Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters











Publication year range
1.
Vaccine ; 42(22): 125836, 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-38772837

ABSTRACT

BACKGROUND: Intranasal administration of respiratory vaccines offers many advantages such as eliciting both systemic and mucosal immunity at the point of viral entry. Immunogenicity of intranasal vaccination can be improved through the use of adjuvants. Bacteria-like particles derived fromLactococcus lactishave the potential to serve as a vaccine adjuvant.This clinical study investigated the safety, reactogenicity and immunogenicity of intranasal seasonal influenza vaccine adjuvanted with gram-positive matrix particles (FluGEM®). METHODS: This was a first-in-human, randomized, double-blind, controlled, dose-escalation study performed at the Centre for Human Drug Research (CHDR), the Netherlands. Participants aged 18-49 were randomized in a 3:1 ratio to receive FluGem® in ascending doses (two-dose regimens) together with a standard trivalent inactivated influenza vaccine or unadjuvanted TIV only. Primary outcomes were safety and tolerability. Secondary outcomes were serum hemagglutination inhibition (HI) antibody titers and mucosal IgA. The most immunogenic dose was used in an additionalelderly cohort (>65 years). RESULTS: Ninty participants were included. Intranasal FluGem®was safe and well tolerated. The majority of adverse events were mild (97.4 %) with (un)solicited adverse events comparable across all dose levels and control groups. All groups showed geometric mean increases ≥ 2.5-fold. Seroconversion (≥40 % participants) was achieved at both day 21 (single-dose) and 42 (two-dose) for the 1.25 mg dose and on day 42 (two-dose only) for the 2.5 mg dose. Highest geometric mean IgA increases were observed in the 1.25 mg group on day 21. Immunogenicity was less pronounced in elderly. CONCLUSIONS: Intranasal vaccination of FluGEM®was safe and tolerable in healthy adult volunteers aged 18-49 years and 65 and older. Highest immunogenicity was observed for 1.25 mg and 2.5 mg doses (compared to 5 mg) suggesting a potential non-linear dose-response relationship.More research is needed to further investigate the capabilities of bacteria-like peptides as adjuvants.


Subject(s)
Administration, Intranasal , Antibodies, Viral , Hemagglutination Inhibition Tests , Influenza Vaccines , Influenza, Human , Humans , Influenza Vaccines/immunology , Influenza Vaccines/administration & dosage , Influenza Vaccines/adverse effects , Adult , Male , Female , Middle Aged , Double-Blind Method , Antibodies, Viral/blood , Young Adult , Influenza, Human/prevention & control , Influenza, Human/immunology , Adolescent , Immunogenicity, Vaccine , Adjuvants, Vaccine/administration & dosage , Aged , Immunoglobulin A/blood , Healthy Volunteers , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/adverse effects , Netherlands , Immunity, Mucosal , Vaccination/methods
4.
J Immunol ; 202(7): 2005-2016, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30787109

ABSTRACT

Yersinia enterocolitica causes a severe enteric infection in infants and young children. There is no vaccine approved for use in humans. We investigated the immunogenicity and protective capacity of Yersinia YopB, a conserved type III secretion system protein, alone or combined with LcrV in adult mice immunized intranasally. YopB or LcrV (5 µg) administered with the Escherichia coli double mutant heat-labile toxin (dmLT) adjuvant afforded modest (10-30%) protection against lethal Y. enterocolitica oral infection. The combination of YopB and LcrV (5 µg each) dramatically improved vaccine efficacy (70-80%). Additionally, it afforded complete protection against Y. pestis pulmonary infection. Immunization with YopB/LcrV+dmLT resulted in Ag-specific serum IgG, systemic and mucosal Ab-secreting cells, as well as IFN-γ, TNF-α, IL-2, IL-6, IL-17A, and KC production by spleen cells. Serum Abs elicited by YopB/LcrV+dmLT had enhanced bactericidal and opsonophagocytic killing activity. After Y. enterocolitica challenge, YopB/LcrV+dmLT-vaccinated mice exhibited intact intestinal tissue, active germinal centers in mesenteric lymph nodes, IgG+ and IgA+ plasmablasts in the lamina propria, and Abs in intestinal fluid. On the contrary, complete tissue destruction and abscesses were seen in placebo recipients that succumbed to infection. Mice immunized as infants with YopB+dmLT or LcrV+dmLT achieved 60% protection against lethal Y. enterocolitica infection, and vaccine efficacy increased to 90-100% when they received YopB/LcrV+dmLT. YopB+dmLT also afforded substantial (60%) protection when administered intradermally to infant mice. YopB/LcrV+dmLT is a promising subunit vaccine candidate with the potential to elicit broad protection against Yersinia spp.


