Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 316
Filter
1.
Nat Commun ; 11(1): 4994, 2020 10 05.
Article in English | MEDLINE | ID: mdl-33020485

ABSTRACT

Serogroup B meningococcus (MenB) is a leading cause of meningitis and sepsis across the world and vaccination is the most effective way to protect against this disease. 4CMenB is a multi-component vaccine against MenB, which is now licensed for use in subjects >2 months of age in several countries. In this study, we describe the development and use of an ad hoc protein microarray to study the immune response induced by the three major 4CMenB antigenic components (fHbp, NHBA and NadA) in individual sera from vaccinated infants, adolescents and adults. The resulting 4CMenB protein antigen fingerprinting allowed the identification of specific human antibody repertoire correlating with the bactericidal response elicited in each subject. This work represents an example of epitope mapping of the immune response induced by a multicomponent vaccine in different age groups with the identification of protective signatures. It shows the high flexibility of this microarray based methodology in terms of high-throughput information and minimal volume of biological samples needed.


Subject(s)
Antibodies, Bacterial/immunology , Antigens, Bacterial/immunology , Meningococcal Infections/immunology , Meningococcal Vaccines/immunology , Neisseria meningitidis, Serogroup B/immunology , Adolescent , Adult , Antibodies, Bacterial/blood , Child , Child, Preschool , Epitope Mapping , Humans , Infant , Meningococcal Infections/prevention & control , Peptide Library , Protein Array Analysis , Serum Bactericidal Antibody Assay , Young Adult
3.
Sci Rep ; 8(1): 3700, 2018 02 27.
Article in English | MEDLINE | ID: mdl-29487324

ABSTRACT

4CMenB is the first broad coverage vaccine for the prevention of invasive meningococcal disease caused by serogroup B strains. To gain a comprehensive picture of the antibody response induced upon 4CMenB vaccination and to obtain relevant translational information directly from human studies, we have isolated a panel of human monoclonal antibodies from adult vaccinees. Based on the Ig-gene sequence of the variable region, 37 antigen-specific monoclonal antibodies were identified and produced as recombinant Fab fragments, and a subset also produced as full length recombinant IgG1 and functionally characterized. We found that the monoclonal antibodies were cross-reactive against different antigen variants and recognized multiple epitopes on each of the antigens. Interestingly, synergy between antibodies targeting different epitopes enhanced the potency of the bactericidal response. This work represents the first extensive characterization of monoclonal antibodies generated in humans upon 4CMenB immunization and contributes to further unraveling the immunological and functional properties of the vaccine antigens. Moreover, understanding the mechanistic nature of protection induced by vaccination paves the way to more rational vaccine design and implementation.


Subject(s)
Meningococcal Infections/prevention & control , Meningococcal Vaccines/immunology , Antibodies, Bacterial/immunology , Antibodies, Monoclonal/immunology , Antigens, Bacterial/immunology , Cells, Cultured , Cross Reactions , Epitopes/immunology , Humans , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Meningococcal Infections/immunology
4.
J Viral Hepat ; 21(7): 458-65, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24750327

