Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 463
Filter
1.
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
2.
Vaccine ; 42(16): 3564-3571, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38692955

ABSTRACT

BACKGROUND: Group B Streptococcus (GBS) is a leading cause of morbidity and mortality in young infants worldwide. This study aimed to investigate candidate GBS vaccine targets, virulence factors, and antimicrobial resistance determinants. METHODS: We used whole-genome sequencing to characterize invasive GBS isolates from infants < 3 months of age obtained from a multicenter population-based study conducted from 2015 to 2021 in China. RESULTS: Overall, seven serotypes were detected from 278 GBS isolates, four (Ia, Ib, III, V) of which accounted for 97.8 %. We detected 30 sequence types (including 10 novel types) that were grouped into six clonal complexes (CCs: CC1, CC10, CC17, CC19, CC23 and CC651); three novel ST groups in CC17 were detected, and the rate of CC17, considered a hyperinvasive neonatal clone complex, was attached to 40.6 % (113/278). A total of 98.9 % (275/278) of isolates harbored at least one alpha-like protein gene. All GBS isolates contained at least one of three pilus backbone determinants and the pilus types PI-2b and PI-1 + PI-2a accounted for 79.8 % of the isolates. The 112 serotype III/CC17 GBS isolates were all positive for hvgA. Most of the isolates (75.2 %) were positive for serine-rich repeat glycoprotein determinants (srr1or srr2). Almost all isolates possessed cfb (99.6 %), c1IE (100 %), lmb (95.3 %) or pavA (100 %) gene. Seventy-seven percent of isolates harboured more than three antimicrobial resistance genes with 28.4 % (79/278) gyrA quinoloneresistancedeterminants mutation, 83.8 % (233/278) carrying tet cluster genes and 77.3 % (215/278) carrying erm genes which mediated fluoroquinolone, tetracycline and clindamycin resistance, respectively." CONCLUSIONS: The findings from this large whole-genome sequence of GBS isolates establish important baseline data required for further surveillance and evaluating the impact of future vaccine candidates.


Subject(s)
Streptococcal Infections , Streptococcal Vaccines , Streptococcus agalactiae , Virulence Factors , Whole Genome Sequencing , Humans , Streptococcus agalactiae/genetics , Streptococcus agalactiae/pathogenicity , Streptococcus agalactiae/drug effects , Streptococcus agalactiae/immunology , Streptococcus agalactiae/isolation & purification , Streptococcus agalactiae/classification , Whole Genome Sequencing/methods , Virulence Factors/genetics , Infant , Streptococcal Infections/microbiology , Streptococcal Infections/epidemiology , Streptococcal Infections/prevention & control , Streptococcal Vaccines/immunology , Infant, Newborn , China/epidemiology , Female , Serogroup , Male , Drug Resistance, Bacterial/genetics , Genome, Bacterial , Anti-Bacterial Agents/pharmacology
3.
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
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.
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
6.
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
7.
Drugs R D ; 24(1): 1-12, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38494581

ABSTRACT

Superficial infections with Streptococcus pyogenes (Strep A), pharyngitis and impetigo can induce acute rheumatic fever, an autoimmune sequela manifesting mostly with arthritis and rheumatic carditis. Valvular heart damage can persist or advance following repeated episodes of acute rheumatic fever, causing rheumatic heart disease. Acute rheumatic fever and rheumatic heart disease disproportionately affect children and young adults in developing countries and disadvantaged communities in developed countries. People living with rheumatic heart disease are at risk of experiencing potentially fatal complications such as heart failure, bacterial endocarditis or stroke. Transthoracic echocardiography plays a central role in diagnosing both rheumatic carditis and rheumatic heart disease. Despite the obvious medical need, no licensed Strep A vaccines are currently available, as their clinical development process faces several challenges, including concerns for cardiac safety. However, the development of Strep A vaccines has been recently relaunched by many vaccine developers. In this context, a reliable and consistent safety evaluation of Strep A vaccine candidates, including the use of transthoracic echocardiography for detecting cardiac adverse events, could greatly contribute to developing a safe and efficacious product in the near future. Here, we propose a framework for the consistent use of transthoracic echocardiography to proactively detect cardiac safety events in clinical trials of Strep A vaccine candidates.


