Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters











Publication year range
1.
Wellcome Open Res ; 9: 113, 2024.
Article in English | MEDLINE | ID: mdl-38800518

ABSTRACT

Background: Major histocompatibility class I (MHC-I, human leukocyte antigen [HLA]-I in humans) molecules present small fragments of the proteome on the cell surface for immunosurveillance, which is pivotal to control infected and malignant cells. Immunogenic peptides are generated and selected in the MHC-I antigen processing and presentation pathway. In this pathway, two homologous molecules, tapasin and TAPBPR, optimise the MHC-I peptide repertoire that is ultimately presented at the plasma membrane. Peptide exchange on HLA-I by human TAPBPR involves the flexible loop region K22-D35, with the leucine at position 30 (L30) involved in mediating peptide dissociation. However, our understanding of the exact molecular mechanisms governing TAPBPR-mediated peptide exchange on HLA-I allotypes remains incomplete. Methods: Here, in-depth re-analyses of published immunopeptidomics datasets was used to further examine TAPBPR peptide editing activity and mechanism of action on HLA-I. The role of the TAPBPR editing loop in opening the HLA-I peptide binding groove was assessed using a molecular dynamics simulation. Results: We show that TAPBPR shapes the peptide repertoire on HLA-A, -B and -C allotypes. The TAPBPR editing loop was not essential to allow HLA-I to adopt an open state. L30 in the TAPBPR editing loop was typically sufficient to mediate peptide repertoire restriction on the three HLA-I allotypes expressed by HeLa cells. TAPBPR was also able to load peptides onto HLA-I in a loop-dependent manner. Conclusions: These results unify the previously hypothesised scoop loop and peptide trap mechanisms of TAPBPR-mediated peptide exchange, with the former involved in peptide filtering and the latter in peptide loading.


Major histocompatibility complex (MHC) class I molecules play an essential role in alerting the immune system to infection and cellular changes. They do this by displaying small fragments of proteins (peptides) from pathogen-infected cells and tumours on the cell surface to immune cells. When activated, immune cells can then destroy the target cell. In 2015, we discovered that a novel accessory protein, called TAPBPR, assists in the selection of peptides displayed on MHC class I molecules for immune surveillance. A specific region in the TAPBPR protein ­ the editing loop ­ is known to be involved in removing peptides from MHC class I. However, our understanding of the process of peptide selection on MHC class I molecules remains incomplete. Here, we show that TAPBPR is not only involved in removing peptides from MHC class I molecules but also assists in peptide loading. Additionally, we demonstrate that the TAPBPR editing loop is involved in both removing and loading of peptides. Our results suggest that TAPBPR fine-tunes the peptide repertoire displayed on three different types of MHC class I molecules. Developing our understanding of the mechanisms of peptide selection on MHC class I molecules has important implications in disease and the development of new therapies.

2.
PLoS Pathog ; 18(6): e1010612, 2022 06.
Article in English | MEDLINE | ID: mdl-35727847

ABSTRACT

The interaction between immune cells and virus-infected targets involves multiple plasma membrane (PM) proteins. A systematic study of PM protein modulation by vaccinia virus (VACV), the paradigm of host regulation, has the potential to reveal not only novel viral immune evasion mechanisms, but also novel factors critical in host immunity. Here, >1000 PM proteins were quantified throughout VACV infection, revealing selective downregulation of known T and NK cell ligands including HLA-C, downregulation of cytokine receptors including IFNAR2, IL-6ST and IL-10RB, and rapid inhibition of expression of certain protocadherins and ephrins, candidate activating immune ligands. Downregulation of most PM proteins occurred via a proteasome-independent mechanism. Upregulated proteins included a decoy receptor for TRAIL. Twenty VACV-encoded PM proteins were identified, of which five were not recognised previously as such. Collectively, this dataset constitutes a valuable resource for future studies on antiviral immunity, host-pathogen interaction, poxvirus biology, vector-based vaccine design and oncolytic therapy.


