Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Eur J Immunol ; 46(4): 874-84, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26681580

ABSTRACT

Respiratory syncytial virus (RSV) causes severe bronchiolitis in infants worldwide. The immunological factors responsible for RSV susceptibility in infants are poorly understood. Here, we used the BALB/c mouse model of neonatal RSV infection to study the mechanisms leading to severe disease upon reexposure to the virus when adults. Two major deficiencies in neonatal lung innate responses were found: a poor DCs mobilization, and a weak engagement of the IFNI pathway. The administration of Flt3 ligand (Flt3-L), a growth factor that stimulates the proliferation of hematopoietic cells, to neonates before RSV-infection, resulted in increased lung DC number, and reconditioned the IFNI pathway upon RSV neonatal infection. Besides, neonates treated with Flt3-L were protected against exacerbated airway disease upon adult reexposure to RSV. This was associated with a reorientation of RSV-specific responses toward Th1-mediated immunity. Thus, the poor lung DCs and IFNI responses to RSV in neonates may be partly responsible for the deleterious long-term consequences revealed upon adult reexposure to RSV, which could be prevented by Flt3-L treatment. These results open new perspectives for developing neonatal immuno-modulating strategies to reduce the burden of bronchiolitis.


Subject(s)
Bronchiolitis, Viral/immunology , Immunity, Innate/immunology , Membrane Proteins/therapeutic use , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Viruses/immunology , Animals , Animals, Newborn , Bronchiolitis, Viral/prevention & control , Bronchiolitis, Viral/virology , Cell Proliferation/drug effects , Dendritic Cells/drug effects , Dendritic Cells/immunology , Disease Models, Animal , Interferon Type I/immunology , Mice , Mice, Inbred BALB C , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Infections/virology , Signal Transduction/immunology , Th1 Cells/immunology
2.
J Virol ; 88(1): 325-38, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24155388

ABSTRACT

In this study, subnucleocapsid nanorings formed by the recombinant nucleoprotein (N) of the respiratory syncytial virus were evaluated as a platform to anchor heterologous antigens. The ectodomain of the influenza virus A matrix protein 2 (M2e) is highly conserved and elicits protective antibodies when it is linked to an immunogenic carrier, making it a promising target to develop universal influenza vaccines. In this context, one or three M2e copies were genetically linked to the C terminus of N to produce N-M2e and N-3M2e chimeric recombinant nanorings. Mice were immunized intranasally with N-M2e or N-3M2e or with M2e or 3M2e control peptides. N-3M2e-vaccinated mice showed the strongest mucosal and systemic antibody responses. These mice presented a reduced viral load and minor weight loss, and all survived upon challenge with influenza virus A/PR8/34 (H1N1) (PR8). We compared the intranasal route to the subcutaneous route of N-3M2e immunization. Only the intranasal route induced a strong local IgA response and led to the protection of mice upon challenge. Finally, we demonstrated that the induction of anti-M2e antibodies by N-3M2e is not impaired by preexisting anti-N immunity. Overall, these results show that the N nanoring is a potent carrier for mucosal delivery of vaccinal antigens.


Subject(s)
Influenza A Virus, H1N1 Subtype/immunology , Influenza Vaccines/administration & dosage , Mucous Membrane/immunology , Nanostructures , Viral Matrix Proteins/immunology , Animals , Antibodies, Viral/biosynthesis , Electrophoresis, Polyacrylamide Gel , Enzyme-Linked Immunosorbent Assay , Influenza Vaccines/immunology , Mice , Microscopy, Electron, Transmission , Nucleocapsid , Real-Time Polymerase Chain Reaction , Viral Load
3.
PLoS One ; 7(5): e37722, 2012.
Article in English | MEDLINE | ID: mdl-22655066