Subject(s)
Antigens, Bacterial/immunology , Bacterial Outer Membrane Proteins/immunology , Bacterial Vaccines/immunology , Pore Forming Cytotoxic Proteins/immunology , Yersinia Infections/prevention & control , Animals , Female , Mice , Mice, Inbred BALB C , Vaccines, Subunit/immunology
5.
Am J Respir Crit Care Med ; 200(4): 481-492, 2019 08 15.
Article in English | MEDLINE | ID: mdl-30753101

ABSTRACT

Rationale: Needle-free intranasal vaccines offer major potential advantages, especially against pathogens entering via mucosal surfaces. As yet, there is no effective vaccine against respiratory syncytial virus (RSV), a ubiquitous pathogen of global importance that preferentially infects respiratory epithelial cells; new strategies are urgently required.Objectives: Here, we report the safety and immunogenicity of a novel mucosal RSV F protein vaccine linked to an immunostimulatory bacterium-like particle (BLP).Methods: In this phase I, randomized, double-blind, placebo-controlled trial, 48 healthy volunteers, aged 18-49 years, were randomly assigned to receive placebo or SynGEM (low or high dose) intranasally by prime-boost administration. The primary outcome was safety and tolerability, with secondary objectives assessing virus-specific immunogenicity.Measurements and Main Results: There were no significant differences in adverse events between placebo and vaccinated groups. SynGEM induced systemic plasmablast responses and significant, durable increases in RSV-specific serum antibody in healthy, seropositive adults. Volunteers given low-dose SynGEM (140 µg F, 2 mg BLP) required a boost at Day 28 to achieve plateau responses with a maximum fold change of 2.4, whereas high-dose recipients (350 µg F, 5 mg BLP) achieved plateau responses with a fold change of 1.5 after first vaccination that remained elevated up to 180 days after vaccination, irrespective of further boosting. Palivizumab-like antibodies were consistently induced, but F protein site ∅-specific antibodies were not detected, and virus-specific nasal IgA responses were heterogeneous, with the strongest responses in individuals with lower pre-existing antibody levels.Conclusions: SynGEM is thus the first nonreplicating intranasal RSV subunit vaccine to induce persistent antibody responses in human volunteers.Clinical trials registered with www.clinicaltrials.gov (NCT02958540).


Subject(s)
Antibodies, Viral/immunology , Immunogenicity, Vaccine , Immunoglobulin A/immunology , Nasal Mucosa/immunology , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Vaccines/administration & dosage , Administration, Intranasal , Adolescent , Adult , Antibodies, Viral/blood , Dose-Response Relationship, Drug , Double-Blind Method , Female , Healthy Volunteers , Humans , Immunization, Secondary , Male , Middle Aged , Respiratory Syncytial Virus Vaccines/adverse effects , Young Adult
6.
Immunol Invest ; 47(4): 403-415, 2018 May.
Article in English | MEDLINE | ID: mdl-29498560

ABSTRACT

BACKGROUND: Streptococcus pneumoniae is a major pathogen accounting for a large number of pneumococcal disease in worldwide. Due to the mucosal immune pathway induces both systemic and mucosal immune responses, the potential strategy to prevent pneumococcal disease may be to develop a mucosal vaccine. METHOD: In this study, we developed an intranasal pneumococcal protein vaccine based on a bacterium-like particle (BLP) delivery system. PspA is expressed and exposed on the surface of all pneumococcal strains, which confers the potential to induce immune responses to protect against pneumococcal infection. We fused one of the pneumococcal surface proteins (PspA, family2 clade4) with the protein anchor (PA) protein in order to display PspA on the surface of BLPs. RESULT: The current results showed that intranasal immunization with BLPs/PspA-PA efficiently induced both PspA-specific IgG in the serum and PspA-specific IgA in mucosal washes. And intranasal immunization of BLPs/PspA-PA could provide complete protection in a mouse challenge model with pneumococci of different two clades of both homologous and heterologous PspA families. DISCUSSION AND CONCLUSION: Thus, targeted delivery of multiple bacterial antigens via BLPs may prevent pneumococcal disease by inducing both systemic and mucosal immune responses.