ABSTRACT

Hepatitis C virus (HCV) vaccines may be able to increase viral clearance in combination with antiviral therapy. We analysed viral dynamics and HCV-specific immune response during retreatment for experienced patients in a phase Ib study with E1E2MF59 vaccine. Seventy-eight genotype 1a/1b patients [relapsers (30), partial responders (16) and nonresponders (32) to interferon-(IFN)/ribavirin-(RBV)] were randomly assigned to vaccine (V:23), Peg-IFNα2a-180-ug/qw and ribavirin 1000-1200-mg/qd for 48 weeks (P/R:25), or their combination (P/R + V:30). Vaccine (100 µg/0.5 mL) was administered intramuscularly at week 0-4-8-12-24-28-32-36. Neutralizing of binding (NOB) antibodies and lymphocyte proliferation assay (LPA) for E1E2-specific-CD4 + T cells were performed at week 0-12-16-48. Viral kinetics were analysed up to week 16. The vaccine was safe, and a sustained virological response (SVR) was achieved in 4 P/R + V and 2 P/R patients. Higher SVR rates were observed in prior relapsers (P/R + V = 27.3%; P/R = 12.5%). Higher NOB titres and LPA indexes were found at week 12 and 16 in P/R + V as compared to P/R patients (P = 0.023 and 0.025, P = 0.019 and <0.001, respectively). Among the 22 patients with the strongest direct antiviral effects of IFN (ε ≥ 0.800), those treated with P/R + V (10) reached lower HCV-RNA levels (P = 0.026) at week 16. HCV E1E2MF59 vaccine in combination with Peg-IFNα2a + RBV was safe and elicited E1E2 neutralizing antibodies and specific CD4 + T cell proliferation. Upon early response to IFN, vaccinations were associated with an enhanced second phase viral load decline. These results prompt phase II trials in combination with new antiviral therapies.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Antiviral Agents/therapeutic use , Hepatitis C, Chronic/therapy , Interferon-alpha/therapeutic use , Polyethylene Glycols/therapeutic use , Polysorbates/administration & dosage , Ribavirin/therapeutic use , Squalene/administration & dosage , Viral Hepatitis Vaccines/immunology , Adjuvants, Immunologic/adverse effects , Antibodies, Neutralizing/blood , CD4-Positive T-Lymphocytes/immunology , Cell Proliferation , Combined Modality Therapy/adverse effects , Combined Modality Therapy/methods , Drug-Related Side Effects and Adverse Reactions/epidemiology , Hepatitis C Antibodies/blood , Humans , Injections, Intramuscular , Polysorbates/adverse effects , RNA, Viral/blood , Recombinant Proteins/therapeutic use , Squalene/adverse effects , Treatment Outcome , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/adverse effects , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Viral Hepatitis Vaccines/administration & dosage , Viral Hepatitis Vaccines/adverse effects , Viral Hepatitis Vaccines/genetics , Viral Load
6.
Vaccine ; 31(42): 4827-33, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-23965218

ABSTRACT

Glycoconjugate vaccines are among the most effective and safest vaccines ever developed. Diphtheria toxoid (DT), tetanus toxoid (TT) and CRM197 have been mostly used as protein carriers in licensed vaccines. We evaluated the immunogenicity of serogroup A, C, W-135 and Y meningococcal oligosaccharides conjugated to CRM197, DT and TT in naïve mice. The three carriers were equally efficient in inducing an immune response against the carbohydrate moiety in immunologically naïve mice. The effect of previous exposure to different dosages of the carrier protein on the anti-carbohydrate response was studied using serogroup A meningococcal (MenA) saccharide conjugates as a model. CRM197 showed a strong propensity to positively prime the anti-carbohydrate response elicited by its conjugates or those with the antigenically related carrier DT. Conversely in any of the tested conditions TT priming did not result in enhancement of the anti-carbohydrate response elicited by the corresponding conjugates. Repeated exposure of mice to TT or to CRM197 before immunization with the respective MenA conjugates resulted in a drastic suppression of the anti-carbohydrate response in the case of TT conjugate and only in a slight reduction in the case of CRM197. The effect of carrier priming on the anti-MenA response of DT-based conjugates varied depending on their carbohydrate to protein ratio. These data may have implications for human vaccination since conjugate vaccines are widely used in individuals previously immunized with DT and TT carrier proteins.


Subject(s)
Bacterial Proteins/administration & dosage , Diphtheria Toxoid/administration & dosage , Drug Carriers/administration & dosage , Meningococcal Vaccines/immunology , Tetanus Toxoid/administration & dosage , Animals , Meningococcal Vaccines/administration & dosage , Mice , Neisseria meningitidis, Serogroup A/immunology , Neisseria meningitidis, Serogroup C/immunology , Neisseria meningitidis, Serogroup W-135/immunology , Neisseria meningitidis, Serogroup Y/immunology , Polysaccharides, Bacterial/immunology , Vaccines, Conjugate/administration & dosage , Vaccines, Conjugate/immunology
7.
Vaccine ; 31 Suppl 2: B209-15, 2013 Apr 18.
Article in English | MEDLINE | ID: mdl-23598484