Subject(s)
Communicable Diseases , Poxviridae , Vaccinia , Humans , Immune Evasion , Membrane Proteins/metabolism , Vaccinia virus
3.
J Infect Dis ; 218(4): 614-623, 2018 07 13.
Article in English | MEDLINE | ID: mdl-29912453

ABSTRACT

Background: High-pathogenicity avian influenza viruses continue to circulate in poultry and wild birds and occasionally infect humans, sometimes with fatal outcomes. Development of vaccines is a priority to prepare for potential pandemics but is complicated by antigenic variation of the surface glycoprotein hemagglutinin. We report the immunological profile induced by human immunization with modified vaccinia virus Ankara (MVA) expressing the hemagglutinin gene of influenza A(H5N1) virus A/Vietnam/1194/04 (rMVA-H5). Methods: In a double-blinded phase 1/2a clinical trial, 79 individuals received 1 or 2 injections of rMVA-H5 or vector control. Twenty-seven study subjects received a booster immunization after 1 year. The breadth, magnitude, and properties of vaccine-induced antibody and T-cell responses were characterized. Results: rMVA-H5 induced broadly reactive antibody responses, demonstrated by protein microarray, hemagglutination inhibition, virus neutralization, and antibody-dependent cellular cytotoxicity assays. Antibodies cross-reacted with antigenically distinct H5 viruses, including the recently emerged subtypes H5N6 and H5N8 and the currently circulating subtype H5N1. In addition, the induction of T cells specific for H5 viruses of 2 different clades was demonstrated. Conclusions: rMVA-H5 induced immune responses that cross-reacted with H5 viruses of various clades. These findings validate rMVA-H5 as vaccine candidate against antigenically distinct H5 viruses. Clinical Trials Registration: NTR3401.


Subject(s)
Antibodies, Viral/blood , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Influenza A Virus, H5N1 Subtype/immunology , Influenza Vaccines/immunology , Influenza, Human/prevention & control , T-Lymphocytes/immunology , Adult , Antibody-Dependent Cell Cytotoxicity , Cross Reactions , Double-Blind Method , Drug Carriers , Female , Hemagglutination Inhibition Tests , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Humans , Immunization Schedule , Influenza A Virus, H5N1 Subtype/genetics , Influenza Vaccines/administration & dosage , Influenza Vaccines/genetics , Male , Neutralization Tests , Protein Array Analysis , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Vaccinia virus/genetics , Young Adult
4.
Sci Rep ; 8(1): 6474, 2018 04 24.
Article in English | MEDLINE | ID: mdl-29692427

ABSTRACT

The replication-deficient orthopoxvirus modified vaccinia virus Ankara (MVA) is a promising vaccine vector against various pathogens and has an excellent safety record. However, pre-existing vector-specific immunity is frequently suggested to be a drawback of MVA-based vaccines. To address this issue, mice were vaccinated with MVA-based influenza vaccines in the presence or absence of orthopoxvirus-specific immunity. Importantly, protective efficacy of an MVA-based influenza vaccine against a homologous challenge was not impaired in the presence of orthopoxvirus-specific pre-existing immunity. Nonetheless, orthopoxvirus-specific pre-existing immunity reduced the induction of antigen-specific antibodies under specific conditions and completely prevented induction of antigen-specific T cell responses by rMVA-based vaccination. Notably, antibodies induced by vaccinia virus vaccination, both in mice and humans, were not capable of neutralizing MVA. Thus, when using rMVA-based vaccines it is important to consider the main correlate of protection induced by the vaccine, the vaccine dose and the orthopoxvirus immune status of vaccine recipients.


