Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 117
Filter
1.
Int J Biol Macromol ; 182: 648-658, 2021 Jul 01.
Article in English | MEDLINE | ID: mdl-33862071

ABSTRACT

Vaccination is the most effective means of controlling infectious disease-related morbidity and mortality. However, due to low immunogenicity of viral antigens, nanomedicine as a new opportunity in new generation of vaccine advancement attracted researcher encouragement. Virosome is a lipidic nanomaterial emerging as FDA approved nanocarriers with promising bioinspiration and biomimetic potency against viral infections. Virosome surface modification with critical viral fusion proteins is the cornerstone of vaccine development. Surface antigens at virosomes innovatively interact with targeted receptors on host cells that evoke humoral or cellular immune responses through antibody-producing B cell and internalization by endocytosis-mediated pathways. To date, several nanovaccine based on virosome formulations have been commercialized against widespread and life-threatening infections. Recently, Great efforts were made to fabricate a virosome-based vaccine platform against a new severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus. Thus, this review provides a novel overview of the virosome based nanovaccine production, properties, and application on the viral disease, especially its importance in SARS-CoV-2 vaccine discovery.


Subject(s)
Biomimetic Materials/therapeutic use , COVID-19 Vaccines/therapeutic use , COVID-19/prevention & control , SARS-CoV-2/immunology , Virosomes/therapeutic use , Animals , COVID-19/immunology , COVID-19 Vaccines/immunology , Humans , Virosomes/immunology
2.
Biomed Res Int ; 2021: 8879277, 2021.
Article in English | MEDLINE | ID: mdl-33575353

ABSTRACT

Newcastle disease (ND) is a highly fatal, infectious, viral disease, and despite immunization with live and inactivated vaccines, the disease is still endemic, causing heavy morbidity and mortality leading to huge economic losses to the poultry industry in Pakistan. Therefore, the present study was aimed for the first time in the country at using novel virosomal technology to develop the ND vaccine using an indigenous highly virulent strain of the virus. ND virosome was prepared using Triton X-100, and SM2 Bio-Beads were used to remove the detergent and reconstitute the viral membrane into virosome. Confirmation was done by transmission electron microscopy and protein analysis by SDS-PAGE. In vitro cell adhesion property was observed by incorporating green fluorescent protein (GFP), producing plasmid into virosome and in vitro cell culture assay. Sterility, safety, and stability of the vaccine were tested before in vivo evaluation of immunogenicity and challenge protection study in commercial broiler. The virosome vaccine was administered (30 µg/bird) at days 7 and 14 through the intranasal route in comparison with commercially available live and inactivated ND vaccines. Results revealed significantly high (p < 0.05) and clinically protective hemagglutination inhibition (HI) antibody titers at 7, 14, 21, and 28 days postimmunization with the virosome vaccine in comparison to the negative control. The GMTs were comparable to live and inactivated vaccines with nonsignificant (p > 0.05) differences throughout the experiment. Antibody levels increased in all vaccinated groups gradually from the 7th day and were maximum at 28th-day postvaccination. In the virosome-administered group, GMT was 83.18 and 77.62 at 21st and 28th-days postvaccination, respectively. Challenge revealed 100%, 90%, and 80% protection in virosome, live, and inactivated vaccinated groups, respectively. Under given experimental conditions, we can conclude that ND virosome vaccine prepared from the indigenous virus was found to be safe and immunogenic.


Subject(s)
Newcastle Disease , Poultry Diseases , Vaccines, Virosome , Animals , Antibodies, Viral/blood , Antibodies, Viral/immunology , Chickens , Newcastle Disease/immunology , Newcastle Disease/prevention & control , Newcastle disease virus/genetics , Newcastle disease virus/immunology , Pakistan , Poultry Diseases/immunology , Poultry Diseases/prevention & control , Vaccines, Virosome/chemistry , Vaccines, Virosome/immunology , Vaccines, Virosome/metabolism , Virosomes/immunology
3.
J Immunol ; 203(2): 441-452, 2019 07 15.
Article in English | MEDLINE | ID: mdl-31182479

ABSTRACT

Protease-activated receptor 2 (PAR-2) is expressed in various tissues, including lung, and when activated, promotes inflammation, differentiation, and migration of dendritic cells. We found that combining influenza virosomes containing hemagglutinin and neuraminidase with a PAR-2 agonist peptide (PAR-2AP) in an intranasal prime boost approach increased survival of mice challenged weeks later with lethal influenza virus over that by virosome or PAR-2AP prime boost alone. No weight loss occurred from influenza challenge after virosome-plus-PAR-2AP prime boost compared with either virosomes or PAR-2AP alone. Thus, virosomes plus PAR-2AP prevented morbidity as well as mortality. Through adoptive transfer, CD8+ lung T cells but not CD4+ T cells from virosomes plus PAR-2AP-primed mice protected from lethal influenza virus challenge and enhanced survival with less weight loss and faster recovery. Virosome-plus-PAR-2AP prime boost resulted in greater percentages of T effector memory phenotype cells (Tem) in lung, and higher frequencies of CD8 Tem and T central memory cells displayed effector functions in response to virus challenge in vivo. Virosome-plus-PAR-2AP prime boost also resulted in greater percentages of Ag-specific CD8+ T cells, both Tem and T central memory cells, in lungs of animals subsequently challenged with live influenza virus. Our findings indicate that PAR-2AP, a short peptide, may be a new and useful mucosal adjuvant.