ABSTRACT

Although RSV has been a high priority for vaccine development, efforts to develop a safe and effective vaccine have yet to lead to a licensed product. Clinical and epidemiologic features of RSV disease suggest there are at least 4 distinct target populations for vaccines, the RSV naïve young infant, the RSV naïve child ≥ 6 months of age, pregnant women (to provide passive protection to newborns), and the elderly. These target populations raise different safety and efficacy concerns and may require different vaccination strategies. The highest priority target population is the RSV naïve child. The occurrence of serious adverse events associated with the first vaccine candidate for young children, formalin inactivated RSV (FI-RSV), has focused vaccine development for the young RSV naïve child on live virus vaccines. Enhanced disease is not a concern for persons previously primed by a live virus infection. A variety of live-attenuated viruses have been developed with none yet achieving licensure. New live-attenuated RSV vaccines are being developed and evaluated that maybe sufficiently safe and efficacious to move to licensure. A variety of subunit vaccines are being developed and evaluated primarily for adults in whom enhanced disease is not a concern. An attenuated parainfluenza virus 3 vector expressing the RSV F protein was evaluated in RSV naïve children. Most of these candidate vaccines have used the RSV F protein in various vaccine platforms including virus-like particles, nanoparticles, formulated with adjuvants, and expressed by DNA or virus vectors. The other surface glycoprotein, the G protein, has also been used in candidate vaccines. We now have tools to make and evaluate a wide range of promising vaccines. Costly clinical trials in the target population are needed to evaluate and select candidate vaccines for advancement to efficacy trials. Better data on RSV-associated mortality in developing countries, better estimates of the risk of long term sequelae such as wheezing after infection, better measures of protection in target populations, and data on the costs and benefits of vaccines for target populations are needed to support and justify funding this process. Addressing these challenges and needs should improve the efficiency and speed of achieving a safe and effective, licensed RSV vaccine.


Subject(s)
Biomedical Research/trends , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Vaccines/therapeutic use , Aged , Child , Female , Humans , Infant , Pregnancy , Respiratory Syncytial Virus Vaccines/adverse effects , Vaccines, Attenuated/therapeutic use , Vaccines, Inactivated/adverse effects , Vaccines, Inactivated/therapeutic use , Vaccines, Subunit/therapeutic use , Viral Fusion Proteins/immunology
8.
Public Health Genomics ; 16(1-2): 62-8, 2013.
Article in English | MEDLINE | ID: mdl-23548719

ABSTRACT

Vaccine research has experienced a quantum leap after the beginning of the genomics era. High-throughput sequencing techniques, unlimited computing resources, as well as new bioinformatic algorithms are now changing the way we perform genomic studies. Whole genome sequencing will soon become the gold standard for phylogenetic and epidemiology studies and is already shedding new light on the dynamics of bacterial evolution. We believe that deep sequencing projects, together with structural studies on vaccine candidates, will allow targeting constant epitopes and avoid vaccine failure due to antigenic variability. Systems biology, which is expected to revolutionize vaccine research and clinical studies, greatly relies on high-throughput technologies such as RNA-seq. Furthermore, genomics is a key element to develop safer vaccines, and the accuracy of deep sequencing will allow monitoring vaccine coverage after their introduction on the market.


Subject(s)
High-Throughput Nucleotide Sequencing/methods , Vaccines/therapeutic use , Algorithms , Biomedical Research/methods , Computational Biology/methods , Epitopes/chemistry , Genetic Variation , Genomics , Humans , Phylogeny , Polymorphism, Single Nucleotide , Reproducibility of Results , Systems Biology
9.
Clin Microbiol Infect ; 18 Suppl 5: 109-16, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22882709

ABSTRACT

Vaccines have a significant impact on public health, and vaccinology in the era of genomics is taking advantage of new technologies to tackle diseases for which vaccine development has so far been unsuccessful. Almost all existing vaccines were developed based on traditional vaccinology methods, which relied on empirical screening of a few candidates at a time, based on known features of the pathogen. However, the ability to sequence a pathogen's genome provides access to its entire antigenic repertoire. As such, genomics has catalysed a shift in vaccine development towards sequence-based 'Reverse Vaccinology' approaches, which use high-throughput in silico screening of the entire genome of a pathogen to identify genes that encode proteins with the attributes of good vaccine targets. Furthermore, the increasing availability of genome sequences has led to the development and application of additional technologies to vaccine discovery, including comparative genomics, transcriptomics, proteomics, immunomics and structural genomics. Vaccine candidates identified from a pathogen's genome or proteome can then be expressed as recombinant proteins and tested in appropriate in vitro or in vivo models to assess immunogenicity and protection. The process of reverse vaccinology has been applied to several pathogens, including serogroup B Neisseria meningitidis, Streptococcus agalactiae, Streptococcus pyogenes, Streptococcus pneumoniae and pathogenic Escherichia coli, and has provided scores of new candidate antigens for preclinical and clinical investigation. As novel genome-based technologies continue to emerge, it is expected that new vaccines for unmet diseases will be within reach.


