Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
J Virol ; 75(24): 12421-30, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11711632

ABSTRACT

A BALB/c mouse model of enhanced pulmonary pathology following vaccination with formalin-inactivated alum-adsorbed respiratory syncytial virus (FI-RSV) and live RSV challenge was used to determine the type and kinetics of histopathologic lesions induced and chemokine gene expression profiles in lung tissues. These data were compared and contrasted with data generated following primary and/or secondary RSV infection or RSV challenge following vaccination with a promising subunit vaccine, BBG2Na. Severe peribronchiolitis and perivascularitis coupled with alveolitis and interstitial inflammation were the hallmarks of lesions in the lungs of FI-RSV-primed mice, with peak histopathology evident on days 5 and 9. In contrast, primary RSV infection resulted in no discernible lesions, while challenge of RSV-primed mice resulted in rare but mild peribronchiolitis and perivascularitis, with no evidence of alveolitis or interstitial inflammation. Importantly, mice vaccinated with a broad dose range (20 to 0.02 microg) of a clinical formulation of BBG2Na in aluminium phosphate demonstrated histopathology similar to that observed in secondary RSV infection. At the molecular level, FI-RSV priming was characterized by a rapid and strong up-regulation of eotaxin and monocyte chemotactic protein 3 (MCP-3) relative gene expression (potent lymphocyte and eosinophil chemoattractants) that was sustained through late time points, early but intermittent up-regulation of GRO/melanoma growth stimulatory activity gene and inducible protein 10 gene expression, while macrophage inflammatory protein 2 (MIP-2) and especially MCP-1 were up-regulated only at late time points. By comparison, primary RSV infection or BBG2Na priming resulted in considerably lower eotaxin and MCP-3 gene expression increases postchallenge, while expression of lymphocyte or monocyte chemoattractant chemokine genes (MIP-1beta, MCP-1, and MIP-2) were of higher magnitude and kinetics at early, but not late, time points. Our combined histopathologic and chemokine gene expression data provide a basis for differentiating between aberrant FI-RSV-induced immune responses and normal responses associated with RSV infection in the mouse model. Consequently, our data suggest that BBG2Na may constitute a safe RSV subunit vaccine for use in seronegative infants.


Subject(s)
Chemokines/genetics , Lung/pathology , Respiratory Syncytial Virus Vaccines/immunology , Animals , Chemokine CCL2/genetics , Chemokine CCL4 , Chemokine CCL5/genetics , Chemokine CXCL2 , Female , Immunization , Lung/metabolism , Macrophage Inflammatory Proteins/genetics , Mice , Mice, Inbred BALB C , Reverse Transcriptase Polymerase Chain Reaction , Vaccines, Inactivated/immunology , Vaccines, Subunit/immunology
2.
Infect Immun ; 69(10): 6434-44, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11553588

ABSTRACT

Administration of vaccines by the nasal route has recently proven to be one of the most efficient ways for inducing both mucosal and systemic antibody responses in experimental animals. Our results demonstrate that P40, a well-defined outer membrane protein A from Klebsiella pneumoniae, is indeed a carrier molecule suitable for nasal immunization. Using fragments from the respiratory syncytial virus subgroup A (RSV-A) G protein as antigen models, it has been shown that P40 is able to induce both systemic and mucosal immunity when fused or coupled to a protein or a peptide and administered intranasally (i.n.) to naive or K. pneumoniae-primed mice. Confocal analyses of nasal mucosa-associated lymphoid tissue after i.n. instillation of P40 showed that this molecule is able to cross the nasal epithelium and target CD11c-positive cells likely to be murine dendritic cells or macrophages. More importantly, this targeting of antigen-presenting cells following i.n. immunization with a subunit of the RSV-A molecule in the absence of any mucosal adjuvant results in both upper and lower respiratory tract protection against RSV-A infection.


