by whole human blood. However, loss of the B75KN region significantly diminished bacterial survival in both assays. Although our results show that FH recruited to the bacterial surface through a high-affinity interaction maintains key complement-regulatory functions, they raise questions about the importance of FH binding to immune evasion by GBS as a whole.


Subject(s)
Bacterial Proteins/metabolism , Immune Evasion/immunology , Membrane Proteins/metabolism , Streptococcus agalactiae/immunology , Binding Sites/physiology , Complement C3b/metabolism , Complement Factor H/metabolism , Humans , Neutrophils/immunology , Opsonization/immunology , Protein Binding/immunology , Protein Domains/genetics , Protein Domains/immunology , Streptococcal Infections/immunology , Streptococcal Infections/pathology
17.
Elife ; 112022 02 15.
Article in English | MEDLINE | ID: mdl-35166205

ABSTRACT

New therapeutic strategies to reduce sepsis-related mortality are urgently needed, as sepsis accounts for one in five deaths worldwide. Since hematopoietic stem and progenitor cells (HSPCs) are responsible for producing blood and immune cells, including in response to immunological stress, we explored their potential for treating sepsis. In a mouse model of Group A Streptococcus (GAS)-induced sepsis, severe immunological stress was associated with significant depletion of bone marrow HSPCs and mortality within approximately 5-7 days. We hypothesized that the inflammatory environment of GAS infection drives rapid HSPC differentiation and depletion that can be rescued by infusion of donor HSPCs. Indeed, infusion of 10,000 naïve HSPCs into GAS-infected mice resulted in rapid myelopoiesis and a 50-60% increase in overall survival. Surprisingly, mice receiving donor HSPCs displayed a similar pathogen load compared to untreated mice. Flow cytometric analysis revealed a significantly increased number of myeloid-derived suppressor cells in HSPC-infused mice, which correlated with reduced inflammatory cytokine levels and restored HSPC levels. These findings suggest that HSPCs play an essential immunomodulatory role that may translate into new therapeutic strategies for sepsis.


Subject(s)
Cell Differentiation/immunology , Hematopoietic Stem Cells/immunology , Immunomodulation , Sepsis/immunology , Stem Cells/immunology , Streptococcal Infections/blood , Animals , Cytokines/immunology , Female , Hematopoietic Stem Cell Transplantation/methods , Male , Mice , Mice, Inbred C57BL , Sepsis/therapy , Stem Cell Transplantation/methods , Streptococcal Infections/immunology , Streptococcus/immunology , Streptococcus/pathogenicity
18.
Fish Shellfish Immunol ; 120: 67-74, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34774734

ABSTRACT

The study was executed to find out the potential effects spent coffee ground (SCG) on Nile tilapia's skin mucosal and serum immunities, disease prevention, and growth rate reared in a biofloc system. Nile tilapia fingerlings (average weight 15.25 ± 0.07 g) were disseminated into 15 aquaria (150 L tank-1) at a density of 20 fish per aquarium and treated five diets: SCG1 (control), SCG2 (10 g kg-1), SCG3 (20 g kg-1), SCG4 (40 g kg-1), and SCG5 (80 g kg-1) for eight weeks. A Completely Randomized Design (CRD) with three replications was applied. Growth rate, skin mucus, and serum immunities were quantified every 4 weeks; whereas the challenge study was conducted at the termination of the feeding trial. The outputs indicated that dietary incorporation of SCG give rise to the enhancement of SGR and FCR in comparison with the control, with best levels noted in fish fed SCG2 diet. Similarly, significant enhancements in skin mucosal and serum immunities were revealed in fish treated SCG2 over the control and other SCG diets. Likewise, higher survival rates against Streptococcus agalactiae were displayed in fish fed SCG, with the maximum level displayed in the fish treated SCG2. In conclusion, dietary supplementation of SCG2 (10 g kg-1) can be potential used as immunostimulants in tilapia aquaculture.