Subject(s)
Bacterial Vaccines/genetics , Bacterial Vaccines/immunology , Biotechnology/methods , Reverse Genetics/methods , Technology, Pharmaceutical/methods , Vaccination/methods , Computational Biology/methods , Humans
10.
Vaccine ; 29(4): 712-20, 2011 Jan 17.
Article in English | MEDLINE | ID: mdl-21115057

ABSTRACT

An efficacious, low cost vaccine against typhoid fever, especially for young children, would make a major impact on disease burden in developing countries. The virulence capsular polysaccharide of Salmonella Typhi (Vi) coupled to recombinant mutant Pseudomonas aeruginosa exoprotein A (Vi-rEPA) has been shown to be highly efficacious. We investigated the use of carrier proteins included in infant vaccines, standardized the conjugation process and developed key assays required for routine lot release at production scale. Vi from a BSL1 organism, Citrobacter freundii, strain WR7011, was used as an alternative to Vi from S. Typhi. We showed that Vi conjugated to CRM(197), a non-toxic mutant of diphtheria toxin, widely used in commercial vaccines, was produced at high yield. Vi-CRM(197) proved immunogenic in animal studies, even without adjuvant. Thus, Vi-CRM(197) appears to be a suitable candidate for the development of a commercially viable, effective typhoid vaccine for developing countries.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Bacterial Proteins/administration & dosage , Polysaccharides, Bacterial/immunology , Rickettsial Vaccines/immunology , Typhoid Fever/prevention & control , Animals , Antibodies, Bacterial/blood , Citrobacter freundii/chemistry , Citrobacter freundii/immunology , Female , Immunization, Secondary/methods , Mice , Mice, Inbred BALB C , Polysaccharides, Bacterial/administration & dosage , Polysaccharides, Bacterial/isolation & purification , Rickettsial Vaccines/administration & dosage , Salmonella typhi/chemistry , Salmonella typhi/immunology , Typhoid Fever/immunology , Vaccination/methods , Vaccines, Conjugate/administration & dosage , Vaccines, Conjugate/immunology
11.
Infect Immun ; 77(1): 292-9, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18852235

ABSTRACT

Factor H-binding protein (fHBP; GNA1870) is one of the antigens of the recombinant vaccine against serogroup B Neisseria meningitidis, which has been developed using reverse vaccinology and is the basis of a meningococcal B vaccine entering phase III clinical trials. Binding of factor H (fH), an inhibitor of the complement alternative pathway, to fHBP enables N. meningitidis to evade killing by the innate immune system. All fHBP null mutant strains analyzed were sensitive to killing in ex vivo human whole blood and serum models of meningococcal bacteremia with respect to the isogenic wild-type strains. The fHBP mutant strains of MC58 and BZ83 (high fHBP expressors) survived in human blood and serum for less than 60 min (decrease of >2 log(10) CFU), while NZ98/254 (intermediate fHBP expressor) and 67/00 (low fHBP expressor) showed decreases of >1 log(10) CFU after 60 to 120 min of incubation. In addition, fHBP is important for survival in the presence of the antimicrobial peptide LL-37 (decrease of >3 log(10) CFU after 2 h of incubation), most likely due to electrostatic interactions between fHBP and the cationic LL-37 molecule. Hence, the expression of fHBP by N. meningitidis strains is important for survival in human blood and human serum and in the presence of LL-37, even at low levels. The functional significance of fHBP in mediating resistance to the human immune response, in addition to its widespread distribution and its ability to induce bactericidal antibodies, indicates that it is an important component of the serogroup B meningococcal vaccine.