Subject(s)
Adjuvants, Immunologic , Antigen-Presenting Cells/immunology , Antigens, Viral/immunology , Bacterial Outer Membrane Proteins/immunology , Klebsiella pneumoniae/immunology , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Vaccines/immunology , Vaccines, Synthetic/immunology , Viral Proteins/immunology , Administration, Intranasal , Amino Acid Sequence , Animals , Antibodies, Bacterial/blood , Antibodies, Bacterial/immunology , Antibodies, Viral/blood , Antibodies, Viral/immunology , Bacterial Outer Membrane Proteins/genetics , Bacterial Outer Membrane Proteins/metabolism , Biological Transport , Disease Models, Animal , Female , Humans , Immunity, Mucosal , Lymphoid Tissue/immunology , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Nasal Mucosa/immunology , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/metabolism , Respiratory Syncytial Virus Infections/immunology , Vaccination/methods
3.
Vaccine ; 19(30): 4143-52, 2001 Jul 20.
Article in English | MEDLINE | ID: mdl-11457539

ABSTRACT

Human respiratory syncytial virus (hRSV) is a major pathogen responsible for bronchiolitis and severe pulmonary disease in very young children, immunodeficient patients and the elderly. BBG2Na, a recombinant chimeric protein produced in Escherichia coli, is a promising subunit vaccine candidate against this respiratory pathogen, composed of G2Na, the central domain of RSV G glycoprotein, and BB, an albumin binding domain of streptococcal protein G. BBG2Na has a basic isoelectric point (pI 9.3) and as expected, is strongly adsorbed by aluminium phosphate (AP). Surprisingly, BBG2Na is also strongly adsorbed by aluminium hydroxide (AH), which normally binds molecules with acidic isoelectric points. This behaviour was unexpected according to the well established adsorption model of Hem and co-workers. Our observations may be explained by the bipolar two-domain structure of the BBG2Na chimera which is not reflected by the global basic isoelectric point of the whole protein: the BB domain has an acidic isoelectric point (pI 5.5) and the G2Na domain a highly basic one (pI 10.0). Importantly, formulation in either aluminium salt resulted in equally high immunogenicity and protective efficacy against RSV in mice. From a physicochemical point of view, this unique property of BBG2Na makes it eminently suitable for combination to either paediatric or elderly multivalent AH- or AP-containing vaccines already in the market or in development.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Aluminum Compounds/administration & dosage , Aluminum Hydroxide/administration & dosage , Phosphates/administration & dosage , Respiratory Syncytial Virus, Human/immunology , Vaccines, Synthetic/immunology , Viral Vaccines/immunology , Adsorption , Amino Acid Sequence , Animals , Buffers , Ethylene Glycol/pharmacology , Hydrogen-Ion Concentration , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Vaccines, Subunit/immunology
4.
Vaccine ; 19(28-29): 4036-42, 2001 Jul 16.
Article in English | MEDLINE | ID: mdl-11427280

ABSTRACT

Respiratory syncytial virus (RSV) is a major respiratory pathogen responsible for severe pulmonary disease. We have developed a parenterally administered vaccine, BBG2Na, which is currently in a phase III clinical trial. BBG2Na comprises residues 130--230 of RSV-A G protein (G2Na) fused to the BB carrier protein. In this study, we show that BBG2Na can be delivered by the nasal route and generates both mucosal and systemic antibody responses when co-administered with cholera toxin B or a newly described delivery system, zwittergent 3--14. We found that nasal BBG2Na administration protects against RSV challenge and does not induce lung immunopathology upon subsequent RSV challenge.


