Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
J Transl Med ; 17(1): 242, 2019 07 25.
Article in English | MEDLINE | ID: mdl-31345237

ABSTRACT

BACKGROUND: Current human influenza vaccines lack the adaptability to match the mutational rate of the virus and therefore require annual revisions. Because of extensive manufacturing times and the possibility that antigenic alterations occur during viral vaccine strain production, an inherent risk exists for antigenic mismatch between the new influenza vaccine and circulating viruses. Targeting more conserved antigens such as nucleoprotein (NP) could provide a more sustainable vaccination strategy by inducing long term and heterosubtypic protection against influenza. We previously demonstrated that intranodal mRNA injection can induce potent antigen-specific T-cell responses. In this study, we investigated whether intranodal administration of mRNA encoding NP can induce T-cell responses capable of protecting against a heterologous influenza virus challenge. METHODS: BALB/c mice were immunized in the inguinal lymph nodes with different vaccination regimens of mRNA encoding NP. Immune responses were compared with NP DNA vaccination via IFN-γ ELISPOT and in vivo cytotoxicity. For survival experiments, mice were prime-boost vaccinated with 17 µg NP mRNA and infected with 1LD50 of H1N1 influenza virus 8 weeks after boost. Weight was monitored and viral titers, cytokines and immune cell populations in the bronchoalveolar lavage, and IFN-γ responses in the spleen were analyzed. RESULTS: Our results demonstrate that NP mRNA induces superior systemic T-cell responses against NP compared to classical DNA vaccination. These responses were sustained for several weeks even at low vaccine doses. Upon challenge infection, vaccination with NP mRNA resulted in reduced lung viral titers and improved recovery from infection. Finally, we show that vaccination with NP mRNA affects the immune response in infected lungs by lowering immune cell infiltration while increasing the fraction of T cells, monocytes and MHC II+ alveolar macrophages within immune infiltrates. This change was associated with altered levels of both pro- and anti-inflammatory cytokines. CONCLUSIONS: These findings suggest that intranodal vaccination with NP mRNA induces cross-strain immunity against influenza, but also highlight a paradox of influenza immunity, whereby robust immune responses can provide protection, but can also transiently exacerbate symptoms during infection.


Subject(s)
Influenza Vaccines/immunology , Nucleoproteins/administration & dosage , Orthomyxoviridae Infections/prevention & control , RNA, Messenger/administration & dosage , Animals , Antibodies, Viral/immunology , Antigens/chemistry , Bronchoalveolar Lavage , Dogs , Female , Humans , Influenza A Virus, H3N2 Subtype , Interferon-gamma/immunology , Interferon-gamma/metabolism , Madin Darby Canine Kidney Cells , Mice , Mice, Inbred BALB C , Plasmids , T-Lymphocytes/cytology
2.
Front Immunol ; 9: 2875, 2018.
Article in English | MEDLINE | ID: mdl-30581437

ABSTRACT

The Human Respiratory Syncytial Virus (hRSV) and the Human Metapneumovirus (hMPV) are two pneumoviruses that are leading agents causing acute lower respiratory tract infections (ALRTIs) affecting young infants, the elderly, and immunocompromised patients worldwide. Since these pathogens were first discovered, many approaches for the licensing of safe and effective vaccines have been explored being unsuccessful to date. We have previously described that immunization with recombinant strains of Mycobacterium bovis Bacillus Calmette-Guérin (rBCG) expressing the hRSV nucleoprotein (rBCG-N) or the hMPV phosphoprotein (rBCG-P) induced immune protection against each respective virus. These vaccines efficiently promoted viral clearance without significant lung damage, mainly through the induction of a T helper 1 cellular immunity. Here we show that upon viral challenge, rBCG-immunized mice developed a protective humoral immunity, characterized by production of antibodies specific for most hRSV and hMPV proteins. Further, isotype switching from IgG1 to IgG2a was observed in mice immunized with rBCG vaccines and correlated with an increased viral clearance, as compared to unimmunized animals. Finally, sera obtained from animals immunized with rBCG vaccines and infected with their respective viruses exhibited virus neutralizing capacity and protected naïve mice from viral replication and pulmonary disease. These results support the notion that the use of rBCG strains could be considered as an effective vaccination approach against other respiratory viruses with similar biology as hRSV and hMPV.


Subject(s)
BCG Vaccine/immunology , Immunity, Humoral , Mycobacterium bovis/immunology , Respiratory Tract Infections/prevention & control , Animals , BCG Vaccine/administration & dosage , Cell Line, Tumor , Disease Models, Animal , Humans , Metapneumovirus/genetics , Metapneumovirus/immunology , Mice , Mice, Inbred BALB C , Nucleoproteins/administration & dosage , Nucleoproteins/genetics , Nucleoproteins/immunology , Recombinant Proteins/administration & dosage , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Respiratory Syncytial Virus, Human/genetics , Respiratory Syncytial Virus, Human/immunology , Respiratory Tract Infections/immunology , Respiratory Tract Infections/virology , Th1 Cells/immunology , Th1 Cells/metabolism , Vaccination/methods , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/immunology , Viral Proteins/administration & dosage , Viral Proteins/genetics , Viral Proteins/immunology , Virus Replication/immunology
3.
Viral Immunol ; 31(4): 306-314, 2018 05.
Article in English | MEDLINE | ID: mdl-29373084

