Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
EBioMedicine ; 98: 104878, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38016322

ABSTRACT

BACKGROUND: SARS-CoV-2 booster vaccination should ideally enhance protection against variants and minimise immune imprinting. This Phase I trial evaluated two vaccines targeting SARS-CoV-2 beta-variant receptor-binding domain (RBD): a recombinant dimeric RBD-human IgG1 Fc-fusion protein, and an mRNA encoding a membrane-anchored RBD. METHODS: 76 healthy adults aged 18-64 y, previously triple vaccinated with licensed SARS-CoV-2 vaccines, were randomised to receive a 4th dose of either an adjuvanted (MF59®, CSL Seqirus) protein vaccine (5, 15 or 45 µg, N = 32), mRNA vaccine (10, 20, or 50 µg, N = 32), or placebo (saline, N = 12) at least 90 days after a 3rd boost vaccination or SARS-CoV-2 infection. Bleeds occurred on days 1 (prior to vaccination), 8, and 29. CLINICALTRIALS: govNCT05272605. FINDINGS: No vaccine-related serious or medically-attended adverse events occurred. The protein vaccine reactogenicity was mild, whereas the mRNA vaccine was moderately reactogenic at higher dose levels. Best anti-RBD antibody responses resulted from the higher doses of each vaccine. A similar pattern was seen with live virus neutralisation and surrogate, and pseudovirus neutralisation assays. Breadth of immune response was demonstrated against BA.5 and more recent omicron subvariants (XBB, XBB.1.5 and BQ.1.1). Binding antibody titres for both vaccines were comparable to those of a licensed bivalent mRNA vaccine. Both vaccines enhanced CD4+ and CD8+ T cell activation. INTERPRETATION: There were no safety concerns and the reactogenicity profile was mild and similar to licensed SARS-CoV-2 vaccines. Both vaccines showed strong immune boosting against beta, ancestral and omicron strains. FUNDING: Australian Government Medical Research Future Fund, and philanthropies Jack Ma Foundation and IFM investors.


Subject(s)
COVID-19 Vaccines , COVID-19 , Adult , Humans , Antibodies, Neutralizing , Antibodies, Viral , Australia , COVID-19/prevention & control , COVID-19 Vaccines/adverse effects , mRNA Vaccines , SARS-CoV-2 , Adolescent , Young Adult , Middle Aged
2.
EBioMedicine ; 92: 104574, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37148585

ABSTRACT

BACKGROUND: The SARS-CoV-2 global pandemic has fuelled the generation of vaccines at an unprecedented pace and scale. However, many challenges remain, including: the emergence of vaccine-resistant mutant viruses, vaccine stability during storage and transport, waning vaccine-induced immunity, and concerns about infrequent adverse events associated with existing vaccines. METHODS: We report on a protein subunit vaccine comprising the receptor-binding domain (RBD) of the ancestral SARS-CoV-2 spike protein, dimerised with an immunoglobulin IgG1 Fc domain. These were tested in conjunction with three different adjuvants: a TLR2 agonist R4-Pam2Cys, an NKT cell agonist glycolipid α-Galactosylceramide, or MF59® squalene oil-in-water adjuvant, using mice, rats and hamsters. We also developed an RBD-human IgG1 Fc vaccine with an RBD sequence of the immuno-evasive beta variant (N501Y, E484K, K417N). These vaccines were also tested as a heterologous third dose booster in mice, following priming with whole spike vaccine. FINDINGS: Each formulation of the RBD-Fc vaccines drove strong neutralising antibody (nAb) responses and provided durable and highly protective immunity against lower and upper airway infection in mouse models of COVID-19. The 'beta variant' RBD vaccine, combined with MF59® adjuvant, induced strong protection in mice against the beta strain as well as the ancestral strain. Furthermore, when used as a heterologous third dose booster, the RBD-Fc vaccines combined with MF59® increased titres of nAb against other variants including alpha, delta, delta+, gamma, lambda, mu, and omicron BA.1, BA.2 and BA.5. INTERPRETATION: These results demonstrated that an RBD-Fc protein subunit/MF59® adjuvanted vaccine can induce high levels of broadly reactive nAbs, including when used as a booster following prior immunisation of mice with whole ancestral-strain spike vaccines. This vaccine platform offers a potential approach to augment some of the currently approved vaccines in the face of emerging variants of concern, and it has now entered a phase I clinical trial. FUNDING: This work was supported by grants from the Medical Research Future Fund (MRFF) (2005846), The Jack Ma Foundation, National Health and Medical Research Council of Australia (NHMRC; 1113293) and Singapore National Medical Research Council (MOH-COVID19RF-003). Individual researchers were supported by an NHMRC Senior Principal Research Fellowship (1117766), NHMRC Investigator Awards (2008913 and 1173871), Australian Research Council Discovery Early Career Research Award (ARC DECRA; DE210100705) and philanthropic awards from IFM investors and the A2 Milk Company.