Throat and skin infections caused by certain types of bacteria, named Streptococcus pyogenes, are frequent worldwide; however, in many children from less developed countries and disadvantaged communities, infections with S. pyogenes lead to a condition called acute rheumatic fever, which usually affects the joints and the heart. Damage to the heart valves may evolve to rheumatic heart disease, a permanent condition with often life-threatening complications. Rheumatic heart disease is an important health problem in places and communities where S. pyogenes infections occur frequently. A vaccine against these bacteria would help lower the number of people with valvular heart disease; however, no such vaccine exists yet. Research on vaccines against S. pyogenes was on hold for almost 30 years because of initial concerns that vaccinated children might develop acute rheumatic fever more frequently. Recently, researchers started working again on vaccines against S. pyogenes, but concerns about the safety of such vaccines persist. Doctors can reliably use echocardiography to diagnose cases of rheumatic carditis (as a sign of acute rheumatic fever) and rheumatic heart disease. Here, we propose a simple approach for the consistent use of echocardiography in clinical research of vaccines against S. pyogenes that will allow the detection of any potential heart-related side effects of the vaccine.


Subject(s)
Echocardiography , Streptococcal Infections , Streptococcal Vaccines , Streptococcus pyogenes , Humans , Streptococcus pyogenes/immunology , Echocardiography/methods , Streptococcal Vaccines/administration & dosage , Streptococcal Vaccines/adverse effects , Streptococcal Vaccines/immunology , Streptococcal Infections/prevention & control , Rheumatic Heart Disease/diagnostic imaging
9.
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
10.
Cell Rep Med ; 3(2): 100536, 2022 02 15.
Article in English | MEDLINE | ID: mdl-35243427

ABSTRACT

Clinical trials of protein-based vaccines to prevent Group B streptococcal infections are underway. In this issue of Cell Reports Medicine, Pawlowski et al.1 provide an extensive characterization of the immune response generated by the recently tested GBS-NN vaccine.


Subject(s)
Streptococcal Infections , Streptococcal Vaccines , Humans , Protein Subunits , Streptococcal Infections/prevention & control , Streptococcal Vaccines/immunology , Streptococcus agalactiae/immunology , Vaccines, Subunit
11.
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
12.
S Afr Med J ; 111(5): 487-490, 2021 Apr 30.
Article in English | MEDLINE | ID: mdl-34852893

ABSTRACT

BACKGROUND: Group A streptococcus (GAS) is a human pathogen responsible for a wide range of invasive and non-invasive infections. Pharyngitis caused by GAS may have complications such as acute rheumatic fever subsequently leading to rheumatic heart disease (RHD). RHD continues to have high morbidity and mortality and affects millions of children and young adults, mostly in developing countries. An effective preventive vaccine against GAS may reduce the morbidity and mortality. A 30-valent M-protein-based vaccine is currently at the clinical trials stage of development. Potential vaccine coverage will depend on the geographical distribution of GAS emm (M protein) types. OBJECTIVES: To determine the emm types of GAS isolates circulating in the north-west of Pretoria, South Africa. METHODS: Throat swabs were collected from patients aged 3 - 20 years presenting with pharyngitis at one local clinic. In addition, GAS clinical isolates were collected from the National Health Laboratory Service diagnostic laboratory. Emm genotyping was done on the GAS isolates by amplification of the emm gene followed by sequencing of the 5' portion of the gene. The emm types were correlated with the types in the vaccine. RESULTS: A total of 54 GAS isolates were collected, comprising 19 pharyngitis and 35 clinical isolates. We found 15 different emm types among the 43 GAS isolates that were successfully sequenced. Eleven isolates (20%) could not be typed. The most prevalent emm type was 92 (26%), which is part of the 30-valent vaccine. This was followed by emm 25 and 75, each accounting for 12% of the isolates. Up to 67% of the emm types are not covered in the 30-valent vaccine. CONCLUSIONS: Fifteen emm types were identified, of which 92 was the most prevalent. It is concerning that 67% of the emm types are not covered in the vaccine currently under development. It is recommended that surveillance studies be extended to include other parts of the country in order to expand knowledge of the circulating emm types.