ABSTRACT

The human metapneumovirus (hMPV) is the second leading cause globally of acute infection of the respiratory tract in children, infecting the upper and lower airways. The hMPV may induce an inappropriate Th2-type immune response, which causes severe pulmonary inflammation, leading to the obstruction of airways. Despite its severe epidemiological relevance, no vaccines are currently available for the prevention of hMPV-induced illness. In this investigation, we demonstrated that immunization of mice with the recombinant hMPV nucleoprotein (hMPV-N) mixed with the AbISCO-100 adjuvant reduced viral replication in lungs following challenge with the virus. We found that immunized mice had reduced weight loss, decreased granulocytes in the lung, an increased level of specific nucleoprotein antibodies of IgG1 and IgG2a-isotypes, and a local profile of Th1/Th17-type cytokines. Our results suggest that immunization with the hMPV-N and the AbISCO-100 adjuvant induces a reduction of viral infection and could be considered for the development of an hMPV vaccine.


Subject(s)
Immunization , Metapneumovirus/immunology , Nucleoproteins/administration & dosage , Paramyxoviridae Infections/immunology , Viral Vaccines/immunology , Adjuvants, Immunologic/pharmacology , Animals , Antibodies, Viral/blood , Antibodies, Viral/classification , Cytokines/analysis , Dendritic Cells/classification , Disease Models, Animal , Gene Expression/drug effects , Granulocytes , Humans , Lung/drug effects , Lung/pathology , Lung/virology , Mice , Mice, Inbred BALB C , Nucleoproteins/genetics , Nucleoproteins/isolation & purification , Paramyxoviridae Infections/prevention & control , Pneumonia/virology , RNA, Viral/analysis , Viral Vaccines/pharmacology , Weight Loss
4.
Virology ; 500: 209-217, 2017 01.
Article in English | MEDLINE | ID: mdl-27829176

ABSTRACT

This study sought to improve an existing live attenuated influenza vaccine (LAIV) by including nucleoprotein (NP) from wild-type virus rather than master donor virus (MDV). H7N9 LAIV reassortants with 6:2 (NP from MDV) and 5:3 (NP from wild-type virus) genome compositions were compared with regard to their growth characteristics, induction of humoral and cellular immune responses in mice, and ability to protect mice against homologous and heterologous challenge viruses. Although, in general, the 6:2 reassortant induced greater cell-mediated immunity in C57BL6 mice than the 5:3 vaccine, mice immunized with the 5:3 LAIV were better protected against heterologous challenge. The 5:3 LAIV-induced CTLs also had better in vivo killing activity against target cells loaded with the NP366 epitope of recent influenza viruses. Modification of the genome of reassortant vaccine viruses by incorporating the NP gene from wild-type viruses represents a simple strategy to improve the immunogenicity and cross-protection of influenza vaccines.


Subject(s)
Influenza A Virus, H7N9 Subtype/immunology , Influenza A Virus, H7N9 Subtype/pathogenicity , Influenza Vaccines/immunology , Influenza, Human/immunology , Nucleoproteins/immunology , Vaccines, Attenuated/immunology , Animals , Antibodies, Viral/immunology , Cold Temperature , Cross Protection , Female , Humans , Immunity, Cellular , Influenza A Virus, H7N9 Subtype/genetics , Influenza A Virus, H7N9 Subtype/physiology , Influenza Vaccines/administration & dosage , Influenza, Human/prevention & control , Influenza, Human/virology , Mice , Mice, Inbred C57BL , Nucleoproteins/administration & dosage , Nucleoproteins/genetics , Reassortant Viruses/immunology , Reassortant Viruses/pathogenicity , Reassortant Viruses/physiology , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/genetics , Virulence
5.
J Control Release ; 237: 35-41, 2016 09 10.
Article in English | MEDLINE | ID: mdl-27381247

ABSTRACT

DNA vaccines have many advantages such as thermostability and the ease and rapidity of manufacture; for example, in an influenza pandemic situation where rapid production of vaccine is essential. However, immunogenicity of DNA vaccines was shown to be poor in humans unless large doses of DNA are used. If a highly efficacious DNA vaccine delivery system could be identified, then DNA vaccines have the potential to displace protein vaccines. In this study, we show in a C57BL/6 mouse model, that the Nanopatch, a microprojection array of high density (>21,000 projections/cm(2)), could be used to deliver influenza nucleoprotein DNA vaccine to skin, to generate enhanced antigen specific antibody and CD8(+) T cell responses compared to the conventional intramuscular (IM) delivery by the needle and syringe. Antigen specific antibody was measured using ELISA assays of mice vaccinated with a DNA plasmid containing the nucleoprotein gene of influenza type A/WSN/33 (H1N1). Antigen specific CD8(+) T cell responses were measured ex-vivo in splenocytes of mice using IFN-γ ELISPOT assays. These results and our previous antibody and CD4(+) T cell results using the Nanopatch delivered HSV DNA vaccine indicate that the Nanopatch is an effective delivery system of general utility that could potentially be used in humans to increase the potency of the DNA vaccines.