Subject(s)
Bacterial Proteins/immunology , Pneumococcal Infections/prevention & control , Pneumococcal Vaccines/immunology , Pneumonia, Pneumococcal/immunology , Administration, Intranasal , Animals , Antibodies, Bacterial/immunology , Antibody Specificity , Bacterial Proteins/genetics , Disease Models, Animal , Immunity, Mucosal , Immunization , Mice , Pneumococcal Vaccines/administration & dosage , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/immunology
7.
Immunol Lett ; 187: 41-46, 2017 07.
Article in English | MEDLINE | ID: mdl-28487097

ABSTRACT

Pneumolysin (Ply) is an important virulence factor in pneumococcal infection and a conserved cholesterol-binding cytotoxin expressed by all serotypes of Streptococcus pneumoniae. We previously developed a highly detoxified Ply mutant designated Plym2 by replacement of two amino acids (C428G and W433F), which lost cytotoxicity but retained the ability to induce neutralizing antibodies. In the present work, we applied bacterium-like particles (BLPs) as a carrier and immunostimulant for the development of a Plym2 intranasal vaccine, in which the Plym2 protein was displayed on the surface of BLPs. Intranasal immunization of mice with BLP-Plym2 not only induced a high level of serum IgG antibodies but also a high level of mucosal SIgA antibodies in lung lavages. Antiserum induced by the BLP-Plym2 vaccine elicited high-titer neutralization activity which could inhibit the hemolysis of wild-type Ply. In conclusion, the BLP-Plym2 vaccine was demonstrated to be a promising strategy for intranasal immunization to enhance both systemic and mucosal immune responses.


Subject(s)
Immunity, Mucosal/drug effects , Immunization , Immunoglobulin A/immunology , Mutation, Missense , Pneumococcal Vaccines , Streptolysins , Administration, Intranasal , Amino Acid Substitution , Animals , Antibodies, Bacterial/immunology , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Female , Mice , Mice, Inbred BALB C , Pneumococcal Vaccines/genetics , Pneumococcal Vaccines/immunology , Pneumococcal Vaccines/pharmacology , Streptococcus pneumoniae/genetics , Streptococcus pneumoniae/immunology , Streptolysins/genetics , Streptolysins/immunology
8.
J Virol ; 90(13): 5965-5977, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27099320

ABSTRACT

UNLABELLED: Antibodies against the fusion (F) protein of respiratory syncytial virus (RSV) play an important role in the protective immune response to this important respiratory virus. Little is known, however, about antibody levels against multiple F-specific epitopes induced by infection or after vaccination against RSV, while this is important to guide the evaluation of (novel) vaccines. In this study, we analyzed antibody levels against RSV proteins and F-specific epitopes in human sera and in sera of vaccinated and experimentally infected cotton rats and the correlation thereof with virus neutralization. Analysis of human sera revealed substantial diversity in antibody levels against F-, G (attachment)-, and F-specific epitopes between individuals. The highest correlation with virus neutralization was observed for antibodies recognizing prefusion-specific antigenic site Ø. Nevertheless, our results indicate that high levels of antibodies targeting other parts of the F protein can also mediate a potent antiviral antibody response. In agreement, sera of experimentally infected cotton rats contained high neutralizing activity despite lacking antigenic site Ø-specific antibodies. Strikingly, vaccination with formalin-inactivated RSV (FI-RSV) exclusively resulted in the induction of poorly neutralizing antibodies against postfusion-specific antigenic site I, although antigenic sites I, II, and IV were efficiently displayed in FI-RSV. The apparent immunodominance of antigenic site I in FI-RSV likely explains the low levels of neutralizing antibodies upon vaccination and challenge and may play a role in the vaccination-induced enhancement of disease observed with such preparations. IMPORTANCE: RSV is an importance cause of hospitalization of infants. The development of a vaccine against RSV has been hampered by the disastrous results obtained with FI-RSV vaccine preparations in the 1960s that resulted in vaccination-induced enhancement of disease. To get a better understanding of the antibody repertoire induced after infection or after vaccination against RSV, we investigated antibody levels against fusion (F) protein, attachment (G) protein, and F-specific epitopes in human and animal sera. The results indicate the importance of prefusion-specific antigenic site Ø antibodies as well as of antibodies targeting other epitopes in virus neutralization. However, vaccination of cotton rats with FI-RSV specifically resulted in the induction of weakly neutralizing, antigenic site I-specific antibodies, which may play a role in the enhancement of disease observed after vaccination with such preparations.