Subject(s)
COVID-19 , Carrier Proteins , Cricetinae , Humans , Mice , Rats , Animals , COVID-19 Vaccines , SARS-CoV-2 , Protein Subunits , COVID-19/prevention & control , Australia , Adjuvants, Immunologic , Antibodies, Neutralizing , Antibodies, Viral
3.
Expert Rev Vaccines ; 21(8): 1055-1070, 2022 08.
Article in English | MEDLINE | ID: mdl-35652289

ABSTRACT

INTRODUCTION: Evaluation of immunogenicity and efficacy in animal models provide critical data in vaccine development. Nonhuman primates (NHPs) have been used extensively in the evaluation of SARS-CoV-2 vaccines. AREAS COVERED: A critical synthesis of SARS-CoV-2 vaccine development with a focus on challenge studies in NHPs is provided. The benefits and drawbacks of the NHP models are discussed. The citations were selected by the authors based on PubMed searches of the literature, summaries from national public health bodies, and press-release information provided by vaccine developers. EXPERT OPINION: We identify several aspects of NHP models that limit their usefulness for vaccine-challenge studies and numerous variables that constrain comparisons across vaccine platforms. We propose that studies conducted in NHPs for vaccine development should use a standardized protocol and, where possible, be substituted with smaller animal models. This will ensure continued rapid progression of vaccines to clinical trials without compromising assessments of safety or efficacy.


Subject(s)
COVID-19 Vaccines , COVID-19 , Animals , COVID-19/prevention & control , Disease Models, Animal , Humans , Primates , SARS-CoV-2
4.
Int J Mol Sci ; 23(2)2022 Jan 13.
Article in English | MEDLINE | ID: mdl-35055020

ABSTRACT

The global urgency to uncover medical countermeasures to combat the COVID-19 pandemic caused by the severe acute respiratory syndrome-coronavirus 2 (SARS-CoV-2) has revealed an unmet need for robust tissue culture models that faithfully recapitulate key features of human tissues and disease. Infection of the nose is considered the dominant initial site for SARS-CoV-2 infection and models that replicate this entry portal offer the greatest potential for examining and demonstrating the effectiveness of countermeasures designed to prevent or manage this highly communicable disease. Here, we test an air-liquid-interface (ALI) differentiated human nasal epithelium (HNE) culture system as a model of authentic SARS-CoV-2 infection. Progenitor cells (basal cells) were isolated from nasal turbinate brushings, expanded under conditionally reprogrammed cell (CRC) culture conditions and differentiated at ALI. Differentiated cells were inoculated with different SARS-CoV-2 clinical isolates. Infectious virus release into apical washes was determined by TCID50, while infected cells were visualized by immunofluorescence and confocal microscopy. We demonstrate robust, reproducible SARS-CoV-2 infection of ALI-HNE established from different donors. Viral entry and release occurred from the apical surface, and infection was primarily observed in ciliated cells. In contrast to the ancestral clinical isolate, the Delta variant caused considerable cell damage. Successful establishment of ALI-HNE is donor dependent. ALI-HNE recapitulate key features of human SARS-CoV-2 infection of the nose and can serve as a pre-clinical model without the need for invasive collection of human respiratory tissue samples.


Subject(s)
COVID-19/virology , Nasal Mucosa/cytology , Nasal Mucosa/virology , Tissue Culture Techniques/methods , Adolescent , Adult , Angiotensin-Converting Enzyme 2/metabolism , Cell Culture Techniques , Cell Differentiation , Epithelial Cells/cytology , Epithelial Cells/virology , Female , Humans , Male , Middle Aged , Models, Biological , SARS-CoV-2 , Virus Internalization
5.
JCI Insight ; 6(5)2021 03 08.
Article in English | MEDLINE | ID: mdl-33561017

ABSTRACT

The impact of respiratory virus infections on global health is felt not just during a pandemic, but endemic seasonal infections pose an equal and ongoing risk of severe disease. Moreover, vaccines and antiviral drugs are not always effective or available for many respiratory viruses. We investigated how induction of effective and appropriate antigen-independent innate immunity in the upper airways can prevent the spread of respiratory virus infection to the vulnerable lower airways. Activation of TLR2, when restricted to the nasal turbinates, resulted in prompt induction of innate immune-driven antiviral responses through action of cytokines, chemokines, and cellular activity in the upper but not the lower airways. We have defined how nasal epithelial cells and recruitment of macrophages work in concert and play pivotal roles to limit progression of influenza virus to the lungs and sustain protection for up to 7 days. These results reveal underlying mechanisms of how control of viral infection in the upper airways can occur and support the implementation of strategies that can activate TLR2 in nasal passages to provide rapid protection, especially for at-risk populations, against severe respiratory infection when vaccines and antiviral drugs are not always effective or available.