Subject(s)
Influenza A Virus, H1N1 Subtype/immunology , Influenza Vaccines/administration & dosage , Influenza, Human/prevention & control , Nucleoproteins/administration & dosage , Orthomyxoviridae Infections/prevention & control , Vaccination/instrumentation , Vaccines, DNA/administration & dosage , Administration, Cutaneous , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Equipment Design , Female , Humans , Influenza Vaccines/immunology , Influenza, Human/immunology , Mice, Inbred C57BL , Needles , Nucleoproteins/immunology , Orthomyxoviridae Infections/immunology , Vaccines, DNA/immunology
6.
J Mol Neurosci ; 55(3): 803-11, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25303858

ABSTRACT

Parkinson's disease (PD) is an obstinate progressive neurodegenerative disease and characterized by locomotor impairment and dopaminergic neuronal degeneration in the substantia nigra pars compacta (SNc). We examined in here the dietary effect of nucleoprotein (NP) extracted from salmon soft roe on 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-injected PD-like mice model to prevent the symptom as an alternative medicine. Male C57/BL6 mice were given either an artificially modified NP-free diet (NF) or NF supplied with 1.2% NP for 1 week. Then, mice were injected intraperitoneally four times with 20 mg/kg MPTP. Seven days later, locomotor activity was examined, and the brains were immunostained with tyrosine hydroxylase (TH) and Iba1 antibodies. Moreover, in situ detection of superoxide anion (O2(-)) and gene expression of mitochondrial electron transfer chain gene, Cox8b was evaluated in midbrains. NP-fed animals showed significantly reduced locomotor impairment and an increased number of TH-positive cells in the SNc compared with NF animals. The NP-fed animals also showed reduced lower levels of O2(-) and up-regulation of Cox8b levels and Iba1 immunoreactivity, suggesting that inflammation and oxidative stress were suppressed and mitochondrial impairment was relieved in these animals. Supplementation of the diet with NP may serve as a useful preventive measure to slow the onset of PD.


Subject(s)
Dietary Supplements , Dopaminergic Neurons/drug effects , MPTP Poisoning/drug therapy , Nucleoproteins/therapeutic use , Animals , Dopaminergic Neurons/metabolism , Electron Transport Complex IV/genetics , Electron Transport Complex IV/metabolism , Locomotion , MPTP Poisoning/prevention & control , Male , Mesencephalon/cytology , Mesencephalon/drug effects , Mesencephalon/metabolism , Mice , Mice, Inbred C57BL , Nucleoproteins/administration & dosage , Nucleoproteins/pharmacology , Superoxides/metabolism , Tyrosine 3-Monooxygenase/genetics , Tyrosine 3-Monooxygenase/metabolism
7.
PLoS One ; 9(5): e97270, 2014.
Article in English | MEDLINE | ID: mdl-24824623

ABSTRACT

Recent reports highlight the potential for integrase-defective lentiviral vectors (IDLV) to be developed as vaccines due to their ability to elicit cell-mediated and humoral immune responses after intramuscular administration. Differently from their integrase-competent counterpart, whose utility for vaccine development is limited by the potential for insertional mutagenesis, IDLV possess a mutation in their integrase gene that prevents genomic integration. Instead, they are maintained as episomal DNA circles that retain the ability to stably express functional proteins. Despite their favorable profile, it is unknown whether IDLV elicit immune responses after intranasal administration, a route that could be advantageous in the case of infection with a respiratory agent. Using influenza as a model, we constructed IDLV expressing the influenza virus nucleoprotein (IDLV-NP), and tested their ability to generate NP-specific immune responses and protect from challenge in vivo. We found that administration of IDLV-NP elicited NP-specific T cell and antibody responses in BALB/c mice. Importantly, IDLV-NP was protective against homologous and heterosubtypic influenza virus challenge only when given by the intranasal route. This is the first report demonstrating that IDLV can induce protective immunity after intranasal administration, and suggests that IDLV may represent a promising vaccine platform against infectious agents.


Subject(s)
Genetic Vectors/pharmacology , Influenza A virus , Integrases/metabolism , Nucleoproteins/pharmacology , Orthomyxoviridae Infections/prevention & control , Administration, Intranasal , Analysis of Variance , Animals , Blotting, Western , Dogs , Enzyme-Linked Immunosorbent Assay , Enzyme-Linked Immunospot Assay , Flow Cytometry , Genetic Vectors/administration & dosage , HEK293 Cells , Humans , Immunity, Mucosal , Lentivirus , Madin Darby Canine Kidney Cells , Mice , Mice, Inbred BALB C , Nucleoproteins/administration & dosage , Plasmids/genetics
8.
PLoS One ; 7(9): e46166, 2012.
Article in English | MEDLINE | ID: mdl-23029425

ABSTRACT

Influenza primed mice are protected against lethal infection with H1N1 A/CA/04/E3/09 virus, and T depletion and serum transfer studies suggest a T-dependent mechanism. We therefore set out to investigate the quality of the cross-reactive T cell response to CA/E3/09 in mice primed with H3N2 influenza A/Hong Kong/X31 virus. Sequences of the immunodominant nucleoprotein (NP) NP366-374 and acid polymerase (PA) PA224-233 CD8 epitopes from X31 each differ from the CA/E3/09 virus by one amino acid: an M371V substitution at position 6 of the NP peptide, and an S224P substitution at position 1 of the PA peptide, raising questions about the role of these epitopes in protection. PA224-233 peptides from either virus could elicit IFN-γ spot forming cells from mice infected with X31, indicating cross-reactivity of these two peptides. However, no T cell responses to either PA224-233 peptide were detectable after primary CA/E3/09 infection, suggesting it is cryptic in this virus. In contrast, primary responses to the NP366 peptides were detectable after infection with either virus, but did not cross-react in vitro. Similarly, H2-D(b) tetramers of each NP epitope stained CD8+ T cells from each respective virus infection, but did not obviously cross-react. Early after lethal CA/E3/09 challenge, X31 primed mice had enhanced IFN-γ responses toward both NP366 peptides, as well as recall responses to a set of subdominant NP and PA peptides not detectable after primary X31 infection alone. Furthermore, dual-tetramer staining revealed an expanded population of CD8 T cells reactive to both NP366 variant peptides also not seen after the priming infection alone. These observations demonstrate unusual CD8+ T cell cross-reactivity and specificity are elicited after primary and secondary CA/E3/09 influenza virus infections.