Subject(s)
Respiratory Syncytial Virus Vaccines/administration & dosage , Respiratory Syncytial Viruses/immunology , Administration, Intranasal , Animals , Antibodies, Viral/biosynthesis , Cholera Toxin/administration & dosage , Female , HN Protein/immunology , Humans , Immunity, Mucosal , Injections, Intramuscular , Mice , Mice, Inbred BALB C , Nasal Mucosa/drug effects , Nasal Mucosa/pathology , Quaternary Ammonium Compounds/administration & dosage , Quaternary Ammonium Compounds/toxicity , Vaccines, Subunit/administration & dosage , Viral Envelope Proteins
5.
Virology ; 285(1): 82-90, 2001 Jun 20.
Article in English | MEDLINE | ID: mdl-11414808

ABSTRACT

To understand the lack of protective immunity observed after infection with parainfluenza virus type 3 (PIV3), we tested the effect of the virus on human monocytes and monocyte-derived immature dendritic cells (DCs). Expression of viral antigens on the cell surfaces correlated with replication of the virus, which was marginal in monocytes but extremely efficient in DCs. The virus increased monocyte survival at least in part through the production of granulocyte-macrophage colony-stimulating factor but, in contrast, accelerated DC apoptosis. In addition, PIV3 infection failed to activate monocytes but induced maturation of DCs with increased expression of CD54, HLA-DR, CD86, and CD83 and production of bioactive IL-12. However, PIV3-infected DCs demonstrated low stimulatory properties in DC-T cell cocultures, a finding that could not be attributed to the production of infectious virus or IL-10. These results demonstrate for the first time that PIV3 dramatically modifies the survival and/or the function of antigen-presenting cells and might therefore prevent the development of efficient antiviral immune responses.


Subject(s)
Dendritic Cells/virology , Leukocytes, Mononuclear/virology , Parainfluenza Virus 3, Human/physiology , Antigens, CD/analysis , Antigens, Viral/analysis , Apoptosis , B7-2 Antigen , Cell Differentiation , Cell Survival , Cells, Cultured , Dendritic Cells/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/analysis , Granulocyte-Macrophage Colony-Stimulating Factor/biosynthesis , HLA-DR Antigens/analysis , Humans , Immunoglobulins/analysis , Intercellular Adhesion Molecule-1/analysis , Interleukin-12/analysis , Leukocytes, Mononuclear/immunology , Membrane Glycoproteins/analysis , RNA, Messenger/biosynthesis , Virus Replication , CD83 Antigen
6.
Vaccine ; 19(17-19): 2345-51, 2001 Mar 21.
Article in English | MEDLINE | ID: mdl-11257359

ABSTRACT

Respiratory syncytial virus (RSV) is an important respiratory pathogen in man, against which no vaccine is available. However, recent evidence suggests that antibodies to the RSV F and G proteins may play an important role in disease prevention. We previously demonstrated that BBG2Na, a subunit vaccine candidate including residues 130-230 of the Long strain G protein, protects rodents against RSV challenge. Using a panel of monoclonal antibodies (MAb) and synthetic peptides, five linear B cell epitopes were identified that mapped to residues 152-163, 165-172, 171-187 (two over-lapping epitopes) and 196-204. Antibody passive transfer and peptide immunisation studies revealed that all were protective. Pepscan analyses of anti-RSV-A and BBG2Na murine polyclonal sera suggested stronger immunogenicity of some protective epitopes (protectopes) in the context of BBG2Na compared with live virus. However, all the identified murine B cell protectopes were conserved in RSV seropositive humans. Should these protectopes correspond with protection in humans, BBG2Na may constitute a very interesting vaccine candidate against RSV.


Subject(s)
B-Lymphocytes/immunology , Respiratory Syncytial Viruses/immunology , Viral Proteins/immunology , Amino Acid Sequence , Animals , Antibodies, Monoclonal , Antibodies, Viral , Antigens, Viral/genetics , Epitope Mapping , Epitopes , Humans , Immunization, Passive , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Respiratory Syncytial Viruses/genetics , Vaccines, Subunit/genetics , Vaccines, Subunit/pharmacology , Viral Proteins/genetics , Viral Vaccines/genetics , Viral Vaccines/pharmacology
7.
Vaccine ; 18(24): 2648-55, 2000 Jun 01.
Article in English | MEDLINE | ID: mdl-10781850