Subject(s)
Anti-Infective Agents/pharmacology , Antigens, Bacterial/physiology , Antimicrobial Cationic Peptides/pharmacology , Bacterial Proteins/physiology , Blood/microbiology , Microbial Viability , Neisseria meningitidis/physiology , Serum/microbiology , Antigens, Bacterial/genetics , Bacterial Proteins/genetics , Blood/immunology , Blood Bactericidal Activity , Colony Count, Microbial , Gene Deletion , Humans , Neisseria meningitidis/drug effects , Neisseria meningitidis/genetics , Serum/immunology , Cathelicidins
12.
Proc Natl Acad Sci U S A ; 105(30): 10501-6, 2008 Jul 29.
Article in English | MEDLINE | ID: mdl-18650390

ABSTRACT

Oil-in-water emulsions are potent human adjuvants used for effective pandemic influenza vaccines; however, their mechanism of action is still unknown. By combining microarray and immunofluorescence analysis, we monitored the effects of the adjuvants MF59 oil-in-water emulsion, CpG, and alum in the mouse muscle. MF59 induced a time-dependent change in the expression of 891 genes, whereas CpG and alum regulated 387 and 312 genes, respectively. All adjuvants modulated a common set of 168 genes and promoted antigen-presenting cell recruitment. MF59 was the stronger inducer of cytokines, cytokine receptors, adhesion molecules involved in leukocyte migration, and antigen-presentation genes. In addition, MF59 triggered a more rapid influx of CD11b+ blood cells compared with other adjuvants. The early biomarkers selected by microarray, JunB and Ptx3, were used to identify skeletal muscle as a direct target of MF59. We propose that oil-in-water emulsions are the most efficient human vaccine adjuvants, because they induce an early and strong immunocompetent environment at the injection site by targeting muscle cells.


Subject(s)
Adjuvants, Immunologic/chemistry , Gene Expression Regulation , Influenza Vaccines/chemistry , Alum Compounds/chemistry , Animals , CD11b Antigen/biosynthesis , CpG Islands , Cytokines/metabolism , Genes, MHC Class II , Histocompatibility Antigens Class II/biosynthesis , Humans , Mice , Muscle, Skeletal/metabolism , Polysorbates/pharmacology , Quadriceps Muscle/metabolism , Squalene/pharmacology
13.
Mol Microbiol ; 66(2): 329-40, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17850254

ABSTRACT

Adherence to host cells is important in microbial colonization of a mucosal surface, and Streptococcus pneumoniae adherence was significantly enhanced by expression of an extracellular pilus composed of three subunits, RrgA, RrgB and RrgC. We sought to determine which subunit(s) confers adherence. Bacteria deficient in RrgA are significantly less adherent than wild-type organisms, while overexpression of RrgA enhances adherence. Recombinant monomeric RrgA binds to respiratory cells, as does RrgC with less affinity, and pre-incubation of epithelial cells with RrgA reduces adherence of wild-type piliated pneumococci. Non-adherent RrgA-negative, RrgB- and RrgC-positive organisms produce pili, suggesting that pilus-mediated adherence is due to expression of RrgA, rather than the pilus backbone itself. In contrast, RrgA-positive strains with disrupted rrgB and rrgC genes exhibit wild-type adherence despite failure to produce pili by Western blot or immunoelectron microscopy. The density of bacteria colonizing the upper respiratory tract of mice inoculated with piliated RrgA-negative pneumococci was significantly less compared with wild-type; in contrast, non-piliated pneumococci expressing non-polymeric RrgA had similar numbers of bacteria in the nasopharynx as piliated wild-type bacteria. These data suggest that RrgA is central in pilus-mediated adherence and disease, even in the absence of polymeric pilus production.