Subject(s)
Influenza Vaccines/immunology , Influenza Vaccines/metabolism , Orthopoxvirus/immunology , Adaptive Immunity/physiology , Adolescent , Adult , Animals , Antibodies, Viral/immunology , Cross Reactions/immunology , Disease Models, Animal , Dogs , Female , Humans , Influenza Vaccines/pharmacology , Influenza, Human/prevention & control , Madin Darby Canine Kidney Cells , Male , Mice , Mice, Inbred C57BL , Orthomyxoviridae/immunology , Orthomyxoviridae Infections/immunology , Orthopoxvirus/metabolism , Vaccination , Vaccinia/immunology , Vaccinia virus/metabolism , Young Adult
5.
Immunol Res ; 66(2): 224-233, 2018 04.
Article in English | MEDLINE | ID: mdl-29594879

ABSTRACT

Influenza viruses continuously circulate in the human population and escape recognition by virus neutralizing antibodies induced by prior infection or vaccination through accumulation of mutations in the surface proteins hemagglutinin (HA) and neuraminidase (NA). Various strategies to develop a vaccine that provides broad protection against different influenza A viruses are under investigation, including use of recombinant (r) viral vectors and adjuvants. The replication-deficient modified vaccinia virus Ankara (MVA) is a promising vaccine vector that efficiently induces B and T cell responses specific for the antigen of interest. It is assumed that live vaccine vectors do not require an adjuvant to be immunogenic as the vector already mediates recruitment and activation of immune cells. To address this topic, BALB/c mice were vaccinated with either protein- or rMVA-based HA influenza vaccines, formulated with or without the saponin-based Matrix-M™ adjuvant. Co-formulation with Matrix-M significantly increased HA vaccine immunogenicity, resulting in antigen-specific humoral and cellular immune responses comparable to those induced by unadjuvanted rMVA-HA. Of special interest, rMVA-HA immunogenicity was also enhanced by addition of Matrix-M, demonstrated by enhanced HA inhibition antibody titres and cellular immune responses. Matrix-M added to either protein- or rMVA-based HA vaccines mediated recruitment and activation of antigen-presenting cells and lymphocytes to the draining lymph node 24 and 48 h post-vaccination. Taken together, these results suggest that adjuvants can be used not only with protein-based vaccines but also in combination with rMVA to increase vaccine immunogenicity, which may be a step forward to generate new and more effective influenza vaccines.


Subject(s)
Adjuvants, Immunologic/pharmacology , Immunogenicity, Vaccine , Influenza Vaccines/immunology , Influenza Vaccines/pharmacology , Vaccinia virus/immunology , Animals , Antigen-Presenting Cells/immunology , Female , Lymphocytes/immunology , Mice , Mice, Inbred BALB C , Vaccinia virus/genetics
6.
Sci Rep ; 7(1): 8580, 2017 08 17.
Article in English | MEDLINE | ID: mdl-28819261

ABSTRACT

Modified Vaccinia virus Ankara (MVA) is a promising vaccine vector with an excellent safety profile. However, despite extensive pre-clinical and clinical testing, surprisingly little is known about the cellular tropism of MVA, especially in relevant animal species. Here, we performed in vitro, ex vivo and in vivo experiments with recombinant MVA expressing green fluorescent protein (rMVA-GFP). In both human peripheral blood mononuclear cells and mouse lung explants, rMVA-GFP predominantly infected antigen presenting cells. Subsequent in vivo experiments performed in mice, ferrets and non-human primates indicated that preferential targeting of dendritic cells and alveolar macrophages was observed after respiratory administration, although subtle differences were observed between the respective animal species. Following intramuscular injection, rMVA-GFP was detected in interdigitating cells between myocytes, but also in myocytes themselves. These data are important in advancing our understanding of the basis for the immunogenicity of MVA-based vaccines and aid rational vaccine design and delivery strategies.