ABSTRACT

Respiratory syncytial virus is one of the major respiratory pathogens for infants and immunocompromized children. With the exception of young children, all the population has encountered RSV and is seropositive. Recent reports have demonstrated however that the virus also affects the elderly and represents a major cause of illness associated with an excess of morbidity and mortality. We have generated a recombinant RSV vaccine, BBG2Na, which is highly protective in rodents against RSV infection. The aim of this study was to evaluate the ability of the vaccine to increase anti-RSV protection in RSV-primed mice and to characterize the induced immune responses. Immunization with BBG2Na increased the anti-RSV-A serum antibody titers of RSV-primed mice with induction of both IgG1 and IgG2a antibodies attesting for a mixed Th response. Moreover, the level of the induced anti-G2Na antibodies was greater in seropositive mice. Finally, sera from RSV-primed mice displayed a higher protective efficacy after transfer into naive mice following subsequent immunization with BBG2Na than sera of mice immunized with RSV-A only. Our results demonstrate that BBG2Na is immunogenic and increases the protective efficacy of serum antibodies in RSV-primed mice; they support the possibility of performing clinical trials in the seropositive human population.


Subject(s)
Antibodies, Viral/biosynthesis , Antigens, Viral/immunology , Bacterial Proteins/immunology , HN Protein , Respiratory Syncytial Viruses/immunology , Viral Envelope Proteins/immunology , Viral Fusion Proteins/immunology , Viral Proteins/immunology , Viral Vaccines/immunology , Animals , Cytokines/biosynthesis , Enzyme-Linked Immunosorbent Assay , Epitope Mapping , Humans , Mice , Recombinant Proteins/immunology , Respiratory Syncytial Viruses/isolation & purification
8.
Vaccine ; 18(24): 2735-42, 2000 Jun 01.
Article in English | MEDLINE | ID: mdl-10781861

ABSTRACT

The immunogenicity and protective efficacy of BBG2Na, a novel recombinant respiratory syncytial virus subunit vaccine candidate, was assessed in BALB/c mice under various conditions of dose, administration route and number of immunisations. A single intra-peritoneal (i.p.) dose of 2 microg, or two doses of 0.2 microg, were sufficient to induce elevated RSV-A serum antibodies and sterilising lung protective immunity. Serum antibody titres were significantly boosted following second immunisations, but not a third. Of three routes of immunisation, i.p. induced the highest RSV-A antibody titres, followed in efficacy by the intra-muscular (i. m.) and subcutaneous (s.c.) routes. Nonetheless, all three routes induced comparable and sterilising lung protection. In contrast, upper respiratory tract protection was observed only after i.p. vaccination, although significant viral titre reductions were evident following i.m. or s.c. immunisations. Interestingly, Pepscan analyses indicated that antibody epitope usage was highest in i.p. and lowest in i.m. immunised mice, respectively. Nonetheless, all routes resulted in antibody responses to known lung protective epitopes (protectopes). Thus, the prevention of serious lower respiratory tract disease, the principle goal of a RSV vaccine, but not URT infection, is dose dependent but unlikely to be influenced by the route of BBG2Na administration.


Subject(s)
Antigens, Viral/immunology , Bacterial Proteins/immunology , HN Protein , Respiratory Syncytial Viruses/immunology , Viral Envelope Proteins/immunology , Viral Fusion Proteins/immunology , Viral Proteins/immunology , Viral Vaccines/administration & dosage , Animals , Antibodies, Viral/biosynthesis , Dose-Response Relationship, Drug , Female , Humans , Injections, Intraperitoneal , Mice , Mice, Inbred BALB C , Recombinant Fusion Proteins/immunology , Respiratory Syncytial Viruses/isolation & purification , Viral Vaccines/immunology
9.
Vaccine ; 18(24): 2743-52, 2000 Jun 01.
Article in English | MEDLINE | ID: mdl-10781862