Subject(s)
Adhesins, Bacterial/physiology , Bacterial Adhesion/physiology , Fimbriae Proteins/physiology , Streptococcus pneumoniae/metabolism , Adhesins, Bacterial/genetics , Adhesins, Bacterial/metabolism , Animals , Bacterial Adhesion/genetics , Blotting, Western , Cell Line, Tumor , Epithelial Cells/microbiology , Fimbriae Proteins/genetics , Fimbriae Proteins/metabolism , Gene Expression Regulation, Bacterial , Humans , Mice , Mice, Inbred C57BL , Microscopy, Immunoelectron , Protein Binding , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Streptococcus pneumoniae/genetics , Streptococcus pneumoniae/ultrastructure
15.
Proc Natl Acad Sci U S A ; 103(8): 2857-62, 2006 Feb 21.
Article in English | MEDLINE | ID: mdl-16481624

ABSTRACT

Streptococcus pneumoniae (pneumococcus) is a major cause of morbidity and mortality world-wide. The initial event in invasive pneumococcal disease is the attachment of encapsulated pneumococci to epithelial cells in the upper respiratory tract. This work provides evidence that initial bacterial adhesion and subsequent ability to cause invasive disease is enhanced by pili, long organelles able to extend beyond the polysaccharide capsule, previously unknown to exist in pneumococci. These adhesive pili-like appendages are encoded by the pneumococcal rlrA islet, present in some, but not all, clinical isolates. Introduction of the rlrA islet into an encapsulated rlrA-negative isolate allowed pilus expression, enhanced adherence to lung epithelial cells, and provided a competitive advantage upon mixed intranasal challenge of mice. Furthermore, a pilus-expressing rlrA islet-positive clinical isolate was more virulent than a nonpiliated deletion mutant, and it out-competed the mutant in murine models of colonization, pneumonia, and bacteremia. Additionally, piliated pneumococci evoked a higher TNF response during systemic infection, compared with nonpiliated derivatives, suggesting that pneumococcal pili not only contribute to adherence and virulence but also stimulate the host inflammatory response.


Subject(s)
Fimbriae, Bacterial/physiology , Genes, Bacterial/physiology , Genomic Islands , Pneumonia, Bacterial/microbiology , Streptococcus pneumoniae/genetics , Streptococcus pneumoniae/pathogenicity , Animals , Bacterial Adhesion/genetics , Bacterial Proteins/genetics , Fimbriae, Bacterial/genetics , Fimbriae, Bacterial/ultrastructure , Genes, Bacterial/genetics , Genomic Islands/genetics , Genomic Islands/physiology , Mice , Mice, Inbred C57BL , Mutation , Respiratory Mucosa/microbiology , Streptococcus pneumoniae/ultrastructure , Trans-Activators/genetics , Virulence
16.
Scand J Immunol ; 63(3): 223-31, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16499576

ABSTRACT

Sixty-five healthy adult volunteers were immunized four times at 1-week intervals with an inactivated whole-virus influenza vaccine based on the strain A/New Caledonia/20/99 (H1N1) without adjuvant. The vaccine was administered as nasal spray with a newly developed device to secure intranasal delivery (OptiMist, OptiNose AS, Oslo, Norway), as regular nasal spray, nasal drops or as an oral spray. Significant IgA-antibody responses in nasal secretions were induced in volunteers immunized intranasally but not after oral spray immunization. In saliva, IgA antibodies were only marginally amplified even after oral spray immunizations. At least 73% of the volunteers belonging to any group of vaccine delivery reached serum haemagglutination inhibition titres of 40 or higher, considered protective against influenza, after only two vaccine doses. Those who had the vaccine delivered intranasally also showed evidence from in vitro secretion of granzyme B that cytotoxic T cells had been stimulated. Although immunization with the breath-actuated OptiMist device and nasal drops were superior with respect to both mucosal and systemic immune responses, oral spray immunization might still be considered for studies of mucosal adjuvants that are not yet acceptable for intranasal use.


Subject(s)
Antibodies/metabolism , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Immunization/methods , Influenza Vaccines/administration & dosage , Influenza, Human/prevention & control , Administration, Intranasal , Adolescent , Adult , Dose-Response Relationship, Drug , Female , Humans , Immunity, Mucosal , Immunoglobulin A/metabolism , Male , Middle Aged , Nasal Mucosa/immunology , Nasal Mucosa/metabolism , Saliva/immunology , T-Lymphocytes/immunology
17.
Curr Mol Med ; 5(7): 677-97, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16305493

ABSTRACT

In late 2002 a new disease, severe atypical respiratory syndrome (SARS), emerged in China. A hitherto unknown animal coronavirus (CoV) that had crossed the species barrier through close contact of humans with infected animals was identified as the etiological agent. It rapidly adapted to the new host and not only became readily transmissible between humans but also more pathogenic. Air travel spread it rapidly around the world and ultimately the virus infected 8096 people and caused 774 deaths in 26 countries on 5 continents. Aggressive quarantine measures successfully terminated the disease. Currently, there are no SARS cases recorded and most likely the virus no longer circulates in the human population. In this review we present an overview over SARS-Co virus biology, the disease and discuss strategies to develop antiviral drugs and vaccines.