Subject(s)
Adjuvants, Immunologic/pharmacology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Immunologic Memory/immunology , Receptor, PAR-2/agonists , Virosomes/immunology , Adoptive Transfer/methods , Animals , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , Cell Line , Dogs , Female , Immunologic Memory/drug effects , Lung/immunology , Lung/virology , Madin Darby Canine Kidney Cells , Mice , Mice, Inbred C57BL , Orthomyxoviridae/immunology , Virosomes/drug effects
4.
Viruses ; 11(6)2019 06 14.
Article in English | MEDLINE | ID: mdl-31207978

ABSTRACT

Rabbit haemorrhagic disease virus (RHDV) type 2 (GI.2/RHDV2/b) is an emerging pathogen in wild rabbits and in domestic rabbits vaccinated against RHDV (GI.1). Here we report the genome sequence of a contemporary RHDV2 isolate from the Netherlands and investigate the immunogenicity of virus-like particles (VLPs) produced in insect cells. RHDV2 RNA was isolated from the liver of a naturally infected wild rabbit and the complete viral genome sequence was assembled from sequenced RT-PCR products. Phylogenetic analysis based on the VP60 capsid gene demonstrated that the RHDV2 NL2016 isolate clustered with other contemporary RHDV2 strains. The VP60 gene was cloned in a baculovirus expression vector to produce VLPs in Sf9 insect cells. Density-gradient purified RHDV2 VLPs were visualized by transmission electron microscopy as spherical particles of around 30 nm in diameter with a morphology resembling authentic RHDV. Immunization of rabbits with RHDV2 VLPs resulted in high production of serum antibodies against VP60, and the production of cytokines (IFN-γ and IL-4) was significantly elevated in the immunized rabbits compared to the control group. The results demonstrate that the recombinant RHDV2 VLPs are highly immunogenic and may find applications in serological detection assays and might be further developed as a vaccine candidate to protect domestic rabbits against RHDV2 infection.


Subject(s)
Caliciviridae Infections/veterinary , Hemorrhagic Disease Virus, Rabbit/immunology , Vaccines, Virus-Like Particle/immunology , Virosomes/immunology , Animals , Antibodies, Viral/blood , Antibody Formation , Baculoviridae , Caliciviridae Infections/immunology , Caliciviridae Infections/prevention & control , Cluster Analysis , Cytokines/analysis , Genetic Vectors , Hemorrhagic Disease Virus, Rabbit/classification , Hemorrhagic Disease Virus, Rabbit/genetics , Hemorrhagic Disease Virus, Rabbit/isolation & purification , Immunity, Cellular , Netherlands , Phylogeny , Rabbits , Sequence Analysis, DNA , Sequence Homology , Sf9 Cells , Spodoptera , Vaccines, Virus-Like Particle/administration & dosage , Vaccines, Virus-Like Particle/genetics , Virosomes/genetics
5.
Diagn Microbiol Infect Dis ; 91(3): 233-238, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29530349

ABSTRACT

West Nile virus is a globally spread zoonotic arbovirus. The laboratory diagnosis of WNV infection relies on virus identification by RT-PCR or on specific antibody detection by serological tests, such as ELISA or virus-neutralization. These methods usually require a preparation of the whole virus as antigen, entailing biosafety issues and therefore requiring BSL-3 facilities. For this reason, recombinant antigenic structures enabling effective antibody recognition comparable to that of the native virions, would be advantageous as diagnostic reagents. WNV virions are enveloped spherical particles made up of 3 structural proteins (C, capsid; M, membrane and E, envelope) enclosing the viral RNA. This study describes the co-expression of these 3 proteins yielding non-infectious virus-like particles (VLPs) and the results of the initial assessment of these VLPs, used instead of the whole virus, that were shown to perform correctly in two different ELISAs for WNV diagnosis.


Subject(s)
Antibodies, Viral/blood , Antigens, Viral/immunology , Horse Diseases/diagnosis , Virosomes/immunology , West Nile Fever/veterinary , Animals , Antigens, Viral/genetics , Cell Line , Gene Expression , Horses , Insecta , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Viral Structural Proteins/genetics , Viral Structural Proteins/immunology , Virosomes/genetics , Virosomes/isolation & purification , West Nile Fever/diagnosis , West Nile virus/genetics , West Nile virus/immunology
6.
Biotechnol J ; 13(4): e1700645, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29278302