ABSTRACT

A live bacterial vaccine-delivery system based on the food-grade bacterium Staphylococcus carnosus was used for delivery of peptides from the G glycoprotein of human respiratory syncytial virus, subtype A (RSV-A). Three peptides, corresponding to the G protein amino acids, 144-159 (denoted G5), 190-203 (G9) and 171-188 (G4 S), the latter with four cysteine residues substituted for serines, were expressed by recombinant means as surface-exposed on three different bacteria, and their surface accessibility on the bacteria was verified by fluorescence-activated cell sorting (FACS). Intranasal immunization of mice with the live recombinant staphylococci elicited significant anti-peptide as well as anti-virus serum IgG responses of balanced IgG1/IgG2a isotype profiles, and upon viral challenge with 10(5) tissue culture infectious doses(50) (TCID(50)), lung protection was demonstrated for approximately half of the mice in the G9 and G4 S immunization groups. To our knowledge, this is the first study in which protective immunity to a viral pathogen has been evoked using food-grade bacteria as vaccine-delivery vehicles.


Subject(s)
Antigens, Viral/immunology , HN Protein , Respiratory Syncytial Viruses/immunology , Staphylococcus , Viral Envelope Proteins/immunology , Viral Fusion Proteins/immunology , Viral Proteins/immunology , Viral Vaccines/immunology , Administration, Intranasal , Amino Acid Sequence , Animals , Antibodies, Viral/biosynthesis , Cell Separation , Flow Cytometry , Humans , Immunoglobulin G/biosynthesis , Mice , Molecular Sequence Data , Protein Structure, Secondary , Recombinant Fusion Proteins , Surface Properties , Viral Envelope Proteins/genetics , Viral Fusion Proteins/genetics , Viral Proteins/genetics , Viral Vaccines/administration & dosage
10.
J Virol ; 74(8): 3455-63, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10729118

ABSTRACT

We analyzed the protective mechanisms induced against respiratory syncytial virus subgroup A (RSV-A) infection in the lower and upper respiratory tracts (LRT and URT) of BALB/c mice after intraperitoneal immunization with a recombinant fusion protein incorporating residues 130 to 230 of RSV-A G protein (BBG2Na). Mother-to-offspring antibody (Ab) transfer and adoptive transfer of BBG2Na-primed B cells into SCID mice demonstrated that Abs are important for LRT protection but have no effect on URT infection. In contrast, RSV-A clearance in the URT was achieved in a dose-dependent fashion after adoptive transfer of BBG2Na-primed T cells, while it was abolished in BBG2Na-immunized mice upon in vivo depletion of CD4(+), but not CD8(+), T cells. Furthermore, the conserved RSV-A G protein cysteines and residues 193 and 194, overlapping the recently identified T helper cell epitope on the G protein (P. W. Tebbey et al., J. Exp. Med. 188:1967-1972, 1998), were found to be essential for URT but not LRT protection. Taken together, these results demonstrate for the first time that CD4(+) T cells induced upon parenteral immunization with an RSV G protein fragment play a critical role in URT protection of normal mice against RSV infection.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , HN Protein , Immunization , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Viruses/immunology , Respiratory Tract Infections/prevention & control , Viral Proteins/immunology , Adoptive Transfer , Animals , B-Lymphocytes/immunology , Female , Immunity, Maternally-Acquired , Mice , Mice, Inbred BALB C , Mice, SCID , Pregnancy , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/immunology , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Viruses/genetics , Respiratory Tract Infections/virology , T-Lymphocytes/immunology , Viral Envelope Proteins , Viral Proteins/administration & dosage , Viral Proteins/genetics
11.
J Virol ; 73(7): 5637-45, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10364313