Subject(s)
Antigens, Viral/immunology , CD8-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/immunology , Orthomyxoviridae Infections/prevention & control , T-Cell Antigen Receptor Specificity , Amino Acid Substitution , Animals , Antigens, Viral/administration & dosage , Antigens, Viral/genetics , CD8-Positive T-Lymphocytes/cytology , Cross Protection , Cross Reactions , DNA-Directed DNA Polymerase/genetics , DNA-Directed DNA Polymerase/immunology , Epitopes, T-Lymphocyte/genetics , Humans , Immunologic Memory , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H3N2 Subtype/genetics , Influenza A Virus, H3N2 Subtype/immunology , Mice , Nucleoproteins/administration & dosage , Nucleoproteins/genetics , Nucleoproteins/immunology , Oligopeptides/administration & dosage , Oligopeptides/genetics , Oligopeptides/immunology , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology
9.
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
10.
J Microbiol Biotechnol ; 22(3): 416-21, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22450799

ABSTRACT

Current influenza vaccines elicit antibodies effective against homologous strains, but new strategies are urgently needed for protection against emerging epidemic or pandemic strains. Although influenza vaccine candidates based on the viral nucleoprotein (NP) or matrix protein do not elicit sterilizing immunity, they have the advantage of inducing immunity that may cover a larger number of viral strains. In this study, recombinant NP produced in Escherichia coli was purified and formulated in combination with the adjuvant ISCOMATRIX. This formulation increased a NP-specific immunity in mice, with a Th1 profile, and may constitute a promising low-cost influenza vaccine candidate, with ability to stimulate humoral and cellular immune responses..


Subject(s)
Cholesterol/immunology , Influenza A Virus, H1N1 Subtype/immunology , Influenza Vaccines/immunology , Influenza, Human/prevention & control , Nucleoproteins/immunology , Phospholipids/immunology , Saponins/immunology , Viral Proteins/immunology , Adjuvants, Immunologic/administration & dosage , Animals , Antibodies, Viral , Cholesterol/administration & dosage , Drug Combinations , Female , Humans , Immunization , Influenza A Virus, H1N1 Subtype/genetics , Influenza Vaccines/administration & dosage , Influenza Vaccines/genetics , Influenza, Human/immunology , Influenza, Human/virology , Mice , Mice, Inbred BALB C , Nucleoproteins/administration & dosage , Nucleoproteins/genetics , Phospholipids/administration & dosage , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Saponins/administration & dosage , Viral Proteins/administration & dosage , Viral Proteins/genetics
11.
Mem Inst Oswaldo Cruz ; 105(7): 879-88, 2010 Nov.
Article in English | MEDLINE | ID: mdl-21120357

ABSTRACT

Despite effective chemotherapy, schistosomiasis remains the second largest public health problem in the developing world. Currently, vaccination is the new strategy for schistosomiasis control. The presence of common antigenic fractions between Schistosoma mansoni and its intermediate host provides a source for the preparation of a proper vaccine. The objective of this paper is to evaluate the nucleoprotein extracted from either susceptible or resistant snails to protect against schistosomiasis. The vaccination schedule consisted of a subcutaneous injection of 50 µg protein of each antigen followed by another inoculation 15 days later. Analyses of marker enzymes for different cell organelles [succinate dehydrogenase, lactate dehydrogenase (LDH), glucose-6-phosphatase, acid phosphatase and 5'-nucleotidase] were carried out. Energetic parameters (ATP, ADP, AMP, phosphate potentials, inorganic phosphate, amino acids and LDH isoenzymes) were also investigated. The work was extended to record worm and ova counts, oogram determination in the liver and intestine and the histopathological pattern of the liver. The nucleoprotein of susceptible snails showed reduction in worm and ova counts by 70.96% and 51.31%, respectively, whereas the nucleoprotein of resistant snails showed reductions of 9.67% and 16.77%, respectively. In conclusion, we found that the nucleoprotein of susceptible snails was more effective in protecting against schistosomiasis.


Subject(s)
Amino Acids/analysis , Biomphalaria/parasitology , Liver/parasitology , Nucleoproteins/immunology , Schistosoma mansoni/immunology , Schistosomiasis mansoni/immunology , Animals , Biomphalaria/immunology , Host-Parasite Interactions , Liver/chemistry , Liver/enzymology , Liver/pathology , Male , Mice , Nucleoproteins/administration & dosage , Parasite Egg Count , Schistosomiasis mansoni/parasitology
12.
Respir Res ; 8: 44, 2007 Jun 27.
Article in English | MEDLINE | ID: mdl-17597533