ABSTRACT

Induction of CD8+ cytotoxic T cells (CTLs) to conserved internal influenza antigens, such as nucleoprotein (NP), is a promising strategy for the development of cross-protective influenza vaccines. However, influenza NP protein alone cannot induce CTL immunity due to its low capacity to activate antigen-presenting cells (APCs) and get access to the MHC class I antigen processing pathway. To facilitate the generation of NP-specific CTL immunity the authors develop a novel influenza vaccine consisting of virosomes with the Toll-like receptor 4 (TLR4) ligand monophosphoryl lipid A (MPLA) and the metal-ion-chelating lipid DOGS-NTA-Ni incorporated in the membrane. In vitro, virosomes with incorporated MPLA induce stronger activation of APCs than unadjuvanted virosomes. Virosomes modified with DOGS-NTA-Ni show high conjugation efficacy for his-tagged proteins and facilitate efficient uptake of conjugated proteins by APCs. Immunization of mice with MPLA-adjuvanted virosomes with attached NP results in priming of NP-specific CTLs while MPLA-adjuvanted virosomes with admixed NP are inefficient in priming CTLs. Both vaccines induce equally high titers of NP-specific antibodies. When challenged with heterosubtypic influenza virus, mice immunized with virosomes with attached or admixed NP are protected from severe weight loss. Yet, unexpectedly, they show more weight loss and more severe disease symptoms than mice immunized with MPLA-virosomes without NP. Taken together, these results indicate that virosomes with conjugated antigen and adjuvant incorporated in the membrane are effective in priming of CTLs and eliciting antigen-specific antibody responses in vivo. However, for protection from influenza infection NP-specific immunity appears not to be advantageous.


Subject(s)
Adjuvants, Immunologic/chemistry , Lipid A/analogs & derivatives , RNA-Binding Proteins/immunology , Viral Core Proteins/immunology , Virosomes/immunology , Animals , CD8-Positive T-Lymphocytes/metabolism , Influenza Vaccines/immunology , Lipid A/chemistry , Mice , Nickel/chemistry , Nucleocapsid Proteins , RAW 264.7 Cells , T-Lymphocytes, Cytotoxic/metabolism , Virosomes/chemistry
7.
J Med Virol ; 90(4): 671-676, 2018 04.
Article in English | MEDLINE | ID: mdl-29236287

ABSTRACT

Noroviruses (NoVs) are increasingly recognized as the leading cause of acute non-bacterial gastroenteritis worldwide. To screen for NoV-specific monoclonal antibodies (mAbs) with wide spectrum binding activities that could be used for the development of NoV-related detection reagents, we immunized mice with a combination of virus like particles (VLPs) derived from eight different genotypes (two from genogroup I and six from genogroup II), of which two (GI.7 and GII.2) were newly produced VLPs. Indirect enzyme-linked immunosorbent assay (ELISA) confirmed that two mAbs (8D8 and 10B11) bound to all eight major capsid proteins (VP1) with varied binding abilities. Epitope mapping using short peptides covering the N-terminal half of GII.3 VP1 indicated that the binding site of mAb 8D8 was located between amino acid 31 and 60. Multiple amino acid sequence alignment of VP1 suggested that this site harbors conservative sequences across all genogroups. Indirect and sandwich ELISA indicated that mAb 8D8 was unable bind intact VLPs. In summary, we successfully produced GI.7 and GII.2 VLPs using recombinant baculovirus expression system and a cross-reactive mAb by immunizing mice with eight different VLPs that might be useful in the studying and detecting NoVs.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Viral/immunology , Norovirus/immunology , Animals , Antibodies, Monoclonal/isolation & purification , Antibodies, Viral/isolation & purification , Baculoviridae/genetics , Binding Sites , Enzyme-Linked Immunosorbent Assay , Epitope Mapping , Genetic Vectors , Genotype , Mice, Inbred BALB C , Norovirus/genetics , Protein Binding , Virosomes/genetics , Virosomes/immunology
8.
Virology ; 511: 114-122, 2017 11.
Article in English | MEDLINE | ID: mdl-28843813

ABSTRACT

Recombinant proteins produced by baculovirus (BV) expression systems contain residual BV after crude purification. We studied adjuvant effect of BV on antibody and T cell responses against two model antigens, monomeric ovalbumin (OVA) protein and oligomeric norovirus (NoV) virus-like particles (VLPs). BALB/c mice were immunized intradermally with OVA alone or OVA formulated with live or inactivated BV, and VLP formulations comprised of chromatographically purified NoV GII.4 VLPs alone or mixed with BV, or of crude purified VLPs containing BV impurities from expression system. Live BV improved immunogenicity of NoV VLPs, sparing VLP dose up to 10-fold. Moreover, soluble OVA protein induced IgG2a antibodies and T cell response only when co-administered with live BV. BV adjuvant effect was completely abrogated by removal or inactivation of BV. These findings support the usage of crude purified proteins containing residual BV as vaccine antigens.


Subject(s)
Adjuvants, Immunologic/metabolism , B-Lymphocytes/immunology , Baculoviridae/metabolism , Norovirus/immunology , Ovalbumin/immunology , T-Lymphocytes/immunology , Virosomes/immunology , Animals , Injections, Intradermal , Mice, Inbred BALB C , Ovalbumin/administration & dosage , Virosomes/administration & dosage
9.
J Virol ; 91(16)2017 08 15.
Article in English | MEDLINE | ID: mdl-28566373