Subject(s)
Immunity, Innate/drug effects , Immunologic Factors/pharmacology , Influenza, Human , Lipopeptides/pharmacology , Lung , Respiratory Tract Infections , Toll-Like Receptor 2/metabolism , Animals , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Disease Models, Animal , Female , Humans , Immunologic Factors/therapeutic use , Influenza A virus , Influenza, Human/drug therapy , Influenza, Human/immunology , Influenza, Human/metabolism , Influenza, Human/virology , Lipopeptides/therapeutic use , Lung/drug effects , Lung/immunology , Lung/metabolism , Lung/virology , Male , Mice, Inbred C57BL , Orthomyxoviridae Infections/drug therapy , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/metabolism , Orthomyxoviridae Infections/virology , Respiratory System/drug effects , Respiratory System/immunology , Respiratory System/metabolism , Respiratory System/virology , Respiratory Tract Infections/drug therapy , Respiratory Tract Infections/immunology , Respiratory Tract Infections/metabolism , Respiratory Tract Infections/virology , Toll-Like Receptor 2/agonists
6.
Arch Virol ; 163(9): 2451-2457, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29752559

ABSTRACT

Ixodes holocyclus, the eastern paralysis tick, is a significant parasite in Australia in terms of animal and human health. However, very little is known about its virome. In this study, next-generation sequencing of I. holocyclus salivary glands yielded a full-length genome sequence which phylogenetically groups with viruses classified in the Iflaviridae family and shares 45% amino acid similarity with its closest relative Bole hyalomma asiaticum virus 1. The sequence of this virus, provisionally named Ixodes holocyclus iflavirus (IhIV) has been identified in tick populations from northern New South Wales and Queensland, Australia and represents the first virus sequence reported from I. holocyclus.


Subject(s)
Ixodes/virology , RNA Viruses/isolation & purification , Amino Acid Sequence , Animals , Australia , Cats/parasitology , Dogs/parasitology , Ixodes/genetics , Ixodes/physiology , Molecular Sequence Data , Phylogeny , RNA Viruses/chemistry , RNA Viruses/classification , RNA Viruses/genetics , Sequence Alignment , Viral Proteins/chemistry , Viral Proteins/genetics
7.
J Virol ; 91(14)2017 07 15.
Article in English | MEDLINE | ID: mdl-28446669

ABSTRACT

It is possible to model the progression of influenza virus from the upper respiratory tract to the lower respiratory tract in the mouse using viral inoculum delivered in a restricted manner to the nose. In this model, infection with the A/Udorn/307/72 (Udorn) strain of virus results ultimately in high viral titers in both the trachea and lungs. In contrast, the A/Puerto Rico/8/34 (PR8) strain causes an infection that is almost entirely limited to the nasal passages. The factors that govern the progression of virus down the respiratory tract are not well understood. Here, we show that, while PR8 virus grows to high titers in the nose, an inhibitor present in the saliva blocks further progression of infection to the trachea and lungs and renders an otherwise lethal dose of virus completely asymptomatic. In vitro, the salivary inhibitor was capable of potent neutralization of PR8 virus and an additional 20 strains of type A virus and two type B strains that were tested. The exceptions were Udorn virus and the closely related H3N2 strains A/Port Chalmers/1/73 and A/Victoria/3/75. Characterization of the salivary inhibitor showed it to be independent of sialic acid and other carbohydrates for its function. This and other biochemical properties, together with its virus strain specificity and in vivo function, indicate that the mouse salivary inhibitor is a previously undescribed innate inhibitory molecule that may have evolved to provide pulmonary protection of the species from fatal influenza virus infection.IMPORTANCE Influenza A virus occasionally jumps from aquatic birds, its natural host, into mammals to cause outbreaks of varying severity, including pandemics in humans. Despite the laboratory mouse being used as a model to study influenza virus pathogenesis, natural outbreaks of influenza have not been reported in the species. Here, we shed light on one mechanism that might allow mice to be protected from influenza in the wild. We show that virus deposited in the mouse upper respiratory tract will not progress to the lower respiratory tract due to the presence of a potent inhibitor of the virus in saliva. Containing inhibitor-sensitive virus to the upper respiratory tract renders an otherwise lethal infection subclinical. This knowledge sheds light on how natural inhibitors may have evolved to improve survival in this species.