ABSTRACT

A recombinant fusion protein (BBG2Na) comprising the central conserved domain of the respiratory syncytial virus subgroup A (RSV-A) (Long) G protein (residues 130 to 230) and an albumin binding domain of streptococcal protein G was shown previously to protect mouse upper (URT) and lower (LRT) respiratory tracts against intranasal RSV challenge (U. F. Power, H. Plotnicky-Gilquin, T. Huss, A. Robert, M. Trudel, S. Stahl, M. Uhlén, T. N. Nguyen, and H. Binz, Virology 230:155-166, 1997). Panels of monoclonal antibodies (MAbs) and synthetic peptides were generated to facilitate dissection of the structural elements of this domain implicated in protective efficacy. All MAbs recognized native RSV-A antigens, and five linear B-cell epitopes were identified; these mapped to residues 152 to 163, 165 to 172, 171 to 187 (two overlapping epitopes), and 196 to 204, thereby covering the highly conserved cysteine noose domain. Antibody passive-transfer and peptide immunization studies revealed that all epitopes were implicated in protection of the LRT, but not likely the URT, against RSV-A challenge. Pepscan analyses of anti-RSV-A and anti-BBG2Na murine polyclonal sera revealed lower-level epitope usage within the central conserved region in the former, suggesting diminished immunogenicity of the implicated epitopes in the context of the whole virus. However, Pepscan analyses of RSV-seropositive human sera revealed that all of the murine B-cell protective epitopes (protectopes) that mapped to the central conserved domain were recognized in man. Should these murine protectopes also be implicated in human LRT protection, their clustering around the highly conserved cysteine noose region will have important implications for the development of RSV vaccines.


Subject(s)
Antigens, Viral/immunology , Epitopes, B-Lymphocyte/immunology , HN Protein , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus, Human/immunology , Viral Proteins/immunology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Viral/immunology , Binding Sites , Disease Models, Animal , Epitope Mapping , Female , Humans , Mice , Mice, Inbred BALB C , Peptides/chemical synthesis , Peptides/immunology , Recombinant Fusion Proteins/immunology , Respiratory Syncytial Virus Infections/prevention & control , Viral Envelope Proteins
12.
J Infect Dis ; 179(6): 1326-33, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10228051

ABSTRACT

Alum-adsorbed BBG2Na, a recombinant vaccine derived in part from the respiratory syncytial virus (RSV) subgroup A G protein, induced moderate antibody titers after 1 immunization in 1-week-old mice but conferred complete lung protection upon RSV challenge. The anti-BBG2Na IgG1-IgG2a neonatal isotype profile was suggestive of dominant Th2 responses compared with those in adults. Formulation of BBG2Na with a Th1-driving adjuvant efficiently shifted neonatal responses toward a more balanced and adultlike IgG1-IgG2a profile without compromising its protective efficacy. BBG2Na-induced protective immunity was maintained even after early life immunization in the presence of high titers of maternal antibodies. Under these conditions, the protective efficacy (86%-100%) reflected the high capacity of the nonglycosylated G2Na immunogen to escape inhibition by RSV-A-induced maternal antibodies. Thus, immunization with BBG2Na protected against viral challenge despite neonatal immunologic immaturity and the presence of maternal antibodies, two major obstacles to neonatal RSV vaccine development.


Subject(s)
HN Protein , Respiratory Syncytial Virus Infections/prevention & control , Vaccination , Viral Proteins/therapeutic use , Viral Vaccines/therapeutic use , Adjuvants, Immunologic , Animals , Animals, Newborn , Immunity, Maternally-Acquired , Mice , Mice, Inbred BALB C , Vaccines, Synthetic/therapeutic use , Viral Envelope Proteins
13.
Virology ; 258(1): 128-40, 1999 May 25.
Article in English | MEDLINE | ID: mdl-10329575