Subject(s)
Antigen-Presenting Cells/immunology , Leukocytes, Mononuclear/immunology , Vaccinia virus/immunology , Viral Vaccines/immunology , Animals , Dendritic Cells/immunology , Dendritic Cells/metabolism , Dendritic Cells/virology , Ferrets , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/virology , Macaca fascicularis , Macrophages, Alveolar/immunology , Macrophages, Alveolar/metabolism , Macrophages, Alveolar/virology , Mice , Microscopy, Confocal , Muscle Cells/immunology , Muscle Cells/metabolism , Muscle Cells/virology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Vaccinia virus/genetics , Vaccinia virus/physiology
7.
J Virol ; 90(22): 10209-10219, 2016 Nov 15.
Article in English | MEDLINE | ID: mdl-27581985

ABSTRACT

Due to antigenic drift of influenza viruses, seasonal influenza vaccines need to be updated annually. These vaccines are based on predictions of strains likely to circulate in the next season. However, vaccine efficacy is greatly reduced in the case of a mismatch between circulating and vaccine strains. Furthermore, novel antigenically distinct influenza viruses are introduced into the human population from animal reservoirs occasionally and may cause pandemic outbreaks. To dampen the impact of seasonal and pandemic influenza, vaccines that induce broadly protective and long-lasting immunity are preferred. Because influenza virus-specific CD8+ T cells are directed mainly against relatively conserved internal proteins, like nucleoprotein (NP), they are highly cross-reactive and afford protection against infection with antigenically distinct influenza virus strains, so-called heterosubtypic immunity. Here, we used modified vaccinia virus Ankara (MVA) as a vaccine vector for the induction of influenza virus NP-specific CD8+ T cells. To optimize the induction of CD8+ T cell responses, we made several modifications to NP, aiming at retaining the protein in the cytosol or targeting it to the proteasome. We hypothesized that these strategies would increase antigen processing and presentation and thus improve the induction of CD8+ T cell responses. We showed that NP with increased degradation rates improved CD8+ T cell activation in vitro if the amount of antigen was limited or if CD8+ T cells were of low functional avidity. However, after immunization of C57BL/6 mice, no differences were detected between modified NP and wild-type NP (NPwt), since NPwt already induced optimal CD8+ T cell responses. IMPORTANCE: Due to the continuous antigenic drift of seasonal influenza viruses and the threat of a novel pandemic, there is a great need for the development of novel influenza vaccines that offer broadly protective immunity against multiple subtypes. CD8+ T cells can provide immunity against multiple subtypes of influenza viruses by the recognition of relatively conserved internal antigens. In this study, we aimed at optimizing the CD8+ T cell response to influenza A virus by making modifications to influenza A virus nucleoprotein (NP) expressed from the modified vaccinia virus Ankara (MVA) vaccine vector. These modifications resulted in increased antigen degradation, thereby producing elevated levels of peptides that can be presented on major histocompatibility complex (MHC) class I molecules to CD8+ T cells. Although we were unable to increase the NP-specific immune response in the mouse strain used, this approach may have benefits for vaccine development using less-immunogenic proteins.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Influenza A virus/immunology , Influenza A virus/metabolism , Lymphocyte Activation/immunology , RNA-Binding Proteins/metabolism , Viral Core Proteins/metabolism , Animals , Antibodies, Viral/metabolism , Antigens, Viral/immunology , Cell Line , Cell Line, Tumor , Chickens , Cross Reactions/immunology , Dogs , Female , HeLa Cells , Humans , Influenza Vaccines/immunology , Madin Darby Canine Kidney Cells , Mice , Mice, Inbred C57BL , Nucleocapsid Proteins , Orthomyxoviridae Infections/virology , Proteolysis , RNA-Binding Proteins/immunology , Vaccination/methods , Vaccinia virus/immunology , Viral Core Proteins/immunology
8.
Expert Rev Vaccines ; 14(10): 1299-301, 2015.
Article in English | MEDLINE | ID: mdl-26104835

ABSTRACT

Currently used influenza vaccines are only effective when the vaccine strains match the epidemic strains antigenically. To this end, seasonal influenza vaccines must be updated almost annually. Furthermore, seasonal influenza vaccines fail to afford protection against antigenically distinct pandemic influenza viruses. Because of an ever-present threat of the next influenza pandemic and the continuous emergence of drift variants of seasonal influenza A viruses, there is a need for an universal influenza vaccine that induces protective immunity against all influenza A viruses. Here, we summarize some of the efforts that are ongoing to develop universal influenza vaccines.