Subject(s)
Influenza A virus/immunology , Orthomyxoviridae Infections/immunology , Respiratory Tract Infections/immunology , Saliva/immunology , Animals , Disease Models, Animal , Immunity, Innate , Lung/virology , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, SCID , Nasal Cavity/virology , Trachea/virology
8.
Immunol Cell Biol ; 91(1): 96-104, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23146941

ABSTRACT

In this study, we examined the reactivity of human peripheral blood mononuclear cells to a panel of influenza A virus (IAV) CD8(+) T-cell epitopes that are recognised by the major human leukocyte antigen (HLA) groups represented in the human population. We examined the level of recognition in a sample of the human population and the potential coverage that could be achieved if these were incorporated into a T-cell epitope-based vaccine. We then designed a candidate influenza vaccine that incorporated three of the examined HLA-A2-restricted influenza epitopes into Pam2Cys-based lipopeptides. These lipopeptides do not require the addition of an adjuvant and can be delivered directly to the respiratory mucosa enabling the generation of local memory cell populations that are crucial for clearance of influenza. Intranasal administration of a mixture of three lipopeptides to HLA-A2 transgenic HHD mice elicited multiple CD8(+) T-cell specificities in the spleen and lung that closely mimicked the response generated following natural infection with influenza. These CD8(+) T cells were associated with viral reduction following H3N1 influenza virus challenge for as long as 3 months after lipopeptide administration. In addition, lipopeptides containing IAV-targeting epitopes conferred substantial benefit against death following infection with a virulent H1N1 strain. Because CD8(+) T cell epitopes are often derived from highly conserved regions of influenza viruses, such vaccines need not be reformulated annually and unlike current antibody-inducing vaccines could provide cross-protective immunity against newly emerging pandemic viruses.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/immunology , Influenza A Virus, H1N1 Subtype/immunology , Influenza Vaccines/immunology , Influenza, Human/immunology , Administration, Intranasal , Animals , Cross Reactions , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/pharmacology , Female , HLA-A2 Antigen/genetics , HLA-A2 Antigen/immunology , Humans , Influenza A Virus, H1N1 Subtype/genetics , Influenza Vaccines/genetics , Influenza, Human/genetics , Influenza, Human/prevention & control , Lipopeptides/immunology , Lipopeptides/pharmacology , Male , Mice , Mice, Transgenic
9.
Antiviral Res ; 94(2): 168-78, 2012 May.
Article in English | MEDLINE | ID: mdl-22504097

ABSTRACT

In hepatitis C virus (HCV) infection, CD8(+) T cell responses have been shown to be important in viral clearance. Examining the efficacy of CD8(+) T cell vaccines against HCV has been limited by the lack of an HCV infectious model in mice and the differences between MHC restriction in humans and mice. Using HLA-A2 transgenic HHD mice, we demonstrate that intranasally delivered Pam2Cys-based lipopeptides containing HLA-A2-restricted HCV epitopes can induce polyfunctional CD8(+) T cell responses in several organs including the liver. To examine the activity of these responses in an infectious context, we developed a recombinant influenza virus that expresses the NS5B(2594-2602) epitope from non-structural protein 5B of hepatitis C virus (PR8-HCV(NS5B)). We showed that mice inoculated with a lipopeptide containing the NS5B epitope had reduced viral loads following challenge with the PR8-HCV(NS5B) virus. This reduction was associated with the induction of NS5B(2594-2602)-specific IFN-γ and TNF-α co-producing CD8(+) T cells. The T cell receptor usage in the NS5B(2594-2602) response was found to exhibit a Vß8.1/8.2 bias that was characterized by a narrow repertoire and a common CDR3ß motif. This work has identified CD8(+) T cell functions induced by lipopeptides that are associated with viral control and demonstrate the potential of lipopeptide-based vaccines as candidates for treatment of HCV infection.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Epitopes/immunology , Genetic Vectors , Hepacivirus/immunology , Orthomyxoviridae/pathogenicity , Viral Hepatitis Vaccines/immunology , Administration, Intranasal , Animals , Disease Models, Animal , Epitopes/genetics , Female , Hepacivirus/genetics , Interferon-gamma/metabolism , Mice , Mice, Transgenic , Orthomyxoviridae/genetics , Orthomyxoviridae/immunology , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/pathology , Tumor Necrosis Factor-alpha/metabolism , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Viral Hepatitis Vaccines/administration & dosage , Viral Hepatitis Vaccines/genetics
10.
Vaccine ; 27(18): 2475-82, 2009 Apr 21.
Article in English | MEDLINE | ID: mdl-19368789

ABSTRACT

The effectiveness of single dose, intranasally delivered vaccines comprising detergent-disrupted inactivated influenza virus (split virus) and ISCOMATRIX adjuvant was examined in mice. Vaccines formulated with adjuvant required 10- to 100-fold less split virus antigen to induce pulmonary protection following viral challenge when compared to vaccines containing split virus alone. Furthermore, those formulated with ISCOMATRIX adjuvant elicited specific antibody in serum, saliva, vaginal, nasal and lung fluids when delivered to the entire respiratory tract. No specific antibody was detected in serum or mucosal samples, however, when the same vaccines were delivered using a procedure that restricted the inoculum to the nasal passages. Good protective responses can thus be achieved with only a single intranasal inoculation of influenza vaccine formulated with adjuvant, providing the vaccine can access sites of immune induction in the lower respiratory tract.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Antibodies, Viral/immunology , Cholesterol/administration & dosage , Influenza Vaccines/administration & dosage , Phospholipids/administration & dosage , Saponins/administration & dosage , Administration, Intranasal , Animals , Drug Combinations , Immunization , Influenza Vaccines/immunology , Mice , Mice, Inbred BALB C
11.
Vaccine ; 26(44): 5641-8, 2008 Oct 16.
Article in English | MEDLINE | ID: mdl-18761047