ABSTRACT

The relative immunopathogenic potential of a recombinant fusion protein incorporating residues 130-230 of respiratory syncytial virus (RSV-A) G protein (BBG2Na), formalin-inactivated RSV-A (FI-RSV), and phosphate-buffered saline (PBS) was investigated in mice after immunization and RSV challenge. FI-RSV priming resulted in massive infiltration of B cells and activated CD4(+) and CD8(+) T lymphocytes in mediastinal lymph nodes (MLN) and lungs, where eosinophilia and elevated IFN-gamma, IL-2, -4, -5, -10, and -13 mRNA transcripts were also detected. PBS-primed mice showed only elevated pulmonary IL-2 and IFN-gamma mRNAs, while an activated CD8(+) T cell peak was detected in MLN and lungs. Cell infiltration also occurred in MLN of BBG2Na-immunized mice. However, there was no evidence of T cell, B cell, or granulocyte infiltration or activation in lungs, while transient transcription of Th1-type cytokine genes was evident. The absence of pulmonary infiltration is unlikely due to insufficient viral antigen. Thus, this recombinant fusion RSV G fragment does not prime for adverse pulmonary immunopathologic responses.


Subject(s)
HN Protein , Lung/immunology , Respiratory Syncytial Virus, Human/immunology , Viral Proteins/immunology , Animals , B-Lymphocytes/immunology , Cytokines/genetics , Female , Humans , Lymph Nodes/immunology , Mice , Mice, Inbred BALB C , RNA, Messenger , Recombinant Fusion Proteins/immunology , T-Lymphocytes/immunology , Tumor Cells, Cultured , Vaccination , Viral Envelope Proteins
14.
J Infect Dis ; 176(4): 884-91, 1997 Oct.
Article in English | MEDLINE | ID: mdl-9333145

ABSTRACT

Maternal and neonatal immunization were evaluated for their capacity to induce protective immunity against respiratory syncytial virus (RSV) lower respiratory tract infections in early life. Murine models were studied by use of a novel recombinant vaccine candidate, designated BBG2Na, which was derived in part from the RSV (Long) G protein. Maternal immunization resulted in the passive transfer of high levels of RSV-A antibodies to the offspring, which protected them from RSV challenge for up to 14 weeks. Indeed, protection correlated with the detection of RSV antibodies in the serum. Neonatal immunization with BBG2Na induced significant antibody responses even in the first week of life. Most importantly, these neonatal responses were not inhibited by the presence of RSV maternal antibodies. Consequently, the combination of maternal and neonatal immunization with BBG2Na resulted in the continual presence of protective levels of antibodies in the offspring.


Subject(s)
Respiratory Syncytial Virus Infections/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/immunology , Animals , Antibodies, Viral/analysis , Antibodies, Viral/immunology , Immunity, Active , Immunity, Maternally-Acquired , Mice , Mice, Inbred BALB C , Models, Immunological , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus, Human/immunology , Specific Pathogen-Free Organisms , Vaccination , Viral Proteins/immunology
15.
Virology ; 230(2): 155-66, 1997 Apr 14.
Article in English | MEDLINE | ID: mdl-9143271

ABSTRACT

A subunit approach to the development of a respiratory syncytial virus (RSV) vaccine was investigated. It involved the production, in Escherichia coli, of an RSV (Long) G protein fragment (G2Na) as a C-terminal fusion partner to an albumin binding region (BB) of streptococcal protein G. G2Na incorporated amino acid residues 130-230 and was specifically recognized by murine anti-RSV-A polyclonal serum. In mice, intraperitoneal immunization with BBG2Na induced high anti-RSV-A serum ELISA titers and low to moderate neutralization activity. The immune response induced by BBG2Na demonstrated a potent protective efficacy against upper and lower respiratory tract RSV-A infection. The immunogenicity and protective efficacy of BBG2Na was maintained for at least 47 and 48 weeks, respectively, and was as potent and durable as live RSV-A administered in a similar fashion. Intramuscular immunization of cotton rats with BBG2Na protected lungs from both homologous and heterologous virus challenge. In contrast to mice, however, cotton rat nasal tracts were not protected after BBG2Na immunization. Consistent with antibody-mediated protection, virus was cleared within 24 hr from the lungs of BBG2Na-immunized mice. The anti-RSV-A antibodies induced in mice were exclusively of the IgG1 isotype and were detected in the serum, lungs, and nasal tracts. Passive transfer of these antibodies prevented acute, and eliminated chronic, RSV-A lung infection in normal and immunodeficient mice, respectively, confirming that such antibodies are important and sufficient for BBG2Na-induced pulmonary protection. Our results clearly demonstrate that BBG2Na contains an important immunogenic domain of the RSV G protein. The prokaryotic origin of this protein indicates that glycosylation of the RSV G protein is not necessary for protective efficacy. Thus, BBG2Na has potential as an RSV subunit vaccine.