ABSTRACT

Human papillomavirus (HPV) infection is the most common viral infection of the reproductive tract, with virtually all cases of cervical cancer being attributable to infection by oncogenic HPVs. However, the exact mechanism and receptors used by HPV to infect epithelial cells are controversial. The current entry model suggests that HPV initially attaches to heparan sulfate proteoglycans (HSPGs) at the cell surface, followed by conformational changes, cleavage by furin convertase, and subsequent transfer of the virus to an as-yet-unidentified high-affinity receptor. In line with this model, we established an in vitro infection system using the HSPG-deficient cell line pgsD677 together with HPV16 pseudovirions (HPV16-PsVs). While pgsD677 cells were nonpermissive for untreated HPV16-PsVs, furin cleavage of the particles led to a substantial increase in infection. Biochemical pulldown assays followed by mass spectrometry analysis showed that furin-precleaved HPV16-PsVs specifically interacted with surface-expressed vimentin on pgsD677 cells. We further demonstrated that both furin-precleaved and uncleaved HPV16-PsVs colocalized with surface-expressed vimentin on pgsD677, HeLa, HaCaT, and NIKS cells, while binding of incoming viral particles to soluble vimentin protein before infection led to a substantial decrease in viral uptake. Interestingly, decreasing cell surface vimentin by small interfering RNA (siRNA) knockdown in HeLa and NIKS cells significantly increased HPV16-PsV infectious internalization, while overexpression of vimentin had the opposite effect. The identification of vimentin as an HPV restriction factor enhances our understanding of the initial steps of HPV-host interaction and may lay the basis for the design of novel antiviral drugs preventing HPV internalization into epithelial cells.IMPORTANCE Despite HPV being a highly prevalent sexually transmitted virus causing significant disease burden worldwide, particularly cancer of the cervix, cell surface events preceding oncogenic HPV internalization are poorly understood. We herein describe the identification of surface-expressed vimentin as a novel molecule not previously implicated in the infectious internalization of HPV16. Contrary to our expectations, vimentin was found to act not as a receptor but rather as a restriction factor dampening the initial steps of HPV16 infection. These results importantly contribute to our current understanding of the molecular events during the infectious internalization of HPV16 and open a new direction in the development of alternative drugs to prevent HPV infection.


Subject(s)
Epithelial Cells/virology , Host-Pathogen Interactions , Human papillomavirus 16/immunology , Human papillomavirus 16/physiology , Vimentin/metabolism , Virosomes/immunology , Virus Internalization , Cell Line , Centrifugation , Humans , Mass Spectrometry , Protein Interaction Mapping , Proteomics
10.
J Virol ; 91(14)2017 07 15.
Article in English | MEDLINE | ID: mdl-28446665

ABSTRACT

HIV-1 is rare among viruses for having a low number of envelope glycoprotein (Env) spikes per virion, i.e., ∼7 to 14. This exceptional feature has been associated with avoidance of humoral immunity, i.e., B cell activation and antibody neutralization. Virus-like particles (VLPs) with increased density of Env are being pursued for vaccine development; however, these typically require protein engineering that alters Env structure. Here, we used instead a strategy that targets the producer cell. We employed fluorescence-activated cell sorting (FACS) to sort for cells that are recognized by trimer cross-reactive broadly neutralizing antibody (bnAb) and not by nonneutralizing antibodies. Following multiple iterations of FACS, cells and progeny virions were shown to display higher levels of antigenically correct Env in a manner that correlated between cells and cognate virions (P = 0.027). High-Env VLPs, or hVLPs, were shown to be monodisperse and to display more than a 10-fold increase in spikes per particle by electron microscopy (average, 127 spikes; range, 90 to 214 spikes). Sequencing revealed a partial truncation in the C-terminal tail of Env that had emerged in the sort; however, iterative rounds of "cell factory" selection were required for the high-Env phenotype. hVLPs showed greater infectivity than standard pseudovirions but largely similar neutralization sensitivity. Importantly, hVLPs also showed superior activation of Env-specific B cells. Hence, high-Env HIV-1 virions, obtained through selection of producer cells, represent an adaptable platform for vaccine design and should aid in the study of native Env.IMPORTANCE The paucity of spikes on HIV is a unique feature that has been associated with evasion of the immune system, while increasing spike density has been a goal of vaccine design. Increasing the density of Env by modifying it in various ways has met with limited success. Here, we focused instead on the producer cell. Cells that stably express HIV spikes were screened on the basis of high binding by bnAbs and low binding by nonneutralizing antibodies. Levels of spikes on cells correlated well with those on progeny virions. Importantly, high-Env virus-like particles (hVLPs) were produced with a manifest array of well-defined spikes, and these were shown to be superior in activating desirable B cells. Our study describes HIV particles that are densely coated with functional spikes, which should facilitate the study of HIV spikes and their development as immunogens.