Subject(s)
Pharyngitis/epidemiology , Streptococcal Infections/epidemiology , Streptococcus pyogenes/isolation & purification , Adolescent , Adult , Aged , Aged, 80 and over , Child , Child, Preschool , Cross-Sectional Studies , Genotype , Humans , Infant , Middle Aged , Pharyngitis/microbiology , South Africa/epidemiology , Streptococcal Infections/microbiology , Streptococcal Vaccines/immunology , Streptococcus pyogenes/genetics , Young Adult
13.
J Immunol ; 207(4): 1138-1149, 2021 08 15.
Article in English | MEDLINE | ID: mdl-34341168

ABSTRACT

Group A streptococcal infections are a significant cause of global morbidity and mortality. A leading vaccine candidate is the surface M protein, a major virulence determinant and protective Ag. One obstacle to the development of M protein-based vaccines is the >200 different M types defined by the N-terminal sequences that contain protective epitopes. Despite sequence variability, M proteins share coiled-coil structural motifs that bind host proteins required for virulence. In this study, we exploit this potential Achilles heel of conserved structure to predict cross-reactive M peptides that could serve as broadly protective vaccine Ags. Combining sequences with structural predictions, six heterologous M peptides in a sequence-related cluster were predicted to elicit cross-reactive Abs with the remaining five nonvaccine M types in the cluster. The six-valent vaccine elicited Abs in rabbits that reacted with all 11 M peptides in the cluster and functional opsonic Abs against vaccine and nonvaccine M types in the cluster. We next immunized mice with four sequence-unrelated M peptides predicted to contain different coiled-coil propensities and tested the antisera for cross-reactivity against 41 heterologous M peptides. Based on these results, we developed an improved algorithm to select cross-reactive peptide pairs using additional parameters of coiled-coil length and propensity. The revised algorithm accurately predicted cross-reactive Ab binding, improving the Matthews correlation coefficient from 0.42 to 0.74. These results form the basis for selecting the minimum number of N-terminal M peptides to include in potentially broadly efficacious multivalent vaccines that could impact the overall global burden of group A streptococcal diseases.


Subject(s)
Antigens, Bacterial/immunology , Bacterial Outer Membrane Proteins/immunology , Bacterial Proteins/immunology , Carrier Proteins/immunology , Cross Reactions/immunology , Streptococcal Vaccines/immunology , Animals , Antibodies, Bacterial/immunology , Epitopes/immunology , Female , Humans , Male , Mice , Peptides/immunology , Vaccines, Synthetic/immunology
14.
Vet Res ; 52(1): 112, 2021 Aug 25.
Article in English | MEDLINE | ID: mdl-34433500

ABSTRACT

A vaccine protecting against different Streptococcus suis serotypes is highly needed in porcine practice to improve animal welfare and reduce the use of antibiotics. We hypothesized that immunogens prominently recognized by convalescence sera but significantly less so by sera of susceptible piglets are putative protective antigens. Accordingly, we investigated immunogenicity and protective efficacy of a multicomponent vaccine including six main conserved immunogens, namely SSU0934, SSU1869, SSU0757, SSU1950, SSU1664 and SSU0187. Flow cytometry confirmed surface expression of all six immunogens in S. suis serotypes 2, 9 and 14. Although prime-booster vaccination after weaning resulted in significantly higher specific IgG levels against all six immunogens compared to the placebo-treated group, no significant differences between bacterial survival in blood from either vaccinated or control animals were recorded for serotype 2, 9 and 14 strains. Furthermore, vaccinated piglets were not protected against morbidity elicited through intranasal challenge with S. suis serotype 14. As ~50% of animals in both groups did not develop disease, we investigated putative other correlates of protection. Induction of reactive oxygen species (ROS) in blood granulocytes was not associated with vaccination but correlated with protection as all piglets with >5% ROS survived the challenge. Based on these findings we discuss that the main immunogens of S. suis might actually not be a priori good candidates for protective antigens. On the contrary, expression of immunogens that evoke antibodies that do not mediate killing of this pathogen might constitute an evolutionary advantage conserved in many different S. suis strains.