ABSTRACT

BACKGROUND: MHC class I-restricted CD8+ cytotoxic T lymphocytes (CTL) are thought to play a major role in clearing virus and promoting recovery from influenza infection and disease. This has been demonstrated for clearance of influenza virus from the lungs of infected mice. However, human influenza infection is primarily a respiratory mucosal infection involving the nasopharynx and tracheobronchial tree. The role of CD8+ CTL directed toward the influenza nucleoprotein (NP) in defense against influenza virus infection at the respiratory mucosa was evaluated in two separate adoptive transfer experiments. METHODS: Influenza nucleoprotein (NP)-specific CD8+ CTL were generated from splenocytes obtained from Balb/c mice previously primed with influenza A/Taiwan/1/86 (H1N1) infection or with influenza A/PR/8/34 (H1N1)-derived NP plasmid DNA vaccine followed by infection with A/Hong Kong/68 (H3N2) virus. After in vitro expansion by exposure to an influenza NP-vaccinia recombinant, highly purified CD8+ T cells exhibited significant lysis in vitro of P815 target cells infected with A/Hong Kong/68 (H3N2) virus while the CD8- fraction (CD4+ T cells, B cells and macrophages) had no CTL activity. Purified CD8+ and CD8- T cells (1 x 107) were injected intravenously or interperitoneally into naive mice four hours prior to intranasal challenge with A/HK/68 (H3N2) virus. RESULTS: The adoptively transferred NP-vaccinia-induced CD8+ T cells caused significant reduction of virus titers in both the lungs and nasal passages when compared to CD8- cells. Neither CD8+ nor CD8- T cells from cultures stimulated with HIV gp120-vaccinia recombinant reduced virus titers. CONCLUSION: The present data demonstrate that influenza NP-specific CD8+ CTL can play a direct role in clearance of influenza virus from the upper respiratory mucosal surfaces.


Subject(s)
Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H3N2 Subtype/immunology , Influenza, Human/immunology , Nucleoproteins/administration & dosage , Respiratory Tract Infections/immunology , T-Lymphocyte Subsets , T-Lymphocytes, Cytotoxic , Viral Proteins/administration & dosage , Animals , Disease Models, Animal , Humans , Injections, Intraperitoneal , Injections, Intravenous , Mice , Mice, Inbred BALB C , Respiratory Mucosa/immunology , Treatment Outcome
13.
J Biol Chem ; 281(52): 39925-34, 2006 Dec 29.
Article in English | MEDLINE | ID: mdl-17088258

ABSTRACT

CD8(+) T lymphocytes recognize infected cells that display virus-derived antigenic peptides complexed with major histocompatibility complex class I molecules. Peptides are mainly byproducts of cellular protein turnover by cytosolic proteasomes. Cytosolic tripeptidyl-peptidase II (TPPII) also participates in protein degradation. Several peptidic epitopes unexpectedly do not require proteasomes, but it is unclear which proteases generate them. We studied antigen processing of influenza virus nucleoprotein epitope NP(147-155), an archetype epitope that is even destroyed by a proteasome-mediated mechanism. TPPII, with the assistance of endoplasmic reticulum trimming metallo-aminopeptidases, probably ERAAP (endoplasmic reticulum aminopeptidase associated with antigen processing), was crucial for nucleoprotein epitope generation both in the presence of functional proteasomes and when blocked by lactacystin, as shown with specific chemical inhibitors and gene silencing. Different protein contexts and subcellular targeting all allowed epitope processing by TPPII as well as trimming. The results show the plasticity of the cell's assortment of proteases for providing ligands for recognition by antiviral CD8(+) T cells. Our observations identify for the first time a set of proteases competent for antigen processing of an epitope that is susceptible to destruction by proteasomes.


Subject(s)
Antigen Presentation/immunology , Antigens, Viral/metabolism , Histocompatibility Antigens Class I/metabolism , Proteasome Endopeptidase Complex/physiology , Serine Endopeptidases/physiology , Acetylcysteine/analogs & derivatives , Acetylcysteine/pharmacology , Amino Acid Sequence , Aminopeptidases , Animals , Antigens, Viral/genetics , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases , Epitopes, T-Lymphocyte/metabolism , Hydrolysis , Influenza A virus/enzymology , Influenza A virus/immunology , Influenza A virus/metabolism , L Cells , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Nucleocapsid Proteins , Nucleoproteins/administration & dosage , Nucleoproteins/immunology , Nucleoproteins/metabolism , Proteasome Inhibitors , RNA-Binding Proteins/administration & dosage , RNA-Binding Proteins/immunology , RNA-Binding Proteins/metabolism , Serine Proteinase Inhibitors/pharmacology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , Vaccinia virus/enzymology , Vaccinia virus/immunology , Viral Core Proteins/administration & dosage , Viral Core Proteins/immunology , Viral Core Proteins/metabolism
14.
J Immunol ; 175(6): 3882-91, 2005 Sep 15.
Article in English | MEDLINE | ID: mdl-16148134

ABSTRACT

Improving DNA vaccination remains a fundamental goal in vaccine research. Theoretically, this could be achieved by molecules encoded by DNA capable of activating TLRs to mimic inflammatory responses generated by infection. Therefore, we constructed an expression vector that allows mammalian cells to express the TLR5 agonist flagellin (FliC) at the cell surface. In vitro, cell lines expressing FliC stimulated production of proinflammatory cytokines and the up-regulation of costimulatory molecules on monocytes. Mice given the FliC expression vector intradermally exhibited site-specific inflammation and, in combination with vectors expressing Ags, developed dramatic increases in Ag-specific IgG as well as IgA. Surprisingly, mice also developed strong Ag-specific MHC class I-restricted cellular immunity. To determine whether vaccination using FliC vectors could elicit protective immunity to an infectious agent, mice were given dermal injections of FliC expression vector together with a vector encoding the influenza A virus nucleoprotein. This vaccination strategy elicited protective immunity to lethal influenza A virus infection. These results demonstrate that expression of DNA-encoded TLR agonists by mammalian cells greatly enhance and broaden immune responses, imposing new possibilities on DNA vaccination to infectious agents and cancer.