Subject(s)
HIV-1/ultrastructure , Virion/ultrastructure , Virosomes/ultrastructure , env Gene Products, Human Immunodeficiency Virus/metabolism , B-Lymphocytes/immunology , Cells, Cultured , HIV-1/growth & development , HIV-1/immunology , Humans , Microscopy, Electron, Transmission , Neutralization Tests , Virosomes/immunology , Virosomes/metabolism , env Gene Products, Human Immunodeficiency Virus/genetics , env Gene Products, Human Immunodeficiency Virus/immunology
11.
Vaccine ; 35(10): 1448-1454, 2017 03 07.
Article in English | MEDLINE | ID: mdl-28190741

ABSTRACT

PURPOSE: This was a 20-year follow-up study to assess long-term persistence of protective antibody levels against the hepatitis A virus (HAV) in healthy participants vaccinated with 2 doses of inactivated hepatitis A vaccine (Epaxal®) between 1992 and 1995. METHODS: Blood samples for anti-HAV antibody concentrations were obtained during a follow-up visit 20years after vaccination and were analyzed in parallel with samples still available from previous visits using AxSYM® HAVAB 2.0 assay. RESULTS: Mean (SD) age of the participants was 44.71 (3.905) years at year 20 follow-up (N=95). Participants completing 0/12-month Epaxal® immunization regimen (N=94) had seroprotection rate of 100% (95% CI: 96.2, 100.0) with ⩾10mIU/mL seropositivity cut-off and 98.9% (95% CI: 94.2, 100.0) with ⩾20mIU/mL cut-off. With ⩾10mIU/mL cut-off, the estimated median duration of protection was 77.3years (95% CI: 71.8, 83.5) with 95% of the vaccinated participants predicted to be protected for at least 41.5years. At ⩾20mIU/mL cut-off, the estimated median duration of protection was 64.8years (95% CI: 60.1, 68.4) with 95% of the vaccinated participants predicted to be protected for at least 33years. Anti-HAV antibody geometric mean concentrations were higher in women (277.9; 95% CI: 217.7, 354.7) than in men (167.7; 95% CI: 125.2, 224.6). CONCLUSION: The data from this 20-year follow-up study confirm previous observations that two doses of Epaxal® provide protection against hepatitis A infection for at least 30years in over 95% of healthy participants.


Subject(s)
Hepatitis A Antibodies/blood , Hepatitis A Vaccines/immunology , Hepatitis A virus/immunology , Immunity, Humoral , Virosomes/immunology , Adolescent , Adult , Female , Follow-Up Studies , Healthy Volunteers , Hepatitis A Vaccines/administration & dosage , Humans , Immunization Schedule , Male , Prospective Studies , Time Factors , Young Adult
12.
J Virol Methods ; 243: 146-150, 2017 05.
Article in English | MEDLINE | ID: mdl-28131868

ABSTRACT

Although porcine circovirus type 2 (PCV2) virus-like particles (VLPs) have been successfully harvested from various protein expression systems, conditions to promote their stability and integrity during long-term storage have not been well defined since only the intact VLPs, instead of the monomeric capsid protein (Cap), can induce neutralizing antibodies in pigs in previous studies. In this study, freshly prepared PCV2 VLPs were stored in several media (various concentrations of NaCl, sorbitol, sucrose and trehalose) at three temperatures (4°C, -20°C and -80°C) and their stability and integration were evaluated after 7 month. Addition of 15% trehalose in storage buffer promoted long-term preservation of PCV2 VLPs. In contrast, storage buffer with 5% osmolytes (sucrose, trehalose and sorbitol) did not confer stabilization for long-term storage. These refined storage conditions for stabilization of PCV2 VLPs should enhance their use in vaccines.


Subject(s)
Circovirus/ultrastructure , Drug Storage/methods , Virosomes/ultrastructure , Animals , Circovirus/immunology , Cryoprotective Agents , Microscopy, Electron, Transmission , Sodium Chloride/metabolism , Swine , Temperature , Viral Vaccines/immunology , Virosomes/immunology
13.
J Virol ; 90(19): 8720-8, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27440895

ABSTRACT

UNLABELLED: Ebola virus (EBOV) is a highly contagious lethal pathogen. As a biosafety level 4 (BSL-4) agent, however, EBOV is restricted to costly BSL-4 laboratories for experimentation, thus significantly impeding the evaluation of EBOV vaccines and drugs. Here, we report an EBOV-like particle (EBOVLP)-based luciferase reporter system that enables the evaluation of anti-EBOV agents in vitro and in vivo outside BSL-4 facilities. Cotransfection of HEK293T cells with four plasmids encoding the proteins VP40, NP, and GP of EBOV and firefly luciferase (Fluc) resulted in the production of Fluc-containing filamentous particles that morphologically resemble authentic EBOV. The reporter EBOVLP was capable of delivering Fluc into various cultured cells in a GP-dependent manner and was recognized by a conformation-dependent anti-EBOV monoclonal antibody (MAb). Significantly, inoculation of mice with the reporter EBOVLP led to the delivery of Fluc protein into target cells and rapid generation of intense bioluminescence signals that could be blocked by the administration of EBOV neutralizing MAbs. This BSL-4-free reporter system should facilitate high-throughput screening for anti-EBOV drugs targeting viral entry and efficacy testing of candidate vaccines. IMPORTANCE: Ebola virus (EBOV) researches have been limited to costly biosafety level 4 (BSL-4) facilities due to the lack of animal models independent of BSL-4 laboratories. In this study, we reveal that a firefly luciferase-bearing EBOV-like particle (EBOVLP) with typical filamentous EBOV morphology is capable of delivering the reporter protein into murine target cells both in vitro and in vivo Moreover, we demonstrate that the reporter delivery can be inhibited both in vitro and in vivo by a known anti-EBOV protective monoclonal antibody, 13C6. Our work provides a BSL-4-free system that can facilitate the in vivo evaluation of anti-EBOV antibodies, drugs, and vaccines. The system may also be useful for mechanistic study of the viral entry process.