ABSTRACT

We report here on the use of a Sindbis virus-based DNA-launch RNA replicon vector (pSIN-HA) that expresses influenza hemagglutinin (HA) as an immunogen. Immunization of mice with pSIN-HA generated anti-HA antibody and CTL responses and resulted in lower lung viral titers after influenza challenge when compared to controls. Importantly, immunization with a low dose of pSIN-HA mediated significantly reduced lung viral titers following challenge at 43 weeks after the final immunization. In contrast, immunization with a non-replicon DNA vector expressing HA failed to mediate reduced lung viral titer at the same dose. This demonstrated the dose-sparing capacity of the SIN vector system and its ability to stimulate long-term memory responses, properties that are highly desirable in any vaccine formulation.


Subject(s)
Genetic Vectors , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Influenza A Virus, H1N1 Subtype/immunology , Influenza Vaccines/administration & dosage , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/prevention & control , Sindbis Virus/genetics , Animals , Antibodies, Viral/blood , Cricetinae , Female , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Immunization , Influenza A Virus, H1N1 Subtype/genetics , Lung/virology , Mice , Mice, Inbred BALB C , Orthomyxoviridae Infections/virology , Plasmids , Replicon , Sindbis Virus/immunology , T-Lymphocytes, Cytotoxic/immunology , Vaccination
12.
Vaccine ; 26(44): 5649-53, 2008 Oct 16.
Article in English | MEDLINE | ID: mdl-18725260

ABSTRACT

Vaccination strategies against Klebsiella pneumoniae have largely focussed on the polysaccharide capsule. However, the large number and high prevalence of individual capsular serotypes limits the widespread applicability of capsule-based vaccines. This study establishes that immunization with purified LPS can protect mice against lethal challenge with K. pneumoniae, and that subcapsular antibodies directed against purified LPS can be used to treat and/or prevent experimental K. pneumoniae infection in mice. This approach offers potential for prophylaxis and/or therapy against drug-resistant strains of K. pneumoniae.


Subject(s)
Antibodies, Bacterial/therapeutic use , Antibodies, Monoclonal/therapeutic use , Bacterial Capsules/immunology , Bacterial Vaccines/administration & dosage , Klebsiella Infections/prevention & control , Klebsiella pneumoniae/immunology , Lipopolysaccharides/administration & dosage , Animals , Antibodies, Bacterial/blood , Antibodies, Bacterial/immunology , Antibodies, Monoclonal/blood , Antibodies, Monoclonal/immunology , Antigens, Bacterial/immunology , Bacterial Vaccines/immunology , Klebsiella Infections/immunology , Klebsiella Infections/microbiology , Klebsiella pneumoniae/classification , Lipopolysaccharides/immunology , Mice , Mice, Inbred BALB C , O Antigens/immunology , Serotyping , Vaccination
13.
Respir Res ; 9: 53, 2008 Jul 15.
Article in English | MEDLINE | ID: mdl-18627612

ABSTRACT

BACKGROUND: Cigarette smoke has both pro-inflammatory and immunosuppressive effects. Both active and passive cigarette smoke exposure are linked to an increased incidence and severity of respiratory virus infections, but underlying mechanisms are not well defined. We hypothesized, based on prior gene expression profiling studies, that upregulation of pro-inflammatory mediators by short term smoke exposure would be protective against a subsequent influenza infection. METHODS: BALB/c mice were subjected to whole body smoke exposure with 9 cigarettes/day for 4 days. Mice were then infected with influenza A (H3N1, Mem71 strain), and analyzed 3 and 10 days later (d3, d10). These time points are the peak and resolution (respectively) of influenza infection. RESULTS: Inflammatory cell influx into the bronchoalveolar lavage (BALF), inflammatory mediators, proteases, histopathology, viral titres and T lymphocyte profiles were analyzed. Compared to smoke or influenza alone, mice exposed to smoke and then influenza had more macrophages, neutrophils and total lymphocytes in BALF at d3, more macrophages in BALF at d10, lower net gelatinase activity and increased activity of tissue inhibitor of metalloprotease-1 in BALF at d3, altered profiles of key cytokines and CD4+ and CD8+ T lymphocytes, worse lung pathology and more virus-specific, activated CD8+ T lymphocytes in BALF. Mice smoke exposed before influenza infection had close to 10-fold higher lung virus titres at d3 than influenza alone mice, although all mice had cleared virus by d10, regardless of smoke exposure. Smoke exposure caused temporary weight loss and when smoking ceased after viral infection, smoke and influenza mice regained significantly less weight than smoke alone mice. CONCLUSION: Smoke induced inflammation does not protect against influenza infection.In most respects, smoke exposure worsened the host response to influenza. This animal model may be useful in studying how smoke worsens respiratory viral infections.