Subject(s)
Flagellin/pharmacology , Immunity, Innate/drug effects , Inflammation/chemically induced , Vaccines, DNA/genetics , Animals , Antigens, Viral/administration & dosage , Antigens, Viral/genetics , Cell Line , Flagellin/administration & dosage , Flagellin/genetics , Genetic Vectors , Humans , Immunity, Cellular/drug effects , Immunoglobulin A/biosynthesis , Immunoglobulin G/biosynthesis , Influenza A virus/immunology , Influenza, Human/prevention & control , Influenza, Human/therapy , Mice , Mice, Inbred C57BL , Nucleocapsid Proteins , Nucleoproteins/administration & dosage , Nucleoproteins/genetics , RNA-Binding Proteins/administration & dosage , RNA-Binding Proteins/genetics , Vaccines, DNA/administration & dosage , Vaccines, DNA/pharmacology , Viral Core Proteins/administration & dosage , Viral Core Proteins/genetics
15.
J Immunol ; 175(2): 1153-60, 2005 Jul 15.
Article in English | MEDLINE | ID: mdl-16002717

ABSTRACT

During viral infection, constitutive proteasomes are largely replaced by immunoproteasomes, which display distinct cleavage specificities, resulting in different populations of potential CD8(+) T cell epitope peptides. Immunoproteasomes are believed to be important for the generation of many viral CD8(+) T cell epitopes and have been implicated in shaping the immunodominance hierarchies of CD8(+) T cell responses to influenza virus infection. However, it remains unclear whether these conclusions are generally applicable. In this study we investigated the CD8(+) T cell responses to lymphocytic choriomeningitis virus infection and DNA immunization in wild-type mice and in mice lacking the immunoproteasome subunits LMP2 or LMP7. Although the total number of virus-specific cells was lower in LMP2 knockout mice, consistent with their having lower numbers of naive cells before infection, the kinetics of virus clearance were similar in all three mouse strains, and LMP-deficient mice mounted strong primary and secondary lymphocytic choriomeningitis virus-specific CD8(+) T cell responses. Furthermore, the immunodominance hierarchy of the four investigated epitopes (nuclear protein 396 (NP(396)) > gp33 > gp276 > NP(205)) was well maintained. We observed a slight reduction in the NP(205)-specific response in LMP2-deficient mice, but this had no demonstrable biological consequence. DNA vaccination of LMP2- and LMP7-deficient mice induced CD8(+) T cell responses that were slightly lower than, although not significantly different from, those induced in wild-type mice. Taken together, our results challenge the notion that immunoproteasomes are generally needed for effective antiviral CD8(+) T cell responses and for the shaping of immunodominance hierarchies. We conclude that the immunoproteasome may affect T cell responses to only a limited number of viral epitopes, and we propose that its main biological function may lie elsewhere.


Subject(s)
CD8-Positive T-Lymphocytes/enzymology , CD8-Positive T-Lymphocytes/immunology , Cysteine Endopeptidases/deficiency , Cysteine Endopeptidases/genetics , Lymphocytic Choriomeningitis/immunology , Multienzyme Complexes/deficiency , Multienzyme Complexes/genetics , Vaccines, DNA/immunology , Amino Acid Sequence , Animals , CD8-Positive T-Lymphocytes/virology , Cysteine Endopeptidases/physiology , Dose-Response Relationship, Immunologic , Histocompatibility Antigens Class I/biosynthesis , Immunodominant Epitopes/administration & dosage , Immunodominant Epitopes/biosynthesis , Immunodominant Epitopes/immunology , Lymphocyte Count , Lymphocytic Choriomeningitis/enzymology , Lymphocytic Choriomeningitis/genetics , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Multienzyme Complexes/physiology , Nucleoproteins/administration & dosage , Nucleoproteins/immunology , Peptide Fragments/administration & dosage , Peptide Fragments/immunology , Proteasome Endopeptidase Complex , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/enzymology , T-Lymphocyte Subsets/virology , Vaccines, DNA/administration & dosage , Viral Load
16.
J Immunol ; 173(10): 5929-33, 2004 Nov 15.
Article in English | MEDLINE | ID: mdl-15528326

ABSTRACT

The ability for the professional APC to cross-present Ag to MHC class I from parenchymal cells is essential for priming as well as tolerance of CD8+ T cells against intracellular Ags. Since cross-presentations of non-cell-associated free Ags are inefficient, the roles of molecular chaperones or heat shock proteins (HSPs) in chaperoning Ags to APCs have been postulated. We herein genetically addressed this hypothesis using mice that were defective of heat shock factor 1 (Hsf1), a major transcription factor for HSPs. Hsf1(-/-) mice have a decreased expression of several HSPs including HSP90 and HSP70. Using multiple Ag systems, we demonstrated that cross-priming of Ag-specific CD8+ T cells was inefficient when Ag expression was restricted to Hsf1(-/-) non-APCs. Our study provides the first genetic evidence for the roles of Hsf1 in regulating cross-presentation of MHC class I-associated Ags.