Subject(s)
Cichlids , Coffee , Diet , Fish Diseases , Streptococcal Infections , Animal Feed/analysis , Animals , Aquaculture , Cichlids/immunology , Diet/veterinary , Dietary Supplements , Disease Resistance , Fish Diseases/immunology , Fish Diseases/microbiology , Streptococcal Infections/immunology , Streptococcal Infections/veterinary
19.
J Immunol Methods ; 500: 113194, 2022 01.
Article in English | MEDLINE | ID: mdl-34801540

ABSTRACT

Group A Streptococcus (GAS) is a major human pathogen responsible for superficial infections through to life-threatening invasive disease and the autoimmune sequelae acute rheumatic fever (ARF). Despite a significant global economic and health burden, there is no licensed vaccine available to prevent GAS disease. Several pre-clinical vaccines that target conserved GAS antigens are in development. Assays that measure antigen-specific antibodies are essential for vaccine research. The aim of this study was to develop a multiplex beadbased immunoassay that can detect and quantify antibody responses to multiple GAS antigen targets in small volume blood samples. This builds on our existing triplex assay comprised of antigens used in clinical serology for the diagnosis of ARF (SLO, DNase B and SpnA). Five additional conserved putative GAS vaccine antigens (Spy0843, SCPA, SpyCEP, SpyAD and the Group A carbohydrate), were coupled to spectrally unique beads to form an 8-plex antigen panel. After optimisation of the assay protocol, standard curves were generated, and assessments of assay specificity, precision and reproducibility were conducted. A broad range of antibody (IgG) titres were able to be quickly and accurately quantified from a single serum dilution. Assay utility was assessed using a panel of 62 clinical samples including serum from adults with GAS bacteraemia and children with ARF. Circulating IgG to all eight antigens was elevated in patients with GAS disease (n = 23) compared to age-matched controls (n = 39) (P < 0.05). The feasibility of using dried blood samples to quantify antigen-specific IgG was also demonstrated. In summary, a robust and reproducible 8-plex assay has been developed that simultaneously quantifies IgG antibodies to GAS vaccine and diagnostic antigens.


Subject(s)
Antigens, Bacterial/immunology , Autoimmune Diseases/diagnosis , Rheumatic Fever/diagnosis , Serologic Tests/methods , Streptococcal Infections/diagnosis , Streptococcal Vaccines/immunology , Streptococcus pyogenes/physiology , Adult , Antibodies, Bacterial/blood , Autoimmune Diseases/immunology , Child , Enzyme-Linked Immunosorbent Assay , Humans , Immunoglobulin G/blood , Microspheres , Rheumatic Fever/immunology , Streptococcal Infections/immunology , Vaccine Development
20.
Front Immunol ; 12: 786602, 2021.
Article in English | MEDLINE | ID: mdl-34899755

ABSTRACT

Streptococcus agalactiae, also known as group B streptococcus (GBS), can cause pneumonia, meningitis, and bacteremia, making it a pathogen that can increase the risk of death in newborns and immunodeficient individuals. Neutrophils are the first barrier to a host's innate immune defense against these infections. Fpr2(Formyl peptide receptor 2) is an important chemotactic receptor of neutrophils, though its activation would cause pro- and anti-inflammatory effects. In this study, we found that mice without Fpr2 receptor were highly susceptible to GBS infections. These mice demonstrated decreased chemotaxis to neutrophils, decreased bactericidal ability of neutrophils, and high mortality. RNA-seq and Luminex assay indicated that Fpr2 activates key signal molecules downstream and produces chemokines CXCL1/2 to chemotaxis neutrophils. Like Fpr2-/-, CXCL1/2 or neutrophil depletion impairs host's ability to defend against GBS infection. Altogether, these data indicate that Fpr2 contributes to a host's ability to control GBS infection and that a lack of Fpr2 was associated with selective impairment during the production of chemokines CXCL1 and CXCL2 as well as neutrophil recruitment. Here, We clarified that Fpr2, as a chemotactic receptor, could not only directly chemotactic neutrophils, but also regulate the production of chemokines to control infection by chemotactic neutrophils.


Subject(s)
Chemokine CXCL1/metabolism , Chemokine CXCL2/metabolism , Chemotaxis, Leukocyte , Neutrophil Infiltration , Neutrophils/immunology , Receptors, Formyl Peptide/metabolism , Streptococcal Infections/prevention & control , Streptococcus agalactiae/immunology , Animals , Disease Models, Animal , Host-Pathogen Interactions , Immunity, Innate , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/metabolism , Neutrophils/microbiology , Receptors, Formyl Peptide/genetics , Signal Transduction , Streptococcal Infections/immunology , Streptococcal Infections/metabolism , Streptococcal Infections/microbiology , Streptococcus agalactiae/pathogenicity , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...