Subject(s)
Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Epitopes/immunology , Immunity, Innate , Respiratory Syncytial Virus Vaccines/immunology , Respiratory Syncytial Virus, Human/immunology , Animals , Antibodies, Viral/immunology , Formaldehyde , Humans , Immunodominant Epitopes/blood , Immunodominant Epitopes/immunology , Rats , Respiratory Syncytial Virus Vaccines/administration & dosage , Respiratory Syncytial Virus Vaccines/adverse effects , Respiratory Syncytial Virus Vaccines/chemistry , Respiratory Syncytial Virus, Human/chemistry , Sigmodontinae , Vaccination/adverse effects , Vaccines, Inactivated/administration & dosage , Vaccines, Inactivated/adverse effects , Viral Envelope Proteins/immunology , Viral Fusion Proteins/genetics , Viral Fusion Proteins/immunology , Viral Proteins/immunology
9.
J Pharm Sci ; 104(12): 4065-4073, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26422758

ABSTRACT

Diarrhea caused by Shigella, Salmonella, and Yersinia is an important public health problem, but development of safe and effective vaccines against such diseases is challenging. A new antigen delivery platform called bacterium-like particles (BLPs) was explored as a means for delivering protective antigens from the type III secretion systems (T3SS) of these pathogens. BLPs are peptidoglycan skeletons derived from Lactococcus lactis that are safe for newborns and can carry multiple antigens. Hydrophobic T3SS translocator proteins were fused to a peptidoglycan anchor (PA) for BLP attachment. The proteins and protein-BLP complexes associated with BLPs were characterized and the resulting data used to create three-index empirical phase diagrams (EPDs). On the basis of these EPDs, IpaB (Shigella) and SipB (Salmonella) behave distinctly from YopB (Yersinia) under different environmental stresses. Adding the PA domain appears to enhance the stability of both the PA and translocator proteins, which was confirmed using differential scanning calorimetry, and although the particles dominated the spectroscopic signals in the protein-loaded BLPs, structural changes in the proteins were still detected. The protein-BLPs were most stable near neutral pH, but these proteins' hydrophobicity made them sensitive to environmental stresses.


Subject(s)
Bacterial Proteins/metabolism , Protein Transport/physiology , Type III Secretion Systems/metabolism , Antigens, Bacterial/metabolism , Bacteria/metabolism , Biophysics/methods , Lactococcus lactis/metabolism , Peptidoglycan/metabolism
10.
PLoS One ; 10(6): e0130829, 2015.
Article in English | MEDLINE | ID: mdl-26107504

ABSTRACT

The respiratory syncytial virus (RSV) fusion protein F is considered an attractive vaccine candidate especially in its prefusion conformation. We studied whether recombinant soluble RSV F proteins could be stabilized in a prefusion-like conformation by mutation of heptad repeat B (HRB). The results show that soluble, trimeric, non-cleaved RSV F protein, produced by expression of the furin cleavage site-mutated F ectodomain extended with a GCN4 trimerization sequence, is efficiently recognized by pre- as well as postfusion-specific antibodies. In contrast, a similar F protein completely lacking HRB displayed high reactivity with prefusion-specific antibodies recognizing antigenic site Ø, but did not expose postfusion-specific antigenic site I, in agreement with this protein maintaining a prefusion-like conformation. These features were dependent on the presence of the GCN4 trimerization domain. Absence of cleavage also contributed to binding of prefusion-specific antibodies. Similar antibody reactivity profiles were observed when the prefusion form of F was stabilized by the introduction of cysteine pairs in HRB. To study whether the inability to form the 6HB was responsible for the prefusion-like antibody reactivity profile, alanine mutations were introduced in HRB. Although introduction of alanine residues in HRB inhibited the formation of the 6HB, the exposure of postfusion-specific antigenic site I was not prevented. In conclusion, proteins that are not able to form the 6HB, due to mutation of HRB, may still display postfusion-specific antigenic site I. Replacement of HRB by the GCN4 trimerization domain in a non-cleaved soluble F protein resulted, however, in a protein with prefusion-like characteristics, suggesting that this HRB-lacking protein may represent a potential prefusion F protein subunit vaccine candidate.


Subject(s)
Antibodies, Viral/pharmacology , Epithelial Cells/metabolism , Respiratory Mucosa/metabolism , Respiratory Syncytial Virus, Human/genetics , Viral Fusion Proteins/genetics , Antibodies, Neutralizing/pharmacology , Binding Sites , Cell Line, Tumor , Epithelial Cells/pathology , Epithelial Cells/virology , Gene Expression , HEK293 Cells , Humans , Models, Molecular , Protein Binding , Protein Multimerization , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Respiratory Mucosa/pathology , Respiratory Mucosa/virology , Respiratory Syncytial Virus, Human/metabolism , Viral Fusion Proteins/antagonists & inhibitors , Viral Fusion Proteins/chemistry , Viral Fusion Proteins/metabolism
11.
Immunol Cell Biol ; 93(7): 641-52, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25776843