Subject(s)
Antigen Presentation , DNA-Binding Proteins/physiology , H-2 Antigens/metabolism , Heat-Shock Proteins/physiology , Transcription Factors/physiology , Adoptive Transfer , Animals , Antigen Presentation/genetics , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/virology , Cell Line , Cell Line, Transformed , Chickens , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Egg Proteins/immunology , Egg Proteins/metabolism , H-2 Antigens/biosynthesis , H-2 Antigens/immunology , Heat Shock Transcription Factors , Heat-Shock Proteins/biosynthesis , Heat-Shock Proteins/deficiency , Hematopoiesis/genetics , Hematopoiesis/immunology , Influenza A virus/immunology , Influenza Vaccines/administration & dosage , Influenza Vaccines/immunology , Lymphocyte Activation/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Nucleoproteins/administration & dosage , Nucleoproteins/immunology , Ovalbumin/immunology , Ovalbumin/metabolism , Peptide Fragments/administration & dosage , Peptide Fragments/immunology , Transcription Factors/deficiency , Transcription Factors/genetics
17.
J Immunol ; 171(11): 6032-8, 2003 Dec 01.
Article in English | MEDLINE | ID: mdl-14634115

ABSTRACT

Memory CD8 T cells play a critical role in protective immunity against intracellular pathogens. In addition to their ability to specifically recognize and lyse infected targets, activated CD8 T cells secrete cytokines that induce phagocytic cells to engulf and kill bacterial pathogens. In this study, we asked whether activation of Ag-specific CD8 T cells results in nonspecific killing of bystander bacteria during a mixed infection. Mice with epitope-specific memory CD8 T cells were coinfected with two isogenic strains of recombinant Listeria monocytogenes that differ in the cognate epitope. Recall responses by epitope-specific CD8 T cells rapidly inhibited the growth of epitope-bearing bacteria, impeding the course of infection within 6 h after challenge. This rapid inhibition was highly specific and did not affect the growth of coinfecting bacteria without the epitope. CTL recall did not enhance activation of innate immune cells, as evidenced by the absence of inducible NO synthase production in infectious foci. Our observations demonstrate the remarkable specificity of the bactericidal mechanisms of CTL and reveal the possibility for escape mutants to prevail in the hostile environment of a specific immune response. This implication has a bearing on subunit vaccine design strategies and understanding failure of immunization against bacterial infection.


Subject(s)
Bystander Effect/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/microbiology , Cytotoxicity, Immunologic/immunology , Epitopes, T-Lymphocyte/immunology , Listeria monocytogenes/growth & development , Listeria monocytogenes/immunology , Lymphocyte Activation/immunology , Amino Acid Sequence , Animals , CD8-Positive T-Lymphocytes/cytology , Female , Immunologic Memory , Listeria monocytogenes/genetics , Listeriosis/immunology , Listeriosis/microbiology , Listeriosis/pathology , Lymphocyte Count , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/microbiology , Lymphocytic Choriomeningitis/pathology , Lymphocytic choriomeningitis virus/genetics , Lymphocytic choriomeningitis virus/immunology , Macrophage Activation/immunology , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Nucleoproteins/administration & dosage , Nucleoproteins/genetics , Nucleoproteins/immunology , Peptide Fragments/administration & dosage , Peptide Fragments/genetics , Peptide Fragments/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology
18.
J Immunol ; 171(4): 1999-2005, 2003 Aug 15.
Article in English | MEDLINE | ID: mdl-12902504

ABSTRACT

DNA-based immunizations have been used to determine the patterns of type 1 and type 2 cytokines that can be induced in vivo for Ag-specific CD4(+) and CD8(+) T cells. IL-4 was used as a signature cytokine for a type 2 T cell response and IFN-gamma as the signature cytokine for a type 1 response. Gene gun deliveries of secreted Ags were used to bias responses toward type 2 and saline injections of cell-associated Ags to bias responses toward type 1. The studies revealed that gene gun bombardments of DNAs expressing secreted Ags strongly biased responses toward type 2, inducing IL-4-producing CD8(+) as well as CD4(+) T cells. Saline injections of DNAs expressing cell-associated Ags strongly biased responses toward type 1, inducing IFN-gamma-producing CD8(+) and CD4(+) cells. A mixed type 1/type 2 response of IFN-gamma-producing CD8(+) T cells and IL-4-producing CD4(+) T cells was found for gene gun deliveries of cell-associated Ags. Saline injections of secreted Ags raised a weakly type 1-biased response characterized by only slightly higher frequencies of IFN-gamma- than IL-4-producing CD4(+) and CD8(+) T cells. Studies in B cell knockout and hen egg lysozyme Ig transgenic mice revealed that B cells were required for the generation of IL-4-producing CD8(+) T cells.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Interferon-gamma/biosynthesis , Interleukin-4/biosynthesis , RNA-Binding Proteins , Vaccines, DNA/administration & dosage , Vaccines, DNA/immunology , Amino Acid Sequence , Animals , Antigen Presentation/genetics , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Biolistics , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cell Line , Epitopes, T-Lymphocyte/administration & dosage , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/immunology , Female , Hemagglutinin Glycoproteins, Influenza Virus/administration & dosage , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Humans , Influenza A virus/immunology , Injections, Intramuscular , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Molecular Sequence Data , Nucleocapsid Proteins , Nucleoproteins/administration & dosage , Nucleoproteins/genetics , Nucleoproteins/immunology , Ovalbumin/administration & dosage , Ovalbumin/genetics , Ovalbumin/immunology , Sodium Chloride/administration & dosage , Vaccines, DNA/genetics , Viral Core Proteins/administration & dosage , Viral Core Proteins/genetics , Viral Core Proteins/immunology
19.
J Immunol ; 170(3): 1498-503, 2003 Feb 01.
Article in English | MEDLINE | ID: mdl-12538713