Subject(s)
Antiviral Agents/isolation & purification , Ebola Vaccines/immunology , Ebolavirus/drug effects , Endocytosis , Genes, Reporter , Luciferases/analysis , Virosomes/metabolism , Animals , Antiviral Agents/pharmacology , Drug Evaluation, Preclinical/methods , Ebolavirus/genetics , Luciferases/genetics , Mice , Virosomes/drug effects , Virosomes/genetics , Virosomes/immunology
14.
J Virol ; 90(17): 7618-27, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27279622

ABSTRACT

UNLABELLED: ZMapp, a cocktail of three monoclonal antibodies (MAbs; c2G4, c4G7, and c13C6) against the ebolavirus (EBOV) glycoprotein (GP), shows promise for combatting outbreaks of EBOV, as occurred in West Africa in 2014. Prior studies showed that Fabs from these MAbs bind a soluble EBOV GP ectodomain and that MAbs c2G4 and c4G7, but not c13C6, neutralize infections in cell cultures. Using cryo-electron tomography, we extended these findings by characterizing the structures of c2G4, c4G7, and c13C6 IgGs bound to native, full-length GP from the West African 2014 isolate embedded in filamentous viruslike particles (VLPs). As with the isolated ectodomain, c13C6 bound to the glycan cap, whereas c2G4 and c4G7 bound to the base region of membrane-bound GP. The tomographic data suggest that all three MAbs bind with high occupancy and that the base-binding antibodies can potentially bridge neighboring GP spikes. Functional studies indicated that c2G4 and c4G7, but not c13C6, competitively inhibit entry of VLPs bearing EBOV GP into the host cell cytoplasm, without blocking trafficking of VLPs to NPC1(+) endolysosomes, where EBOV fuses. Moreover, c2G4 and c4G7 bind to and can block entry mediated by the primed (19-kDa) form of GP without impeding binding of the C-loop of NPC1, the endolysosomal receptor for EBOV. The most likely mode of action of c2G4 and c4G7 is therefore by inhibiting conformational changes in primed, NPC1-bound GP that initiate fusion between the viral and target membranes, similar to the action of certain broadly neutralizing antibodies against influenza hemagglutinin and HIV Env. IMPORTANCE: The recent West African outbreak of ebolavirus caused the deaths of more than 11,000 individuals. Hence, there is an urgent need to be prepared with vaccines and therapeutics for similar future disasters. ZMapp, a cocktail of three MAbs directed against the ebolavirus glycoprotein, is a promising anti-ebolavirus therapeutic. Using cryo-electron tomography, we provide structural information on how each of the MAbs in this cocktail binds to the ebolavirus glycoprotein as it is displayed-embedded in the membrane and present at high density-on filamentous viruslike particles that recapitulate the surface structure and entry functions of ebolavirus. Moreover, after confirming that two of the MAbs bind to the same region in the base of the glycoprotein, we show that they competitively block the entry function of the glycoprotein and that they can do so after the glycoprotein is proteolytically primed and bound to its intracellular receptor, Niemann-Pick C1. These findings should inform future developments of ebolavirus therapeutics.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Ebolavirus/immunology , Ebolavirus/physiology , Viral Envelope Proteins/immunology , Virus Internalization/drug effects , Antibodies, Monoclonal/metabolism , Antibodies, Neutralizing/metabolism , Electron Microscope Tomography , Protein Binding , Viral Envelope Proteins/metabolism , Virosomes/immunology , Virosomes/metabolism
15.
J Gen Virol ; 97(8): 1865-1876, 2016 08.
Article in English | MEDLINE | ID: mdl-27147296

ABSTRACT

An effective immune response against hepatitis C virus (HCV) requires the early development of multi-specific class 1 CD8+ and class II CD4+ T-cells together with broad neutralizing antibody responses. We have produced mammalian-cell-derived HCV virus-like particles (VLPs) incorporating core, E1 and E2 of HCV genotype 1a to produce such immune responses. Here we describe the biochemical and morphological characterization of the HCV VLPs and study HCV core-specific T-cell responses to the particles. The E1 and E2 glycoproteins in HCV VLPs formed non-covalent heterodimers and together with core protein assembled into VLPs with a buoyant density of 1.22 to 1.28 g cm-3. The HCV VLPs could be immunoprecipited with anti-ApoE and anti-ApoC. On electron microscopy, the VLPs had a heterogeneous morphology and ranged in size from 40 to 80 nm. The HCV VLPs demonstrated dose-dependent binding to murine-derived dendritic cells and the entry of HCV VLPs into Huh7 cells was blocked by anti-CD81 antibody. Vaccination of BALB/c mice with HCV VLPs purified from iodixanol gradients resulted in the production of neutralizing antibody responses while vaccination of humanized MHC class I transgenic mice resulted in the prodution of HCV core-specific CD8+ T-cell responses. Furthermore, IgG purified from the sera of patients chronically infected with HCV genotypes 1a and 3a blocked the binding and entry of the HCV VLPs into Huh7 cells. These results show that our mammalian-cell-derived HCV VLPs induce humoral and HCV-specific CD8+ T-cell responses and will have important implications for the development of a preventative vaccine for HCV.