ABSTRACT

BACKGROUND: The human respiratory syncytial virus (hRSV) is the leading cause of severe bronchiolitis in infants worldwide. The most severe RSV diseases occur between 2 and 6 months-of-age, so pediatric vaccination will have to be started within the first weeks after birth, when the immune system is prone to Th2 responses that may turn deleterious upon exposure to the virus. So far, the high risk to prime for immunopathological responses in infants has hampered the development of vaccine. In the present study we investigated the safety and efficacy of ring-nanostructures formed by the recombinant nucleoprotein N of hRSV (N(SRS)) as a mucosal vaccine candidate against RSV in BALB/c neonates, which are highly sensitive to immunopathological Th2 imprinting. METHODOLOGY AND PRINCIPAL FINDINGS: A single intranasal administration of N(SRS) with detoxified E. coli enterotoxin LT(R192G) to 5-7 day old neonates provided a significant reduction of the viral load after an RSV challenge at five weeks of age. However, neonatal vaccination also generated an enhanced lung infiltration by neutrophils and eosinophils following the RSV challenge. Analysis of antibody subclasses and cytokines produced after an RSV challenge or a boost administration of the vaccine suggested that neonatal vaccination induced a Th2 biased local immune memory. This Th2 bias and the eosinophilic reaction could be prevented by adding CpG to the vaccine formulation, which, however did not prevent pulmonary inflammation and neutrophil infiltration upon viral challenge. CONCLUSIONS/SIGNIFICANCE: In conclusion, protective vaccination against RSV can be achieved in neonates but requires an appropriate combination of adjuvants to prevent harmful Th2 imprinting.


Subject(s)
Adjuvants, Immunologic/therapeutic use , Nanostructures/therapeutic use , Nucleoproteins/therapeutic use , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Vaccines/therapeutic use , Respiratory Syncytial Viruses/immunology , Viral Proteins/therapeutic use , Adjuvants, Immunologic/administration & dosage , Administration, Intranasal , Animals , CpG Islands , Humans , Infant, Newborn , Lung/immunology , Lung/pathology , Mice , Mice, Inbred BALB C , Nanostructures/administration & dosage , Nanostructures/chemistry , Nucleoproteins/administration & dosage , Nucleoproteins/chemistry , Nucleoproteins/immunology , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/pathology , Respiratory Syncytial Virus Vaccines/administration & dosage , Respiratory Syncytial Virus Vaccines/chemistry , Respiratory Syncytial Virus Vaccines/immunology , Th2 Cells/immunology , Th2 Cells/pathology , Viral Proteins/administration & dosage , Viral Proteins/chemistry , Viral Proteins/immunology
4.
Eur J Immunol ; 41(10): 2852-61, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21770043

ABSTRACT

The high incidence of lung-damaging life-threatening respiratory infections in infants may be related to the immaturity of their immune systems. To determine whether lung immune features differ in early life compared with those in adulthood, whole lung as well as lung T lymphocyte and DC responses were investigated in BALB/c neonates versus adults. Higher expression of GATA-3 and rapid and sustained production of type 2 cytokines by lung explants after in vitro exposure to anti-CD3 was the hallmark of the neonatal period, suggestive of a Th2 bias. Neonatal lung GATA-3-producing cells were identified as CD3(+), CD4 and CD8 double-negative T lymphocytes, a subset found at a higher frequency in neonatal than adult lung. The neonatal lungs contained fewer conventional DCs, with a lower ratio of CD103(+) to CD11b(+) DCs, and a much lower number of plasmacytoid DCs in comparison with adult lungs. Yet, when stimulated in vivo by BCG, neonatal lung DCs matured and primed adult naïve CD4(+) T cells toward Th1 as efficiently as adult BCG-primed lung DCs. Conversely, both adult and neonatal BCG-primed lung DCs induced a Th2 cytokine response from neonatal naïve lymph node T cells, suggestive of an intrinsic feature of neonatal T lymphocytes.