ABSTRACT

Linear peptides (SynB vectors) with specific sequence motifs have been identified that are capable of enhancing the transport of a wide range of molecules into cells. These peptide vectors have been used to deliver exogenous peptides and protein Ags across the cell membrane and into the cytoplasm of cells. Specifically, in vitro analysis indicated that these SynB peptides enhanced the uptake of two 9-mer peptide Ags, NP(147-155) and Mtb(250-258) (T cell epitopes of influenza nucleoprotein and Mycobacterium tuberculosis, respectively) and the M. tuberculosis Ag Mtb8.4 protein, into K562 cells when covalently linked to the respective Ags. Furthermore, selected SynB vectors, when conjugated to these same Ags and used as immunogens, resulted in considerably enhanced Ag-specific CTL responses. Several SynB vectors were tested and resulted in varying levels of cellular uptake. The efficiency of uptake correlated with the ability of the SynB construct to deliver each epitope in vivo and induce specific CTL responses in mice. These data suggest that peptide vectors, such as SynB that transport target Ags across the cell membrane in a highly efficient manner, have significant potential for vaccine delivery.


Subject(s)
Antigens, Bacterial/administration & dosage , Cytotoxicity, Immunologic , Epitopes, T-Lymphocyte/administration & dosage , Lymphocyte Activation , Nucleoproteins/administration & dosage , Peptide Fragments/administration & dosage , RNA-Binding Proteins , T-Lymphocytes, Cytotoxic/immunology , Viral Core Proteins/administration & dosage , Amino Acid Sequence , Animals , Antigen Presentation/genetics , Antigen Presentation/immunology , Antigens, Bacterial/immunology , Antigens, Bacterial/metabolism , Cytotoxicity, Immunologic/genetics , Epitopes, T-Lymphocyte/immunology , Female , Genetic Vectors/administration & dosage , Genetic Vectors/immunology , Humans , K562 Cells , Lymphocyte Activation/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred DBA , Molecular Sequence Data , Mycobacterium tuberculosis/immunology , Nucleocapsid Proteins , Nucleoproteins/immunology , Nucleoproteins/metabolism , Peptide Fragments/immunology , Peptide Fragments/metabolism , Recombinant Proteins/administration & dosage , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Tumor Cells, Cultured , Viral Core Proteins/immunology , Viral Core Proteins/metabolism
20.
Vaccine ; 20(25-26): 3123-9, 2002 Aug 19.
Article in English | MEDLINE | ID: mdl-12163263

ABSTRACT

Mucosal immunity is critical for protection from viral infections. We attempted to activate mucosal cytotoxic T lymphocytes (CTLs) specific for influenza A virus nucleoprotein (NP) which play an important role in protective immunity. It has been shown that dendritic cells (DCs) activated by signaling via CD40-CD40 ligand (CD40L) interaction are required for the differentiation of naive CD8(+) T cells into antigen-specific CTLs in a non-mucosal environment. We herein inoculated mice intranasally with an anti-CD40 monoclonal antibody (anti-CD40 mAb) and NP366-374 peptide, corresponding to a CTL epitope on NP, encapsulated in liposome (liposomal NP366-374) to induce protective CTL responses against influenza A virus. Intranasal but not subcutaneous immunization with liposomal NP366-374 effectively induced mucosal immunity to reduce virus replication in the lung, suggesting that anti-CD40 mAb also functioned as a mucosal adjuvant. Interestingly, neither MHC class I- nor class II-deficient mice immunized intranasally with these materials were resistant to the infection. Since anti-CD40 mAb was considered to help replace CD4(+) T cells, another help of CD4(+) T cells are presumably required for the induction of CTL activity in the lung. This approach may prove promising for developing vaccines to induce mucosal CTL responses, and seems to highlight differences between mucosal and non-mucosal immunity.


Subject(s)
Antibodies, Monoclonal/administration & dosage , CD40 Antigens/immunology , Influenza A virus/immunology , Influenza Vaccines/administration & dosage , Lung/immunology , Nucleoproteins/administration & dosage , Peptide Fragments/administration & dosage , T-Lymphocytes, Cytotoxic/immunology , Viral Core Proteins/administration & dosage , Adjuvants, Immunologic , Administration, Intranasal , Animals , Antibodies, Monoclonal/immunology , CD4-Positive T-Lymphocytes/immunology , Epitopes/administration & dosage , Epitopes/immunology , Female , Genes, MHC Class II , Histocompatibility Antigens Class II/genetics , Immunity, Mucosal/immunology , Immunization Schedule , Influenza A virus/physiology , Influenza Vaccines/immunology , Kinetics , Liposomes/administration & dosage , Lung/virology , Mice , Mice, Inbred C57BL , Mice, Knockout , Nucleocapsid Proteins , Nucleoproteins/immunology , Peptide Fragments/immunology , Vaccination/methods , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/immunology , Viral Core Proteins/immunology , Virus Replication , beta 2-Microglobulin/deficiency , beta 2-Microglobulin/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...