ABSTRACT

Shigella spp. are among the enteric pathogens with the highest attributable incidence of moderate-to-severe diarrhea in children under 5 years of age living in endemic areas. There are no vaccines available to prevent this disease. In this work, we investigated a new Shigella vaccine concept consisting of nonliving, self-adjuvanted, Lactococcus lactis bacterium-like particles (BLP) displaying Shigella invasion plasmid antigen (Ipa) B and IpaD and examined its immunogenicity and protective efficacy in adult and newborn/infant mice immunized via the nasal route. Unique advantages of this approach include the potential for broad protection due to the highly conserved structure of the Ipas and the safety and practicality of a probiotic-based mucosal/adjuvant delivery platform. Immunization of adult mice with BLP-IpaB and BLP-IpaD (BLP-IpaB/D) induced high levels of Ipa-specific serum IgG and stool IgA in a dose-dependent manner. Immune responses and protection were enhanced by BLP delivery. Vaccine-induced serum antibodies exhibited opsonophagocytic and cytotoxic neutralizing activity, and IpaB/D IgG titers correlated with increased survival post-challenge. Ipa-specific antibody secreting cells were detected in nasal tissue and lungs, as well as IgG in bronchoalveolar lavage. Bone marrow cells produced IpaB/D-specific antibodies and contributed to protection after adoptive transfer. The BLP-IpaB/D vaccine conferred 90% and 80% protection against S. flexneri and S. sonnei, respectively. Mice immunized with BLP-IpaB/D as newborns also developed IpaB and IpaD serum antibodies; 90% were protected against S. flexneri and 44% against S. sonnei. The BLP-IpaB/D vaccine is a promising candidate for safe, practical and potentially effective immunization of children against shigellosis.


Subject(s)
Antigens, Bacterial/immunology , Bacterial Proteins/immunology , Lactococcus lactis , Shigella Vaccines/immunology , Adjuvants, Immunologic , Administration, Intranasal , Adoptive Transfer , Aging/immunology , Animals , Animals, Newborn/immunology , Antibodies, Bacterial/biosynthesis , Antibodies, Bacterial/immunology , Bone Marrow/immunology , Bronchoalveolar Lavage Fluid/immunology , Female , Immunoglobulin A/biosynthesis , Immunoglobulin A/immunology , Immunoglobulin G/biosynthesis , Immunoglobulin G/immunology , Lung/immunology , Mice , Mice, Inbred BALB C , Nasal Mucosa/immunology , Shigella Vaccines/administration & dosage , Vaccination , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/immunology
12.
J Pharm Sci ; 104(2): 424-32, 2015 Feb.
Article in English | MEDLINE | ID: mdl-24916512

ABSTRACT

Bacterium-like particles (BLPs), derived from Lactococcus lactis, offer a self-adjuvanting delivery vehicle for subunit protein vaccines. Proteins can be specifically loaded onto the BLPs via a peptidoglycan anchoring (PA) domain. In this study, the tip proteins IpaD, SipD, and LcrV belonging to type III secretion systems of Shigella flexneri, Salmonella enterica, and Yersinia enterocolitica, respectively, were fused to the PA and loaded onto the BLPs. Herein, we biophysically characterized these nine samples and condensed the spectroscopic results into three-index empirical phase diagrams (EPDs). The EPDs show distinctions between the IpaD/SipD and LcrV subfamilies of tip proteins, based on their physical stability, even upon addition of the PA. Upon attachment to the BLPs, the BLPs become defining moiety in the spectroscopic measurements, leaving the tip proteins to have a subtle yet modulating effect on the structural integrity of the tip proteins-BLPs binding. In summary, this work provides a comprehensive view of physical stability of the tip proteins and tip protein-BLPs and serves as a baseline for screening of excipients to increase the stability of the tip protein-BLPs for future vaccine formulation.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Secretion Systems , Lactococcus lactis/chemistry , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/chemistry , Biophysical Phenomena , Particle Size
13.
Appl Microbiol Biotechnol ; 98(10): 4331-45, 2014 May.
Article in English | MEDLINE | ID: mdl-24652063

ABSTRACT

The lysin motif (LysM) was first identified by Garvey et al. in 1986 and, in subsequent studies, has been shown to bind noncovalently to peptidoglycan and chitin by interacting with N-acetylglucosamine moieties. The LysM sequence is present singly or repeatedly in a large number of proteins of prokaryotes and eukaryotes. Since the mid-1990s, domains containing one or more of these LysM sequences originating from different LysM-containing proteins have been examined for purely scientific reasons as well as for their possible use in various medical and industrial applications. These studies range from detecting localized binding of LysM-containing proteins onto bacteria to actual bacterial cell surface analysis. On a more applied level, the possibilities of employing the LysM domains for cell immobilization, for the display of peptides, proteins, or enzymes on (bacterial) surfaces as well as their utility in the development of novel vaccines have been scrutinized. To serve these purposes, the chimeric proteins containing one or more of the LysM sequences have been produced and isolated from various prokaryotic and eukaryotic expression hosts. This review gives a succinct overview of the characteristics of the LysM domain and of current developments in its application potential.