Subject(s)
Antibodies, Viral/immunology , Cross Protection/immunology , Influenza Vaccines/immunology , Influenza, Human/immunology , Influenza, Human/prevention & control , Antigenic Variation/immunology , Antigens, Viral/immunology , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Humans , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H3N2 Subtype/immunology , Pandemics/prevention & control , Viral Matrix Proteins/immunology
9.
Vaccine ; 33(4): 500-6, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25498210

ABSTRACT

Since inactivated influenza vaccines mainly confer protective immunity by inducing strain-specific antibodies to the viral hemagglutinin, these vaccines only afford protection against infection with antigenically matching influenza virus strains. Due to the continuous emergence of antigenic drift variants of seasonal influenza viruses and the inevitable future emergence of pandemic influenza viruses, there is considerable interest in the development of influenza vaccines that induce broader protective immunity. It has long been recognized that influenza virus-specific CD8(+) T cells directed to epitopes located in the relatively conserved internal proteins can cross-react with various subtypes of influenza A virus. This implies that these CD8(+) T cells, induced by prior influenza virus infections or vaccinations, could afford heterosubtypic immunity. Furthermore, influenza virus-specific CD4(+) T cells have been shown to be important in protection from infection, either via direct cytotoxic effects or indirectly by providing help to B cells and CD8(+) T cells. In the present paper, we review the induction of virus-specific T cell responses by influenza virus infection and the role of virus-specific CD4(+) and CD8(+) T cells in viral clearance and conferring protection from subsequent infections with homologous or heterologous influenza virus strains. Furthermore, we discuss vector-based vaccination strategies that aim at the induction of a cross-reactive virus-specific T cell response.


Subject(s)
Antigens, Viral/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cross Protection , Influenza, Human/immunology , Influenza, Human/virology , Orthomyxoviridae/immunology , Antigens, Viral/genetics , Conserved Sequence , Drug Discovery/trends , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/immunology , Humans , Influenza Vaccines/genetics , Influenza Vaccines/immunology , Orthomyxoviridae/genetics
10.
Viruses ; 6(7): 2735-61, 2014 Jul 17.
Article in English | MEDLINE | ID: mdl-25036462

ABSTRACT

Respiratory viruses infections caused by influenza viruses, human parainfluenza virus (hPIV), respiratory syncytial virus (RSV) and coronaviruses are an eminent threat for public health. Currently, there are no licensed vaccines available for hPIV, RSV and coronaviruses, and the available seasonal influenza vaccines have considerable limitations. With regard to pandemic preparedness, it is important that procedures are in place to respond rapidly and produce tailor made vaccines against these respiratory viruses on short notice. Moreover, especially for influenza there is great need for the development of a universal vaccine that induces broad protective immunity against influenza viruses of various subtypes. Modified Vaccinia Virus Ankara (MVA) is a replication-deficient viral vector that holds great promise as a vaccine platform. MVA can encode one or more foreign antigens and thus functions as a multivalent vaccine. The vector can be used at biosafety level 1, has intrinsic adjuvant capacities and induces humoral and cellular immune responses. However, there are some practical and regulatory issues that need to be addressed in order to develop MVA-based vaccines on short notice at the verge of a pandemic. In this review, we discuss promising novel influenza virus vaccine targets and the use of MVA for vaccine development against various respiratory viruses.


Subject(s)
Antibodies, Viral/biosynthesis , Influenza, Human/prevention & control , Respiratory Tract Infections/prevention & control , Vaccinia virus/genetics , Viral Vaccines/immunology , Animals , Cross Protection , Genetic Vectors , Humans , Influenza, Human/immunology , Influenza, Human/virology , Orthomyxoviridae/drug effects , Orthomyxoviridae/immunology , Orthomyxoviridae/pathogenicity , Respiratory Syncytial Viruses/drug effects , Respiratory Syncytial Viruses/immunology , Respiratory Syncytial Viruses/pathogenicity , Respiratory Tract Infections/immunology , Respiratory Tract Infections/virology , Respirovirus/drug effects , Respirovirus/immunology , Respirovirus/pathogenicity , Severe acute respiratory syndrome-related coronavirus/drug effects , Severe acute respiratory syndrome-related coronavirus/immunology , Severe acute respiratory syndrome-related coronavirus/pathogenicity , Vaccines, Synthetic , Vaccinia virus/immunology , Viral Vaccines/administration & dosage , Viral Vaccines/genetics
11.
Cell Host Microbe ; 14(5): 510-21, 2013 Nov 13.
Article in English | MEDLINE | ID: mdl-24237697