Subject(s)
Orthomyxoviridae Infections/physiopathology , Pneumonia/physiopathology , Pneumonia/virology , Smoking/adverse effects , Animals , Bronchoalveolar Lavage Fluid , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/pathology , Disease Models, Animal , Immunosuppression Therapy , Influenza A virus/immunology , Influenza A virus/pathogenicity , Lung/enzymology , Lung/pathology , Lung/virology , Macrophages/pathology , Male , Mice , Mice, Inbred BALB C , Neutrophils/pathology , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/pathology , Peptide Hydrolases/metabolism , Pneumonia/pathology , Tissue Inhibitor of Metalloproteinases/metabolism
14.
Int Immunol ; 18(12): 1801-13, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17077175

ABSTRACT

Anti-viral CD8(+) T cell responses can be induced using synthetic lipopeptides and a range of different lipid moieties have been examined in a variety of model systems and in man for this purpose. Nevertheless, only limited data exist on comparative efficacy of different lipopeptides in a single model of protection so that the optimal composition for vaccination purposes remains unknown. In this study, we examined different lipid structures from bacterial or non-bacterial sources coupled to peptides representing influenza viral epitopes recognized by CD8(+) and CD4(+) T cells. These were assessed in the context of intra-nasal (i.n.) immunization in the absence of added adjuvant. The strongest immunogens were those containing bacterially derived lipids that induced dendritic cell (DC) maturation via Toll-like receptor 2 (TLR2) binding. The number of DCs induced to mature in vitro was directly associated with the strength of the CD8(+) T cell-mediated viral clearing responses in primed mice. Mice immunized with the TLR2-binding lipopeptides showed greatly enhanced numbers of specific IFN-gamma-secreting CD8(+) T cells at the site of infection after i.n. exposure to virus, which resulted in enhanced protection of the pneumonic lung. Importantly, lipopeptide-pulsed DCs were able to induce the appropriate T cells, indicating that the self-adjuvanting effects could occur in the absence of free lipopeptide interacting with additional TLR2-bearing cells in vivo. This study defines a hierarchy of lipopeptide constructs that can program DC to prime memory CD8(+) T cells that on recall function to clear influenza virus from the infected lung.


Subject(s)
Biomimetics , Immunity, Innate , Influenza Vaccines/immunology , Lipoproteins , Lymphocyte Activation , T-Lymphocytes, Cytotoxic/immunology , Amino Acid Sequence , Animals , Female , Humans , Immunization , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H1N1 Subtype/pathogenicity , Influenza A Virus, H3N2 Subtype/genetics , Influenza A Virus, H3N2 Subtype/pathogenicity , Influenza Vaccines/administration & dosage , Influenza Vaccines/chemistry , Influenza, Human/immunology , Influenza, Human/prevention & control , Influenza, Human/virology , Lipoproteins/administration & dosage , Lipoproteins/chemical synthesis , Lipoproteins/chemistry , Lipoproteins/immunology , Lung/immunology , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Peptides/administration & dosage , Peptides/chemical synthesis , Peptides/chemistry , Peptides/immunology , Reassortant Viruses/genetics , Reassortant Viruses/pathogenicity
15.
Eur J Immunol ; 36(3): 770-8, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16435281

ABSTRACT

The longevity of the influenza virus-specific CD8+ T cell response following intranasal delivery of a synthetic lipopeptide was investigated and the characteristics and location of the cells associated with viral clearance examined. The lipopeptide, incorporating an epitope for CD8+ T cells and another for CD4+ T cells with the lipid moiety S-[2,3-bis(palmitoyloxy)propyl]cysteine (Pam2Cys) attached, induced potent and long-lived pulmonary protection. Both the lipopeptide and its largely unprotective non-lipidated counterpart elicited comparable numbers of CD8+ T cells in the spleen, which was the main location of the memory pool. However, the lipopeptide, unlike the non-lipidated peptide, also induced a substantial memory population that remained in the lungs and was rapidly activated upon viral challenge months later. These lipopeptide-induced lung-resident CD8+ T cells were also very similar in number and IFN-gamma-secreting potential to those induced by prior exposure to the virus itself and are likely mediators of initial viral clearance prior to recruitment from the expanding lymph node T cell pool. Significant clearing responses were demonstrated as late as 9 months post-lipopeptide vaccination. This study shows that CD8+ T cells primed by the lipopeptide are not only long-lived but can take up residence in the lung where they are important early mediators of pulmonary protection.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/immunology , Immunologic Memory/immunology , Influenza A virus/immunology , Influenza Vaccines/immunology , Influenza, Human/immunology , Lipoproteins/immunology , Vaccines, Synthetic/immunology , Administration, Intranasal , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Epitopes, T-Lymphocyte/administration & dosage , Female , Humans , Influenza Vaccines/administration & dosage , Influenza, Human/prevention & control , Interferon-gamma/immunology , Interferon-gamma/metabolism , Lipoproteins/administration & dosage , Lung/immunology , Lymph Nodes/immunology , Mice , Mice, Inbred BALB C , Vaccines, Synthetic/administration & dosage
16.
Int Immunol ; 18(2): 291-300, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16361310