Subject(s)
Carrier Proteins/metabolism , Peptidoglycan/metabolism , Bacteria/chemistry , Bacteria/genetics , Carrier Proteins/genetics , Cell Surface Display Techniques/methods , Eukaryota/chemistry , Eukaryota/genetics , Membrane Proteins/metabolism , Protein Binding , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
14.
Front Immunol ; 4: 282, 2013.
Article in English | MEDLINE | ID: mdl-24062748

ABSTRACT

The successful development of a mucosal vaccine depends critically on the use of a safe and effective immunostimulant and/or carrier system. This review describes the effectiveness and mode of action of an immunostimulating particle, derived from bacteria, used in mucosal subunit vaccines. The non-living particles, designated bacterium-like particles are based on the food-grade bacterium Lactococcus lactis. The focus of the overview is on the development of intranasal BLP-based vaccines to prevent diseases caused by influenza and respiratory syncytial virus, and includes a selection of Phase I clinical data for the intranasal FluGEM vaccine.

15.
PLoS One ; 8(8): e71072, 2013.
Article in English | MEDLINE | ID: mdl-23951084

ABSTRACT

Respiratory syncytial virus (RSV) is an important cause of respiratory tract disease in infants and the elderly. Currently, no licensed vaccine against RSV is available. Here we describe the development of a safe and effective intranasal subunit vaccine that is based on recombinant fusion (F) protein bound to the surface of immunostimulatory bacterium-like particles (BLPs) derived from the food-grade bacterium Lactococcus lactis. Different variants of F were analyzed with respect to their conformation and reactivity with neutralizing antibodies, assuming that F proteins mimicking the metastable prefusion form of RSV F expose a more extensive and relevant epitope repertoire than F proteins corresponding to the postfusion structure. Our results indicate that the recombinant soluble ectodomain of RSV F readily adopts a postfusion conformation, generation of which cannot be prevented by C-terminal addition of a trimerization motif, but whose formation is prevented by mutation of the two furin cleavage sites in F. While the putative postfusion form of F is recognized well by the monoclonal antibody Palivizumab, this is much less so for the more potently neutralizing, prefusion-specific antibodies D25 and AM22. Both addition of the trimerization motif and mutation of the furin cleavage sites increased the reactivity of F with D25 and AM22, with the highest reactivity being observed for F proteins in which both these features were combined. Intranasal vaccination of mice or cotton rats with BLPs loaded with this latter prefusion-like F protein (BLP-F), resulted in the potent induction of F-specific immunoglobulins and in significantly decreased virus titers in the lungs upon RSV challenge. Moreover, and in contrast to animals vaccinated with formalin-inactivated RSV, animals that received BLP-F exhibited high levels of F-specific secretory IgA in the nose and RSV-neutralizing antibodies in sera, but did not show symptoms of enhanced disease after challenge with RSV.


Subject(s)
Recombinant Fusion Proteins/immunology , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Vaccines/immunology , Respiratory Syncytial Virus, Human/immunology , Viral Fusion Proteins/immunology , Administration, Intranasal , Animals , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Antibodies, Viral/blood , Antibodies, Viral/immunology , Gene Expression , Gene Order , Genetic Vectors/genetics , Humans , Lactococcus lactis/immunology , Lung/immunology , Lung/pathology , Lung/virology , Mice , Recombinant Fusion Proteins/genetics , Respiratory Syncytial Virus Vaccines/administration & dosage , Respiratory Syncytial Virus Vaccines/adverse effects , Respiratory Syncytial Virus, Human/genetics , Sigmodontinae , Vaccination , Viral Fusion Proteins/genetics
16.
PLoS One ; 8(8): e72167, 2013.
Article in English | MEDLINE | ID: mdl-23951292

ABSTRACT

Lactococcus lactis expresses the homologous glucosaminidases AcmB, AcmC, AcmA and AcmD. The latter two have three C-terminal LysM repeats for peptidoglycan binding. AcmD has much shorter intervening sequences separating the LysM repeats and a lower iso-electric point (4.3) than AcmA (10.3). Under standard laboratory conditions AcmD was mainly secreted into the culture supernatant. An L. lactis acmAacmD double mutant formed longer chains than the acmA single mutant, indicating that AcmD contributes to cell separation. This phenotype could be complemented by plasmid-encoded expression of AcmD in the double mutant. No clear difference in cellular lysis and protein secretion was observed between both mutants. Nevertheless, overexpression of AcmD resulted in increased autolysis when AcmA was present (as in the wild type strain) or when AcmA was added to the culture medium of an AcmA-minus strain. Possibly, AcmD is mainly active within the cell wall, at places where proper conditions are present for its binding and catalytic activity. Various fusion proteins carrying either the three LysM repeats of AcmA or AcmD were used to study and compare their cell wall binding characteristics. Whereas binding of the LysM domain of AcmA took place at pHs ranging from 4 to 8, LysM domain of AcmD seems to bind strongest at pH 4.