Subject(s)
Hepacivirus/immunology , Hepatitis C Antibodies/blood , T-Lymphocytes/immunology , Vaccines, Virus-Like Particle/immunology , Animals , Antibodies, Neutralizing/blood , Cell Line , Cells, Cultured , Hepacivirus/genetics , Hepatocytes/virology , Humans , Mice, Inbred BALB C , Mice, Transgenic , Microscopy, Electron , Vaccines, Virus-Like Particle/administration & dosage , Vaccines, Virus-Like Particle/genetics , Vaccines, Virus-Like Particle/isolation & purification , Viral Core Proteins/genetics , Viral Core Proteins/immunology , Viral Core Proteins/metabolism , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology , Viral Envelope Proteins/metabolism , Virosomes/genetics , Virosomes/immunology , Virosomes/metabolism , Virosomes/ultrastructure
16.
J Virol ; 90(3): 1169-77, 2016 02 01.
Article in English | MEDLINE | ID: mdl-26537684

ABSTRACT

UNLABELLED: Chikungunya virus is a positive-stranded RNA alphavirus. Structures of chikungunya virus-like particles in complex with strongly neutralizing antibody Fab fragments (8B10 and 5F10) were determined using cryo-electron microscopy and X-ray crystallography. By fitting the crystallographically determined structures of these Fab fragments into the cryo-electron density maps, we show that Fab fragments of antibody 8B10 extend radially from the viral surface and block receptor binding on the E2 glycoprotein. In contrast, Fab fragments of antibody 5F10 bind the tip of the E2 B domain and lie tangentially on the viral surface. Fab 5F10 fixes the B domain rigidly to the surface of the virus, blocking exposure of the fusion loop on glycoprotein E1 and therefore preventing the virus from becoming fusogenic. Although Fab 5F10 can neutralize the wild-type virus, it can also bind to a mutant virus without inhibiting fusion or attachment. Although the mutant virus is no longer able to propagate by extracellular budding, it can, however, enter the next cell by traveling through junctional complexes without being intercepted by a neutralizing antibody to the wild-type virus, thus clarifying how cell-to-cell transmission can occur. IMPORTANCE: Alphaviral infections are transmitted mainly by mosquitoes. Chikungunya virus (CHIKV), which belongs to the Alphavirus genus, has a wide distribution in the Old World that has expanded in recent years into the Americas. There are currently no vaccines or drugs against alphaviral infections. Therefore, a better understanding of CHIKV and its associated neutralizing antibodies will aid in the development of effective treatments.


Subject(s)
Antibodies, Neutralizing/metabolism , Antibodies, Viral/metabolism , Chikungunya virus/immunology , Chikungunya virus/ultrastructure , Virosomes/immunology , Virosomes/ultrastructure , Chikungunya virus/chemistry , Chikungunya virus/physiology , Cryoelectron Microscopy , Crystallography, X-Ray , Humans , Immunoglobulin Fab Fragments/metabolism , Models, Molecular , Protein Binding , Virosomes/chemistry , Virus Attachment
17.
J Virol ; 90(5): 2706-9, 2015 Dec 16.
Article in English | MEDLINE | ID: mdl-26676791

ABSTRACT

Nipah virus (NiV) is a highly pathogenic paramyxovirus that causes pronounced infection of brain endothelia and central nervous system (CNS) inflammation. Using primary porcine brain microvascular endothelial cells, we showed that upregulation of E-selectin precedes cytokine induction and is induced not only by infectious NiV but also by NiV-glycoprotein-containing virus-like particles. This demonstrates that very early events in NiV brain endothelial infection do not depend on NiV replication but can be triggered by the NiV glycoproteins alone.


Subject(s)
Cytokines/biosynthesis , Endothelial Cells/immunology , Glycoproteins/immunology , Host-Pathogen Interactions , Nipah Virus/immunology , Viral Proteins/immunology , Virosomes/immunology , Animals , Cells, Cultured , E-Selectin/biosynthesis , Endothelial Cells/drug effects , Swine , Up-Regulation
18.
J Virol ; 90(5): 2664-75, 2015 Dec 23.
Article in English | MEDLINE | ID: mdl-26699644