Subject(s)
Immunogenicity, Vaccine , Streptococcal Infections/veterinary , Streptococcal Vaccines/immunology , Streptococcus suis/immunology , Swine Diseases/prevention & control , Animals , Streptococcal Infections/microbiology , Streptococcal Infections/prevention & control , Streptococcal Vaccines/administration & dosage , Sus scrofa , Swine , Swine Diseases/microbiology , Treatment Outcome
15.
Vet Microbiol ; 259: 109165, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34225054

ABSTRACT

Streptococcus equi subspecies equi is a pathogenic bacterium that causes strangles, a highly contagious respiratory infection in horses and other equines. The limitations of current vaccines against S. equi infection warrants the development of an affordable, safe, and effective vaccine. Because gram-positive extracellular vesicles (EVs) transport various immunogenic antigens, they are attractive vaccine candidates. Here, we purified the EVs of S. equi ATCC 39506 and evaluated them as a vaccine candidate against S. equi infection in mice. As an initial step, comparative proteomic analysis was performed to characterize the functional features of the EVs. Reverse vaccinology and knowledge-based annotations were then used to screen potential vaccine candidates (PVCs) for S. equi ATCC 39506. Finally, 32 PVCs were found to be enriched in the EV fraction, suggesting the usefulness of this fraction as a vaccine. Importantly, a significantly higher survival rate after S. equi infection was detected in mice immunized with S. equi-derived EVs via the intraperitoneal route than in mice immunized with heat-killed bacteria. Of note, immunoprecipitation-mass spectrometry results validated various immunogenic antigens within the EV proteome. In conclusion, our results suggest that S. equi-derived EVs can serve as a vaccine candidate against S. equi infection.


Subject(s)
Antigens, Bacterial/immunology , Bacterial Proteins/immunology , Extracellular Vesicles/immunology , Streptococcal Infections/prevention & control , Streptococcal Vaccines/immunology , Streptococcus equi/immunology , Animals , Antigens, Bacterial/administration & dosage , Antigens, Bacterial/analysis , Bacterial Proteins/administration & dosage , Bacterial Proteins/analysis , Extracellular Vesicles/chemistry , Female , Horse Diseases/microbiology , Horse Diseases/prevention & control , Horses , Immunoprecipitation , Mass Spectrometry/methods , Mice , Mice, Inbred BALB C , Proteomics , Streptococcal Infections/immunology , Streptococcal Vaccines/administration & dosage , Vaccination
16.
Glycoconj J ; 38(4): 447-457, 2021 08.
Article in English | MEDLINE | ID: mdl-33956253

ABSTRACT

The capsular polysaccharide of the human pathogen Group B Streptococcus is a key virulence factor and vaccine candidate that induces protective antibodies when conjugated to carrier proteins. It consists of long polymeric chains of oligosaccharide repeating units, and each of the ten capsular serotypes described so far presents a unique chemical structure with distinct antigenic properties; therefore, broad protection against this pathogen could be achieved by a combination of ten glycoconjugates. Capsular polysaccharide biosynthesis and assembly follow a polymerase-dependent pathway that is widespread in encapsulated bacteria and is encoded by a polycistronic operon. Here we exploited the sequence similarity between the capsule operons of types V and IX to generate hybrid polysaccharides incorporating epitopes of both serotypes in a single molecule, by co-expressing their specific CpsM, O, I glycosyltransferases in a single isolate. Physicochemical and immunochemical methods confirmed that an engineered strain produced a high molecular weight chimeric polysaccharide, combining antigenic specificities of both type V and IX. By optimizing the copy number of key glycosyltransferase genes, we were able to modulate the ratio between type-specific epitopes. Finally, vaccination with chimeric glycoconjugates significantly decreased the incidence of disease in pups born from immunized mice challenged with either serotype. This study provides proof of concept for a new generation of glycoconjugate vaccines that combine the antigenic specificity of different polysaccharide variants in a single molecule, eliciting a protective immune response against multiple serotype variants.


Subject(s)
Bacterial Capsules/immunology , Polysaccharides, Bacterial/immunology , Streptococcal Vaccines/immunology , Streptococcus agalactiae/immunology , Vaccines, Combined/immunology , Animals , Antibodies, Monoclonal , Bacterial Proteins/immunology , Female , Genetic Engineering , Glycoconjugates , Humans , Immunity, Maternally-Acquired , Mice
17.
Res Vet Sci ; 137: 201-207, 2021 Jul.
Article in English | MEDLINE | ID: mdl-34020335

ABSTRACT

Streptococcus suis is an important zoonotic pathogen that leads to huge economic losses in the swine industry. Because of the enormous genetic and phenotypic diversity within S. suis, it is necessary to develop effective vaccines to control this zoonotic pathogen. SBP2' is a major pili subunit in S. suis that belongs to an srtBCD pili cluster and has already been reported to be associated with the pathogenesis of this bacterium. In this study, we aimed to evaluate the immunogenicity and protective ability of SBP2'. The rSBP2' protein was expressed by an Escherichia coli expression system and emulsified with Montanide ISA 201 adjuvant to prepare the subunit vaccine. Through active immune assays, the results showed that rSBP2' exhibited good immunogenicity and could protect mice from a lethal dose challenge. Additionally, the qRT-PCR data showed that the transcription levels of cytokines associated with systemic symptoms caused by S. suis were decreased, indicating that immunization with rSBP2' could protect the host from cytokine storms caused by S. suis. Furthermore, the passive immune assay showed that the humoral immunity induced by rSBP2' played an important role against S. suis infection. Taken together, SBP2' could provide proper immune protection against S. suis challenge and could be a candidate for S. suis subunit vaccine. The results of this study could provide new ideas for the development of effective vaccines against S. suis.


Subject(s)
Fimbriae, Bacterial/immunology , Immunogenicity, Vaccine , Streptococcal Infections/prevention & control , Streptococcal Vaccines/immunology , Streptococcus suis/immunology , Animals , Escherichia coli/genetics , Mice , Mice, Inbred ICR , Serogroup , Streptococcal Infections/microbiology , Streptococcus suis/genetics , Vaccines, Synthetic/immunology
18.
J Immunol ; 206(8): 1806-1816, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33811104

ABSTRACT

CD4+ T cells enable the critical B cell humoral immune protection afforded by most effective vaccines. We and others have recently identified an alternative source of help for B cells in mice, invariant NK T (iNKT) cells. iNKT cells are innate glycolipid-specific T cells restricted to the nonpolymorphic Ag-presenting molecule CD1d. As such, iNKT cells respond to glycolipids equally well in all people, making them an appealing adjuvant for universal vaccines. We tested the potential for the iNKT glycolipid agonist, α-galactosylceramide (αGC), to serve as an adjuvant for a known human protective epitope by creating a nanoparticle that delivers αGC plus antigenic polysaccharides from Streptococcus pneumoniae αGC-embedded nanoparticles activate murine iNKT cells and B cells in vitro and in vivo, facilitate significant dose sparing, and avoid iNKT anergy. Nanoparticles containing αGC plus S. pneumoniae polysaccharides elicits robust IgM and IgG in vivo and protect mice against lethal systemic S. pneumoniae However, codelivery of αGC via nanoparticles actually eliminated Ab protection elicited by a T-independent S. pneumoniae vaccine. This is consistent with previous studies demonstrating iNKT cell help for B cells following acute activation, but negative regulation of B cells during chronic inflammation. αGC-containing nanoparticles represent a viable platform for broadly efficacious vaccines against deadly human pathogens, but their potential for eliminating B cells under certain conditions suggests further clarity on iNKT cell interactions with B cells is warranted.


Subject(s)
B-Lymphocytes/immunology , Galactosylceramides/metabolism , Nanoparticles/metabolism , Natural Killer T-Cells/immunology , Pneumococcal Infections/immunology , Polysaccharides, Bacterial/metabolism , Streptococcal Vaccines/immunology , Streptococcus pneumoniae/immunology , Animals , Cells, Cultured , Galactosylceramides/immunology , Humans , Immunity, Humoral , Immunoglobulin G/metabolism , Immunoglobulin M/metabolism , Lymphocyte Activation , Mice , Polysaccharides, Bacterial/immunology , T-Lymphocytes/immunology
19.
Res Vet Sci ; 136: 422-429, 2021 May.
Article in English | MEDLINE | ID: mdl-33812285

ABSTRACT

Glyceraldehyde-3-phosphate dehydrogenase C (GapC) of Streptococcus dysgalactiae (S. dysgalactiae) is a highly conserved surface protein that can induce a protective immune response against S. dysgalactiae infection. To investigate the immune response and protective efficacy induced by epitope-vaccines against S. dysgalactiae infection, we constructed epitope-vaccines GTB1, GB1B2, and GTB1B2 using a T cell epitope (GapC63-77, abbreviated as GT) and two B cell epitopes (GapC30-36, abbreviated as GB1, and GapC97-103, abbreviated as GB2), which were identified in GapC1-150 of S. dysgalactiae in tandem by a GSGSGS linker. BALB/c mice were immunized via an intramuscular injection with the epitope vaccines. The levels of the cytokines, IFN-γ, IL-4, and IL-17, secreted by splenic lymphocytes and the antibody levels in the sera of the immunized mice were detected by ELISA. The immunized mice were subsequently challenged with S. dysgalactiae, and the bacterial colonization in the immunized-mouse organs was examined using the plate counting method. The results showed that the level of the cytokines induced by GTB1B2 was lower than that induced by GapC1-150, but higher than that induced by other epitope vaccines. The level of IgG induced by GTB1B2 was lower than that induced by GapC1-150, but higher than the levels induced by other epitope vaccines. The bacterial colonization numbers in the organs of the mice immunized with GTB1B2 were higher those of the mice immunized with GapC1-150, but significantly lower than those from the mice immunized with other epitope-vaccines. Our results demonstrated that the T cell and B cell epitopes in the epitope-vaccines worked synergistically against bacterial challenge. The multi-epitope vaccine, GTB1B2, could induce stronger cellular and humoral immune responses, and provide a better protective effect against S. dysgalactiae infection.