ABSTRACT

CD45 is a receptor tyrosine phosphatase essential for TCR signaling. One isoform, CD45RB, is down-regulated in memory cells and targeting CD45RB with a specific antibody has been shown to inhibit graft rejection. Its role in immunity to infection, however, has not been tested. Here, we report the effect of anti-CD45RB antibody treatment on the induction of anti-influenza CD8+ T cells and viral clearance. Anti-CD45RB-treated mice had delayed pulmonary viral clearance compared with untreated mice whose infection was completely cleared by day 8 post-infection. In anti-CD45RB-treated mice, the total CD4+ and CD8+ T cell numbers in both the lungs and mediastinal nodes were substantially reduced at days 5 and 8; this effect was less marked for the spleen. CD8+ T cells specific for influenza virus were also reduced compared with the control group in all three organs at day 8. By day 11, when both treated and control groups showed no virus remaining in the lungs, specific CD8+ T cell numbers were at similar low levels. Homing to lymph nodes and lung of dye-labeled T cells was greatly inhibited (by >80%) by anti-CD45RB treatment. This reduced homing corresponded with reduced CD62L and beta1-integrin expression in both uninfected and infected mice. Since CD62L plays a critical role in homing lymphocytes to lymph nodes, and high levels of CD62L and alpha4beta1-integrin are expressed by lymphocytes that home to bronchus-associated lymphoid tissue, we suggest that reduced expression of these molecules is a key explanation for the delay in immune responses.


Subject(s)
Influenza, Human/immunology , Leukocyte Common Antigens/immunology , Animals , Antibodies, Viral/immunology , Antibody Formation , CD8-Positive T-Lymphocytes/immunology , Female , Humans , Influenza, Human/therapy , Influenza, Human/virology , L-Selectin/biosynthesis , Mice , Mice, Inbred C57BL , Mice, Transgenic , Protein Isoforms/immunology
17.
Immunol Cell Biol ; 83(2): 119-28, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15748208

ABSTRACT

The immunostimulating complex or 'iscom' was first described 20 years ago as an antigen delivery system with powerful immunostimulating activity. Iscoms are cage-like structures, typically 40 nm in diameter, that are comprised of antigen, cholesterol, phospholipid and saponin. ISCOM-based vaccines have been shown to promote both antibody and cellular immune responses in a variety of experimental animal models. This review focuses on the evaluation of ISCOM-based vaccines in animals over the past 10 years, as well as examining the progress that has been achieved in the development of human vaccines based on ISCOM adjuvant technology.


Subject(s)
ISCOMs/immunology , Animals , Antigens/administration & dosage , CD8-Positive T-Lymphocytes/immunology , Cholesterol/immunology , Drug Combinations , Drug Delivery Systems , Humans , Immune System/immunology , Mice , Microscopy, Electron , Phospholipids/immunology , Saponins/immunology
18.
Proc Natl Acad Sci U S A ; 101(43): 15440-5, 2004 Oct 26.
Article in English | MEDLINE | ID: mdl-15489266

ABSTRACT

A simple generic peptide-based vaccine structure that targets Toll-like receptor 2-expressing dendritic cells and causes their activation is described. The vaccines are totally synthetic, serve as their own adjuvant, and are composed of (i) a single helper T cell epitope, (ii) a target epitope that is either recognized by CD8+ T cells or B cells, and (iii) a Toll-like receptor 2-targeting lipid moiety, S-[2,3-bis(palmitoyloxy)propyl]cysteine, that is situated between the peptide epitopes to form a branched configuration. The different CD8+ T cell epitopes examined were from (i) influenza virus, (ii) the intracellular bacterium Listeria monocytogenes, and (iii) ovalbumin as a model tumor antigen. Vaccines containing a B cell epitope from gastrin or luteinizing hormone-releasing hormone as a B cell epitope were also examined for their ability to elicit antibody against the parent hormones. Each of the vaccines was capable of inducing either CD8+ T cell or antibody-mediated immune responses. The lipidated vaccines, but not the nonlipidated vaccines, were able to mediate protection against viral or bacterial infection and mediate prophylactic and therapeutic anticancer activity. The two hormone-based vaccines induced high antibody titers, which in the case of luteinizing hormone-releasing hormone resulted in abrogation of reproductive function. These results highlight the utility of simple, totally synthetic, epitope-based vaccines.