ABSTRACT

UNLABELLED: The capsid protein (VP1) of all caliciviruses forms an icosahedral particle with two principal domains, shell (S) and protruding (P) domains, which are connected via a flexible hinge region. The S domain forms a scaffold surrounding the nucleic acid, while the P domains form a homodimer that interacts with receptors. The P domain is further subdivided into two subdomains, termed P1 and P2. The P2 subdomain is likely an insertion in the P1 subdomain; consequently, the P domain is divided into the P1-1, P2, and P1-2 subdomains. In order to investigate capsid antigenicity, N-terminal (N-term)/S/P1-1 and P2/P1-2 were switched between two sapovirus genotypes GI.1 and GI.5. The chimeric VP1 constructs were expressed in insect cells and were shown to self-assemble into virus-like particles (VLPs) morphologically similar to the parental VLPs. Interestingly, the chimeric VLPs had higher levels of cross-reactivities to heterogeneous antisera than the parental VLPs. In order to better understand the antigenicity from a structural perspective, we determined an intermediate-resolution (8.5-Å) cryo-electron microscopy (cryo-EM) structure of a chimeric VLP and developed a VP1 homology model. The cryo-EM structure revealed that the P domain dimers were raised slightly (∼5 Å) above the S domain. The VP1 homology model allowed us predict the S domain (67-229) and P1-1 (229-280), P2 (281-447), and P1-2 (448-567) subdomains. Our results suggested that the raised P dimers might expose immunoreactive S/P1-1 subdomain epitopes. Consequently, the higher levels of cross-reactivities with the chimeric VLPs resulted from a combination of GI.1 and GI.5 epitopes. IMPORTANCE: We developed sapovirus chimeric VP1 constructs and produced the chimeric VLPs in insect cells. We found that both chimeric VLPs had a higher level of cross-reactivity against heterogeneous VLP antisera than the parental VLPs. The cryo-EM structure of one chimeric VLP (Yokote/Mc114) was solved to 8.5-Å resolution. A homology model of the VP1 indicated for the first time the putative S and P (P1-1, P2, and P1-2) domains. The overall structure of Yokote/Mc114 contained features common among other caliciviruses. We showed that the P2 subdomain was mainly involved in the homodimeric interface, whereas a large gap between the P1 subdomains had fewer interactions.


Subject(s)
Cryoelectron Microscopy , Sapovirus/chemistry , Sapovirus/ultrastructure , Virosomes/chemistry , Virosomes/ultrastructure , Amino Acid Sequence , Antibodies, Viral/immunology , Antigens, Viral/genetics , Antigens, Viral/immunology , Antigens, Viral/metabolism , Antigens, Viral/ultrastructure , Capsid Proteins/genetics , Capsid Proteins/immunology , Capsid Proteins/metabolism , Capsid Proteins/ultrastructure , Cross Reactions , Molecular Sequence Data , Protein Multimerization , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Recombinant Proteins/ultrastructure , Recombination, Genetic , Sapovirus/genetics , Sapovirus/immunology , Virosomes/genetics , Virosomes/immunology
19.
Curr Protoc Microbiol ; 38: 14B.5.1-26, 2015 Aug 03.
Article in English | MEDLINE | ID: mdl-26237105

ABSTRACT

Immunization with Human Papillomavirus (HPV) L1 virus-like particles or L2 capsid protein elicits neutralizing antibodies that mediate protection. A high-throughput and sensitive in vitro neutralization assay is therefore valuable for prophylactic HPV vaccine studies. Over several hours during infection of the genital tract, virions take on a distinct intermediate conformation, including a required furin cleavage of L2 at its N-terminus. This intermediate is an important target for neutralization by L2-specific antibody, but it is very transiently exposed during in vitro infection of most cell lines resulting in insensitive measurement for L2, but not L1-specific neutralizing antibodies. To model this intermediate, we describe a protocol to generate furin-cleaved HPV pseudovirions (fc-PsV), which deliver an encapsidated reporter plasmid to facilitate infectivity measurements. We also describe a protocol for use of fc-PsV in a high-throughput in vitro neutralization assay for the sensitive measurement of both L1 and L2-specific neutralizing antibodies.


Subject(s)
Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Furin/metabolism , Neutralization Tests/methods , Papillomaviridae/immunology , Virosomes/immunology , High-Throughput Screening Assays , Humans , Papillomaviridae/genetics , Virosomes/genetics
20.
J Virol Methods ; 222: 55-61, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26025459

ABSTRACT

Chikungunya fever (CHIKF) has re-emerged as an arboviral disease that mimics clinical symptoms of other diseases such as dengue, malaria, as well as other alphavirus-related illnesses leading to problems with definitive diagnosis of the infection. Herein we describe the development and evaluation of a sensitive epitope-blocking ELISA (EB-ELISA) capable of specifically detecting anti-chikungunya virus (CHIKV) antibodies in clinical samples. The assay uses a monoclonal antibody (mAb) that binds an epitope on the E2 protein of CHIKV and does not exhibit cross-reactivity to other related alphaviruses. We also demonstrated the use of recombinant CHIK virus-like particles (VLPs) as a safe alternative antigen to infectious virions in the assay. Based on testing of 60 serum samples from patients in the acute or convalescent phase of CHIKV infection, the EB-ELISA provided us with 100% sensitivity, and exhibited 98.5% specificity when Ross River virus (RRV)- or Barmah Forest virus (BFV)-immune serum samples were included. This assay meets the public health demands of a rapid, robust, sensitive and specific, yet simple assay for specifically diagnosing CHIK-infections in humans.


Subject(s)
Antibodies, Viral/blood , Antigens, Viral/immunology , Chikungunya Fever/diagnosis , Chikungunya virus/immunology , Enzyme-Linked Immunosorbent Assay/methods , Epitopes/immunology , Antibodies, Monoclonal/immunology , Antibodies, Viral/immunology , Chikungunya virus/isolation & purification , Humans , Sensitivity and Specificity , Virosomes/genetics , Virosomes/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...