Subject(s)
Antigens, Bacterial/immunology , Bacterial Proteins/immunology , Immunogenicity, Vaccine , Streptococcal Vaccines/immunology , Streptococcus/immunology , Animals , Cytokines/immunology , Enzyme-Linked Immunosorbent Assay/veterinary , Epitopes, B-Lymphocyte/immunology , Epitopes, T-Lymphocyte/immunology , Female , Mice , Mice, Inbred BALB C
20.
Fish Shellfish Immunol ; 113: 162-175, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33857622

ABSTRACT

Streptococcosis and motile aeromonad septicemia (MAS) are well-known diseases in tilapia culture, which cause mass mortality with significant economic losses. The development of feed-based bivalent vaccines in controlling these diseases has been initiated, however, the mechanisms of immunities and cross-protection in fish remain unclear. This study was conducted to assess the immuno-protective as well as the cross-protective efficacy of a newly developed feed-based bivalent vaccine against Streptococcus and Aeromonas infections in red hybrid tilapia. A total of five groups of fish were vaccinated orally through two different techniques; bivalent vaccine (inactivated Streptococcus iniae and Aeromonas hydrophila) sprayed on feed pellets (BS group); bivalent vaccine (inactivated S. iniae and A. hydrophila) incorporated in feed (BI group); monovalent inactivated S. iniae and A. hydrophila vaccine separately incorporated into feed as monovalent S. iniae (MS group) and monovalent A. hydrophila (MA group); and control group (without vaccine). The feed-based vaccine was delivered orally at 5% of body weight for five consecutive days. The booster doses were given in the same manner on weeks 2 and 6. Serum and skin mucus samples were collected to assess the IgM responses using indirect ELISA. The first administration of the feed-based vaccine stimulated the IgM levels that lasted until week 3, while the second booster ensured that the IgM levels remained high for a period of 16 weeks in the BI, MS and MA groups. The BI group developed a strong and significantly (P < 0.05) higher systemic and mucosal IgM responses against both S. iniae and A. hydrophila and also cross-protective antigen S. agalactiae and A. veronii compared to the BS and control groups. Quantitative real-time PCR results also showed that the relative expressions of IL-8, INF-γ and IgM in the BI immunized fish spleen, head kidney and hindgut exhibited various significant (P < 0.05) rising trends following both the vaccination and the challenge phase. On weeks 10, all fish were challenged through the intraperitoneal route, where relative percent survivals (RPS) of 82.22 ± 3.85% when challenged with S. iniae, 77.78 ± 3.85% when challenged with A. hydrophila and 77.78 ± 3.85% when co-challenged with both S. iniae and A. hydrophila were observed in the BI group, which were significantly higher (P < 0.05) compared to the other groups. The BI group also showed significantly (P < 0.05) higher partial cross-protections following challenges with S. agalactiae (RPS at 60.00 ± 6.67%) and A. veronii (RPS at 57.78 ± 7.70%). This study demonstrated that immunization with feed-based BI vaccine elicited immune responses that were capable of protecting red hybrid tilapia against streptococcosis and MAS.


Subject(s)
Bacterial Vaccines/immunology , Cichlids/immunology , Gram-Negative Bacterial Infections/immunology , Vaccines, Combined/immunology , Animals , Gram-Negative Bacterial Infections/microbiology , Streptococcal Infections/immunology , Streptococcal Infections/microbiology , Streptococcal Infections/veterinary , Streptococcal Vaccines/immunology , Vaccines, Inactivated/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...