Subject(s)
Cytokines/biosynthesis , Dendritic Cells/immunology , Lung/immunology , Th2 Cells/immunology , Transcription Factors/biosynthesis , Animals , Animals, Newborn , Antigens, CD/analysis , CD11b Antigen/analysis , CD3 Complex/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cytokines/immunology , Dendritic Cells/metabolism , GATA3 Transcription Factor/biosynthesis , Integrin alpha Chains/analysis , Lung/metabolism , Lymphocyte Activation , Lymphocyte Count , Mice , Mice, Inbred BALB C , Mycobacterium bovis/immunology , Th2 Cells/metabolism
5.
Vaccine ; 28(21): 3722-34, 2010 May 07.
Article in English | MEDLINE | ID: mdl-20307593

ABSTRACT

Human and bovine respiratory syncytial viruses (HRSV and BRSV) are two closely related, worldwide prevalent viruses that are the leading cause of severe airway disease in children and calves, respectively. Efficacy of commercial bovine vaccines needs improvement and no human vaccine is licensed yet. We reported that nasal vaccination with the HRSV nucleoprotein produced as recombinant ring-shaped nanoparticles (N(SRS)) protects mice against a viral challenge with HRSV. The aim of this work was to evaluate this new vaccine that uses a conserved viral antigen, in calves, natural hosts for BRSV. Calves, free of colostral or natural anti-BRSV antibodies, were vaccinated with N(SRS) either intramuscularly, or both intramuscularly and intranasally using Montanide ISA71 and IMS4132 as adjuvants and challenged with BRSV. All vaccinated calves developed anti-N antibodies in blood and nasal secretions and N-specific cellular immunity in local lymph nodes. Clinical monitoring post-challenge demonstrated moderate respiratory pathology with local lung tissue consolidations for the non-vaccinated calves that were significantly reduced in the vaccinated calves. Vaccinated calves had lower viral loads than the non-vaccinated control calves. Thus N(SRS) vaccination in calves provided cross-protective immunity against BRSV infection without adverse inflammatory reaction.


Subject(s)
Cattle Diseases/prevention & control , Nucleoproteins/immunology , Respiratory Syncytial Virus Infections/veterinary , Respiratory Syncytial Virus Vaccines/immunology , Viral Proteins/immunology , Adjuvants, Immunologic/pharmacology , Amino Acid Sequence , Animals , Antibodies, Viral/blood , Antibody Formation , Cattle , Cattle Diseases/immunology , Cross Protection , Immunity, Cellular , Lung/immunology , Lung/pathology , Lung/virology , Male , Molecular Sequence Data , Nanoparticles , Recombinant Proteins/immunology , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus, Bovine/immunology , Vaccines, Subunit/immunology , Viral Load
6.
PLoS One ; 3(3): e1766, 2008 Mar 12.
Article in English | MEDLINE | ID: mdl-18335041

ABSTRACT

BACKGROUND: Bronchiolitis caused by the respiratory syncytial virus (RSV) in infants less than two years old is a growing public health concern worldwide, and there is currently no safe and effective vaccine. A major component of RSV nucleocapsid, the nucleoprotein (N), has been so far poorly explored as a potential vaccine antigen, even though it is a target of protective anti-viral T cell responses and is remarkably conserved between human RSV A and B serotypes. We recently reported a method to produce recombinant N assembling in homogenous rings composed of 10-11 N subunits enclosing a bacterial RNA. These nanoparticles were named sub-nucleocapsid ring structure (N SRS). METHODOLOGY AND PRINCIPAL FINDINGS: The vaccine potential of N SRS was evaluated in a well-characterized and widely acknowledged mouse model of RSV infection. BALB/c adult mice were immunized intranasally with N SRS adjuvanted with the detoxified E. coli enterotoxin LT(R192G). Upon RSV challenge, vaccinated mice were largely protected against virus replication in the lungs, with a mild inflammatory lymphocytic and neutrophilic reaction in their airways. Mucosal immunization with N SRS elicited strong local and systemic immunity characterized by high titers of IgG1, IgG2a and IgA anti-N antibodies, antigen-specific CD8(+) T cells and IFN-gamma-producing CD4(+) T cells. CONCLUSIONS/SIGNIFICANCE: This is the first report of using nanoparticles formed by the recombinant nucleocapsid protein as an efficient and safe intra-nasal vaccine against RSV.


Subject(s)
Nanoparticles , Respiratory Syncytial Viruses/immunology , Viral Vaccines/administration & dosage , Administration, Intranasal , Animals , Base Sequence , Cell Line , DNA Primers , Flow Cytometry , Mice , Mice, Inbred BALB C , Microscopy, Confocal , Microscopy, Electron , Mutagenesis, Site-Directed , Viral Vaccines/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...