Subject(s)
Bacterial Proteins/metabolism , Glycoside Hydrolases/metabolism , Lactococcus lactis/cytology , Lactococcus lactis/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacteriolysis , Cell Wall/metabolism , Glycoside Hydrolases/chemistry , Glycoside Hydrolases/genetics , Hexosaminidases/chemistry , Hexosaminidases/genetics , Hexosaminidases/metabolism , Lactococcus lactis/chemistry , Lactococcus lactis/genetics , Mutation , Peptidoglycan/metabolism , Protein Binding , Protein Structure, Tertiary
17.
Amino Acids ; 44(5): 1329-36, 2013 May.
Article in English | MEDLINE | ID: mdl-23404517

ABSTRACT

Biosynthetic incorporation of tryptophan (Trp) analogues in recombinant proteins using an E. coli Trp auxotroph expression host is limited to analogues modified with a small substituent like a fluoro atom or a hydroxyl or amine group. We report here the efficient incorporation (>89 %) of chloro- and bromo atoms containing Trp analogues in alloproteins at high expression levels using a Lactococcus lactis Trp auxotroph strain. This result was only obtained after coexpression of the enzyme tryptophanyl-tRNA synthetase (TrpRS) of L. lactis, an enzyme believed to show a more relaxed substrate specificity than TrpRS from E. coli. Chloro- and bromo-Trps are attractive intrinsic phosphorescence probes as these Trp analogues are much less sensitive for quenchers in the medium, like oxygen, than Trp. Coexpression of TrpRS was also essential for the biosynthetic incorporation (94 %) of the Trp analogue 5,6 difluoroTrp. This makes our expression system ideally suited to generate a set of methyl- and fluoro-substituted Trp analogue-containing alloproteins in high yield for investigating the involvement of the Trp residue in cation-pi or pi-pi interactions. Taken together, the presented Trp auxotroph expression system features the most relaxed specificity for Trp analogue structures reported to date and gives a high alloprotein yield.


Subject(s)
Bacterial Proteins/biosynthesis , Lactococcus lactis/enzymology , Tryptophan-tRNA Ligase/biosynthesis , Tryptophan/analogs & derivatives , Tryptophan/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/isolation & purification , Cloning, Molecular , Fluorescent Dyes/chemistry , Fluorescent Dyes/metabolism , Lactococcus lactis/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Spectrometry, Fluorescence , Staining and Labeling , Substrate Specificity
18.
Anal Biochem ; 428(2): 111-8, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22713342

ABSTRACT

The lysin motif (LysM) is a peptidoglycan binding protein domain found in a wide range of prokaryotes and eukaryotes. Various techniques have been used to study the LysM-ligand interaction, but a sensitive spectroscopic method to directly monitor this interaction has not been reported. Here a tryptophan analog fluorescence spectroscopy approach is presented to monitor the LysM-ligand interaction using the LysM of the N-acetylglucosaminidase enzyme of Lactococcus lactis. A three-dimensional model of this LysM protein was built based on available structural information of a homolog. This model allowed choosing the amino acid positions to be labeled with a Trp analog. Four functional single-Trp LysM mutants and one double-Trp LysM mutant were constructed and biosynthetically labeled with 7-azatryptophan or 5-hydroxytryptophan. These Trp analogs feature red-shifted absorption spectra, enabling the monitoring of the LysM-ligand interaction in media with a Trp background. The emission intensities of four of the five LysM constructs were found to change markedly on exposure to either L. lactis bacterium-like particles or peptidoglycan as ligands. The method reported here is suitable to monitor LysM-ligand interactions at (sub)micromolar LysM concentrations and can be used for the detection of low levels of peptidoglycan or microbes in solutions.