Subject(s)
HN Protein , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus, Human/immunology , Viral Proteins/immunology , Viral Vaccines/immunology , Animals , Antibodies, Viral/immunology , Escherichia coli/metabolism , Female , Humans , Immunization, Passive , Kinetics , Mice , Mice, Inbred BALB C , Mice, SCID , Peptide Fragments/genetics , Peptide Fragments/immunology , Respiratory Syncytial Virus, Human/genetics , Sigmodontinae , Tumor Cells, Cultured , Vaccination , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Viral Envelope Proteins , Viral Proteins/genetics , Viral Vaccines/genetics
16.
Clin Exp Immunol ; 105(1): 39-45, 1996 Jul.
Article in English | MEDLINE | ID: mdl-8697633

ABSTRACT

To investigate whether HTLV-I induces the development of complement-dependent cytotoxic antibodies in humans, sera of asymptomatic HTLV-I carriers and of patients suffering from tropical spastic paraparesis/HTLV-I-associated myelopathy (TSP/HAM) or adult T cell leukaemia (ATL) were used in a cytotoxicity assay against a panel of target cells. This panel included uninfected cell lines (CEM, Jurkat, Molt and H9), cell lines chronically infected with HTLV-I (MT2, MT4, C9IPL and HUT102), as well as lines H36 (H9 infected with HTLV-I), H9-IIIB (H9 infected with HIVms) and H9-MN (H9 infected with HIMVMN). HTLV-I+ sera induced lysis of H36 and of lines expressing HTLV-I antigens in the presence of rabbit complement, but did not lyse cells in presence of human complement. The HTLV-I+ sera also failed to lyse the HTLV-I- lines and H9 cells, suggesting that lysis was specific for HTLV-I. H36 cell lysis was prevented by IgG depletion of the sera and by dialysis of rabbit complement against EGTA or EDTA. Rabbit complement-dependent cytotoxic antibodies were present in the sera of 14/14 HTLV-I-infected individuals; the highest titres were predominantly found in the sera of the TSP/HAM patients. Such antibodies were also detected in 5/5 individuals coinfected with HIV-1 and HTLV-I, although no cytotoxic antibody could be found against HIV-infected cells. Vice versa, sera of HIV-1-infected individuals did not exert a lytic effect in the presence of complement (of human or rabbit origin) against HIV-1- or HTLV-I-infected cells. Incubation of the sera of four HTLV-I-infected patients with HTLV-I env-specific synthetic peptides demonstrated that some of the complement-dependent cytotoxic antibodies recognized epitopes located on gp46 between amino acids 190 and 209. There is no correlation of rabbit complement-dependent cytotoxic HTLV-I antibodies with the development of disease.


Subject(s)
Antibodies, Viral/blood , Antibody-Dependent Cell Cytotoxicity , Antilymphocyte Serum/blood , Complement System Proteins/physiology , HTLV-I Infections/blood , HTLV-I Infections/immunology , Human T-lymphotropic virus 1/immunology , Antibodies, Viral/biosynthesis , Cell Line , HTLV-I Infections/etiology , Humans , Leukemia-Lymphoma, Adult T-Cell/etiology , Paraparesis, Tropical Spastic/etiology
SELECTION OF CITATIONS
SEARCH DETAIL
...