Subject(s)
Antibody Formation , Dendritic Cells/immunology , Membrane Glycoproteins/immunology , Receptors, Cell Surface/immunology , T-Lymphocytes, Cytotoxic/immunology , Vaccines, Synthetic/immunology , Amino Acid Sequence , Animals , Cell Line , Female , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Molecular Sequence Data , Toll-Like Receptor 2 , Toll-Like Receptors
19.
Immunol Res ; 30(1): 1-14, 2004.
Article in English | MEDLINE | ID: mdl-15258307

ABSTRACT

The most common models of CD4 T-cell deficiency are mice exogenously injected with anti-CD4 antibody (Ab), CD4 knockout (CD4-/-) and major histocompatibility complex (MHC) class II knockout (class II-/-) mice. We recently described the anti-CD4 Ab transgenic mouse (GK) as an improved CD4 cell-deficient model. This review compares this new GK mouse model with the widely available class II-/- and CD4-/- mice, when exposed to complex antigens (foreign grafts and during bacterial or viral infection). We highlight here the cytometric and functional differences (including Ab isotype, viral or bacterial clearance, and graft survival) among these CD4 cell-deficient models. For example, whereas grafts are generally rejected in class II-/- and CD4-/- mice as quickly as in wild-type mice, they survive longer in GK mice. Also, CD4-/- mice produce IgG against both simple model and complex antigens, but class II-/- and GK mice produce small amounts of IgG2a against complex antigens but not simple model antigens. These differences harbinger the caveats in the use of these various mice.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Lymphocyte Depletion , Animals , Antigens, Bacterial/immunology , Antigens, Viral/immunology , CD4 Antigens/genetics , CD4 Antigens/immunology , CD4-Positive T-Lymphocytes/microbiology , CD4-Positive T-Lymphocytes/virology , Disease Models, Animal , Graft Rejection/immunology , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/immunology , Mice , Mice, Knockout , Mice, Transgenic
20.
Mol Pharm ; 1(3): 220-32, 2004.
Article in English | MEDLINE | ID: mdl-15981925

ABSTRACT

The ability of the short, proline-rich native antibacterial peptides to penetrate bacterial and host cells suggests the utility of these transport systems in delivering peptidic cargo into cells. We studied the uptake of pyrrhocoricin and its most potent dimeric analogue by bacteria as well as human dendritic cells and fibroblasts. Native pyrrhocoricin entered the susceptible organism Escherichia coli very efficiently and the nonsusceptible bacterium Staphylococcus aureus to a significant degree. The antibacterial peptide also penetrated human monocyte-derived dendritic cells. It failed, however, to enter fibroblasts, whereas the designer analogue Pip-pyrr-MeArg dimer penetrated all the cell types that were studied. When glucoincretin hormone Glp-1 fragment 7-36 was cosynthesized with the dimer, the antibacterial peptide derivative lost its ability to cross the bacterial membrane layer. In contrast, a chimera of the Pip-pyrr-MeArg dimer and two copies of a shorter (nine residues) class I major histocompatibility complex epitope successfully entered bacterial and mammalian cells. While the Pip-pyrr-MeArg dimer was not immunogenic when inoculated into mice, the chimera elicited a strong cytotoxic T-cell response, indicating the maintenance of the antigenic integrity of the cargo in the peptide conjugate. The chimera when tested for its immunological properties activated human dendritic cells significantly more strongly than any of the two independent fragments alone, yet lacked mammalian cell toxicity. These results confirm the utility of designed pyrrhocoricin analogues for delivery of peptidic cargo across cell membranes in general, and their potential as carriers for epitope-based vaccines in particular.


Subject(s)
Anti-Bacterial Agents/metabolism , Antimicrobial Cationic Peptides/metabolism , Antimicrobial Cationic Peptides/pharmacology , Drug Delivery Systems , Insect Proteins/metabolism , Insect Proteins/pharmacology , Peptide Fragments/metabolism , Peptide Fragments/pharmacology , Amino Acid Sequence , Animals , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/pharmacology , Antimicrobial Cationic Peptides/chemistry , Cells, Cultured , Chlorocebus aethiops , Dendritic Cells/drug effects , Dimerization , Escherichia coli/drug effects , Escherichia coli/metabolism , Fibroblasts , Humans , Insect Proteins/chemistry , Molecular Sequence Data , Peptide Fragments/chemistry , Staphylococcus aureus/drug effects , Staphylococcus aureus/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...