Subject(s)
Lactococcus lactis/enzymology , Muramidase/chemistry , Spectrometry, Fluorescence/methods , Tryptophan/analogs & derivatives , Amino Acid Motifs , Amino Acid Sequence , Carbohydrates/chemistry , Computational Biology , Enzyme-Linked Immunosorbent Assay , Ligands , Molecular Docking Simulation , Molecular Sequence Data , Muramidase/metabolism , Protein Binding , Protein Structure, Tertiary , Sequence Alignment , Substrate Specificity , Titrimetry , Tryptophan/metabolism
19.
Vaccine ; 30(32): 4884-91, 2012 Jul 06.
Article in English | MEDLINE | ID: mdl-22537989

ABSTRACT

Administration of influenza vaccines through the intranasal (IN) route forms an attractive alternative to conventional intramuscular (IM) injection. It is not only a better accepted form of vaccine administration but it also has the potential to induce, in addition to systemic antibodies, local protective antibodies, i.e. S-IgA. Most commercially available vaccines however are inactivated non-replicating vaccines and have a low immunogenicity when administered intranasally. Local administration of these vaccines would therefore need an adjuvant to boost systemic and local antibody responses. Here we explored the use of a safe adjuvant system, i.e. bacterium-like particles (BLPs) derived from the food-grade bacterium in Lactococcus lactis, in the induction of protective antibody responses after intranasal immunization of mice. Supplementation of H1N1 split vaccine with BLPs significantly increased levels of serum influenza-specific IgG and hemagglutination-inhibiting antibodies: this was dependent on the dose of admixed BLPs and number of immunizations. Admixing BLPs further boosted local influenza-specific S-IgA antibody levels at lung and nasal mucosal sites, but also at distant mucosal sites such as the vaginal mucosal tissue. Mice immunized IN with BLP-adjuvanted vaccine and IM with non-adjuvanted vaccine were protected against weight loss upon homologous infection with H1N1 A/PR/8/34. Full protection against weight loss upon heterologous challenge with H1N1 A/PR/8/34 was seen in mice immunized IN with BLP-adjuvanted H1N1 A/New Caledonia-derived split virus vaccine, but not in those receiving the split virus vaccine IM. Mice immunized IN with BLP-adjuvanted vaccine had significantly lower lung viral titers upon homologous and heterologous challenge when compared to titers detected in mice immunized by IM injection of non-adjuvanted vaccine. Thus, adjuvantation of IN-administered influenza vaccines with BLPs effectively enhances systemic and local antibody responses leading to a superior protection against homologous and heterologous influenza infection compared to conventional IM immunization.


Subject(s)
Antibody Formation , Cross Protection , Influenza Vaccines/immunology , Lactococcus lactis/immunology , Orthomyxoviridae Infections/prevention & control , Adjuvants, Immunologic/administration & dosage , Administration, Intranasal , Animals , Antibodies, Viral/blood , Antibody Specificity , Antigens, Viral/immunology , Hemagglutination Inhibition Tests , Immunity, Mucosal , Immunoglobulin G/blood , Influenza A Virus, H1N1 Subtype , Influenza Vaccines/administration & dosage , Lung/virology , Mice , Mice, Inbred BALB C , Orthomyxoviridae Infections/immunology , Vaccines, Inactivated/administration & dosage , Vaccines, Inactivated/immunology
20.
Malar J ; 11: 50, 2012 Feb 20.
Article in English | MEDLINE | ID: mdl-22348325

ABSTRACT

BACKGROUND: Virus-like particles have been regularly used as an antigen delivery system for a number of Plasmodium peptides or proteins. The present study reports the immunogenicity and protective efficacy of bacterium-like particles (BLPs) generated from Lactococcus lactis and loaded with Plasmodium berghei circumsporozoite protein (PbCSP) peptides. METHODS: A panel of BLP-PbCSP formulations differing in composition and quantity of B-cell, CD4+ and CD8+ T-cell epitopes of PbCSP were tested in BALB/c mice. RESULTS: BLP-PbCSP1 induced specific humoral responses but no IFN-γ ELISPOT response, protecting 30-40% of the immunized mice. BLP-PbCSP2, with reduced length of the non-immunogenic part of the T-cell-epitopes construct, increased induction of IFN-γ responses as well as protection up to 60-70%. Compared to controls, lower parasitaemia was observed in unprotected mice immunized with BLP-PbCSP1 or 2, suggestive for partial immunity. Finally, further increase of the number of B-cell epitopes and codon optimization (BLP-PbCSP4) induced the highest anti-CSP antibody levels and number of IFN-γ spots, resulting in sterile immunity in 100% of the immunized mice. CONCLUSION: Presentation of Plasmodium-derived antigens using BLPs as a delivery system induced complete protection in a murine malaria model. Eventually, BLPs have the potential to be used as a novel versatile delivery platform in malaria vaccine development.


Subject(s)
Epitopes/immunology , Malaria Vaccines/immunology , Malaria/prevention & control , Plasmodium berghei/immunology , Protozoan Proteins/immunology , Animals , Bacteria/genetics , Malaria/immunology , Mice , Mice, Inbred BALB C
SELECTION OF CITATIONS
SEARCH DETAIL