Subject(s)
Severe Acute Respiratory Syndrome/therapy , Severe acute respiratory syndrome-related coronavirus/physiology , Animals , Disease Models, Animal , Disease Reservoirs/virology , Humans , Phylogeny , Severe acute respiratory syndrome-related coronavirus/genetics , Severe acute respiratory syndrome-related coronavirus/metabolism , Severe acute respiratory syndrome-related coronavirus/ultrastructure , Severe Acute Respiratory Syndrome/epidemiology , Severe Acute Respiratory Syndrome/transmission , Severe Acute Respiratory Syndrome/virology , Viral Vaccines/immunology
18.
Cell Biol Int ; 29(8): 605-15, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15985378

ABSTRACT

Understanding the response of chondrocytes to topographical cues and chemical patterns could provide invaluable information to advance the repair of chondral lesions. We studied the response of primary chondrocytes to nano- and micro-grooved surfaces, and sulphated hyaluronic acid (HyalS). Cells were grown on grooves ranging from 80 nm to 9 microm in depth, and from 2 microm to 20 microm in width. Observations showed that the cells did not spread appreciably on any groove size, or alter morphology or F-actin organization, although cells showed accelerated movement on 750 nm deep grooves in comparison to flat surfaces. On chemical patterns, the cells migrated onto, and preferentially attached to, HyalS and showed a greater degree of spreading and F-actin re-arrangement. This study shows that 750 nm deep grooves and sulphated hyaluronic acid elicit responses from primary chondrocytes, and this could have implications for the future direction of cartilage reconstruction and orthopaedic treatments in general.


Subject(s)
Cartilage, Articular/cytology , Chondrocytes/cytology , Hyaluronic Acid/pharmacology , Actins/metabolism , Animals , Biocompatible Materials , Cell Adhesion/physiology , Cell Culture Techniques , Cell Movement/physiology , Chondrocytes/drug effects , Chondrocytes/ultrastructure , Cytoskeleton/metabolism , Hyaluronic Acid/analogs & derivatives , Microscopy, Atomic Force , Sheep , Surface Properties/drug effects
20.
Infect Immun ; 70(9): 4785-90, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12183520

ABSTRACT

Most vaccines are still given parenterally. Mucosal vaccination would offer different advantages over parenteral immunization, including blocking of the pathogens at the portal of entry. In this paper, nontoxic Escherichia coli heat-labile enterotoxin (LT) mutants and Supramolecular Biovector systems (SMBV) were evaluated in mice as mucosal adjuvants and delivery systems, respectively, for intranasal immunization with the conjugated group C meningococcal vaccine. The conjugated vaccine formulated together with the LT mutants and the SMBV induced very high titers of serum and mucosal antibodies specific for the group C meningococcal polysaccharide. This vaccination strategy also induced high titers of antibodies with bactericidal activity, which is known to correlate with efficacy. Importantly, the mucosal vaccination, but not the conventional parenteral vaccination, induced bactericidal antibodies at the mucosal level. These data strongly support the feasibility of development of intranasal vaccines with an enhanced protective efficacy against meningococci and possibly against other encapsulated bacteria.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Bacterial Toxins/administration & dosage , Enterotoxins/administration & dosage , Escherichia coli Proteins , Meningococcal Vaccines/administration & dosage , Administration, Intranasal , Animals , Antibodies, Bacterial/biosynthesis , Antibodies, Bacterial/blood , Bacterial Toxins/genetics , Enterotoxins/genetics , Escherichia coli/genetics , Escherichia coli/immunology , Female , Humans , Immunity, Mucosal , Mice , Mice, Inbred BALB C , Mutation , Neisseria meningitidis/immunology , Vaccines, Conjugate/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...