ABSTRACT

Several enveloped viruses exploit host pathways, such as the cellular endosomal sorting complex required for transport (ESCRT) machinery, for their assembly and release. The influenza A virus (IAV) matrix protein binds to the ESCRT-I complex, although the involvement of early ESCRT proteins such as Tsg101 in IAV trafficking remain to be established. We find that Tsg101 can facilitate IAV trafficking, but this is effectively restricted by the interferon (IFN)-stimulated protein ISG15. Cytosol from type I IFN-treated cells abolished IAV hemagglutinin (HA) transport to the cell surface in infected semi-intact cells. This inhibition required Tsg101 and could be relieved with deISGylases. Tsg101 is itself ISGylated in IFN-treated cells. Upon infection, intact Tsg101-deficient cells obtained by CRISPR-Cas9 genome editing were defective in the surface display of HA and for infectious virion release. These data support the IFN-induced generation of a Tsg101- and ISG15-dependent checkpoint in the secretory pathway that compromises influenza virus release.


Subject(s)
Cytokines/metabolism , DNA-Binding Proteins/metabolism , Endosomal Sorting Complexes Required for Transport/metabolism , Glycoproteins/metabolism , Golgi Apparatus/metabolism , Host-Pathogen Interactions , Influenza A virus/immunology , Interferon Type I/immunology , Transcription Factors/metabolism , Ubiquitins/metabolism , Animals , Humans , Influenza A virus/physiology , Virus Release
12.
Nature ; 503(7476): 406-9, 2013 Nov 21.
Article in English | MEDLINE | ID: mdl-24141948

ABSTRACT

Influenza A virus-specific B lymphocytes and the antibodies they produce protect against infection. However, the outcome of interactions between an influenza haemagglutinin-specific B cell via its receptor (BCR) and virus is unclear. Through somatic cell nuclear transfer we generated mice that harbour B cells with a BCR specific for the haemagglutinin of influenza A/WSN/33 virus (FluBI mice). Their B cells secrete an immunoglobulin gamma 2b that neutralizes infectious virus. Whereas B cells from FluBI and control mice bind equivalent amounts of virus through interaction of haemagglutinin with surface-disposed sialic acids, the A/WSN/33 virus infects only the haemagglutinin-specific B cells. Mere binding of virus is not sufficient for infection of B cells: this requires interactions of the BCR with haemagglutinin, causing both disruption of antibody secretion and FluBI B-cell death within 18 h. In mice infected with A/WSN/33, lung-resident FluBI B cells are infected by the virus, thus delaying the onset of protective antibody release into the lungs, whereas FluBI cells in the draining lymph node are not infected and proliferate. We propose that influenza targets and kills influenza-specific B cells in the lung, thus allowing the virus to gain purchase before the initiation of an effective adaptive response.


Subject(s)
B-Lymphocytes/immunology , B-Lymphocytes/virology , Orthomyxoviridae/physiology , Receptors, Antigen, B-Cell/immunology , Animals , Antibodies/immunology , Antibodies/metabolism , Antibody Specificity/immunology , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Cell Death , Female , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Immunoglobulin G/immunology , Immunoglobulin G/metabolism , Lung/cytology , Lung/immunology , Lung/metabolism , Lung/virology , Lymph Nodes/cytology , Lymph Nodes/immunology , Male , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Neutralization Tests , Nuclear Transfer Techniques , Orthomyxoviridae/pathogenicity , Receptors, Antigen, B-Cell/metabolism , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL